US20190248922A1 - Affinity ligands and methods relating thereto - Google Patents

Affinity ligands and methods relating thereto Download PDF

Info

Publication number
US20190248922A1
US20190248922A1 US16/373,293 US201916373293A US2019248922A1 US 20190248922 A1 US20190248922 A1 US 20190248922A1 US 201916373293 A US201916373293 A US 201916373293A US 2019248922 A1 US2019248922 A1 US 2019248922A1
Authority
US
United States
Prior art keywords
affinity
elution
target
buffer
affinity ligands
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US16/373,293
Inventor
Achim Knappik
Stefan Paschen
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Bio Rad Laboratories Inc
Original Assignee
Bio Rad Laboratories Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Bio Rad Laboratories Inc filed Critical Bio Rad Laboratories Inc
Priority to US16/373,293 priority Critical patent/US20190248922A1/en
Assigned to BIO-RAD LABORATORIES, INC. reassignment BIO-RAD LABORATORIES, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: KNAPPIK, ACHIM, PASCHEN, Stefan
Publication of US20190248922A1 publication Critical patent/US20190248922A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/42Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against immunoglobulins
    • C07K16/4283Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against immunoglobulins against an allotypic or isotypic determinant on Ig
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/02Nasal agents, e.g. decongestants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01DSEPARATION
    • B01D15/00Separating processes involving the treatment of liquids with solid sorbents; Apparatus therefor
    • B01D15/08Selective adsorption, e.g. chromatography
    • B01D15/26Selective adsorption, e.g. chromatography characterised by the separation mechanism
    • B01D15/38Selective adsorption, e.g. chromatography characterised by the separation mechanism involving specific interaction not covered by one or more of groups B01D15/265 - B01D15/36
    • B01D15/3804Affinity chromatography
    • B01D15/3809Affinity chromatography of the antigen-antibody type, e.g. protein A, G, L chromatography
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J20/00Solid sorbent compositions or filter aid compositions; Sorbents for chromatography; Processes for preparing, regenerating or reactivating thereof
    • B01J20/281Sorbents specially adapted for preparative, analytical or investigative chromatography
    • B01J20/286Phases chemically bonded to a substrate, e.g. to silica or to polymers
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J20/00Solid sorbent compositions or filter aid compositions; Sorbents for chromatography; Processes for preparing, regenerating or reactivating thereof
    • B01J20/30Processes for preparing, regenerating, or reactivating
    • B01J20/32Impregnating or coating ; Solid sorbent compositions obtained from processes involving impregnating or coating
    • B01J20/3202Impregnating or coating ; Solid sorbent compositions obtained from processes involving impregnating or coating characterised by the carrier, support or substrate used for impregnation or coating
    • B01J20/3206Organic carriers, supports or substrates
    • B01J20/3208Polymeric carriers, supports or substrates
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01JCHEMICAL OR PHYSICAL PROCESSES, e.g. CATALYSIS OR COLLOID CHEMISTRY; THEIR RELEVANT APPARATUS
    • B01J20/00Solid sorbent compositions or filter aid compositions; Sorbents for chromatography; Processes for preparing, regenerating or reactivating thereof
    • B01J20/30Processes for preparing, regenerating, or reactivating
    • B01J20/32Impregnating or coating ; Solid sorbent compositions obtained from processes involving impregnating or coating
    • B01J20/3231Impregnating or coating ; Solid sorbent compositions obtained from processes involving impregnating or coating characterised by the coating or impregnating layer
    • B01J20/3242Layers with a functional group, e.g. an affinity material, a ligand, a reactant or a complexing group
    • B01J20/3268Macromolecular compounds
    • B01J20/3272Polymers obtained by reactions otherwise than involving only carbon to carbon unsaturated bonds
    • B01J20/3274Proteins, nucleic acids, polysaccharides, antibodies or antigens
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/14Extraction; Separation; Purification
    • C07K1/16Extraction; Separation; Purification by chromatography
    • C07K1/22Affinity chromatography or related techniques based upon selective absorption processes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/06Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies from serum
    • C07K16/065Purification, fragmentation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/42Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against immunoglobulins
    • C07K16/4283Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against immunoglobulins against an allotypic or isotypic determinant on Ig
    • C07K16/4291Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against immunoglobulins against an allotypic or isotypic determinant on Ig against IgE
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/21Immunoglobulins specific features characterized by taxonomic origin from primates, e.g. man
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'

