US20080125580A1 - Refolding of Recombinant Proteins - Google Patents

Refolding of Recombinant Proteins Download PDF

Info

Publication number
US20080125580A1
US20080125580A1 US11/777,997 US77799707A US2008125580A1 US 20080125580 A1 US20080125580 A1 US 20080125580A1 US 77799707 A US77799707 A US 77799707A US 2008125580 A1 US2008125580 A1 US 2008125580A1
Authority
US
United States
Prior art keywords
recombinant protein
protein
vegf
buffered solution
support
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
US11/777,997
Other languages
English (en)
Inventor
Shelly Pizarro
Ailen Sanchez
Charles H. Schmelzer
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.)
Genentech Inc
Original Assignee
Genentech Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Genentech Inc filed Critical Genentech Inc
Priority to US11/777,997 priority Critical patent/US20080125580A1/en
Assigned to GENENTECH, INC. reassignment GENENTECH, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SCHMELZER, CHARLES H., PIZARRO, SHELLY, SANCHEZ, AILEN
Publication of US20080125580A1 publication Critical patent/US20080125580A1/en
Priority to US13/196,680 priority patent/US20110294990A1/en
Priority to US13/668,182 priority patent/US9200030B2/en
Priority to US14/922,802 priority patent/US9994612B2/en
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
    • C07K1/36Extraction; Separation; Purification by a combination of two or more processes of different types
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/14Extraction; Separation; Purification
    • 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
    • C07K1/107General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length by chemical modification of precursor peptides
    • C07K1/113General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length by chemical modification of precursor peptides without change of the primary structure
    • C07K1/1133General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length by chemical modification of precursor peptides without change of the primary structure by redox-reactions involving cystein/cystin side chains
    • 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
    • C07K1/107General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length by chemical modification of precursor peptides
    • C07K1/113General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length by chemical modification of precursor peptides without change of the primary structure
    • C07K1/1136General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length by chemical modification of precursor peptides without change of the primary structure by reversible modification of the secondary, tertiary or quarternary structure, e.g. using denaturating or stabilising agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/14Extraction; Separation; Purification
    • C07K1/16Extraction; Separation; Purification by chromatography
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/14Extraction; Separation; Purification
    • C07K1/16Extraction; Separation; Purification by chromatography
    • C07K1/20Partition-, reverse-phase or hydrophobic interaction chromatography
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators

