US20100267933A1 - Purification of proteins - Google Patents

Purification of proteins Download PDF

Info

Publication number
US20100267933A1
US20100267933A1 US12/448,004 US44800407A US2010267933A1 US 20100267933 A1 US20100267933 A1 US 20100267933A1 US 44800407 A US44800407 A US 44800407A US 2010267933 A1 US2010267933 A1 US 2010267933A1
Authority
US
United States
Prior art keywords
polymers
mixture
biomolecule
polymer
antibody
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
US12/448,004
Inventor
Moya Wilson
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.)
EMD Millipore Corp
Original Assignee
Individual
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 Individual filed Critical Individual
Priority to US12/448,004 priority Critical patent/US20100267933A1/en
Assigned to MILLIPORE CORPORATION reassignment MILLIPORE CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MOYA, WILSON
Publication of US20100267933A1 publication Critical patent/US20100267933A1/en
Assigned to EMD MILLIPORE CORPORATION reassignment EMD MILLIPORE CORPORATION CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: MILLIPORE CORPORATION
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/14Extraction; Separation; Purification
    • C07K1/30Extraction; Separation; Purification by precipitation
    • C07K1/32Extraction; Separation; Purification by precipitation as complexes
    • 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
    • C07K1/30Extraction; Separation; Purification by precipitation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2800/00Properties of cosmetic compositions or active ingredients thereof or formulation aids used therein and process related aspects
    • A61K2800/40Chemical, physico-chemical or functional or structural properties of particular ingredients
    • A61K2800/54Polymers characterized by specific structures/properties
    • A61K2800/542Polymers characterized by specific structures/properties characterized by the charge
    • A61K2800/5426Polymers characterized by specific structures/properties characterized by the charge cationic
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/14Extraction; Separation; Purification
    • C07K1/36Extraction; Separation; Purification by a combination of two or more processes of different types