Definitions

  • the invention relates to affinity ligands, such as antibody affinity ligands, and methods for using, identifying, and making affinity ligands.
  • sequence listing is submitted electronically via EFS-Web as an ASCII formatted sequence listing with a file named 1010888-105410US_ST25.txt, created on May 26, 2016, and having a size of 103,871 bytes and is filed concurrently with the specification.
  • sequence listing contained in this ASCII formatted document is part of the specification and is herein incorporated by reference in its entirety.
  • Affinity chromatography is a method of separating biochemical mixtures based on highly specific interactions between an affinity ligand and its target, such as that between antibody and antigen.
  • An affinity ligand that selectively interacts with the desired target is immobilized onto a solid support in order to create an affinity matrix that can be used in a column format.
  • Affinity chromatography can be used in a number of applications, including nucleic acid purification, protein purification from cell free extracts or cell culture supernatants, and purification from blood.
  • the present disclosure provides methods of generating affinity ligands, uses of such affinity ligands, and specific affinity ligands.
  • affinity ligands that bind specifically to a target molecule, wherein the specific binding strength of the affinity ligand to the target molecule is reduced under buffer conditions including (i) a pH of about 4.0 to about 5.5 or (ii) about 1-2 M MgCl 2 .
  • the buffer condition may have a pH of about 4.0 to about 5.0. In some instances, the buffer condition may have a pH of about 4.0. In some instances, the buffer condition may have a pH of about 5.0. In some instances, the buffer condition may include 2 M MgCl 2 . In some instances, the buffer condition may include 2 M MgCl 2 and a relatively neutral pH.
  • the target molecule may be an immunoglobulin.
  • the target molecule may be an immunoglobulin selected from the group consisting of an immunoglobulin M (IgM), an immunoglobulin A (IgA), or an immunoglobulin E (IgE).
  • the affinity ligand may be an immunoglobulin.
  • the affinity ligand may be a recombinant Fab fragment or Fab fragment derivative.
  • the affinity ligand may be an anti-IgE antibody comprising heavy chain complementarity determining regions CDR1, CDR2, and CDR3 sequences selected from any of the sequences set forth in FIG. 1A .
  • the affinity ligand may be an anti-IgE antibody having a light chain complementarity determining regions CDR1, CDR2, and CDR3 sequences selected from any of the sequences set forth in FIG. 1B .
  • the affinity ligand may be an anti-IgA antibody comprising heavy chain complementarity determining regions CDR1, CDR2, and CDR3 sequences selected from any of the sequences set forth in FIG. 2A . In some instances, the affinity ligand may be an anti-IgA antibody comprising light chain complementarity determining regions CDR1, CDR2, and CDR3 sequences selected from any of the sequences set forth in FIG. 2B . In some instances, the affinity ligand may be an anti-IgM antibody comprising heavy chain complementarity determining regions CDR1, CDR2, and CDR3 sequences selected from any of the sequences set forth in FIG. 3A . In some instances, the affinity ligand may be an anti-IgM antibody comprising light chain complementarity determining regions CDR1, CDR2, and CDR3 sequences selected from any of the sequences set forth in FIG. 3B .
  • the affinity ligand may be linked to a solid support.
  • the solid support may be a bead or a sample plate.
  • the bead may be an agarose bead, a polystyrene bead, a polymethacrylate bead, a polyacrylamide bead, a magnetic bead, or a paramagnetic bead.
  • a target molecule comprising the steps of: providing a solid support linked to an affinity ligand; contacting the solid support with a sample containing the target molecule; washing the solid support with a wash buffer to remove unbound components of the sample; and eluting bound target molecule from the solid support with an elution buffer comprising (i) a pH of about 4.0 to about 5.5 or (ii) about 1-2 M MgCl 2 .
  • the elution buffer may include a pH of about 4.0 to about 5.5 and a relatively low salt concentration. In some instances, the elution buffer may include about 1 M to 2 M MgCl 2 and a relatively neutral pH. In some instances, the elution buffer may include about 1 M to 2 M MgCl 2 and a pH of about 6.0 to 8.0. In some instances, the eluting may be a single-step elution with an elution buffer comprising (i) a pH of about 4.0 to about 5.5 or (ii) about 1-2 M MgCl 2 .
  • the eluting may be a multiple-step elution with a plurality of elution buffers comprising (i) a pH of about 4.0 to about 5.5 or (ii) about 1-2 M MgCl 2 , wherein the plurality of elution buffers are applied to the solid support sequentially, wherein elution buffers having higher salt concentrations are applied after elution buffers having lower salt concentrations and elution buffers having lower pH are applied after elution buffers having higher pH.
  • the affinity ligand may be an immunoglobulin.
  • the target molecule may be an immunoglobulin M (IgM), an immunoglobulin A (IgA), or an immunoglobulin E (IgE).
  • the affinity ligand may be an anti-IgE antibody comprising heavy chain complementarity determining regions CDR1, CDR2, and CDR3 sequences selected from any of the sequences set forth in FIG. 1A .
  • the affinity ligand may be an anti-IgE antibody comprising light chain complementarity determining regions CDR1, CDR2, and CDR3 sequences selected from any of the sequences set forth in FIG. 1B .
  • the affinity ligand may be an anti-IgA antibody comprising heavy chain complementarity determining regions CDR1, CDR2, and CDR3 sequences selected from any of the sequences set forth in FIG. 2A . In some instances, the affinity ligand may be an anti-IgA antibody comprising light chain complementarity determining regions CDR1, CDR2, and CDR3 sequences selected from any of the sequences set forth in FIG. 2B . In some instances, the affinity ligand may be an anti-IgM antibody comprising heavy chain complementarity determining regions CDR1, CDR2, and CDR3 sequences selected from any of the sequences set forth in FIG. 3A . In some instances, the affinity ligand may be an anti-IgM antibody comprising light chain complementarity determining regions CDR1, CDR2, and CDR3 sequences selected from any of the sequences set forth in FIG. 3B .
  • an affinity ligand that specifically binds to a target molecule under neutral buffer conditions and has reduced binding strength to the target molecular under mild elution conditions including the steps of: expressing a naive affinity ligand library to produce a plurality of affinity ligands; providing a solid support linked to a target; contacting the solid support with the plurality of affinity ligands; washing the solid support with a wash buffer to remove unbound affinity ligands, wherein the wash buffer comprises neutral buffer conditions; contacting the solid support with an elution buffer comprising (i) a pH of about 4.0 to about 5.5 or (ii) about 1-2 M MgCl 2 ; and identifying affinity ligands that substantially dissociate from the solid support in the elution buffer.
  • the affinity ligand library may not be preselected for characteristics favoring reduced binding strength to the target molecule under mild elution conditions.
  • the plurality of affinity ligands may be encoded by a plurality of nucleic acid sequences.
  • the plurality of nucleic acid sequences includes a heterologous promoter operably linked thereto.
  • the plurality of affinity ligands may be expressed on a plurality of phage.
  • the elution buffer may have a pH of about 4.0 to about 5.5 and a relatively low salt concentration. In some instances, the elution buffer may include about 1 M to 2 M MgCl 2 and may have a relatively neutral pH. In some instances, the elution buffer may include about 1 M to 2 M MgCl 2 and may have a pH of about 6.0 to 8.0. In some instances, the wash buffer may have a pH of 6.0-8.0. In some instances, the wash buffer may have a relatively low salt concentration.
  • the target may be an immunoglobulin.
  • the target may be an immunoglobulin M (IgM), an immunoglobulin A (IgA), or an immunoglobulin E (IgE).
  • the plurality of affinity ligands may be a plurality of antibodies or derivatives thereof. In some instances, the plurality of affinity ligands is a plurality of Fab fragments or derivatives thereof. In some instances, the affinity ligand identified may be encoded by a polynucleotide comprising a nucleic acid sequence encoding heavy chain complementarity determining regions CDR1, CDR2, and CDR3 sequences selected from any of the sequences set forth in FIG. 1A .
  • the affinity ligand identified may be encoded by a polynucleotide comprising a nucleic acid sequence encoding light chain complementarity determining regions CDR1, CDR2, and CDR3 sequences selected from any of the sequences set forth in FIG. 3B .
  • kits including the affinity ligand described above.
  • FIG. 1A and FIG. 1B show heavy and light chain CDR sequences, respectively, for anti-IgE antibodies according to some examples.
  • FIG. 2A and FIG. 2B show heavy and light chain CDR sequences, respectively, for anti-IgA antibodies according to some examples.
  • FIG. 3A and FIG. 3B show heavy and light chain CDR sequences, respectively, for anti-IgM antibodies according to some examples.
  • FIGS. 4A-4I show heavy and light chain sequences for anti-IgM antibodies according to some examples.
  • FIG. 5 shows ELISA results for a first set of anti-IgM antibody affinity ligands assessing binding to and elution of target molecules according to one example.
  • FIG. 6 shows ELISA results for a second set of anti-IgM antibody affinity ligands assessing binding to and elution of target molecules according to one example.
  • FIG. 7 shows ELISA results for a first set of anti-IgA antibody affinity ligands assessing binding to and elution of target molecules according to one example.
  • FIG. 8 shows ELISA results for a second set of anti-IgA antibody affinity ligands assessing binding to and elution of target molecules according to one example. Controls N1-CD33-6 ⁇ His and BSA are also shown.
  • FIG. 9 shows ELISA results for a set of anti-IgE antibody affinity ligands assessing binding specificity for target molecules according to one example.
  • FIG. 10 shows elution profiles of purified AbD18705 hIgM target molecule from affinity ligand columns according to one example.
  • the affinity column on the left uses anti-IgM antibody ligand AbD20775.2, and the affinity column on the right uses anti-IgM antibody ligand AbD20771.2. Collected fractions (A#) are shown across the bottom of each graph.
  • FIG. 11 shows SDS-PAGE analysis of purified AbD18705 hIgM target molecule elution fractions according to one example.
  • the left gel shows the purified AbD18705 hIgM from the indicated fractions (each fraction shown under reducing and under oxidizing conditions).
  • Purified human IgM (product number OBT1524) is shown as a control in the gel on the right, again under reducing and oxidizing conditions.
  • FIG. 12 shows an overlay of size exclusion chromatography runs for purified AbD18705 hIgM target molecule elution fractions identified in FIG. 10 according to one example.
  • FIG. 13 shows a graph illustrating the results of an ELISA assay assessing the activity and specificity of the purified AbD18705 hIgM target molecule (“His-GFP) for its antigen GFP according to one example. Controls GST, N1-CD33-6 ⁇ His, and BSA are also shown.
  • Affinity ligand refers to a composition (such as, for example, an antibody or non-antibody protein), that binds specifically to a specific substance, such as a protein, protein complex, or organic compound having a defined structure.
  • solid support is used herein to denote a solid inert surface or body to which an agent, such as an antibody or an antigen, that is reactive in any of the binding reactions described herein can be immobilized.
  • agent such as an antibody or an antigen
  • immobilized denotes a molecularly-based coupling that is not dislodged or de-coupled under any of the conditions imposed during any of the steps of the assays described herein. Such immobilization can be achieved through a covalent bond, an ionic bond, an affinity-type bond, or any other covalent or non-covalent bond.
  • antibody refers to an immunoglobulin, composite, or fragmentary form thereof.
  • the term may include but is not limited to polyclonal or monoclonal antibodies of the classes IgA, IgD, IgE, IgG, and IgM, derived from human or other cell lines, including natural or genetically modified forms such as humanized, human, single-chain, chimeric, synthetic, recombinant, hybrid, mutated, grafted, and in vitro generated antibodies and fragments thereof.
  • Antibody may also include composite forms including but not limited to fusion proteins containing an immunoglobulin moiety.
  • Antibody may also include non-quaternary antibody structures (such as camelids and camelid derivatives).
  • Antibody may also include antibody fragments such as Fab (fragment-antigen binding), F(ab′)2, Fv, scFv, Fd, dAb, Fc and other compositions that retain antigen-binding function.
  • antibody includes aggregates, polymers, and conjugates of immunoglobulins or their fragments, where the molecules largely retain binding affinity for their epitope(s).
  • an “antibody” may be modified, such as, for example, by linking to a chemical or peptide moiety or detectable tag moiety.
  • CDR complementarity determining region
  • HVR hypervariable region
  • a “constant region” refers to a region in the heavy and light chains of an antibody having relatively less variability compared to the N′ terminal variable region of the heavy and light chains of an antibody.
  • the constant region is generally the same in all antibodies of the same isotype and differs in antibodies of different isotypes.
  • Neutral buffer or “neutral buffer condition” refers to a buffer having approximately physiological pH. Such buffers/conditions allow binding of proteins to an affinity column without resulting in substantial protein denaturation or aggregation.
  • a neutral buffer or neutral buffer condition may permit near optimal interaction between an affinity ligand and a target molecule.
  • a neutral buffer condition or neutral buffer generally has a pH in the range of about 6.0 to 8.0.
  • Physiological pH refers to the pH of human blood, which is about 7.4. A pH in the range of pH 7.0 to pH 7.8 may be considered approximately physiological pH.
  • “Naive expression library,” for the purposes of expressing affinity ligands, refers to an expression library that expresses a large number of recombinant proteins, polypeptides, or peptides that have a large diversity of structure or specificity in their binding domains.
  • a naive library is one generated from synthetic or natural ligand repertoires that have not previously subjected to selections for increased affinity for a target.
  • a naive expression library is generated from a plurality of nucleic acid sequences that encode a plurality of affinity ligands or at least a plurality of target binding regions.
  • “Relatively neutral pH” refers to a pH of about pH 6.0 to 8.0.
  • a “solid support” refers to a material or group of materials having a rigid or semi-rigid surface or surfaces.
  • the solid support takes the form of thin films or membranes, beads, bottles, dishes, fibers, woven fibers, shaped polymers, particles, and microparticles, including but not limited to, microspheres.
  • a solid support can be formed, for example, from an inert solid support of natural material, such as glass and collagen, or synthetic material, such as acrylamide, cellulose, nitrocellulose, silicone rubber, polystyrene, polyethylene vinyl acetate, polypropylene, polymethacrylate, polyethylene, polysilicates, polyethylene oxide, polycarbonates, teflon, fluorocarbons, nylon, polyanhydrides, polyglycolic acid, polylactic acid, polyorthoesters, polypropylfumarate, glycosaminoglycans, and polyamino acids.
  • natural material such as glass and collagen
  • synthetic material such as acrylamide, cellulose, nitrocellulose, silicone rubber, polystyrene, polyethylene vinyl acetate, polypropylene, polymethacrylate, polyethylene, polysilicates, polyethylene oxide, polycarbonates, teflon, fluorocarbons, nylon, polyanhydrides, polyglycolic acid, polylactic acid
  • some functional groups naturally present on the surface of a carrier for example, carboxylic acid (—COOH), free amine (—NH2), and sulfhydryl (—SH) groups
  • a carrier for example, carboxylic acid (—COOH), free amine (—NH2), and sulfhydryl (—SH) groups
  • carboxylic acid (—COOH) for example, carboxylic acid (—COOH), free amine (—NH2), and sulfhydryl (—SH) groups
  • a desired functional group such as a carboxylic acid group, or a moiety known to be a partner of a binding interaction (such as avidin that is capable of binding biotin) may be attached to such solid support.
  • the solid support is a carboxylated latex or magnetic microsphere.
  • binding refers to a binding reaction where two members of a binding pair (for example, an antibody and a molecule comprising the antibody's target epitope) bind to each other with an affinity that is at least 10-fold better than the members' affinity for other components of a heterogeneous mixture (for example, a hybridoma culture supernatant or other mixture of proteins).
  • a binding pair for example, an antibody and a molecule comprising the antibody's target epitope
  • affinity ligands such as antibodies
  • target molecules such as immunoglobulins M, A, or E
  • mild conditions can be used to elute the target molecule to which the affinity ligand specifically binds, thereby avoiding harsh elution conditions used previously.
  • Certain examples and features of the present disclosure relate to methods to identify affinity ligands, such as antibodies, that are especially useful for mild elution immunoaffinity chromatography.
  • Such ligands when attached to a solid support, allow one-step purification of target molecules using mild elution conditions.
  • the use of mild elution conditions circumvents the typically harsh elution conditions used in this type of chromatography that may lead to denaturing or aggregation of target molecules (such as low pH, for example, pH 3.0).
  • the described methods utilize in vitro enrichment methods to permit selection of affinity ligands for which mild elution conditions can be used. The selection of such ligands is performed by a first binding step under conditions that allow binding in standard/neutral buffer conditions (such as neutral pH and/or low salt), followed by a subsequent elution step that is performed using mild elution conditions.
  • the described methods permit specific selection and enrichment and finally isolation of affinity ligands having the desired characteristics. As such, affinity ligands of interest can be generated more rapidly and efficiently using the described methods.
  • affinity ligands such as antibodies, that exhibit the property of specifically binding to a target under neutral conditions and releasing the target under mild elution conditions.
  • affinity ligands can be selected for using the method described above. Examples of affinity ligands that are specific for human immunoglobulin M (IgM), human immunoglobulin A (IgA), and human immunoglobulin E (IgE) are described.
  • affinity ligands may be used linked to a solid support as an affinity purification column.
  • some targets that are sensitive to harsh elution conditions for example, pH 3.0
  • affinity ligands are useful for isolating IgM, IgA, or IgE molecules as these molecules may be sensitive to denaturation when affinity purified using harsh elution conditions.
  • Other molecules that are sensitive to denaturation can be purified accordingly, once an affinity ligand has been isolated using the methods described in this disclosure.
  • affinity ligand compositions are described herein that bind specifically to a target and from which target can be eluted using mild elution conditions.
  • the affinity ligand may be an antibody, antibody-like molecule, or other affinity-binding molecule.
  • this disclosure will sometimes describe examples and features of the affinity ligands in the context of antibody affinity ligands. However, this disclosure is not limited to affinity ligands that are antibodies.
  • the affinity ligand may be selected from a variety of different types of protein or non-protein compositions.
  • an affinity ligand is an antibody, antibody-like molecule, or any other affinity-binding molecule derived from a naive expression library as described below in Section II.
  • the antibody may be a monoclonal antibody, a Fab fragment, a F(ab′)2, an Fv, a scFv, an Fd, a dAb, an Fc fragment, a VHH, or other fragments thereof that retain antigen-binding function.
  • affinity ligand may be recombinant antibodies having heterologous constant and variable domains; for example, generated by a recombinant protein expression library.
  • the affinity ligand may be a recombinant Fab fragment.
  • the Fab fragment may have the variable domain and the first constant domain (Fd chain) of one heavy chain plus one complete light chain (L chain).
  • the Fd and L chain are linked by strong non-covalent interactions and can be covalently linked by a disulfide bond.
  • the polynucleotide sequence encoding the recombinant Fab fragment may be subcloned into an expression vector containing a heterologous promoter to drive expression of the recombinant Fab fragment.
  • the Fab fragment may monovalent, bivalent, or multivalent. In some instances, the Fab fragment is monovalent.
  • the affinity ligand is a single-chain variable (scFv) fragment or a bivalent scFv fragment (diabody).
  • ScFv fragments typically have one VH and one VL chain expressed as a single polypeptide joined by a peptide linker.
  • the polypeptide linker stabilizes the interaction between the VH and VL chains.
  • the polynucleotide sequence encoding the recombinant scFv fragment may be subcloned into an expression vector containing a heterologous promoter to drive expression of the recombinant scFv fragment.
  • the affinity ligand may be a recombinant antibody or protein that specifically binds to targets in a manner similar to antibodies, or fragment thereof that retains antigen-binding function.
  • the affinity ligand may be a recombinant protein that contains at least three complementarity determining regions (CDRs) that cause the specific binding of the affinity ligand to the target; for example, three heavy chain CDRs and, in some examples, contains six CDRs (three heavy chain and three light chain).
  • the antibody may be a variable domain of heavy chain (VHH) antibody or a nanobody (a monomeric variable domain antibody).
  • VHH or nanobody may be encoded by a singly polypeptide.
  • the affinity ligand is a camelid antibody or camelid nanobody.
  • Camelid antibodies are certain IgG antibodies from the mammalian family of camel and dromedary ( Camelus bactrianus and Camelus dromaderius ) family, including new world members such as llama species (such as Lama paccos, Lama glama and Lama vicugna ), that lack light chains. See, for example, International Appl. WO 94/04678.
  • the small single variable domain (VHH) of the camelid antibody can be used to as the basis of a low molecular weight antibody-derived protein known as a “camelid nanobody” having high affinity for a target. See U.S. Pat. No.
  • the affinity ligand can be a compound or non-antibody protein that specifically binds to targets in a manner similar to antibodies.
  • Certain of these “antibody mimics” use non-immunoglobulin protein scaffolds as alternative protein frameworks for the variable regions of antibodies.
  • the affinity ligand can be a monobody, which are small antibody mimics using the scaffold of a fibronectin type III domain (FN3).
  • FN3 scaffold functions as an effective framework onto which loops for specific building functions can be grafted.
  • the affinity ligand may utilize the tenth FN3 unit of human fibronectin as scaffold. It is small, monomeric, and does not have disulfide bonds.
  • FN3-based antigen-binding molecules can be prepared using methods described in the art. For example, see Koide et al., J. Mol. Biol. 284: 1141-1151, 1998; Koide et al., Proc. Natl Acad. Sci. USA 99:1253-1258, 2002; and Batori et al., Protein Eng. 15:1015-20, 2002, and U.S. Pat. Nos. 6,818,418 and 7,115,396.
  • the affinity ligand may be a single polypeptide chain binding molecule that contains the antigen binding sites of both the heavy and light variable regions of an antibody connected by a peptide linker and will fold into a structure similar to that of the two peptide antibody.
  • the affinity ligand may be a recombinant protein containing derivative sequences of one or more loops of cytochrome b562 that are selected for binding specificity to the target. See, for example, Ku et al., Proc. Natl. Acad. Sci. U.S.A. 92(14):6552-6556 (1995).
  • the affinity ligand may be an antibody mimic based on a lipocalin scaffold, in which one or more of the hypervariable loops of the lipocalin protein are randomized and selected for specific binding to the target. See, for example, Beste et al., Proc. Natl. Acad. Sci.
  • the affinity ligand may be a synthetic antibody mimic using the rigid, non-peptide organic scaffold of calixarene to which are attached multiple variable peptide loops used as binding sites. See, for example, U.S. Pat. No. 5,770,380.
  • the affinity ligand may be an antibody-like binding peptidomimetic. See, for example, Murali et al., Cell. Mol. Biol. 49(2):209-216 (2003).
  • the affinity ligand may include a scaffold derived from one or more A-domains.
  • the affinity ligand may include multiple A-domains, each of which binding independently to a distinct epitope of the target.
  • Such affinity ligands can be generated using methods described in, for example, Gliemann et al., Biol. Chem. 379:951-964, 1998; Koduri et al., Biochemistry 40:12801-12807, 2001 and Silverman et al., Nat Biotechnol. 23:1556-61, 2005.
  • Other exemplary non-antibody scaffolds for use as ligands include darpins, affimers, cystine-knot mini-proteins, affilins, and peptides or non-protein-based scaffolds like aptamers.
  • the affinity ligand may comprise an affinity tag or moiety.
  • the affinity tag or moiety may be useful for purification of the affinity ligand or attachment to a solid support.
  • the affinity ligand may include at least one of a FLAG® peptide, 6 ⁇ -Histidine (6 ⁇ His) peptide, etc.
  • the affinity ligand is chemically modified to facilitate attachment of the ligand to a solid support.
  • the affinity ligand binds specifically to its target under neutral buffer conditions.
  • Neutral buffer conditions may include a pH of approximately physiological pH, or relatively neutral pH, such as, for example a pH of about 6.0, 6.2, 6.5, 6.8, 7.1, 7.3, 7.5, 7.8, 7.9, 8.1, or a pH of about 6.0 to about 8.0.
  • the neutral buffer condition also comprises a relatively low salt concentration and/or ionic strength.
  • the salt concentration may be approximately physiological salt concentration.
  • the neutral buffer conditions include approximately physiological ionic strength.
  • neutral buffer conditions may be used for a wash buffer.
  • the wash buffer may have somewhat higher salt concentration or ionic strength to improve stringency and reduce non-specific binding of non-targets to the affinity ligand.
  • the neutral buffer condition does not include a salt.
  • the neutral buffer condition may include detergent to reduce non-specific binding of non-targets to the affinity ligand.
  • the neutral buffer condition comprises a buffering agent.
  • a neutral wash buffer may be phosphate buffered saline (PBS).
  • the neutral wash buffer may be PBS containing about 0.05-1.0% Tween-20® (for example, 0.1%). Other solutions and detergents are contemplated.
  • the mild elution conditions may comprise a pH of about 4.0 to about 5.5. In another aspect, the mild elution conditions may comprise about 1-2 M MgCl 2 .
  • mild elution conditions may comprise a pH in the range of about 4.0 to about 5.5.
  • mild elution conditions may include a pH of at least about 4.0 but less than or equal to about 5.5.
  • the mild elution condition may include a pH greater than or equal to about 4.0, a pH of about 4.5, a pH of about 5.0, or a pH of about 5.5, or any pH within the range of about 4.0 to about 5.5.
  • the elution buffer may further comprise a relatively low salt concentration.
  • the elution buffer may contain salt conditions relatively similar to physiological salt conditions.
  • the salt concentration may be 25 mM, 50 mM, 100 mM, 125 mM, 150 mM, 175 mM, 200 mM, 225 mM, or 250 mM.
  • the elution buffer may include about 150 mM NaCl.
  • the mild elution conditions include approximately physiological ionic strength.
  • the elution buffer may further include a buffering agent.
  • Exemplary buffering agents include citrate, sodium acetate, and sodium phosphate buffered saline (PBS).
  • the elution buffer may contain 100 mM citrate buffer.
  • the elution buffer may contain 100 mM sodium acetate.
  • the elution buffer may contain 1 ⁇ PBS.
  • the elution buffer may contain 150 mM NaCl and 100 mM citrate buffer.
  • mild elution conditions may comprise a salt concentration of about 1 M to 2 M MgCl 2 .
  • the salt concentration may be about 0.8 M, about 1 M, about 1.2 M, about 1.4 M, about 1.6 M, about 1.8 M, about 2 M, or about 2.2 M.
  • the mild elution condition comprises 2 M MgCl 2 .
  • the mild elution conditions may further comprise a relatively neutral pH, or an approximately physiological pH, or a pH of about 6.8 to about 7.9, or a pH of about 6.0 to about 8.0, or any pH with these ranges.
  • the pH may be about 6.0, about 6.2, about 6.4, about 6.6, about 6.8, about 7.0, about 7.2, about 7.4, about 7.6, about 7.8, or about 8.0, when the mild elution condition includes a salt concentration of about 1 M to 2 M MgCl 2 .
  • the elution buffer may further include a buffering agent.
  • Exemplary buffering agents include citrate, sodium acetate, and phosphate buffered saline (PBS).
  • PBS phosphate buffered saline
  • the buffering agent may be of sufficient concentration to provide pH buffering.
  • the elution buffer may contain 100 mM citrate.
  • the elution buffer may contain 100 mM sodium acetate.