Definitions

  • This invention relates to methods for obtaining heterologous recombinant proteins produced in cell culture.
  • the invention includes methods for recovering and purifying refolded recombinant proteins that have been produced in prokaryotic host cells and are present in these cells, typically in the periplasmic or intracellular space.
  • the recombinant proteins produced in prokaryotic host cells can also be found as soluble proteins or as a mixture of soluble and insoluble proteins.
  • recombinant proteins are produced in a variety of host organisms. Most proteins can be expressed in their native form in eukaryotic hosts such as CHO cells. Animal cell culture generally requires prolonged growing times to achieve maximum cell density and ultimately achieves lower cell density than prokaryotic cell cultures (Cleland, J. (1993) ACS Symposium Series 526, Protein Folding: In Vivo and In Vitro, American Chemical Society). Additionally, animal cell cultures often require expensive media containing growth components that may interfere with the recovery of the desired protein. Bacterial host expression systems provide a cost-effective alternative to the manufacturing scale production of recombinant proteins. Numerous U.S. patents on general bacterial expression of recombinant proteins exist, including U.S. Pat. Nos.
  • a major advantage of the production method is the ability to easily isolate the product from the cellular components by centrifugation or microfiltration. See, e.g., Kipriyanov and Little, (1999) Molecular Biotechnology, 12: 173-201; and, Skerra and Pluckthun, (1988) Science, 240: 1038-1040.
  • bacterial expression systems such as E. coli lack the cellular machinery to facilitate proper refolding of the proteins and generally do not result in the secretion of large proteins into the culture media.
  • Recombinant proteins expressed in bacterial host cells are often found as inclusion bodies consisting of dense masses of partially folded and misfolded reduced protein. See, e.g., Baneyx, (1999) Current Opin. Biotechnology 10:411-421; and, Villaverde and Carrio, (2003) Biotech. Letts. 25:1385-1395. Proteins may also be expressed without forming inclusion bodies. See, e.g., Id. Typically in inclusion bodies, the recombinant protein is generally inactive.
  • Typical downstream processing of proteins recovered from inclusion bodies includes the dissolution of the inclusion body at high concentration of a denaturant such as urea followed by dilution of the denaturant to permit refolding to occur (see, U.S. Pat. Nos. 4,512,922; 4,511,502; and 4,511,503). See also, e.g., Rudolph and Lilie, (1996) FASEB J.
  • HBP heparin binding protein
  • Reversed micelles or ion exchange chromatography have been used to assist refolding of denatured proteins by enclosing a single protein within micelles or isolating them on a resin and then removing the denaturant (Hagen et al., (1990) Biotechnol. Bioeng. 35:966-975; Creighton (1985) in Protein Structure Folding and Design (Oxender, D. L. Ed.) pp.249-251, New York: Alan R. Liss, Inc.). These methods have been useful in preventing protein aggregation and facilitating proper refolding.
  • the invention provides a method for recovering and purifying refolded recombinant proteins from cell culture.
  • the invention provides a method of recovering a recombinant protein from prokaryotic host cells, e.g., bacterial cells.
  • the processes of the invention are broadly applicable to recombinant proteins.
  • the recombinant protein is a growth factor, e.g., vascular endothelial growth factor (VEGF).
  • VEGF vascular endothelial growth factor
  • the growth factor is VEGF 165 .
  • a process includes: (a) isolating said recombinant protein from the prokaryotic cell culture; (b) solubilizing said protein in a first buffered solution, pH greater than 9, comprising a first chaotropic agent; (c) refolding said solubilized protein in a second buffered solution, pH>9 but ⁇ 11, comprising a second chaotropic agent, two or more reducing agents and addition of air or oxygen for such a time and under such conditions that refolding of the recombinant protein occurs; and (d) recovering said refolded recombinant protein.
  • the first buffered solution and/or the second buffered solution further comprises arginine.
  • the first buffered solution comprises 1 M Urea, 300 mM arginine, 10 mM CHES, 5 mM EDTA, pH 11, final concentration. In another embodiment, the first buffered solution comprises 1 M Urea, 300 mM arginine, 10 mM TRIS, 5 mM EDTA, pH 11, final concentration. In one embodiment, the second buffered solution comprises two or more reducing agents, e.g., DTT and cysteine. In one embodiment, the second buffered solution comprises 1 M Urea, 15 mM cysteine, 2 mM DTT, 100 mM arginine, 10 mM CHES, 5 mM EDTA, pH 10, final concentration. In another embodiment, the second buffered solution comprises 1 M Urea, 15 mM cysteine, 0.5-2 mM DTT, 100 mM arginine, 10 mM TRIS, 5 mM EDTA, pH 10, final concentration.
  • a process includes: (a) isolating said recombinant protein from the prokaryotic cell culture; (b) solubilizing and refolding said protein in a combo buffered solution, pH>9 but ⁇ 11 with the addition of air or oxygen; and, (c) recovering said recombinant protein.
  • the combo buffered solution comprises 1 M Urea, 15 mM cysteine, 2 mM DTT, 100 mM arginine, 10 mM CHES, 5 mM EDTA, pH 10, final concentration.
  • the combo buffered solution comprises 1 M Urea, 15 mM cysteine, 0.5-2 mM DTT, 100 mM arginine, 10 mM TRIS, 5 mM EDTA, pH 10, final concentration.
  • the oxygen or air for the refold reaction can be provided by an air source or an oxygen enriched compressed gas supply.
  • a k La of 0.004 min ⁇ 1 is used, e.g., which represents a mixing rate of 200-400 rpm and sparging rate of 0.3 cc/min/L in a 2.5L vessel containing a marine type impeller.
  • k Lz 0.01 min ⁇ 1 or 0.1 min ⁇ 1 are used to produce refolded protein.
  • the solubilization and/or refolding can be done at a variety of temperatures.
  • the incubation temperature for the solubilization and/or refolding is room temperature.
  • the incubation time can vary according to the recombinant protein being recovered and refolded.
  • the recombinant protein is incubated in the first buffered solution for at least 1 hour, or 1 to 2 hours.
  • the solubilized protein is incubated in the second buffered solution for about 3 to 24 hours.
  • the isolated recombinant protein is incubated in the combo buffered solution for 3 to 24 hours.
  • the invention additionally provides processes and methods for refolding of recombinant proteins either alone or in connection with the recovery of the recombinant protein as described herein.
  • purification methods include clarifying the solution containing the recombinant protein and contacting said refolded recombinant protein in the clarified solution with a mixed mode support, a cationic chromatographic support, a first hydrophobic interaction chromatographic support, and optionally, a second hydrophobic chromatographic support or an ion exchange support; and selectively recovering or eluting the refolded recombinant protein from each support.
  • clarifying the solution comprises adding detergent to a final concentration of 1%, adjusting pH to about 8.5-9.5, incubating solution for 1 to 10 hours at 25-30° C., centrifuging the solution; and filtering the liquid recovered from the centrifugation step.
  • the pH is about 8.7. In another embodiment, the pH is about 9.0. It is contemplated that the steps for recovery steps can be performed in any order, e.g., sequentially or altering the order of the chromatographic supports.
  • methods are provided for recovering and purifying refolded recombinant proteins from manufacturing or industrial scale cell culture.
  • FIG. 1 illustrates an example a time course study of the refolding process as described herein, which was evaluated by rpHPLC chromatography.
  • FIG. 2 illustrates a chromatograph from VEGF produced by bacterial strain W3110 loaded on a CaptoMMCTM column.
  • the column is equilibrated with 25 mM HEPES pH 9.0.
  • the VEGF is eluted from the MMC column isocratically with 1 M arginine/25 mM HEPES, pH 6-9.
  • FIG. 3 illustrates a chromatograph from VEGF produced by bacterial strain W3110 loaded on a SPHP column.
  • the SPHP column is equilibrated in 50 mM HEPES, pH 7.5.
  • the column is eluted using a linear gradient from 0.0-1.2 M sodium acetate in, 50 mM HEPES, pH 7.5 over 1 column volume.
  • the eluant is monitored at 280 nm.
  • the protein is recovered from fractions with the highest absorbance at 280 nm (OD max at ⁇ 42 mS/cm), which typically contain >90% of the VEGF, are pooled for further processing.
  • FIG. 4 illustrates a chromatograph from VEGF produced by bacterial strain W3110 loaded on a HiPropyl column.
  • FIG. 5 illustrates a chromatograph from VEGF produced by bacterial strain W3110 loaded on a Phenyl Sepharose column.
  • FIG. 6 illustrates the effect of urea and arginine on refolding conditions.
  • FIG. 7 illustrates the effect of N 2 in stabilizing the refold pool up to 48 hours as evaluated by rpHPLC time course, when N 2 was added 6 hours after refolding started.
  • FIG. 8 illustrates the effect of varying air sparging rates on refolding conditions
  • FIG. 9 illustrates a column assay, where cation exchange HPLC is used to assay for refolded VEGF.
  • FIG. 10 illustrates the amino acid sequence of VEGF 165 with disulfide bonds indicated.
  • polypeptide refers generally to peptides and proteins from any cell source having more than about ten amino acids.
  • heterologous polypeptides are those polypeptides foreign to the host cell being utilized, such as a human protein produced by E. coli. While the heterologous polypeptide may be prokaryotic or eukaryotic, preferably it is eukaryotic, more preferably mammalian, and most preferably human. In certain embodiments of the invention, it is a recombinantly produced (e.g., a recombinant polypeptide or a recombinant protein).
  • mammalian polypeptides include molecules such as, e.g., a growth factor; a heparin-binding growth factor; vascular endothelial growth factor (VEGF), e.g., VEGF-A (isoforms), VEGF-B, VEGF-C and VEGF-D; a receptor and antibody to VEGF such as rhuFab V2 and bevacizumab, ranibizumab; an antibody to VEGF receptors; rennin; a growth hormone, including human growth hormone (hGH); bovine growth hormone; growth hormone releasing factor; parathyroid hormone; thyroid stimulating hormone; lipoproteins; 1-antitrypsin; insulin A-chain; insulin B-chain; proinsulin; thrombopoietin; follicle stimulating hormone; calcitonin; luteinizing hormone; glucagon; growth hormone receptors; growth hormone releasing protein (GHRP); LIV-1 (EP 1263780); TRAIL; clo
  • the term “antibody” herein is used in the broadest sense and specifically covers intact monoclonal antibodies, polyclonal antibodies, multispecific antibodies (e.g. bispecific antibodies) formed from at least two intact antibodies, and antibody fragments, so long as they exhibit the desired biological activity.
  • the recombinant polypeptide is a growth factor.
  • the recombinant polypeptide is the mammalian polypeptide VEGF.
  • the recombinant polypeptide is human VEGF (e.g., VEGF 165 ).
  • the recombinant polypeptide is not angiostatin.
  • the recombinant polypeptide is not IGF-1.
  • vascular endothelial growth factor refers to a mammalian growth factor derived originally from bovine pituitary follicular cells having the amino acid sequence disclosed in Castor, C. W., et al., (1991) Methods in Enzymol. 198:391-405, together with functional derivatives thereof having the qualitative biological activity of a corresponding native VEGF, including, but not limited to, the human VEGF amino acid sequence as reported in Houck et al., (1991) Mol. Endocrin. 5:1806-1814. See also, Leung et al.
  • VEGF-A This is also referred to as VEGF-A.
  • Other members of the family are indicated by a letter notation at the end of VEGF, e.g., VEGF-B, VEGF-C, or VEGF-D.
  • the predominant form of VEGF or VEGF-A is a 165 amino acid homodimer having sixteen cysteine residues that form 7 intramolecular disulfide bonds and two intermolecular disulfide bonds.
  • VEGF 121 variant lacks the heparin binding domain of the other variants. All isoforms of VEGF share a common amino-terminal domain, but differ in the length of the carboxyl-terminal portion of the molecule.
  • VEGF 165 has disulfide bonds between amino acid residues Cys26-Cys68; Cys57-Cys104; Cys61-Cys102; Cys117-Cys135; Cys120-Cys137; Cys139-Cys;158; Cys146-Cys160 in each monomer. See FIG. 10 . See also, e.g., Keck et al., (1997) Archives of Biochemistry and Biophysics 344(1): 103-113.
  • the VEGF 165 molecule is composed of two domains: an amino-terminal receptor-binding domain (amino acids 1-110 disulfide linked homodimer) and a carboxyl-terminal heparin-binding domain (residues 111-165). See, e.g., Keyt et al., (1996) J. Biol. Chem., 271(13):7788-7795.
  • the VEGF 165 isolated and purified is not glycosylated at residue 75 (Asn). See, e.g., Yang et al., (1998) Journal of Pharm. & Experimental Therapeutics, 284:103-110.
  • the VEGF 165 isolated and purified is substantially undeamidated at residue Asn10. In certain embodiments of the invention, the VEGF 165 isolated and purified is a mixture of deamidated (at residue Asn10) and undearnidated protein, typically with majority of the protein being undeamidated. Since VEGF 165 is a homodimer, deamination can occur on one or both polypeptide chains.
  • heparin binding protein refers to a polypeptide capable of binding heparin (as herein defined).
  • the definition includes the mature, pre, pre-pro, and pro forms of native and recombinantly produced heparin-binding proteins.
  • Typical examples of heparin-binding proteins are “heparin binding growth factors,” including but not limited to epidermal growth factor (EGF), platelet derived growth factor (PDGF), basic fibroblast growth factor (bFGF), acidic fibroblast growth factor (aFGF), vascular endothelial growth factor (VEGF), hepatocyte growth factor (HGF) (also known as scatter factor, SF), and nerve growth factor (NGF), IL-8, etc.
  • EGF epidermal growth factor
  • PDGF platelet derived growth factor
  • bFGF basic fibroblast growth factor
  • aFGF acidic fibroblast growth factor
  • VEGF vascular endothelial growth factor
  • HGF hepatocyte growth factor
  • NGF
  • Heparin (also referred to as heparinic acid) is a heterogenous group of highly sulfated, straight-chain anionic mucopolysaccharides, called glycosaminoglycans. Although others may be present, the main sugars in heparin are: ⁇ -L-iduronic acid 2-sulfate, 2-deoxy-2-sulfamino- ⁇ -glucose 6-sulfate, ⁇ -D-glucuronic acid, 2-acetamido-2-deoxy- ⁇ -D-glucose, and L-iduronic acid. These and optionally other sugars are joined by glycosidic linkages, forming polymers of varying sizes.
  • heparin Due to the presence of its covalently linked sulfate and carboxylic acid groups, heparin is strongly acidic. The molecular weight of heparin varies from about 3,000 to about 20,000 daltons depending on the source and the method of determination. Native heparin is a constituent of various tissues, especially liver and lung, and mast cells in several mammalian species. Heparin and heparin salts (heparin sodium) are commercially available and are primarily used as anticoagulants in various clinical situations.
  • VEGF vascular endothelial growth factor
  • biologically active VEGF or other recombinant protein and the like refers to a molecule with a biologically active conformation.
  • the properly folded or biologically active VEGF or recombinant protein corresponds to the native folding pattern of the VEGF (described above) or other recombinant protein.
  • properly folded VEGF has the above noted disulfide pairs, in addition to two intermolecular disulfide bonds in the dimeric molecule, however other intermediates may be produced by bacterial cell culture.
  • VEGF vascular endothelial growth factor
  • VEGF receptor binding and signaling through a VEGF receptor
  • purified or “pure recombinant protein” and the like refer to a material free from substances which normally accompany it as found in its recombinant production and especially in prokaryotic or bacterial cell culture. Thus the terms refer to a recombinant protein which is free of contaminating DNA, host cell proteins or other molecules associated with its in situ environment. The terms refer to a degree of purity that is at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95% or at least about 98% or more.