Definitions

  • the present invention relates to the purification of biomolecules. More particularly, it relates to the purification of biomolecules such as proteins, polypeptides, antibodies and the like, by a solubilized polymer to remove impurities from a solution/suspension by a controlled precipitation mechanism.
  • the general process for the manufacture of biomolecules, such as proteins, particularly recombinant proteins typically involves two main steps: (1) the expression of the protein in a host cell, followed by (2) the purification of the protein.
  • the first step involves growing the desired host cell in a bioreactor to effect the expression of the protein.
  • cell lines used for this purpose include Chinese hamster ovary (CHO) cells, myeloma (NSO) cells, bacterial cells such as e - coli and insect cells.
  • CHO Chinese hamster ovary
  • NSO myeloma
  • bacterial cells such as e - coli and insect cells.
  • Suspended particulates such as cells, cell fragments, lipids and other insoluble matter are typically removed from the protein-containing fluid by filtration or centrifugation, resulting in a clarified fluid containing the protein of interest in solution as well as other soluble impurities.
  • the second step involves the purification of the harvested protein to remove impurities which are inherent to the process.
  • impurities include host cell proteins (HCP, proteins other than the desired or targeted protein), nucleic acids, endotoxins, viruses, protein variants and protein aggregates.
  • This purification typically involves several chromatography steps, which can include affinity, ion exchange hydrophobic interaction, etc.
  • One example of chromatography process train for the purification of proteins involves protein-A affinity, followed by cation exchange, followed by anion exchange.
  • the protein-A column captures the protein of interest or target protein by an affinity mechanism while the bulk of the impurities pass through the column to be discarded.
  • the protein then is recovered by elution from the column. Since most of the proteins of interest have isoelectric points (PI) in the basic range (8-9) and therefore being positively charged under normal processing conditions (pH below the PI of the protein), they are bound to the cation exchange resin in the second column. Other positively charged impurities are also bound to this resin.
  • PI isoelectric points
  • the protein of interest is then recovered by elution from this column under conditions (pH, salt concentration) in which the protein elutes while the impurities remain bound to the resin.
  • the anion exchange column is typically operated in a flow through mode, such that any negatively charged impurities are bound to the resin while the positively charged protein of interest is recovered in the flow through stream. This process results in a highly purified and concentrated protein solution.
  • a flocculation technique In this technique, a soluble polyelectrolyte is added to a clarified or unclarified cell culture broth to capture the impurities thereby forming a flocculant, which is allowed to settle and can be subsequently removed from the protein solution.
  • the present invention relates to a selectively soluble polymer capable of binding to one or more constituents in a biological material containing stream and the methods of using such a material to purify a biomolecule from such a stream.
  • polymer shall mean a polymer capable of binding to one or more constituents in a biological material containing stream unless otherwise stated.
  • biomolecule selected biomolecule, target biomolecule or molecule, target protein, biomolecule or protein of interest, or similar terms all refer to products of a biomolecule manufacturing process.
  • the polymer is soluble in an aqueous based solvent under a certain set of process conditions such as pH or temperature or salt concentration or the like and is rendered insoluble and precipitates out of solution upon a change in conditions (temperature, pH, salt or the like). While in its solubilized state, the polymer is capable of binding to a selected entity within the stream such as impurities (cells, cell fragments, lipids, DNA, RNA, host cell protein, endotoxins, virus, etc) in a cell broth and remains capable of binding to that entity even after the polymer is precipitated out of solution. The precipitate can then be easily removed, such as by being filtered out from the remainder of the stream and the desired biomolecule is recovered and further processed.
  • a selected entity within the stream such as impurities (cells, cell fragments, lipids, DNA, RNA, host cell protein, endotoxins, virus, etc) in a cell broth and remains capable of binding to that entity even after the polymer is precipitated out of solution.
  • the precipitate can then be
  • the polymer being bound to one or more impurities can either be disposed of or the one or more impurities can be eluted from the polymer and the polymer is then cleaned or sanitized and reused if desired. It can also be washed to ensure that any biomolecules of interest have been recovered for further use or processing.
  • polymers such as poly(N-vinyl caprolactam), poly(N-acryloylpiperidine), poly(N-vinylisobutyramide), poly (N-substituted acrylamide) including [poly(N-isopropylacrylamide), poly(N,N′-diethylacrylamide), and poly(N-acryloyl-N′-alkylpiperazine)], hydroxyalkylcellulose, copolymers of acrylic acid and methacrylic acid or methacrylic acid and methyl methacrylate, polymers and copolymers of 2 or 4-vinylpyridine and chitosan to selectively remove one or more impurities from a stream containing impurities along with a desired biomolecule.
  • polymers such as poly(N-vinyl caprolactam), poly(N-acryloylpiperidine), poly(N-vinylisobutyramide), poly (N-substituted acrylamide) including [poly(N-iso
  • the one or more entities are impurities in the mixture selected from host cell protein, cells, cell fragments, nucleic acids and endotoxins.
  • viruses which are either removed or rendered inactive by the polymer process.
  • viruses may be removed by the precipitated polymer or they may be rendered inactive by the polymer itself or by the conditions under which the polymer is dissolved into the mixture or rendered insoluble from the mixture.
  • FIG. 1 shows a block diagram of a first process according to the present invention.
  • FIG. 2 shows a block diagram of a second process according to the present invention.
  • FIG. 3 shows a block diagram of a third process according to the present invention.
  • FIG. 4 shows a block diagram of a fourth process according to the present invention.
  • FIG. 5 shows a block diagram of a fifth process according to the present invention.
  • FIGS. 6A and B show gel electrophoresis data of Example 11 according to the present invention.
  • FIG. 7 shows SDS-PAGE data of Example 11 according to the present invention.
  • the invention is to use a liquid phase or solubilized polymer that has a capability, such as affinity or charge or hydrophobicity and the like, to remove undesirable soluble and suspended impurities from a fluid containing a desirable biomolecule of interest.
  • Preferred polymers have an affinity or electrostatic ability. The biomolecule of interest is then recovered and further processed as desired or required.
  • the idea relates to the process of using one or more polymers soluble in a liquid phase to remove impurities from a solution/suspension by a precipitation mechanism and which polymer can also be removed, if present, in any excess, by the same mechanism.
  • this idea can best be described in the context of protein purification although it can be used to purify any solute from complex mixtures as long as the mechanism of removal applies to the specific solute of interest.
  • the one or more polymers can be used in excess unlike flocculants and can be recovered essentially completely from the mixture by the precipitation action. This allows one to operate the purification step with greater windows of use and without having to calculate the precise amount of material that needs to be used.
  • the present concept is based on the fact that certain polymers undergo changes in properties as a result of changes in the environment in which they are in.
  • the most common polymer property to change as a result of a stimulus is solubility and the most common stimuli are temperature salt concentration and pH.
  • a polymer may remain in solution as long as the pH, salt or temperature is maintained within a certain range but it will precipitate out of solution as soon as the pH, salt or temperature is raised or lowered outside of said range.
  • Certain polymers such as poly (N-isopropylacrylamide), agarose, polyethylene oxide, etc. are examples of polymers that exhibit solubility changes as a result of changes in temperature.
  • Other polymers such as certain catonic and anionic polyelectrolytes, especially poly(4-vinylpyridine), poly(2-vinylpyridine), copolymers of 4-vinyl pyridine or 2-vinyl pyridine with other monomers such as styrene, butyl methacrylate, etc., chitosan and copolymers of acrylic acid or methacrylic acid with other monomers such as methyl methacrylate are examples of polymers that exhibit changes in solubility as a result of changes in pH and/or salt concentration.
  • the precise mechanism is not currently known. It may be that the polymer(s) interact with the entity or entities while in a soluble state and continue to bind to them upon precipitation. It may also be that the polymer and/or entity(s) bind to one another as the polymer is in the process of precipitating. It may be another mechanism as yet unknown to the inventor at this time. The inventor does not wish to be bound to any particular theory of what mechanism is being used but that the invention encompasses any all such mechanisms and phenomena.
  • the process used can vary. However, the processes will generally involve having one or more conditions of the liquid or the mixture, such as a cell broth, at the correct pH, temperature, temperature and salt concentration or other condition used to cause the polymer(s) to become soluble and then adding the polymer(s) either directly or already solubilized in a carrier liquid, such as water, to the mixture.
  • a carrier liquid such as water
  • the mixture will be at the proper condition to allow the polymer(s) to be simply added to the mixture.
  • the mixture may need to be conditioned or modified to be at the desired condition.
  • This modification or conditioning can be by modifying the mixture first and then adding the polymer(s), by adding the polymer(s) to a carrier liquid that is conditioned to the desired state and simply adding it to the mixture such that the carrier liquid is sufficient to cause the mixture to thus reach that condition or to do both.
  • the mixture and the solubilized or soluble polymer(s) are then mixed to ensure the polymer(s) is solubilized, and that the entities of the mixture and the solubilized polymer(s) have sufficient and intimate contact with each other.
  • the conditions of the liquid in the mixture are then changed (pH, temperature, salt content, combinations thereof, etc) that causes the polymer(s) to become insoluble and precipitate out of the mixture as a solid while either still remaining bound to the one or more entities it contacted while soluble in the mixture or to bind to the entities as it precipitates and continue to bind to it thereafter.
  • the precipitate and remaining mixture are then separated such as by centrifugation or filtration or gravity and time with the liquid portion being decanted. Depending on what was bound to the precipitate, it is either disposed of (if it bound to impurities) or treated (such as by elution and or washing) one or more times to remove any residual impurities or contaminants and then sanitized for reuse.
  • One polymer or a blend of polymers may be used in the present invention and it is meant to cover both embodiments whenever the term polymer, polymer(s) or one or more polymers is used hereafter.
  • the polymer may be added directly to the conditioned mixture.
  • it can be added to a carrier liquid in which it is soluble and which carrier preferably is also compatible with the mixture.
  • a carrier liquid is water, water adjusted to the correct pH by the addition of acids or bases, another is an aqueous based solution such as saline, buffered solutions or blends of water with an organic solvent such as alcohol.
  • the selection of carrier liquid is dependent on the mixture to which it is added as to what is preferred and tolerated.
  • the polymer is added to the carrier liquid that either has already been conditioned (such as pH adjusted or heated to a desired temperature or heated to a desired temperature with the addition of one or more salts or cooled to the desired temperature with or without one or more salts) or it can be added and then the carrier is conditioned to cause the solubilizing of the polymer in the carrier.
  • the carrier/ soluble polymer blend is then added to the. mixture.
  • the mixture may be contained in a mixing vessel such as a tapered bottom metal (preferably stainless steel more preferably 304 or 316 L stainless steel) or glass vat or tank.
  • a mixing vessel such as a tapered bottom metal (preferably stainless steel more preferably 304 or 316 L stainless steel) or glass vat or tank.
  • a cell culture or microbial or yeast culture it may be the bioreactor or fermentor in which it has been grown. It may also be a disposable bioreactor or fermentor or a disposable mixing bag such as a plastic bag as is available from Millipore Corporation of Billerica, Mass.
  • the mixture and polymer are brought into intimate contact through a mixing action that may be done by a magnetic stirred bar, a magnetic driven mixer such as a NovAseptic® mixer available from Millipore Corporation of Billerica Mass., a Lightning-type mixer., a recirculation pump, or a rocking motion closed mixing bag or bioreactor or fermentor, such as is shown in US 2005/0063259A1 or an airlift type of mixer or reactor in which rising bubbles in the liquid cause a circulatory pattern to be formed.
  • a magnetic stirred bar such as a NovAseptic® mixer available from Millipore Corporation of Billerica Mass., a Lightning-type mixer., a recirculation pump, or a rocking motion closed mixing bag or bioreactor or fermentor, such as is shown in US 2005/0063259A1 or an airlift type of mixer or reactor in which rising bubbles in the liquid cause a circulatory pattern to be formed.
  • the mixture and polymer can be in separate containers and mixed in line in a static blender.
  • the blend can either then go to a container or to a centrifuge or a filter where the polymer is caused to precipitate and the precipitated polymer and its bound one or more entities is separated from the remainder of the mixture. Then at least the remainder of the mixture is further processed.
  • the mixture and polymer are blended together in the container holding the mixture and further mixed in line in a static blender.
  • the blend is then treated to cause precipitation of the polymer and its bound entity(s). It can either then go to a container or to a centrifuge or to a filter where the precipitated polymer and its bound one or more entities is separated from the remainder of the mixture. Then at least the filtrate is further processed.
  • centrifugation one can easily and quickly separate the precipitated polymer from the remainder of the liquid mixture. After centrifugation, the supernatant, generally the remainder of the mixture is drawn off. Either the drawn off mixture or the precipitated polymer or both if desired is further processed.
  • Simple settling of the precipitated solids and decantation of the supernatant fluid may also be used if desired.
  • Filtration can be accomplished in a variety of manners. Depending upon the size of the polymer as it is precipitated; one may use one or more filters of varying sizes or asymmetries.
  • type and size of filter will depend on the volume of precipitate to be captured and whether one wishes to further process the precipitated polymer or just the remainder of the mixture,
  • Membrane based filters preferably microporous membranes can be used in the present invention.
  • Such filters are generally polymeric in nature and can be made from polymers such as but not limited to olefins such as polyethylene including ultrahigh molecular weight polyethylene, polypropylene, EVA copolymers and alpha olefins, metallocene olefinic polymers, PFA, MFA, FIFE, polycarbonates, vinyl copolymers such as PVC, polyamides such as nylon, polyesters, cellulose, cellulose acetate, regenerated cellulose, cellulose composites, polysulfone, polyethersulfone, polyarylsulfone, polyphenylsulfone, polyacrylonitrile, polyvinylidene fluoride (PVDF), and blends thereof.
  • olefins such as polyethylene including ultrahigh molecular weight polyethylene, polypropylene, EVA copolymers and alpha olefins, metallocene
  • the membrane selected depends upon the application, desired filtration characteristics, particle type and size to be filtered and the flow desired.
  • Preferred membrane based filters include DURAPORE® PVDF membranes available from Millipore Corporation of Billerica Mass., MILLIPORE EXPRESS® and MILLIPORE EXPRESS® PLUS or SH_PES membranes available from Millipore Corporation of Billerica Mass.
  • the membrane may be hydrophilic or hydrophobic.
  • Preferred membranes are hydrophilic and are low in protein binding.
  • the membrane may be symmetric in pore size through out its depth such as DURAPORE® PVDF membranes available from Millipore Corporation of Billerica Mass., or it may be asymmetric in pore size through its thickness as with MILLIPORE EXPRESS® and MILLIPORE EXPRESS® PLUS or SH_PES membranes available from Millipore Corporation of Billerica Mass. It may contain a prefilter layer if desired, either as a separate upstream layer or as an integral upstream portion of the membrane itself.
  • the pore size of the membrane can vary depending upon the polymer and mixture selected. Generally, it has an average pore size of from about 0.05 micron to 5 microns, preferably from about 0.05 micron to about 1 micron, more preferably from about 0.05 to about 0.65 micron.
  • the membrane filter may run in a deadend or normal flow (NF) format or a tangential flow (TFF) format.
  • NF normal flow
  • TFF tangential flow
  • Depth filters such as the MILLISTAK+® depth filters, in either lenticular or POD format, or POLYGARD® wound filters available from Millipore Corporation of Billerica Mass. allows one to trap a large volume of precipitated polymer due to its asymmetric structure and large holding capacity. This can be useful when the polymer is designed to remove impurities and to leave the target or desired biomolecule in the liquid of the remaining mixture.
  • FIG. 1 shows a block diagram of a first process of the present invention.
  • the mixture is either conditioned to the correct parameter(s) to maintain the polymer of choice in solution or if the conditions of the mixture are already such that the polymer(s) become soluble in the mixture, no further conditioning may be required.
  • the polymer(s) may be added as a solid to an unconditioned mixture and then the mixture (containing the solid polymer(s)) may be conditioned to the correct parameters to dissolve the polymer(s) in the mixture.
  • the polymer(s) is then added to the mixture and mixed to cause it to go into solution and to make intimate contact with all the constituents of the mixture.
  • the mixture conditions are changed to cause the polymer(s) to precipitate out of solution while retaining one or more entities of the mixture with it.
  • the mixture and the precipitated polymer(s) are then separated from each other in the fourth step 8 .
  • the precipitate and remaining mixture may be separated by centrifugation or filtration.
  • FIG. 2 shows a block diagram of a second process of the present invention.
  • the mixture is either conditioned to the correct parameter(s) to maintain the polymer of choice in solution before, during or after the introduction of the polymer or it is already at the desired condition.
  • the polymer is added to a carrier liquid under conditions that allow it to go into solution.
  • the polymer in its carrier is then added to the mixture and mixed to make intimate contact with all the constituents of the mixture.
  • the mixture conditions are changed to cause the polymer to precipitate out of solution carrying one or more entities of the mixture with it.
  • the mixture and the precipitated polymer are then separated from each other in the fifth step 18 . As discussed above the precipitate and remaining mixture may be separated by centrifugation or filtration in a sixth step 20 and the target or desired biomolecule is recovered.
  • FIG. 3 shows a block diagram of a third process of the present invention.
  • the mixture is either conditioned to the correct parameter(s) to maintain the polymer of choice in solution before, during or after introduction of the polymer or it is already at the desired condition.
  • the polymer is added to a carrier liquid under conditions that allow it to go into solution.
  • the polymer in its carrier is then added to the mixture through one or more static mixers to make intimate contact with all the constituents of the mixture.
  • the mixture conditions are changed to cause the polymer to precipitate out of solution.
  • the mixture and the precipitated polymer are then separated from each other in the fifth step 38 . As discussed above the precipitate and remaining mixture may be separated by centrifugation or filtration in a sixth step 40 and the target or desired biomolecule is recovered.
  • FIG. 4 shows a block diagram of the first process of the present invention as shown in FIG. 1 with an additional step.
  • the mixture is conditioned to the correct parameter(s), or if the conditions of the mixture are already such that the polymer becomes soluble in the mixture, no further conditioning may be required to maintain the polymer of choice in solution.
  • the polymer may be added as a solid to an unconditioned mixture and then the mixture (containing the solid polymer) may be conditioned to the correct parameters to dissolve the polymer in the mixture and maintain the polymer of choice in solution.
  • the polymer is then added to the mixture and mixed to cause it to go into solution and to make intimate contact with all the constituents of the mixture.
  • the mixture conditions are changed to cause the polymer to precipitate out of solution.
  • the mixture and the precipitated polymer are then separated from each other in the fourth step 8 .
  • the precipitate is separated from the remaining mixture by filtration.
  • the filtration maybe by either normal flow or tangential flow filtration with any of the membranes or depth filters described above.
  • FIG. 5 shows a block diagram of the first process of the present invention as shown in FIG. 1 with an additional step.
  • the mixture is conditioned to the correct parameter(s) or if the conditions of the mixture are already such that the polymer becomes soluble in the mixture, no further conditioning may be required to maintain the polymer of choice in solution.
  • the polymer may be added as a solid to an unconditioned mixture and then the mixture (containing the solid polymer) may be conditioned to the correct parameters to dissolve the polymer in the mixture to maintain the polymer of choice in solution.
  • the polymer is then added to the mixture and mixed to cause it to go into solution and to make intimate contact with all the constituents of the mixture.
  • the mixture conditions are changed to cause the polymer to precipitate out of solution.
  • the mixture and the precipitated polymer are then separated from each other in the fourth step 8 .
  • the precipitate is separated from the remaining mixture by centrifugation.
  • the change in stimuli may be gradual or it may be done substantially instantaneous.
  • a change in pH can be done by slowly adding a pH changing material to the liquid to change the pH slowly over a span of several minutes or even hours.
  • a suitable amount of pH changing material can be added to the liquid at one time to cause the change in pH to occur more rapidly. More control has been found in general with incremental changes rather than immediate changes for most processes.
  • Examples of cationic polyelectrolytes that exhibit this selective solubility behavior include chitosan, polyvinylpyridines (PVPs) and copolymers of PVPs such as poly(2 vinylpyridine) (P2VP) or poly(4 vinylpyridine) (P4VP), polyvinylpyridine-co-styrene (PVP-S), polyvinylpyridine-co-butyl methacrylate (PVP-BMA) as well as other primary, secondary and tertiary amine-containing polymers. These polymers are soluble at a pH lower than about 6-7 and are insoluble at a pH greater than about 5-7.
  • a solution of said cationic polyelectrolyte can be added to a fluid containing a biomolecule of interest, such as a protein in the presence of other impurities.
  • This fluid can be for example a cell culture fluid.
  • the polymer is added to the fluid either as a solution in a carrier liquid at a pH of about 4.5 or as a solid particulate in which the fluid is either modified to a pH of about 4.5 either before, during or after the introduction of the polymer to it (as further described below) so that the polymer binds all the negatively charged impurities, such as cells, cell fragments, nucleic acids, viruses, host cell proteins, pyrogens and endotoxins.
  • the biomolecule of interest does not interact with the polymer given its positive net charge due to its basic PI.
  • the pH is then raised to 5-7 or more if desired and the polymer precipitates out of solution, carrying with it all the impurities as well as any excess polymer.
  • the precipitate can then be easily removed by centrifugation or filtration, resulting in a “purified” biomolecule containing solution.
  • anionic polyelectrolytes that exhibit this solubility behavior is a class of copolymers of acrylic acid and methyl methacrylate or methacrylic acid and methyl methacrylate. These polymers are soluble at a pH greater than about 4-7 and insoluble at a pH lower than about 4-7. These polymers can also be used to purify proteins from complex mixtures in a bind and elute mode. For instance, a solution of these polymers can be added to a fluid containing a protein of interest in the presence of other impurities wherein the pH of the fluid is at or above about 4-7. Under these conditions, the negatively charged polymer binds the positively charged protein of interest (basic PI) while it repels the negatively charged impurities.
  • basic PI positively charged protein of interest
  • the pH of the fluid is then lowered below about 4-7 to effect precipitation of the polymer-protein complex and any excess polymer.
  • the precipitate can then be washed to remove any soluble or loosely bound impurities while the pH is kept below about 4-7.
  • the protein can be subsequently eluted from the polymer with an elution buffer at high salt concentrations and a pH below about 4-7 to recover the purified protein.
  • a temperature sensitive polymer is agarose, which is often used in chromatography, hydroxyalkylcelluloses such as hydroxypropylcellulose; polymers and copolymers containing N-isopropylacrylamide monomer, polyethylene oxide, etc. The temperature can then be reduced or raised to cause the polymer to gel and/or precipitate out of solution.
  • these polymers are generally insoluble at room temperature and are soluble in water or other solvents at temperatures generally between about 80 to 120° C. They can be simply heated to cause them to dissolve, added to the mixture and then cooled to cause them to precipitate. In other cases, such as with polymers and copolymers containing N-isopropylacrylamide monomer, the polymer is soluble at a temperature below about 30 to 35° C. and will precipitate out of solution when the temperature is raised above this range.
  • gel-inhibiting agents such as various salts can depress the solubility temperature to lower temperatures, often to room temperature if desired.
  • Salts that can be used include lithium chloride and zinc chloride.
  • Bases such as sodium hydroxide or lithium hydroxide can also be used to depress the gelling temperature or to eliminate it altogether.
  • the melting point for agarose is about 92 degrees
  • the gelling temperature is about 43 degrees.
  • This gelling temperature can be manipulated by the modification of the agarose molecule as described above or by the addition of the above salts or by a combination thereof.
  • a cationic ligand can be attached to agarose in an amount such that the gelling temperature of the modified polymer is about 20° C. degrees with or without the addition of the above salts.
  • the modified agarose is added, in solution at a temperature about 25° C. degrees, to the mixture (also at a temperature of about 25° C. degrees) to bind the constituents and then the temperature of the mixture is lowered to below 20° C. degrees thereby gelling the modified agarose with the constituents.
  • polymers such as polyvinylpyridine, polyvinylpyridene-co-styrene and the like
  • One such method is to place the polymer as purchased in an oven, preferably with an inert or low oxygen gas atmosphere such as by purging the oven several times during the process with argon or nitrogen, and maintain it at an elevated temperature (generally between 100 and 200° C., preferably about 120° C.) and under a vacuum so as to drive off all monomer present (generally about 24 hours).
  • polymers such as polyvinylpyridine, polyvinylpyridene-co-sytrene and the like
  • higher molecular weight polymers 200,000 molecular weight or higher
  • precipitation by itself may be slow or incomplete.
  • Typical polymer concentrations in the carrier solvent are between 1-20% by weight depending on the viscosity of the solution. It is preferred that the concentration be as high as possible to minimize dilution of the feedstock. Practically, polymer solutions in the 10-20% are preferred to achieve a good balance between viscosity and dilution of the feedstock.
  • the final concentration of the polymer in the feedstock may depend on the amount of impurities in the feedstock but it is typically between 0.01% to 2% by weight and more specifically between 0.05% and 0.1%.
  • the recovered biomolecule may then undergo one or more additional processing steps depending on whether it is contained within the liquid of the mixture or is bound to the precipitated polymer.
  • a method of sequential precipitation may be used to recover the biomolecule of interest.
  • a first precipitation as described above would be used to remove impurities and the precipitated polymer/impurities mass would be separated from the target biomolecule containing solution.
  • the solution would then be mixed with a stimuli responsive polymer containing a ligand capable of binding to the biomolecule of interest at a solution condition at which the polymer is soluble.
  • the solution conditions would be changed so as to precipitate the polymer and bound biomolecule.
  • the polymer/biomolecule would then be separated as previously described, the biomolecule eluted or otherwise separated from the polymer, and the recovered biomolecule further processed as needed.
  • the biomolecule of interest As the biomolecule of interest is in the liquid, it may, if needed or desired undergo one or more known process steps including but not limited to chromatography steps such as ion exchange, hydrophobic interaction or affinity chromatography, various filtration steps such as high performance tangential flow filtration (HPTFF), viral removal/inactivation steps, final filtration and the like.
  • chromatography steps such as ion exchange, hydrophobic interaction or affinity chromatography
  • various filtration steps such as high performance tangential flow filtration (HPTFF), viral removal/inactivation steps, final filtration and the like.
  • HPTFF high performance tangential flow filtration
  • the biomolecule of interest present in the liquid may be used as is without the need for further purification steps.
  • the chromatography may be column based using solid bead media or monoliths or through a membrane absorber or chromatography device.
  • the step if desired can be a classic bind/elute or a flow through mode of chromatography.
  • biomolecule of interest may undergo further purification without the need for chromatography steps such as through the use of high performance tangential flow filtration using one or more charged membranes.
  • no further purification is required.
  • a further variation uses an affinity step to bind and then elute the desired biomolecule.
  • Affinity ligands such as Protein A either on a solid matrix such as a bead or membrane may be used.
  • the current process simply replaces a clarification step and prefilter step in a normal biological product process train.
  • it replaces clarification, prefiltration and at least one chromatography step by directly purifying the biomolecule of interest from the starting materials.
  • it replaces a cell harvest or biomolecule collection step by being added directly to the bioreactor or fermentor. This also eliminates the need for clarification, prefiltration and potentially at least one chromatography step by directly purifying the biomolecule of interest from the starting materials.
  • the protein thus recovered may be formulated in a pharmaceutically acceptable carrier and is used for various diagnostic, therapeutic or other uses known for such molecules.
  • the mixture that is the starting material of the process will vary depending upon the cell line in which it was grown as well as the conditions under which it is grown and harvested.
  • the cells express the molecule outside of the cell wall into the media.
  • some cells during grow and harvesting may rupture due to shear or other handling conditions or die and lyse, spilling their contents into the mixture.
  • the biomolecule is often kept with the cellular wall or it may actually be part of the cellular wall (Protein A). In these systems the cell walls need to be disrupted or lysed in order to recover the biomolecule of interest.
  • the target molecule to be purified can be any biomolecule, preferably a protein, in particular, recombinant protein produced in any host cell, including but not limited to, Chinese hamster ovary (CHO) cells, Per.C6® cell lines available from Crucell of the Netherlands, myeloma cells such as NSO cells, other animal cells such as mouse cells, insect cells, or microbial cells such as E. coli or yeast. Additionally, the mixture may be a fluid derived from an animal modified to produce a transgenic fluid such as milk or blood that contains the biomolecule of interest.
  • Optimal target proteins are antibodies, immunoadhesins and other antibody-like molecules, such as fusion proteins including a C H 2/C H 3 region.
  • this product and process can be used for purification of recombinant humanized monoclonal antibodies such as (RhuMAb) from a conditioned harvested cell culture fluid (HCCF) grown in Chinese hamster ovary (CHO) cells expressing RhuMAb.
  • HCCF conditioned harvested cell culture fluid
  • CHO Chinese hamster ovary
  • Antibodies within the scope of the present invention include, but are not limited to: anti-HER2 antibodies including Trastuzumab (HERCEPTIN®) (Carter et al., Proc. Natl. Acad. Sci. USA, 89:4285-4289 (1992), U.S. Pat. No. 5,725,856); anti-CD20 antibodies such as chimeric anti-CD20 “C2B8” as in U.S. Pat. No. 5,736,137 (RITUXAN.®), a chimeric or humanized variant of the 2H7 antibody as in U.S. Pat. No.