  • the elution buffer may contain 1 ⁇ PBS.
  • the mild elution condition comprises a pH in the range of about 4.0 to 5.5
  • the neutral buffer conditions generally have a higher pH of approximately neutral pH or approximately physiological pH. It is also understood that, where the mild elution condition comprises about 1M to 2M MgCl 2 , the neutral buffer conditions generally have a lower salt concentration or ionic strength.
  • the affinity ligand binds specifically to a target epitope of a target molecule.
  • the target epitope can be a portion of a target molecule, such as a protein, nucleic acid, or other biological molecule.
  • the target may be a protein or other molecule that is sensitive to harsh buffer conditions, including low pH (such as pH 3.0).
  • a target may denature, dissociate into individual subunits, or dissociate from cofactors under harsh buffer conditions.
  • the target molecule may be an immunoglobulin (antibody), such as an immunoglobulin G (IgG), an immunoglobulin M (IgM), an immunoglobulin A (IgA), or an immunoglobulin E (IgE).
  • the affinity ligand may be a Fab fragment that binds specifically to an IgG, an IgM, an IgA, or an IgE.
  • the affinity ligand may be a Fab fragment that binds specifically to an IgE having any of the heavy and light chain CDR sequences as set forth in FIG. 1A and FIG. 1B , respectively.
  • the antibodies specific for IgE may contain a combination of CDR1, CDR2, and CDR3 sequences as set forth in FIG. 1A or FIG. 1B .
  • such affinity ligand antibodies will have reduced binding to IgE under mild elution conditions, such as at a pH equal to or greater than about 4.0 and less than or equal to about 5.5, such that the IgE elutes from the affinity agent.
  • certain anti-IgE antibodies substantially dissociate from their target under elution conditions comprising pH 4.0 and pH 5.0.
  • the elution buffer conditions also comprise relatively low salt, such as 150 mM NaCl, and a buffering agent, such as citrate.
  • relatively low salt such as 150 mM NaCl
  • buffering agent such as citrate.
  • Other suitable salts and buffering agents are also contemplated.
  • the affinity ligand may be a Fab fragment that binds specifically to an IgA having any of the heavy and light chain CDR sequences as set forth in FIG. 2A and FIG. 2B , respectively.
  • the antibodies specific for IgA may contain a combination of CDR1, CDR2, and CDR3 sequences as set forth in FIG. 2A or FIG. 2B .
  • such affinity ligand antibodies will have reduced binding to IgA under mild elution conditions, such as at a pH equal to or greater than about 4.0 and less than or equal to about 5.5, such that the IgE elutes from the affinity agent.
  • certain anti-IgA antibodies substantially dissociate from their target under elution conditions comprising pH 4.0 and pH 5.0.
  • the elution buffer conditions also comprise relatively low salt, such as 150 mM NaCl, and a buffering agent, such as citrate.
  • relatively low salt such as 150 mM NaCl
  • buffering agent such as citrate.
  • Other suitable salts and buffering agents are also contemplated.
  • the affinity ligand may be a Fab fragment that binds specifically to an IgM having any of the heavy and light chain CDR sequences as set forth in FIG. 3A and FIG. 3B , respectively.
  • the affinity ligand may have at least one of the heavy chain sequences and at least one of the light chain sequences shown in FIGS. 4A-4I .
  • the affinity ligand may have at least one heavy and light chain sequences pair as shown in FIGS. 4A-4I .
  • the antibodies specific for IgM may contain a combination of CDR1, CDR2, and CDR3 sequences as set forth in FIG. 3A or FIG. 3B .
  • affinity ligand antibodies will have reduced binding to IgM under mild elution conditions, such as at a pH equal to or greater than about 4.0 and less than or equal to about 5.5, such that the IgM elutes from the affinity agent.
  • mild elution conditions such as at a pH equal to or greater than about 4.0 and less than or equal to about 5.5, such that the IgM elutes from the affinity agent.
  • certain anti-IgM antibodies substantially dissociate from their target under elution conditions comprising pH 4.0 and pH 5.0.
  • the elution buffer conditions also comprise relatively low salt, such as 150 mM NaCl, and a buffering agent, such as citrate. Other suitable salts and buffering agents are also contemplated.
  • the affinity ligand may have CDR sequences similar to those identified in this disclosure except varying in amino acid sequence at one or two amino acid positions. In some instances, the variance in sequence is a conservative amino acid change.
  • affinity ligands that are specific for immunoglobulin targets
  • affinity ligands specific for other types of ligands are also contemplated.
  • affinity ligands in particular antibodies
  • affinity chromatography methods using mild elution conditions.
  • a plurality of potential affinity ligands are provided and processed such that ligands are selected that specifically bind to a target under neutral buffer conditions and display weak specific binding strength to the target under mild elution conditions.
  • naive expression libraries are useful for expressing a large number of recombinant proteins, polypeptides, and peptides, including, for example, antibodies, that have a large diversity of structure or specificity in their binding domains.
  • a naive library is one generated from synthetic or natural ligand repertoires that have not previously subjected to selections for increased affinity for a target.
  • a naive library is generated from a plurality of nucleic acid sequences that encode a plurality of affinity ligands or at least a plurality of target binding regions.
  • the nucleic acid sequences encoding the target binding regions of the plurality of affinity ligands may have randomized sequences to generate binding regions in the plurality of affinity ligands that have randomized amino acid sequences.
  • the expression library expresses proteins or polypeptides having a structural domain with a common amino sequence and a binding domain having variable sequences such that the proteins or polypeptides expressed by the library may display a large number of distinct binding domains based on sequence variability. As such, different proteins or polypeptides expressed by the library may have different binding specificities for different targets under different conditions.
  • the expression libraries express hundreds of thousands, millions, or billions of different proteins or polypeptides, each of which may have a distinct binding affinity and, thus, may bind to different targets with different degrees of specificity under different conditions.
  • Expression libraries useful for expressing a plurality of affinity ligands for use in this method include phage display libraries, yeast display libraries, ribosome display, mRNA display or other selection system, or any other recombinant expression library capable of expressing a plurality of affinity ligands having a range of binding specificities.
  • an example of a useful affinity ligand expression library is the HuCAL Platinum® Platform (AbD Serotec, Bio-Rad), which provides phage display libraries of antibodies in Fab format representing an extensive array (>10 10 members) of CDR sequence variability (Prassler, J., et al. (2011). “HuCAL PLATINUM, a synthetic Fab library optimized for sequence diversity and superior performance in mammalian expression systems.” J. Mol. Biol. 413(1): 261-278).
  • the variability of the plurality of affinity ligands expressed by the library facilitate the identification of ligands that bind to a target with specific binding affinity under neutral buffer conditions and have substantially reduced specific binding under mild elution conditions as described in Section I.
  • a target of interest is linked or adsorbed to a solid support.
  • the target may be such as a protein, nucleic acid, or other biomolecule as described above.
  • the solid support may be the wall or floor of an assay vessel, or a dipstick or other implement to be inserted into an assay vessel, or particles (such as magnetic beads) placed inside or suspended in an assay vessel.
  • Particles, and especially beads can be useful in many embodiments, including beads that are microscopic in size (microparticles) and formed of a polymeric material.
  • the target-linked solid support may be blocked with a blocking buffer.
  • the blocking buffer may contain an animal protein blocker (such a milk or bovine serum albumin), a non-animal protein blocker (such as ChemiBLOCKERTM), or a detergent (such a Tween-20®).
  • the blocking buffer may contain a closely related antigen.
  • the target is an IgM antibody with kappa light chain
  • the blocking buffer may contain an antibody of different isotype (e.g., IgG) with kappa light chain as a blocker.
  • the target-linked solid support may be blocked with one or more types of antibody light chains as a blocker.
  • the ligand is human IgM or human IgA
  • the target-linked solid support may be blocked with human IgG1 lambda or IgG1 kappa as described in Examples 1 and 2.
  • the blocking buffer may contain culture media (for example, media that sustain growth of a eukaryotic cell culture producing immunoglobulin like IgE, IgA or IgM).
  • Culture media for example, media that sustain growth of a eukaryotic cell culture producing immunoglobulin like IgE, IgA or IgM.
  • Blocking with culture medium may be useful to avoid enrichment of affinity ligands with cross-reactivity to components in the culture medium. In some instances, this can be useful because the target proteins are expressed in cells that are grown in culture medium. For example, as described in Examples 1 and 2, where the ligand is human IgE, the target-linked solid support may be blocked with culture medium.
  • the expressed affinity ligands can be brought into contact with the target-linked solid support under neutral buffer conditions to achieve binding of the affinity ligands, if any ligands in the library have affinity for the target.
  • a washing step may be performed using neutral buffer conditions as described above in Section I.
  • An elution step may then be performed using the mild elution conditions as described above in Section I. Under such mild elution conditions, the binding of some of the affinity ligands to its target may be sufficiently reduced such that the affinity ligand and the target substantially dissociate from each other under these conditions.
  • the mild elution conditions may comprise a pH of about 4.0 to about 5.5. In another aspect, the mild elution conditions may comprise about 1-2 M MgCl 2 .
  • the described methods allow for specific selection and enrichment of affinity ligands that specifically bind to a target under neutral buffer conditions and have substantially reduced specific binding to the target under mild elution conditions.
  • Various elution conditions may be assessed to determine the optimal elution condition for a particular affinity ligand-target combination.
  • Control elution conditions in particular harsh elution conditions (such as pH 3.0), can be used for comparison purposes to assess the extent of elution obtained under the various elution conditions tested.
  • affinity ligands identified using the methods may be performed. For example, once affinity ligands of interest are identified using the methods described above, the nucleotide sequences encoding the affinity ligands of interest may be cloned for larger scale production (for example, in vitro eukaryotic or bacterial expression). Cloned affinity ligands of interest may then be further screened to confirm specificity, such as, for example, by assessing binding affinity for other targets.
  • affinity ligands identified that specifically bind to a target under neutral buffer conditions and have substantially reduced binding to the target under mild elution conditions are useful for affinity chromatography.
  • affinity ligands can be used for the affinity purification of targets in samples or expression cultures.
  • the target may be a protein, nucleic acid, or other biological molecule.
  • the affinity ligands are particularly useful for affinity chromatography for targets that are sensitive to harsh buffer conditions, including low pH (such as pH 3.0).
  • affinity chromatography using the affinity ligands described herein are useful for purification or assessment of targets that denature, dissociate into individual subunits, or dissociate from cofactors under harsh buffer conditions.
  • Exemplary targets for the affinity chromatography methods described herein are immunoglobulins.
  • affinity ligands that specifically bind to IgM or IgA are particularly useful for immune-chromatographic purification of IgMs and IgAs.
  • Affinity ligands for use in affinity chromatograph may be expressed and purified. These steps may be performed using conventional subcloning and expression technologies (for example, bacterial expression) using the nucleic acid sequences that encode these ligands or their binding domains.
  • the polynucleotides encoding the affinity ligands may be subcloned with an affinity tag or moiety that is useful for purification of the affinity ligands (for example, at least one of a FLAG® peptide, 6 ⁇ -Histidine peptide, etc.).
  • Affinity ligands are generally prepared in substantially purified form prior to use in generating an affinity chromatography matrix.
  • the affinity ligands may be bound to a solid support to generate an affinity chromatography matrix.
  • the affinity ligand is bound to or linked to the solid support.
  • the affinity ligand binds to the solid support through ionic interaction.
  • the affinity ligand is linked to the solid support through chemical bonds or cross-linking. Any solid support is contemplated for linkage to the affinity ligands.
  • Various commercial matrices can be used to generate the affinity matrix with the affinity ligand bound thereto. Exemplary matrices include NHS-activated Sepharose matrix or Ni-NTA Agarose.
  • the solid support may be selected based on characteristics of the purified affinity ligand, characteristics of the target, or intended uses of the target following affinity chromatography.
  • the solid support can be, for example, porous or non-porous and can be in the form, for example, of a matrix, bead, particle, chip, or other conformation, for example, a membrane or a monolith (such as a single block, pellet, or slab of material).
  • the solid support can be the wall or floor of an assay vessel, or a dipstick or other implement to be inserted into an assay vessel, or particles placed inside or suspended in an assay vessel.
  • Particles, and especially beads can be useful in many embodiments, including beads that are microscopic in size (microparticles) and formed of a polymeric material.
  • Polymers useful as microparticles are those that are chemically inert relative to the components of the biological sample and to the assay reagents other than the affinity ligands that are immobilized on the microparticle surface.
  • suitable polymers are polystyrenes, polyesters, polyethers, polyolefins, polyalkylene oxides, polyamides, polyurethanes, polysaccharides, celluloses, and polyisoprenes.
  • Crosslinking is useful in many polymers for imparting structural integrity and rigidity to the microparticle. The size range of the microparticles can vary.
  • the microparticles range in diameter from about 0.3 micrometers to about 120 micrometers, and other embodiments, from about 0.5 micrometers to about 40 micrometers, and in still other embodiments, from about 2 micrometers to about 10 micrometers.
  • Affinity chromatography may be performed on a wide range of samples that contain the target of interest.
  • the samples are obtained from subjects (including humans, primates, non-primate mammals, birds, reptiles, and amphibians).
  • the sample may be a bodily fluid.
  • the sample may also be cultured bacteria or other cells or tissues that express the target of interest, a cell culture supernatant, or a lysate from bacterial or eukaryotic cells.
  • one or more purification steps may be performed on the sample to enrich the target prior to affinity chromatography using the affinity ligands described herein.
  • the sample is applied to the solid support to allow the target to be bound by the affinity ligand.
  • the conditions for the binding may be neutral or physiological.
  • a washing step may be performed using neutral buffer conditions as described above in Section I.
  • an elution step is performed using mild elution conditions as described above in Section I.
  • mild elution conditions the binding of the affinity ligand to its target is reduced such that the affinity ligand and the target substantially dissociate from each other under these conditions.
  • the mild elution conditions may comprise a pH of about 4.0 to about 5.5.
  • the mild elution conditions may comprise about 1-2 M MgCl 2 .
  • elution may be performed as a single-step elution such that the target bound to the affinity ligand-solid support is eluted by exposing it to the mild elution conditions directly.
  • elution may be performed as a multiple-step elution such that the target bound to the affinity ligand-solid support is eluted by exposing it sequentially to multiple mild elution conditions having increasing salt concentrations or decreasing pH.
  • elution may be performed using a pH or salt gradient such that the target bound to the affinity ligand-solid support is eluted by exposing it to dynamic elution conditions as a gradient of linearly increasing salt concentration or decreasing pH.
  • the eluted target is useful for a variety of applications.
  • the target is an antibody (an immunoglobulin, such as IgM, IgA, or IgE)
  • the eluted immunoglobulin target may be used, for example, as a therapeutic molecule, or as a diagnostic or laboratory reagent.
  • additional steps may be performed on the eluted target to prepare it for subsequent uses (for example, subsequent chromatography steps, filtration like ultrafiltration or diafiltration, dialysis, labeling, etc.).
  • Kits containing affinity ligands that specifically bind to a target under neutral buffer conditions and elute the target under mild elution conditions are also contemplated.
  • Kits may include one or more types of affinity ligands.
  • the affinity ligands in the kit may be specific for the same target or for different targets. If the affinity ligands are specific for the same target, they may be specific for different epitopes of the target.
  • the affinity ligands are labeled (for example, with a peptide tag, or a chemical moiety for site-specific coupling to a matrix).
  • the kits may include affinity ligands bound to a solid support. In some examples, the affinity ligand-bound solid support is packed into a chromatography column.
  • the kit provides the affinity ligand and solid support separately along with instructions for coupling the affinity ligand to the solid support.
  • the kit may further include an equilibration buffer, a washing buffer, an elution buffer, or some combination of these buffers. In some cases, more than one washing buffer or elution buffer is provided. Multiple elution buffers may be provided, with each elution buffer having a different elution condition (such as pH, salt type, or salt concentration). For example, different elution buffers may be provided for different affinity ligands included in the kit.
  • Exemplary buffers as referenced in this disclosure may include, for example, citrate, MES, or Bis-Tris, amongst others.
  • antibodies were generated using the HuCAL PLATINUM® library that includes the CysDisplay® selection technology (Rothe, C., et al., 2008. “The human combinatorial antibody library HuCAL GOLD combines diversification of all six CDRs according to the natural immune system with a novel display method for efficient selection of high-affinity antibodies.” J Mol Biol 376(4): 1182-1200.). The aim was to generate Fab antibodies against human IgM, IgA, and IgE that bind at neutral pH but can be eluted from the antigen under mild conditions (e.g. pH 4 to pH 5). Selection on the antigens using elution under mild conditions resulted in 14 antibodies against IgM, 40 antibodies against IgA, and 17 antibodies against IgE. Assays were performed to identify the antibodies with desirable properties for use as affinity chromatography reagent.
  • the antigens and closely related antigens are listed in Table 1 below.
  • Recombinant antibodies were isolated from the HuCAL PLATINUM® library of human antibody genes by three iterative rounds of panning with the antigens as described in Table 1, using standard protocols.
  • the antigens were coupled to Dynal M-450 Epoxy beads (Invitrogen 140-11).
  • the antigen coupled beads were incubated overnight at room temperature, blocked by addition of Tris, pH7.4, and subsequently re-suspended in PBS.
  • the phage antibody library was incubated with blocking buffer containing the CRAs as set forth in Table 1 and Table 2. The blocked library was then incubated with the antigen coupled beads, and non-specific or blocked antibodies were washed off. Specific antibody phage were eluted as noted in Table 2, by incubation with pH 3 elution buffer as control selection (150 mM NaCl, 100 mM Citrate buffer pH 3), with pH 4 elution buffer (150 mM NaCl, 100 mM Citrate buffer pH 4), or pH 5 elution buffer (150 mM NaCl, 100 mM Citrate buffer or 150 mM NaCl, 100 mM Sodium Acetate buffer) for the pH elution conditions or with high salt elution buffer (2M MgCl 2 in PBS). The pH elution condition buffers were neutralized with 1M Tris before E. coli infection. Phagemid containing bacteria were grown overnight at 37° C. and new antibody displaying phage were produced for the next
  • E. coli TG1F (TG1 depleted for the F pilus; Rauchenberger et al. 2003) was then transformed with the ligated expression vectors. Following transformation, 368 individual colonies were randomly picked for each panning and grown in microtiter plates, which were then stored with 15% glycerol at ⁇ 80° C. These master plates were replicated and the resulting daughter plates were used for expression of the antibodies.
  • the ELISA screening protocol is set forth in the Table 3 below.
  • E. coli TG1F ⁇ cultures 250 mL containing the chosen antibody genes were grown at 30° C. until OD 600nm reached 0.5, and the antibody expression was induced by adding IPTG to a final concentration of 1 mM. After further incubation for at least 14 hours at 30° C., the cells were harvested, chemically lysed, and the soluble crude extract was subjected to one-step affinity chromatography (Ni-NTA Agarose, Qiagen 1018240). After elution of the purified antibodies from the column, the buffer was changed from elution buffer to 3 ⁇ PBS, pH 7.4, and the concentration was determined by UV280 nm measurement. Purity and activity was tested subsequently by Coomassie-stained SDS-PAGE and ELISA, respectively.
  • the quality control ELISA (indirect ELISA) protocol using purified HuCAL® antibodies is set forth in Table 4 below.
  • Wash 2x PBST (PBS with 0.05% Tween-20 ®) Blocking 100 ⁇ L of 5% non-fat dried milk in PBST for 1-2 hr at RT Wash 2x PBST Primary Ab 20 ⁇ L/well of antibody at 2 ⁇ g/mL in PBST for 1 hr at (HuCAL ®-Fab) RT Wash 5x PBST Secondary Ab 20 ⁇ L/well of anti-His tag, HRP conjugate (Qiagen 34460), 1:2000 dilution in HiSpec buffer (AbD Serotec BUF049), 1 hr at RT Wash 5x PBST Optional: manually wash specific wells with elution buffer, incubate for 5 min, remove buffer, repeat 2x Detection 20 ⁇ L/well QuantaBlu ® (Thermo Scientific 15169) Reader Settings Excitation at 320 ⁇ 25 nm, emission at 420 ⁇ 35 nm Background Signal values on unspecific antigens (BSA, GST) were used
  • the outcome of a panning is tested in the primary screening on several antigens and closely related antigens (CRA).
  • the antigen role defines whether positive (+) signals are expected (positive controls for the panning antigen) or whether no signal ( ⁇ ) is wanted (negative controls; CRAs).
  • a signal on a CRA (negative control) is counted, and during the analysis subtracted from the positive hits, if the signal is at least 2-fold above background.
  • a clone is considered a hit if it is positive on all antigens (positive controls) and negative on all CRAs (negative controls).
  • Table 5 indicate the numbers of hits according to the above definition.
  • the Analysis column indicates the number of hits that no longer bind to the antigen under the test elution conditions.
  • the antibodies were expressed, purified via affinity chromatography and tested using ELISA. The results are shown in FIGS. 5-9 and in Tables 6-8 and described further below.
  • FIG. 5 BSA and N1-CD33-6 ⁇ His are unrelated antigens.
  • hIgG1Kap-ctrl and hIgG1lambdaCtrl are CRAs (human IgG1 isotype with kappa and lambda light chain, respectively).
  • hIgM is human IgM from AbD Serotec (5275-5504).
  • H-IgM (Sigma) is human IgM from Sigma (18260).
  • Antigen with “pH4 wash” indicate the residual signal on that antigen after 3 consecutive 5 minute incubations of the corresponding wells with the elution buffer (pH 4).
  • FIG. 6 BSA and N1-CD33-His6 are unrelated antigens.
  • hIgG1Kap-ctrl and hIgG1lambdaCtrl are CRAs (human IgG1 isotype with kappa and lambda light chain, respectively).
  • hIgM is human IgM from AbD Serotec (5275-5504).
  • H-IgM (Sigma) is human IgM from Sigma (18260).
  • Antigen with “2M MgCl 2 wash” indicate the residual signal on that antigen after 3 consecutive 5 minute incubations of the corresponding wells with the elution buffer (2M MgCl 2 in PBS).
  • FIG. 7 BSA and GST are unrelated antigens.
  • hIgG1Kap-ctrl and hIgG1lambdaCtrl are CRAs (human IgG1 isotype with kappa and lambda light chain, respectively).
  • hIgA is human IgA from AbD Serotec (5111-5504).
  • HIgA (Sigma) is human IgA from Sigma (12636).
  • Antigen with “pH4 wash” indicate the residual signal on that antigen after 3 consecutive 5 minute incubations of the corresponding wells with the elution buffer (pH 4). A number of antibodies that were originally positive in the primary screening turned out to be negative here (AbD20801 to AbD20812).
  • FIG. 8 BSA and N1-CD33-His6 are unrelated antigens.
  • hIgG1Kap-ctrl and hIgG1lambdaCtrl are CRAs (human IgG1 isotype with kappa and lambda light chain, respectively).
  • hIgA is human IgA from AbD Serotec (5111-5504).
  • HIgA (Sigma) is human IgA from Sigma (12636).
  • Antigen with “2M MgCl 2 wash” indicate the residual signal on that antigen after 3 consecutive 5 minute incubations of the corresponding wells with the elution buffer (2M MgCl 2 in PBS). A number of antibodies that were originally positive in the primary screening turned out to be negative here (AbD20814-AbD20821).
  • FIG. 9 BSA and GST are unrelated antigens.
  • hIgG1Kap-ctrl and hIgG1lambdaCtrl are CRAs (human IgG1 isotype with kappa and lambda light chain, respectively).
  • hIgG4 kappa is human IgG4 with kappa light chain (Sigma 14639).
  • hIgM is human IgM from AbD Serotec (5275-5504).
  • AbD00264_hIgG1f is a human antibody derived from HuCAL and formatted into the IgG1 isotype.
  • AbD00264_hIgE is the same antibody formatted into the IgE isotype.
  • AbD18705_hIgE is another HuCAL-derived antibody formatted into the IgE isotype.
  • hIgE is human IgE from myeloma (AbD Serotec PHP 142).
  • Selected antibody Fab fragments isolated as described in Example 2 were used as affinity ligands to purify an immunoglobulin target.
  • Two anti-IgM antibody Fab fragments were selected as representative affinity ligands: AbD20771 and AbD20775.
  • the affinity ligands were expressed in E. coli as monomers having a FLAG-His6 tag on the heavy chain. The ligands were purified using Ni-NTA agarose.
  • hIgM molecules were selected as exemplary target molecules to assess the purification capability of the ligands. These ligands are set forth in Table 9. Supernatant containing these ligands was collected during culturing of the cell lines noted in Table 9. OBT1524 hIgM (AbD Serotec, Bio-Rad), an IgM protein purified from myeloma serum, was used as a standard control.
  • the purified affinity ligands were coupled to 1 ml “HiTrap NHS-activated HP” columns according to manufacture's protocol (GE Healthcare #17-0716-01), with 7 mg of the ligand being applied to the column.
  • the columns were run on a GE AKTAxpressTM FPLC instrument.
  • the supernatant sample (200 ml) was loaded using a flow rate of 0.4 ml/min (62 cm/h; retention time of 2.4 min) using PBS (pH about 7.0-7.2) as binding and washing buffer.
  • the elution buffer was 100 mM citrate, 150 mM NaCl, pH 4.0.
  • a neutralization buffer (1 M Tris/HCl pH 9.0) was used to neutralize the pH of the purified IgM targets.
  • Exemplary results are shown for target molecule AbD18705 IgM. Results for the other two target molecules were similar. Elution profiles (UV 280 nm) are shown in FIG. 10 (collected fractions are indicated). The AbD18705 IgM target molecule is captured from cell culture supernatant and eluted under mild conditions (pH 4.0) using both affinity ligand AbD20771 and AbD20775.
  • the purified protein fractions (A3/4/5 for the AbD20775 column and A10/11/12 for the AbD20771 column, as identified in FIG. 10 ; 1 ⁇ g protein/lane) were run on 4-20% Mini-PROTEANTM Stain-free TGX gels (Bio-Rad). Exemplary reducing and non-reducing lanes are shown for each fraction, respectively, in FIG. 11 . Comparisons were made to the purified OBT1524 IgM standard control protein ( FIG. 11 , right panel). The purity of the AbD18705 IgM target molecule in the fractions is high under reducing conditions and both heavy and light chains can be detected. Under non-reducing conditions, assembled IgM is detectable.
  • FIG. 12 shows an overlay of SEC runs (UV280 nm signal) for fractions A4/5/10/11/12 as identified in FIG. 10 .
  • the peak retention volume was 10.8 ml, corresponding to a MW of 916 kDa (calculated MW for IgM AbD18705 in pentameric form without glycosylation is 859 kDa. All fractions show an identical elution behaviour indicating that assembled, non-aggregated IgM is present in the fractions. No degradation or aggregation is visible.
  • a single component may be replaced by multiple components, and multiple components may be replaced by a single component, to provide an element or structure or to perform a given function or functions. Except where such substitution would not be operative to practice certain embodiments of the invention, such substitution is considered within the scope of the invention.