  • inclusion bodies or “refractile bodies” refer to dense intracellular masses of aggregated polypeptide of interest, which constitute a significant portion of the total cell protein, including all cell components. In some cases, but not all cases, these aggregates of polypeptide may be recognized as bright spots visible within the enclosure of the cells under a phase-contrast microscope at magnifications down to 1,000 fold.
  • misfolded protein refers to precipitated or aggregated polypeptides that are contained within refractile bodies.
  • insoluble or misfolded VEGF or other recombinant protein refers to precipitated or aggregated VEGF or recombinant protein that is contained within the periplasm or intracellular space of prokaryotic host cells, or is otherwise prokaryotic host cell associated, and assumes a biologically inactive conformation with mismatched or unformed disulfide bonds.
  • the insoluble recombinant protein is generally, but need not be, contained in refractile bodies, i.e., it may or may not be visible under a phase contrast microscope.
  • chaotropic agent refers to a compound that, in a suitable concentration in aqueous solution, is capable of changing the spatial configuration or conformation of polypeptides through alterations at the surface thereof so as to render the polypeptide soluble in the aqueous medium.
  • the alterations may occur by changing, e.g., the state of hydration, the solvent environment, or the solvent-surface interaction.
  • concentration of chaotropic agent will directly affect its strength and effectiveness.
  • a strongly denaturing chaotropic solution contains a chaotropic agent in large concentrations which, in solution, will effectively unfold a polypeptide present in the solution effectively eliminating the proteins secondary structure. The unfolding will be relatively extensive, but reversible.
  • a moderately denaturing chaotropic solution contains a chaotropic agent which, in sufficient concentrations in solution, permits partial folding of a polypeptide from whatever contorted conformation the polypeptide has assumed through intermediates soluble in the solution, into the spatial conformation in which it finds itself when operating in its active form under endogenous or homologous physiological conditions.
  • chaotropic agents include guanidine hydrochloride, urea, and hydroxides such as sodium or potassium hydroxide. Chaotropic agents include a combination of these reagents, such as a mixture of a hydroxide with urea or guanidine hydrochloride.
  • reducing agent refers to a compound that, in a suitable concentration in aqueous solution, maintains free sulfhydryl groups so that the intra- or intermolecular disulfide bonds are chemically disrupted.
  • suitable reducing agents include dithiothreitol (DTT), dithioerythritol (DTE), beta-mercaptoethanol (BME), cysteine, cysteamine, thioglycolate, glutathione, and sodium borohydride.
  • buffered solution refers to a solution which resists changes in pH by the action of its acid-base conjugate components.
  • bacteria for purposes herein include eubacteria and archaebacteria.
  • eubacteria including gram-positive and gram-negative bacteria, are used in the methods and processes described herein.
  • gram-negative bacteria are used, e.g., Enterobacteriaceae.
  • bacteria belonging to Enterobacteriaceae include Escherichia, Enterobacter, Erwinia, Klebsiella, Proteus, Salmonella, Serratia, and Shigella.
  • Other types of suitable bacteria include Azotobacter, Pseudomonas, Rhizobia, Vitreoscilla, and Paracoccus.
  • E. coli is used.
  • Suitable E. coli hosts include E. coli W3110 (ATCC 27,325), E. coli 294 (ATCC 31,446), E. coli B, and E. coli X1776 (ATCC 31,537). These examples are illustrative rather than limiting, and W3110 is one example. Mutant cells of any of the above-mentioned bacteria may also be employed. It is, of course, necessary to select the appropriate bacteria taking into consideration replicability of the replicon in the cells of a bacterium. For example, E.
  • coli, Serratia, or Salmonella species can be suitably used as the host when well-known plasmids such as pBR322, pBR325, pACYC177, or pKN410 are used to supply the replicon. See further below regarding examples of suitable bacterial host cells.
  • the expressions “cell,” “cell line,” “strain,” and “cell culture” are used interchangeably and all such designations include progeny.
  • the words “transformants” and “transformed cells” include the primary subject cell and cultures derived therefrom without regard for the number of transfers. It is also understood that all progeny may not be precisely identical in DNA content, due to deliberate or inadvertent mutations. Mutant progeny that have the same function or biological activity as screened for in the originally transformed cell are included. Where distinct designations are intended, it will be clear from the context.
  • a mixed mode column refers to a column with a resin that has both cation exchange properties as well as hydrophobic interactions.
  • Recombinant proteins e.g., growth factors such as acidic fibroblast growth factor, basic fibroblast growth factor and vascular endothelial growth factor have been recovered and purified from a number of sources including bacteria (Salter D. H. et al., (1996) Labor. Invest. 74(2):546-556 (VEGF); Sieffle et al., (1996) Biochem. Biophys. Res. Commun. 222(2):249-55 (VEGF); Cao et al., (1996) J. Biol. Chem. 261(6):3154-62 (VEGF); Yang et al., (1994) Gaojishu Tongxun, 4:28-31 (VEGF); Anspach et al., (1995) J.
  • growth factors such as acidic fibroblast growth factor, basic fibroblast growth factor and vascular endothelial growth factor
  • VEGF vascular endothelial growth factor
  • bFGF Gaulandris (1994) J Cell. Physiol. 161(1):149-59 (bFGF); Estape and Rinas (1996) Biotech. Tech. 10(7):481-484 (bFGF); McDonald et al., (1995) FASEB J. 9(3):A410 (bFGF)).
  • VEGF-165 the predominant active form of VEGF is a homodimer of two 165-amino acid polypeptides.
  • each subunit contains 7 pairs of intrachain disulfide bonds and two additional pairs which effect the covalent linkage of the two subunits (Ferrara et al., (1991) J. Cell.
  • the native conformation includes a strongly basic domain which has been shown to readily bind heparin (Ferrara et al (1991) supra). While covalent dimerization of VEGF is needed for effective receptor binding and biological activity (Pötgens et al., (1994) J. Biol. Chem. 269:32879-32885; Claffey et al., (1995) Biochim. et Biophys. Acta 1246:1-9), the bacterial product potentially contains several misfolded and disulfide scrambled intermediates. Procedures are provided which are useful in isolating, purifying, and, reactivating proteins which appear in host cells in the form of “refractile bodies” and as soluble proteins as well.
  • Insoluble, misfolded recombinant protein is isolated from prokaryotic host cells expressing the protein by any of a number of art standard techniques.
  • the insoluble recombinant protein is isolated in a suitable isolation buffer by exposing the cells to a buffer of suitable ionic strength to solubilize most host proteins, but in which the subject protein is substantially insoluble, or disrupting the cells so as to release the inclusion bodies or the protein form the periplasmic or intracellular space and make them available for recovery by, for example, centrifugation.
  • This technique is well known and is described in, for example, U.S. Pat. No. 4,511,503.
  • Kleid et al. disclose purification of refractile bodies by homogenization followed by centrifugation (Kleid et al., (1984) in Developments in Industrial Microbiology, (Society for Industrial Microbiology, Arlington, Va.) 25:217-235). See also, e.g., Fischer et al., (1993) Biotechnology and Bioengineering 41:3-13.
  • U.S. Pat. No. 5,410,026 describes a typical method for recovering protein from inclusion bodies and is summarized as follows.
  • the prokaryotic cells are suspended in a suitable buffer.
  • the buffer consists of a buffering agent suitable for buffering at between pH 5 to 9, or about 6 to 8 and a salt. Any suitable salt, including NaCl, is useful to maintain a sufficient ionic strength in the buffered solution. Typically an ionic strength of about 0.01 to 2 M, or 0.1 to 0.2 M is employed.
  • the cells while suspended in this buffer, are disrupted or lysed using techniques commonly employed such as, for example, mechanical methods, e.g., Homogenizer (Manton-Gaulin press, Microfluidizer, or Niro-Soavi), a French press, a bead mill, or a sonic oscillator, or by chemical or enzymatic methods.
  • mechanical methods e.g., Homogenizer (Manton-Gaulin press, Microfluidizer, or Niro-Soavi), a French press, a bead mill, or a sonic oscillator, or by chemical or enzymatic methods.
  • Examples of chemical or enzymatic methods of cell disruption include spheroplasting, which entails the use of lysozyme to lyse the bacterial wall (H. Neu et al., (1964) Biochem. Biophys. Res. Comm., 17:215), and osmotic shock, which involves treatment of viable cells with a solution of high tonicity and with a cold-water wash of low tonicity to release the polypeptides (H. Neu et al., 1965 J. Biol. Chem., 240(9):3685-3692). Sonication is generally used for disruption of bacteria contained in analytical scale volumes of fermentation broth. At larger scales high pressure homogenization is typically used.
  • the suspension is typically centrifuged at low speed, generally around 500 to 15,000 ⁇ g, e.g., in one embodiment of the invention about 12,000 ⁇ g is used, in a standard centrifuge for a time sufficient to pellet substantially all of the insoluble protein. Such times can be simply determined and depend on the volume being centrifuged as well as the centrifuge design. Typically about 10 minutes to 0.5 hours is sufficient to pellet the insoluble protein. In one embodiment the suspension is centrifuged at 12,000 ⁇ g for 10 minutes.
  • the resulting pellet contains substantially all of the insoluble protein fraction. If the cell disruption process is not complete, the pellet may also contain intact cells or broken cell fragments. Completeness of cell disruption can be assayed by resuspending the pellet in a small amount of the same buffer solution and examining the suspension with a phase contrast microscope. The presence of broken cell fragments or whole cells indicates that further sonication or other means of disruption is necessary to remove the fragments or cells and the associated non-refractile polypeptides. After such further disruption, if required, the suspension is again centrifuged and the pellet recovered, resuspended, and reexamined. The process is repeated until visual examination reveals the absence of broken cell fragments in the pelleted material or until further treatment fails to reduce the size of the resulting pellet.
  • the above process can be employed whether the insoluble protein is intracellular or in the periplasmic space.
  • the conditions given herein for isolating recombinant protein are directed to inclusion bodies precipitated in the periplasmic space or intracellular space and relate particularly to VEGF.
  • the processes and procedures are thought to be applicable to recombinant proteins in general with minor modifications as noted throughout the following text.
  • the processes and procedures are applicable to manufacturing or industrial scale production, refolding, and purification of the recombinant protein.
  • the isolated recombinant protein in the pellet is incubated in a first buffered solution sufficient to substantially solubilize the recombinant protein. This incubation takes place under conditions of concentration, incubation time, and incubation temperature that will allow solubilization of desired amount or substantially all the recombinant protein.
  • the first buffered solution comprises a buffering agent suitable for maintaining the pH range of the buffer at least about 9 or greater, with the typical range being 9-11.
  • the pH for VEGF is pH 11.
  • suitable buffers include TRIS (Tris[hydroxymethyl]aminomethane), HEPPS (N-[2-Hydroxyethyl]piperazine-N′-[3-propane-sulfonic acid]), CAPSO (3-[Cyclohexylamino]-2-hydroxy-1-propanesulfonic acid), AMP (2-Amino-2-methyl-1-propanol), CAPS (3-[Cyclohexylamino]-1-propanesulfonic acid), CHES (2-[N-Cyclohexylamino]ethanesulfonic acid), arginine, lysine, and sodium borate.
  • TRIS Tris[hydroxymethyl]aminomethane
  • HEPPS N-[2-Hydroxyethyl]
  • the buffer herein includes CHES and arginine at about pH 11. In another embodiment, the buffer herein includes Tris and arginine at about pH 11. In one embodiment, the buffer herein includes CHES at about pH 11. In another embodiment, the buffer herein includes Tris at about pH 11. In certain embodiments, the first buffered solution includes a chaotropic agent.
  • Chaotropic agents suitable for practicing this invention include, e.g., urea and salts of guanidine or thiocyanate, e.g., urea, guanidine hydrochloride, sodium thiocyanate, etc.
  • the amount of chaotropic agent necessary to be present in the buffer is an amount sufficient to unfold the recombinant protein in solution.
  • a chaotrope is present at about between about 0.5-5 molar.
  • the chaotropic agent is urea at about 1 M.
  • the concentration of the protein in the buffered solution must be such that the protein will be substantially solubilized as determined by optical density.
  • the exact amount to employ will depend on, e.g., the concentrations and types of other ingredients in the buffered solution, particularly the protein concentration, chaotropic agent, and the pH of the buffer.
  • the concentration of recombinant protein is in the range of 0.5-5.5 mg per ml, or 1.5-5.0 mg/ml.
  • the solubilization is typically carried out at about 0-45° C., or about 2-40° C., or about 20-40° C., or about 23-37° C., or about 25-37° C., or about 25° C. for at least about one to 24 hours.
  • the temperature is not apparently affected by salt, reducing agent and chaotropic agent levels.
  • the solubilization is carried out at atmospheric pressure.
  • Measurement of the degree of solubilization in the buffered solution can be determined and is suitably carried out, for example, by turbidity determination, by analyzing fractionation between the supernatant and pellet after centrifugation, on reduced SDS-PAGE gels, by protein assay (e.g., the Bio-Rad protein assay kit), or by HPLC.
  • turbidity determination by analyzing fractionation between the supernatant and pellet after centrifugation, on reduced SDS-PAGE gels, by protein assay (e.g., the Bio-Rad protein assay kit), or by HPLC.
  • the disrupted cells are not centrifuged but are diluted, e.g., 1:4, 1:6, 1:8 in a second buffered solution described herein (refolding buffer).
  • refolding buffer a second buffered solution described herein.
  • This incubation takes place under conditions of concentration, incubation time, and incubation temperature that will allow solubilization and refolding of the recombinant protein.
  • about 30% or more of recombinant protein is solubilized and refolded.
  • a constant volumetric mass transfer coefficient k La 0.004 to 0.1 min ⁇ 1 (e.g., for a 2.5L vessel with a marine type impeller, air sparging rate is 0.3-10 cc/min/L, 0.3-3 cc/min/L,
  • the oxygen or air for the refold reaction can be provided by an air source or an oxygen enriched compressed gas supply.
  • the efficiency of mass transfer from the gas phase to the liquid phase is controlled by agitation, sparging and pressurization and is captured by the volumetric mass transfer coefficient, k La .
  • k La volumetric mass transfer coefficient
  • a k La of 0.004 min ⁇ 1 is used representing a mixing rate of 200-400 rpm and sparging rate of 0.3 cc/min/L in a 2.5L vessel containing a marine type impeller.
  • k La 0.01 min ⁇ 1 or 0.1 min ⁇ 1 are used to produce properly folded protein.
  • the second buffered solution contains two or more reducing agents.
  • the polypeptide may be diluted with the refolding buffer, e.g., at least five fold, or at least about ten fold, or about 20 fold, or about 40 fold.
  • the conditions of this second incubation of the soluble, unfolded protein will generally be such that desired amount or substantial or complete refolding of the protein will take place. The exact conditions will depend on, for example, the pH of the buffer and the types and concentrations of chaotropic and reducing agents present.
  • the incubation temperature is generally about 0-40° C. and the incubation will generally be carried out for at least about 1 hour to about 48 hours to effect refolding.
  • the reaction is carried out, e.g., at about 0-45° C., or about 2-40° C., or about 20-40° C., or about 23-37° C., or about 25-37° C., or about 25° C., for at least about 3 hours, for at least about 10 hours, or between about 3 and 30 hours, or between about 3 and 24 hours.
  • the reaction is carried out at atmospheric pressure.