  • anti-VEGF antibodies including humanized and/or affinity matured anti-VEGF antibodies such as the humanized anti-VEGF antibody huA4.6.1 AVASTIN®. (Kim et al., Growth Factors, 7:53-64 (1992), International Publication No. WO 96/30046, and WO 98/45331, published Oct.
  • anti-PSCA antibodies WO01/40309
  • anti-CD40 antibodies including S2C6 and humanized variants thereof (WO00/75348)
  • anti-CD11a U.S. Pat. No. 5,622,700, WO 98/23761, Steppe et al., Transplant Intl. 4:3-7 (1991), and Hourmant et al., Transplantation 58:377-380 (1994)
  • anti-IgE Presta et al., J Immunol. 151:2623-2632 (1993), and International Publication No. WO 95/19181
  • anti-CD18 U.S. Pat. No. 5,622,700, issued Apr. 22, 1997, or as in WO 97/26912, published Jul.
  • anti-IgE including E25, E26 and E27; U.S. Pat. No. 5,714,338, issued Feb. 3, 1998 or U.S. Pat. No. 5,091,313, issued Feb. 25, 1992, WO 93/04173 published Mar. 4, 1993, or International Application No. PCT/US98/13410 filed Jun. 30, 1998, U.S. Pat. No. 5,714,338); anti-Apo-2 receptor antibody (WO 98/51793 published Nov. 19, 1998); anti-TNF- ⁇ antibodies including cA2 (REMICADE®), CDP571 and MAK-195 (See, U.S. Pat. No. 5,672,347 issued Sep. 30, 1997, Lorenz et al. J. Immunol.
  • anti-Tissue Factor TF
  • TF tissue Factor
  • anti-human ⁇ 4 ⁇ 7 integrin WO 98/06248 published Feb. 19, 1998
  • anti-EGFR chimerized or humanized 225 antibody as in WO 96/40210 published Dec. 19, 1996
  • anti-CD3 antibodies such as OKT3 (U.S. Pat. No. 4,515,893 issued May 7, 1985); anti-CD25 or anti-tac antibodies such as CHI-621 (SIMULECT®) and (ZENAPAX®) (See U.S. Pat.
  • anti-CD4 antibodies such as the cM-7412 antibody (Choy et al. Arthritis Rheum 39(1):52-56 (1996)); anti-CD52 antibodies such as CAMPATH-1H (Riechmann et al. Nature 332:323-337 (1988)); anti-Fc receptor antibodies such as the M22 antibody directed against Fc ⁇ RI as in Graziano et al. J. Immunol. 155(10):4996-5002 (1995); anti-carcinoembryonic antigen (CEA) antibodies such as hMN-14 (Sharkey et al. Cancer Res.
  • CEA anti-carcinoembryonic antigen
  • anti-CD33 antibodies such as Hu M195 (Jurcic et al. Cancer Res 55(23 Suppl):5908s-5910s (1995) and CMA-676 or CDP771; anti-CD22 antibodies such as LL2 or LymphoCide (Juweid et al.
  • anti-EpCAM antibodies such as 17-1A (PANOREX®); anti-GpIIb/IIIa antibodies such as abciximab or c7E3 Fab (REOPRO®); anti-RSV antibodies such as MEDI-493 (SYNAGIS®); anti-CMV antibodies such as PROTOVIR®; anti-HIV antibodies such as PRO542; anti-hepatitis antibodies such as the anti-Hep B antibody OSTAVIR®; anti-CA 125 antibody OvaRex; anti-idiotypic GD3 epitope antibody BEC2; anti- ⁇ v ⁇ 3 antibody VITAXIN®.; anti-human renal cell carcinoma antibody such as ch-G250; ING-1; anti-human 17-1A antibody (3622W94); anti-human colorectal tumor antibody (A33); anti-human melanoma antibody R24 directed against GD3 ganglioside; anti-human squamous-
  • the antibody herein is directed against an antigen of interest.
  • the antigen is a biologically important polypeptide and administration of the antibody to a mammal suffering from a disease or disorder can result in a therapeutic benefit in that mammal.
  • antibodies directed against non-polypeptide antigens are also contemplated.
  • the antigen is a polypeptide, it may be a transmembrane molecule (e.g. receptor) or ligand such as a growth factor.
  • Exemplary antigens include those proteins described in section (3) below.
  • Exemplary molecular targets for antibodies encompassed by the present invention include CD proteins such as CD3, CD4, CD8, CD19, CD20, CD22, CD34, CD40; members of the ErbB receptor family such as the EGF receptor, HER2, HER3 or HER4 receptor; cell adhesion molecules such as LFA-1, Mac1, p150,95, VLA-4, ICAM-1, VCAM and ⁇ v/ ⁇ 3 integrin including either ⁇ or ⁇ subunits thereof (e.g.
  • anti-CD11a, anti-CD18 or anti-CD11b antibodies growth factors such as VEGF; IgE; blood group antigens; flk2/flt3 receptor; obesity (OB) receptor; mpl receptor; CTLA-4; protein C, or any of the other antigens mentioned herein.
  • Antigens to which the antibodies listed above bind are specifically included within the scope herein.
  • Soluble antigens or fragments thereof, optionally conjugated to other molecules, can be used as immunogens for generating antibodies.
  • immunogens for transmembrane molecules, such as receptors, fragments of these (e.g. the extracellular domain of a receptor) can be used as the immunogen.
  • transmembrane molecules such as receptors
  • fragments of these e.g. the extracellular domain of a receptor
  • cells expressing the transmembrane molecule can be used as the immunogen.
  • Such cells can be derived from a natural source (e.g. cancer cell lines) or may be cells which have been transformed by recombinant techniques to express the transmembrane molecule.
  • Polyclonal antibodies can also be purified in the present invention.
  • Polyclonal antibodies are preferably raised in animals by multiple subcutaneous (sc) or intraperitoneal (ip) injections of the relevant antigen and an adjuvant.
  • a protein that is immunogenic in the species to be immunized e.g., keyhole limpet hemocyanin, serum albumin, bovine thyroglobulin, or soybean trypsin inhibitor using a bifunctional or derivatizing agent, for example, maleimidobenzoyl sulfosuccinimide ester (conjugation through cysteine residues), N-hydroxysuccinimide (through lysine residues), glutaraldehyde, succinic anhydride, SOCl 2 , or R 1 N C ⁇ NR, where R and R 1 are different alkyl groups.
  • Animals are immunized against the antigen, immunogenic conjugates, or derivatives by combining, e.g., 100 ⁇ g or 5 ⁇ g of the protein or conjugate (for rabbits or mice, respectively) with 3 volumes of Freund's complete adjuvant and injecting the solution intradermally at multiple sites.
  • the animals are boosted with 1 ⁇ 5 to 1/10 the original amount of antigen or conjugate in Freund's complete adjuvant by subcutaneous injection at multiple sites.
  • Seven to 14 days later the animals are bled and the serum is assayed for antibody titer. Animals are boosted until the titer plateaus.
  • the animal is boosted with the conjugate of the same antigen, but conjugated to a different protein and/or through a different cross-linking reagent.
  • Conjugates also can be made in recombinant cell culture as protein fusions.
  • aggregating agents such as alum are suitably used to enhance the immune response.
  • Monoclonal antibodies are of interest in the present invention and may be made using the hybridoma method first described by Kohler et al., Nature, 256:495 (1975), or may be made by recombinant DNA methods (U.S. Pat. No. 4,816,567).
  • a mouse or other appropriate host animal such as a hamster or macaque monkey
  • lymphocytes that produce or are capable of producing antibodies that will specifically bind to the protein used for immunization.
  • lymphocytes may be immunized in vitro. Lymphocytes then are fused with myeloma cells using a suitable fusing agent, such as polyethylene glycol, to form a hybridoma cell (Goding, Monoclonal Antibodies: Principles and Practice, pp. 59-103 (Academic Press, 1986)).
  • the hybridoma cells thus prepared are seeded and grown in a suitable culture medium that preferably contains one or more substances that inhibit the growth or survival of the unfused, parental myeloma cells.
  • a suitable culture medium that preferably contains one or more substances that inhibit the growth or survival of the unfused, parental myeloma cells.
  • the culture medium for the hybridomas typically will include hypoxanthine, aminopterin, and thymidine (HAT medium), which substances prevent the growth of HGPRT-deficient cells.
  • Preferred myeloma cells are those that fuse efficiently, support stable high-level production of antibody by the selected antibody-producing cells, and are sensitive to a medium such as HAT medium.
  • preferred myeloma cell lines are murine myeloma lines, such as those derived from MOPC-21 and MPC-11 mouse tumors available from the Salk Institute Cell Distribution Center, San Diego, Calif. USA, and SP-2 or X63-Ag8-653 cells available from the American Type Culture Collection, Rockville, Md. USA.
  • Human myeloma and mouse-human heteromyeloma cell lines also have been described for the production of human monoclonal antibodies (Kozbor, J. Immunol., 133:3001 (1984); Brodeur et al., Monoclonal Antibody Production Techniques and Applications, pp. 51-63 (Marcel Dekker, Inc., New York, 1987)).
  • Culture medium in which hybridoma cells are growing is assayed for production of monoclonal antibodies directed against the antigen.
  • the binding specificity of monoclonal antibodies produced by hybridoma cells is determined by immunoprecipitation or by an in vitro binding assay, such as radioimmunoassay (RIA) or enzyme-linked immunoabsorbent assay (ELISA).
  • RIA radioimmunoassay
  • ELISA enzyme-linked immunoabsorbent assay
  • the clones may be subcloned by limiting dilution procedures and grown by standard methods (Goding, Monoclonal Antibodies: Principles and Practice, pp. 59-103 (Academic Press, 1986)). Suitable culture media for this purpose include, for example, D-MEM or RPMI-1640 medium.
  • the hybridoma cells may be grown in vivo as ascites tumors in an animal.
  • the monoclonal antibodies secreted by the subclones are suitably separated from the culture medium, ascites fluid, or serum by conventional immunoglobulin purification procedures such as, for example, Pro-Sep® Protein A media available from Millipore Corporation of Billerica, Mass., hydroxyapatite chromatography, gel electrophoresis, dialysis, or affinity chromatography.
  • immunoglobulin purification procedures such as, for example, Pro-Sep® Protein A media available from Millipore Corporation of Billerica, Mass., hydroxyapatite chromatography, gel electrophoresis, dialysis, or affinity chromatography.
  • Protein A chromatography procedure described herein is used.
  • DNA encoding the monoclonal antibodies is readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of the monoclonal antibodies).
  • the hybridoma cells serve as a preferred source of such DNA.
  • the DNA may be placed into expression vectors, which are then transfected into host cells such as E. coli cells, simian COS cells, Chinese hamster ovary (CHO) cells, or myeloma cells that do not otherwise produce immunoglobulin protein, to obtain the synthesis of monoclonal antibodies in the recombinant host cells.
  • the DNA also may be modified, for example, by substituting the coding sequence for human heavy- and light-chain constant domains in place of the homologous murine sequences (U.S. Pat. No. 4,816,567; Morrison, et al., Proc. Natl. Acad. Sci. USA, 81:6851 (1984)), or by covalently joining to the immunoglobulin coding sequence all or part of the coding sequence for a non-immunoglobulin polypeptide.
  • non-immunoglobulin polypeptides are substituted for the constant domains of an antibody, or they are substituted for the variable domains of one antigen-combining site of an antibody to create a chimeric bivalent antibody comprising one antigen-combining site having specificity for an antigen and another antigen-combining site having specificity for a different antigen.
  • monoclonal antibodies can be isolated from antibody phage libraries generated using the techniques described in McCafferty et al., Nature, 348:552-554 (1990). Clackson et al., Nature, 352:624-628 (1991) and Marks et al., J. Mol. Biol., 222:581-597 (1991) describe the isolation of murine and human antibodies, respectively, using phage libraries.
  • a humanized antibody has one or more amino acid residues introduced into it from a source which is non-human. These non-human amino acid residues are often referred to as “import” residues, which are typically taken from an “import” variable domain. Humanization can be essentially performed following the method of Winter and co-workers (Jones et al., Nature, 321:522-525 (1986); Riechmann et al., Nature, 332:323-327 (1988); Verhoeyen et al., Science, 239:1534-1536 (1988)), by substituting rodent CDRs or CDR sequences for the corresponding sequences of a human antibody. Accordingly, such “humanized” antibodies are chimeric antibodies (U.S. Pat. No.
  • humanized antibodies are typically human antibodies in which some CDR residues and possibly some FR residues are substituted by residues from analogous sites in rodent antibodies.
  • variable domains both light and heavy
  • sequence of the variable domain of a rodent antibody is screened against the entire library of known human variable-domain sequences.
  • the human sequence which is closest to that of the rodent is then accepted as the human FR for the humanized antibody (Sims et al., J. Immunol., 151:2296 (1993)).
  • Another method uses a particular framework derived from the consensus sequence of all human antibodies of a particular subgroup of light or heavy chains. The same framework may be used for several different humanized antibodies (Carter et al., Proc. Natl. Acad. Sci. USA, 89:4285 (1992); Presta et al., J. Immunol., 151:2623 (1993)).
  • humanized antibodies are prepared by a process of analysis of the parental sequences and various conceptual humanized products using three-dimensional models of the parental and humanized sequences.
  • Three-dimensional immunoglobulin models are commonly available and are familiar to those skilled in the art.
  • Computer programs are available which illustrate and display probable three-dimensional conformational structures of selected candidate immunoglobulin sequences. Inspection of these displays permits analysis of the likely role of the residues in the functioning of the candidate immunoglobulin sequence, i.e., the analysis of residues that influence the ability of the candidate immunoglobulin to bind its antigen.
  • FR residues can be selected and combined from the recipient and import sequences so that the desired antibody characteristic, such as increased affinity for the target antigen(s), is achieved.
  • the CDR residues are directly and most substantially involved in influencing antigen binding.
  • transgenic animals e.g., mice
  • transgenic animals e.g., mice
  • J H antibody heavy-chain joining region
  • Human antibodies can also be derived from phage-display libraries (Hoogenboom et al., J. Mol. Biol., 227:381 (1991); Marks et al., J. Mol. Biol., 222:581-597 (1991); Vaughan et al. Nature Biotech 14:309 (1996)).
  • antibody fragments Traditionally, these fragments were derived via proteolytic digestion of intact antibodies (see, e.g., Morimoto et al., Journal of Biochemical and Biophysical Methods 24:107-117 (1992) and Brennan et al., Science, 229:81 (1985)). However, these fragments can now be produced directly by recombinant host cells. For example, the antibody fragments can be isolated from the antibody phage libraries discussed above. Alternatively, Fab′-SH fragments can be directly recovered from E. coli and chemically coupled to form F(ab′) 2 fragments (Carter et al., Bio/Technology 10:163-167 (1992)).
  • F(ab′) 2 fragments can be isolated directly from recombinant host cell culture.
  • Other techniques for the production of antibody fragments will be apparent to the skilled practitioner.
  • the antibody of choice is a single chain Fv fragment (scFv). See WO 93/16185.
  • Multispecific antibodies have binding specificities for at least two different antigens. While such molecules normally will only bind two antigens (i.e. bispecific antibodies, BsAbs), antibodies with additional specificities such as trispecific antibodies are encompassed by this expression when used herein.
  • bispecific antibodies are known in the art. Traditional production of full length bispecific antibodies is based on the coexpression of two immunoglobulin heavy chain-light chain pairs, where the two chains have different specificities (Millstein et al., Nature, 305:537-539 (1983)). Because of the random assortment of immunoglobulin heavy and light chains, these hybridomas (quadromas) produce a potential mixture of 10 different antibody molecules, of which only one has the correct bispecific structure. Purification of the correct molecule, which is usually done by affinity chromatography steps, is rather cumbersome, and the product yields are low. Similar procedures are disclosed in WO 93/08829, and in Traunecker et al., EMBO J., 10:3655-3659 (1991).
  • the interface between a pair of antibody molecules can be engineered to maximize the percentage of heterodimers which are recovered from recombinant cell culture.
  • the preferred interface comprises at least a part of the C H 3 domain of an antibody constant domain.
  • one or more small amino acid side chains from the interface of the first antibody molecule are replaced with larger side chains (e.g. tyrosine or tryptophan).
  • Compensatory “cavities” of identical or similar size to the large side chain(s) are created on the interface of the second antibody molecule by replacing large amino acid side chains with smaller ones (e.g. alanine or threonine). This provides a mechanism for increasing the yield of the heterodimer over other unwanted end-products such as homodimers.
  • Bispecific antibodies include cross-linked or “heteroconjugate” antibodies.
  • one of the antibodies in the heteroconjugate can be coupled to avidin, the other to biotin.
  • Such antibodies have, for example, been proposed to target immune system cells to unwanted cells (U.S. Pat. No. 4,676,980), and for treatment of HIV infection (WO 91/00360, WO 92/200373, and EP 03089).
  • Heteroconjugate antibodies may be made using any convenient cross-linking methods. Suitable cross-linking agents are well known in the art, and are disclosed in U.S. Pat. No. 4,676,980, along with a number of cross-linking techniques.
  • bispecific antibodies can be prepared using chemical linkage.
  • Brennan et al., Science, 229: 81 (1985) describe a procedure wherein intact antibodies are proteolytically cleaved to generate F(ab′) 2 fragments. These fragments are reduced in the presence of the dithiol complexing agent sodium arsenite to stabilize vicinal dithiols and prevent intermolecular disulfide formation.
  • the Fab′ fragments generated are then converted to thionitrobenzoate (TNB) derivatives.
  • One of the Fab′-TNB derivatives is then reconverted to the Fab′-thiol by reduction with mercaptoethylamine and is mixed with an equimolar amount of the other Fab'-TNB derivative to form the bispecific antibody.
  • the bispecific antibodies produced can be used as agents for the selective immobilization of enzymes.
  • bispecific antibodies have been produced using leucine zippers.
  • the leucine zipper peptides from the Fos and Jun proteins were linked to the Fab' portions of two different antibodies by gene fusion.
  • the antibody homodimers were reduced at the hinge region to form monomers and then re-oxidized to form the antibody heterodimers. This method can also be utilized for the production of antibody homodimers.
  • the fragments comprise a heavy-chain variable domain (V H ) connected to a light-chain variable domain (V L ) by a linker which is too short to allow pairing between the two domains on the same chain. Accordingly, the V H and V L domains of one fragment are forced to pair with the complementary V L and V H domains of another fragment, thereby forming two antigen-binding sites.
  • V H and V L domains of one fragment are forced to pair with the complementary V L and V H domains of another fragment, thereby forming two antigen-binding sites.
  • sFv single-chain Fv
  • the antibodies can be “linear antibodies” as described in Zapata et al. Protein Eng. 8(10):1057-1062 (1995). Briefly, these antibodies comprise a pair of tandem Fd segments (V H -C H 1-V H -C H 1) which form a pair of antigen binding regions. Linear antibodies can be bispecific or monospecific.
  • Antibodies with more than two valencies are contemplated.
  • trispecific antibodies can be prepared. Tutt et al. J. Immunol. 147: 60 (1991).
  • the simplest and most straightforward immunoadhesin design combines the binding domain(s) of the adhesin (e.g. the extracellular domain (ECD) of a receptor) with the hinge and Fc regions of an immunoglobulin heavy chain.
  • ECD extracellular domain
  • nucleic acid encoding the binding domain of the adhesin will be fused C-terminally to nucleic acid encoding the N-terminus of an immunoglobulin constant domain sequence, however N-terminal fusions are also possible.
  • the encoded chimeric polypeptide will retain at least functionally active hinge, C H 2 and C H 3 domains of the constant region of an immunoglobulin heavy chain. Fusions are also made to the C-terminus of the Fc portion of a constant domain, or immediately N-terminal to the C H 1 of the heavy chain or the corresponding region of the light chain.
  • the precise site at which the fusion is made is not critical; particular sites are well known and may be selected in order to optimize the biological activity, secretion, or binding characteristics of the immunoadhesin.
  • the adhesin sequence is fused to the N-terminus of the Fc domain of immunoglobulin G 1 (IgG 1 ). It is possible to fuse the entire heavy chain constant region to the adhesin sequence. However, more preferably, a sequence beginning in the hinge region just upstream of the papain cleavage site which defines IgG Fc chemically (i.e. residue 216, taking the first residue of heavy chain constant region to be 114), or analogous sites of other immunoglobulins is used in the fusion.
  • the adhesin amino acid sequence is fused to (a) the hinge region and C H 2 and C H 3 or (b) the C H 1, hinge, C H 2 and C H 3 domains, of an IgG heavy chain.
  • the immunoadhesins are assembled as multimers, and particularly as heterodimers or heterotetramers.
  • these assembled immunoglobulins will have known unit structures.
  • a basic four chain structural unit is the form in which IgG, IgD, and IgE exist.
  • a four chain unit is repeated in the higher molecular weight immunoglobulins; IgM generally exists as a pentamer of four basic units held together by disulfide bonds.
  • IgA globulin, and occasionally IgG globulin may also exist in multimeric form in serum. In the case of multimer, each of the four units may be the same or different.
  • each A represents identical or different adhesin amino acid sequences
  • V L is an immunoglobulin light chain variable domain
  • V H is an immunoglobulin heavy chain variable domain
  • C L is an immunoglobulin light chain constant domain
  • C H is an immunoglobulin heavy chain constant domain
  • n is an integer greater than 1;
  • Y designates the residue of a covalent cross-linking agent.
  • the adhesin sequences can be inserted between immunoglobulin heavy chain and light chain sequences, such that an immunoglobulin comprising a chimeric heavy chain is obtained.
  • the adhesin sequences are fused to the 3′ end of an immunoglobulin heavy chain in each arm of an immunoglobulin, either between the hinge and the C H 2 domain, or between the C H 2 and C H 3 domains. Similar constructs have been reported by Hoogenboom, et al., Mol. Immunol. 28:1027-1037 (1991).
  • an immunoglobulin light chain might be present either covalently associated to an adhesin-immunoglobulin heavy chain fusion polypeptide, or directly fused to the adhesin.
  • DNA encoding an immunoglobulin light chain is typically coexpressed with the DNA encoding the adhesin-immunoglobulin heavy chain fusion protein.
  • the hybrid heavy chain and the light chain will be covalently associated to provide an immunoglobulin-like structure comprising two disulfide-linked immunoglobulin heavy chain-light chain pairs.
  • Immunoadhesins are most conveniently constructed by fusing the cDNA sequence encoding the adhesin portion in-frame to an immunoglobulin cDNA sequence.
  • fusion to genomic immunoglobulin fragments can also be used (see, e.g. Aruffo et al., Cell 61:1303-1313 (1990); and Stamenkovic et al., Cell 66:1133-1144 (1991)).
  • the latter type of fusion requires the presence of Ig regulatory sequences for expression.
  • cDNAs encoding IgG heavy-chain constant regions can be isolated based on published sequences from cDNA libraries derived from spleen or peripheral blood lymphocytes, by hybridization or by polymerase chain reaction (PCR) techniques.
  • the cDNAs encoding the “adhesin” and the immunoglobulin parts of the immunoadhesin are inserted in tandem into a plasmid vector that directs efficient expression in the chosen host cells.
  • the protein to be purified is one which is fused to, or conjugated with, a C H 2/C H 3 region.
  • fusion proteins may be produced so as to increase the serum half-life of the protein.
  • biologically important proteins which can be conjugated this way include renin; a growth hormone, including human growth hormone and bovine growth hormone; growth hormone releasing factor; parathyroid hormone; thyroid stimulating hormone; lipoproteins; alpha-1-antitrypsin; insulin A-chain; insulin B-chain; proinsulin; follicle stimulating hormone; calcitonin; luteinizing hormone; glucagon; clotting factors such as factor VIIIC, factor IX, tissue factor, and von Willebrands factor; anti-clotting factors such as Protein C; atrial natriuretic factor; lung surfactant; a plasminogen activator, such as urokinase or human urine or tissue-type plasminogen activator (t-PA); bombesin; thrombin; hemopo
  • This example illustrates the removal of residual 4-vinyl pyridine monomer from poly(4-vinylpyridine).
  • the resulting polymer did not have a noticeable odor, whereas the untreated polymer has a distinct odor of 4-vinyl pyridine monomer.
  • the amount of residual 4-vinyl pyridine monomer present in the treated polymer was not detectable by gel permeation chromatography whereas the untreated polymer had 0.05% (w/w) residual 4-vinyl pyridine monomer
  • This example illustrates the removal of residual 2-vinyl pyridine monomer from poly(2-vinylpyridine).
  • This example illustrates the removal of residual 4-vinyl pyridine and styrene monomers from poly(4-vinylpyridine-co-styrene).
  • the resulting polymer did not have a noticeable odor, whereas the untreated polymer has a distinct odor of 4-vinyl pyridine and styrene monomers.
  • This example illustrates the preparation of a poly(4-vinylpyridine) (P4VP) solution.
  • a 20% (w/w) solution of P4VP was prepared by dissolving 20 g purified P4VP, from example 1, in 80 g 1.0 M hydrochloric acid with continuous agitation for 16 hours at room temperature. The resulting viscous solution was clear and had a slight yellow color.
  • This example illustrates the preparation of a (P2VP) solution.
  • a 20% (w/w) solution of P2VP was prepared by dissolving 20 g purified P2VP, from example 2, in 80 g 1.0 M hydrochloric acid with continuous agitation for 16 hours at room temperature. The resulting viscous solution was clear and had a slight yellow color.
  • This example illustrates the preparation of a P4VP-S solution.
  • a 10% (w/w) solution of P4VP-S was prepared by dissolving 10 g purified P4VP-S, from example 3, in 90 g 1.0 M hydrochloric acid with continuous agitation for 16 hours at room temperature. The resulting viscous solution was hazy.
  • This example illustrates the preparation of a chitosan solution.
  • a 2.5% (w/w) solution of chitosan was prepared by dissolving 2.5 g chitosan, high molecular weight, obtained form Sigma-Aldrich, in 97.5 g 0.5 M hydrochloric acid with continuous agitation for 16 hours at room temperature.
  • This example illustrates the preparation of a P4VP solution.
  • a 10% (w/w) solution of P4VP was prepared by dissolving 10 g purified P4VP, from example 1, in 90 g 1.0 M hydrochloric acid with continuous agitation for 16 hours at room temperature. The resulting viscous solution was clear.
  • This example illustrates the response of a “smart” polymer as a result of a pH stimulus.
  • This example illustrates the removal of a negatively charged dye from solution.
  • This example illustrates the removal of negatively charged impurities from a cell culture medium containing a monoclonal antibody
  • the starting feed for the experiment is this example was a raw cell culture medium from a non-expressing line of Chinese Hamster Ovary (CHO) cells incubated in a Wave bioreactor for ten days (sample C), the appearance of this liquid was turbid (due to suspended cell mass) and slightly yellow.
  • Sample C was spiked with a monoclonal antibody (MAb) to a final concentration of about 1 g/L MAb (sample CP), the appearance of this liquid did not change following the spike.
  • MAb monoclonal antibody
  • sample C was adjusted to 4.8 with the addition of HCl.
  • the P4VP solution of example 8 was added in an amount equivalent to about 20% of the starting volume of pH adjusted sample, the appearance of this sample did not change.
  • the pH of the liquid was then slowly raised to 7 by the dropwise addition of 2N NaOH while mixing. A pale yellow precipitate was formed, which was easily removed by decanting, the resulting liquid was completely clear and colorless (sample T). This procedure was repeated with sample CP to yield a completely clear and colorless liquid (sample TP) after the removal of the yellow precipitate.
  • FIGS. 6A and B show SDS-PAGE data for sample C indicates the presence of a multitude of proteins of varying molecular weights due to CHO cell proteins.
  • Sample CP contains the same population of CHO cell proteins as in sample C as well as the MAb (characteristic molecular weight).
  • Data for sample T shows no visible bands indicating the absence of proteins.
  • Data for sample TP shows only the characteristic bands for the MAb indicating the presence of pure MAb.
  • Gel electrophoresis data in FIG. 7 shows the presence of DNA in samples C and CP while no DNA bands were visible in samples T and TP indicating the absence of DNA. This example shows that this technique is capable of purifying a MAb from a complex mixture of suspended material (cell mass) as well as soluble molecules (host cell proteins and DNA) to result in a pure MAb solution.
  • Lanes 3 and 8 are the molecular weight standards. Lane 4 is sample C. Lane 5 is sample CP. Lane 6 is sample T. Lane 7 is sample TP.
  • Lane 5 is the molecular weight standards. Lane 6 is sample C. Lane 7 is sample CP. Lane 8 is sample T. Lane 9 is sample TP.
  • This example illustrates the use of PVP-S for the removal of cell mass from a cell culture fluid containing a monoclonal antibody
  • the starting feed for the experiment is this example was a raw cell culture fluid from an IgG expressing line of Chinese Hamster Ovary (CHO) cells incubated in a Wave bioreactor, the appearance of this liquid was turbid (due to suspended cell mass) and slightly yellow.
  • the pH of the starting feed was adjusted to 4.8 with the addition of HCl.
  • the PVP-S solution of example 6 was added in an amount equivalent to about 0.1% by weight of dry PVP-S relative to the volume of the starting feed, the appearance of this sample did not change.
  • the pH of the liquid was then quickly raised to 7 by addition of 2N NaOH while mixing vigorously. A pale yellow precipitate was formed, which was easily removed by decanting, the resulting liquid was completely clear.
  • This example illustrates the use of chitosan for the removal of cell mass from a cell culture fluid containing a monoclonal antibody
  • the starting feed for the experiment is this example was a raw cell culture fluid from an IgG expressing line of Chinese Hamster Ovary (CHO) cells incubated in a Wave bioreactor, the appearance of this liquid was turbid (due to suspended cell mass) and slightly yellow.
  • the pH of the starting feed was adjusted to 6.5 with the addition of HCl.
  • the chitosan solution of example 7 was added in an amount equivalent to about 0.06% by weight of dry chitosan relative to the volume of the starting feed, the appearance of this sample did not change.
  • the pH of the liquid was then raised to 7.3 by addition of 1N NaOH while mixing. A precipitate was formed, which was easily removed by decanting, the resulting liquid was completely clear.
  • the turbidity of the supernatant was measured to be less than 5 NTU, the turbidity of the starting feed was in excess of 300 NTU.
  • the removal of DNA by this process was measured to be about 1.5 logs.
  • This example illustrates the use of a mixture of P4VP and chitosan for the removal of cell mass from a cell culture fluid containing a monoclonal antibody
  • the starting feed for the experiment is this example was a raw cell culture fluid from an IgG expressing line of Chinese Hamster Ovary (CHO) cells incubated in a Wave bioreactor, the appearance of this liquid was turbid (due to suspended cell mass) and slightly yellow.
  • the pH of the starting feed was adjusted to 4 with the addition of HCl.
  • the P4VP solution of example 8 and the chitosan solution of example 7 were added in amounts equivalent to about 0.1% and 0.06%, respectively, by weight of dry polymer relative to the volume of the starting feed, the appearance of this sample did not change.
  • the pH of the liquid was then raised to 7.3 by addition of 1N NaOH while mixing. A first precipitate was formed at pH 5 and a second precipitate was observed at pH 7. The solids were then easily removed by decanting, the resulting liquid was completely clear. The turbidity of the supernatant was measured to be less than 5 NTU.
  • This example illustrates the use of chitosan for the removal of cell mass from a cell culture fluid containing a monoclonal antibody without first adjusting the pH of the starting feedstock.
  • the starting feed for the experiment is this example was a raw cell culture fluid from an IgG expressing line of Chinese Hamster Ovary (CHO) cells incubated in a Wave bioreactor, the appearance of this liquid was turbid (due to suspended cell mass) and slightly yellow.
  • the pH of the feedstock was about 7.2
  • Chitosan solution of example 7 was added directly to the starting feedstock, without adjusting the pH of the feedstock, in an amount equivalent to about 0.06% by weight of dry chitosan relative to the volume of the starting feed, the appearance of this sample did not change.
  • the pH of the liquid was then raised to 7.3 by addition of 1N NaOH while mixing. A precipitate was formed, which was easily removed by decanting, the resulting liquid was completely clear.
  • the turbidity of the supernatant was measured to be less than 1 NTU.
  • This example illustrates the synthesis of poly(4-vinyl pyridine-co-vinyl pyridinium Sulphopropyl betaine), a water soluble polymer for impurity removal.
  • the polymer was synthesized by the direct reaction of poly(4-vinyl pyridine), PVP, and 1,3 propane sultone, PS, according to a modified literature technique.
  • 0.18 g of PS was dissolved in 2 ml of propylene carbonate and the solution was then added drop wise to the reaction flask. After the complete addition of the PS solution, the reaction was maintained at 110° C. for 12 hr.
  • the mole ratio of the reactants was selected such that the product comprises 5 mol % of 4-vinyl pyridinium Sulphopropyl betaine.
  • the resulting reaction mixture was cooled down to room temperature and precipitated in excess ethylacetate.
  • the polymer was characterized using FTIR which revealed the presence of the sulfonate stretches at 1035 cm ⁇ 1 and 1200 cm ⁇ 1 .
  • This example illustrates the removal of negatively charged impurities from a cell culture medium containing a monoclonal antibody using poly(4-vinyl pyridine-co-vinyl pyridinium sulphopropyl betaine).
  • This example illustrates the synthesis of poly(4-vinyl pyridine-co-vinyl pyridinium ethylene glycol), a water soluble polymer for impurity removal.
  • This example illustrates the removal of negatively charged impurities from a cell culture medium containing a monoclonal antibody using poly(4-vinyl pyridine-co-vinyl pyridinium ethylene glycol).