Abstract

Affinity ligands useful for mild elution affinity chromatography, including affinity ligands specific for immunoglobulins M, A, and E, are disclosed as are method of identifying and using such affinity ligands.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application is a divisional of U.S. patent application Ser. No. 15/166,761, filed May 27, 2016, which claims the benefit of priority of U.S. Provisional Application No. 62/167,387, filed May 28, 2015, the entire content of which is incorporated herein by reference.
  • FIELD OF THE INVENTION
  • The invention relates to affinity ligands, such as antibody affinity ligands, and methods for using, identifying, and making affinity ligands.
  • REFERENCE TO A SEQUENCE LISTING SUBMITTED AS A TEXT FILE VIA EFS-WEB
  • The official copy of the sequence listing is submitted electronically via EFS-Web as an ASCII formatted sequence listing with a file named 1010888-105410US_ST25.txt, created on May 26, 2016, and having a size of 103,871 bytes and is filed concurrently with the specification. The sequence listing contained in this ASCII formatted document is part of the specification and is herein incorporated by reference in its entirety.
  • BACKGROUND OF THE INVENTION
  • Affinity chromatography is a method of separating biochemical mixtures based on highly specific interactions between an affinity ligand and its target, such as that between antibody and antigen. An affinity ligand that selectively interacts with the desired target is immobilized onto a solid support in order to create an affinity matrix that can be used in a column format. Affinity chromatography can be used in a number of applications, including nucleic acid purification, protein purification from cell free extracts or cell culture supernatants, and purification from blood.
  • BRIEF SUMMARY OF THE INVENTION
  • The present disclosure provides methods of generating affinity ligands, uses of such affinity ligands, and specific affinity ligands.
  • In one aspect, provided are affinity ligands that bind specifically to a target molecule, wherein the specific binding strength of the affinity ligand to the target molecule is reduced under buffer conditions including (i) a pH of about 4.0 to about 5.5 or (ii) about 1-2 M MgCl2.
  • In some instances, the buffer condition may have a pH of about 4.0 to about 5.0. In some instances, the buffer condition may have a pH of about 4.0. In some instances, the buffer condition may have a pH of about 5.0. In some instances, the buffer condition may include 2 M MgCl2. In some instances, the buffer condition may include 2 M MgCl2 and a relatively neutral pH.
  • In some instances, the target molecule may be an immunoglobulin. In some instances, the target molecule may be an immunoglobulin selected from the group consisting of an immunoglobulin M (IgM), an immunoglobulin A (IgA), or an immunoglobulin E (IgE).
  • In some instances, the affinity ligand may be an immunoglobulin. For example, in some instances the affinity ligand may be a recombinant Fab fragment or Fab fragment derivative. In some instances, the affinity ligand may be an anti-IgE antibody comprising heavy chain complementarity determining regions CDR1, CDR2, and CDR3 sequences selected from any of the sequences set forth in FIG. 1A. In some instances, the affinity ligand may be an anti-IgE antibody having a light chain complementarity determining regions CDR1, CDR2, and CDR3 sequences selected from any of the sequences set forth in FIG. 1B. In some instances, the affinity ligand may be an anti-IgA antibody comprising heavy chain complementarity determining regions CDR1, CDR2, and CDR3 sequences selected from any of the sequences set forth in FIG. 2A. In some instances, the affinity ligand may be an anti-IgA antibody comprising light chain complementarity determining regions CDR1, CDR2, and CDR3 sequences selected from any of the sequences set forth in FIG. 2B. In some instances, the affinity ligand may be an anti-IgM antibody comprising heavy chain complementarity determining regions CDR1, CDR2, and CDR3 sequences selected from any of the sequences set forth in FIG. 3A. In some instances, the affinity ligand may be an anti-IgM antibody comprising light chain complementarity determining regions CDR1, CDR2, and CDR3 sequences selected from any of the sequences set forth in FIG. 3B.
  • In some instances, the affinity ligand may be linked to a solid support. In some instances, the solid support may be a bead or a sample plate. In some instances, the bead may be an agarose bead, a polystyrene bead, a polymethacrylate bead, a polyacrylamide bead, a magnetic bead, or a paramagnetic bead.
  • In another aspect, provided are methods of isolating a target molecule, the method comprising the steps of: providing a solid support linked to an affinity ligand; contacting the solid support with a sample containing the target molecule; washing the solid support with a wash buffer to remove unbound components of the sample; and eluting bound target molecule from the solid support with an elution buffer comprising (i) a pH of about 4.0 to about 5.5 or (ii) about 1-2 M MgCl2.
  • In some instances, the elution buffer may include a pH of about 4.0 to about 5.5 and a relatively low salt concentration. In some instances, the elution buffer may include about 1 M to 2 M MgCl2 and a relatively neutral pH. In some instances, the elution buffer may include about 1 M to 2 M MgCl2 and a pH of about 6.0 to 8.0. In some instances, the eluting may be a single-step elution with an elution buffer comprising (i) a pH of about 4.0 to about 5.5 or (ii) about 1-2 M MgCl2. In some instances, the eluting may be a multiple-step elution with a plurality of elution buffers comprising (i) a pH of about 4.0 to about 5.5 or (ii) about 1-2 M MgCl2, wherein the plurality of elution buffers are applied to the solid support sequentially, wherein elution buffers having higher salt concentrations are applied after elution buffers having lower salt concentrations and elution buffers having lower pH are applied after elution buffers having higher pH. In some instances, the eluting may be a gradient elution with an elution buffer having a gradient of linearly increasing salt concentration during the time of the eluting, wherein the maximum salt concentration is about 1-2 M MgCl2. In some instances, the eluting may be a gradient elution with an elution buffer having a gradient of linearly decreasing pH during the time of the eluting, wherein the minimum pH is about 4.0. In some instances, the wash buffer may have a pH of 6.0-8.0. In some instances, the wash buffer may have a relatively low salt concentration.
  • In some instances, the affinity ligand may be an immunoglobulin. In some instances, the target molecule may be an immunoglobulin M (IgM), an immunoglobulin A (IgA), or an immunoglobulin E (IgE). In some instances, the affinity ligand may be an anti-IgE antibody comprising heavy chain complementarity determining regions CDR1, CDR2, and CDR3 sequences selected from any of the sequences set forth in FIG. 1A. In some instances, the affinity ligand may be an anti-IgE antibody comprising light chain complementarity determining regions CDR1, CDR2, and CDR3 sequences selected from any of the sequences set forth in FIG. 1B. In some instances, the affinity ligand may be an anti-IgA antibody comprising heavy chain complementarity determining regions CDR1, CDR2, and CDR3 sequences selected from any of the sequences set forth in FIG. 2A. In some instances, the affinity ligand may be an anti-IgA antibody comprising light chain complementarity determining regions CDR1, CDR2, and CDR3 sequences selected from any of the sequences set forth in FIG. 2B. In some instances, the affinity ligand may be an anti-IgM antibody comprising heavy chain complementarity determining regions CDR1, CDR2, and CDR3 sequences selected from any of the sequences set forth in FIG. 3A. In some instances, the affinity ligand may be an anti-IgM antibody comprising light chain complementarity determining regions CDR1, CDR2, and CDR3 sequences selected from any of the sequences set forth in FIG. 3B.
  • In another aspect, provided are methods of selecting an affinity ligand that specifically binds to a target molecule under neutral buffer conditions and has reduced binding strength to the target molecular under mild elution conditions, the method including the steps of: expressing a naive affinity ligand library to produce a plurality of affinity ligands; providing a solid support linked to a target; contacting the solid support with the plurality of affinity ligands; washing the solid support with a wash buffer to remove unbound affinity ligands, wherein the wash buffer comprises neutral buffer conditions; contacting the solid support with an elution buffer comprising (i) a pH of about 4.0 to about 5.5 or (ii) about 1-2 M MgCl2; and identifying affinity ligands that substantially dissociate from the solid support in the elution buffer.
  • In some instances, the affinity ligand library may not be preselected for characteristics favoring reduced binding strength to the target molecule under mild elution conditions. In some instances, the plurality of affinity ligands may be encoded by a plurality of nucleic acid sequences. In some instances, the plurality of nucleic acid sequences includes a heterologous promoter operably linked thereto. In some instances, the plurality of affinity ligands may be expressed on a plurality of phage.
  • In some instances, the elution buffer may have a pH of about 4.0 to about 5.5 and a relatively low salt concentration. In some instances, the elution buffer may include about 1 M to 2 M MgCl2 and may have a relatively neutral pH. In some instances, the elution buffer may include about 1 M to 2 M MgCl2 and may have a pH of about 6.0 to 8.0. In some instances, the wash buffer may have a pH of 6.0-8.0. In some instances, the wash buffer may have a relatively low salt concentration.
  • In some instances, the target may be an immunoglobulin. In some instances, the target may be an immunoglobulin M (IgM), an immunoglobulin A (IgA), or an immunoglobulin E (IgE).
  • In some instances, the plurality of affinity ligands may be a plurality of antibodies or derivatives thereof. In some instances, the plurality of affinity ligands is a plurality of Fab fragments or derivatives thereof. In some instances, the affinity ligand identified may be encoded by a polynucleotide comprising a nucleic acid sequence encoding heavy chain complementarity determining regions CDR1, CDR2, and CDR3 sequences selected from any of the sequences set forth in FIG. 1A. In some instances, the affinity ligand identified may be encoded by a polynucleotide comprising a nucleic acid sequence encoding light chain complementarity determining regions CDR1, CDR2, and CDR3 sequences selected from any of the sequences set forth in FIG. 1B. In some instances, the affinity ligand identified may be encoded by a polynucleotide comprising a nucleic acid sequence encoding heavy chain complementarity determining regions CDR1, CDR2, and CDR3 sequences selected from any of the sequences set forth in FIG. 2A. In some instances, the affinity ligand identified may be encoded by a polynucleotide comprising a nucleic acid sequence encoding light chain complementarity determining regions CDR1, CDR2, and CDR3 sequences selected from any of the sequences set forth in FIG. 2B. In some instances, the affinity ligand identified may be encoded by a polynucleotide comprising a nucleic acid sequence encoding heavy chain complementarity determining regions CDR1, CDR2, and CDR3 sequences selected from any of the sequences set forth in FIG. 3A. In some instances, the affinity ligand identified may be encoded by a polynucleotide comprising a nucleic acid sequence encoding light chain complementarity determining regions CDR1, CDR2, and CDR3 sequences selected from any of the sequences set forth in FIG. 3B.
  • In another aspect, provided are kits including the affinity ligand described above.
  • It will be appreciated from a review of the remainder of this application that further methods and compositions are also part of the invention.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1A and FIG. 1B show heavy and light chain CDR sequences, respectively, for anti-IgE antibodies according to some examples.
  • FIG. 2A and FIG. 2B show heavy and light chain CDR sequences, respectively, for anti-IgA antibodies according to some examples.
  • FIG. 3A and FIG. 3B show heavy and light chain CDR sequences, respectively, for anti-IgM antibodies according to some examples.
  • FIGS. 4A-4I show heavy and light chain sequences for anti-IgM antibodies according to some examples.
  • FIG. 5 shows ELISA results for a first set of anti-IgM antibody affinity ligands assessing binding to and elution of target molecules according to one example.
  • FIG. 6 shows ELISA results for a second set of anti-IgM antibody affinity ligands assessing binding to and elution of target molecules according to one example.
  • FIG. 7 shows ELISA results for a first set of anti-IgA antibody affinity ligands assessing binding to and elution of target molecules according to one example.
  • FIG. 8 shows ELISA results for a second set of anti-IgA antibody affinity ligands assessing binding to and elution of target molecules according to one example. Controls N1-CD33-6×His and BSA are also shown.
  • FIG. 9 shows ELISA results for a set of anti-IgE antibody affinity ligands assessing binding specificity for target molecules according to one example.
  • FIG. 10 shows elution profiles of purified AbD18705 hIgM target molecule from affinity ligand columns according to one example. The affinity column on the left uses anti-IgM antibody ligand AbD20775.2, and the affinity column on the right uses anti-IgM antibody ligand AbD20771.2. Collected fractions (A#) are shown across the bottom of each graph.
  • FIG. 11 shows SDS-PAGE analysis of purified AbD18705 hIgM target molecule elution fractions according to one example. The left gel shows the purified AbD18705 hIgM from the indicated fractions (each fraction shown under reducing and under oxidizing conditions). Purified human IgM (product number OBT1524) is shown as a control in the gel on the right, again under reducing and oxidizing conditions.
  • FIG. 12 shows an overlay of size exclusion chromatography runs for purified AbD18705 hIgM target molecule elution fractions identified in FIG. 10 according to one example.
  • FIG. 13 shows a graph illustrating the results of an ELISA assay assessing the activity and specificity of the purified AbD18705 hIgM target molecule (“His-GFP) for its antigen GFP according to one example. Controls GST, N1-CD33-6×His, and BSA are also shown.
  • DEFINITIONS
  • “Affinity ligand” or “ligand” refers to a composition (such as, for example, an antibody or non-antibody protein), that binds specifically to a specific substance, such as a protein, protein complex, or organic compound having a defined structure.
  • The term “solid support” is used herein to denote a solid inert surface or body to which an agent, such as an antibody or an antigen, that is reactive in any of the binding reactions described herein can be immobilized. The term “immobilized” as used herein denotes a molecularly-based coupling that is not dislodged or de-coupled under any of the conditions imposed during any of the steps of the assays described herein. Such immobilization can be achieved through a covalent bond, an ionic bond, an affinity-type bond, or any other covalent or non-covalent bond.
  • The term “antibody” or “immunoglobulin” refers to an immunoglobulin, composite, or fragmentary form thereof. The term may include but is not limited to polyclonal or monoclonal antibodies of the classes IgA, IgD, IgE, IgG, and IgM, derived from human or other cell lines, including natural or genetically modified forms such as humanized, human, single-chain, chimeric, synthetic, recombinant, hybrid, mutated, grafted, and in vitro generated antibodies and fragments thereof. “Antibody” may also include composite forms including but not limited to fusion proteins containing an immunoglobulin moiety. “Antibody” may also include non-quaternary antibody structures (such as camelids and camelid derivatives). “Antibody” may also include antibody fragments such as Fab (fragment-antigen binding), F(ab′)2, Fv, scFv, Fd, dAb, Fc and other compositions that retain antigen-binding function. In addition, the term “antibody” includes aggregates, polymers, and conjugates of immunoglobulins or their fragments, where the molecules largely retain binding affinity for their epitope(s). Further, an “antibody” may be modified, such as, for example, by linking to a chemical or peptide moiety or detectable tag moiety.
  • The term “complementarity determining region” or “CDR” (also known as “hypervariable region” or “HVR”) refers to an immunoglobulin hypervariable domain that determines specific binding of an immunoglobulin to an epitope. The variable regions of both the heavy and light chains of an antibody each generally contain three CDRs. Antibodies with different specificities have different CDRs, while antibodies of the exact same specificity may have identical CDRs.
  • A “constant region” refers to a region in the heavy and light chains of an antibody having relatively less variability compared to the N′ terminal variable region of the heavy and light chains of an antibody. On the heavy chains, the constant region is generally the same in all antibodies of the same isotype and differs in antibodies of different isotypes. There are two primary types of light chains (kappa and lambda), each with a distinct constant region.
  • “Neutral buffer” or “neutral buffer condition” refers to a buffer having approximately physiological pH. Such buffers/conditions allow binding of proteins to an affinity column without resulting in substantial protein denaturation or aggregation. A neutral buffer or neutral buffer condition may permit near optimal interaction between an affinity ligand and a target molecule. A neutral buffer condition or neutral buffer generally has a pH in the range of about 6.0 to 8.0.
  • “Mild elution condition” or “mild elution buffer” refers to a buffer in which an affinity ligand that specifically binds to a target molecule dissociates from the target molecule (such as on an affinity matrix) without resulting in substantial protein denaturation or aggregation of the target molecule. This is in contrast to the harsh conditions that are typically applied in elution of a target molecule, such as low pH (about pH 2 to 3.5, for example). Such mild elution buffers may include relatively high pH (pH≥4.0) or relatively high salt or ionic strength.
  • “Physiological pH” refers to the pH of human blood, which is about 7.4. A pH in the range of pH 7.0 to pH 7.8 may be considered approximately physiological pH.
  • “Naive expression library,” for the purposes of expressing affinity ligands, refers to an expression library that expresses a large number of recombinant proteins, polypeptides, or peptides that have a large diversity of structure or specificity in their binding domains. A naive library is one generated from synthetic or natural ligand repertoires that have not previously subjected to selections for increased affinity for a target. A naive expression library is generated from a plurality of nucleic acid sequences that encode a plurality of affinity ligands or at least a plurality of target binding regions.
  • “Relatively neutral pH” refers to a pH of about pH 6.0 to 8.0.
  • A “solid support” refers to a material or group of materials having a rigid or semi-rigid surface or surfaces. In some embodiments, the solid support takes the form of thin films or membranes, beads, bottles, dishes, fibers, woven fibers, shaped polymers, particles, and microparticles, including but not limited to, microspheres. A solid support can be formed, for example, from an inert solid support of natural material, such as glass and collagen, or synthetic material, such as acrylamide, cellulose, nitrocellulose, silicone rubber, polystyrene, polyethylene vinyl acetate, polypropylene, polymethacrylate, polyethylene, polysilicates, polyethylene oxide, polycarbonates, teflon, fluorocarbons, nylon, polyanhydrides, polyglycolic acid, polylactic acid, polyorthoesters, polypropylfumarate, glycosaminoglycans, and polyamino acids. In some cases, some functional groups naturally present on the surface of a carrier (for example, carboxylic acid (—COOH), free amine (—NH2), and sulfhydryl (—SH) groups) can be used for peptide linkage. In case no such functional group is naturally available, a desired functional group, such as a carboxylic acid group, or a moiety known to be a partner of a binding interaction (such as avidin that is capable of binding biotin) may be attached to such solid support. In some embodiments, the solid support is a carboxylated latex or magnetic microsphere.
  • The phrase “specific binding” or “binds specifically” refers to a binding reaction where two members of a binding pair (for example, an antibody and a molecule comprising the antibody's target epitope) bind to each other with an affinity that is at least 10-fold better than the members' affinity for other components of a heterogeneous mixture (for example, a hybridoma culture supernatant or other mixture of proteins).
  • The term “variable region” refers to an N′ terminal region of each of the heavy and light chains of an antibody that has relatively more variability compared to the constant region(s) of the heavy and light chains of an antibody. The variable region contains the CDRs.
  • DETAILED DESCRIPTION OF THE INVENTION