  • the second buffered solution comprises a buffering agent suitable for maintaining the pH range of the buffer at least about 9 or greater than 9, with the typical range being 9-11, a chaotropic agent, and at least one reducing agent.
  • the second buffered solution comprises two or more reducing agents.
  • the pH for VEGF is pH 10. Examples of suitable buffers that will provide a pH within this latter range include TRIS.
  • the second buffered solution herein comprises CHES and arginine at about pH 10 (at about a concentration of 10 mM and 100 mM final concentration, respectively), with two or more reducing agents and at least one chaotropic agent.
  • the second buffered solution herein comprises Tris and arginine at about pH 10 (at about a concentration of 10 mM and 100 mM final concentration, respectively), with two or more reducing agents and at least one chaotropic agent.
  • Arginine (or another positively charged amino acid), e.g., L-arginine/HCl, can be present in the first buffered solution and the second buffered solution.
  • the concentration of arginine is e.g., about 50-500 mM, about 75-300 mM, or about 100-300 mM, or about 100 mM or 300 mM final concentration, etc.
  • the protein is in a first buffered solution at pH greater than 9 and 0.5-3 M urea, 50-500 mM arginine and 5 mM EDTA, final concentration. In one embodiment, 10 mM CHES final concentration is used.
  • the first buffered solution comprises 1 M Urea, 300 mM arginine, 10 mM CHES, 5 mM EDTA, pH 11, final concentration. In another embodiment, the first buffered solution comprises 1 M Urea, 300 mM arginine, 10 mM Tris, 5 mM EDTA, pH 11, final concentration.
  • the protein is in a second buffered solution (refolding buffered solution) at pH>9 but ⁇ 11 containing 0.5-3 M urea, 50-500 mM arginine, 0.25-1 mM DTT, 5-20 mM cysteine, and 2-10 mM EDTA, final concentration.
  • 10 mM CHES final concentration is used.
  • 10 mM Tris final concentration is used.
  • the protein is in a refolding buffer solution with 1 M urea, 15 mM cysteine, 2 mM DTT, 100 mM arginine, 10 mM CHES, 5 mM EDTA, pH 9-10, final concentration.
  • the protein is in a refolding buffer solution with 1 M urea, 15 mM cysteine, 0.5-2 mM DTT, 100 mM arginine, 10 mM Tris, 5 mM EDTA, pH 9-10, final concentration.
  • the solution also contains at least one reducing agent.
  • suitable reducing agents include, but are not limited to, dithiothreitol (DTT), ⁇ -mercaptoethonol (BME), cysteine, DTE, etc.
  • DTT dithiothreitol
  • BME ⁇ -mercaptoethonol
  • cysteine DTE
  • the amount of reducing agent to be present in the buffer will depend mainly on the type of reducing agent and chaotropic agent, the type and pH of the buffer employed, the amount of oxygen entrained in or introduced to the solution, and the concentration of the protein in the buffer.
  • the reducing agent is suitably selected from those described above in the concentration range of about 0.5 to about 20 mM for cysteine, 0.25-3.0 mM for DTT (e.g., 0.5-2 mM DTT), and less than about 0.2 mM for BME.
  • there are two or more reducing agents e.g., DTT at about 0.5-2 mM and 0.5 to about 20 mM cysteine.
  • DTT and BME can be used in connection with the procedures provided herein for recombinant proteins in general, a combination of cysteine at about 15 mM and DTT as described above is an example for the recovery of VEGF.
  • the reducing agent is DTT at about 2 mM, with 15 mM cysteine, final concentration.
  • the reducing agent is DTT at about 0.5 mM, with 15 mM cysteine, final concentration.
  • the second buffered solution contains at least one chaotropic agent at a concentration such that refolding of the recombinant protein occurs.
  • a chaotrope is present at about between about 0.5 and 2 molar final concentration.
  • the chaotropic agent herein is urea at about 0.5-2 M, 0.5-2 M, or at about 1 M, final concentration.
  • the chaotropic agent is guanidine hydrochloride at about 0.1-1 M final concentration.
  • the refolding buffer can optionally contain additional agents such as any of a variety of non-ionic detergents such as TRITONTM X-100, NONIDETTM P-40, the TWEENTM series and the BRIJTM series.
  • the non-ionic detergent is present at about between 0.01% and 1.0% final concentration. In one example, the concentrations for non ionic detergent are between about 0.025% and 0.05%, or about 0.05% final concentration.
  • the degree of refolding is suitably determined by high performance liquid chromatography (HPLC) analysis using e.g., rpHPLC chromatography column, a cation exchange HPLC (SP-5PW TSK gel column, Tosoh Bioscience LLC), or other appropriate heparin affinity column.
  • HPLC high performance liquid chromatography
  • rpHPLC chromatography column e.g., rpHPLC chromatography column
  • a cation exchange HPLC SP-5PW TSK gel column, Tosoh Bioscience LLC
  • Heparin binding HPLC assay directly correlates with increasing amounts of folded, biologically active recombinant protein present in the buffer.
  • the incubation is carried out to maximize the ratio of correctly folded recombinant protein to misfolded recombinant protein recovered, as determined by rpHPLC assay.
  • the quality and quantity of properly-folded VEGF is assessed using a heparin-binding assay.
  • Samples containing the diluted recombinant protein are loaded on a e.g., Heparin-5PW column (7.5 ⁇ 75 mm, Tosoh Biosciences LLC, Tokyo, Japan) or other suitable heparin affinity column.
  • the Heparin-5PW column is equilibrated in 10 mM sodium phosphate, pH 7.4 containing 0.15 M sodium chloride. At a flow rate of 1 ml/min or 2 m/min, the column is eluted using a linear gradient from 0.15-2 M sodium chloride in, 10 mM sodium phosphate, pH 7.4 over 10 minutes.
  • the eluant is monitored at 280 nm.
  • the protein is recovered in a single peak corresponding to the biologically active properly refolded VEGF.
  • an assay for determining properly refolded VEGF is RPHPLC. Disulfide linkages can optionally be confirmed by peptide map. Circular dichroism can also be used in for determining 2 & 3D structure/folding.
  • solubilization and refolding is performed in one step. After obtaining the disrupted cell pellet, it is placed or diluted into the second buffered solution described above (in this case entitled a combo buffered solution).
  • the polypeptide may be diluted with the combo buffered solution, e.g., at least five fold, or at least about ten fold, or about 20 fold, or about 40 fold.
  • the conditions of this incubation of the pellet will generally be such that desired amount or substantial or complete solubilization and refolding of the protein will take place, with the addition of air or oxygen.
  • a k La of 0.004 min ⁇ 1 is used representing a mixing rate of 200-400 rpm and sparging rate of 0.3 cc/min/L in a 2.5L vessel containing a marine type impeller.
  • k La 0.01 min ⁇ 1 or 0.1 min ⁇ 1 are used to produce properly folded protein.
  • the exact conditions will depend on, for example, the pH of the buffer and the types and concentrations of chaotropic and reducing agents present.
  • the incubation temperature is generally about 0-40° C. and the incubation will generally be carried out for at least about 1 to 48 hours to effect solubilization and refolding.
  • the reaction is carried out, e.g., at about 0-45° C., or about 2-40° C., or about 20-40° C., or about 23-37° C., or about 25-37° C., or about 25° C., for at least about 3 hours, for at least about 10 hours, or between about 3 and 48 hours, or between about 3 and 30 hours.
  • the reaction is carried out at atmospheric temperature.
  • the clarification step comprises adding detergent to a final concentration of 1% (e.g., Triton-X-100), adjusting pH to about 8.5-9.5 (or about 8.7 or about 9), incubating solution for 1 to 10 hours at 25-30° C., centrifuging the solution; and filtering liquid recovered from the centrifugation step.
  • 1% e.g., Triton-X-100
  • the multi-step chromatographic procedure comprises contacting said refolded recombinant protein with a mixed mode resin, a cationic chromatographic support, a first hydrophobic chromatography support, and optionally, a second hydrophobic chromatography support or an ion exchange support; and selectively recovering or eluting the recombinant protein from each support. It is contemplated that the steps of either procedure can be performed in any order. In one embodiment of the invention, the steps are performed sequentially.
  • a suitable first step in the further recovery and purification of the recombinant protein characteristically provides for the concentration of the recombinant protein and a reduction in sample volume.
  • the second incubation step described above may result in a large increase in the volume of the recovered recombinant protein and concommitant dilution of the protein in the refolding buffer.
  • Suitable first chromatographic supports provide a reduction in volume of recovered recombinant protein and may advantageously provide some purification of the protein from unwanted contaminating proteins.
  • Suitable first chromatographic steps include chromatographic supports which can be eluted and loaded directly onto a second chromatographic support.
  • first chromatographic supports include, but are not limited to, mixed mode resin (e.g., CaptoMMCTM, GE Healthcare, or MEP Hypercel, Pall Corporation), hydroxyapatite chromatographic supports, e.g., CHT ceramic type I and type II (formally known as MacroPrep ceramic), Bio-Gel HT, Bio-Gel HTP, Biorad, Hercules, Calif., etc.; metal chelating chromatographic supports consisting of an inert resin of immobilized metal ions such as copper, nickel, etc.; as well as non-derivatized silica gels.
  • the first chromatographic supports for the purification and recovery of VEGF are mixed ion exchange chromatographic supports. Elution from the first chromatographic support is accomplished according to art standard practices. Suitable elution conditions and buffers will facilitate the loading of the eluted recombinant protein directly onto the cationic chromatographic support as described below.
  • anionic constituents may be attached to matrices in order to form cationic supports for chromatography.
  • Anionic constituents include carboxymethyl, sulfethyl groups, sulfopropyl groups, phosphate and sulfonate (S).
  • Cellulosic ion exchange resins such as SE52 SE53, SE92, CM32, CM52, CM92, P11, DE23, DE32, DE52, EXPRESS IONTM S and EXPRESS IONTM C are available from Whatman LTD, Maidstone Kent U.K.
  • SEPHADEXTM and SEPHAROSETM based and cross linked ion exchangers are also known under the product names CM SEPHADEXTM C-25, CM SEPHADEXTM C-50 and SP SEPHADEXTM C-25 SP SEPHADEXTM C-50 and SP-SEPHAROSETM High Performance, SP-SEPHAROSETM-XL SP-SEPHAROSETM Fast Flow, CM-SEPHAROSETM Fast Flow, and CM-SEPHAROSETM, CL-6B, all available from Pharmacia AB.
  • ion exchangers for the practice of the invention include but are not limited to, e.g., ion exchangers under the product names MACROPREPTM such as for example MACROPREPTM S support, MACROPREPTM High S support and MACROPREPTM CM support from BioRad, Hercules, Calif.
  • MACROPREPTM such as for example MACROPREPTM S support, MACROPREPTM High S support and MACROPREPTM CM support from BioRad, Hercules, Calif.
  • Elution from cationic chromatographic supports is generally accomplished by increasing salt concentrations. Because the elution from ionic columns involves addition of salt and because, as mentioned herein, HIC is enhanced in salt concentration the introduction of HIC step following the ionic step or other salt step is optionally used.
  • a cationic exchange chromatographic step precedes at least the HIC step, e.g., a first hydrophobic interaction chromatographic support and/or a second hydrophobic interaction.
  • Hydrophobic columns can be used in the purification of the recombinant protein, e.g., in the 2 nd , 3 rd , and/or 4 th purification steps.
  • Hydrophobic interaction chromatography is well known in the art and is predicated on the interaction of hydrophobic portions of the molecule interacting with hydrophobic ligands attached to “chromatographic supports.”
  • a hydrophobic ligand coupled to a matrix is variously referred to as an HIC chromatographic support, HIC gel, or HIC column and the like. It is further appreciated that the strength of the interaction between the protein and the HIC column is not only a function of the proportion of non-polar to polar surfaces on the protein but of the distribution of the non-polar surfaces as well.
  • a number of matrices may be employed in the preparation of HIC columns. The most extensively used is agarose, although silica and organic polymer resins may be used.
  • Useful hydrophobic ligands include but are not limited to alkyl groups having from about 2 to about 10 carbon atoms, such as butyl, propyl, or octyl, or aryl groups such as phenyl.
  • HIC supports for gels and columns may be obtained commercially from suppliers such as Pharmacia, Uppsala, Sweden under the product names butyl-SEPHAROSETM, buty-SEPHAROSETM-Fast Flow, phenyl-SEPHAROSETM CL-4B, octyl SEPHAROSETM FF and phenyl SEPHAROSETM FF and Tosoh Corporation, Tokyo, Japan under the product names TOYOPEARLTM butyl 650M (Fractogel TSK Butyl-650) or TSK-GEL phenyl 5PW.
  • Ligand density is an important parameter in that it influences not only the strength of the interaction of the protein but the capacity of the column as well.
  • the ligand density of the commercially available phenyl or octyl phenyl gels is on the order of 5-40 ⁇ moles/mil gel bed.
  • Gel capacity is a function of the particular protein in question as well as pH, temperature and salt concentration but generally can be expected to fall in the range of 3-20 mg/ml gel.
  • the choice of particular gel can be determined by the skilled artisan.
  • the strength of the interaction of the protein and the HIC ligand increases with the chain length of the alkyl ligands but ligands having from about 4 to about 8 carbon atoms are suitable for most separations.
  • a phenyl group has about the same hydrophobicity as a pentyl group, although the selectivity can be different owing to the possibility of pi-pi interaction with aromatic groups of the protein.
  • Adsorption of the protein to a HIC column is favored by high salt concentration, but the actual concentration can vary over a wide range depending of the nature of the protein and the particular HIC ligand chosen. In general salt concentration between about 1 and 4 M are useful.
  • Elution from an HIC support can be accomplished in a variety of ways such as a) by changing the salt concentration, b) by changing the polarity of the solvent or c) by adding detergents.
  • a) by changing the salt concentration By decreasing salt concentrations adsorbed proteins are eluted in order of increasing hydrophobicity.
  • Changes in polarity may be effected by additions of solvents such as ethylene glycol or isopropanol thereby decreasing the strength of the hydrophobic interactions.
  • Detergents function as displacers of proteins and have been used primarily in connection with the purification of membrane proteins.
  • expression vectors capable of autonomous replication and protein expression relative to the host prokaryotic cell genome are introduced into the host cell. Construction of appropriate expression vectors is well known in the art including the nucleotide sequences of the recombinant proteins described herein. See, e.g., Sambrook et al., Molecular Cloning, A Laboratory Manual, Cold Spring Harbor Laboratory Press (Cold Spring Harbor, New York) (2001); Ausubel et al., Short Protocols in Molecular Biology, Current Protocols John Wiley and Sons (New Jersey) (2002); and, Baneyx, (1999) Current Opinion in Biotechnology, 10:411-421.
  • prokaryotic cell including bacteria
  • expression vectors are available commercially through, for example, the American Type Culture Collection (ATCC), Rockville, Md. Methods for the large scale growth of prokaryotic cells, and especially bacterial cell culture are well known in the art and these methods can be used in the context of the invention.
  • prokaryotic host cells are transfected with expression or cloning vectors encoding the recombinant protein of interest and cultured in conventional nutrient media modified as appropriate for inducing promoters, selecting transformants, or amplifying the genes encoding the desired sequences.
  • the nucleic acid encoding the polypeptide of interest is suitably RNA, cDNA, or genomic DNA from any source, provided it encodes the polypeptide(s) of interest. Methods are well known for selecting the appropriate nucleic acid for expression of heterologous polypeptides (including variants thereof) in microbial hosts.