Abstract

The present invention relates to a selectively soluble polymer capable of binding to one or more constituents in a mixture containing various biological materials and the methods of using such a polymer to purify a biomolecule from such a mixture. The polymer is soluble in the mixture under a certain set of process conditions such as pH or temperature and is rendered insoluble and precipitates out of solution upon a change in the process conditions. While in its solubilized state, the polymer is capable of binding to a selected entity within the stream such as impurities (DNA, RNA, host cell protein, endotoxins, etc) in a cell broth and remains capable of binding to that entity even after the polymer is precipitated out of solution. The precipitate can then be filtered out from the remainder of the stream and the desired biomolecule is recovered and further processed.

Description

  • This application claims priority of U.S. Provisional Application Ser. No. 60/876,330 filed Dec. 21, 2006, the disclosure of which is incorporated herein by reference.
  • The present invention relates to the purification of biomolecules. More particularly, it relates to the purification of biomolecules such as proteins, polypeptides, antibodies and the like, by a solubilized polymer to remove impurities from a solution/suspension by a controlled precipitation mechanism.
  • BACKGROUND OF THE INVENTION
  • The general process for the manufacture of biomolecules, such as proteins, particularly recombinant proteins typically involves two main steps: (1) the expression of the protein in a host cell, followed by (2) the purification of the protein. The first step involves growing the desired host cell in a bioreactor to effect the expression of the protein, Some examples of cell lines used for this purpose include Chinese hamster ovary (CHO) cells, myeloma (NSO) cells, bacterial cells such as e-coli and insect cells. Once the protein is expressed at the desired levels, the protein is removed from the host cell and harvested. In some instances the protein has been expressed outside of the cell and in others it is still within the cell that must be lysed to allow one access to the protein of interest. Suspended particulates, such as cells, cell fragments, lipids and other insoluble matter are typically removed from the protein-containing fluid by filtration or centrifugation, resulting in a clarified fluid containing the protein of interest in solution as well as other soluble impurities.
  • The second step involves the purification of the harvested protein to remove impurities which are inherent to the process. Examples of impurities include host cell proteins (HCP, proteins other than the desired or targeted protein), nucleic acids, endotoxins, viruses, protein variants and protein aggregates.
  • This purification typically involves several chromatography steps, which can include affinity, ion exchange hydrophobic interaction, etc. One example of chromatography process train for the purification of proteins involves protein-A affinity, followed by cation exchange, followed by anion exchange. The protein-A column captures the protein of interest or target protein by an affinity mechanism while the bulk of the impurities pass through the column to be discarded. The protein then is recovered by elution from the column. Since most of the proteins of interest have isoelectric points (PI) in the basic range (8-9) and therefore being positively charged under normal processing conditions (pH below the PI of the protein), they are bound to the cation exchange resin in the second column. Other positively charged impurities are also bound to this resin. The protein of interest is then recovered by elution from this column under conditions (pH, salt concentration) in which the protein elutes while the impurities remain bound to the resin. The anion exchange column is typically operated in a flow through mode, such that any negatively charged impurities are bound to the resin while the positively charged protein of interest is recovered in the flow through stream. This process results in a highly purified and concentrated protein solution.
  • Other alternative methods for purifying proteins have been investigated in recent years, one such method involves a flocculation technique. In this technique, a soluble polyelectrolyte is added to a clarified or unclarified cell culture broth to capture the impurities thereby forming a flocculant, which is allowed to settle and can be subsequently removed from the protein solution.
  • The main drawback of this flocculation technique is that it requires that the polyelectrolyte be added in the exact amount needed to remove the impurities. If too little flocculent is added, impurities will remain in the protein solution and if too much flocculent is added, the excess polyelectrolyte needs to be removed from the resulting solution. The exact level of impurities in the broth is extremely difficult to predict due to the relatively large degree of variability in the process (from batch to batch) as well as the vast differences between processes to produce different proteins. Removing any excess polyelectrolyte is practically impossible because it is a soluble material and thus it is carried through the process as an undesirable impurity.
  • What is needed is a better process for purifying biomolecules.
  • SUMMARY OF THE INVENTION
  • The present invention relates to a selectively soluble polymer capable of binding to one or more constituents in a biological material containing stream and the methods of using such a material to purify a biomolecule from such a stream.
  • In the following description, the term polymer shall mean a polymer capable of binding to one or more constituents in a biological material containing stream unless otherwise stated.
  • The terms selected biomolecule, target biomolecule or molecule, target protein, biomolecule or protein of interest, or similar terms all refer to products of a biomolecule manufacturing process.
  • The polymer is soluble in an aqueous based solvent under a certain set of process conditions such as pH or temperature or salt concentration or the like and is rendered insoluble and precipitates out of solution upon a change in conditions (temperature, pH, salt or the like). While in its solubilized state, the polymer is capable of binding to a selected entity within the stream such as impurities (cells, cell fragments, lipids, DNA, RNA, host cell protein, endotoxins, virus, etc) in a cell broth and remains capable of binding to that entity even after the polymer is precipitated out of solution. The precipitate can then be easily removed, such as by being filtered out from the remainder of the stream and the desired biomolecule is recovered and further processed.
  • The polymer being bound to one or more impurities, it can either be disposed of or the one or more impurities can be eluted from the polymer and the polymer is then cleaned or sanitized and reused if desired. It can also be washed to ensure that any biomolecules of interest have been recovered for further use or processing.
  • It is an object of the present invention to provide a polymer that is capable of being selectively solubilized in a liquid under certain conditions and to be insoluble and to precipitate out of solution under different conditions in that liquid.
  • It is another object of the present invention to provide a polymer that is capable of being selectively solubilized in a liquid under certain conditions and to be insoluble and to precipitate out of solution under different conditions in that liquid and to allow any excess polymer in solution to be recovered from the solution by precipitation.
  • It is a further object of the present invention to provide a polymer that is capable of being solubilized under a first certain set of conditions in the liquid and to be capable of binding to one or more entities either in the liquid while in its solubilized form and/or to retain the one or more entities or bind to the impurities or have the impurities bind to it as/after being precipitated from the liquid under different conditions in the liquid.
  • It is another object of the present invention to provide a polymer capable of being solubilized under certain ranges of pH, temperature, temperature and salt concentration or the like and to have it bind to one or more entities either in the liquid while in its solubilized form and/or to retain the one or more entities or bind to the impurities or have the impurities bind to it as/after being precipitated under a different set of ranges of pH, temperature, temperature and salt concentration or the like.
  • It is a further object of the present invention to provide a process for purifying a selected biomolecule from a biomolecule containing stream by either having the stream at a given condition or modifying the stream to a given condition and adding a polymer soluble in the stream at that given condition, allowing the solubilized polymer to circulate throughout the stream, further changing the given condition of the stream so as to cause the polymer to become insoluble in the stream and precipitate out with one or more entities of the stream, separating the stream from the polymer and processing one or both further.
  • It is an object to use one or more polymers such as poly(N-vinyl caprolactam), poly(N-acryloylpiperidine), poly(N-vinylisobutyramide), poly (N-substituted acrylamide) including [poly(N-isopropylacrylamide), poly(N,N′-diethylacrylamide), and poly(N-acryloyl-N′-alkylpiperazine)], hydroxyalkylcellulose, copolymers of acrylic acid and methacrylic acid or methacrylic acid and methyl methacrylate, polymers and copolymers of 2 or 4-vinylpyridine and chitosan to selectively remove one or more impurities from a stream containing impurities along with a desired biomolecule.
  • It is an object to do the process with an overabundance of polymer and recover substantially all of the polymer as a precipitate from the mixture.
  • It is an additional object of the present invention to provide the process based on a polymer which is soluble based upon a condition selected from temperature, temperature and salt content or pH.
  • It is another object of the present invention to provide a polymer selected from N-isopropylacrylamide-containing polymers, functionalized agarose, functionalized polyethylene oxide, cationic and anionic polyelectrolytes.
  • It is a further object of the present invention to provide a process for purifying a selected biomolecule from a biomolecule containing mixture by setting the mixture to a given condition, modifying a carrier liquid compatible with the mixture to the same given condition, adding a polymer soluble in the carrier liquid at that given condition to the carrier liquid, allowing the carrier liquid with the solubilized polymer to the mixture and allowing it to circulate throughout the mixture, changing the given condition of the stream so as to cause the polymer to become insoluble in the stream and precipitate out along with one or more entities of the mixture, separating the mixture from the polymer and processing one or both further.
  • It is an additional object of the present invention to provide a static mixer for causing the mixture and solubilized polymer to mix and to allow the polymer to bind to the one or more entities.
  • It is another object of the present invention to provide that the one or more entities are impurities in the mixture.
  • It is a further object of the present invention to provide that the one or more entities are impurities in the mixture selected from host cell protein, cells, cell fragments, nucleic acids and endotoxins.
  • It is an object of the present invention to provide that the one or more entities are viruses which are either removed or rendered inactive by the polymer process. For example, viruses may be removed by the precipitated polymer or they may be rendered inactive by the polymer itself or by the conditions under which the polymer is dissolved into the mixture or rendered insoluble from the mixture.
  • It is an additional object of the present invention to provide a process for the purification of a mixture of biological constituents in a single step.
  • It is another object of the present invention to provide a process for the purification of a mixture of biological constituents selected from proteins, polypeptides, monoclonal antibodies, humanized, chimeral or animal monoclonal antibodies polyclonal antibodies, antibody fragments, multispecific antibodies, immunoadhesins, and C H2/CH3 region-containing proteins.
  • It is a further object of the present invention to provide a process of having a mixture containing a biomolecule of interest at a set range of conditions that will cause one or more polymers of choice to go into solution, adding the one or more polymers and having one or more polymers go into solution, mixing the one or more polymers with entities of the mixture, changing the conditions of the mixture to cause the one or more polymers to precipitate out of solution while retaining one or more entities of the mixture and then separating the precipitate from the remainder of the mixture.
  • It is a further object of the present invention to provide a carrier liquid for the polymer having conditions suitable to cause the polymer to go into solution in the carrier liquid and then to add the carrier liquid with the dissolved polymer to the mixture.
  • It is an additional object of the present invention to provide one or more static mixers to mix the polymer and the mixture.
  • It is another object of the present invention to provide a process for recovering a biomolecule of interest directly from a fermentor or bioreactor in which it has been made.
  • It is a further object of the present invention to provide a process for recovering a biomolecule of interest from a mixture obtained from a fermentor or bioreactor in which it has been made.
  • It is an additional object of the present invention to provide a filtration step to separate the precipitate from the remainder of the mixture.
  • It is another object of the present invention to provide a normal flow filtration step to separate the precipitate from the remainder of the mixture.
  • It is a further object of the present invention to provide a tangential flow filtration step to separate the precipitate from the remainder of the mixture.
  • It is an additional object of the present invention to provide a centrifugation step to separate the precipitate from the remainder of the mixture.
  • It is another object of the present invention to provide a decantation step to separate the precipitate from the remainder of the mixture.
  • It is an additional object of the present invention to provide a further step to recover the one or more constituents of the mixture from the precipitated polymer.
  • It is a further object of the present invention to provide additional processing to the biomolecule of interest.
  • It is an additional object of the present invention to provide a further step of formulating the biomolecule in a pharmaceutically acceptable carrier and using it for various diagnostic, therapeutic or other uses known for such biomolecules.
  • It is an object of the present invention to provide a purified biomolecule in one step, directly out of the bioreactor with no further processing required.
  • It is an additional object of the present invention to improve the efficiency of the clarification (centrifuge or prefiltration) step so that the clarifier (centrifuge or prefilter) exhibits enhanced throughput or capacity.
  • It is another object of the present invention to improve the efficiency of the clarification (centrifuge or prefiltration) step so that the resulting clarified mixture is “cleaner” (going into the sterile membrane filtration step) than a conventional process in which the precipitation technique has not been effected.
  • It is an additional object of the present invention to improve the efficiency of the sterile membrane filtration step so that the membrane filter exhibits enhanced throughput (capacity).
  • It is an additional object of the present invention to provide a purified mixture which enables the improved cleanabiliity of membrane filters and/or chromatography resins that may be used in additional processing following the precipitation step.
  • It is a further object of the present invention to compress the additional processing steps into one continuous process.
  • It is an additional object of the present invention to effect the purification and recovery of a target molecule with additional processing but without the need for bind and elute chromatography steps.
  • It is a further object of the present invention to effect the purification and recovery of a target molecule with additional processing but without the need for any chromatography steps.
  • It is a further object of the present invention to effect the purification and recovery of a target molecule with additional processing in a flow through mode.
  • It is a further object of the present invention to effect the purification and recovery of a target molecule with additional processing using membrane adsorbers.
  • It is a further object of the present invention to effect the purification and recovery of a target molecule with additional processing using a disposable process.
  • It is a further object of the present invention to effect virus inactivation as part of the precipitation process.
  • It is a further object of the present invention to use a UF step to concentrate the target protein after it has been purified and recovered with the precipitation technique.
  • It is an additional object of the present invention to effect the purification and recovery of a target molecule with additional processing using an enhanced UF (charged UF) process.
  • IN THE DRAWINGS
  • FIG. 1 shows a block diagram of a first process according to the present invention.
  • FIG. 2 shows a block diagram of a second process according to the present invention.
  • FIG. 3 shows a block diagram of a third process according to the present invention.
  • FIG. 4 shows a block diagram of a fourth process according to the present invention.
  • FIG. 5 shows a block diagram of a fifth process according to the present invention.
  • FIGS. 6A and B show gel electrophoresis data of Example 11 according to the present invention.
  • FIG. 7 shows SDS-PAGE data of Example 11 according to the present invention.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The invention is to use a liquid phase or solubilized polymer that has a capability, such as affinity or charge or hydrophobicity and the like, to remove undesirable soluble and suspended impurities from a fluid containing a desirable biomolecule of interest. Preferred polymers have an affinity or electrostatic ability. The biomolecule of interest is then recovered and further processed as desired or required.
  • More specifically, the idea relates to the process of using one or more polymers soluble in a liquid phase to remove impurities from a solution/suspension by a precipitation mechanism and which polymer can also be removed, if present, in any excess, by the same mechanism. By way of example, this idea can best be described in the context of protein purification although it can be used to purify any solute from complex mixtures as long as the mechanism of removal applies to the specific solute of interest.
  • The one or more polymers can be used in excess unlike flocculants and can be recovered essentially completely from the mixture by the precipitation action. This allows one to operate the purification step with greater windows of use and without having to calculate the precise amount of material that needs to be used.
  • The present concept is based on the fact that certain polymers undergo changes in properties as a result of changes in the environment in which they are in. The most common polymer property to change as a result of a stimulus is solubility and the most common stimuli are temperature salt concentration and pH. As an example, a polymer may remain in solution as long as the pH, salt or temperature is maintained within a certain range but it will precipitate out of solution as soon as the pH, salt or temperature is raised or lowered outside of said range.
  • Certain polymers, such as poly (N-isopropylacrylamide), agarose, polyethylene oxide, etc. are examples of polymers that exhibit solubility changes as a result of changes in temperature. Other polymers, such as certain catonic and anionic polyelectrolytes, especially poly(4-vinylpyridine), poly(2-vinylpyridine), copolymers of 4-vinyl pyridine or 2-vinyl pyridine with other monomers such as styrene, butyl methacrylate, etc., chitosan and copolymers of acrylic acid or methacrylic acid with other monomers such as methyl methacrylate are examples of polymers that exhibit changes in solubility as a result of changes in pH and/or salt concentration.
  • The precise mechanism is not currently known. It may be that the polymer(s) interact with the entity or entities while in a soluble state and continue to bind to them upon precipitation. It may also be that the polymer and/or entity(s) bind to one another as the polymer is in the process of precipitating. It may be another mechanism as yet unknown to the inventor at this time. The inventor does not wish to be bound to any particular theory of what mechanism is being used but that the invention encompasses any all such mechanisms and phenomena.
  • Depending upon the polymer used, the process used can vary. However, the processes will generally involve having one or more conditions of the liquid or the mixture, such as a cell broth, at the correct pH, temperature, temperature and salt concentration or other condition used to cause the polymer(s) to become soluble and then adding the polymer(s) either directly or already solubilized in a carrier liquid, such as water, to the mixture. In many instances the mixture will be at the proper condition to allow the polymer(s) to be simply added to the mixture. In other instances, the mixture may need to be conditioned or modified to be at the desired condition. This modification or conditioning can be by modifying the mixture first and then adding the polymer(s), by adding the polymer(s) to a carrier liquid that is conditioned to the desired state and simply adding it to the mixture such that the carrier liquid is sufficient to cause the mixture to thus reach that condition or to do both. The mixture and the solubilized or soluble polymer(s) are then mixed to ensure the polymer(s) is solubilized, and that the entities of the mixture and the solubilized polymer(s) have sufficient and intimate contact with each other. The conditions of the liquid in the mixture are then changed (pH, temperature, salt content, combinations thereof, etc) that causes the polymer(s) to become insoluble and precipitate out of the mixture as a solid while either still remaining bound to the one or more entities it contacted while soluble in the mixture or to bind to the entities as it precipitates and continue to bind to it thereafter. The precipitate and remaining mixture are then separated such as by centrifugation or filtration or gravity and time with the liquid portion being decanted. Depending on what was bound to the precipitate, it is either disposed of (if it bound to impurities) or treated (such as by elution and or washing) one or more times to remove any residual impurities or contaminants and then sanitized for reuse.
  • One polymer or a blend of polymers may be used in the present invention and it is meant to cover both embodiments whenever the term polymer, polymer(s) or one or more polymers is used hereafter.
  • As discussed above the polymer may be added directly to the conditioned mixture. Alternatively, it can be added to a carrier liquid in which it is soluble and which carrier preferably is also compatible with the mixture. One such carrier liquid is water, water adjusted to the correct pH by the addition of acids or bases, another is an aqueous based solution such as saline, buffered solutions or blends of water with an organic solvent such as alcohol. The selection of carrier liquid is dependent on the mixture to which it is added as to what is preferred and tolerated. The polymer is added to the carrier liquid that either has already been conditioned (such as pH adjusted or heated to a desired temperature or heated to a desired temperature with the addition of one or more salts or cooled to the desired temperature with or without one or more salts) or it can be added and then the carrier is conditioned to cause the solubilizing of the polymer in the carrier. The carrier/ soluble polymer blend is then added to the. mixture.
  • The mixture may be contained in a mixing vessel such as a tapered bottom metal (preferably stainless steel more preferably 304 or 316 L stainless steel) or glass vat or tank. Alternatively, especially when a cell culture or microbial or yeast culture is used, it may be the bioreactor or fermentor in which it has been grown. It may also be a disposable bioreactor or fermentor or a disposable mixing bag such as a plastic bag as is available from Millipore Corporation of Billerica, Mass. The mixture and polymer are brought into intimate contact through a mixing action that may be done by a magnetic stirred bar, a magnetic driven mixer such as a NovAseptic® mixer available from Millipore Corporation of Billerica Mass., a Lightning-type mixer., a recirculation pump, or a rocking motion closed mixing bag or bioreactor or fermentor, such as is shown in US 2005/0063259A1 or an airlift type of mixer or reactor in which rising bubbles in the liquid cause a circulatory pattern to be formed.
  • Alternatively, the mixture and polymer (either by itself or in a carrier) can be in separate containers and mixed in line in a static blender. The blend can either then go to a container or to a centrifuge or a filter where the polymer is caused to precipitate and the precipitated polymer and its bound one or more entities is separated from the remainder of the mixture. Then at least the remainder of the mixture is further processed.
  • In another embodiment, the mixture and polymer (either by itself or in a carrier) are blended together in the container holding the mixture and further mixed in line in a static blender. The blend is then treated to cause precipitation of the polymer and its bound entity(s). It can either then go to a container or to a centrifuge or to a filter where the precipitated polymer and its bound one or more entities is separated from the remainder of the mixture. Then at least the filtrate is further processed.
  • Using centrifugation, one can easily and quickly separate the precipitated polymer from the remainder of the liquid mixture. After centrifugation, the supernatant, generally the remainder of the mixture is drawn off. Either the drawn off mixture or the precipitated polymer or both if desired is further processed.
  • Simple settling of the precipitated solids and decantation of the supernatant fluid may also be used if desired.
  • Filtration can be accomplished in a variety of manners. Depending upon the size of the polymer as it is precipitated; one may use one or more filters of varying sizes or asymmetries.
  • The selection of type and size of filter will depend on the volume of precipitate to be captured and whether one wishes to further process the precipitated polymer or just the remainder of the mixture,
  • Membrane based filters, preferably microporous membranes can be used in the present invention. Such filters are generally polymeric in nature and can be made from polymers such as but not limited to olefins such as polyethylene including ultrahigh molecular weight polyethylene, polypropylene, EVA copolymers and alpha olefins, metallocene olefinic polymers, PFA, MFA, FIFE, polycarbonates, vinyl copolymers such as PVC, polyamides such as nylon, polyesters, cellulose, cellulose acetate, regenerated cellulose, cellulose composites, polysulfone, polyethersulfone, polyarylsulfone, polyphenylsulfone, polyacrylonitrile, polyvinylidene fluoride (PVDF), and blends thereof. The membrane selected depends upon the application, desired filtration characteristics, particle type and size to be filtered and the flow desired. Preferred membrane based filters include DURAPORE® PVDF membranes available from Millipore Corporation of Billerica Mass., MILLIPORE EXPRESS® and MILLIPORE EXPRESS® PLUS or SH_PES membranes available from Millipore Corporation of Billerica Mass.
  • Depending on the mixture, polymer and the nature of component(s) being removed the membrane may be hydrophilic or hydrophobic. Preferred membranes are hydrophilic and are low in protein binding.
  • The membrane may be symmetric in pore size through out its depth such as DURAPORE® PVDF membranes available from Millipore Corporation of Billerica Mass., or it may be asymmetric in pore size through its thickness as with MILLIPORE EXPRESS® and MILLIPORE EXPRESS® PLUS or SH_PES membranes available from Millipore Corporation of Billerica Mass. It may contain a prefilter layer if desired, either as a separate upstream layer or as an integral upstream portion of the membrane itself.
  • The pore size of the membrane can vary depending upon the polymer and mixture selected. Generally, it has an average pore size of from about 0.05 micron to 5 microns, preferably from about 0.05 micron to about 1 micron, more preferably from about 0.05 to about 0.65 micron.
  • The membrane filter may run in a deadend or normal flow (NF) format or a tangential flow (TFF) format. The choice is dependent on a number of factors, primarily the user's preference or installed filtration equipment as either works with the present invention.
  • Depth filters such as the MILLISTAK+® depth filters, in either lenticular or POD format, or POLYGARD® wound filters available from Millipore Corporation of Billerica Mass. allows one to trap a large volume of precipitated polymer due to its asymmetric structure and large holding capacity. This can be useful when the polymer is designed to remove impurities and to leave the target or desired biomolecule in the liquid of the remaining mixture.
  • FIG. 1 shows a block diagram of a first process of the present invention. In the first step 2, the mixture is either conditioned to the correct parameter(s) to maintain the polymer of choice in solution or if the conditions of the mixture are already such that the polymer(s) become soluble in the mixture, no further conditioning may be required. Also, the polymer(s) may be added as a solid to an unconditioned mixture and then the mixture (containing the solid polymer(s)) may be conditioned to the correct parameters to dissolve the polymer(s) in the mixture. In the second step 4, the polymer(s) is then added to the mixture and mixed to cause it to go into solution and to make intimate contact with all the constituents of the mixture. This may occur in the bioreactor especially if the reactor is a disposable item or in a separate holding tank if desired. In the third step 6, the mixture conditions are changed to cause the polymer(s) to precipitate out of solution while retaining one or more entities of the mixture with it. The mixture and the precipitated polymer(s) are then separated from each other in the fourth step 8. As discussed above the precipitate and remaining mixture may be separated by centrifugation or filtration.
  • FIG. 2 shows a block diagram of a second process of the present invention. In the first step 10, the mixture is either conditioned to the correct parameter(s) to maintain the polymer of choice in solution before, during or after the introduction of the polymer or it is already at the desired condition. In the second step 12 which may occur separately before, simultaneously or after the first step 10, the polymer is added to a carrier liquid under conditions that allow it to go into solution. In the third step 14, the polymer in its carrier is then added to the mixture and mixed to make intimate contact with all the constituents of the mixture. In the fourth step 16, the mixture conditions are changed to cause the polymer to precipitate out of solution carrying one or more entities of the mixture with it. The mixture and the precipitated polymer are then separated from each other in the fifth step 18. As discussed above the precipitate and remaining mixture may be separated by centrifugation or filtration in a sixth step 20 and the target or desired biomolecule is recovered.
  • FIG. 3 shows a block diagram of a third process of the present invention. In the first step 30, the mixture is either conditioned to the correct parameter(s) to maintain the polymer of choice in solution before, during or after introduction of the polymer or it is already at the desired condition. In the second step 32 which may occur separately before, simultaneously or after the first step 30, the polymer is added to a carrier liquid under conditions that allow it to go into solution. In the third step 34, the polymer in its carrier is then added to the mixture through one or more static mixers to make intimate contact with all the constituents of the mixture. In the fourth step 36, the mixture conditions are changed to cause the polymer to precipitate out of solution. The mixture and the precipitated polymer are then separated from each other in the fifth step 38. As discussed above the precipitate and remaining mixture may be separated by centrifugation or filtration in a sixth step 40 and the target or desired biomolecule is recovered.
  • FIG. 4 shows a block diagram of the first process of the present invention as shown in FIG. 1 with an additional step. In the first step 2, the mixture is conditioned to the correct parameter(s), or if the conditions of the mixture are already such that the polymer becomes soluble in the mixture, no further conditioning may be required to maintain the polymer of choice in solution. Also, the polymer may be added as a solid to an unconditioned mixture and then the mixture (containing the solid polymer) may be conditioned to the correct parameters to dissolve the polymer in the mixture and maintain the polymer of choice in solution. In the second step 4, the polymer is then added to the mixture and mixed to cause it to go into solution and to make intimate contact with all the constituents of the mixture. In the third step 6, the mixture conditions are changed to cause the polymer to precipitate out of solution. The mixture and the precipitated polymer are then separated from each other in the fourth step 8. In the fifth step 9, the precipitate is separated from the remaining mixture by filtration. The filtration maybe by either normal flow or tangential flow filtration with any of the membranes or depth filters described above.
  • FIG. 5 shows a block diagram of the first process of the present invention as shown in FIG. 1 with an additional step. In the first step 2, the mixture is conditioned to the correct parameter(s) or if the conditions of the mixture are already such that the polymer becomes soluble in the mixture, no further conditioning may be required to maintain the polymer of choice in solution. Also, the polymer may be added as a solid to an unconditioned mixture and then the mixture (containing the solid polymer) may be conditioned to the correct parameters to dissolve the polymer in the mixture to maintain the polymer of choice in solution. In the second step 4, the polymer is then added to the mixture and mixed to cause it to go into solution and to make intimate contact with all the constituents of the mixture. In the third step 6, the mixture conditions are changed to cause the polymer to precipitate out of solution. The mixture and the precipitated polymer are then separated from each other in the fourth step 8. In the fifth step 7, the precipitate is separated from the remaining mixture by centrifugation.
  • The change in stimuli may be gradual or it may be done substantially instantaneous. For example, a change in pH can be done by slowly adding a pH changing material to the liquid to change the pH slowly over a span of several minutes or even hours. Alternatively, for example, a suitable amount of pH changing material can be added to the liquid at one time to cause the change in pH to occur more rapidly. More control has been found in general with incremental changes rather than immediate changes for most processes.
  • Examples of cationic polyelectrolytes that exhibit this selective solubility behavior include chitosan, polyvinylpyridines (PVPs) and copolymers of PVPs such as poly(2 vinylpyridine) (P2VP) or poly(4 vinylpyridine) (P4VP), polyvinylpyridine-co-styrene (PVP-S), polyvinylpyridine-co-butyl methacrylate (PVP-BMA) as well as other primary, secondary and tertiary amine-containing polymers. These polymers are soluble at a pH lower than about 6-7 and are insoluble at a pH greater than about 5-7. When in solution, these polymers will precipitate if the pH is raised above this critical range (ph=5˜7). In the context of protein purification, a solution of said cationic polyelectrolyte can be added to a fluid containing a biomolecule of interest, such as a protein in the presence of other impurities. This fluid can be for example a cell culture fluid. The polymer is added to the fluid either as a solution in a carrier liquid at a pH of about 4.5 or as a solid particulate in which the fluid is either modified to a pH of about 4.5 either before, during or after the introduction of the polymer to it (as further described below) so that the polymer binds all the negatively charged impurities, such as cells, cell fragments, nucleic acids, viruses, host cell proteins, pyrogens and endotoxins. The biomolecule of interest does not interact with the polymer given its positive net charge due to its basic PI. The pH is then raised to 5-7 or more if desired and the polymer precipitates out of solution, carrying with it all the impurities as well as any excess polymer. The precipitate can then be easily removed by centrifugation or filtration, resulting in a “purified” biomolecule containing solution.
  • An example of anionic polyelectrolytes that exhibit this solubility behavior is a class of copolymers of acrylic acid and methyl methacrylate or methacrylic acid and methyl methacrylate. These polymers are soluble at a pH greater than about 4-7 and insoluble at a pH lower than about 4-7. These polymers can also be used to purify proteins from complex mixtures in a bind and elute mode. For instance, a solution of these polymers can be added to a fluid containing a protein of interest in the presence of other impurities wherein the pH of the fluid is at or above about 4-7. Under these conditions, the negatively charged polymer binds the positively charged protein of interest (basic PI) while it repels the negatively charged impurities. The pH of the fluid is then lowered below about 4-7 to effect precipitation of the polymer-protein complex and any excess polymer. The precipitate can then be washed to remove any soluble or loosely bound impurities while the pH is kept below about 4-7. The protein can be subsequently eluted from the polymer with an elution buffer at high salt concentrations and a pH below about 4-7 to recover the purified protein.
  • Examples of a temperature sensitive polymer is agarose, which is often used in chromatography, hydroxyalkylcelluloses such as hydroxypropylcellulose; polymers and copolymers containing N-isopropylacrylamide monomer, polyethylene oxide, etc. The temperature can then be reduced or raised to cause the polymer to gel and/or precipitate out of solution.
  • In some cases, such as with agarose, these polymers are generally insoluble at room temperature and are soluble in water or other solvents at temperatures generally between about 80 to 120° C. They can be simply heated to cause them to dissolve, added to the mixture and then cooled to cause them to precipitate. In other cases, such as with polymers and copolymers containing N-isopropylacrylamide monomer, the polymer is soluble at a temperature below about 30 to 35° C. and will precipitate out of solution when the temperature is raised above this range.
  • In the case of agarose the use of gel-inhibiting agents such as various salts can depress the solubility temperature to lower temperatures, often to room temperature if desired.
  • Salts that can be used include lithium chloride and zinc chloride. Bases, such as sodium hydroxide or lithium hydroxide can also be used to depress the gelling temperature or to eliminate it altogether. Although the melting point for agarose is about 92 degrees, the gelling temperature is about 43 degrees. This gelling temperature can be manipulated by the modification of the agarose molecule as described above or by the addition of the above salts or by a combination thereof. For example, a cationic ligand can be attached to agarose in an amount such that the gelling temperature of the modified polymer is about 20° C. degrees with or without the addition of the above salts. The modified agarose is added, in solution at a temperature about 25° C. degrees, to the mixture (also at a temperature of about 25° C. degrees) to bind the constituents and then the temperature of the mixture is lowered to below 20° C. degrees thereby gelling the modified agarose with the constituents.
  • With some polymers, such as polyvinylpyridine, polyvinylpyridene-co-styrene and the like, there may be residual monomer left in the polymer as supplied. It is desirable to remove any free monomer before using the polymer. One such method is to place the polymer as purchased in an oven, preferably with an inert or low oxygen gas atmosphere such as by purging the oven several times during the process with argon or nitrogen, and maintain it at an elevated temperature (generally between 100 and 200° C., preferably about 120° C.) and under a vacuum so as to drive off all monomer present (generally about 24 hours).
  • Additionally, with some polymers, such as polyvinylpyridine, polyvinylpyridene-co-sytrene and the like, it is desirable to select higher molecular weight polymers (200,000 molecular weight or higher) as they have been found to more freely precipitate out of solution than lower molecular weight polymers. This means that one can be sure that no residual polymer is left in the solution after precipitation.
  • In some instances, precipitation by itself may be slow or incomplete. In those instances, one can repeat the process of changing the stimuli conditions two or more times, add precipitant enhancers such as glass beads, salts and the like, vary the temperature of the process and the like to enhance the precipitation.
  • Typical polymer concentrations in the carrier solvent are between 1-20% by weight depending on the viscosity of the solution. It is preferred that the concentration be as high as possible to minimize dilution of the feedstock. Practically, polymer solutions in the 10-20% are preferred to achieve a good balance between viscosity and dilution of the feedstock. The final concentration of the polymer in the feedstock may depend on the amount of impurities in the feedstock but it is typically between 0.01% to 2% by weight and more specifically between 0.05% and 0.1%.
  • In some processes one may use two or more polymers either simultaneously or sequentially to enhance the impurity removal. For example, one may use chitosan as the first polymer and conduct a first purification step. This fluid is separated from the precipitated chitosan/impurities and then treated with a second polymer such as a polyvinylpyridine to further remove impurities.
  • The recovered biomolecule may then undergo one or more additional processing steps depending on whether it is contained within the liquid of the mixture or is bound to the precipitated polymer.
  • A method of sequential precipitation may be used to recover the biomolecule of interest. In such a method, a first precipitation as described above would be used to remove impurities and the precipitated polymer/impurities mass would be separated from the target biomolecule containing solution. The solution would then be mixed with a stimuli responsive polymer containing a ligand capable of binding to the biomolecule of interest at a solution condition at which the polymer is soluble. Following methods described previously, the solution conditions would be changed so as to precipitate the polymer and bound biomolecule. The polymer/biomolecule would then be separated as previously described, the biomolecule eluted or otherwise separated from the polymer, and the recovered biomolecule further processed as needed.
  • As the biomolecule of interest is in the liquid, it may, if needed or desired undergo one or more known process steps including but not limited to chromatography steps such as ion exchange, hydrophobic interaction or affinity chromatography, various filtration steps such as high performance tangential flow filtration (HPTFF), viral removal/inactivation steps, final filtration and the like. Alternatively, the biomolecule of interest present in the liquid may be used as is without the need for further purification steps.
  • The chromatography may be column based using solid bead media or monoliths or through a membrane absorber or chromatography device. The step if desired can be a classic bind/elute or a flow through mode of chromatography.
  • Also the biomolecule of interest may undergo further purification without the need for chromatography steps such as through the use of high performance tangential flow filtration using one or more charged membranes.
  • Additionally, in several embodiments, no further purification is required. One may if desired add additional steps to ensure that viruses have been removed or inactivated or to be sure no residual precipitate remains.
  • A further variation uses an affinity step to bind and then elute the desired biomolecule. Affinity ligands such as Protein A either on a solid matrix such as a bead or membrane may be used.
  • In one embodiment of the present invention, the current process simply replaces a clarification step and prefilter step in a normal biological product process train.
  • In another embodiment, it replaces clarification, prefiltration and at least one chromatography step by directly purifying the biomolecule of interest from the starting materials.
  • In an additional embodiment, it replaces a cell harvest or biomolecule collection step by being added directly to the bioreactor or fermentor. This also eliminates the need for clarification, prefiltration and potentially at least one chromatography step by directly purifying the biomolecule of interest from the starting materials.
  • In any of the embodiments of the present invention the protein thus recovered may be formulated in a pharmaceutically acceptable carrier and is used for various diagnostic, therapeutic or other uses known for such molecules.
  • The mixture that is the starting material of the process will vary depending upon the cell line in which it was grown as well as the conditions under which it is grown and harvested. For example, in most CHO cell processes the cells express the molecule outside of the cell wall into the media. One tries not to rupture the cells during harvest in order to reduce the amount impurities in the mixture. However, some cells during grow and harvesting may rupture due to shear or other handling conditions or die and lyse, spilling their contents into the mixture. In bacteria cell systems, the biomolecule is often kept with the cellular wall or it may actually be part of the cellular wall (Protein A). In these systems the cell walls need to be disrupted or lysed in order to recover the biomolecule of interest.
  • The target molecule to be purified can be any biomolecule, preferably a protein, in particular, recombinant protein produced in any host cell, including but not limited to, Chinese hamster ovary (CHO) cells, Per.C6® cell lines available from Crucell of the Netherlands, myeloma cells such as NSO cells, other animal cells such as mouse cells, insect cells, or microbial cells such as E. coli or yeast. Additionally, the mixture may be a fluid derived from an animal modified to produce a transgenic fluid such as milk or blood that contains the biomolecule of interest. Optimal target proteins are antibodies, immunoadhesins and other antibody-like molecules, such as fusion proteins including a C H2/CH3 region. In particular, this product and process can be used for purification of recombinant humanized monoclonal antibodies such as (RhuMAb) from a conditioned harvested cell culture fluid (HCCF) grown in Chinese hamster ovary (CHO) cells expressing RhuMAb.