  • It has been discovered that affinity ligands (such as antibodies) can be identified for use in immunoaffinity purification of target molecules (such as immunoglobulins M, A, or E) such that mild conditions can be used to elute the target molecule to which the affinity ligand specifically binds, thereby avoiding harsh elution conditions used previously. Certain examples and features of the present disclosure relate to methods to identify affinity ligands, such as antibodies, that are especially useful for mild elution immunoaffinity chromatography. Such ligands, when attached to a solid support, allow one-step purification of target molecules using mild elution conditions. The use of mild elution conditions circumvents the typically harsh elution conditions used in this type of chromatography that may lead to denaturing or aggregation of target molecules (such as low pH, for example, pH 3.0). The described methods utilize in vitro enrichment methods to permit selection of affinity ligands for which mild elution conditions can be used. The selection of such ligands is performed by a first binding step under conditions that allow binding in standard/neutral buffer conditions (such as neutral pH and/or low salt), followed by a subsequent elution step that is performed using mild elution conditions. The described methods permit specific selection and enrichment and finally isolation of affinity ligands having the desired characteristics. As such, affinity ligands of interest can be generated more rapidly and efficiently using the described methods.
  • Certain examples and features of the present disclosure relate to affinity ligands, such as antibodies, that exhibit the property of specifically binding to a target under neutral conditions and releasing the target under mild elution conditions. These affinity ligands can be selected for using the method described above. Examples of affinity ligands that are specific for human immunoglobulin M (IgM), human immunoglobulin A (IgA), and human immunoglobulin E (IgE) are described.
  • Certain examples and features of the present disclosure relate to methods of using such affinity ligands to purify targets using mild elution conditions. Affinity ligands may be used linked to a solid support as an affinity purification column. For example, some targets that are sensitive to harsh elution conditions (for example, pH 3.0) may be successfully purified using an affinity column generated using the affinity ligands that allow elution of targets under mild elution conditions. For example, such affinity ligands are useful for isolating IgM, IgA, or IgE molecules as these molecules may be sensitive to denaturation when affinity purified using harsh elution conditions. Other molecules that are sensitive to denaturation can be purified accordingly, once an affinity ligand has been isolated using the methods described in this disclosure.
  • I. Affinity Ligands
  • Affinity ligand compositions are described herein that bind specifically to a target and from which target can be eluted using mild elution conditions. As described further below, the affinity ligand may be an antibody, antibody-like molecule, or other affinity-binding molecule. For ease of description, this disclosure will sometimes describe examples and features of the affinity ligands in the context of antibody affinity ligands. However, this disclosure is not limited to affinity ligands that are antibodies.
  • The affinity ligand may be selected from a variety of different types of protein or non-protein compositions. In certain cases, an affinity ligand is an antibody, antibody-like molecule, or any other affinity-binding molecule derived from a naive expression library as described below in Section II. For example, the antibody may be a monoclonal antibody, a Fab fragment, a F(ab′)2, an Fv, a scFv, an Fd, a dAb, an Fc fragment, a VHH, or other fragments thereof that retain antigen-binding function. In some examples, affinity ligand may be recombinant antibodies having heterologous constant and variable domains; for example, generated by a recombinant protein expression library. In some instances, the affinity ligand may be a recombinant Fab fragment. For example, the Fab fragment may have the variable domain and the first constant domain (Fd chain) of one heavy chain plus one complete light chain (L chain). The Fd and L chain are linked by strong non-covalent interactions and can be covalently linked by a disulfide bond. The polynucleotide sequence encoding the recombinant Fab fragment may be subcloned into an expression vector containing a heterologous promoter to drive expression of the recombinant Fab fragment. The Fab fragment may monovalent, bivalent, or multivalent. In some instances, the Fab fragment is monovalent. In some examples, the affinity ligand is a single-chain variable (scFv) fragment or a bivalent scFv fragment (diabody). ScFv fragments typically have one VH and one VL chain expressed as a single polypeptide joined by a peptide linker. The polypeptide linker stabilizes the interaction between the VH and VL chains. The polynucleotide sequence encoding the recombinant scFv fragment may be subcloned into an expression vector containing a heterologous promoter to drive expression of the recombinant scFv fragment. In some instances, the affinity ligand may be a recombinant antibody or protein that specifically binds to targets in a manner similar to antibodies, or fragment thereof that retains antigen-binding function. The affinity ligand may be a recombinant protein that contains at least three complementarity determining regions (CDRs) that cause the specific binding of the affinity ligand to the target; for example, three heavy chain CDRs and, in some examples, contains six CDRs (three heavy chain and three light chain). In some instances, the antibody may be a variable domain of heavy chain (VHH) antibody or a nanobody (a monomeric variable domain antibody). The VHH or nanobody may be encoded by a singly polypeptide.
  • In some examples, the affinity ligand is a camelid antibody or camelid nanobody. Camelid antibodies are certain IgG antibodies from the mammalian family of camel and dromedary (Camelus bactrianus and Camelus dromaderius) family, including new world members such as llama species (such as Lama paccos, Lama glama and Lama vicugna), that lack light chains. See, for example, International Appl. WO 94/04678. The small single variable domain (VHH) of the camelid antibody can be used to as the basis of a low molecular weight antibody-derived protein known as a “camelid nanobody” having high affinity for a target. See U.S. Pat. No. 5,759,808; see also Stijlemans, B. et al., 2004 J Biol Chem 279: 1256-1261; Dumoulin, M. et al., 2003 Nature 424: 783-788; Pleschberger, M. et al. 2003 Bioconjugate Chem 14: 440-448; Cortez-Retamozo, V. et al. 2002 Int J Cancer 89: 456-62; and Lauwereys, M. et al. 1998 EMBO J 17: 3512-3520. Engineered libraries of camelid antibodies and antibody fragments are commercially available.
  • In some cases, the affinity ligand can be a compound or non-antibody protein that specifically binds to targets in a manner similar to antibodies. Certain of these “antibody mimics” use non-immunoglobulin protein scaffolds as alternative protein frameworks for the variable regions of antibodies. For example, the affinity ligand can be a monobody, which are small antibody mimics using the scaffold of a fibronectin type III domain (FN3). FN3 scaffold functions as an effective framework onto which loops for specific building functions can be grafted. For example, the affinity ligand may utilize the tenth FN3 unit of human fibronectin as scaffold. It is small, monomeric, and does not have disulfide bonds. FN3-based antigen-binding molecules can be prepared using methods described in the art. For example, see Koide et al., J. Mol. Biol. 284: 1141-1151, 1998; Koide et al., Proc. Natl Acad. Sci. USA 99:1253-1258, 2002; and Batori et al., Protein Eng. 15:1015-20, 2002, and U.S. Pat. Nos. 6,818,418 and 7,115,396. In another example, the affinity ligand may be a single polypeptide chain binding molecule that contains the antigen binding sites of both the heavy and light variable regions of an antibody connected by a peptide linker and will fold into a structure similar to that of the two peptide antibody. See, for example, U.S. Pat. No. 5,260,203. Also, the affinity ligand may be a recombinant protein containing derivative sequences of one or more loops of cytochrome b562 that are selected for binding specificity to the target. See, for example, Ku et al., Proc. Natl. Acad. Sci. U.S.A. 92(14):6552-6556 (1995). In another example, the affinity ligand may be an antibody mimic based on a lipocalin scaffold, in which one or more of the hypervariable loops of the lipocalin protein are randomized and selected for specific binding to the target. See, for example, Beste et al., Proc. Natl. Acad. Sci. U.S.A. 96(5):1898-1903 (1999). An example of such antibody mimetics are Anticalins®, which are small, single chain peptides, typically between 160 and 180 residues. In addition, the affinity ligand may be a synthetic antibody mimic using the rigid, non-peptide organic scaffold of calixarene to which are attached multiple variable peptide loops used as binding sites. See, for example, U.S. Pat. No. 5,770,380. In some examples, the affinity ligand may be an antibody-like binding peptidomimetic. See, for example, Murali et al., Cell. Mol. Biol. 49(2):209-216 (2003). Also, in some examples, the affinity ligand may include a scaffold derived from one or more A-domains. For example, the affinity ligand may include multiple A-domains, each of which binding independently to a distinct epitope of the target. Such affinity ligands can be generated using methods described in, for example, Gliemann et al., Biol. Chem. 379:951-964, 1998; Koduri et al., Biochemistry 40:12801-12807, 2001 and Silverman et al., Nat Biotechnol. 23:1556-61, 2005. Other exemplary non-antibody scaffolds for use as ligands include darpins, affimers, cystine-knot mini-proteins, affilins, and peptides or non-protein-based scaffolds like aptamers.
  • In some instances, the affinity ligand may comprise an affinity tag or moiety. The affinity tag or moiety may be useful for purification of the affinity ligand or attachment to a solid support. For example, the affinity ligand may include at least one of a FLAG® peptide, 6×-Histidine (6×His) peptide, etc. In some cases, the affinity ligand is chemically modified to facilitate attachment of the ligand to a solid support.
  • In one aspect, the affinity ligand binds specifically to its target under neutral buffer conditions. Neutral buffer conditions may include a pH of approximately physiological pH, or relatively neutral pH, such as, for example a pH of about 6.0, 6.2, 6.5, 6.8, 7.1, 7.3, 7.5, 7.8, 7.9, 8.1, or a pH of about 6.0 to about 8.0. In some instances, the neutral buffer condition also comprises a relatively low salt concentration and/or ionic strength. For example, the salt concentration may be approximately physiological salt concentration. In some cases, the neutral buffer conditions include approximately physiological ionic strength. In some instances, neutral buffer conditions may be used for a wash buffer. In such instances, the wash buffer may have somewhat higher salt concentration or ionic strength to improve stringency and reduce non-specific binding of non-targets to the affinity ligand. In some instances, the neutral buffer condition does not include a salt. In certain instances, the neutral buffer condition may include detergent to reduce non-specific binding of non-targets to the affinity ligand. In some instances, the neutral buffer condition comprises a buffering agent. In one example, a neutral wash buffer may be phosphate buffered saline (PBS). In another example, the neutral wash buffer may be PBS containing about 0.05-1.0% Tween-20® (for example, 0.1%). Other solutions and detergents are contemplated.
  • In another aspect, under mild elution conditions, the specific binding of the affinity ligand to its target is reduced such that the affinity ligand and the target substantially dissociate from each other. In one aspect, the mild elution conditions may comprise a pH of about 4.0 to about 5.5. In another aspect, the mild elution conditions may comprise about 1-2 M MgCl2.
  • In one aspect, mild elution conditions may comprise a pH in the range of about 4.0 to about 5.5. For example, mild elution conditions may include a pH of at least about 4.0 but less than or equal to about 5.5. The mild elution condition may include a pH greater than or equal to about 4.0, a pH of about 4.5, a pH of about 5.0, or a pH of about 5.5, or any pH within the range of about 4.0 to about 5.5. Where the pH is in the range of about 4.0 to 5.5, the elution buffer may further comprise a relatively low salt concentration. In some instances, the elution buffer may contain salt conditions relatively similar to physiological salt conditions. In some instances, the salt concentration may be 25 mM, 50 mM, 100 mM, 125 mM, 150 mM, 175 mM, 200 mM, 225 mM, or 250 mM. In one example, the elution buffer may include about 150 mM NaCl. In some cases, the mild elution conditions include approximately physiological ionic strength. In some instances the elution buffer may further include a buffering agent. Exemplary buffering agents include citrate, sodium acetate, and sodium phosphate buffered saline (PBS). In one example, the elution buffer may contain 100 mM citrate buffer. In another example, the elution buffer may contain 100 mM sodium acetate. In another example, the elution buffer may contain 1×PBS. In one example, the elution buffer may contain 150 mM NaCl and 100 mM citrate buffer.
  • In another aspect, mild elution conditions may comprise a salt concentration of about 1 M to 2 M MgCl2. For example, the salt concentration may be about 0.8 M, about 1 M, about 1.2 M, about 1.4 M, about 1.6 M, about 1.8 M, about 2 M, or about 2.2 M. In one example, the mild elution condition comprises 2 M MgCl2. In some instances, the mild elution conditions may further comprise a relatively neutral pH, or an approximately physiological pH, or a pH of about 6.8 to about 7.9, or a pH of about 6.0 to about 8.0, or any pH with these ranges. For example, the pH may be about 6.0, about 6.2, about 6.4, about 6.6, about 6.8, about 7.0, about 7.2, about 7.4, about 7.6, about 7.8, or about 8.0, when the mild elution condition includes a salt concentration of about 1 M to 2 M MgCl2. In some instances, the elution buffer may further include a buffering agent. Exemplary buffering agents include citrate, sodium acetate, and phosphate buffered saline (PBS). The buffering agent may be of sufficient concentration to provide pH buffering. In one example, the elution buffer may contain 100 mM citrate. In another example, the elution buffer may contain 100 mM sodium acetate. In another example, the elution buffer may contain 1×PBS.
  • It is understood that, where the mild elution condition comprises a pH in the range of about 4.0 to 5.5, the neutral buffer conditions generally have a higher pH of approximately neutral pH or approximately physiological pH. It is also understood that, where the mild elution condition comprises about 1M to 2M MgCl2, the neutral buffer conditions generally have a lower salt concentration or ionic strength.
  • In one aspect, the affinity ligand binds specifically to a target epitope of a target molecule. The target epitope can be a portion of a target molecule, such as a protein, nucleic acid, or other biological molecule. For example, the target may be a protein or other molecule that is sensitive to harsh buffer conditions, including low pH (such as pH 3.0). In some instances, a target may denature, dissociate into individual subunits, or dissociate from cofactors under harsh buffer conditions. In some examples, the target molecule may be an immunoglobulin (antibody), such as an immunoglobulin G (IgG), an immunoglobulin M (IgM), an immunoglobulin A (IgA), or an immunoglobulin E (IgE). In some instances, the affinity ligand may be a Fab fragment that binds specifically to an IgG, an IgM, an IgA, or an IgE.
  • For example, the affinity ligand may be a Fab fragment that binds specifically to an IgE having any of the heavy and light chain CDR sequences as set forth in FIG. 1A and FIG. 1B, respectively. In some instances, the antibodies specific for IgE may contain a combination of CDR1, CDR2, and CDR3 sequences as set forth in FIG. 1A or FIG. 1B. In some examples, such affinity ligand antibodies will have reduced binding to IgE under mild elution conditions, such as at a pH equal to or greater than about 4.0 and less than or equal to about 5.5, such that the IgE elutes from the affinity agent. For example, as described in Example 2, certain anti-IgE antibodies substantially dissociate from their target under elution conditions comprising pH 4.0 and pH 5.0. In some instances, the elution buffer conditions also comprise relatively low salt, such as 150 mM NaCl, and a buffering agent, such as citrate. Other suitable salts and buffering agents are also contemplated.
  • In another example, the affinity ligand may be a Fab fragment that binds specifically to an IgA having any of the heavy and light chain CDR sequences as set forth in FIG. 2A and FIG. 2B, respectively. In some instances, the antibodies specific for IgA may contain a combination of CDR1, CDR2, and CDR3 sequences as set forth in FIG. 2A or FIG. 2B. In some examples, such affinity ligand antibodies will have reduced binding to IgA under mild elution conditions, such as at a pH equal to or greater than about 4.0 and less than or equal to about 5.5, such that the IgE elutes from the affinity agent. For example, as described in Example 2, certain anti-IgA antibodies substantially dissociate from their target under elution conditions comprising pH 4.0 and pH 5.0. In some instances, the elution buffer conditions also comprise relatively low salt, such as 150 mM NaCl, and a buffering agent, such as citrate. Other suitable salts and buffering agents are also contemplated.
  • In another example, the affinity ligand may be a Fab fragment that binds specifically to an IgM having any of the heavy and light chain CDR sequences as set forth in FIG. 3A and FIG. 3B, respectively. In some examples, the affinity ligand may have at least one of the heavy chain sequences and at least one of the light chain sequences shown in FIGS. 4A-4I. In some instances, the affinity ligand may have at least one heavy and light chain sequences pair as shown in FIGS. 4A-4I. In some instances, the antibodies specific for IgM may contain a combination of CDR1, CDR2, and CDR3 sequences as set forth in FIG. 3A or FIG. 3B. In some examples, such affinity ligand antibodies will have reduced binding to IgM under mild elution conditions, such as at a pH equal to or greater than about 4.0 and less than or equal to about 5.5, such that the IgM elutes from the affinity agent. For example, as described in Example 2, certain anti-IgM antibodies substantially dissociate from their target under elution conditions comprising pH 4.0 and pH 5.0. In some instances, the elution buffer conditions also comprise relatively low salt, such as 150 mM NaCl, and a buffering agent, such as citrate. Other suitable salts and buffering agents are also contemplated.
  • In certain examples, the affinity ligand may have CDR sequences similar to those identified in this disclosure except varying in amino acid sequence at one or two amino acid positions. In some instances, the variance in sequence is a conservative amino acid change.
  • While this disclosure describes specific examples and features of the affinity ligands that are specific for immunoglobulin targets, affinity ligands specific for other types of ligands are also contemplated.
  • II. Selection Methods
  • The described selection methods allow for fast and efficient identification of affinity ligands, in particular antibodies, for use in affinity chromatography methods using mild elution conditions. In some embodiments, a plurality of potential affinity ligands are provided and processed such that ligands are selected that specifically bind to a target under neutral buffer conditions and display weak specific binding strength to the target under mild elution conditions.
  • A variety of methods are known and can be used for expressing a plurality of affinity ligands. In particular, naive expression libraries are useful for expressing a large number of recombinant proteins, polypeptides, and peptides, including, for example, antibodies, that have a large diversity of structure or specificity in their binding domains. A naive library is one generated from synthetic or natural ligand repertoires that have not previously subjected to selections for increased affinity for a target. In some instances, a naive library is generated from a plurality of nucleic acid sequences that encode a plurality of affinity ligands or at least a plurality of target binding regions. In some instances, the nucleic acid sequences encoding the target binding regions of the plurality of affinity ligands may have randomized sequences to generate binding regions in the plurality of affinity ligands that have randomized amino acid sequences. In some instances, the expression library expresses proteins or polypeptides having a structural domain with a common amino sequence and a binding domain having variable sequences such that the proteins or polypeptides expressed by the library may display a large number of distinct binding domains based on sequence variability. As such, different proteins or polypeptides expressed by the library may have different binding specificities for different targets under different conditions. In some examples, the expression libraries express hundreds of thousands, millions, or billions of different proteins or polypeptides, each of which may have a distinct binding affinity and, thus, may bind to different targets with different degrees of specificity under different conditions. Expression libraries useful for expressing a plurality of affinity ligands for use in this method include phage display libraries, yeast display libraries, ribosome display, mRNA display or other selection system, or any other recombinant expression library capable of expressing a plurality of affinity ligands having a range of binding specificities. An example of a useful affinity ligand expression library is the HuCAL Platinum® Platform (AbD Serotec, Bio-Rad), which provides phage display libraries of antibodies in Fab format representing an extensive array (>1010 members) of CDR sequence variability (Prassler, J., et al. (2011). “HuCAL PLATINUM, a synthetic Fab library optimized for sequence diversity and superior performance in mammalian expression systems.” J. Mol. Biol. 413(1): 261-278). In some instances, the variability of the plurality of affinity ligands expressed by the library facilitate the identification of ligands that bind to a target with specific binding affinity under neutral buffer conditions and have substantially reduced specific binding under mild elution conditions as described in Section I.