  • Nucleic acid molecules encoding the polypeptide are prepared by a variety of methods known in the art. For example, a DNA encoding VEGF is isolated and sequenced, e.g., by using oligonucleotide probes that are capable of binding specifically to the gene encoding VEGF.
  • the heterologous nucleic acid (e.g., cDNA or genomic DNA) is suitably inserted into a replicable vector for expression in the microorganism under the control of a suitable promoter.
  • a replicable vector for expression in the microorganism under the control of a suitable promoter.
  • Many vectors are available for this purpose, and selection of the appropriate vector will depend mainly on the size of the nucleic -acid to be inserted into the vector and the particular host cell to be transformed with the vector.
  • Each vector contains various components depending on the particular host cell with which it is compatible. Depending on the particular type of host, the vector components generally include, but are not limited to, one or more of the following: a signal sequence, an origin of replication, one or more marker genes, a promoter, and a transcription termination sequence.
  • plasmid vectors containing replicon and control sequences that are derived from species compatible with the host cell are used in connection with microbial hosts.
  • the vector ordinarily carries a replication site, as well as marking sequences that are capable of providing phenotypic selection in transformed cells.
  • E. coli is typically transformed using pBR322, a plasmid derived from an E. coli species (see, e.g., Bolivar et al., (1977) Gene, 2: 95).
  • pBR322 contains genes for ampicillin and tetracycline resistance and thus provides easy means for identifying transformed cells.
  • the pBR322 plasmid, or other bacterial plasmid or phage also generally contains, or is modified to contain, promoters that can be used by the host for expression of the selectable marker genes.
  • Polypeptides of the invention may be produced recombinantly not only directly, but also as a fusion polypeptide with a heterologous polypeptide, which is typically a signal sequence or other polypeptide having a specific cleavage site at the N-terminus of the mature protein or polypeptide.
  • a heterologous polypeptide typically is one that is recognized and processed (i.e., cleaved by a signal peptidase) by the host cell.
  • the signal sequence is substituted by a prokaryotic signal sequence selected, for example, from the group of the alkaline phosphatase, penicillinase, lpp, or heat-stable enterotoxin II leaders.
  • Expression vectors contain a nucleic acid sequence that enables the vector to replicate in one or more selected host cells. Such sequences are well known for a variety of microbes.
  • the origin of replication from the plasmid pBR322 is suitable for most Gram-negative bacteria such as E. coli.
  • Expression vectors generally contain a selection gene, also termed a selectable marker. This gene encodes a protein necessary for the survival or growth of transformed host cells grown in a selective culture medium. Host cells not transformed with the vector containing the selection gene will not survive in the culture medium. This selectable marker is separate from the genetic markers as utilized and defined by this invention.
  • Typical selection genes encode proteins that (a) confer resistance to antibiotics or other toxins, e.g., ampicillin, neomycin, methotrexate, or tetracycline, (b) complement auxotrophic deficiencies other than those caused by the presence of the genetic marker(s), or (c) supply critical nutrients not available from complex media, e.g., the gene encoding D-alanine racemase for Bacilli.
  • One example of a selection scheme utilizes a drug to arrest growth of a host cell.
  • those cells that are successfully transformed with the nucleic acid of interest produce a polypeptide conferring drug resistance and thus survive the selection regimen.
  • Examples of such dominant selection use the drugs neomycin (Southern et al., (1982) J. Molec. Appl. Genet., 1: 327), mycophenolic acid (Mulligan et al., (1980) Science 209: 1422) or hygromycin (Sugden et al., (1985) Mol. Cell. Biol., 5: 410-413).
  • the three examples given above employ bacterial genes under eukaryotic control to convey resistance to the appropriate drug G418 or neomycin (geneticin), xgpt (mycophenolic acid), or hygromycin, respectively.
  • the expression vector for producing the recombinant protein of interest contains a suitable promoter that is recognized by the host organism and is operably linked to the nucleic acid encoding the polypeptide of interest.
  • Promoters suitable for use with prokaryotic hosts include the beta-lactamase and lactose promoter systems (Chang et al., (1978) Nature, 275: 615; Goeddel et al., (1979) Nature, 281: 544), the arabinose promoter system (Guzman et al., (1992) J.
  • Promoters for use in bacterial systems also generally contain a Shine-Dalgarno (S.D.) sequence operably linked to the DNA encoding the polypeptide of interest.
  • the promoter can be removed from the bacterial source DNA by restriction enzyme digestion and inserted into the vector containing the desired DNA.
  • Plasmids containing one or more of the above-listed components employs standard ligation techniques. Isolated plasmids or DNA fragments are cleaved, tailored, and re-ligated in the form desired to generate the plasmids required.
  • the ligation mixtures are used to transform E. coli K12 strain 294 (ATCC 31,446) or other strains, and successful transformants are selected by ampicillin or tetracycline resistance where appropriate. Plasmids from the transformants are prepared, analyzed by restriction endonuclease digestion, and/or sequenced by the method of Sanger et al., (1977) Proc. Natl. Acad. Sci. USA, 74: 5463-5467 or Messing et al., (1981) Nucleic Acids Res., 9: 309, or by the method of Maxam et al., (1980) Methods in Enzvmology, 65: 499. See also, e.g., Sambrook et al., supra; and Ausubel et al., supra.
  • the nucleic acid encoding the recombinant protein of interest is inserted into the host cells. Typically, this is accomplished by transforming the host cells with the above-described expression vectors and culturing in conventional nutrient media modified as appropriate for inducing the various promoters.
  • Suitable prokayotic cells for the practice of the invention are well known in the art. Host cells that express the recombinant protein abundantly in the form of inclusion bodies or in the perplasmic or intracellular space are typically used. Suitable prokaryotes include bacteria, e.g., eubacteria, such as Gram-negative or Gram-positive organisms, for example, E. coli, Bacilli such as B. subtilis, Pseudomonas species such as P. aeruginosa, Salmonella typhimurium, or Serratia marcescens. One example of an E. coli host is E. coli 294 (ATCC 31,446). Other strains such as E. coli B, E.
  • Strain W3110 is a typical host because it is a common host strain for recombinant DNA product fermentations. In one aspect of the invention, the host cell should secrete minimal amounts of proteolytic enzymes.
  • strain W3110 may be modified to effect a genetic mutation in the genes encoding proteins, with examples of such hosts including E. coli W3110 strains 1A2, 27A7, 27B4, and 27C7 described in U.S. Pat. No. 5,410,026 issued Apr. 25, 1995.
  • a strain for the production of VEGF is E.
  • Prokaryotic cells used to produce the recombinant protein of interest are grown in media known in the art and suitable for culture of the selected host cells, including the media generally described by Sambrook et al., Molecular Cloning, A Laboratory Manual, Cold Spring Harbor Laboratory Press (Cold Spring Harbor, N.Y.) (2001).
  • Media that are suitable for bacteria include, but are not limited to, AP5 medium, nutrient broth, Luria-Bertani (LB) broth, Neidhardt's minimal medium, and C.R.A.P. minimal or complete medium, plus necessary nutrient supplements.
  • the media also contains a selection agent, chosen based on the construction of the expression vector, to selectively permit growth of prokaryotic cells containing the expression vector.
  • ampicillin is added to media for growth of cells expressing ampicillin resistant gene.
  • Any necessary supplements besides carbon, nitrogen, and inorganic phosphate sources may also be included at appropriate concentrations introduced alone or as a mixture with another supplement or medium such as a complex nitrogen source.
  • the culture medium may contain one or more reducing agents selected from the group consisting of glutathione, cysteine, cystamine, thioglycollate, dithioerythritol, and dithiothreitol.
  • C.R.A.P. phosphate-limiting media consists of 3.57 g (NH 4 ) 2 (SO 4 ), 0.71 g Na citrate-2H 2 O, 1.07 g KCl, 5.36 g Yeast Extract (certified), 5.36 g HycaseSFTM-Sheffield, adjusted pH with KOH to 7.3, volume adjusted to 872 ml with deionized H 2 O and autoclaved; cooled to 55° C. and supplemented with 110 ml 1 M MOPS pH 7.3, 11 ml 50% glucose, 7 ml 1M MgSO 4 ). Carbenicillin may then be added to the induction culture at a concentration of 50 ⁇ g/ml.
  • the prokaryotic host cells are cultured at suitable temperatures.
  • the temperature ranges from, e.g., about 20° C. to about 39° C., or from about 25° C. to about 37° C., or at about 30° C.
  • E. coli cells used to produce the polypeptide of interest of this invention are cultured in suitable media in which the alkaline phosphatase promoter can be partially or completely induced as described generally, e.g., in Sambrook et al., Molecular Cloning, A Laboratory Manual, Cold Spring Harbor Laboratory Press (Cold Spring Harbor, New York) (2001).
  • the culturing need never take place in the absence of inorganic phosphate or at phosphate starvation levels.
  • the medium contains inorganic phosphate in an amount above the level of induction of protein synthesis and sufficient for the growth of the bacterium. As the cells grow and utilize phosphate, they decrease the level of phosphate in the medium, thereby causing induction of synthesis of the polypeptide.
  • the cells are cultured until a certain optical density is achieved, e.g., a A 550 of about 200 using a high cell density process, at which point induction is initiated (e.g., by addition of an inducer, by depletion of a medium component, etc.), to induce expression of the gene encoding the polypeptide of interest.
  • a certain optical density e.g., a A 550 of about 200 using a high cell density process, at which point induction is initiated (e.g., by addition of an inducer, by depletion of a medium component, etc.), to induce expression of the gene encoding the polypeptide of interest.
  • any necessary supplements may also be included at appropriate concentrations that would be known to those skilled in the art, introduced alone or as a mixture with another supplement or medium such as a complex nitrogen source.
  • the pH of the medium may be any pH from about 5-9, depending mainly on the host organism. For E. coli, the pH is, e.g., from about 6.8 to about 7.4, or about 7.0.
  • polypeptide thus recovered may be formulated in a pharmaceutically acceptable carrier and is used for various diagnostic, therapeutic, or other uses known for such molecules.
  • proteins described herein can be used in immunoassays, such as enzyme immunoassays.
  • a growth factor or hormone e.g., VEGF
  • VEGF can be used to enhance growth as desired.
  • VEGF can be used to promote wound healing of, e.g., an acute wound (e.g., burn, surgical wound, normal wound, etc.) or a chronic wound (e.g., diabetic ulcer, pressure ulcer, a decubitus ulcer, a venous ulcer, etc.), to promote hair growth, to promote tissue growth and repair, etc.
  • Therapeutic formulations of recombinant proteins are prepared for storage by mixing a molecule, e.g., a polypeptide, having the desired degree of purity with optional pharmaceutically acceptable carriers, excipients or stabilizers (Remington's Pharmaceutical Sciences 18 th edition, Osol, A. Ed. (1995)), in the form of lyophilized formulations or aqueous solutions.
  • a molecule e.g., a polypeptide, having the desired degree of purity
  • optional pharmaceutically acceptable carriers, excipients or stabilizers Remington's Pharmaceutical Sciences 18 th edition, Osol, A. Ed. (1995)
  • Acceptable carriers, excipients, or stabilizers are nontoxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, and other organic acids; antioxidants including ascorbic acid and methionine; preservatives (such as octadecyldimethylbenzyl ammonium chloride; hexamethonium chloride; benzalkonium chloride, benzethonium chloride; phenol, butyl or benzyl alcohol; alkyl parabens such as methyl or propyl paraben; catechol; resorcinol; cyclohexanol; 3-pentanol; and m-cresol); low molecular weight (less than about 10 residues) polypeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids such as glycine, glutamine, asparagine, histidine,
  • the formulations to be used for in vivo administration are sterile. This is readily accomplished by filtration through sterile filtration membranes.
  • Recombinant proteins can be stored in lyophilized form or as an aqueous solution or gel form.
  • the pH of the recombinant protein preparations can be, e.g., from about 4 to 8 (in one embodiment, pH 5.0), although higher or lower pH values may also be appropriate in certain instances. It will be understood that use of certain of the excipients, carriers, or stabilizers can result in the formation of salts of the recombinant protein.
  • the route of polypeptide administration is in accord with known methods, e.g., topical administration, injection or infusion by intravenous, intraperitoneal, intracerebral, intramuscular, intraocular, intraarterial, or intralesional routes, or by sustained-release systems as noted below.
  • the polypeptide can be administered continuously by infusion or by bolus injection.
  • recombinant protein is formulated for site-specific delivery.
  • the recombinant protein is suitably combined with other ingredients, such as carriers and/or adjuvants.
  • suitable vehicles include ointments, creams, gels, sprays, or suspensions, with or without purified collagen.
  • the compositions also may be impregnated into sterile dressings, transdermal patches, plasters, and bandages, optionally in liquid or semi-liquid form.
  • the recombinant protein formulated in a liquid composition may be mixed with an effective amount of a water-soluble polysaccharide or synthetic polymer such as polyethylene glycol to form a gel of the proper viscosity to be applied topically.
  • a water-soluble polysaccharide or synthetic polymer such as polyethylene glycol
  • the polysaccharide that may be used includes, for example, cellulose derivatives such as etherified cellulose derivatives, including alkyl celluloses, hydroxyalkyl celluloses, and alkylhydroxyalkyl celluloses, for example, methylcellulose, hydroxyethyl cellulose, carboxymethyl cellulose, hydroxypropyl methylcellulose, and hydroxypropyl cellulose; starch and fractionated starch; agar; alginic acid and alginates; gum arabic; pullullan; agarose; carrageenan; dextrans; dextrins; fructans; inulin; mannans; xylans; arabinans; chitosans; glycogens; glucans; and synthetic biopolymers; as well as gums such as xanthan gum; guar gum; locust bean gum; gum arabic; tragacanth gum; and karaya gum; and derivatives and mixtures thereof.
  • cellulose derivatives such as etherified cellulose derivative
  • the gelling agent herein is one that is, e.g., inert to biological systems, nontoxic, simple to prepare, and/or not too runny or viscous, and will not destabilize the recombinant protein held within it.
  • the polysaccharide is an etherified cellulose derivative, in another embodiment one that is well defined, purified, and listed in USP, e.g., methylcellulose and the hydroxyalkyl cellulose derivatives, such as hydroxypropyl cellulose, hydroxyethyl cellulose, and hydroxypropyl methylcellulose.
  • methylcellulose is the polysaccharide.
  • the polyethylene glycol useful for gelling is typically a mixture of low and high molecular weight polyethylene glycols to obtain the proper viscosity.
  • a mixture of a polyethylene glycol of molecular weight 400-600 with one of molecular weight 1500 would be effective for this purpose when mixed in the proper ratio to obtain a paste.
  • water soluble as applied to the polysaccharides and polyethylene glycols is meant to include colloidal solutions and dispersions.
  • solubility of the cellulose derivatives is determined by the degree of substitution of ether groups, and the stabilizing derivatives useful herein should have a sufficient quantity of such ether groups per anhydroglucose unit in the cellulose chain to render the derivatives water soluble.
  • a degree of ether substitution of at least 0.35 ether groups per anhydroglucose unit is generally sufficient.