  • Antibodies within the scope of the present invention include, but are not limited to: anti-HER2 antibodies including Trastuzumab (HERCEPTIN®) (Carter et al., Proc. Natl. Acad. Sci. USA, 89:4285-4289 (1992), U.S. Pat. No. 5,725,856); anti-CD20 antibodies such as chimeric anti-CD20 “C2B8” as in U.S. Pat. No. 5,736,137 (RITUXAN.®), a chimeric or humanized variant of the 2H7 antibody as in U.S. Pat. No. 5,721,108, B1, or Tositumomab (BEXXAR.®); anti-IL-8 (St John et al., Chest, 103:932 (1993), and International Publication No. WO 95/23865); anti-VEGF antibodies including humanized and/or affinity matured anti-VEGF antibodies such as the humanized anti-VEGF antibody huA4.6.1 AVASTIN®. (Kim et al., Growth Factors, 7:53-64 (1992), International Publication No. WO 96/30046, and WO 98/45331, published Oct. 15, 1998); anti-PSCA antibodies (WO01/40309); anti-CD40 antibodies, including S2C6 and humanized variants thereof (WO00/75348); anti-CD11a (U.S. Pat. No. 5,622,700, WO 98/23761, Steppe et al., Transplant Intl. 4:3-7 (1991), and Hourmant et al., Transplantation 58:377-380 (1994)); anti-IgE (Presta et al., J Immunol. 151:2623-2632 (1993), and International Publication No. WO 95/19181); anti-CD18 (U.S. Pat. No. 5,622,700, issued Apr. 22, 1997, or as in WO 97/26912, published Jul. 31, 1997); anti-IgE (including E25, E26 and E27; U.S. Pat. No. 5,714,338, issued Feb. 3, 1998 or U.S. Pat. No. 5,091,313, issued Feb. 25, 1992, WO 93/04173 published Mar. 4, 1993, or International Application No. PCT/US98/13410 filed Jun. 30, 1998, U.S. Pat. No. 5,714,338); anti-Apo-2 receptor antibody (WO 98/51793 published Nov. 19, 1998); anti-TNF-α antibodies including cA2 (REMICADE®), CDP571 and MAK-195 (See, U.S. Pat. No. 5,672,347 issued Sep. 30, 1997, Lorenz et al. J. Immunol. 156(4):1646-1653 (1996), and Dhainaut et al. Crit. Care Med. 23(9):1461-1469 (1995)); anti-Tissue Factor (TF) (European Patent No. 0 420 937 B1 granted Nov. 9, 1994); anti-human α4β7 integrin (WO 98/06248 published Feb. 19, 1998); anti-EGFR (chimerized or humanized 225 antibody as in WO 96/40210 published Dec. 19, 1996); anti-CD3 antibodies such as OKT3 (U.S. Pat. No. 4,515,893 issued May 7, 1985); anti-CD25 or anti-tac antibodies such as CHI-621 (SIMULECT®) and (ZENAPAX®) (See U.S. Pat. No. 5,693,762 issued Dec. 2, 1997); anti-CD4 antibodies such as the cM-7412 antibody (Choy et al. Arthritis Rheum 39(1):52-56 (1996)); anti-CD52 antibodies such as CAMPATH-1H (Riechmann et al. Nature 332:323-337 (1988)); anti-Fc receptor antibodies such as the M22 antibody directed against FcγRI as in Graziano et al. J. Immunol. 155(10):4996-5002 (1995); anti-carcinoembryonic antigen (CEA) antibodies such as hMN-14 (Sharkey et al. Cancer Res. 55(23Suppl): 5935s-5945s (1995); antibodies directed against breast epithelial cells including huBrE-3, hu-Mc 3 and CRL6 (Ceriani et al. Cancer Res. 55(23): 5852s-5856s (1995); and Richman et al. Cancer Res. 55(23 Supp): 5916s-5920s (1995)); antibodies that bind to colon carcinoma cells such as C242 (Litton et al. Eur J. Immunol. 26(1):1-9 (1996)); anti-CD38 antibodies, e.g. AT 13/5 (Ellis et al. J Immunol. 155(2):925-937 (1995)); anti-CD33 antibodies such as Hu M195 (Jurcic et al. Cancer Res 55(23 Suppl):5908s-5910s (1995) and CMA-676 or CDP771; anti-CD22 antibodies such as LL2 or LymphoCide (Juweid et al. Cancer Res 55(23 Suppl):5899s-5907s (1995)); anti-EpCAM antibodies such as 17-1A (PANOREX®); anti-GpIIb/IIIa antibodies such as abciximab or c7E3 Fab (REOPRO®); anti-RSV antibodies such as MEDI-493 (SYNAGIS®); anti-CMV antibodies such as PROTOVIR®; anti-HIV antibodies such as PRO542; anti-hepatitis antibodies such as the anti-Hep B antibody OSTAVIR®; anti-CA 125 antibody OvaRex; anti-idiotypic GD3 epitope antibody BEC2; anti-αvβ3 antibody VITAXIN®.; anti-human renal cell carcinoma antibody such as ch-G250; ING-1; anti-human 17-1A antibody (3622W94); anti-human colorectal tumor antibody (A33); anti-human melanoma antibody R24 directed against GD3 ganglioside; anti-human squamous-cell carcinoma (SF-25); and anti-human leukocyte antigen (HLA) antibodies such as Smart ID10 and the anti-HLA DR antibody Oncolym (Lym-1). The preferred target antigens for the antibody herein are: HER2 receptor, VEGF, IgE, CD20, CD11a, and CD40.
  • Aside from the antibodies specifically identified above, the skilled practitioner could generate antibodies directed against an antigen of interest, e.g., using the techniques described below.
  • The antibody herein is directed against an antigen of interest. Preferably, the antigen is a biologically important polypeptide and administration of the antibody to a mammal suffering from a disease or disorder can result in a therapeutic benefit in that mammal. However, antibodies directed against non-polypeptide antigens (such as tumor-associated glycolipid antigens; see U.S. Pat. No. 5,091,178) are also contemplated. Where the antigen is a polypeptide, it may be a transmembrane molecule (e.g. receptor) or ligand such as a growth factor. Exemplary antigens include those proteins described in section (3) below. Exemplary molecular targets for antibodies encompassed by the present invention include CD proteins such as CD3, CD4, CD8, CD19, CD20, CD22, CD34, CD40; members of the ErbB receptor family such as the EGF receptor, HER2, HER3 or HER4 receptor; cell adhesion molecules such as LFA-1, Mac1, p150,95, VLA-4, ICAM-1, VCAM and αv/β3 integrin including either α or β subunits thereof (e.g. anti-CD11a, anti-CD18 or anti-CD11b antibodies); growth factors such as VEGF; IgE; blood group antigens; flk2/flt3 receptor; obesity (OB) receptor; mpl receptor; CTLA-4; protein C, or any of the other antigens mentioned herein. Antigens to which the antibodies listed above bind are specifically included within the scope herein.
  • Soluble antigens or fragments thereof, optionally conjugated to other molecules, can be used as immunogens for generating antibodies. For transmembrane molecules, such as receptors, fragments of these (e.g. the extracellular domain of a receptor) can be used as the immunogen. Alternatively, cells expressing the transmembrane molecule can be used as the immunogen. Such cells can be derived from a natural source (e.g. cancer cell lines) or may be cells which have been transformed by recombinant techniques to express the transmembrane molecule.
  • Other antigens and forms thereof useful for preparing antibodies will be apparent to those in the art.
  • Polyclonal antibodies can also be purified in the present invention. Polyclonal antibodies are preferably raised in animals by multiple subcutaneous (sc) or intraperitoneal (ip) injections of the relevant antigen and an adjuvant. It may be useful to conjugate the antigen to a protein that is immunogenic in the species to be immunized, e.g., keyhole limpet hemocyanin, serum albumin, bovine thyroglobulin, or soybean trypsin inhibitor using a bifunctional or derivatizing agent, for example, maleimidobenzoyl sulfosuccinimide ester (conjugation through cysteine residues), N-hydroxysuccinimide (through lysine residues), glutaraldehyde, succinic anhydride, SOCl2, or R1N C═NR, where R and R1 are different alkyl groups.
  • Animals are immunized against the antigen, immunogenic conjugates, or derivatives by combining, e.g., 100 μg or 5 μg of the protein or conjugate (for rabbits or mice, respectively) with 3 volumes of Freund's complete adjuvant and injecting the solution intradermally at multiple sites. One month later the animals are boosted with ⅕ to 1/10 the original amount of antigen or conjugate in Freund's complete adjuvant by subcutaneous injection at multiple sites. Seven to 14 days later the animals are bled and the serum is assayed for antibody titer. Animals are boosted until the titer plateaus. Preferably, the animal is boosted with the conjugate of the same antigen, but conjugated to a different protein and/or through a different cross-linking reagent. Conjugates also can be made in recombinant cell culture as protein fusions. Also, aggregating agents such as alum are suitably used to enhance the immune response.
  • Monoclonal antibodies are of interest in the present invention and may be made using the hybridoma method first described by Kohler et al., Nature, 256:495 (1975), or may be made by recombinant DNA methods (U.S. Pat. No. 4,816,567).
  • In the hybridoma method, a mouse or other appropriate host animal, such as a hamster or macaque monkey, is immunized as hereinabove described to elicit lymphocytes that produce or are capable of producing antibodies that will specifically bind to the protein used for immunization. Alternatively, lymphocytes may be immunized in vitro. Lymphocytes then are fused with myeloma cells using a suitable fusing agent, such as polyethylene glycol, to form a hybridoma cell (Goding, Monoclonal Antibodies: Principles and Practice, pp. 59-103 (Academic Press, 1986)).
  • The hybridoma cells thus prepared are seeded and grown in a suitable culture medium that preferably contains one or more substances that inhibit the growth or survival of the unfused, parental myeloma cells. For example, if the parental myeloma cells lack the enzyme hypoxanthine guanine phosphoribosyl transferase (HGPRT or HPRT), the culture medium for the hybridomas typically will include hypoxanthine, aminopterin, and thymidine (HAT medium), which substances prevent the growth of HGPRT-deficient cells.
  • Preferred myeloma cells are those that fuse efficiently, support stable high-level production of antibody by the selected antibody-producing cells, and are sensitive to a medium such as HAT medium. Among these, preferred myeloma cell lines are murine myeloma lines, such as those derived from MOPC-21 and MPC-11 mouse tumors available from the Salk Institute Cell Distribution Center, San Diego, Calif. USA, and SP-2 or X63-Ag8-653 cells available from the American Type Culture Collection, Rockville, Md. USA. Human myeloma and mouse-human heteromyeloma cell lines also have been described for the production of human monoclonal antibodies (Kozbor, J. Immunol., 133:3001 (1984); Brodeur et al., Monoclonal Antibody Production Techniques and Applications, pp. 51-63 (Marcel Dekker, Inc., New York, 1987)).
  • Culture medium in which hybridoma cells are growing is assayed for production of monoclonal antibodies directed against the antigen. Preferably, the binding specificity of monoclonal antibodies produced by hybridoma cells is determined by immunoprecipitation or by an in vitro binding assay, such as radioimmunoassay (RIA) or enzyme-linked immunoabsorbent assay (ELISA).
  • After hybridoma cells are identified that produce antibodies of the desired specificity, affinity, and/or activity, the clones may be subcloned by limiting dilution procedures and grown by standard methods (Goding, Monoclonal Antibodies: Principles and Practice, pp. 59-103 (Academic Press, 1986)). Suitable culture media for this purpose include, for example, D-MEM or RPMI-1640 medium. In addition, the hybridoma cells may be grown in vivo as ascites tumors in an animal.
  • The monoclonal antibodies secreted by the subclones are suitably separated from the culture medium, ascites fluid, or serum by conventional immunoglobulin purification procedures such as, for example, Pro-Sep® Protein A media available from Millipore Corporation of Billerica, Mass., hydroxyapatite chromatography, gel electrophoresis, dialysis, or affinity chromatography. Preferably the Protein A chromatography procedure described herein is used.
  • DNA encoding the monoclonal antibodies is readily isolated and sequenced using conventional procedures (e.g., by using oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of the monoclonal antibodies). The hybridoma cells serve as a preferred source of such DNA. Once isolated, the DNA may be placed into expression vectors, which are then transfected into host cells such as E. coli cells, simian COS cells, Chinese hamster ovary (CHO) cells, or myeloma cells that do not otherwise produce immunoglobulin protein, to obtain the synthesis of monoclonal antibodies in the recombinant host cells.
  • The DNA also may be modified, for example, by substituting the coding sequence for human heavy- and light-chain constant domains in place of the homologous murine sequences (U.S. Pat. No. 4,816,567; Morrison, et al., Proc. Natl. Acad. Sci. USA, 81:6851 (1984)), or by covalently joining to the immunoglobulin coding sequence all or part of the coding sequence for a non-immunoglobulin polypeptide.
  • Typically such non-immunoglobulin polypeptides are substituted for the constant domains of an antibody, or they are substituted for the variable domains of one antigen-combining site of an antibody to create a chimeric bivalent antibody comprising one antigen-combining site having specificity for an antigen and another antigen-combining site having specificity for a different antigen.
  • In a further embodiment, monoclonal antibodies can be isolated from antibody phage libraries generated using the techniques described in McCafferty et al., Nature, 348:552-554 (1990). Clackson et al., Nature, 352:624-628 (1991) and Marks et al., J. Mol. Biol., 222:581-597 (1991) describe the isolation of murine and human antibodies, respectively, using phage libraries. Subsequent publications describe the production of high affinity (nM range) human antibodies by chain shuffling (Marks et al., Bio/Technology, 10:779-783 (1992)), as well as combinatorial infection and in vivo recombination as a strategy for constructing very large phage libraries (Waterhouse et al., Nuc. Acids. Res., 21:2265-2266 (1993)). Thus, these techniques are viable alternatives to traditional hybridoma techniques for isolation of monoclonal antibodies.
  • A humanized antibody has one or more amino acid residues introduced into it from a source which is non-human. These non-human amino acid residues are often referred to as “import” residues, which are typically taken from an “import” variable domain. Humanization can be essentially performed following the method of Winter and co-workers (Jones et al., Nature, 321:522-525 (1986); Riechmann et al., Nature, 332:323-327 (1988); Verhoeyen et al., Science, 239:1534-1536 (1988)), by substituting rodent CDRs or CDR sequences for the corresponding sequences of a human antibody. Accordingly, such “humanized” antibodies are chimeric antibodies (U.S. Pat. No. 4,816,567) wherein substantially less than an intact human variable domain has been substituted by the corresponding sequence from a non-human species. In practice, humanized antibodies are typically human antibodies in which some CDR residues and possibly some FR residues are substituted by residues from analogous sites in rodent antibodies.
  • The choice of human variable domains, both light and heavy, to be used in making the humanized antibodies is very important to reduce antigenicity. According to the so-called “best-fit” method, the sequence of the variable domain of a rodent antibody is screened against the entire library of known human variable-domain sequences. The human sequence which is closest to that of the rodent is then accepted as the human FR for the humanized antibody (Sims et al., J. Immunol., 151:2296 (1993)). Another method uses a particular framework derived from the consensus sequence of all human antibodies of a particular subgroup of light or heavy chains. The same framework may be used for several different humanized antibodies (Carter et al., Proc. Natl. Acad. Sci. USA, 89:4285 (1992); Presta et al., J. Immunol., 151:2623 (1993)).
  • It is further important that antibodies be humanized with retention of high affinity for the antigen and other favorable biological properties. To achieve this goal, according to a preferred method, humanized antibodies are prepared by a process of analysis of the parental sequences and various conceptual humanized products using three-dimensional models of the parental and humanized sequences. Three-dimensional immunoglobulin models are commonly available and are familiar to those skilled in the art. Computer programs are available which illustrate and display probable three-dimensional conformational structures of selected candidate immunoglobulin sequences. Inspection of these displays permits analysis of the likely role of the residues in the functioning of the candidate immunoglobulin sequence, i.e., the analysis of residues that influence the ability of the candidate immunoglobulin to bind its antigen. In this way, FR residues can be selected and combined from the recipient and import sequences so that the desired antibody characteristic, such as increased affinity for the target antigen(s), is achieved. In general, the CDR residues are directly and most substantially involved in influencing antigen binding.
  • Alternatively, it is now possible to produce transgenic animals (e.g., mice) that are capable, upon immunization, of producing a full repertoire of human antibodies in the absence of endogenous immunoglobulin production. For example, it has been described that the homozygous deletion of the antibody heavy-chain joining region (JH) gene in chimeric and germ-line mutant mice results in complete inhibition of endogenous antibody production. Transfer of the human germ-line immunoglobulin gene array in such germ-line mutant mice will result in the production of human antibodies upon antigen challenge. See, e.g., Jakobovits et al., Proc. Natl. Acad. Sci. USA, 90:2551 (1993); Jakobovits et al., Nature, 362:255-258 (1993); Bruggermann et al., Year in Immuno., 7:33 (1993); and Duchosal et al. Nature 355:258 (1992). Human antibodies can also be derived from phage-display libraries (Hoogenboom et al., J. Mol. Biol., 227:381 (1991); Marks et al., J. Mol. Biol., 222:581-597 (1991); Vaughan et al. Nature Biotech 14:309 (1996)).
  • Various techniques have been developed for the production of antibody fragments. Traditionally, these fragments were derived via proteolytic digestion of intact antibodies (see, e.g., Morimoto et al., Journal of Biochemical and Biophysical Methods 24:107-117 (1992) and Brennan et al., Science, 229:81 (1985)). However, these fragments can now be produced directly by recombinant host cells. For example, the antibody fragments can be isolated from the antibody phage libraries discussed above. Alternatively, Fab′-SH fragments can be directly recovered from E. coli and chemically coupled to form F(ab′)2 fragments (Carter et al., Bio/Technology 10:163-167 (1992)). According to another approach, F(ab′)2 fragments can be isolated directly from recombinant host cell culture. Other techniques for the production of antibody fragments will be apparent to the skilled practitioner. In other embodiments, the antibody of choice is a single chain Fv fragment (scFv). See WO 93/16185.
  • Multispecific antibodies have binding specificities for at least two different antigens. While such molecules normally will only bind two antigens (i.e. bispecific antibodies, BsAbs), antibodies with additional specificities such as trispecific antibodies are encompassed by this expression when used herein.
  • Methods for making bispecific antibodies are known in the art. Traditional production of full length bispecific antibodies is based on the coexpression of two immunoglobulin heavy chain-light chain pairs, where the two chains have different specificities (Millstein et al., Nature, 305:537-539 (1983)). Because of the random assortment of immunoglobulin heavy and light chains, these hybridomas (quadromas) produce a potential mixture of 10 different antibody molecules, of which only one has the correct bispecific structure. Purification of the correct molecule, which is usually done by affinity chromatography steps, is rather cumbersome, and the product yields are low. Similar procedures are disclosed in WO 93/08829, and in Traunecker et al., EMBO J., 10:3655-3659 (1991).
  • According to another approach described in WO96/27011, the interface between a pair of antibody molecules can be engineered to maximize the percentage of heterodimers which are recovered from recombinant cell culture. The preferred interface comprises at least a part of the CH3 domain of an antibody constant domain. In this method, one or more small amino acid side chains from the interface of the first antibody molecule are replaced with larger side chains (e.g. tyrosine or tryptophan). Compensatory “cavities” of identical or similar size to the large side chain(s) are created on the interface of the second antibody molecule by replacing large amino acid side chains with smaller ones (e.g. alanine or threonine). This provides a mechanism for increasing the yield of the heterodimer over other unwanted end-products such as homodimers.
  • Bispecific antibodies include cross-linked or “heteroconjugate” antibodies. For example, one of the antibodies in the heteroconjugate can be coupled to avidin, the other to biotin. Such antibodies have, for example, been proposed to target immune system cells to unwanted cells (U.S. Pat. No. 4,676,980), and for treatment of HIV infection (WO 91/00360, WO 92/200373, and EP 03089). Heteroconjugate antibodies may be made using any convenient cross-linking methods. Suitable cross-linking agents are well known in the art, and are disclosed in U.S. Pat. No. 4,676,980, along with a number of cross-linking techniques.
  • Techniques for generating bispecific antibodies from antibody fragments have also been described in the literature. For example, bispecific antibodies can be prepared using chemical linkage. Brennan et al., Science, 229: 81 (1985) describe a procedure wherein intact antibodies are proteolytically cleaved to generate F(ab′)2 fragments. These fragments are reduced in the presence of the dithiol complexing agent sodium arsenite to stabilize vicinal dithiols and prevent intermolecular disulfide formation. The Fab′ fragments generated are then converted to thionitrobenzoate (TNB) derivatives. One of the Fab′-TNB derivatives is then reconverted to the Fab′-thiol by reduction with mercaptoethylamine and is mixed with an equimolar amount of the other Fab'-TNB derivative to form the bispecific antibody. The bispecific antibodies produced can be used as agents for the selective immobilization of enzymes.
  • Recent progress has facilitated the direct recovery of Fab′-SH fragments from E. coli, which can be chemically coupled to form bispecific antibodies. Shalaby et al., J Exp. Med., 175: 217-225 (1992) describe the production of a fully humanized bispecific antibody F(ab′)2 molecule. Each Fab′ fragment was separately secreted from E. coli and subjected to directed chemical coupling in vitro to form the bispecific antibody. The bispecific antibody thus formed was able to bind to cells overexpressing the ErbB2 receptor and normal human T cells, as well as trigger the lytic activity of human cytotoxic lymphocytes against human breast tumor targets.
  • Various techniques for making and isolating bispecific antibody fragments directly from recombinant cell culture have also been described. For example, bispecific antibodies have been produced using leucine zippers. Kostelny et al., J. Immunol., 148(5):1547-1553 (1992). The leucine zipper peptides from the Fos and Jun proteins were linked to the Fab' portions of two different antibodies by gene fusion. The antibody homodimers were reduced at the hinge region to form monomers and then re-oxidized to form the antibody heterodimers. This method can also be utilized for the production of antibody homodimers. The “diabody” technology described by Hollinger et al., Proc. Natl. Acad. Sci. USA, 90:6444-6448 (1993) has provided an alternative mechanism for making bispecific antibody fragments. The fragments comprise a heavy-chain variable domain (VH) connected to a light-chain variable domain (VL) by a linker which is too short to allow pairing between the two domains on the same chain. Accordingly, the VH and VL domains of one fragment are forced to pair with the complementary VL and VH domains of another fragment, thereby forming two antigen-binding sites. Another strategy for making bispecific antibody fragments by the use of single-chain Fv (sFv) dimers has also been reported. See Gruber et al., J. Immunol., 152:5368 (1994). Alternatively, the antibodies can be “linear antibodies” as described in Zapata et al. Protein Eng. 8(10):1057-1062 (1995). Briefly, these antibodies comprise a pair of tandem Fd segments (VH-CH1-VH-CH1) which form a pair of antigen binding regions. Linear antibodies can be bispecific or monospecific.
  • Antibodies with more than two valencies are contemplated. For example, trispecific antibodies can be prepared. Tutt et al. J. Immunol. 147: 60 (1991).
  • The simplest and most straightforward immunoadhesin design combines the binding domain(s) of the adhesin (e.g. the extracellular domain (ECD) of a receptor) with the hinge and Fc regions of an immunoglobulin heavy chain. Ordinarily, when preparing the immunoadhesins of the present invention, nucleic acid encoding the binding domain of the adhesin will be fused C-terminally to nucleic acid encoding the N-terminus of an immunoglobulin constant domain sequence, however N-terminal fusions are also possible.
  • Typically, in such fusions the encoded chimeric polypeptide will retain at least functionally active hinge, C H2 and CH3 domains of the constant region of an immunoglobulin heavy chain. Fusions are also made to the C-terminus of the Fc portion of a constant domain, or immediately N-terminal to the C H1 of the heavy chain or the corresponding region of the light chain. The precise site at which the fusion is made is not critical; particular sites are well known and may be selected in order to optimize the biological activity, secretion, or binding characteristics of the immunoadhesin.
  • In a preferred embodiment, the adhesin sequence is fused to the N-terminus of the Fc domain of immunoglobulin G1 (IgG1). It is possible to fuse the entire heavy chain constant region to the adhesin sequence. However, more preferably, a sequence beginning in the hinge region just upstream of the papain cleavage site which defines IgG Fc chemically (i.e. residue 216, taking the first residue of heavy chain constant region to be 114), or analogous sites of other immunoglobulins is used in the fusion. In a particularly preferred embodiment, the adhesin amino acid sequence is fused to (a) the hinge region and C H2 and CH3 or (b) the C H1, hinge, C H2 and CH3 domains, of an IgG heavy chain.
  • For bispecific immunoadhesins, the immunoadhesins are assembled as multimers, and particularly as heterodimers or heterotetramers. Generally, these assembled immunoglobulins will have known unit structures. A basic four chain structural unit is the form in which IgG, IgD, and IgE exist. A four chain unit is repeated in the higher molecular weight immunoglobulins; IgM generally exists as a pentamer of four basic units held together by disulfide bonds. IgA globulin, and occasionally IgG globulin, may also exist in multimeric form in serum. In the case of multimer, each of the four units may be the same or different.
  • Various exemplary assembled immunoadhesins within the scope herein are schematically diagrammed below:
  • (a) ACL-ACL;
  • (b) ACH-(ACH, ACL-ACH, ACL-VHCH, or VLCL-ACH);
  • (c) ACL-ACH-(ACL-ACH, ACL-VHCH, VLCL-ACH, or VLCL-VHCH)
  • (d) ACL-VHCH-(ACH, Or ACL-VHCH, or VLCL-ACH);
  • (e) VLCL-ACH-(ACL-VHCH, or VLCL-ACH); and
  • (f) (A-Y)n-(VLCL-VHCH)2,
  • wherein each A represents identical or different adhesin amino acid sequences;
  • VL is an immunoglobulin light chain variable domain;
  • VH is an immunoglobulin heavy chain variable domain;
  • CL is an immunoglobulin light chain constant domain;
  • CH is an immunoglobulin heavy chain constant domain;
  • n is an integer greater than 1;
  • Y designates the residue of a covalent cross-linking agent.
  • In the interests of brevity, the foregoing structures only show key features; they do not indicate joining (J) or other domains of the immunoglobulins, nor are disulfide bonds shown. However, where such domains are required for binding activity, they shall be constructed to be present in the ordinary locations which they occupy in the immunoglobulin molecules.
  • Alternatively, the adhesin sequences can be inserted between immunoglobulin heavy chain and light chain sequences, such that an immunoglobulin comprising a chimeric heavy chain is obtained. In this embodiment, the adhesin sequences are fused to the 3′ end of an immunoglobulin heavy chain in each arm of an immunoglobulin, either between the hinge and the C H2 domain, or between the C H2 and CH3 domains. Similar constructs have been reported by Hoogenboom, et al., Mol. Immunol. 28:1027-1037 (1991).
  • Although the presence of an immunoglobulin light chain is not required in the immunoadhesins of the present invention, an immunoglobulin light chain might be present either covalently associated to an adhesin-immunoglobulin heavy chain fusion polypeptide, or directly fused to the adhesin. In the former case, DNA encoding an immunoglobulin light chain is typically coexpressed with the DNA encoding the adhesin-immunoglobulin heavy chain fusion protein. Upon secretion, the hybrid heavy chain and the light chain will be covalently associated to provide an immunoglobulin-like structure comprising two disulfide-linked immunoglobulin heavy chain-light chain pairs. Methods suitable for the preparation of such structures are, for example, disclosed in U.S. Pat. No. 4,816,567, issued 28 Mar. 1989.
  • Immunoadhesins are most conveniently constructed by fusing the cDNA sequence encoding the adhesin portion in-frame to an immunoglobulin cDNA sequence. However, fusion to genomic immunoglobulin fragments can also be used (see, e.g. Aruffo et al., Cell 61:1303-1313 (1990); and Stamenkovic et al., Cell 66:1133-1144 (1991)). The latter type of fusion requires the presence of Ig regulatory sequences for expression. cDNAs encoding IgG heavy-chain constant regions can be isolated based on published sequences from cDNA libraries derived from spleen or peripheral blood lymphocytes, by hybridization or by polymerase chain reaction (PCR) techniques. The cDNAs encoding the “adhesin” and the immunoglobulin parts of the immunoadhesin are inserted in tandem into a plasmid vector that directs efficient expression in the chosen host cells.
  • In other embodiments, the protein to be purified is one which is fused to, or conjugated with, a C H2/CH3 region. Such fusion proteins may be produced so as to increase the serum half-life of the protein. Examples of biologically important proteins which can be conjugated this way include renin; a growth hormone, including human growth hormone and bovine growth hormone; growth hormone releasing factor; parathyroid hormone; thyroid stimulating hormone; lipoproteins; alpha-1-antitrypsin; insulin A-chain; insulin B-chain; proinsulin; follicle stimulating hormone; calcitonin; luteinizing hormone; glucagon; clotting factors such as factor VIIIC, factor IX, tissue factor, and von Willebrands factor; anti-clotting factors such as Protein C; atrial natriuretic factor; lung surfactant; a plasminogen activator, such as urokinase or human urine or tissue-type plasminogen activator (t-PA); bombesin; thrombin; hemopoietic growth factor; tumor necrosis factor-alpha and -beta; enkephalinase; RANTES (regulated on activation normally T-cell expressed and secreted); human macrophage inflammatory protein (MIP-1-alpha); a serum albumin such as human serum albumin; Muellerian-inhibiting substance; relaxin A-chain; relaxin B-chain; prorelaxin; mouse gonadotropin-associated peptide; a microbial protein, such as beta-lactamase; DNase; IgE; a cytotoxic T-lymphocyte associated antigen (CTLA), such as CTLA-4; inhibin; activin; vascular endothelial growth factor (VEGF); receptors for hormones or growth factors; Protein A or D; rheumatoid factors; a neurotrophic factor such as bone-derived neurotrophic factor (BDNF), neurotrophin-3, -4, -5, or -6 (NT-3, NT-4, NT-5, or NT-6), or a nerve growth factor such as NGF-β; platelet-derived growth factor (PDGF); fibroblast growth factor such as aFGF and bFGF; epidermal growth factor (EGF); transforming growth factor (TGF) such as TGF-alpha and TGF-beta, including TGF-β1, TGF-β2, TGF-β3, TGF-β4, or TGF-β5; insulin-like growth factor-I and -II (IGF-I and IGF-II); des(1-3)-IGF-I (brain IGF-I), insulin-like growth factor binding proteins; CD proteins such as CD3, CD4, CD8, CD19, CD20, CD34, and CD40; erythropoietin; osteoinductive factors; immunotoxins; a bone morphogenetic protein (BMP); an interferon such as interferon-alpha, -beta, and -gamma; colony stimulating factors (CSFs), e.g., M-CSF, GM-CSF, and G-CSF; interleukins (ILs), e.g., IL-1 to IL-10; superoxide dismutase; T-cell receptors; surface membrane proteins; decay accelerating factor; viral antigen such as, for example, a portion of the AIDS envelope; transport proteins; homing receptors; addressins; regulatory proteins; integrins such as CD11a, CD11b, CD11c, CD18, an ICAM, VLA-4 and VCAM; a tumor associated antigen such as HER2, HER3 or HER4 receptor; and fragments of any of the above-listed polypeptides.
  • Examples
  • The following examples are offered by way of illustration and not by way of limitation. The disclosures of all citations in the specification are expressly incorporated herein by reference.
  • Example 1
  • This example illustrates the removal of residual 4-vinyl pyridine monomer from poly(4-vinylpyridine).
  • Linear poly(4-vinylpyridine), (P4VP) MW 200,000 obtained form Scientific Polymer Products, Inc., was spread evenly on a glass dish and placed in a vacuum oven. The atmosphere inside the oven was purged with argon for 5 minutes several times to remove oxygen. The pressure in the oven was reduced to 0.1 in mercury using a mechanical vacuum pump and subsequently the temperature was raised to 120° C. The polymer was subjected to these conditions for a total of 24 hours. During this time, the atmosphere inside the oven was purged with argon for 5 minutes several times. At the end of the heating period, the oven temperature was lowered to room temperature and the oven was purged with argon several times before opening the door. The resulting polymer did not have a noticeable odor, whereas the untreated polymer has a distinct odor of 4-vinyl pyridine monomer. The amount of residual 4-vinyl pyridine monomer present in the treated polymer was not detectable by gel permeation chromatography whereas the untreated polymer had 0.05% (w/w) residual 4-vinyl pyridine monomer
  • Example 2
  • This example illustrates the removal of residual 2-vinyl pyridine monomer from poly(2-vinylpyridine).
  • Linear poly(2-vinylpyridine), (P2VP) MW 200,000 obtained form Scientific Polymer Products, Inc., was treated exactly according to the process of example 1. The resulting polymer did not have a noticeable odor, whereas the untreated polymer has a distinct odor of 2-vinyl pyridine monomer.
  • Example 3
  • This example illustrates the removal of residual 4-vinyl pyridine and styrene monomers from poly(4-vinylpyridine-co-styrene).
  • Linear poly(4-vinylpyridine-co-styrene), (P4VP-S), 10% styrene content, obtained form Scientific Polymer Products, Inc., was treated exactly according to the process of example 1. The resulting polymer did not have a noticeable odor, whereas the untreated polymer has a distinct odor of 4-vinyl pyridine and styrene monomers.
  • Example 4
  • This example illustrates the preparation of a poly(4-vinylpyridine) (P4VP) solution.
  • A 20% (w/w) solution of P4VP was prepared by dissolving 20 g purified P4VP, from example 1, in 80 g 1.0 M hydrochloric acid with continuous agitation for 16 hours at room temperature. The resulting viscous solution was clear and had a slight yellow color.
  • Example 5
  • This example illustrates the preparation of a (P2VP) solution.
  • A 20% (w/w) solution of P2VP was prepared by dissolving 20 g purified P2VP, from example 2, in 80 g 1.0 M hydrochloric acid with continuous agitation for 16 hours at room temperature. The resulting viscous solution was clear and had a slight yellow color.
  • Example 6
  • This example illustrates the preparation of a P4VP-S solution.
  • A 10% (w/w) solution of P4VP-S was prepared by dissolving 10 g purified P4VP-S, from example 3, in 90 g 1.0 M hydrochloric acid with continuous agitation for 16 hours at room temperature. The resulting viscous solution was hazy.
  • Example 7
  • This example illustrates the preparation of a chitosan solution.
  • A 2.5% (w/w) solution of chitosan was prepared by dissolving 2.5 g chitosan, high molecular weight, obtained form Sigma-Aldrich, in 97.5 g 0.5 M hydrochloric acid with continuous agitation for 16 hours at room temperature.
  • Example 8
  • This example illustrates the preparation of a P4VP solution.
  • A 10% (w/w) solution of P4VP was prepared by dissolving 10 g purified P4VP, from example 1, in 90 g 1.0 M hydrochloric acid with continuous agitation for 16 hours at room temperature. The resulting viscous solution was clear.
  • Example 9
  • This example illustrates the response of a “smart” polymer as a result of a pH stimulus.
  • About 5 ml of the P4VP solution from example 8 were placed in a test tube, the pH was measured to be below 5. The pH of this solution was slowly raised to 7 by a dropwise addition of 2N NaOH. The polymer precipitated out of solution, the color of the precipitate was white and the decanted liquid was completely clear and colorless suggesting the absence of any polymer.
  • Example 10
  • This example illustrates the removal of a negatively charged dye from solution.
  • About 3 ml of the P4VP solution from example 8 were added to about 5 ml of a solution containing 0.1 ml Ponceau-S (negatively charged dye, red in color) in water. After the addition, the solution was clear and red in color. The pH of the solution was slowly raised to 7 with a few drops of 2N NaOH, a red precipitate was formed and a clear and colorless liquid was recovered by decanting. The red precipitate contains all the Ponceau S dye that was effectively removed from the liquid phase.
  • Example 11
  • This example illustrates the removal of negatively charged impurities from a cell culture medium containing a monoclonal antibody
  • The starting feed for the experiment is this example was a raw cell culture medium from a non-expressing line of Chinese Hamster Ovary (CHO) cells incubated in a Wave bioreactor for ten days (sample C), the appearance of this liquid was turbid (due to suspended cell mass) and slightly yellow. Sample C was spiked with a monoclonal antibody (MAb) to a final concentration of about 1 g/L MAb (sample CP), the appearance of this liquid did not change following the spike.
  • The pH of sample C was adjusted to 4.8 with the addition of HCl. To this liquid, the P4VP solution of example 8 was added in an amount equivalent to about 20% of the starting volume of pH adjusted sample, the appearance of this sample did not change. The pH of the liquid was then slowly raised to 7 by the dropwise addition of 2N NaOH while mixing. A pale yellow precipitate was formed, which was easily removed by decanting, the resulting liquid was completely clear and colorless (sample T). This procedure was repeated with sample CP to yield a completely clear and colorless liquid (sample TP) after the removal of the yellow precipitate.
  • All samples were filtered through 0.2 μm PVDF membranes and analyzed for the presence of host cell proteins (CHO cell proteins), MAb and DNA. FIGS. 6A and B show SDS-PAGE data for sample C indicates the presence of a multitude of proteins of varying molecular weights due to CHO cell proteins. Sample CP contains the same population of CHO cell proteins as in sample C as well as the MAb (characteristic molecular weight). Data for sample T shows no visible bands indicating the absence of proteins. Data for sample TP shows only the characteristic bands for the MAb indicating the presence of pure MAb. Gel electrophoresis data in FIG. 7 shows the presence of DNA in samples C and CP while no DNA bands were visible in samples T and TP indicating the absence of DNA. This example shows that this technique is capable of purifying a MAb from a complex mixture of suspended material (cell mass) as well as soluble molecules (host cell proteins and DNA) to result in a pure MAb solution.
  • SDS-PAGE data as shown in FIGS. 6A and B:
  • Lanes 3 and 8 are the molecular weight standards. Lane 4 is sample C. Lane 5 is sample CP. Lane 6 is sample T. Lane 7 is sample TP.
  • Gel electrophoresis data as shown in FIG. 7:
  • Lane 5 is the molecular weight standards. Lane 6 is sample C. Lane 7 is sample CP. Lane 8 is sample T. Lane 9 is sample TP.
  • Example 12
  • This example illustrates the use of PVP-S for the removal of cell mass from a cell culture fluid containing a monoclonal antibody
  • The starting feed for the experiment is this example was a raw cell culture fluid from an IgG expressing line of Chinese Hamster Ovary (CHO) cells incubated in a Wave bioreactor, the appearance of this liquid was turbid (due to suspended cell mass) and slightly yellow.
  • The pH of the starting feed was adjusted to 4.8 with the addition of HCl. To this fluid, the PVP-S solution of example 6 was added in an amount equivalent to about 0.1% by weight of dry PVP-S relative to the volume of the starting feed, the appearance of this sample did not change. The pH of the liquid was then quickly raised to 7 by addition of 2N NaOH while mixing vigorously. A pale yellow precipitate was formed, which was easily removed by decanting, the resulting liquid was completely clear.
  • Example 13
  • This example illustrates the use of chitosan for the removal of cell mass from a cell culture fluid containing a monoclonal antibody
  • The starting feed for the experiment is this example was a raw cell culture fluid from an IgG expressing line of Chinese Hamster Ovary (CHO) cells incubated in a Wave bioreactor, the appearance of this liquid was turbid (due to suspended cell mass) and slightly yellow.
  • The pH of the starting feed was adjusted to 6.5 with the addition of HCl. To this fluid, the chitosan solution of example 7 was added in an amount equivalent to about 0.06% by weight of dry chitosan relative to the volume of the starting feed, the appearance of this sample did not change. The pH of the liquid was then raised to 7.3 by addition of 1N NaOH while mixing. A precipitate was formed, which was easily removed by decanting, the resulting liquid was completely clear. The turbidity of the supernatant was measured to be less than 5 NTU, the turbidity of the starting feed was in excess of 300 NTU. The removal of DNA by this process was measured to be about 1.5 logs.
  • Example 14
  • This example illustrates the use of a mixture of P4VP and chitosan for the removal of cell mass from a cell culture fluid containing a monoclonal antibody
  • The starting feed for the experiment is this example was a raw cell culture fluid from an IgG expressing line of Chinese Hamster Ovary (CHO) cells incubated in a Wave bioreactor, the appearance of this liquid was turbid (due to suspended cell mass) and slightly yellow.
  • The pH of the starting feed was adjusted to 4 with the addition of HCl. To this fluid, the P4VP solution of example 8 and the chitosan solution of example 7 were added in amounts equivalent to about 0.1% and 0.06%, respectively, by weight of dry polymer relative to the volume of the starting feed, the appearance of this sample did not change. The pH of the liquid was then raised to 7.3 by addition of 1N NaOH while mixing. A first precipitate was formed at pH 5 and a second precipitate was observed at pH 7. The solids were then easily removed by decanting, the resulting liquid was completely clear. The turbidity of the supernatant was measured to be less than 5 NTU.
  • Example 15
  • This example illustrates the use of chitosan for the removal of cell mass from a cell culture fluid containing a monoclonal antibody without first adjusting the pH of the starting feedstock.
  • The starting feed for the experiment is this example was a raw cell culture fluid from an IgG expressing line of Chinese Hamster Ovary (CHO) cells incubated in a Wave bioreactor, the appearance of this liquid was turbid (due to suspended cell mass) and slightly yellow. The pH of the feedstock was about 7.2
  • Chitosan solution of example 7 was added directly to the starting feedstock, without adjusting the pH of the feedstock, in an amount equivalent to about 0.06% by weight of dry chitosan relative to the volume of the starting feed, the appearance of this sample did not change. The pH of the liquid was then raised to 7.3 by addition of 1N NaOH while mixing. A precipitate was formed, which was easily removed by decanting, the resulting liquid was completely clear. The turbidity of the supernatant was measured to be less than 1 NTU.
  • Example 16
  • This example illustrates the synthesis of poly(4-vinyl pyridine-co-vinyl pyridinium Sulphopropyl betaine), a water soluble polymer for impurity removal.
  • The polymer was synthesized by the direct reaction of poly(4-vinyl pyridine), PVP, and 1,3 propane sultone, PS, according to a modified literature technique. The polymer was initially dried in a vacuum oven at T=100° C. for 48 hr to remove residual monomer. 3 g of PVP were transferred to the reaction flask and dissolved in 50 ml propylene carbonate (99%) maintained at T=80° C. under a nitrogen atmosphere. 0.18 g of PS was dissolved in 2 ml of propylene carbonate and the solution was then added drop wise to the reaction flask. After the complete addition of the PS solution, the reaction was maintained at 110° C. for 12 hr. The mole ratio of the reactants was selected such that the product comprises 5 mol % of 4-vinyl pyridinium Sulphopropyl betaine. The resulting reaction mixture was cooled down to room temperature and precipitated in excess ethylacetate. The precipitate was dried in a vacuum oven a T=70° C. for 24 hr.
  • The polymer was characterized using FTIR which revealed the presence of the sulfonate stretches at 1035 cm−1 and 1200 cm−1.
  • Example 17
  • This example illustrates the removal of negatively charged impurities from a cell culture medium containing a monoclonal antibody using poly(4-vinyl pyridine-co-vinyl pyridinium sulphopropyl betaine).
  • 0.07 g of the copolymer (10 wt %) from example 16 were mixed with 10 ml feedstock after adjusting the pH of the latter to 3.5-4.0 using 0.4 g of 1M HCl. Following 30 min incubation time, the polymer-cell complexes were precipitated by increasing the pH gradually to 5.2 using NaOH (1.0 M). The precipitate was removed by filtration and the pH of the supernatant was adjusted to 7.0.
  • Following this procedure, 85 wt % of IgG was recovered in the clarified supernatant. The quoted numbers are relative to the initial amount of IgG present in the starting unclarified feedstock.
  • Example 18
  • This example illustrates the synthesis of poly(4-vinyl pyridine-co-vinyl pyridinium ethylene glycol), a water soluble polymer for impurity removal.
  • The polymer was synthesized by the direct reaction of poly(4-vinyl pyridine), P4VP, and polyethylene glycol diglycidyl ether (Mn 540 g mol−1). The polymer was initially dried in a vacuum oven at T=100° C. for 48 hr to remove residual monomer. 10 g of P4VP were transferred to the reaction flask and dissolved in 100 ml methanol, followed by the drop wise addition of 0.1 g of lithium hydroxide dissolved in 10 ml deionized water. A precipitate was observed upon the addition of the lithium hydroxide solution but re-dissolved within minutes of continuous stirring. After adding 0.15 g of polyethylene glycol diglycidyl ether to the reaction flask, the mixture was maintained at 80° C. for 24 hrs. The resulting reaction mixture was then cooled down to room temperature and precipitated in water. The product was further purified by precipitation from an acidic solution by adjusting the solution pH to neutral conditions. The purified polymer was stored in the solution state as 10 wt % in 1M HCl.
  • Example 19
  • This example illustrates the removal of negatively charged impurities from a cell culture medium containing a monoclonal antibody using poly(4-vinyl pyridine-co-vinyl pyridinium ethylene glycol).
  • 0.04 g of the copolymer (10 wt %) from example 18 were mixed with 8 ml feedstock after adjusting the pH of the latter to 3.5 using 0.4 g of 1M HCl. Following 10 min incubation time, the polymer-cell complexes were precipitated by increasing the pH gradually to 5.2 using NaOH (1M). The precipitate was removed by filtration and the pH of the supernatant was adjusted to 7.0. The turbidity of the supernatant was around 90 NTU, while the turbidity of the starting feedstock was 300 NTU.