  • To perform the method, a target of interest is linked or adsorbed to a solid support. The target may be such as a protein, nucleic acid, or other biomolecule as described above. The solid support may be the wall or floor of an assay vessel, or a dipstick or other implement to be inserted into an assay vessel, or particles (such as magnetic beads) placed inside or suspended in an assay vessel. Particles, and especially beads, can be useful in many embodiments, including beads that are microscopic in size (microparticles) and formed of a polymeric material.
  • To prevent non-specific binding of affinity ligands, the target-linked solid support may be blocked with a blocking buffer. For example, the blocking buffer may contain an animal protein blocker (such a milk or bovine serum albumin), a non-animal protein blocker (such as ChemiBLOCKER™), or a detergent (such a Tween-20®). In some instances, the blocking buffer may contain a closely related antigen. For example, where the target is an IgM antibody with kappa light chain, the blocking buffer may contain an antibody of different isotype (e.g., IgG) with kappa light chain as a blocker. Inclusion of this blocker may help to avoid enrichment of affinity ligands that bind specifically to the kappa light chain or to heavy chain epitopes that are similar in IgM and IgG. In particular, where the target protein is an immunoglobulin and the plurality of affinity ligands are antibodies, the target-linked solid support may be blocked with one or more types of antibody light chains as a blocker. For example, where the ligand is human IgM or human IgA, the target-linked solid support may be blocked with human IgG1 lambda or IgG1 kappa as described in Examples 1 and 2. In some instances, the blocking buffer may contain culture media (for example, media that sustain growth of a eukaryotic cell culture producing immunoglobulin like IgE, IgA or IgM). Blocking with culture medium may be useful to avoid enrichment of affinity ligands with cross-reactivity to components in the culture medium. In some instances, this can be useful because the target proteins are expressed in cells that are grown in culture medium. For example, as described in Examples 1 and 2, where the ligand is human IgE, the target-linked solid support may be blocked with culture medium.
  • The expressed affinity ligands can be brought into contact with the target-linked solid support under neutral buffer conditions to achieve binding of the affinity ligands, if any ligands in the library have affinity for the target. To remove non-specifically or weakly bound affinity ligands, a washing step may be performed using neutral buffer conditions as described above in Section I.
  • An elution step may then be performed using the mild elution conditions as described above in Section I. Under such mild elution conditions, the binding of some of the affinity ligands to its target may be sufficiently reduced such that the affinity ligand and the target substantially dissociate from each other under these conditions. In one aspect, the mild elution conditions may comprise a pH of about 4.0 to about 5.5. In another aspect, the mild elution conditions may comprise about 1-2 M MgCl2.
  • The described methods allow for specific selection and enrichment of affinity ligands that specifically bind to a target under neutral buffer conditions and have substantially reduced specific binding to the target under mild elution conditions. Various elution conditions may be assessed to determine the optimal elution condition for a particular affinity ligand-target combination. Control elution conditions, in particular harsh elution conditions (such as pH 3.0), can be used for comparison purposes to assess the extent of elution obtained under the various elution conditions tested.
  • Additional assessment of affinity ligands identified using the methods may be performed. For example, once affinity ligands of interest are identified using the methods described above, the nucleotide sequences encoding the affinity ligands of interest may be cloned for larger scale production (for example, in vitro eukaryotic or bacterial expression). Cloned affinity ligands of interest may then be further screened to confirm specificity, such as, for example, by assessing binding affinity for other targets.
  • III. Affinity Chromatography Methods
  • Affinity ligands identified that specifically bind to a target under neutral buffer conditions and have substantially reduced binding to the target under mild elution conditions are useful for affinity chromatography. For example, such affinity ligands can be used for the affinity purification of targets in samples or expression cultures. The target may be a protein, nucleic acid, or other biological molecule. The affinity ligands are particularly useful for affinity chromatography for targets that are sensitive to harsh buffer conditions, including low pH (such as pH 3.0). For example, affinity chromatography using the affinity ligands described herein are useful for purification or assessment of targets that denature, dissociate into individual subunits, or dissociate from cofactors under harsh buffer conditions. Exemplary targets for the affinity chromatography methods described herein are immunoglobulins. As IgMs and IgAs are relatively sensitive to low pH conditions, affinity ligands that specifically bind to IgM or IgA are particularly useful for immune-chromatographic purification of IgMs and IgAs.
  • Affinity ligands for use in affinity chromatograph may be expressed and purified. These steps may be performed using conventional subcloning and expression technologies (for example, bacterial expression) using the nucleic acid sequences that encode these ligands or their binding domains. In some instances, the polynucleotides encoding the affinity ligands may be subcloned with an affinity tag or moiety that is useful for purification of the affinity ligands (for example, at least one of a FLAG® peptide, 6×-Histidine peptide, etc.). Affinity ligands are generally prepared in substantially purified form prior to use in generating an affinity chromatography matrix.
  • To perform affinity chromatography using the affinity ligands, the affinity ligands may be bound to a solid support to generate an affinity chromatography matrix. The affinity ligand is bound to or linked to the solid support. In some examples, the affinity ligand binds to the solid support through ionic interaction. In some instances, the affinity ligand is linked to the solid support through chemical bonds or cross-linking. Any solid support is contemplated for linkage to the affinity ligands. Various commercial matrices can be used to generate the affinity matrix with the affinity ligand bound thereto. Exemplary matrices include NHS-activated Sepharose matrix or Ni-NTA Agarose. The solid support may be selected based on characteristics of the purified affinity ligand, characteristics of the target, or intended uses of the target following affinity chromatography. The solid support can be, for example, porous or non-porous and can be in the form, for example, of a matrix, bead, particle, chip, or other conformation, for example, a membrane or a monolith (such as a single block, pellet, or slab of material). The solid support can be the wall or floor of an assay vessel, or a dipstick or other implement to be inserted into an assay vessel, or particles placed inside or suspended in an assay vessel. Particles, and especially beads, can be useful in many embodiments, including beads that are microscopic in size (microparticles) and formed of a polymeric material. Polymers useful as microparticles are those that are chemically inert relative to the components of the biological sample and to the assay reagents other than the affinity ligands that are immobilized on the microparticle surface. Examples of suitable polymers are polystyrenes, polyesters, polyethers, polyolefins, polyalkylene oxides, polyamides, polyurethanes, polysaccharides, celluloses, and polyisoprenes. Crosslinking is useful in many polymers for imparting structural integrity and rigidity to the microparticle. The size range of the microparticles can vary. In some embodiments, the microparticles range in diameter from about 0.3 micrometers to about 120 micrometers, and other embodiments, from about 0.5 micrometers to about 40 micrometers, and in still other embodiments, from about 2 micrometers to about 10 micrometers.
  • Affinity chromatography may be performed on a wide range of samples that contain the target of interest. In some instances, the samples are obtained from subjects (including humans, primates, non-primate mammals, birds, reptiles, and amphibians). For example, the sample may be a bodily fluid. The sample may also be cultured bacteria or other cells or tissues that express the target of interest, a cell culture supernatant, or a lysate from bacterial or eukaryotic cells. In addition, one or more purification steps may be performed on the sample to enrich the target prior to affinity chromatography using the affinity ligands described herein.
  • The sample is applied to the solid support to allow the target to be bound by the affinity ligand. The conditions for the binding may be neutral or physiological. To remove non-specifically or weakly bound non-target compounds, a washing step may be performed using neutral buffer conditions as described above in Section I.
  • To elute the target bound to the affinity ligand-solid support, an elution step is performed using mild elution conditions as described above in Section I. Under mild elution conditions, the binding of the affinity ligand to its target is reduced such that the affinity ligand and the target substantially dissociate from each other under these conditions. In one aspect, the mild elution conditions may comprise a pH of about 4.0 to about 5.5. In another aspect, the mild elution conditions may comprise about 1-2 M MgCl2. In some instances, elution may be performed as a single-step elution such that the target bound to the affinity ligand-solid support is eluted by exposing it to the mild elution conditions directly. In some instances, elution may be performed as a multiple-step elution such that the target bound to the affinity ligand-solid support is eluted by exposing it sequentially to multiple mild elution conditions having increasing salt concentrations or decreasing pH. In some instances, elution may be performed using a pH or salt gradient such that the target bound to the affinity ligand-solid support is eluted by exposing it to dynamic elution conditions as a gradient of linearly increasing salt concentration or decreasing pH.
  • The eluted target is useful for a variety of applications. For example, in the case that the target is an antibody (an immunoglobulin, such as IgM, IgA, or IgE), the eluted immunoglobulin target may be used, for example, as a therapeutic molecule, or as a diagnostic or laboratory reagent. In some instances, additional steps may be performed on the eluted target to prepare it for subsequent uses (for example, subsequent chromatography steps, filtration like ultrafiltration or diafiltration, dialysis, labeling, etc.).
  • IV. Kits
  • Kits containing affinity ligands that specifically bind to a target under neutral buffer conditions and elute the target under mild elution conditions are also contemplated. Kits may include one or more types of affinity ligands. The affinity ligands in the kit may be specific for the same target or for different targets. If the affinity ligands are specific for the same target, they may be specific for different epitopes of the target. In some instances, the affinity ligands are labeled (for example, with a peptide tag, or a chemical moiety for site-specific coupling to a matrix). The kits may include affinity ligands bound to a solid support. In some examples, the affinity ligand-bound solid support is packed into a chromatography column. Sometimes, the chromatography column is provided separately and the affinity ligand-bound solid support will be packed into the chromatography column prior to use. In some cases, the kit provides the affinity ligand and solid support separately along with instructions for coupling the affinity ligand to the solid support. The kit may further include an equilibration buffer, a washing buffer, an elution buffer, or some combination of these buffers. In some cases, more than one washing buffer or elution buffer is provided. Multiple elution buffers may be provided, with each elution buffer having a different elution condition (such as pH, salt type, or salt concentration). For example, different elution buffers may be provided for different affinity ligands included in the kit.
  • Exemplary buffers as referenced in this disclosure may include, for example, citrate, MES, or Bis-Tris, amongst others.
  • The foregoing description of certain embodiments, including illustrated embodiments, has been presented only for the purpose of illustration and description and is not intended to be exhaustive or to limit the disclosure to the precise forms disclosed. Numerous modifications, adaptations, and uses thereof will be apparent to those skilled in the art without departing from the scope of the disclosure. Certain features that are described in this specification in the context of separate embodiments can also be implemented in combination in a single implementation. Conversely, various features that are described in the context of a single implementation can also be implemented in multiple ways separately or in any suitable subcombination. Moreover, although features may be described above as acting in certain combinations, one or more features from a combination can in some cases be excised from the combination, and the combination may be directed to a subcombination or variation of a subcombination. Thus, particular embodiments have been described. Other embodiments are within the scope of the disclosure.
  • EXAMPLES
  • The following examples are offered to illustrate, but not to limit the claimed invention.
  • In the examples described below, antibodies were generated using the HuCAL PLATINUM® library that includes the CysDisplay® selection technology (Rothe, C., et al., 2008. “The human combinatorial antibody library HuCAL GOLD combines diversification of all six CDRs according to the natural immune system with a novel display method for efficient selection of high-affinity antibodies.” J Mol Biol 376(4): 1182-1200.). The aim was to generate Fab antibodies against human IgM, IgA, and IgE that bind at neutral pH but can be eluted from the antigen under mild conditions (e.g. pH 4 to pH 5). Selection on the antigens using elution under mild conditions resulted in 14 antibodies against IgM, 40 antibodies against IgA, and 17 antibodies against IgE. Assays were performed to identify the antibodies with desirable properties for use as affinity chromatography reagent.
  • Example 1
  • This example provides a description of the materials and methods used in performing the experiments described in Example 2.
  • The antigens and closely related antigens (CRAs) are listed in Table 1 below.
  • TABLE 1
    Antigens and CRAs
    Antigen/CRA Reference Source
    hIgM Ag05450 Human IgM, human plasma (AbD Serotec
    5275-5504)
    hIgA Ag5451 Human IgA, human colostrum (AbD Serotec
    5111-5504)
    hIgM Ag04815 Human IgM, human serum (Sigma I8260)
    hIgA Ag4813 Human IgA, human colostrum (Sigma
    I2636)
    hIgE Ag05681 Human IgE with lambda light chain,
    AbD00264
    hIgE Ag05682 Human IgE with kappa light chain, (AbD
    Serotec, HCA190)
    hIgE Ag05711 Human IgE, myeloma (AbD Serotec
    PHP142)
    hIgG1lambda Ag05029 Human IgG1 lambda, human myeloma
    plasma (Sigma I5029)
    hIgG1lambda Ag05419 Human IgG1 lambda, human myeloma
    plasma (Sigma I5029)
    hIgG1kappa Ag05153 Human IgG1 kappa, human myeloma
    plasma (Sigma I5154)
  • Recombinant antibodies were isolated from the HuCAL PLATINUM® library of human antibody genes by three iterative rounds of panning with the antigens as described in Table 1, using standard protocols. (Knappik et al., J. Mol. Biol. 2000, 296:57-86; Prassler et al., J. Mol. Biol. 2011, 413:261-278; Krebs et al., J. Immunol. Methods 2001, 254:67-84; Jarutat et al., Biol. Chem. 2006, 387:995-1003.)
  • For pannings 274.17-274.22 (see Table 2), the antigens were passively adsorbed to microtiter plates (F96 Maxisorp™ Nunc-Immuno Plate #442404) for use in “solid phase panning” as described below.
  • For pannings 274.4-274.9 and 289.12-14, the antigens were coupled to Dynal M-450 Epoxy beads (Invitrogen 140-11). The antigen coupled beads were incubated overnight at room temperature, blocked by addition of Tris, pH7.4, and subsequently re-suspended in PBS.
  • The phage antibody library was incubated with blocking buffer containing the CRAs as set forth in Table 1 and Table 2. The blocked library was then incubated with the antigen coupled beads, and non-specific or blocked antibodies were washed off. Specific antibody phage were eluted as noted in Table 2, by incubation with pH 3 elution buffer as control selection (150 mM NaCl, 100 mM Citrate buffer pH 3), with pH 4 elution buffer (150 mM NaCl, 100 mM Citrate buffer pH 4), or pH 5 elution buffer (150 mM NaCl, 100 mM Citrate buffer or 150 mM NaCl, 100 mM Sodium Acetate buffer) for the pH elution conditions or with high salt elution buffer (2M MgCl2 in PBS). The pH elution condition buffers were neutralized with 1M Tris before E. coli infection. Phagemid containing bacteria were grown overnight at 37° C. and new antibody displaying phage were produced for the next panning round.
  • After 3 rounds of panning, the enriched pool of Fab genes was isolated and inserted into the E. coli expression vector pMx11-FH that leads to functional periplasmic expression of monovalent Fab fragments. (Rauchenberger et al., J. Biol. Chem. 2003, 278:38194-38205.) Each Fab includes a FLAG tag and a 6×His tag in tandem at the C-terminus of the heavy chain.
  • E. coli TG1F (TG1 depleted for the F pilus; Rauchenberger et al. 2003) was then transformed with the ligated expression vectors. Following transformation, 368 individual colonies were randomly picked for each panning and grown in microtiter plates, which were then stored with 15% glycerol at −80° C. These master plates were replicated and the resulting daughter plates were used for expression of the antibodies. After induction of antibody expression with 1 mM Isopropyl-β-D-thiogalactopyranosid (IPTG) overnight at 22° C., the cultures were chemically lysed, and the crude extracts were tested in Enzyme-Linked Immunosorbent Assay (ELISA) with the immobilized antigens and closely related antigens for the presence of antibody fragments that bind specifically to the immobilized antigens. In addition binding to IgM or IgA from a different source (Sigma) was tested. These antigens were also used to determine which antibodies can be washed of the ELISA plate after incubation with the different elution buffers for 20 min. The sequences of the antibody VH and VL complementarity-determining regions (CDR) were determined from a selection of the clones that gave a strong signal on the antigens in the ELISA (at least 5-fold above the background signal, antigens from AbD Serotec and Sigma) and a weak or no signal on the CRAs and on the antigen after elution buffer treatment (below 5-fold above the background signal). Clones containing antibodies with unique sequence were chosen for antibody production.
  • TABLE 2
    Panning, Blocking and Screening Antigens
    Pan- Panning Elution Primary ELISA
    code Antigens Strategy CRAs Used for Blocking Screening on Antigens
    274.4 hIgM pH 3 Elution Ag05029 & Ag05153 Ag05450 (hIgM)
    (Ag05450) (hIgG1 lambda & kappa) to Ag05029 (hIgG1lambda)
    final conc. of 50 μg/mL Ag05153 (hIgG1kappa)
    100 μl cell culture medium Ag04815 (hIgM, Sigma)
    Ag04815 (elution buffer
    incubation)
    274.5 hIgA Bead Ag05029 & Ag05153 Ag05451 (hIgA)
    (Ag05451) Panning (hIgG1 lambda & kappa) to Ag05029 (hIgG1lambda)
    pH 3 Elution final conc. of 50 μg/mL Ag05153 (hIgG1kappa)
    100 μl cell culture medium Ag04813 (hIgA, Sigma)
    Ag04813 (elution buffer
    incubation)
    274.6 hIgM Bead Ag05029 & Ag05153 Ag05450 (hIgM)
    (Ag05450) Panning (hIgG1 lambda & kappa) to Ag05029 (hIgG1lambda)
    pH 4 Elution final conc. of 50 μg/mL Ag05153 (hIgG1kappa)
    100 μl cell culture medium Ag04815 (hIgM, Sigma)