  • the cellulose derivatives may be in the form of alkali metal salts, for example, the Li, Na, K, or Cs salts.
  • methylcellulose is employed in the gel, e.g., it comprises about 2-5%, or about 3%, or about 4% or about 5%, of the gel, and the recombinant protein is present in an amount of about 300-1000 mg per ml of gel.
  • the active ingredients may also be entrapped in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly-(methylmethacylate) microcapsules, respectively, in colloidal drug delivery systems (for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules) or in macroemulsions.
  • colloidal drug delivery systems for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules
  • macroemulsions for example, liposomes, albumin microspheres, microemulsions, nano-particles and nanocapsules
  • Sustained-release preparations may be prepared. Suitable examples of sustained-release preparations include semipermeable matrices of solid hydrophobic polymers containing a polypeptide of the invention, which matrices are in the form of shaped articles, e.g. films, or microcapsules. Examples of sustained-release matrices include polyesters, hydrogels (for example, poly(2-hydroxyethyl-methacrylate), or poly(vinylalcohol)), polylactides (U.S. Pat. No.
  • copolymers of L-glutamic acid and ⁇ ethyl-L-glutamate copolymers of L-glutamic acid and ⁇ ethyl-L-glutamate, non-degradable ethylene-vinyl acetate, degradable lactic acid-glycolic acid copolymers such as the LUPRON DEPOTTM (injectable microspheres composed of lactic acid-glycolic acid copolymer and leuprolide acetate), poly-lactic-coglycolic acid (PLGA) polymer, and poly-D-( ⁇ )-3-hydroxybutyric acid. While polymers such as ethylene-vinyl acetate and lactic acid-glycolic acid enable release of molecules for over 100 days, certain hydrogels release proteins for shorter time periods.
  • encapsulated proteins When encapsulated proteins remain in the body for a long time, they may denature or aggregate as a result of exposure to moisture at 37° C., resulting in a loss of biological activity and possible changes in immunogenicity. Rational strategies can be devised for stabilization depending on the mechanism involved. For example, if the aggregation mechanism is discovered to be intermolecular S—S bond formation through thio-disulfide interchange, stabilization may be achieved by modifying sulfhydryl residues, lyophilizing from acidic solutions, controlling moisture content, using appropriate additives, and developing specific polymer matrix compositions.
  • Sustained-release polypeptide compositions also include liposomally entrapped polypeptides.
  • Liposomes containing the protein are prepared by methods known per se: DE 3,218,121; Epstein et al., (1985) Proc. Natl. Acad. Sci. USA, 82: 3688-3692; Hwang et al., (1980) Proc. Natl. Acad. Sci. USA, 77: 40304034; EP 52,322; EP 36,676; EP 88,046; EP 143,949; EP 142,641; Japanese patent application 83-118008; U.S. Pat. Nos. 4,485,045 and 4,544,545; and EP 102,324.
  • the liposomes are of the small (about 200-800 Angstroms) unilamellar type in which the lipid content is greater than about 30 mol. % cholesterol, the selected proportion being adjusted for the most effective therapy with polypeptide.
  • An effective amount of recombinant protein to be employed therapeutically will depend, for example, upon the therapeutic objectives, the route of administration, and the condition of the patient. Accordingly, it will be necessary for the therapist to titer the dosage and modify the route of administration as required to obtain the most beneficial therapeutic effect.
  • a typical daily dosage might range from about 1 ⁇ g/kg to up to 10 mg/kg or more, depending on the factors mentioned above.
  • the clinician will administer polypeptide until a dosage is reached that achieves the desired effect.
  • a patient can also administer the polypeptide under the guidance of the clinician. The progress of this therapy is easily monitored by conventional assays.
  • Plasmid for VEGF 165 expression The plasmid pVEGF171 is designed for the expression of human VEGF 165 (see, e.g., Leung et al., (1989) Science, 246: 1306-1309) in the E. coli periplasm.
  • VEGF coding sequence Transcription of the VEGF coding sequence is placed under tight control of the alkaline phosphatase (AP) promoter (see, e.g., Kikuchi et al., (1981) Nucleic Acids Research, 9:5671-8), while sequences required for translation initiation are provided by the trp Shine-Dalgarno region (see, e.g., Yanofsky et al., (1981) Nucleic Acids Research, 9:6647-68).
  • the VEGF coding sequence is fused downstream of the bacterial heat-stable enterotoxin II (STII) signal sequence (see, e.g., Lee et al., (1983) Infect. Immun.
  • STII bacterial heat-stable enterotoxin II
  • Cell Homogenization and Refractile body preparation Whole cell broth from Escherichia coli cells producing recombinant protein are homogenized with a microfluidizer or Niro Soavi at pressures greater than 8000 psid. The homogenate is diluted 1:1 with 160 mM MgSO 4 , 0.0375 dextran sulfate and 1% Triton X-100 prior to harvesting the pellet by centrifugation (BTUX centrifuge, Alfa Laval, Sweden).
  • Solubilization and Refolding The pellet (e.g., 1 gram) is suspended in 4 volumes (e.g., 4 ml) of solubilization buffer: 1M Urea/300 mM arginine, 10 mM Tris or CHES, 5 mM EDTA, pH 11, final concentration, (4 L/kg pellet). The suspension is thoroughly mixed for 1-2 hours at room temperature (15-30° C.).
  • Refolding is initiated by addition of 3 volumes (1:4 v/v) of buffer per volume of solubilization buffer, which results in the final concentration of the refolding buffer being 1 M Urea, 15 mM cysteine, 0.5-2 mM DTT, 100 mM arginine, 10 mM Tris or CHES, 5 mM EDTA, pH 9-10.
  • VEGF can be stabilized in the refold buffer by adding nitrogen (e.g., 0.3-3 cc/min/L for 2.5L tank) in place of air after 6 hours. See FIG. 7 .
  • nitrogen e.g., 0.3-3 cc/min/L for 2.5L tank
  • the folding is monitored by SDS-PAGE, cation exchange HPLC and rpHPLC chromatography, and/or Heparin HPLC.
  • Solubilization and Refolding The pellet is suspended in 10-39 liter volumes of refolding buffer (in this case termed “combo buffered solution”) for every kg of cell pellet, where the combo buffered solution contains 1 M Urea, 15 mM cysteine, 0.5 or 2 mM DTT, 100 mM arginine, 10 mM Tris or CHES, 5 mM EDTA, pH 9.5-10.5, final concentration. See FIG. 6 for the effect of urea and arginine addition in the refolding buffered solution.
  • FIG. 6 shows the results of a 1-step pellet refold (combo buffered solution) as described in this example at pH 9.5 for 15 hours at room temperature.
  • the denaturant concentrations are varied as follows: (1) 1 M urea and 100 mM arginine; (2) 1 M urea (and 0 mM arginine); (3) 2 M urea (and 0 mM arginine), while all the other buffer components (e.g., Tris or CHES, DTT, etc.) remain in the same concentration.
  • the VEGF titer extracted from these is equivalent as determined by the cation exchange HPLC assay.
  • FIG. 6 shows that the rpHPLC profiles are comparable with or without the presence of arginine.
  • the VEGF containing pellet was added to the combo buffered solution at pH 10 at a ratio of 1:39 (pellet kg to buffer L).
  • the refolding buffer contains 1 M Urea, 15 mM cysteine, 2 mM DTT, 100 mM arginine, 10 mM Tris or CHES, 5 mM EDTA, pH 9-10, final concentration.
  • Refold incubation is conducted at room temperature for 3-24 hours.
  • VEGF can be stabilized in the refold buffer by adding nitrogen in place of air after 3 hours refold incubation.
  • the folding is monitored by cation exchange HPLC, rpHPLC chromatography, and/or Heparin HPLC.
  • the refold pool is clarified by adding Triton X-100 to a final concentration of 1%, adjusting to pH 9 and then centrifugation (10,000 ⁇ g for 20 minutes at 4° C.). The supernatant is then filtered (Cuno depth filter+0.22 or 0.45 ⁇ membrane filter) prior to capture on a mixed mode resin (CaptoMMCTM, GE Healthcare, Piscataway, N.J.) at pH 9 and conductivity ⁇ 10 mS/cm.
  • Triton X-100 Triton X-100
  • the supernatant is then filtered (Cuno depth filter+0.22 or 0.45 ⁇ membrane filter) prior to capture on a mixed mode resin (CaptoMMCTM, GE Healthcare, Piscataway, N.J.) at pH 9 and conductivity ⁇ 10 mS/cm.
  • the refold pool is diluted at least 1:5 in equilibration loading buffer and then filtered (Cuno depth filter+0.22 or 0.45 ⁇ membrane filter) prior to capture on a mixed mode resin (CaptoMMCTM, GE Healthcare, Piscataway, N.J.) at pH 9 and conductivity ⁇ 10 mS/cm.
  • a mixed mode resin CaptoMMCTM, GE Healthcare, Piscataway, N.J.
  • the packed column is equilibrated with 25 mM HEPES pH 9 prior to loading the sample on the column.
  • the VEGF is eluted from the MMC column isocratically with 1 M arginine/25 mM HEPES, at pH 6-9 (e.g., pH 7.5). See FIG. 2 .
  • the CaptoMMCTM pool is adjusted to pH 7.5 with 0.1 N sodium hydroxide and diluted with WFI to 20 mS/cm conductivity prior to loading onto a SP-Sepharose HP column. (equilibrated with 50 mM HEPES pH 7.5).
  • the VEGF is eluted using a linear salt gradient composed of 50 mM HEPES/0-1M sodium acetate pH 7.5 over 10-20 column volumes (e.g., 15 column volumes) and fractions are collected (1 column volume).
  • the fractions with the highest absorbance @ 280 nm typically contain >90% of the VEGF and are pooled for further processing. See FIG. 3 .
  • the third chromatography step includes a hydrophobic resin (e.g., Hi Propyl, J. T. Baker, Phenyl Sepharose Fast Flow (low sub), GE Healthcare, Piscataway, N.J.).
  • the SP-Sepharose HP elution pool is conditioned to 50 mS/cm conductivity using either sodium acetate or sodium sulfate prior to loading onto the equilibrated column (50 mM HEPES, 1.2 M sodium acetate, pH 7.5). See FIG. 4 .
  • the VEGF elutes isocratically into 50 mM HEPES, pH 7.5. and the pool is analyzed for remaining host cell impurities and soluble aggregates.
  • Fractions are collected and those which contained properly-folded VEGF, as determined by assays described herein are pooled.
  • an additional chromatography step is performed, e.g., using a second hydrophobic resin (e.g., Phenyl TSK) or ion exchange resin.
  • the pooled VEGF can be ultrafiltered on a 5 kD regenerated cellulose membrane on a labscale TFF system to a concentration of 6 g/L (UFI).
  • the sample is diafiltrated with 7-14 DV (Diavolume) with 5 mM sodium succinate via TFF system to 10 g/L and then formulated at 5 g/L for storage at ⁇ 80° C.
  • the formulation buffer used is 5 mM sodium succinate/275 mM trehalose dehydrate/0.01% polysorbate 20/pH 5.0.
  • the refold pool is clarified by adding Triton X-100 to a final concentration of 1%, adjusting to pH 8.5-9.5 (e.g., pH 8.7) and holding at 25-30° C. for 1 to 10 hours prior to centrifugation. After processing on the centrifuge (10,000 ⁇ g for 20 minutes at 4° C.) to remove the large density particles, the recovered liquid (centrate) is passed thru a series of depth filters and sterile guard (0.22 or 0.45 ⁇ membrane) filters to remove the fine particles. rhVEGF is then captured on a mixed mode resin (CaptoMMCTM, GE Healthcare, Piscataway, N.J.) at pH 8.7 and conductivity ⁇ 10 mS/cm.
  • Triton X-100 adjusting to pH 8.5-9.5 (e.g., pH 8.7) and holding at 25-30° C. for 1 to 10 hours prior to centrifugation. After processing on the centrifuge (10,000 ⁇ g for 20 minutes at 4° C.) to remove the large density particles, the recovered
  • the packed column is equilibrated with 25 mM CHES pH 8.7 prior to loading the sample on the column.
  • the VEGF is eluted from the MMC column isocratically with 0.9 M L-arginine HCl/25 mM HEPES, at pH 6-9 (e.g., pH 7.5).
  • the CaptoMMCTM pool is adjusted to pH 7.5 with 0.1 N sodium hydroxide and diluted with WFI to 20 mS/cm conductivity prior to loading onto a SP-Sepharose High Performance column (equilibrated with 25 mM HEPES pH 7.5).
  • the VEGF is eluted using a linear salt gradient composed of 50 mM HEPES/0-1.2 M sodium acetate pH 7.5 over 10-20 column volumes (e.g., 15 column volumes) and fractions are collected (1 column volume).
  • the fractions with the highest absorbance @ 280 nm typically contain >90% of the VEGF and are pooled for further processing.
  • the third chromatography step includes a hydrophobic resin (e.g., Hi Propyl, J. T. Baker, Phenyl Sepharose Fast Flow (low sub), GE Healthcare, Piscataway, N.J.).
  • the SP-Sepharose HP elution pool is loaded directly onto the equilibrated HIC column (25 mM HEPES, 0.75 M sodium acetate, pH 7.5). See FIG. 5 .
  • the VEGF elutes isocratically into 50 mM HEPES, pH 7.5 and the pool is analyzed for remaining host cell impurities and soluble aggregates. Fractions are collected and those which contained properly-folded VEGF, as determined by assays described herein are pooled.
  • an additional chromatography step is performed to further remove host impurities, e.g., using a second hydrophobic resin (e.g., Phenyl TSK) or ion exchange resin.
  • a second hydrophobic resin e
  • the pooled VEGF can be ultrafiltered on a 5 kD regenerated cellulose membrane in commercial TFF system (Pellicon 2 casettes, Millipore, Billerica, Mass.) to a concentration of 10 g/L then diafiltered with 7-14 diavolumes (eg., 10 DV) into the formulation buffer.
  • Final conditioning produces a solution containing 5 g/L VEGF in 5 mM sodium succinate/275 mM trehalose dehydrate/0.01% polysorbate 20/pH 5.0 that can be stored at ⁇ 80° C.
  • final purity and/or activity can be assessed by peptide mapping, disulfide mapping, SDS-PAGE (both reduced and non-reduced), circular dichroism, limulus amobocyte lysate (LAL), Cation exchange HPLC, heparin HPLC (e.g., Heparin HPLC can be used to determine VEGF dimer concentration and level of misfolded species), reverse phase (rp) HPLC chromatography (e.g., rpHPLC of reduced samples can be used to determine total VEGF concentration whereas rpHPLC of native samples can assess the quality of refolded VEGF), receptor binding (for example for VEGF e.g., KDR receptor binding-Bioanalytic R&D, and/or Flt1 receptor binding), SEC Analysis, cell assays, HUVEC potency assays, ELISAs with VEGF antibodies, mass spec analysis, etc.
  • LAL limulus amobocyte lysate
  • Cation exchange HPLC
  • (1) rpHPLC of reduced samples The quantity of expressed VEGF is measured using a reverse phase HPLC assay on a C18 column (Jupiter C18 column (4.6 ⁇ 250 mm, 5 micron, by Phenomenex, Torrance, Calif.). The column is equilibrated in 0.22% trifluoroacetic acid and eluted using a linear gradient of 25% to 45% acetonitrile containing 0.2% trifluoroacetic acid in 30 min with a flow rate of 1 mL/min. The eluant is monitored at 280 nm. The sample is treated and fully reduced in guanidine and DTT prior to injection. The reduced VEGF protein elutes around 26 min and the peak area is used to calculate the amount of total VEGF in the sample from a known standard curve.
  • Cation Exchange HPLC assay The quantity of properly refolded VEGF dimers is determined using an analytical cation exchange column, e.g., SP-5PW column (TSK gel SP-5PW, 7.5 ⁇ 75 mm, 10 micron, by Tosoh Biosciences LLC, Japan).
  • the column is equilibrated in 50 mM sodium phosphate pH 7.5. At a flow rate of 1 mL/min the column is eluted using a linear gradient from 0 to 2 M sodium chloride in equilibration buffer over 60 min.
  • the eluant is monitored at 280 nm or 214 nm. Typically, the majority of protein is eluted in the first 30 min and VEGF is eluted around 40 min. See FIG. 9 .
  • rp-HPLC assay The quality of properly refolded VEGF is determined using a Zorbax 300SB-C8 column (4.6 ⁇ 150 mm, 3.5 micron, by Agilent Technologies, Santa Clara, Calif.). The column is equilibrated in 0.1% trifluoroacetic acid and eluted using a linear gradient of 0 to 50% acetonitrile containing 0.08% trifluoroacetic acid over 50 min with a flow rate of 1 mL/min. The eluant is monitored at 214 nm. Typically, VEGF elutes around 35 min and the peak profile is evaluated for the percent content of the leading edge hydrophobic species relative to the main peak. Unfolded VEGF monomer elutes 2-3 min later.
  • Heparin-binding HPLC Assay The quality and quantity of properly refolded VEGF is determined using a column containing immobilized heparin.
  • the column Heparin-5PW (7.5 ⁇ 75 mm, 10 micron, TSK gel by Tosoh Biosciences LLC, Japan) is equilibrated in 10 mM sodium phosphate, pH 7.4 containing 0.15 M sodium chloride. At a flow rate of 1 mL/min the column is eluted using a linear gradient from 0.15 M to 1.6 M sodium chloride in equilibration buffer over 20 min. In some assays, elution is done in 16 min. The eluant is monitored at 280 nm. Typically, the majority of protein is eluted in the void volume and 3 classes of VEGF could be identified. The highest affinity, latest-eluting species is identified as correctly folded VEGF and is sometimes identified as “Peak 3 VEGF.”