Claims (41)

1) A method for purifying a biomolecule from a mixture containing impurities comprising:
a. providing the mixture at a set of conditions,
b. adding one or more polymers solubilizable in said mixture under the set of conditions, the one or more polymers being capable of binding to one or more of the impurities in the mixture,
c. mixing the one or more solubilized polymers throughout the mixture;
d. precipitating the one or more polymers and one or more bound impurities out of solution by changing the set of conditions in the mixture; and
e. recovering the biomolecule.
2) The method of claim 1 wherein the one or more polymers are solubilized in carrier liquid before addition to the mixture.
3) The method of claim 1 wherein the one or more polymers is caused to be soluble in the aqueous solution due to a pH level and the polymer is precipitated by a pH change.
4) The method of claim 1 wherein the one or more polymers is caused to be soluble in the aqueous solution due to a pH level below 7.
5) The method of claim 1 wherein the one or more polymers is caused to be soluble in the mixture due to a temperature of the one or more polymers and the mixture and the one or more polymers precipitates by a change in the temperature of the one or more polymers and mixture.
6) The method of claim 1 wherein the biomolecule is a protein.
7) The method of claim 1 wherein the biomolecule is an antibody.
8) The method of claim 1 wherein the biomolecule is a recombinant antibody.
9) The method of claim 1 wherein the biomolecule is a monoclonal antibody.
10) The method of claim 1 wherein the biomolecule is a recombinant monoclonal antibody.
11) The method of claim 1 wherein the biomolecule is selected from the group consisting of a polyclonal antibody, a humanized antibody and an antibody fragment.
12) The method of claim 1 wherein said biomolecule is an antibody fragment selected from the group consisting of Fab, Fab!, f(ab!)2 and Fv fragments, single-chain antibody molecules diabodies, linear antibodies, bispecific antibodies and multispecific antibodies formed from antibody fragments.
13) The method of claim 1 wherein said biomolecule is an antibody that specifically binds to an antigen selected from the group consisting of CD3, CD4, CD8, CD19, CD20, CD34, CD40, EGF receptor, HER2, HER3, HER4 receptor, LFA-1, Mac1, p150,95, VLA-4, ICAM-1, VCAM, av/b3 integrin, CD11a, CD18, CD11b, VEGF, IgE, flk2/flt3 receptor, obesity (OB) receptor, mpl receptor, CTLA-4 and polypeptide C.
14) The method of claim 1 wherein said biomolecule is an antibody selected from the group consisting of anti-HER2; anti-CD20; anti-IL-8; anti-VEGF; anti-PSCA; anti-CD11a; anti-IgE; anti-Apo-2 receptor; anti-TNF-α, anti-Tissue Factor(TF); anti-CD3; anti-CD25; anti-CD34; anti-CD40; anti-tac; anti-CD4; anti-CD52; anti-Fc receptor; anti-carcinoembryonic antigen (CEA) antibodies; antibodies directed against breast epithelial cells; antibodies that bind to colon carcinoma cells; anti-CD33; anti-CD22; anti-EpCAM; anti-GpIIb/IIIa; anti-RSV; anti-CMV; anti-HIV; anti-hepatitis; anti-αvβ3; anti-human renal cell carcinoma; anti-human 17-1A; anti-human colorectal tumor; anti-human melanoma; anti-human squamous-cell carcinoma; and anti-human leukocyte antigen (HLA) antibodies.
15) The method of claim 1 wherein said biomolecule is an antibody selected from the group consisting of anti-HER2 receptor, anti-VEGF, anti-IgE, anti-CD20, anti-CD11a, and anti-CD40 antibodies.
16) The method of claim 1 wherein the biomolecule is an immunoadhesin.
17) The method of claim 1 wherein the biomolecule is an antibody-like molecule.
18) The method of claim 1 wherein the biomolecule is an antibody-like molecule and the antibody-like molecule is a protein fused to, or conjugated with, a CH2/CH3 region.
19) The method of claim 1 wherein the biomolecule is an antibody-like molecule and the antibody-like molecule is a protein fused to, or conjugated with, a CH2/CH3 region and said protein is selected from the group consisting of rennin; growth hormones; growth hormone releasing factor; parathyroid hormone; thyroid stimulating hormone; lipoproteins; alpha-l-antitrypsin; insulin A-chain; insulin B-chain; proinsulin; follicle stimulating hormone; calcitonin; luteinizing hormone; glucagons; factor VIIIC; factor IX; tissue factor; von Willebrands factor; Protein C; atrial natriuretic factor; lung surfactant; urokinase; human urine and tissue-type plasminogen activator (t-PA); bombesin; thrombin; hemopoietic growth factor; tumor necrosis factor-alpha and -beta; enkephalinase; RANTES; human macrophage inflammatory protein (MIP-1alpha); serum albumins; Muellerian-inhibiting substance; relaxin A-chain; relaxin B-chain; prorelaxin; mouse gonadotropin-associated peptide; beta-lactamase; DNase; IgE, cytotoxic T-lymphocyte associated antigens (CTLAs); inhibin; activin; vascular endothelial growth factor (VEGF); receptors for hormones or growth factors; Protein A or D; rheumatoid factors; bone-derived neurotrophic factor (BDNF); neurotrophin-3. -4, -5, and -6 (NT-3, NT-4, NT-5, and NT-6), nerve growth factors; platelet-derived growth factor (PDGF); fibroblast growth factors; epidermal growth factor (EGF); transforming growth factors (TGF); insulin like growth factor-I and -II (IGF-I and IGF-II); des(1-3)-IGF-I (brain IGF-I) insulin-like growth factor binding proteins (IGFBPs); CD proteins; erythropoietin; osteoinductive factors; immunotoxins; bone morphogenetic proteins (BMPs); interferons-alpha, -beta, and -gamma; colony stimulating factors (CSFs); interleukins IL-1 to IL-10; superoxide dismutase; T-cell receptors; surface membrane proteins; decay accelerating factor; viral antigens; transport proteins; homing receptors; addressing; regulatory proteins; integrins; tumor associated antigens; and fragments and thereof.
20) The method of claim 1 further comprising the step of incorporating the recovered biomolecule into a pharmaceutical formulation.
21) The method of claim 1 wherein the one or more polymers are selected from the group consisting of poly(N-isopropylacrylamide), agarose, dextran, polyethylene oxide, cationic polyelectrolytes and anionic polyelectrolytes.
22) The method of claim 1 wherein the one or more polymers are precipitated by a change in temperature.
23) The method of claim 1 wherein the one or more polymers are precipitated by a change in temperature and are selected from the group consisting of poly(N-isopropylacrylamide), agarose Hydroxyalkylcellulose and polyethylene oxide.
24) The method of claim 1 wherein the one or more polymers are precipitated by a change in pH.
25) The method of claim 1 wherein the one or more polymers are precipitated by a change in pH and are selected from the group consisting of cationic polyelectrolytes and anionic polyelectrolytes.
26) The method of claim 1 wherein the one or more polymers are precipitated by a change in pH and are cationic polyelectrolytes selected from the group consisting of chitosan, polyvinylpyridine, copolymers of vinylpyridine, primary amine containing polymers, secondary amine containing polymers and tertiary amine containing polymers.
27) The method of claim 1 wherein the one or more polymers are precipitated by a change in pH and are anionic polyelectrolytes selected from the group consisting of copolymers of acrylic acid and methyl methacrylate and copolymers of methacrylic acid and methyl methacrylate.
28) The method of claim 1 wherein the one or more polymers are rendered soluble by the condition of temperature and the mixture is maintained at the desired temperature for period of time to maintain the one or more polymers in solution and the mixture temperature is then changed to precipitate out the one or more polymers.
29) The method of claim 1 wherein the one or more polymers are solubilized and precipitated by a change in pH, are cationic polyelectrolytes selected from the group consisting of chitosan, polyvinylpyridine, copolymers of vinyl pyridine, primary amine containing polymers, secondary amine containing polymers and tertiary amine containing polymers and are soluble at a first pH and are insoluble at a second pH.
30) The method of claim 1 wherein the one or more polymers are solubilized and precipitated by a change in pH, are cationic polyelectrolytes selected from the group consisting of chitosan, polyvinylpyridine, copolymers of vinyl pyridine, primary amine containing polymers, secondary amine containing polymers and tertiary amine containing polymers.
31) The method of claim 1 wherein the one or more polymers are solubilized and precipitated by a change in pH, are anionic polyelectrolytes selected from the group consisting of copolymers of acrylic acid and methyl methacrylate and copolymers of methacrylic acid and methyl methacrylate and are soluble at a first pH and are insoluble at a second pH.
32) The method of claim 1 wherein the one or more polymers are added to a carrier liquid under conditions to cause the one or more polymers to go into solution and the carrier liquid containing the one or more polymers in solution is added to the mixture through a static mixer.
33) A method for purifying a biomolecule from a mixture containing a host cell protein as an impurity comprising subjecting said mixture to:
a. providing a mixture of a biomolecule and a host cell protein as an impurity,
b. conducting a purification step by adding a carrier liquid containing one or more polymers solubilized in said carrier liquid to the mixture, the polymer being soluble under a set of conditions within the carrier liquid and the mixture and being capable of binding to the impurity,
c. allowing the one or more polymers to mix with the constituents of the mixture;
d. precipitating the one or more polymers and host cell protein of the mixture out of solution by changing the set of conditions in the mixture which causes the one or more polymers to be insoluble;
e. filtering the precipitated one or more polymers from the mixture, and
f. recovering the biomolecule at a purity at least 1 LRV better than the initial mixture.
34) The method of claim 1 further comprising the recovered biomolecule is formulated in a pharmaceutically acceptable carrier.
35) The method of claim 1 further comprising the recovered biomolecule is formulated in a pharmaceutically acceptable carrier a purpose selected from the group consisting of research, diagnostic and therapeutic purposes.
36) The method of claim 1 wherein the one or more polymers are added in excess to the mixture and recovered as the precipitate.
37) The method of claim 1 further comprising subjecting the recovered biomolecule to a second purification step in which a stimuli responsive soluble polymer which is capable of binding to the biomolecule is added to a mixture containing the recovered biomolecule from the method of claim 1 under conditions to cause the polymer to be in solution, the conditions are changed so as to cause the polymer to precipitate out of solution with the recovered biomolecule, recovering the precipitate, eluting the biomolecule from the precipitate and recovering the biomolecule.
38) The method of claim 1 further comprising subjecting the recovered biomolecule to a second purification step in which a stimuli responsive soluble polymer which is capable of binding to the biomolecule is added to a mixture containing the recovered biomolecule from the method of claim 1 under conditions to cause the polymer to be in solution, the conditions are changed so as to cause the polymer to precipitate out of solution with the recovered biomolecule, recovering the precipitate, washing the precipitate at least one time, eluting the biomolecule from the precipitate and recovering the biomolecule.
39) The method of claim 1 wherein the impurity is selected from the group consisting of DNA, RNA, virus, host cell proteins, endotoxins, cells, cell fragments and combination thereof.
40) The method of claim 1 wherein the biomolecule is recovered by separating the one or more precipitated polymers and one or more impurities from the mixture.
41) The method of claim 1 wherein the one or more polymers are capable of binding with one or more of the impurities in the solubilized and precipitated state.
US12/448,004 2006-12-21 2007-12-20 Purification of proteins Abandoned US20100267933A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/448,004 US20100267933A1 (en) 2006-12-21 2007-12-20 Purification of proteins

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US87633006P 2006-12-21 2006-12-21
US12/448,004 US20100267933A1 (en) 2006-12-21 2007-12-20 Purification of proteins
PCT/US2007/026090 WO2008079302A2 (en) 2006-12-21 2007-12-20 Purification of proteins

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2007/026090 A-371-Of-International WO2008079302A2 (en) 2006-12-21 2007-12-20 Purification of proteins

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/732,613 Continuation US10793593B2 (en) 2006-12-21 2013-01-02 Purification of proteins

Publications (1)

Publication Number Publication Date
US20100267933A1 true US20100267933A1 (en) 2010-10-21

Family

ID=39562839

Family Applications (3)

Application Number Title Priority Date Filing Date
US12/448,004 Abandoned US20100267933A1 (en) 2006-12-21 2007-12-20 Purification of proteins
US12/004,314 Active 2028-09-01 US8163886B2 (en) 2006-12-21 2007-12-20 Purification of proteins
US13/732,613 Active 2028-03-25 US10793593B2 (en) 2006-12-21 2013-01-02 Purification of proteins

Family Applications After (2)

Application Number Title Priority Date Filing Date
US12/004,314 Active 2028-09-01 US8163886B2 (en) 2006-12-21 2007-12-20 Purification of proteins
US13/732,613 Active 2028-03-25 US10793593B2 (en) 2006-12-21 2013-01-02 Purification of proteins

Country Status (2)

Country Link
US (3) US20100267933A1 (en)
WO (2) WO2008079280A1 (en)

Cited By (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20130005950A1 (en) * 2008-12-16 2013-01-03 Emd Millipore Corporation Stirred Tank Reactor And Method
US8362217B2 (en) 2006-12-21 2013-01-29 Emd Millipore Corporation Purification of proteins
US8569464B2 (en) 2006-12-21 2013-10-29 Emd Millipore Corporation Purification of proteins
WO2014004281A1 (en) 2012-06-29 2014-01-03 Emd Millipore Corporation Purification of biological molecules
US8691918B2 (en) 2010-05-17 2014-04-08 Emd Millipore Corporation Stimulus responsive polymers for the purification of biomolecules
US8772459B2 (en) 2009-12-02 2014-07-08 Imaginab, Inc. J591 minibodies and Cys-diabodies for targeting human prostate specific membrane antigen (PSMA) and methods for their use
US8940871B2 (en) 2006-03-20 2015-01-27 The Regents Of The University Of California Engineered anti-prostate stem cell antigen (PSCA) antibodies for cancer targeting
US8940298B2 (en) 2007-09-04 2015-01-27 The Regents Of The University Of California High affinity anti-prostate stem cell antigen (PSCA) antibodies for cancer targeting and detection
US8951737B2 (en) 1996-05-06 2015-02-10 Cornell Research Foundation, Inc. Treatment and diagnosis of cancer
US8999702B2 (en) 2008-06-11 2015-04-07 Emd Millipore Corporation Stirred tank bioreactor
US9085622B2 (en) 2010-09-03 2015-07-21 Glaxosmithkline Intellectual Property Development Limited Antigen binding proteins
US9090930B2 (en) 2006-06-27 2015-07-28 Emd Millipore Corporation Method and unit for preparing a sample for the microbiological analysis of a liquid
US9926341B2 (en) 2012-12-20 2018-03-27 Merck Patent Gmbh Copolymers for protein precipitation
US10517969B2 (en) 2009-02-17 2019-12-31 Cornell University Methods and kits for diagnosis of cancer and prediction of therapeutic value
US10793593B2 (en) 2006-12-21 2020-10-06 Emd Millipore Corporation Purification of proteins
US11254744B2 (en) 2015-08-07 2022-02-22 Imaginab, Inc. Antigen binding constructs to target molecules
US11266745B2 (en) 2017-02-08 2022-03-08 Imaginab, Inc. Extension sequences for diabodies

Families Citing this family (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008091740A2 (en) * 2007-01-22 2008-07-31 Genentech, Inc. Polyelectrolyte precipitation and purification of antibodies
JP5053045B2 (en) * 2007-03-30 2012-10-17 富士フイルム株式会社 Ink composition, printed matter, and inkjet recording method
WO2009067666A2 (en) 2007-11-21 2009-05-28 Tennessee Technological University Thermoresponsive microparticle composite hydrogels for electrophoresis
GB0818228D0 (en) 2008-10-06 2008-11-12 Avecia Biolog Ltd Purification process
WO2010043703A1 (en) * 2008-10-17 2010-04-22 Dsm Ip Assets B.V. Removal of host cell proteins
CA2738499A1 (en) * 2008-10-20 2010-04-29 Abbott Laboratories Viral inactivation during purification of antibodies
TW201024318A (en) * 2008-10-20 2010-07-01 Abbott Lab Isolation and purification of antibodies using protein A affinity chromatography
CA2739455A1 (en) * 2008-10-20 2010-04-29 Abbott Laboratories Antibodies that bind to il-12 and methods of purifying the same
US20110020327A1 (en) * 2008-12-16 2011-01-27 Millipore Corporation Purification of proteins
WO2012030512A1 (en) 2010-09-03 2012-03-08 Percivia Llc. Flow-through protein purification process
EP2578286A1 (en) 2011-10-04 2013-04-10 Merck Patent GmbH Method and apparatus for chromatographic purification
CN102492027A (en) * 2011-12-09 2012-06-13 福建省农业科学院农业生物资源研究所 Method for extracting Bt insecticidal protein crystal
DK2791176T3 (en) * 2011-12-15 2018-10-15 Prestige Biopharma Pte Ltd PROCEDURE FOR CLEANING ANTIBODIES
EP2656892A1 (en) 2012-04-23 2013-10-30 Merck Patent GmbH Chromatography method
CN102671637B (en) * 2012-05-16 2013-10-30 华南理工大学 Biomimetic specific immune adsorption material with PAMAM (Polyamidoamine) as spacer arm, and preparation method and application thereof
CN102731417B (en) * 2012-06-28 2014-07-30 华南理工大学 Dendrimer with alkynyl core and outer amino acid shell, and Huisgen 1,3-dipolar cycloaddition synthetic method and application thereof
CN104492395B (en) * 2014-11-28 2016-10-05 珠海健帆生物科技股份有限公司 Bionical immunoadsorbent with PAMAM as spacerarm and preparation method and application
WO2017022651A1 (en) * 2015-07-31 2017-02-09 中外製薬株式会社 Method for purifying composition comprising antibodies with anionic polymer
CN108435002B (en) * 2018-03-27 2021-03-19 东华大学 Preparation method of functionalized carbon quantum dot modified composite nanofiltration membrane
CN113004479B (en) * 2021-03-15 2022-07-08 吉林师范大学 Magnetic graphene oxide-based hydrophilic molecularly imprinted nanocomposite resin, and preparation method and application thereof