    Ag04815 (elution buffer
    incubation)
    274.7 hIgA Bead Ag05029 & Ag05153 Ag05451 (hIgA)
    (Ag05451) Panning (hIgG1 lambda & kappa) to Ag05029 (hIgG1lambda)
    pH 4 Elution final conc. of 50 μg/mL Ag05153 (hIgG1kappa)
    100 μl cell culture medium Ag04813 (hIgA, Sigma)
    Ag04813 (elution buffer
    incubation)
    274.8 hIgM Bead Ag05029 & Ag05153 Ag05450 (hIgM)
    (Ag05450) Panning (hIgG1 lambda & kappa) to Ag05029 (hIgG1lambda)
    High Salt final conc. of 50 μg/mL Ag05153 (hIgG1kappa)
    Elution (2M 100 μl cell culture medium Ag04815 (hIgM, Sigma)
    MgCl2) Ag04815 (elution buffer
    incubation)
    274.9 hIgA Bead Ag05029 & Ag05153 Ag05451 (hIgA)
    (Ag05451) Panning (hIgG1 lambda & kappa) to Ag05029 (hIgG1lambda)
    High Salt final conc. of 50 μg/mL Ag05153 (hIgG1kappa)
    Elution (2M 100 μl cell culture medium Ag04813 (hIgA, Sigma)
    MgCl2) Ag04813 (elution buffer
    incubation)
    274.17 hIgM Bead Ag05029 & Ag05153 Ag05450 (hIgM)
    (Ag05450) Panning (hIgG1 lambda & kappa) to Ag05029 (hIgG1lambda)
    pH 3 Elution final conc. of 50 μg/mL Ag05153 (hIgG1kappa)
    100 μl cell culture medium Ag04815 (hIgM, Sigma)
    Ag04815 (elution buffer
    incubation)
    274.18 hIgA Bead Ag05029 & Ag05153 Ag05451 (hIgA)
    (Ag05451) Panning (hIgG1 lambda & kappa) to Ag05029 (hIgG1lambda)
    pH 3 Elution final conc. of 50 μg/mL Ag05153 (hIgG1kappa)
    100 μl cell culture medium Ag04813 (hIgA, Sigma)
    Ag04813 (elution buffer
    incubation)
    274.19 hIgM Bead Ag05029 & Ag05153 Ag05450 (hIgM)
    (Ag05450) Panning (hIgG1 lambda & kappa) to Ag05029 (hIgG1lambda)
    pH 4 Elution final conc. of 50 μg/mL Ag05153 (hIgG1kappa)
    100 μl cell culture medium Ag04815 (hIgM, Sigma)
    Ag04815 (elution buffer
    incubation)
    274.20 hIgA Bead Ag05029 & Ag05153 Ag05451 (hIgA)
    (Ag05451) Panning (hIgG1 lambda & kappa) to Ag05029 (hIgG1lambda)
    pH 4 Elution final conc. of 50 μg/mL Ag05153 (hIgG1kappa)
    100 μl cell culture medium Ag04813 (hIgA, Sigma)
    Ag04813 (elution buffer
    incubation)
    274.21 hIgM Bead Ag05029 & Ag05153 Ag05450 (hIgM)
    (Ag05450) Panning (hIgG1 lambda & kappa) to Ag05029 (hIgG1lambda)
    High Salt final conc. of 50 μg/mL Ag05153 (hIgG1kappa)
    Elution (2M 100 μl cell culture medium Ag04815 (hIgM, Sigma)
    MgCl2) Ag04815 (elution buffer
    incubation)
    274.22 hIgA Bead Ag05029 & Ag05153 Ag05451 (hIgA)
    (Ag05451) Panning (hIgG1 lambda & kappa) to Ag05029 (hIgG1lambda)
    High Salt final conc. of 50 μg/mL Ag05153 (hIgG1kappa)
    Elution (2M 100 μl cell culture medium Ag04813 (hIgA, Sigma)
    MgCl2) Ag04813 (elution buffer
    incubation)
    289.12 hIgE Bead Ag05419 & Ag05153 Ag05681 (hIgE/lambda)
    lambda Panning (hIgG1 lambda & kappa) to Ag05682 (hIgE/kappa)
    (Ag05681) pH 4 final conc. of 50 μg/mL Ag05419 (hIgG1lambda)
    hIgE/kappa Elution, 100 μl cell culture medium Ag05153 (hIgG1kappa)
    (Ag05682) 10 min Ag05681 (elution buffer
    Elution Step incubation)
    289.13 hIgE Bead Ag05419 & Ag05153 Ag05681 (hIgE/lambda)
    lambda Panning (hIgG1 lambda & kappa) to Ag05682 (hIgE/kappa)
    (Ag05681) pH 4 final conc. of 50 μg/mL Ag05419 (hIgG1lambda)
    hIgE/kappa Elution, 100 μl cell culture medium Ag05153 (hIgG1kappa)
    (Ag05682) 5 min Ag05681 (elution buffer
    Elution Step incubation)
    289.14 hIgE Bead Ag05419 & Ag05153 Ag05681 (hIgE/lambda)
    lambda Panning (hIgG1 lambda & kappa) to Ag05682 (hIgE/kappa)
    (Ag05681) pH 5 final conc. of 50 μg/mL Ag05419 (hIgG1lambda)
    hIgE/kappa Elution, 100 μl cell culture medium Ag05153 (hIgG1kappa)
    (Ag05682) 10 min Ag05681 (elution buffer
    Elution Step incubation)
  • The ELISA screening protocol is set forth in the Table 3 below.
  • TABLE 3
    ELISA Screening Protocol
    Plates 384 well Maxisorp microtiter plates (MTP), black,
    flat bottom, Polystyrene (Nunc 460518)
    Coating 20 μL/well of antigens at 5 μg/mL in PBS pH 7.4;
    overnight (o/n) incubation at 4° C.
    Wash 2x PBST (PBS with 0.05% Tween ® 20)
    Blocking 100 μL of 5% non-fat dried milk in PBST for 1-2 h
    at room temperature (RT)
    Wash 2x PBST
    Primary Ab
    20 μL/well of crude E. coli lysate of expression
    (HuCAL ®-Fab) culture containing HuCAL ®-Fab (pre-blocked with
    a final concentration of 5% non-fat dried milk in
    PBST), 1 h at RT
    Wash 5x PBST
    Secondary Ab
    20 μL/well of anti-His tag, HRP conjugate (Qiagen
    34460), 1:2000 dilution in HiSpec buffer (AbD
    Serotec BUF049), 1 h at RT
    Wash 5x PBST
    Detection
    20 μL/well QuantaBlu ® (Thermo Scientific 15169)
    Reader Settings Excitation at 320 ± 25 nm, emission at 420 ± 35 nm
  • E. coli TG1F cultures (250 mL) containing the chosen antibody genes were grown at 30° C. until OD600nm reached 0.5, and the antibody expression was induced by adding IPTG to a final concentration of 1 mM. After further incubation for at least 14 hours at 30° C., the cells were harvested, chemically lysed, and the soluble crude extract was subjected to one-step affinity chromatography (Ni-NTA Agarose, Qiagen 1018240). After elution of the purified antibodies from the column, the buffer was changed from elution buffer to 3×PBS, pH 7.4, and the concentration was determined by UV280 nm measurement. Purity and activity was tested subsequently by Coomassie-stained SDS-PAGE and ELISA, respectively.
  • The quality control ELISA (indirect ELISA) protocol using purified HuCAL® antibodies is set forth in Table 4 below.
  • TABLE 4
    QC ELISA Protocol
    Plate 384 well Maxisorp MTP, black, flat bottom,
    Polystyrene (Nunc 460518)
    Coating 20 μL/well of antigen at 5 μg/mL in PBS, o/n at 4° C.
    Wash 2x PBST (PBS with 0.05% Tween-20 ®)
    Blocking 100 μL of 5% non-fat dried milk in PBST for 1-2 hr at
    RT
    Wash 2x PBST
    Primary Ab
    20 μL/well of antibody at 2 μg/mL in PBST for 1 hr at
    (HuCAL ®-Fab) RT
    Wash 5x PBST
    Secondary Ab
    20 μL/well of anti-His tag, HRP conjugate (Qiagen
    34460), 1:2000 dilution in HiSpec buffer (AbD Serotec
    BUF049), 1 hr at RT
    Wash 5x PBST
    Optional: manually wash specific wells with elution
    buffer, incubate for 5 min, remove buffer, repeat 2x
    Detection
    20 μL/well QuantaBlu ® (Thermo Scientific 15169)
    Reader Settings Excitation at 320 ± 25 nm, emission at 420 ± 35 nm
    Background Signal values on unspecific antigens (BSA, GST) were
    used for the calculation of background signal
  • Example 2
  • The results of the methods described in Example 1 are described in this example.
  • Primary ELISA Screening
  • The outcome of a panning is tested in the primary screening on several antigens and closely related antigens (CRA). The antigen role defines whether positive (+) signals are expected (positive controls for the panning antigen) or whether no signal (−) is wanted (negative controls; CRAs).
  • An ELISA signal on an antigen plate (positive control) is considered a hit if the signal is 5-fold above background. Background is the average value of a number of wells on the plate that were not treated with antigen or antibody.
  • A signal on a CRA (negative control) is counted, and during the analysis subtracted from the positive hits, if the signal is at least 2-fold above background.
  • A clone is considered a hit if it is positive on all antigens (positive controls) and negative on all CRAs (negative controls).
  • The values in Table 5 indicate the numbers of hits according to the above definition. The Analysis column indicates the number of hits that no longer bind to the antigen under the test elution conditions.
  • TABLE 5
    Primary screening ELISA results
    Antigen Role
    + +
    hIgG1 hIgG1 hIgM,
    hIgM, hIgM, lambda, kappa, Ag04815 &
    Panning Ag05450 Ag04815 Ag05029 Ag05153 elution buffer Analysis
    274.4  234 43 0 2 11, pH 3 38
    274.6  191 59 1 2 5, pH 4 55
    274.8  234 163 3 2 7, high salt 153
    274.17 239 8 0 15 1, pH 3 4
    274.19 228 106 2 0 1, pH 4 93
    274.21 28 0 1 1 7, high salt 0
    hIgG1 hIgG1 hIgA,
    hIgA, hIgA, lambda, kappa, Ag04813 &
    Ag05451 Ag04813 Ag05029 Ag05153 elution buffer Analysis
    274.5  205 200 2 1 71, pH 3 125
    274.7  223 201 1 31 96, pH 4 94
    274.9  285 286 1 8 271, high salt 5
    274.18 193 215 3 1 52, pH 3 132
    274.20 173 184 0 6 119, pH 4 58
    274.22 174 220 6 1 166, high salt 16
    hIgG1 hIgG1 hIgE,
    hIgE, hIgE, lambda, kappa, Ag05681 &
    Ag05681 Ag05682 Ag05419 Ag05153 elution buffer Analysis
    289.12 9 1 0 3 4, pH 4 1
    289.13 20 24 24 164 208, pH 4, 9
    not used*
    289.14 25 186 147 10 21, pH 5 10
    *The number of hits was too high, so this result was considered to be an artifact and was not used to pick clones for further characterization.
  • Sequencing and Identification of Unique Antibodies
  • For the IgE project, 20 clones (289.12: 1, 289.13: 9, 289.14: 10) were sequenced and resulted in 17 different antibodies (AbD22512, AbD22628-22643). The heavy and light chain CDR1, CDR2, and CDR3 region sequences of these antibodies are shown in FIG. 1A and FIG. 1B.
  • For the IgA project, a total of 45 clones (274.7: 18, 274.9: 3, 274.20: 15, 274.22: 9) were selected for further analysis. Sequencing of the gene regions coding for VH and VL revealed 40 different antibodies (AbD20776-20791, AbD20797-20799, AbD20801-20812, AbD20813-20821). The heavy and light chain CDR1, CDR2, and CDR3 region sequences of these antibodies are shown in FIG. 2A and FIG. 2B.
  • For the IgM project, a total of 50 clones (274.6: 15, 274.8: 20, 274.19: 15) were selected for further analysis. Sequencing of the gene regions coding for VH and VL revealed 14 different antibodies (AbD20768-20775, AbD20792-20796, AbD20800). The heavy and light chain CDR1, CDR2, and CDR3 region sequences of these antibodies are shown in FIG. 3A FIG. 3B; full heavy and light chain sequences for certain of these antibodies is shown in FIGS. 4A-4I.
  • The antibodies were expressed, purified via affinity chromatography and tested using ELISA. The results are shown in FIGS. 5-9 and in Tables 6-8 and described further below.
  • For each clone shown, a number of antigens and conditions were tested. The bars indicate the signal strength plotted as specific fluorescence divided by background fluorescence. The designation of the clones is “clone-name.batch-number.” For example, AbD20768.1 is the first batch of AbD20768
  • FIG. 5: BSA and N1-CD33-6×His are unrelated antigens. hIgG1Kap-ctrl and hIgG1lambdaCtrl are CRAs (human IgG1 isotype with kappa and lambda light chain, respectively). hIgM is human IgM from AbD Serotec (5275-5504). H-IgM (Sigma) is human IgM from Sigma (18260). Antigen with “pH4 wash” indicate the residual signal on that antigen after 3 consecutive 5 minute incubations of the corresponding wells with the elution buffer (pH 4).
  • FIG. 6: BSA and N1-CD33-His6 are unrelated antigens. hIgG1Kap-ctrl and hIgG1lambdaCtrl are CRAs (human IgG1 isotype with kappa and lambda light chain, respectively). hIgM is human IgM from AbD Serotec (5275-5504). H-IgM (Sigma) is human IgM from Sigma (18260). Antigen with “2M MgCl2 wash” indicate the residual signal on that antigen after 3 consecutive 5 minute incubations of the corresponding wells with the elution buffer (2M MgCl2 in PBS).
  • FIG. 7: BSA and GST are unrelated antigens. hIgG1Kap-ctrl and hIgG1lambdaCtrl are CRAs (human IgG1 isotype with kappa and lambda light chain, respectively). hIgA is human IgA from AbD Serotec (5111-5504). HIgA (Sigma) is human IgA from Sigma (12636). Antigen with “pH4 wash” indicate the residual signal on that antigen after 3 consecutive 5 minute incubations of the corresponding wells with the elution buffer (pH 4). A number of antibodies that were originally positive in the primary screening turned out to be negative here (AbD20801 to AbD20812).
  • FIG. 8: BSA and N1-CD33-His6 are unrelated antigens. hIgG1Kap-ctrl and hIgG1lambdaCtrl are CRAs (human IgG1 isotype with kappa and lambda light chain, respectively). hIgA is human IgA from AbD Serotec (5111-5504). HIgA (Sigma) is human IgA from Sigma (12636). Antigen with “2M MgCl2 wash” indicate the residual signal on that antigen after 3 consecutive 5 minute incubations of the corresponding wells with the elution buffer (2M MgCl2 in PBS). A number of antibodies that were originally positive in the primary screening turned out to be negative here (AbD20814-AbD20821).
  • FIG. 9: BSA and GST are unrelated antigens. hIgG1Kap-ctrl and hIgG1lambdaCtrl are CRAs (human IgG1 isotype with kappa and lambda light chain, respectively). hIgG4 kappa is human IgG4 with kappa light chain (Sigma 14639). hIgM is human IgM from AbD Serotec (5275-5504). AbD00264_hIgG1f is a human antibody derived from HuCAL and formatted into the IgG1 isotype. AbD00264_hIgE is the same antibody formatted into the IgE isotype. AbD18705_hIgE is another HuCAL-derived antibody formatted into the IgE isotype. hIgE is human IgE from myeloma (AbD Serotec PHP 142).
  • TABLE 6
    Overview of Specific Anti-hIgM Antibodies
    Antigen Elution
    Antibody Antigen Number Name conc. [mg/ml] buffer
    AbD20768.1 Ag05450 hIgM 1.56 pH 4
    AbD20769.1 Ag05450 hIgM 1.65 pH 4
    AbD20770.1 Ag05450 hIgM 1.6 pH 4
    AbD20771.1 Ag05450 hIgM 1.7 pH 4
    AbD20772.1 Ag05450 hIgM 0.88 pH 4
    AbD20773.1 Ag05450 hIgM 0.82 pH 4
    AbD20774.1 Ag05450 hIgM 1.48 pH 4
    AbD20775.1 Ag05450 hIgM 1.11 pH 4
    AbD20800.1 Ag05450 hIgM 0.72 pH 4
    AbD20792.1 Ag05450 hIgM 0.97 2M MgCl2
    AbD20793.1 Ag05450 hIgM 0.3 2M MgCl2
    AbD20794.1 Ag05450 hIgM 0.93 2M MgCl2
    AbD20795.1 Ag05450 hIgM 0.73 2M MgCl2
    AbD20796.1 Ag05450 hIgM 0.93 2M MgCl2
  • TABLE 7
    Overview of Specific Anti-hIgA Antibodies
    Antigen conc.
    Antibody Number Antigen Name [mg/ml] Elution buffer
    AbD20776.1 Ag05451 hIgA 1.13 pH 4
    AbD20777.1 Ag05451 hIgA 0.96 pH 4
    AbD20778.1 Ag05451 hIgA 1.32 pH 4
    AbD20779.1 Ag05451 hIgA 1.38 pH 4
    AbD20780.1 Ag05451 hIgA 1.54 pH 4
    AbD20781.1 Ag05451 hIgA 0.99 pH 4
    AbD20782.1 Ag05451 hIgA 0.99 pH 4
    AbD20783.1 Ag05451 hIgA 1.25 pH 4
    AbD20784.1 Ag05451 hIgA 1.03 pH 4
    AbD20785.1 Ag05451 hIgA 1.46 pH 4
    AbD20786.1 Ag05451 hIgA 1.22 pH 4
    AbD20787.1 Ag05451 hIgA 1.35 pH 4
    AbD20788.1 Ag05451 hIgA 1.5 pH 4
    AbD20789.1 Ag05451 hIgA 1.68 pH 4
    AbD20790.1 Ag05451 hIgA 1.61 pH 4
    AbD20791.1 Ag05451 hIgA 1.25 pH 4
    AbD20797.1 Ag05451 hIgA 1.43 2M MgCl2
    AbD20798.1 Ag05451 hIgA 1.43 2M MgCl2
    AbD20799.1 Ag05451 hIgA 1.04 2M MgCl2
    AbD20813.1 Ag05451 hIgA 1.21 2M MgCl2
  • TABLE 8
    Overview of Specific Anti-hIgE Antibodies
    Antigen Antigen conc. Elution
    Antibody Number Name [mg/ml] buffer
    AbD22512.1 Ag05681 hIgE 1.76 pH 4
    AbD22628.1 Ag05681 hIgE 1.41 pH 4
    AbD22629.1 Ag05681 hIgE 1.83 pH 4
    AbD22630.1 Ag05681 hIgE 1.78 pH 4
    AbD22631.1 Ag05681 hIgE 1.75 pH 4
    AbD22632.1 Ag05681 hIgE 1.77 pH 4
    AbD22633.1 Ag05681 hIgE 1.59 pH 4
    AbD22634.1 Ag05681 hIgE 0.69 pH 4
    AbD22635.1 Ag05681 hIgE 1.72 pH 5
    AbD22636.1 Ag05681 hIgE 1.70 pH 5
    AbD22637.1 Ag05681 hIgE 1.20 pH 5
    AbD22638.1 Ag05681 hIgE 1.70 pH 5
    AbD22639.1 Ag05681 hIgE 1.37 pH 5
    AbD22640.1 Ag05681 hIgE 1.58 pH 5
    AbD22641.1 Ag05681 hIgE 1.76 pH 5
    AbD22642.1 Ag05681 hIgE 0.90 pH 5
    AbD22643.1 Ag05681 hIgE 1.69 pH 5
  • Example 3
  • Selected antibody Fab fragments isolated as described in Example 2 were used as affinity ligands to purify an immunoglobulin target. Two anti-IgM antibody Fab fragments were selected as representative affinity ligands: AbD20771 and AbD20775. The affinity ligands were expressed in E. coli as monomers having a FLAG-His6 tag on the heavy chain. The ligands were purified using Ni-NTA agarose.
  • Four hIgM molecules were selected as exemplary target molecules to assess the purification capability of the ligands. These ligands are set forth in Table 9. Supernatant containing these ligands was collected during culturing of the cell lines noted in Table 9. OBT1524 hIgM (AbD Serotec, Bio-Rad), an IgM protein purified from myeloma serum, was used as a standard control.
  • TABLE 9
    IgM Target Molecules For Purification
    Target Molecule Cell Line Expression
    AbD00264 hIgM lambda (AbD Expression in a stable human HKB11
    Serotec, BioRad) cell pool (AbD Serotec, BioRad)
    AbD18705 hIgM kappa (AbD Expression in a stable human HKB11
    Serotec, BioRad) cell pool (AbD Serotec, BioRad)
    AbD18777 hIgM kappa (AbD Transient expression in human HKB11
    Serotec, BioRad) cell line (AbD Serotec, BioRad)
  • The purified affinity ligands were coupled to 1 ml “HiTrap NHS-activated HP” columns according to manufacture's protocol (GE Healthcare #17-0716-01), with 7 mg of the ligand being applied to the column. The columns were run on a GE AKTAxpress™ FPLC instrument. The supernatant sample (200 ml) was loaded using a flow rate of 0.4 ml/min (62 cm/h; retention time of 2.4 min) using PBS (pH about 7.0-7.2) as binding and washing buffer. The elution buffer was 100 mM citrate, 150 mM NaCl, pH 4.0. Following elution, a neutralization buffer (1 M Tris/HCl pH 9.0) was used to neutralize the pH of the purified IgM targets.
  • Exemplary results are shown for target molecule AbD18705 IgM. Results for the other two target molecules were similar. Elution profiles (UV 280 nm) are shown in FIG. 10 (collected fractions are indicated). The AbD18705 IgM target molecule is captured from cell culture supernatant and eluted under mild conditions (pH 4.0) using both affinity ligand AbD20771 and AbD20775.
  • To assess the purity and integrity of the purified target molecules, the purified protein fractions (A3/4/5 for the AbD20775 column and A10/11/12 for the AbD20771 column, as identified in FIG. 10; 1 μg protein/lane) were run on 4-20% Mini-PROTEAN™ Stain-free TGX gels (Bio-Rad). Exemplary reducing and non-reducing lanes are shown for each fraction, respectively, in FIG. 11. Comparisons were made to the purified OBT1524 IgM standard control protein (FIG. 11, right panel). The purity of the AbD18705 IgM target molecule in the fractions is high under reducing conditions and both heavy and light chains can be detected. Under non-reducing conditions, assembled IgM is detectable.
  • Size exclusion chromatography (SEC) using a Superose 6 SEC column (GE Healthcare) was also performed to assess the integrity of the purified target molecules. FIG. 12 shows an overlay of SEC runs (UV280 nm signal) for fractions A4/5/10/11/12 as identified in FIG. 10. The peak retention volume was 10.8 ml, corresponding to a MW of 916 kDa (calculated MW for IgM AbD18705 in pentameric form without glycosylation is 859 kDa. All fractions show an identical elution behaviour indicating that assembled, non-aggregated IgM is present in the fractions. No degradation or aggregation is visible.
  • Finally, ELISA analysis of the various fractions was performed to assess activity and specificity of the purified AbD18705 hIgM target molecule. Negative control antigens (BSA, N1-CD33-6×His, GST) were coated at 5 μg/ml onto the ELISA plate along with the specific antigen His-GFP, which is the antigen of AbD18705. The six fractions A3/4/5 and A10/11/12 of the purified IgM AbD18705, numbered 1 to 6 here, were added (20 μl each) after washing, blocking and additional washing. Detection was performed using an anti-human IgM HRP conjugate (AbD Serotec) in combination with Quantablu™ substrate. As shown in FIG. 13, the purified AbD18705 hIgM antibody fractions all recognize the His-GFP antigen specifically and, thus, have a native active conformation.
  • All patents, patent publications, patent applications, journal articles, books, technical references, and the like discussed in the instant disclosure are incorporated herein by reference in their entirety for all purposes.
  • It is to be understood that the figures and descriptions of the invention have been simplified to illustrate elements that are relevant for a clear understanding of the invention. It should be appreciated that the figures are presented for illustrative purposes and not as construction drawings. Omitted details and modifications or alternative embodiments are within the purview of persons of ordinary skill in the art.
  • It can be appreciated that, in certain aspects of the invention, a single component may be replaced by multiple components, and multiple components may be replaced by a single component, to provide an element or structure or to perform a given function or functions. Except where such substitution would not be operative to practice certain embodiments of the invention, such substitution is considered within the scope of the invention.
  • The examples presented herein are intended to illustrate potential and specific implementations of the invention. It can be appreciated that the examples are intended primarily for purposes of illustration of the invention for those skilled in the art. There may be variations to these diagrams or the operations described herein without departing from the spirit of the invention. For instance, in certain cases, method steps or operations may be performed or executed in differing order, or operations may be added, deleted or modified.
  • Different arrangements of the components depicted in the drawings or described above, as well as components and steps not shown or described are possible. Similarly, some features and sub-combinations are useful and may be employed without reference to other features and sub-combinations. Embodiments of the invention have been described for illustrative and not restrictive purposes, and alternative embodiments will become apparent to readers of this patent. Accordingly, the present invention is not limited to the embodiments described above or depicted in the drawings, and various embodiments and modifications can be made without departing from the scope of the claims below.