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Analytical Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Crystallography & Structural Chemistry (AREA)
  • Toxicology (AREA)
  • Zoology (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Peptides Or Proteins (AREA)
US11/777,997 2006-07-14 2007-07-13 Refolding of Recombinant Proteins Abandoned US20080125580A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
US11/777,997 US20080125580A1 (en) 2006-07-14 2007-07-13 Refolding of Recombinant Proteins
US13/196,680 US20110294990A1 (en) 2006-07-14 2011-08-02 Refolding of Recombinant Proteins
US13/668,182 US9200030B2 (en) 2006-07-14 2012-11-02 Refolding of recombinant proteins
US14/922,802 US9994612B2 (en) 2006-07-14 2015-10-26 Refolding of recombinant proteins

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US83083106P 2006-07-14 2006-07-14
US11/777,997 US20080125580A1 (en) 2006-07-14 2007-07-13 Refolding of Recombinant Proteins

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/196,680 Continuation US20110294990A1 (en) 2006-07-14 2011-08-02 Refolding of Recombinant Proteins

Publications (1)

Publication Number Publication Date
US20080125580A1 true US20080125580A1 (en) 2008-05-29

Family

ID=38722613

Family Applications (4)

Application Number Title Priority Date Filing Date
US11/777,997 Abandoned US20080125580A1 (en) 2006-07-14 2007-07-13 Refolding of Recombinant Proteins
US13/196,680 Abandoned US20110294990A1 (en) 2006-07-14 2011-08-02 Refolding of Recombinant Proteins
US13/668,182 Active 2027-09-19 US9200030B2 (en) 2006-07-14 2012-11-02 Refolding of recombinant proteins
US14/922,802 Active US9994612B2 (en) 2006-07-14 2015-10-26 Refolding of recombinant proteins

Family Applications After (3)

Application Number Title Priority Date Filing Date
US13/196,680 Abandoned US20110294990A1 (en) 2006-07-14 2011-08-02 Refolding of Recombinant Proteins
US13/668,182 Active 2027-09-19 US9200030B2 (en) 2006-07-14 2012-11-02 Refolding of recombinant proteins
US14/922,802 Active US9994612B2 (en) 2006-07-14 2015-10-26 Refolding of recombinant proteins

Country Status (18)

Country Link
US (4) US20080125580A1 (pt)
EP (1) EP2041154B1 (pt)
JP (2) JP5566104B2 (pt)
KR (1) KR101520105B1 (pt)
CN (1) CN101506222B (pt)
AR (1) AR062069A1 (pt)
AU (1) AU2007272412B2 (pt)
BR (1) BRPI0713252C1 (pt)
CA (1) CA2656835C (pt)
IL (1) IL196004A (pt)
MX (1) MX2009000278A (pt)
MY (1) MY169472A (pt)
NZ (1) NZ573639A (pt)
RU (1) RU2439076C2 (pt)
SG (1) SG162834A1 (pt)
TW (1) TWI476207B (pt)
WO (1) WO2008008975A2 (pt)
ZA (1) ZA200900229B (pt)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100273216A1 (en) * 2009-04-17 2010-10-28 Paul Morin Methods for protein refolding
KR101063347B1 (ko) 2009-04-06 2011-09-07 인하대학교 산학협력단 이온성 액체를 이용한 이황화 결합을 포함하는 단백질의 재접힘 방법
WO2012054679A1 (en) * 2010-10-20 2012-04-26 Medimmune, Llc Methods for processing inclusion bodies
WO2016106229A1 (en) * 2014-12-23 2016-06-30 Armo Biosciences, Inc. Methods of improving yield in recombinant protein production
US20160266016A1 (en) 2013-08-14 2016-09-15 Riken Composition for preparing biomaterial with excellent light-transmitting property, and use thereof
US9920124B2 (en) * 2012-12-20 2018-03-20 Medimmune, Llc Methods of producing immunoconjugates
WO2018211529A1 (en) 2017-05-19 2018-11-22 Council Of Scientific & Industrial Research A method for producing refolded recombinant humanized ranibizumab
US10444124B2 (en) 2011-05-20 2019-10-15 Riken Clarifying reagent for biological materials and use thereof
US11345722B2 (en) * 2013-02-12 2022-05-31 Bristol-Myers Squibb Company High pH protein refolding methods

Families Citing this family (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR101804136B1 (ko) * 2008-06-24 2017-12-04 옥타파마 아게 응고 인자 viii을 정제하는 방법
MX2011001624A (es) 2008-08-21 2011-03-28 Octapharma Ag Factor viii y ix humano producido en forma recombinante.
MX2011013898A (es) 2009-06-22 2012-05-22 Amgen Inc Proteínas replegadas que usan un estado de oxido-reduccion quimicamente controlado.
EP2445924B2 (en) 2009-06-25 2023-12-13 Amgen Inc. Capture purification processes for proteins expressed in a non-mammalian system
WO2012147965A1 (ja) * 2011-04-28 2012-11-01 独立行政法人理化学研究所 生物材料を透明化する方法、及びその利用
CN102443055B (zh) * 2011-10-26 2017-07-28 浙江海正药业股份有限公司 一种重组人肿瘤坏死因子相关凋亡诱导配体的纯化工艺
RU2492177C2 (ru) * 2011-12-15 2013-09-10 Федеральное государственное унитарное предприятие "Государственный научно-исследовательский институт генетики и селекции промышленных микроорганизмов" (ФГУП ГосНИИгенетика) СПОСОБ ВЫДЕЛЕНИЯ И ОЧИСТКИ РЕКОМБИНАНТНОГО ГОРМОНА РОСТА ЧЕЛОВЕКА, СЕКРЕТИРУЕМОГО ДРОЖЖАМИ Saccharomyces cerevisiae
CN104350156B (zh) * 2012-03-27 2019-04-02 弗·哈夫曼-拉罗切有限公司 用于重组蛋白质的改进的收获操作
CN103588871B (zh) * 2012-08-16 2016-08-03 深圳新鹏生物工程有限公司 一种rhTRAIL菌体的分离纯化方法
EP3981380A1 (en) 2013-03-12 2022-04-13 Primal Therapies, Inc. Antimicrobial composition comprising a chelator and a basic aminoacid
WO2014155349A2 (en) * 2013-03-29 2014-10-02 Dr. Reddy's Laboratories Refolding of proteins
CN103694339B (zh) * 2013-12-04 2017-10-13 珠海联邦制药股份有限公司 一种甘精胰岛素前体的复性方法
KR101651330B1 (ko) * 2014-07-28 2016-08-25 가톨릭대학교 산학협력단 세포투과성이 우수한 tat-a20 융합단백질의 제조방법 및 이의 용도
EP3221449B1 (en) * 2014-11-18 2023-08-30 Merck Sharp & Dohme LLC Process for refolding recombinant chymotrypsin
EP3221448B1 (en) 2014-11-18 2021-01-06 Merck Sharp & Dohme Corp. Process for producing recombinant trypsin
EA039002B1 (ru) 2014-12-22 2021-11-19 Юсб Биофарма Спрл Способ получения белка
AU2016307976A1 (en) * 2015-08-17 2018-03-08 Lupin Limited An improved refolding process for antibody's fragments
CN106008702B (zh) * 2016-07-22 2019-11-26 江苏江山聚源生物技术有限公司 一种规模化分离纯化重组人源胶原蛋白的方法
EP3959222A4 (en) * 2019-04-24 2023-06-14 Tanvex Biopharma Usa, Inc. METHOD FOR PRODUCTION OF GRANULOCYTE COLONY STIMULATING FACTOR
CN116710568A (zh) * 2020-06-26 2023-09-05 特里福伊尔治疗公司 重组修饰成纤维细胞生长因子及其治疗用途
CN111848774B (zh) * 2020-08-05 2022-05-10 武汉海特生物制药股份有限公司 一种美曲普汀的制备方法
WO2023017540A1 (en) * 2021-08-11 2023-02-16 Tata Institute For Genetics And Society Methods for refolding a solubilized recombinant protein and implementation thereof

Citations (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4490473A (en) * 1983-03-28 1984-12-25 Panab Labeled antibodies and methods
US4511502A (en) * 1982-12-22 1985-04-16 Genentech, Inc. Purification and activity assurance of precipitated heterologous proteins
US4511503A (en) * 1982-12-22 1985-04-16 Genentech, Inc. Purification and activity assurance of precipitated heterologous proteins
US4512922A (en) * 1982-12-22 1985-04-23 Genentech, Inc. Purification and activity assurance of precipitated heterologous proteins
US4565785A (en) * 1978-06-08 1986-01-21 The President And Fellows Of Harvard College Recombinant DNA molecule
US4673641A (en) * 1982-12-16 1987-06-16 Molecular Genetics Research And Development Limited Partnership Co-aggregate purification of proteins
US4795706A (en) * 1985-01-31 1989-01-03 Eli Lilly And Company Novel expression control sequences
US4929700A (en) * 1987-04-16 1990-05-29 Cetus Corporation Production of purified, biologically active, bacterially produced recombinant human CSF-1
US5288931A (en) * 1991-12-06 1994-02-22 Genentech, Inc. Method for refolding insoluble, misfolded insulin-like growth factor-I into an active conformation
US5322699A (en) * 1991-02-04 1994-06-21 The Rockefeller University Leukocyte-derived CR3 modulator, integrin modulating factor-1 (IMF-1)
US5593865A (en) * 1985-10-23 1997-01-14 Boehringer Mannheim Gmbh Process for the activating of gene-technologically produced, heterologous, disulphide bridge-containing eukaryotic proteins after expression in prokaryotes
US5618927A (en) * 1990-11-22 1997-04-08 Boehringer Mannheim Gmbh Process for the reactivation of denatured protein
US20010041521A1 (en) * 1999-07-29 2001-11-15 Chartered Semiconductor Manufacturing Ltd. Adjustable and extended guide rings
US6583268B2 (en) * 2000-01-25 2003-06-24 Oklahoma Medical Research Foundation Universal procedure for refolding recombinant proteins
US20030120042A1 (en) * 2000-03-30 2003-06-26 Takao Yamada Process for producing recombinant protein
US20030190316A1 (en) * 2000-08-11 2003-10-09 Masaya Kakuta Stabilized antibody-containing preparations
US20050176109A1 (en) * 2004-01-21 2005-08-11 Ryosuke Yumioka Purification method which prevents denaturation of an antibody
US20060199948A1 (en) * 2005-03-03 2006-09-07 Daisuke Ejima Method for improving recovery yield in protein purification with gel filtration chromatography