Citations (94)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3556302A (en) * 1969-01-06 1971-01-19 Amicon Corp Filtration apparatus having flow distributor
US3565973A (en) * 1967-11-14 1971-02-23 Amicon Corp Purifying cross-linked polyelectrolytes
US3632507A (en) * 1970-06-30 1972-01-04 Standard Brands Chem Ind Inc Flocculation of particles dispersed in aqueous media and flocculants used therein
US3702806A (en) * 1970-09-03 1972-11-14 William Emil Oliva Disposable culture media container
US3737377A (en) * 1971-01-27 1973-06-05 Miles Lab Purification of lactase
US3859212A (en) * 1971-10-12 1975-01-07 Allied Colloids Mfg Flocculating agents
US3968037A (en) * 1972-09-01 1976-07-06 Calgon Corporation Emulsion polymerization of cationic monomers
US4045377A (en) * 1975-10-20 1977-08-30 Hercules Incorporated Cationic polymer prepared from dicyandiamide, a polyamide, a dialkylamine, and an epoxide
US4055469A (en) * 1976-12-10 1977-10-25 Eastman Kodak Company Purification of microbial enzyme extracts using synthetic polyelectrolytes
US4200695A (en) * 1978-09-19 1980-04-29 Rohm And Haas Company Flocs for filtration and deionization prepared from cationic and anionic emulsion ion exchange resins
US4305829A (en) * 1979-06-29 1981-12-15 Union Carbide Corporation Process for flocculating an aqueous suspension of particles with quaternary ammonium graft copolymers
US4359537A (en) * 1978-09-19 1982-11-16 Rohm And Haas Company Emulsion copolymer anion exchange resins
US4371674A (en) * 1979-08-29 1983-02-01 Otto Hertel Water soluble crosslinked ethyleneimine grafted polyamidoamine
US4380590A (en) * 1978-09-19 1983-04-19 Rohm And Haas Company Emulsion copolymer cation exchange resins
US4382028A (en) * 1982-07-19 1983-05-03 Monsanto Company Separation of plasma proteins from cell culture systems
US4515893A (en) * 1979-04-26 1985-05-07 Ortho Pharmaceutical Corporation Hybrid cell line for producing complement-fixing monoclonal antibody to human T cells
US4536294A (en) * 1982-03-23 1985-08-20 Guillet James E Polymeric flocculants
US4676980A (en) * 1985-09-23 1987-06-30 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Target specific cross-linked heteroantibodies
US4780409A (en) * 1985-05-02 1988-10-25 Genetic Systems Corporation Thermally induced phase separation immunoassay
US4816567A (en) * 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4828701A (en) * 1983-08-25 1989-05-09 Regents Of The University Of Minnesota Temperature-sensitive method of size-selective extraction from solution
US4839046A (en) * 1987-08-20 1989-06-13 The United States Of America As Represented By The Administrator Of The National Aeronautics And Space Administration Bio-reactor chamber
US4863613A (en) * 1985-10-25 1989-09-05 Regents Of The University Of Minnesota Soy protein isolation process using swellable poly(N-isopropylacrylamide) gels
US4904385A (en) * 1985-05-23 1990-02-27 The Dow Chemical Company Porous filter media and membrane support means
US4912032A (en) * 1986-04-17 1990-03-27 Genetec Systems Corporation Methods for selectively reacting ligands immobilized within a temperature-sensitive polymer gel
US4925785A (en) * 1986-03-07 1990-05-15 Biotechnica Diagnostics, Inc. Nucleic acid hybridization assays
US4968435A (en) * 1988-12-19 1990-11-06 American Cyanamid Company Cross-linked cationic polymeric microparticles
US5003047A (en) * 1989-01-10 1991-03-26 Massachusetts Institute Of Technology Method for purifying biologically active ligate
US5047511A (en) * 1989-08-28 1991-09-10 Pitman-Moore, Inc. Method for recovering recombinant proteins
US5091313A (en) * 1988-08-05 1992-02-25 Tanox Biosystems, Inc. Antigenic epitopes of IgE present on B cell but not basophil surface
US5091178A (en) * 1986-02-21 1992-02-25 Oncogen Tumor therapy with biologically active anti-tumor antibodies
US5152903A (en) * 1988-12-19 1992-10-06 American Cyanamid Company Cross-linked cationic polymeric microparticles
US5164057A (en) * 1990-04-13 1992-11-17 W. R. Grace & Co.-Conn. Electrophoretic matrices and electrophoretic method using same
US5238545A (en) * 1991-02-27 1993-08-24 W. R. Grace & Co.-Conn. Electrophoretic gel for separation and recovery of substances and its use
US5258122A (en) * 1992-04-16 1993-11-02 Amicon, Inc. Cross-flow filter device with pressure-balancing feature
US5324787A (en) * 1992-11-18 1994-06-28 Air Products And Chemicals, Inc. Modification of poly (vinylamine)
US5340865A (en) * 1988-12-19 1994-08-23 Cytec Technology Corp. Cross-linked cationic polyermic microparticles
US5354481A (en) * 1988-12-19 1994-10-11 Cytec Technology Corp. Water-soluble highly branched polymeric microparticles
US5354801A (en) * 1993-08-12 1994-10-11 Cytec Technology Corp. Process for producing small polymer phase droplet microemulsions by multistep aqueous phase addition
US5430110A (en) * 1992-07-22 1995-07-04 Hoechst Aktiengesellschaft Polyvinylamine derivatives having hydrophilic centers, processes for their preparation and the use of the compounds as a medicament, active compound carrier and foodstuff auxiliary
US5512480A (en) * 1994-03-11 1996-04-30 Baxter International Inc. Flow-through bioreactor with grooves for cell retention
US5573675A (en) * 1995-05-11 1996-11-12 Nalco Chemical Company Clarification of deinking process waters using polymers containing vinylamine
US5599719A (en) * 1992-01-07 1997-02-04 Middlesex Sciences, Inc. Method for isolating biomolecules from a biological sample with linear polymers
US5622700A (en) * 1992-08-21 1997-04-22 Genentech, Inc. Method for treating a LFA-1-mediated disorder
US5672347A (en) * 1984-07-05 1997-09-30 Genentech, Inc. Tumor necrosis factor antagonists and their use
US5684107A (en) * 1991-02-09 1997-11-04 Basf Aktiengesellschaft Agglomerated polymer particles of finely divided, water-soluble or water-swellable polymers, the preparation thereof and the use thereof
US5693762A (en) * 1988-12-28 1997-12-02 Protein Design Labs, Inc. Humanized immunoglobulins
US5714338A (en) * 1993-12-10 1998-02-03 Genentech, Inc. Methods for diagnosis of allergy
US5721108A (en) * 1987-01-08 1998-02-24 Xoma Corporation Chimeric antibody with specificity to human B cell surface antigen
US5725856A (en) * 1988-01-12 1998-03-10 Genentech, Inc. Monoclonal antibodies directed to the HER2 receptor
US5736137A (en) * 1992-11-13 1998-04-07 Idec Pharmaceuticals Corporation Therapeutic application of chimeric and radiolabeled antibodies to human B lymphocyte restricted differentiation antigen for treatment of B cell lymphoma
US5739383A (en) * 1996-08-29 1998-04-14 Hoechst Celanese Corp. Resolution of racemic mixtures using polymers containing chiral units
US5807489A (en) * 1995-11-14 1998-09-15 Cytec Technology Corp. High performance polymer flocculating agents
US5840804A (en) * 1994-03-01 1998-11-24 Roehm Gmbh Chemische Fabrik Crosslinked water-soluble polymer dispersions
US5840851A (en) * 1993-07-23 1998-11-24 Plomer; J. Jeffrey Purification of hemoglobin
US5879564A (en) * 1995-11-14 1999-03-09 Cytec Technology Corp. High performance polymer flocculating agents
US5929214A (en) * 1997-02-28 1999-07-27 Cornell Research Foundation, Inc. Thermally responsive polymer monoliths
US5998588A (en) * 1995-09-01 1999-12-07 University Of Washington Interactive molecular conjugates
US6024955A (en) * 1995-11-01 2000-02-15 Toagosei Co. Ltd. Peptides and monoclonal antibodies
US6127526A (en) * 1996-11-27 2000-10-03 Genentech, Inc. Protein purification by Protein A chromatography
US6133047A (en) * 1996-05-24 2000-10-17 Bio Merieux Superparamagnetic monodisperse particles
US6139746A (en) * 1999-02-22 2000-10-31 Kopf; Henry B. Method and apparatus for purification of biological substances
US6147176A (en) * 1988-12-19 2000-11-14 Cytec Technology Corp. High performance polymer flocculating agents nonionic polymers
US6191242B1 (en) * 1988-12-19 2001-02-20 Cytec Technology Corp. Process for making high performance anionic polymeric flocculating agents
US6245555B1 (en) * 1998-09-01 2001-06-12 The Penn State Research Foundation Method and apparatus for aseptic growth or processing of biomass
US6258275B1 (en) * 1999-10-01 2001-07-10 Ecole Polytechnique Federale De Lausanne Affinity macroligands
US6294622B1 (en) * 1999-09-27 2001-09-25 Ecole Polytechnique Federale De Lausanne (Epfl) Polymer flocculants with improved dewatering characteristics
US6307013B1 (en) * 1998-07-21 2001-10-23 Monsanto Technology Llc Clarification of protein precipitate suspensions using anionic polymeric flocculants
US6638918B2 (en) * 2001-11-09 2003-10-28 The Procter & Gamble Company Chitosan compositions
US6641735B1 (en) * 2000-03-23 2003-11-04 Japan Chemical Innovation Institute Separatory material with the use of stimulus-responsive polymer and separation method by using the separatory material
US6706187B1 (en) * 1998-05-22 2004-03-16 Teruo Okano Packing material for chromatography having novel characteristic and method for isolation of substance using the same
US6821515B1 (en) * 1995-07-27 2004-11-23 Genentech, Inc. Protein formulation
US6858694B2 (en) * 1997-12-09 2005-02-22 Agency Of Industrial Science And Technology Miti Stimuli-responsive polymer utilizing keto-enol tautomerization and stimuli-responsive separating material and chemical-releasing capsule comprising the same
US20050063259A1 (en) * 2003-09-22 2005-03-24 Fumio Isshiki Optical information recording apparatus
US6926832B2 (en) * 2002-01-04 2005-08-09 Nalco Company Method of using water soluble polymers in a membrane biological reactor
US20050224415A1 (en) * 1999-01-29 2005-10-13 Amersham Biosciences Kk Temperature-responsive polymer compound and process for producing the same
US6967085B1 (en) * 1999-08-14 2005-11-22 Ciba Specialty Chemicals Water Treatments Ltd. Flocculation of cell material
US20050282169A1 (en) * 2004-01-29 2005-12-22 Turner Allen C Signatory sequences
US7001953B2 (en) * 2001-04-16 2006-02-21 Wsp Chemicals & Technology, Llc Water-soluble polymer complexes
US20060121519A1 (en) * 2003-07-24 2006-06-08 Affisink Biotechnology Ltd. Compositions and methods for purifying and crystallizing molecules of interest
US7070696B2 (en) * 2001-04-05 2006-07-04 Ciba Specialty Chemicals Water Treatments Ltd. Process for flocculating suspensions
US20060162882A1 (en) * 2002-06-21 2006-07-27 Takumi Ohara Water-soluble polymer dispersion, process for producing the same and method of use therefor
US7160971B2 (en) * 2002-02-15 2007-01-09 Iowa State University Research Foundation pH-sensitive methacrylic copolymers and the production thereof
US7169908B2 (en) * 2003-01-09 2007-01-30 Genentech, Inc. Purification of polypeptides
US7377686B2 (en) * 2003-09-04 2008-05-27 Millipore Corporation Disposable mixing system
US20080193981A1 (en) * 2007-01-22 2008-08-14 Fahrner Robert L Polyelectrolyte precipitation and purification of proteins
US7547747B2 (en) * 2003-04-01 2009-06-16 Nitto Boseki Co., Ltd. Modified polyallylamine and process for producing the same
US20090155201A1 (en) * 1994-06-10 2009-06-18 Genzyme Corporation Alkylated poly(allylamine) polymers and methods of use
US7767399B2 (en) * 2005-01-31 2010-08-03 Merck & Co., Inc. Purification process for plasmid DNA
US20100193148A1 (en) * 2009-01-30 2010-08-05 Mckay Jonathan M Quaternary Vinylamine-Containing Polymers as Additives in Papermaking
US20100282425A1 (en) * 2006-04-24 2010-11-11 Asko Karppi Cationic polysaccharide, its preparation and use
US20110020327A1 (en) * 2008-12-16 2011-01-27 Millipore Corporation Purification of proteins
US20110313066A1 (en) * 2010-05-17 2011-12-22 Millipore Corporation Stimulus responsive polymers for the purification of biomolecules
US20120070836A1 (en) * 2010-06-08 2012-03-22 Millipore Corporation Methods of detecting residual amounts of polymers used in the purification of biomolecules

Family Cites Families (178)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2923669A (en) 1954-11-22 1960-02-02 Millipore Filter Corp Method of bacterial analysis
US3211645A (en) 1962-07-17 1965-10-12 Technicon Instr Method and apparatus for filtering sanguineous liquid streams
US4215198A (en) 1978-09-15 1980-07-29 Gordon Maurice R Sterility testing unit
US4317726A (en) 1981-02-12 1982-03-02 The United States Of America As Represented By The Secretary Of The Army Microbial filter assembly
DE3145320A1 (en) 1981-11-14 1983-05-26 Biotest-Serum-Institut Gmbh, 6000 Frankfurt MICROFILTRATION DEVICE FOR FILTRATION OF COAGELS AND MICROAGGREGATES OF BLOOD
US4528933A (en) 1983-05-11 1985-07-16 Robert Allen Container with indicating closure
US4533496A (en) 1984-05-08 1985-08-06 Monsanto Company Method of isolating monoclonal antibodies from hybridoma cultures
DE3879209D1 (en) 1987-08-27 1993-04-15 Polyfiltronics Ltd FILTER UNITS FOR THE PREPARATION OF BIOLOGICAL SAMPLES.
US5429952A (en) 1988-02-02 1995-07-04 Biocode, Inc. Marking of products to establish identity and source
DE68919361T2 (en) 1988-06-21 1995-05-24 Genentech Inc THERAPEUTIC COMPOSITIONS FOR THE TREATMENT OF MYOCARD INFARTS.
US5139031A (en) 1989-09-18 1992-08-18 La Mina Ltd. Method and device for cytology and microbiological testing
ATE144793T1 (en) 1989-06-29 1996-11-15 Medarex Inc BISPECIFIC REAGENTS FOR AIDS THERAPY
JPH046463A (en) 1990-04-16 1992-01-10 W R Grace & Co Carrier for liquid chromatography and liquid chromatographic method using said carrier
DE69132709T2 (en) 1990-06-29 2002-06-20 Large Scale Biology Corp MELANINE PRODUCTION BY TRANSFORMED MICROORGANISMS
US5116754A (en) 1990-10-04 1992-05-26 Fraser Ann D E Separation of bacteria from organic matter
US5171450A (en) 1991-03-20 1992-12-15 Nalco Chemical Company Monitoring and dosage control of tagged polymers in cooling water systems
WO1992020373A1 (en) 1991-05-14 1992-11-26 Repligen Corporation Heteroconjugate antibodies for treatment of hiv infection
WO1993004173A1 (en) 1991-08-14 1993-03-04 Genentech, Inc. Immunoglobulin variants for specific fc epsilon receptors
AU669489B2 (en) 1991-09-18 1996-06-13 Affymax Technologies N.V. Method of synthesizing diverse collections of oligomers
WO1993008829A1 (en) 1991-11-04 1993-05-13 The Regents Of The University Of California Compositions that mediate killing of hiv-infected cells
EP1997894B1 (en) 1992-02-06 2011-03-30 Novartis Vaccines and Diagnostics, Inc. Biosynthetic binding protein for cancer marker
JP3253730B2 (en) 1992-08-21 2002-02-04 科学技術振興事業団 Temperature-responsive physiologically active substance-oligomer complex and method for producing the same
SE9300090D0 (en) 1993-01-14 1993-01-14 Bo Gustav Mattiasson AFFINITY CLEANING WITH COMPLEX BOND LIGAND
US5374971A (en) 1993-03-12 1994-12-20 Picturetel Corporation Two-view video camera stand and support method
US5342581A (en) 1993-04-19 1994-08-30 Sanadi Ashok R Apparatus for preventing cross-contamination of multi-well test plates
DK83093D0 (en) 1993-07-09 1993-07-09 Novo Nordisk As COURSE OF ACTION
WO1995019181A1 (en) 1994-01-18 1995-07-20 Genentech, Inc. A METHOD OF TREATMENT OF PARASITIC INFECTION USING IgE ANTAGONISTS
US5622857A (en) 1995-08-08 1997-04-22 Genespan Corporation High performance cell culture bioreactor and method
WO1995023865A1 (en) 1994-03-03 1995-09-08 Genentech, Inc. Anti-il-8 monoclonal antibodies for treatment of inflammatory disorders
WO1996002577A1 (en) 1994-07-18 1996-02-01 Gel Sciences, Inc. Responsive polymer gel beads
CA2157219C (en) 1994-08-31 2010-10-05 Munehiro Noda Process for purifying recombinant human serum albumin
GB9422504D0 (en) 1994-11-08 1995-01-04 Robertson Patricia M B Blood testing
AUPN060095A0 (en) 1995-01-13 1995-02-09 Enviro Research Pty Ltd Apparatus for biomass production
GB9503109D0 (en) 1995-02-17 1995-04-05 Hampshire Advisory Tech Serv Diagnostic test tube and kits
US5731168A (en) 1995-03-01 1998-03-24 Genentech, Inc. Method for making heteromultimeric polypeptides
US5939485A (en) 1995-06-19 1999-08-17 Medlogic Global Corporation Responsive polymer networks and methods of their use
IL117645A (en) 1995-03-30 2005-08-31 Genentech Inc Vascular endothelial cell growth factor antagonists for use as medicaments in the treatment of age-related macular degeneration
GB9510634D0 (en) 1995-05-25 1995-07-19 Sev Trent Water Ltd Filtration and culture methods and apparatus
US5733507A (en) 1995-06-07 1998-03-31 Inphocyte, Inc. Biological cell sample holder for use in infrared and/or Raman spectroscopy analysis holder
JPH11507535A (en) 1995-06-07 1999-07-06 イムクローン システムズ インコーポレイテッド Antibodies and antibody fragments that suppress tumor growth
JPH0912598A (en) 1995-06-29 1997-01-14 Sekisui Chem Co Ltd Separation and purification of hemoglobin
GB2305936B (en) 1995-10-06 1997-09-03 Jonathan William Lewis Sterile, disposable culture vessels for the rapid growth of bacteria
ZA97248B (en) 1996-01-18 1997-07-18 Rohm & Haas Method for identifying and quantifying polymers utilizing immunoassay techniques
DE69714966T2 (en) 1996-01-23 2003-04-24 Genentech Inc ANTIBODIES AGAINST CD 18 FOR USE AGAINST BRAIN SHOCK
EP0842184B1 (en) 1996-03-20 2003-01-29 Bio Merieux Nucleic acid isolation
US7147851B1 (en) 1996-08-15 2006-12-12 Millennium Pharmaceuticals, Inc. Humanized immunoglobulin reactive with α4β7 integrin
DE69729209T2 (en) 1996-11-27 2005-05-19 Genentech, Inc., South San Francisco HUMANIZED ANTI-KOERPER AGAINST CD11A
US6372141B1 (en) 1997-01-24 2002-04-16 Amersham Pharmacia Biotech K.K. Method for separating PTH amino acids
US5942444A (en) 1997-01-27 1999-08-24 Biocode, Inc. Marking of products to establish identity, source and fate
JP4040107B2 (en) 1997-01-29 2008-01-30 ポール・コーポレーション Filtration assembly
WO1998045331A2 (en) 1997-04-07 1998-10-15 Genentech, Inc. Anti-vegf antibodies
PT1860187E (en) 1997-05-15 2011-10-04 Genentech Inc Apo-2 receptor
WO1998056808A1 (en) 1997-06-13 1998-12-17 Genentech, Inc. Protein recovery by chromatography followed by filtration upon a charged layer
US5994511A (en) 1997-07-02 1999-11-30 Genentech, Inc. Anti-IgE antibodies and methods of improving polypeptides
US6007803A (en) 1997-09-19 1999-12-28 Geltex Pharmaceuticals, Inc. Ionic polymers as toxin binding agents
NZ504768A (en) 1997-11-28 2002-11-26 Provalis Diagnostics Ltd Device and apparatus for conducting an assay
US6914137B2 (en) * 1997-12-06 2005-07-05 Dna Research Innovations Limited Isolation of nucleic acids
JP4217804B2 (en) 1998-09-14 2009-02-04 独立行政法人産業技術総合研究所 Thermally responsive separation material and drug release capsule using thermoresponsive polymer derivative having upper limit solution critical temperature
FR2773416B1 (en) 1998-01-06 2000-02-11 Bio Merieux IMPROVED MAGNETIC PARTICLES, PROCESSES FOR OBTAINING SAME AND THEIR USES IN THE SEPARATION OF MOLECULES
US6756217B1 (en) 1998-05-29 2004-06-29 Southern Illinois University Glass composite materials containing alkoxosilane derivative having alterable charge, hydrophobic and hydrophilic groups
US6221655B1 (en) 1998-08-01 2001-04-24 Cytosignal Spin filter assembly for isolation and analysis
SE9802882D0 (en) 1998-08-28 1998-08-28 Amersham Pharm Biotech Ab Composite material and its use
AU1218000A (en) 1998-10-21 2000-05-08 Sunol Molecular Corporation Polyspecific binding molecules and uses thereof
FR2788008B1 (en) 1998-12-30 2001-03-23 Inst Curie THERMOSENSITIVE MEDIUM FOR THE ELECTROKINETIC SEPARATION OF SPECIES WITHIN A SEPARATION CHANNEL
CA2356704A1 (en) 1998-12-30 2000-07-13 Folke Tjerneld Separation method utilizing liquid-liquid partition
WO2000040974A1 (en) 1999-01-07 2000-07-13 Quidel Corporation Pyridinium crosslinks assay
SE9900378D0 (en) 1999-02-05 1999-02-05 Forskarpatent I Syd Ab Gels with shape memory
US6565872B2 (en) 1999-02-16 2003-05-20 Xiao Yu Wu Polymeric system for drug delivery and solute separation
WO2000056774A1 (en) 1999-03-19 2000-09-28 Duke University Methods of using bioelastomers
JP2003524680A (en) 1999-05-11 2003-08-19 財団法人化学技術戦略推進機構 Affinity control type material using stimulus-responsive polymer and separation / purification method using the material
US6946129B1 (en) 1999-06-08 2005-09-20 Seattle Genetics, Inc. Recombinant anti-CD40 antibody and uses thereof
US6211140B1 (en) 1999-07-26 2001-04-03 The Procter & Gamble Company Cationic charge boosting systems
DE60022762T2 (en) 1999-07-29 2006-07-13 National Institute Of Advanced Industrial Science And Technology POLYMERIZABLE BIOTIN DERIVATIVES, BIOPTIN POLYMER AND POLYMER REACTIVE ON STIMULATION BY AVIDIN
US6420487B1 (en) 1999-09-08 2002-07-16 Council Of Scientific And Industrial Research Process for the preparation of thermoprecipitating affinity polymers
DK1226177T3 (en) 1999-10-29 2008-10-06 Genentech Inc Antibody Compositions for Anti-Prostate Stem Cell Antigen (PSCA) and Applications thereof
FR2804117B1 (en) 2000-01-21 2004-08-20 Bio Merieux PROCESS FOR ISOLATING PROTEINS AND / OR NUCLEIC ACIDS, PARTICLE COMPLEXES AND PROTEINS AND / OR NUCLEIC ACIDS, REAGENT AND APPLICATIONS
AU2001246843A1 (en) 2000-04-05 2001-10-15 Japan Chemical Innovation Institute Novel material for use in separation and separating method using the same
US6372145B1 (en) 2000-06-05 2002-04-16 Power Engineering Company Fat and protein removal from process streams
JP4887530B2 (en) 2000-08-21 2012-02-29 独立行政法人産業技術総合研究所 Magnetic fine particles and method for producing the same
WO2002016571A1 (en) 2000-08-21 2002-02-28 National Institute Of Advanced Industrial Science And Technology Magnetic particles having lower limit critical solution temperature
WO2002016454A1 (en) 2000-08-21 2002-02-28 National Institute Of Advanced Industrial Science And Technology Polymers
US6863437B2 (en) 2000-08-23 2005-03-08 National Institute Of Advanced Industrial Science And Technology Temperature-responsive polymer/polymer complex
US6374684B1 (en) 2000-08-25 2002-04-23 Cepheid Fluid control and processing system
ES2320736T3 (en) 2000-09-06 2009-05-28 University Of Massachusetts EXTRACTION OF HIGH EFFICIENCY OF PROTEINS.
JP5109003B2 (en) 2000-10-13 2012-12-26 岡野 光夫 Separation material such as stimulus-responsive affinity chromatography material and separation purification method
US6605714B2 (en) 2000-11-29 2003-08-12 Council Of Scientific And Industrial Research Thermoprecipitating polymer containing enzyme specific ligands, process for the preparation thereof, and use thereof for the separation of enzymes
US6367749B2 (en) 2001-03-21 2002-04-09 Southern Imperial, Inc. Stand base
US6688487B2 (en) 2001-04-13 2004-02-10 The Coca-Cola Company Locking cup and lid with negative draft sealing surfaces
US7157603B2 (en) 2001-05-14 2007-01-02 Polytag Technology Sa Method for the separation of oligomeric N-substituted (meth)acrylamide compounds and conjugates thereof which are reversibly thermally precipitating
JP3907508B2 (en) 2001-07-30 2007-04-18 松下エコシステムズ株式会社 Microorganism collection chip, microorganism collection kit, microorganism measurement method, and microorganism measurement apparatus
FR2829500B1 (en) 2001-09-13 2003-12-12 Hemosystem PROCESS FOR THE CONCENTRATION AND DETECTION OF PATHOGENIC SPROUTS FROM BLOOD PRODUCTS AND / OR DERIVATIVES THEREOF AND DEVICE FOR CARRYING OUT SAID METHOD
US6541124B1 (en) 2001-11-13 2003-04-01 Rhino Metals, Inc. Drill resistant hard plate
TWI288758B (en) 2001-12-19 2007-10-21 Ind Tech Res Inst Thermal responsive, water-soluble polymers
CA2473376A1 (en) 2002-01-16 2003-07-31 Dynal Biotech Asa Method for isolating nucleic acids and protein from a single sample
WO2003074571A2 (en) 2002-02-28 2003-09-12 Keck Graduate Institute Cationic polyelectrolytes in biomolecule purification and analysis
WO2003078947A2 (en) 2002-03-15 2003-09-25 The Penn State Research Foundation Method for control of temperature-sensitivity of polymers in solution
DK1499382T3 (en) 2002-04-26 2007-05-07 Gl Tool And Mfg Co Inc A valve
DE10224352A1 (en) 2002-06-01 2003-12-11 Mueller Schulte Detlef Thermosensitive polymer carrier with changeable physical structure for biochemical analysis, diagnostics and therapy
US20040009473A1 (en) 2002-06-07 2004-01-15 Christopher Pease Kit and process for microbiological for on-site examination of a liquid sample
AU2003259289A1 (en) 2002-07-30 2004-02-16 University Of Washington Apparatus and methods for binding molecules and cells
US7422724B1 (en) 2002-08-07 2008-09-09 Sandia Corporation Biological preconcentrator
SE0202552D0 (en) 2002-08-27 2002-08-27 Amersham Biosciences Ab Recovery of plasmids in an aqueous two-phase system
CN1300297C (en) 2002-09-20 2007-02-14 华东理工大学 Digestive reactor for animal cell extension inoculation
US6837610B2 (en) 2002-09-27 2005-01-04 Ilc Dover Lpp Bioprocess container, bioprocess container mixing device and method of use thereof
US6673598B1 (en) 2002-10-29 2004-01-06 Synthecon, Inc. Disposable culture bag
BRPI0316779B8 (en) 2002-12-16 2023-02-28 Genentech Inc HUMAN ANTI-CD20 ANTIBODY OR ANTIGEN-BINDING FRAGMENT THEREOF, ITS USES, COMPOSITION, MANUFACTURED ARTICLE AND LIQUID FORMULATION
AU2003299873A1 (en) 2002-12-23 2004-07-22 Univ Rice William M Methods of detecting the inhibition of fibrocyte formation and methods and compositions for enhancing fibrocyte formation
US7083948B1 (en) 2002-12-24 2006-08-01 Immunex Corporation Polypeptide purification reagents and methods for their use
TWI257928B (en) 2002-12-27 2006-07-11 Ind Tech Res Inst Method for the separation of polysaccharides
CA2515213A1 (en) 2003-02-11 2004-08-26 University Of Washington Stimuli-responsive polymer conjugates and related methods
JP5362943B2 (en) 2003-03-04 2013-12-11 株式会社セルシード Drug metabolic capacity evaluation system and method of using the same
SE0300791D0 (en) 2003-03-20 2003-03-20 Amersham Biosciences Ab Use of ph-responsive polymers
US6949625B2 (en) 2003-05-12 2005-09-27 Khorionyx Injectable implant of insoluble globin
WO2006085321A2 (en) 2005-02-10 2006-08-17 Affisink Biotechnology Ltd. Compositions and methods for purifying and crystallizing molecules of interest
CA2560901C (en) 2003-08-29 2012-08-21 The University Of Newcastle Research Associates Limited Stimulant sensitive flocculation and consolidation
DE10350248A1 (en) 2003-10-28 2005-06-16 Magnamedics Gmbh Thermosensitive, biocompatible polymer carriers with variable physical structure for therapy, diagnostics and analytics
US20090130704A1 (en) 2003-11-13 2009-05-21 Gyure Dale C Novel bioreactor
US7875448B2 (en) 2004-01-12 2011-01-25 Single Use Brx, Llc Bioreactor systems and disposable bioreactor
US8105849B2 (en) 2004-02-27 2012-01-31 Board Of Regents, The University Of Texas System Integration of fluids and reagents into self-contained cartridges containing sensor elements
WO2005090272A1 (en) 2004-03-12 2005-09-29 University Of Utah Cyclone reactor and associated methods
SE0400916D0 (en) 2004-04-05 2004-04-05 Amersham Biosciences Ab Polymeric ligands
BRPI0510291A (en) 2004-04-27 2007-10-30 Baxter Int stirred tank reactor system
AU2005250500B2 (en) 2004-06-04 2011-06-23 Global Life Sciences Solutions Usa Llc Disposable bioreactor systems and methods
US8288169B2 (en) 2005-01-21 2012-10-16 Argylla Technologies Surface mediated self-assembly of nanoparticles
US8603805B2 (en) 2005-04-22 2013-12-10 Hyclone Laboratories, Inc. Gas spargers and related container systems
JP2006312117A (en) 2005-05-06 2006-11-16 Canon Inc Separation material for physiologically active substance and its production method
WO2006135673A2 (en) 2005-06-10 2006-12-21 Nanologix, Inc. Production of hydrogen gas and isolation of hydrogen producing microorganisms using replenishing coated substrates
WO2006138143A1 (en) 2005-06-15 2006-12-28 Amprotein Corporation Suspension culture vessels
WO2007002690A2 (en) 2005-06-24 2007-01-04 William Marsh Rice University Nano-encapsulated triggered-release viscosity breakers
NL1030538C1 (en) 2005-11-28 2007-05-30 Eurocore Trading & Consultancy Device for indirectly cooling an air stream through evaporation.
CN101296742A (en) 2005-08-26 2008-10-29 最佳空气控股公司 Method and device for separating a substance from a process gas
JP4942753B2 (en) 2005-09-27 2012-05-30 センター フォア アプライド プロテオミクス アンド モレキュラー メディスン How to isolate an analyte from a sample
US20070095666A1 (en) 2005-10-27 2007-05-03 Applera Corporation Surface Modification in a Manipulation Chamber
CA2632328A1 (en) 2005-12-22 2007-06-28 Ge Healthcare Bio-Sciences Ab Preparation of biomolecules
EP1832341A1 (en) 2006-03-10 2007-09-12 MPG Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. Highly efficient desalination and ion exchange using a thermoreversible polymer
US7718193B2 (en) 2006-03-16 2010-05-18 University Of Washington Temperature- and pH-responsive polymer compositions
WO2007148230A2 (en) 2006-04-14 2007-12-27 Interface Biologics Incorporated Grafted polymers and uses thereof
EP1873205A1 (en) 2006-06-12 2008-01-02 Corning Incorporated Thermo-responsive blends and uses thereof
FR2902799B1 (en) 2006-06-27 2012-10-26 Millipore Corp METHOD AND UNIT FOR PREPARING A SAMPLE FOR THE MICROBIOLOGICAL ANALYSIS OF A LIQUID
WO2008004988A1 (en) 2006-07-06 2008-01-10 Agency For Science, Technology And Research Thermally responsive micelles
US20080032396A1 (en) 2006-08-02 2008-02-07 Becton, Dickinson And Company Bioreactor and Method
US7935518B2 (en) 2006-09-27 2011-05-03 Alessandra Luchini Smart hydrogel particles for biomarker harvesting
EP1923461A1 (en) 2006-11-15 2008-05-21 Millipore Corporation A bioreactor
US8057092B2 (en) 2006-11-30 2011-11-15 Corning Incorporated Disposable spinner flask
US8569464B2 (en) 2006-12-21 2013-10-29 Emd Millipore Corporation Purification of proteins
WO2008079280A1 (en) 2006-12-21 2008-07-03 Millipore Corporation Purification of proteins
US8362217B2 (en) 2006-12-21 2013-01-29 Emd Millipore Corporation Purification of proteins
US8137559B2 (en) 2007-02-09 2012-03-20 Ge Healthcare Bio-Sciences Ab Liquid clarification
US7981688B2 (en) 2007-03-08 2011-07-19 University Of Washington Stimuli-responsive magnetic nanoparticles and related methods
US20080284163A1 (en) 2007-05-15 2008-11-20 Millipore Corporation Connector for flexible tubing
EP2152405B2 (en) 2007-05-25 2017-04-05 Merck Patent GmbH Graft copolymer for cation-exchange chromatography
US8105493B2 (en) 2007-06-29 2012-01-31 Jnc Corporation Aggregation and dispersion methods of magnetic particles, separation and detection methods using the same and detection kit
US9433922B2 (en) 2007-08-14 2016-09-06 Emd Millipore Corporation Media for membrane ion exchange chromatography based on polymeric primary amines, sorption device containing that media, and chromatography scheme and purification method using the same
WO2009047587A1 (en) 2007-10-12 2009-04-16 Ecole Polytechnique Federale De Lausanne (Epfl) Magnetic, paramagnetic and/or superparamagnetic nanoparticles
CA2712104A1 (en) 2008-01-14 2009-07-23 The University Of Melbourne Flotation aids and processes for using the same
AU2008354927B2 (en) 2008-04-18 2012-10-04 S.P.C.M. Sa Functionalized cationic polyamines and their use to reduce the NDMA formation during the treatment of aqueous systems, and applications in the water treatment industry, including wastewater and drinking water treatment processes
GB0809404D0 (en) 2008-05-23 2008-07-02 Univ Sheffield Method
WO2009145722A1 (en) 2008-05-30 2009-12-03 Ge Healthcare Bio-Sciences Ab Separation method utilizing polyallylamine ligands
US8999702B2 (en) 2008-06-11 2015-04-07 Emd Millipore Corporation Stirred tank bioreactor
EP2307350A4 (en) 2008-06-27 2013-12-04 Stc Unm Structure, synthesis, and applications for oligo phenylene ethynylenes
FR2934049B1 (en) 2008-07-16 2010-10-15 Millipore Corp UNIT AND METHOD FOR PREPARING A SAMPLE FOR THE MICROBIOLOGICAL ANALYSIS OF A LIQUID
AU2008221604B2 (en) 2008-09-22 2010-04-22 Commonwealth Scientific And Industrial Research Organisation Temperature-responsive polymer particles in protein separation applications
SG171446A1 (en) 2008-12-16 2011-07-28 Millipore Corp Stirred tank reactor and method
CN102272146A (en) 2009-01-13 2011-12-07 通用电气健康护理生物科学股份公司 Precipitation of biomolecules with negatively charged polymers
US9080933B2 (en) 2009-11-09 2015-07-14 University Of Washington Through Its Center For Commercialization Stimuli-responsive polymer diagnostic assay comprising magnetic nanoparticles and capture conjugates
US20120077249A1 (en) 2010-04-20 2012-03-29 Millipore Corporation Separation Of Virus And/Or Protein From Nucleic Acids By Primary Amines
US20120283419A1 (en) 2011-05-03 2012-11-08 Avantor Performance Materials, Inc. Separation of protein monomers from aggregates by solid weak anion exchange support functionalized with amine moieties
JP6138116B2 (en) 2011-05-03 2017-05-31 アバンター・パフォーマンス・マテリアルズ・インコーポレイテッドAvantor Performance Materials, Inc. Novel chromatographic media based on allylamine and allylamine derivatives for protein purification
US20150239956A1 (en) 2012-06-27 2015-08-27 Asahi Kasei Medical Co., Ltd. High-affinity antibody and method for manufacturing the same
EP2682168A1 (en) 2012-07-02 2014-01-08 Millipore Corporation Purification of biological molecules
US20150218208A1 (en) 2012-08-27 2015-08-06 Asahi Kasei Medical Co., Ltd. Method for purifying antibody by temperature-responsive chromatography
AR093330A1 (en) 2012-11-01 2015-06-03 Novozymes As METHOD FOR DNA REMOVAL
JP6255034B2 (en) 2012-12-11 2017-12-27 キアゲン ゲーエムベーハー Preparation of silica particles
JP6410734B2 (en) 2013-02-06 2018-10-24 エイジェンシー・フォー・サイエンス,テクノロジー・アンド・リサーチ Method for reducing the aggregate content of a protein preparation
CN105026418B (en) 2013-02-06 2019-01-11 新加坡科技研究局 Method of purifying protein
CN105008383A (en) 2013-02-26 2015-10-28 新加坡科技研究局 Protein purification in the presence of nonionic organic polymers and electropositive surfaces
CN105189530B (en) 2013-02-28 2019-06-07 新加坡科技研究局 The chromatogram purification antibody from shortage chromatinic cell culture harvest object
CN105008384A (en) 2013-02-28 2015-10-28 新加坡科技研究局 Protein purification in the presence of nonionic organic polymers at elevated conductivity