Claims (16)

1-37. (canceled)
38. A method of selecting an affinity ligand that specifically binds to a target molecule under neutral buffer conditions and has reduced binding strength to the target molecular under mild elution conditions, the method comprising the steps of:
a) expressing a naive affinity ligand library to produce a plurality of affinity ligands;
b) providing a solid support linked to a target;
c) contacting the solid support with the plurality of affinity ligands;
d) washing the solid support with a wash buffer to remove unbound affinity ligands, wherein the wash buffer comprises neutral buffer conditions;
e) contacting the solid support with an elution buffer comprising (i) a pH of about 4.0 to about 5.5 or (ii) about 1-2 M MgCl2; and
f) identifying affinity ligands that substantially dissociate from the solid support in the elution buffer.
39. The method of claim 38, wherein the plurality of affinity ligands is encoded by a plurality of nucleic acid sequences.
40. The method of claim 39, wherein the plurality of nucleic acid sequences comprise a heterologous promoter operably linked thereto.
41. The method of claim 38, wherein the plurality of affinity ligands are expressed on a plurality of phage.
42. The method of claim 38, wherein the elution buffer comprises a pH of about 4.0 to about 5.5 and a relatively low salt concentration.
43. The method of claim 38, wherein the elution buffer comprises about 1 M to 2 M MgCl2 and a relatively neutral pH.
44. The method of claim 38, wherein the elution buffer comprises about 1 M to 2 M MgCl2 and a pH of about 6.0 to 8.0.
45. The method of claim 38, wherein the wash buffer comprises a pH of 6.0-8.0.
46. The method of claim 38, wherein the wash buffer comprises a relatively low salt concentration.
47. The method of claim 38, wherein the target is an immunoglobulin.
48. The method of claim 38, wherein the target is an immunoglobulin M (IgM), an immunoglobulin A (IgA), or an immunoglobulin E (IgE).
49. The method of claim 38, wherein the plurality of affinity ligands is a plurality of antibodies or derivatives thereof.
50. The method of claim 38, wherein the plurality of affinity ligands is a plurality of Fab fragments or derivatives thereof.
51. The method of claim 38, wherein the affinity ligand library is not preselected for characteristics favoring reduced binding strength to the target molecule under mild elution conditions.
52-58. (canceled)
US16/373,293 2015-05-28 2019-04-02 Affinity ligands and methods relating thereto Abandoned US20190248922A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US16/373,293 US20190248922A1 (en) 2015-05-28 2019-04-02 Affinity ligands and methods relating thereto

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201562167387P 2015-05-28 2015-05-28
US15/166,761 US10294306B2 (en) 2015-05-28 2016-05-27 Affinity ligands and methods relating thereto
US16/373,293 US20190248922A1 (en) 2015-05-28 2019-04-02 Affinity ligands and methods relating thereto

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US15/166,761 Division US10294306B2 (en) 2015-05-28 2016-05-27 Affinity ligands and methods relating thereto

Publications (1)

Publication Number Publication Date
US20190248922A1 true US20190248922A1 (en) 2019-08-15

Family

ID=57393716

Family Applications (2)

Application Number Title Priority Date Filing Date
US15/166,761 Active 2036-10-15 US10294306B2 (en) 2015-05-28 2016-05-27 Affinity ligands and methods relating thereto
US16/373,293 Abandoned US20190248922A1 (en) 2015-05-28 2019-04-02 Affinity ligands and methods relating thereto

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US15/166,761 Active 2036-10-15 US10294306B2 (en) 2015-05-28 2016-05-27 Affinity ligands and methods relating thereto

Country Status (4)

Country Link
US (2) US10294306B2 (en)
EP (2) EP3549606A1 (en)
CN (2) CN107614014B (en)
WO (1) WO2016191659A1 (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP7037148B2 (en) * 2015-11-09 2022-03-16 国立大学法人京都工芸繊維大学 A separating agent for human serum-derived IgG polyclonal antibody and a method for separating human serum-derived IgG polyclonal antibody using the same.
WO2021222546A1 (en) * 2020-05-01 2021-11-04 University Of Pittsburgh-Of The Commonwealth System Of Higher Education Compositions and methods for identifying nanobodies and nanobody affinities

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006059904A1 (en) * 2004-12-02 2006-06-08 Unilever N.V. Method for affinity purification
US20100105569A1 (en) * 2008-09-30 2010-04-29 Abbott Laboratories Methods of RNA Display
US20100216187A1 (en) * 2006-10-11 2010-08-26 Ablynx N.V. Amino acid sequences that bind to a desired molecule in a conditional manner
US20130078741A1 (en) * 2010-05-24 2013-03-28 Otsuka Chemical Co., Ltd Iga-binding peptide and purification of iga using the same
US20130336963A1 (en) * 2008-04-11 2013-12-19 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to two or more antigen molecules repeatedly
US20160024147A1 (en) * 2014-07-26 2016-01-28 Regeneron Pharmaceuticals, Inc. Purification Platform for Bispecific Antibodies

Family Cites Families (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5260203A (en) 1986-09-02 1993-11-09 Enzon, Inc. Single polypeptide chain binding molecules
ES2162823T5 (en) 1992-08-21 2010-08-09 Vrije Universiteit Brussel IMMUNOGLOBULINS DESPROVISTAS OF LIGHT CHAINS.
US6372425B1 (en) 1994-10-26 2002-04-16 Merck & Co., Inc. Large scale affinity chromatography of macromolecules
US5770380A (en) 1996-09-13 1998-06-23 University Of Pittsburgh Synthetic antibody mimics--multiple peptide loops attached to a molecular scaffold
US7115396B2 (en) 1998-12-10 2006-10-03 Compound Therapeutics, Inc. Protein scaffolds for antibody mimics and other binding proteins
US6818418B1 (en) 1998-12-10 2004-11-16 Compound Therapeutics, Inc. Protein scaffolds for antibody mimics and other binding proteins
CA2492631C (en) * 2002-05-31 2013-03-12 Genetic Technologies Limited Maternal antibodies as fetal cell markers to identify and enrich fetal cells from maternal blood
WO2004039337A2 (en) * 2002-10-31 2004-05-13 Protein Design Labs, Inc. Stable liquid pharmaceutical formulation of antibodies that are prone to isomerization
SG149759A1 (en) * 2007-07-10 2009-02-27 Millipore Corp Media for affinity chromatography
CA2725919A1 (en) * 2008-05-28 2009-12-03 Roland Antonius Paulus Romijn Method for the production of proteins or protein fragments
JP5654986B2 (en) * 2008-06-30 2015-01-14 モーフオテク・インコーポレーテツド Anti-GD2 antibodies and related methods and uses
WO2011111007A2 (en) 2010-03-11 2011-09-15 Rinat Neuroscience Corporation ANTIBODIES WITH pH DEPENDENT ANTIGEN BINDING
AU2012313594C1 (en) * 2011-09-30 2018-05-10 Chugai Seiyaku Kabushiki Kaisha Ion concentration-dependent binding molecule library
CN107827979A (en) 2012-03-16 2018-03-23 瑞泽恩制药公司 The engineered light chain antibody of histidine and the non-human animal through genetic modification for generating the antibody
JP2013253842A (en) * 2012-06-06 2013-12-19 Univ Of Tokyo Screening method for peptide connected to target molecule depending on ph
WO2014028354A1 (en) * 2012-08-13 2014-02-20 Regeneron Pharmaceuticals, Inc. Anti-pcsk9 antibodies with ph-dependent binding characteristics

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006059904A1 (en) * 2004-12-02 2006-06-08 Unilever N.V. Method for affinity purification
US20100216187A1 (en) * 2006-10-11 2010-08-26 Ablynx N.V. Amino acid sequences that bind to a desired molecule in a conditional manner
US20130336963A1 (en) * 2008-04-11 2013-12-19 Chugai Seiyaku Kabushiki Kaisha Antigen-binding molecule capable of binding to two or more antigen molecules repeatedly
US20100105569A1 (en) * 2008-09-30 2010-04-29 Abbott Laboratories Methods of RNA Display
US20130078741A1 (en) * 2010-05-24 2013-03-28 Otsuka Chemical Co., Ltd Iga-binding peptide and purification of iga using the same
US20160024147A1 (en) * 2014-07-26 2016-01-28 Regeneron Pharmaceuticals, Inc. Purification Platform for Bispecific Antibodies

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
Enzelberger et al. "HuCAL PLATINUM, a synthetic fab library optimized for sequence diversity and superior performance in mammalian expression systems" J. Molecular Biology (2011) 413, 216-278 (Year: 2011) *
Fassina et al. "Affinity purification of mouse monoclonal IgE using a protein A mimetic ligand (TG19318) immobilized on solid supports, J. Mol. Recogn. 11, 247-249, 1998 (Year: 1998) *
Horn et al. "A combinatorial histidine scanning library approach to engineer highly pH-dependent protein switches" Protein Science 2011, 20: 1619-1631 (Year: 2011) *
Park, "IgM antibody purification and analysis by immuno-affinity chromatography" 227(1), 2004; Abstracts of papers --American Chemical Society (Year: 2004) *

Also Published As

Publication number Publication date
EP3549606A1 (en) 2019-10-09
CN115925961A (en) 2023-04-07
US10294306B2 (en) 2019-05-21
EP3302553A1 (en) 2018-04-11
CN107614014A (en) 2018-01-19
EP3302553A4 (en) 2019-03-06
WO2016191659A1 (en) 2016-12-01
US20160347826A1 (en) 2016-12-01
CN107614014B (en) 2022-07-12

Similar Documents

Publication Publication Date Title
JP7001474B2 (en) Non-immunogenic single domain antibody
JP6009431B2 (en) Affinity purification method
US20130085072A1 (en) Recombinant renewable polyclonal antibodies
CN108341870B (en) anti-BSA nano antibody, production method and application thereof
US20190248922A1 (en) Affinity ligands and methods relating thereto
JP2023109916A (en) Mrna display antibody libraries and methods
JP6985572B2 (en) Single-chain antibody screening method and single-chain antibody
Finlay et al. Phage display: a powerful technology for the generation of high-specificity affinity reagents from alternative immune sources
JP7062134B2 (en) Magnetic-based bio-panning method using the attachment of magnetic beads on cells
CN113677702A (en) Antibodies against liraglutide and uses thereof
JP2019502705A (en) Method for generating antibody naive library, said library and use thereof
WO2023051972A1 (en) Method for the generation and selection of a producer cell
CN116120440A (en) Monoclonal antibody for resisting SARS-CoV-2 spike protein S1 and application thereof
TW202028239A (en) Antibodies against soluble bcma
JP6245688B2 (en) IgY-specific binding peptide and method for purifying IgY thereby
CN111518210B (en) Fully human monoclonal antibody for specifically recognizing FAIM3 receptor
CN108003238B (en) Fully human monoclonal antibody or antibody fragment capable of specifically recognizing CTLA-4, and method and application thereof
CN111201239A (en) Methods and compositions for developing antibodies specific for epitope post-translational modification states
CN113999306B (en) Method for obtaining antibody for recognizing space conformation epitope
CN116987194B (en) Anti-idiotype nano antibody of mimic epitope peptide of human ST2 antigen and application thereof
EP3521307B1 (en) Method for acquiring antibodies
US10793619B2 (en) Preparation and selection of cells for producing bispecific antibodies
Thompson et al. Immunoaffinity Chromatography: Advantages and Limitations
Marano Optimization of a pipeline for the development of recombinant monoclonal antibodies for diagnostics
CN117836308A (en) Means and methods for selecting specific binding agents

Legal Events

Date Code Title Description
AS Assignment

Owner name: BIO-RAD LABORATORIES, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:KNAPPIK, ACHIM;PASCHEN, STEFAN;REEL/FRAME:048794/0949

Effective date: 20160704

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

Free format text: NON FINAL ACTION MAILED

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

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

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

Free format text: FINAL REJECTION MAILED

STCB Information on status: application discontinuation

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