Family Cites Families (45)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4169950A (en) 1977-12-08 1979-10-02 Research Organics Amino-hydroxy-alkyl sulfonic acid zwitterions
EP0104771B1 (en) 1982-09-02 1990-05-23 Nellcor Incorporated Pulse oximeter monitor
US4710473A (en) 1983-08-10 1987-12-01 Amgen, Inc. DNA plasmids
US4652630A (en) 1985-02-22 1987-03-24 Monsanto Company Method of somatotropin naturation
ES8800957A1 (es) * 1985-02-22 1987-12-01 Monsanto Co Un metodo para la solubilizacion y renaturalizacion de proteina somatotropina
DE3537708A1 (de) 1985-10-23 1987-04-23 Boehringer Mannheim Gmbh Verfahren zur aktivierung von t-pa nach expression in prokaryonten
US4734362A (en) * 1986-02-03 1988-03-29 Cambridge Bioscience Corporation Process for purifying recombinant proteins, and products thereof
DE3835350A1 (de) 1988-10-17 1990-04-19 Boehringer Mannheim Gmbh Aktivierung von gentechnologisch hergestellten, in prokaryonten exprimierten antikoerpern
US5064943A (en) * 1988-12-16 1991-11-12 American Cyanamid Company Method for solubilization and naturation of somatotropin
US5332671A (en) 1989-05-12 1994-07-26 Genetech, Inc. Production of vascular endothelial cell growth factor and DNA encoding same
US5194596A (en) 1989-07-27 1993-03-16 California Biotechnology Inc. Production of vascular endothelial cell growth factor
GB8927546D0 (en) * 1989-12-06 1990-02-07 Ciba Geigy Process for the production of biologically active tgf-beta
US5331095A (en) 1993-04-12 1994-07-19 Scios Nova Inc. Process for purification of basic fibroblast growth factor
JP4098372B2 (ja) 1993-07-28 2008-06-11 三菱化学株式会社 ヘパリン結合性増殖因子の生産方法
US5663304A (en) * 1993-08-20 1997-09-02 Genentech, Inc. Refolding of misfolded insulin-like growth factor-I
US5464815A (en) 1993-09-08 1995-11-07 Genentech, Inc. Inhibition of heparin-binding
EP0751992B1 (en) 1994-03-08 2005-11-09 Human Genome Sciences, Inc. Vascular endothelial growth factor 2
FR2729972B1 (fr) 1995-01-31 1997-04-18 Sanofi Sa Procede d'extraction de proteines periplasmiques de microorganismes procaryotes en presence d'arginine
SE9504019D0 (sv) 1995-11-13 1995-11-13 Pharmacia Ab Method for production of proteins
US6750044B1 (en) 1996-10-17 2004-06-15 Genentech, Inc. Variants of vascular endothelial cell growth factor having antagonistic properties, nucleic acids encoding the same and host cells comprising those nucleic acids
US6632425B1 (en) 1997-03-20 2003-10-14 Human Genome Sciences, Inc. Chemokine compositions
DE19735711C2 (de) 1997-08-18 2001-04-26 Aventis Pharma Gmbh Verfahren zur Herstellung eines Vorläufers von Insulin oder Insulinderivaten mit korrekt verbundenen Cystinbrücken
US6653098B1 (en) * 1998-02-23 2003-11-25 G. D. Searle & Co. Method of producing mouse and human endostatin
WO1999050302A1 (en) 1998-03-31 1999-10-07 Tonghua Gantech Biotechnology Ltd. Chimeric protein containing an intramolecular chaperone-like sequence and its application to insulin production
ATE274522T1 (de) 1998-06-01 2004-09-15 Genentech Inc Abtrennung von antikörper-monomeren von deren multimeren mittels ionaustausch-chromatographie
ES2207340T3 (es) 1998-10-28 2004-05-16 Genentech, Inc. Procedimiento para la recuperacion de polipeptidos heterologos a partir de celulas bacterianas.
US6783953B1 (en) 1998-12-22 2004-08-31 Janssen Pharmaceutica N.V. Vascular endothelial growth factor-X
EP1183357A2 (en) 1999-05-20 2002-03-06 Scios Inc. Vascular endothelial growth factor dimers
AUPQ568100A0 (en) 2000-02-16 2000-03-09 Amrad Operations Pty. Limited A method for producing recombinant molecules
JP4873818B2 (ja) 2000-05-16 2012-02-08 ボルダー バイオテクノロジー, インコーポレイテッド 遊離システイン残基を含有するタンパク質をリフォールディングする方法
DE10054059A1 (de) 2000-10-31 2002-05-08 Max Delbrueck Centrum Verfahren zur Ermittlung von Faltungsbedingungen für rekombinante Proteine
WO2002057296A1 (en) 2001-01-16 2002-07-25 Københavns Universitet A method for refolding of proteins
US7611711B2 (en) 2001-01-17 2009-11-03 Vegenics Limited VEGFR-3 inhibitor materials and methods
CZ20032208A3 (cs) 2001-02-23 2004-01-14 Immunex Corporation Zvýšený výtěžek aktivních proteinů
DK1501369T3 (en) * 2002-04-26 2015-09-28 Genentech Inc Non-affinity purification of proteins
CN1207307C (zh) * 2002-11-21 2005-06-22 李越希 重组人巨细胞病毒融合蛋白及其制备方法、应用
ES2287687T3 (es) 2003-01-09 2007-12-16 Genentech, Inc. Purificacion de polipeptidos.
EP1449848A1 (en) 2003-02-20 2004-08-25 GBF German Research Centre for Biotechnology Method for the production of cystine-knot proteins
CN1473932A (zh) * 2003-06-30 2004-02-11 中山大学 一种重组石斑鱼腺苷酸环化酶激活多肽基因及其表达系统、表达产物、生产方法和应用
BRPI0416683A (pt) * 2003-12-10 2007-01-30 Lilly Co Eli muteìna de fator de crescimento de fibroblasto de humano 21 (fgf-21), ou um seu peptìdeo biologicamente ativo, polinucleotìdeo, vetor de expressão, célula hospedeira, processo para produzir um polipeptìdeo, composição farmacêutica, método para tratar um paciente, e, uso da muteìna fgf-21
AU2004309177A1 (en) 2003-12-30 2005-07-14 Bharat Biotech International Limited A process for the preparation and purification of recombinant proteins
DE102004015965A1 (de) * 2004-04-01 2005-10-20 Aventis Pharma Gmbh Verfahren zur Gewinnung von Insulinen durch verbesserte Luftbegasung der Faltung
EP1817417B1 (en) * 2004-06-28 2016-05-18 Yeda Research And Development Co., Ltd. Chimeric proteins and uses thereof
AU2005306168A1 (en) * 2004-11-22 2006-05-26 Anaphore, Inc. TNF antagonists
DE602007012412D1 (de) 2005-12-22 2011-03-24 Genentech Inc Rekombinante produktion heparinbindender proteine

Patent Citations (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4565785A (en) * 1978-06-08 1986-01-21 The President And Fellows Of Harvard College Recombinant DNA molecule
US4673641A (en) * 1982-12-16 1987-06-16 Molecular Genetics Research And Development Limited Partnership Co-aggregate purification of proteins
US4511502A (en) * 1982-12-22 1985-04-16 Genentech, Inc. Purification and activity assurance of precipitated heterologous proteins
US4511503A (en) * 1982-12-22 1985-04-16 Genentech, Inc. Purification and activity assurance of precipitated heterologous proteins
US4512922A (en) * 1982-12-22 1985-04-23 Genentech, Inc. Purification and activity assurance of precipitated heterologous proteins
US4490473A (en) * 1983-03-28 1984-12-25 Panab Labeled antibodies and methods
US4795706A (en) * 1985-01-31 1989-01-03 Eli Lilly And Company Novel expression control sequences
US5593865A (en) * 1985-10-23 1997-01-14 Boehringer Mannheim Gmbh Process for the activating of gene-technologically produced, heterologous, disulphide bridge-containing eukaryotic proteins after expression in prokaryotes
US4929700A (en) * 1987-04-16 1990-05-29 Cetus Corporation Production of purified, biologically active, bacterially produced recombinant human CSF-1
US5618927A (en) * 1990-11-22 1997-04-08 Boehringer Mannheim Gmbh Process for the reactivation of denatured protein
US5322699A (en) * 1991-02-04 1994-06-21 The Rockefeller University Leukocyte-derived CR3 modulator, integrin modulating factor-1 (IMF-1)
US5410026A (en) * 1991-12-06 1995-04-25 Genentech, Inc. Method for refolding insoluble, misfolded insulin-like growth factor-1 into an active conformation
US5288931A (en) * 1991-12-06 1994-02-22 Genentech, Inc. Method for refolding insoluble, misfolded insulin-like growth factor-I into an active conformation
US20010041521A1 (en) * 1999-07-29 2001-11-15 Chartered Semiconductor Manufacturing Ltd. Adjustable and extended guide rings
US6583268B2 (en) * 2000-01-25 2003-06-24 Oklahoma Medical Research Foundation Universal procedure for refolding recombinant proteins
US20030199676A1 (en) * 2000-01-25 2003-10-23 Oklahoma Medical Research Foundation Universal procedure for refolding recombinant proteins
US20030120042A1 (en) * 2000-03-30 2003-06-26 Takao Yamada Process for producing recombinant protein
US20030190316A1 (en) * 2000-08-11 2003-10-09 Masaya Kakuta Stabilized antibody-containing preparations
US20050176109A1 (en) * 2004-01-21 2005-08-11 Ryosuke Yumioka Purification method which prevents denaturation of an antibody
US20060199948A1 (en) * 2005-03-03 2006-09-07 Daisuke Ejima Method for improving recovery yield in protein purification with gel filtration chromatography

Cited By (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR101063347B1 (ko) 2009-04-06 2011-09-07 인하대학교 산학협력단 이온성 액체를 이용한 이황화 결합을 포함하는 단백질의 재접힘 방법
US20100273216A1 (en) * 2009-04-17 2010-10-28 Paul Morin Methods for protein refolding
US8067201B2 (en) 2009-04-17 2011-11-29 Bristol-Myers Squibb Company Methods for protein refolding
WO2012054679A1 (en) * 2010-10-20 2012-04-26 Medimmune, Llc Methods for processing inclusion bodies
CN103168046A (zh) * 2010-10-20 2013-06-19 米迪缪尼有限公司 用于加工包涵体的方法
US20130273607A1 (en) * 2010-10-20 2013-10-17 Medimmune, Llc Methods for processing inclusion bodies
JP2013540157A (ja) * 2010-10-20 2013-10-31 メディミューン,エルエルシー 封入体を処理する方法
EP2630153A4 (en) * 2010-10-20 2015-06-03 Medimmune Llc PROCESS FOR PROCESSING INCLUSIVE BODIES
US10444124B2 (en) 2011-05-20 2019-10-15 Riken Clarifying reagent for biological materials and use thereof
US9920124B2 (en) * 2012-12-20 2018-03-20 Medimmune, Llc Methods of producing immunoconjugates
US11345722B2 (en) * 2013-02-12 2022-05-31 Bristol-Myers Squibb Company High pH protein refolding methods
US10267714B2 (en) 2013-08-14 2019-04-23 Riken Composition for preparing biomaterial with excellent light-transmitting property, and use thereof
US20160266016A1 (en) 2013-08-14 2016-09-15 Riken Composition for preparing biomaterial with excellent light-transmitting property, and use thereof
WO2016106229A1 (en) * 2014-12-23 2016-06-30 Armo Biosciences, Inc. Methods of improving yield in recombinant protein production
WO2018211529A1 (en) 2017-05-19 2018-11-22 Council Of Scientific & Industrial Research A method for producing refolded recombinant humanized ranibizumab
US11524996B2 (en) 2017-05-19 2022-12-13 Council Of Scientific & Industrial Research Method for producing refolded recombinant humanized ranibizumab
US11578122B2 (en) 2017-05-19 2023-02-14 Council Of Scientific & Industrial Research IPTG-free induction process for expression of biosimilar rHu Ranibizumab antibody fragment using E. coli

Also Published As

Publication number Publication date
CA2656835A1 (en) 2008-01-17
JP2013121362A (ja) 2013-06-20
MY169472A (en) 2019-04-12
CN101506222B (zh) 2013-10-02
SG162834A1 (en) 2010-07-29
ZA200900229B (en) 2010-04-28
MX2009000278A (es) 2009-01-26
RU2009105080A (ru) 2010-08-27
NZ573639A (en) 2012-03-30
KR101520105B1 (ko) 2015-05-21
AU2007272412A1 (en) 2008-01-17
BRPI0713252C1 (pt) 2021-05-25
TWI476207B (zh) 2015-03-11
AU2007272412B2 (en) 2013-11-07
IL196004A0 (en) 2011-08-01
WO2008008975A8 (en) 2008-10-16
US9200030B2 (en) 2015-12-01
WO2008008975A3 (en) 2008-04-10
EP2041154A2 (en) 2009-04-01
US20110294990A1 (en) 2011-12-01
JP6120356B2 (ja) 2017-04-26
KR20090039772A (ko) 2009-04-22
JP5566104B2 (ja) 2014-08-06
CN101506222A (zh) 2009-08-12
IL196004A (en) 2014-06-30
US20160137690A1 (en) 2016-05-19
CA2656835C (en) 2017-12-19
BRPI0713252A2 (pt) 2012-04-10
BRPI0713252B1 (pt) 2018-12-18
TW200813093A (en) 2008-03-16
BRPI0713252B8 (pt) 2019-04-09
AR062069A1 (es) 2008-10-15
RU2439076C2 (ru) 2012-01-10
JP2009543882A (ja) 2009-12-10
US20130123474A1 (en) 2013-05-16
WO2008008975A2 (en) 2008-01-17
EP2041154B1 (en) 2018-12-12
US9994612B2 (en) 2018-06-12

Similar Documents

Publication Publication Date Title
US9994612B2 (en) Refolding of recombinant proteins
US8906648B2 (en) Recombinant production of vascular endothelial growth factor
ZA200904274B (en) Recombinant production of heparin binding proteins
MX2008008037A (en) Diaminophenothiazine compositions and uses thereof

Legal Events

Date Code Title Description
AS Assignment

Owner name: GENENTECH, INC., CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:PIZARRO, SHELLY;SANCHEZ, AILEN;SCHMELZER, CHARLES H.;REEL/FRAME:019729/0870;SIGNING DATES FROM 20070810 TO 20070813

STCB Information on status: application discontinuation

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