Patent Citations (99)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3565973A (en) * 1967-11-14 1971-02-23 Amicon Corp Purifying cross-linked polyelectrolytes
US3556302A (en) * 1969-01-06 1971-01-19 Amicon Corp Filtration apparatus having flow distributor
US3632507A (en) * 1970-06-30 1972-01-04 Standard Brands Chem Ind Inc Flocculation of particles dispersed in aqueous media and flocculants used therein
US3702806A (en) * 1970-09-03 1972-11-14 William Emil Oliva Disposable culture media container
US3737377A (en) * 1971-01-27 1973-06-05 Miles Lab Purification of lactase
US3859212A (en) * 1971-10-12 1975-01-07 Allied Colloids Mfg Flocculating agents
US3968037A (en) * 1972-09-01 1976-07-06 Calgon Corporation Emulsion polymerization of cationic monomers
US4045377A (en) * 1975-10-20 1977-08-30 Hercules Incorporated Cationic polymer prepared from dicyandiamide, a polyamide, a dialkylamine, and an epoxide
US4055469A (en) * 1976-12-10 1977-10-25 Eastman Kodak Company Purification of microbial enzyme extracts using synthetic polyelectrolytes
US4200695A (en) * 1978-09-19 1980-04-29 Rohm And Haas Company Flocs for filtration and deionization prepared from cationic and anionic emulsion ion exchange resins
US4359537A (en) * 1978-09-19 1982-11-16 Rohm And Haas Company Emulsion copolymer anion exchange resins
US4380590A (en) * 1978-09-19 1983-04-19 Rohm And Haas Company Emulsion copolymer cation exchange resins
US4515893A (en) * 1979-04-26 1985-05-07 Ortho Pharmaceutical Corporation Hybrid cell line for producing complement-fixing monoclonal antibody to human T cells
US4305829A (en) * 1979-06-29 1981-12-15 Union Carbide Corporation Process for flocculating an aqueous suspension of particles with quaternary ammonium graft copolymers
US4371674A (en) * 1979-08-29 1983-02-01 Otto Hertel Water soluble crosslinked ethyleneimine grafted polyamidoamine
US4536294A (en) * 1982-03-23 1985-08-20 Guillet James E Polymeric flocculants
US4382028A (en) * 1982-07-19 1983-05-03 Monsanto Company Separation of plasma proteins from cell culture systems
US4816567A (en) * 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4828701A (en) * 1983-08-25 1989-05-09 Regents Of The University Of Minnesota Temperature-sensitive method of size-selective extraction from solution
US5672347A (en) * 1984-07-05 1997-09-30 Genentech, Inc. Tumor necrosis factor antagonists and their use
US4780409A (en) * 1985-05-02 1988-10-25 Genetic Systems Corporation Thermally induced phase separation immunoassay
US4904385A (en) * 1985-05-23 1990-02-27 The Dow Chemical Company Porous filter media and membrane support means
US4676980A (en) * 1985-09-23 1987-06-30 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Target specific cross-linked heteroantibodies
US4863613A (en) * 1985-10-25 1989-09-05 Regents Of The University Of Minnesota Soy protein isolation process using swellable poly(N-isopropylacrylamide) gels
US5091178A (en) * 1986-02-21 1992-02-25 Oncogen Tumor therapy with biologically active anti-tumor antibodies
US4925785A (en) * 1986-03-07 1990-05-15 Biotechnica Diagnostics, Inc. Nucleic acid hybridization assays
US4912032A (en) * 1986-04-17 1990-03-27 Genetec Systems Corporation Methods for selectively reacting ligands immobilized within a temperature-sensitive polymer gel
US5721108A (en) * 1987-01-08 1998-02-24 Xoma Corporation Chimeric antibody with specificity to human B cell surface antigen
US4839046A (en) * 1987-08-20 1989-06-13 The United States Of America As Represented By The Administrator Of The National Aeronautics And Space Administration Bio-reactor chamber
US5725856A (en) * 1988-01-12 1998-03-10 Genentech, Inc. Monoclonal antibodies directed to the HER2 receptor
US5091313A (en) * 1988-08-05 1992-02-25 Tanox Biosystems, Inc. Antigenic epitopes of IgE present on B cell but not basophil surface
US5152903A (en) * 1988-12-19 1992-10-06 American Cyanamid Company Cross-linked cationic polymeric microparticles
US6191242B1 (en) * 1988-12-19 2001-02-20 Cytec Technology Corp. Process for making high performance anionic polymeric flocculating agents
US6147176A (en) * 1988-12-19 2000-11-14 Cytec Technology Corp. High performance polymer flocculating agents nonionic polymers
US4968435A (en) * 1988-12-19 1990-11-06 American Cyanamid Company Cross-linked cationic polymeric microparticles
US5340865A (en) * 1988-12-19 1994-08-23 Cytec Technology Corp. Cross-linked cationic polyermic microparticles
US5354481A (en) * 1988-12-19 1994-10-11 Cytec Technology Corp. Water-soluble highly branched polymeric microparticles
US5693762A (en) * 1988-12-28 1997-12-02 Protein Design Labs, Inc. Humanized immunoglobulins
US5003047A (en) * 1989-01-10 1991-03-26 Massachusetts Institute Of Technology Method for purifying biologically active ligate
US5047511A (en) * 1989-08-28 1991-09-10 Pitman-Moore, Inc. Method for recovering recombinant proteins
US5164057A (en) * 1990-04-13 1992-11-17 W. R. Grace & Co.-Conn. Electrophoretic matrices and electrophoretic method using same
US5684107A (en) * 1991-02-09 1997-11-04 Basf Aktiengesellschaft Agglomerated polymer particles of finely divided, water-soluble or water-swellable polymers, the preparation thereof and the use thereof
US5238545A (en) * 1991-02-27 1993-08-24 W. R. Grace & Co.-Conn. Electrophoretic gel for separation and recovery of substances and its use
US5599719A (en) * 1992-01-07 1997-02-04 Middlesex Sciences, Inc. Method for isolating biomolecules from a biological sample with linear polymers
US5258122A (en) * 1992-04-16 1993-11-02 Amicon, Inc. Cross-flow filter device with pressure-balancing feature
US5430110A (en) * 1992-07-22 1995-07-04 Hoechst Aktiengesellschaft Polyvinylamine derivatives having hydrophilic centers, processes for their preparation and the use of the compounds as a medicament, active compound carrier and foodstuff auxiliary
US5622700A (en) * 1992-08-21 1997-04-22 Genentech, Inc. Method for treating a LFA-1-mediated disorder
US5736137A (en) * 1992-11-13 1998-04-07 Idec Pharmaceuticals Corporation Therapeutic application of chimeric and radiolabeled antibodies to human B lymphocyte restricted differentiation antigen for treatment of B cell lymphoma
US5324787A (en) * 1992-11-18 1994-06-28 Air Products And Chemicals, Inc. Modification of poly (vinylamine)
US5840851A (en) * 1993-07-23 1998-11-24 Plomer; J. Jeffrey Purification of hemoglobin
US5354801A (en) * 1993-08-12 1994-10-11 Cytec Technology Corp. Process for producing small polymer phase droplet microemulsions by multistep aqueous phase addition
US5714338A (en) * 1993-12-10 1998-02-03 Genentech, Inc. Methods for diagnosis of allergy
US5840804A (en) * 1994-03-01 1998-11-24 Roehm Gmbh Chemische Fabrik Crosslinked water-soluble polymer dispersions
US5512480A (en) * 1994-03-11 1996-04-30 Baxter International Inc. Flow-through bioreactor with grooves for cell retention
US20090155201A1 (en) * 1994-06-10 2009-06-18 Genzyme Corporation Alkylated poly(allylamine) polymers and methods of use
US5573675A (en) * 1995-05-11 1996-11-12 Nalco Chemical Company Clarification of deinking process waters using polymers containing vinylamine
US6821515B1 (en) * 1995-07-27 2004-11-23 Genentech, Inc. Protein formulation
US5998588A (en) * 1995-09-01 1999-12-07 University Of Washington Interactive molecular conjugates
US6024955A (en) * 1995-11-01 2000-02-15 Toagosei Co. Ltd. Peptides and monoclonal antibodies
US5879564A (en) * 1995-11-14 1999-03-09 Cytec Technology Corp. High performance polymer flocculating agents
US5807489A (en) * 1995-11-14 1998-09-15 Cytec Technology Corp. High performance polymer flocculating agents
US6133047A (en) * 1996-05-24 2000-10-17 Bio Merieux Superparamagnetic monodisperse particles
US5994560A (en) * 1996-08-29 1999-11-30 Hoechst Celanese Corp. Resolution of racemic mixtures using polymers containing chiral units
US5739383A (en) * 1996-08-29 1998-04-14 Hoechst Celanese Corp. Resolution of racemic mixtures using polymers containing chiral units
US6127526A (en) * 1996-11-27 2000-10-03 Genentech, Inc. Protein purification by Protein A chromatography
US5929214A (en) * 1997-02-28 1999-07-27 Cornell Research Foundation, Inc. Thermally responsive polymer monoliths
US6858694B2 (en) * 1997-12-09 2005-02-22 Agency Of Industrial Science And Technology Miti Stimuli-responsive polymer utilizing keto-enol tautomerization and stimuli-responsive separating material and chemical-releasing capsule comprising the same
US6706187B1 (en) * 1998-05-22 2004-03-16 Teruo Okano Packing material for chromatography having novel characteristic and method for isolation of substance using the same
US6307013B1 (en) * 1998-07-21 2001-10-23 Monsanto Technology Llc Clarification of protein precipitate suspensions using anionic polymeric flocculants
US6245555B1 (en) * 1998-09-01 2001-06-12 The Penn State Research Foundation Method and apparatus for aseptic growth or processing of biomass
US6956077B1 (en) * 1999-01-29 2005-10-18 Amersham Biosciences Kk Temperature-responsive polymer compound and process for producing the same
US20050224415A1 (en) * 1999-01-29 2005-10-13 Amersham Biosciences Kk Temperature-responsive polymer compound and process for producing the same
US6139746A (en) * 1999-02-22 2000-10-31 Kopf; Henry B. Method and apparatus for purification of biological substances
US6967085B1 (en) * 1999-08-14 2005-11-22 Ciba Specialty Chemicals Water Treatments Ltd. Flocculation of cell material
US6294622B1 (en) * 1999-09-27 2001-09-25 Ecole Polytechnique Federale De Lausanne (Epfl) Polymer flocculants with improved dewatering characteristics
US6258275B1 (en) * 1999-10-01 2001-07-10 Ecole Polytechnique Federale De Lausanne Affinity macroligands
US6641735B1 (en) * 2000-03-23 2003-11-04 Japan Chemical Innovation Institute Separatory material with the use of stimulus-responsive polymer and separation method by using the separatory material
US6805793B2 (en) * 2000-03-23 2004-10-19 Japan Chemical Innovation Institute Separatory material with the use of stimulus-responsive polymer and separation method by using the separatory material
US7070696B2 (en) * 2001-04-05 2006-07-04 Ciba Specialty Chemicals Water Treatments Ltd. Process for flocculating suspensions
US7514007B2 (en) * 2001-04-16 2009-04-07 Wsp Chemicals & Technology, Llc Use of water-soluble polymer complexes in aqueous systems
US7001953B2 (en) * 2001-04-16 2006-02-21 Wsp Chemicals & Technology, Llc Water-soluble polymer complexes
US6638918B2 (en) * 2001-11-09 2003-10-28 The Procter & Gamble Company Chitosan compositions
US6926832B2 (en) * 2002-01-04 2005-08-09 Nalco Company Method of using water soluble polymers in a membrane biological reactor
US7160971B2 (en) * 2002-02-15 2007-01-09 Iowa State University Research Foundation pH-sensitive methacrylic copolymers and the production thereof
US7300545B2 (en) * 2002-06-21 2007-11-27 Hymo Corporation Water-soluble polymer dispersion, process for producing the same and method of use therefor
US20060162882A1 (en) * 2002-06-21 2006-07-27 Takumi Ohara Water-soluble polymer dispersion, process for producing the same and method of use therefor
US7169908B2 (en) * 2003-01-09 2007-01-30 Genentech, Inc. Purification of polypeptides
US7547747B2 (en) * 2003-04-01 2009-06-16 Nitto Boseki Co., Ltd. Modified polyallylamine and process for producing the same
US20060121519A1 (en) * 2003-07-24 2006-06-08 Affisink Biotechnology Ltd. Compositions and methods for purifying and crystallizing molecules of interest
US7377686B2 (en) * 2003-09-04 2008-05-27 Millipore Corporation Disposable mixing system
US20050063259A1 (en) * 2003-09-22 2005-03-24 Fumio Isshiki Optical information recording apparatus
US20050282169A1 (en) * 2004-01-29 2005-12-22 Turner Allen C Signatory sequences
US7767399B2 (en) * 2005-01-31 2010-08-03 Merck & Co., Inc. Purification process for plasmid DNA
US20100282425A1 (en) * 2006-04-24 2010-11-11 Asko Karppi Cationic polysaccharide, its preparation and use
US20080193981A1 (en) * 2007-01-22 2008-08-14 Fahrner Robert L Polyelectrolyte precipitation and purification of proteins
US20110020327A1 (en) * 2008-12-16 2011-01-27 Millipore Corporation Purification of proteins
US20100193148A1 (en) * 2009-01-30 2010-08-05 Mckay Jonathan M Quaternary Vinylamine-Containing Polymers as Additives in Papermaking
US20110313066A1 (en) * 2010-05-17 2011-12-22 Millipore Corporation Stimulus responsive polymers for the purification of biomolecules
US20120070836A1 (en) * 2010-06-08 2012-03-22 Millipore Corporation Methods of detecting residual amounts of polymers used in the purification of biomolecules

Non-Patent Citations (6)

* Cited by examiner, † Cited by third party
Title
Agarwal et al. (Protein Expr. Purif. 1996 May; 7 (3): 294-8). *
Fujii et al. (Trends Biotechnol. 1991 Jun; 9 (6): 191-6) *
Galaev et al. (J. Chromatogr. A. 1994 Oct 28; 684 (1): 45-54) *
Galaev et al. (Trends Biotechnol. 1999 Aug; 17 (8): 335-40). *
Lali et al. (Bioseparation. 1999; 7: 195-205). *
Lehermayr et al. (J. Pharm. Sci. 2011 Jul; 100 (7): 2551-62) *

Cited By (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8951737B2 (en) 1996-05-06 2015-02-10 Cornell Research Foundation, Inc. Treatment and diagnosis of cancer
US8940871B2 (en) 2006-03-20 2015-01-27 The Regents Of The University Of California Engineered anti-prostate stem cell antigen (PSCA) antibodies for cancer targeting
US9410181B2 (en) 2006-06-27 2016-08-09 Emd Millipore Corporation Method and unit for preparing a sample for the microbiological analysis of a liquid
US9090930B2 (en) 2006-06-27 2015-07-28 Emd Millipore Corporation Method and unit for preparing a sample for the microbiological analysis of a liquid
US9376464B2 (en) 2006-12-21 2016-06-28 Emd Millipore Corporation Purification of proteins
US8362217B2 (en) 2006-12-21 2013-01-29 Emd Millipore Corporation Purification of proteins
US8569464B2 (en) 2006-12-21 2013-10-29 Emd Millipore Corporation Purification of proteins
US10793593B2 (en) 2006-12-21 2020-10-06 Emd Millipore Corporation Purification of proteins
US10233211B2 (en) 2006-12-21 2019-03-19 Emd Millipore Corporation Purification of proteins
US9527919B2 (en) 2007-09-04 2016-12-27 The Regents Of The University Of California High affinity anti-prostate stem cell antigen (PSCA) antibodies for cancer targeting and detection
US8940298B2 (en) 2007-09-04 2015-01-27 The Regents Of The University Of California High affinity anti-prostate stem cell antigen (PSCA) antibodies for cancer targeting and detection
US8999702B2 (en) 2008-06-11 2015-04-07 Emd Millipore Corporation Stirred tank bioreactor
US9803165B2 (en) * 2008-12-16 2017-10-31 Emd Millipore Corporation Stirred tank reactor and method
US20130005950A1 (en) * 2008-12-16 2013-01-03 Emd Millipore Corporation Stirred Tank Reactor And Method
US10517969B2 (en) 2009-02-17 2019-12-31 Cornell University Methods and kits for diagnosis of cancer and prediction of therapeutic value
US8772459B2 (en) 2009-12-02 2014-07-08 Imaginab, Inc. J591 minibodies and Cys-diabodies for targeting human prostate specific membrane antigen (PSMA) and methods for their use
US11180570B2 (en) 2009-12-02 2021-11-23 Imaginab, Inc. J591 minibodies and cys-diabodies for targeting human prostate specific membrane antigen (PSMA) and methods for their use
US9731288B2 (en) 2010-05-17 2017-08-15 Emd Millipore Corporation Stimulus responsive polymers for the purification of biomolecules
US9217048B2 (en) 2010-05-17 2015-12-22 Emd Millipore Corporation Stimulus responsive polymers for the purification of biomolecules
US8691918B2 (en) 2010-05-17 2014-04-08 Emd Millipore Corporation Stimulus responsive polymers for the purification of biomolecules
US9085622B2 (en) 2010-09-03 2015-07-21 Glaxosmithkline Intellectual Property Development Limited Antigen binding proteins
WO2014004281A1 (en) 2012-06-29 2014-01-03 Emd Millipore Corporation Purification of biological molecules
US10865224B2 (en) 2012-06-29 2020-12-15 Emd Millipore Corporation Purification of biological molecules
EP3130384A1 (en) 2012-06-29 2017-02-15 EMD Millipore Corporation Purification of biological molecules
US9926341B2 (en) 2012-12-20 2018-03-27 Merck Patent Gmbh Copolymers for protein precipitation
US11254744B2 (en) 2015-08-07 2022-02-22 Imaginab, Inc. Antigen binding constructs to target molecules
US11266745B2 (en) 2017-02-08 2022-03-08 Imaginab, Inc. Extension sequences for diabodies

Also Published As

Publication number Publication date
US8163886B2 (en) 2012-04-24
US20090036651A1 (en) 2009-02-05
WO2008079280A1 (en) 2008-07-03
WO2008079302A2 (en) 2008-07-03
US20130123476A1 (en) 2013-05-16
WO2008079302A3 (en) 2008-09-04
US10793593B2 (en) 2020-10-06

Similar Documents

Publication Publication Date Title
US10793593B2 (en) Purification of proteins
US9376464B2 (en) Purification of proteins
US10233211B2 (en) Purification of proteins
US20110020327A1 (en) Purification of proteins
US20220324905A1 (en) Enhanced protein purification through a modified protein a elution
US9731288B2 (en) Stimulus responsive polymers for the purification of biomolecules
JP4319979B2 (en) Non-affinity purification of proteins
US20130203969A1 (en) Use of small molecules in methods for purification of biomolecules
WO2015038205A1 (en) Surface plasmon resonance spectroscopy method to detect residual polymer flocculants in cell culture feed streams

Legal Events

Date Code Title Description
AS Assignment

Owner name: MILLIPORE CORPORATION, MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:MOYA, WILSON;REEL/FRAME:024593/0186

Effective date: 20100603

AS Assignment

Owner name: EMD MILLIPORE CORPORATION, MASSACHUSETTS

Free format text: CHANGE OF NAME;ASSIGNOR:MILLIPORE CORPORATION;REEL/FRAME:027620/0891

Effective date: 20120101

STCB Information on status: application discontinuation

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