US20100075375A1 - Methods for the purification of polypeptide conjugates - Google Patents

Methods for the purification of polypeptide conjugates Download PDF

Info

Publication number
US20100075375A1
US20100075375A1 US12/444,380 US44438007A US2010075375A1 US 20100075375 A1 US20100075375 A1 US 20100075375A1 US 44438007 A US44438007 A US 44438007A US 2010075375 A1 US2010075375 A1 US 2010075375A1
Authority
US
United States
Prior art keywords
polypeptide
epo
peg
kda
buffer
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/444,380
Other languages
English (en)
Inventor
Shawn DeFrees
Kyle Kinealy
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.)
Novo Nordisk AS
Original Assignee
Novo Nordisk AS
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 Novo Nordisk AS filed Critical Novo Nordisk AS
Priority to US12/444,380 priority Critical patent/US20100075375A1/en
Assigned to NOVO NORDISK A/S reassignment NOVO NORDISK A/S ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: NEOSE TECHNOLOGIES, INC.
Assigned to NOVO NORDISK A/S reassignment NOVO NORDISK A/S ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: KINEALY, KYLE, DEFREES, SHAWN
Publication of US20100075375A1 publication Critical patent/US20100075375A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12PFERMENTATION OR ENZYME-USING PROCESSES TO SYNTHESISE A DESIRED CHEMICAL COMPOUND OR COMPOSITION OR TO SEPARATE OPTICAL ISOMERS FROM A RACEMIC MIXTURE
    • C12P21/00Preparation of peptides or proteins
    • C12P21/005Glycopeptides, glycoproteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • A61K47/60Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes the organic macromolecular compound being a polyoxyalkylene oligomer, polymer or dendrimer, e.g. PEG, PPG, PEO or polyglycerol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • 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
    • C07K1/13Labelling of peptides
    • 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
    • C07K14/505Erythropoietin [EPO]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/14Extraction; Separation; Purification
    • C07K1/16Extraction; Separation; Purification by chromatography
    • C07K1/18Ion-exchange chromatography
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S930/00Peptide or protein sequence
    • Y10S930/01Peptide or protein sequence
    • Y10S930/09Erythropoietin; related peptides

Definitions

  • the invention pertains to the field of polypeptide manufacturing.
  • the invention relates to processes for the purification of polypeptide conjugates, especially those conjugates including poly(alkylene oxide)-based modification groups.
  • a glycosylated or non-glycosylated polypeptide When a glycosylated or non-glycosylated polypeptide is subjected to a chemical modification reaction, side-products may be formed in addition to the desired modified polypeptide.
  • the process In order to isolate a desired product from a reaction mixture, the process must not only be suitable to remove chemical reagents, but must also be capable of removing unwanted side-products. This is especially important when the polypeptide is to be used as a therapeutic agent.
  • Polypeptide modification technologies which rely on the specificity of enzymes, may result in a reaction product that is characterized by improved homogeneity when compared to other chemical methods.
  • a recombinant polypeptide in a cell typically results in a polypeptide population that, at leas to some extend, is characterized by a variety of glycan structures. Subsequent modification of the polypeptide, e.g., via those glycans, results in a heterogenous product. Although remodeling glycan structures prior to chemical or enzymatic modification of the polypeptide can significantly improve the quality of the product, a certain degree of heterogeneity remains.
  • the present invention addresses these and other needs.
  • the present invention provides processes for the isolation (e.g., large-scale purification) of polypeptide conjugates.
  • the polypeptide conjugates of the present invention include a polypeptide that is modified with a modifying group, such as a polymer.
  • Exemplary polymers include water-soluble polymers.
  • the methods of the invention are particularly useful for the isolation of polypeptide conjugates that include poly(alkylene oxide)-based polymers, such as poly(ethylene glycol) and poly(propylene glycol). While reverse-phase (RP) chromatography can be used to purify polypeptides that are derivatized with such highly polar, water-soluble polymers, the technique is not desirable because it requires the use of water-soluble organic solvents, such as acetonitrile.
  • RP reverse-phase
  • the current invention provides methods that allow for the isolation of polypeptide conjugates essentially without the use of organic solvents.
  • An exemplary method of the invention involves at least one chromatographic procedure that is effective in separating polypeptide conjugates having at least one poly(alkylene oxide) moiety from other components of a mixture.
  • the methods of the invention can be used to isolate such polypeptide conjugate from any mixture.
  • the mixture is a reaction mixture (e.g., the product of a chemical PEGylation reaction or an ezymatically catalyzed PEGylation reaction, e.g., glycoPEGylation reaction) and may optionally include other polypeptide conjugates.
  • Preferred methods of the invention utilize hydrophobic interaction chromatography (HIC) media.
  • HIC is used in conjunction with at least one additional chromatography step selected from anion exchange chromatography, mixed-mode chromatography, cation exchange chromatography and hydroxyapatite or fluoroapatite chromatography.
  • HIC is used in conjunction with at least one of anion exchange chromatography, mixed-mode chromatography and cation exchange chromatography.
  • HIC in conjunction with cation chromatography represents an efficient method for the resolution of polypeptide conjugates that include at least one poly(alkylene oxide moiety).
  • HIC, followed by cation exchange can resolve EPO-PEG 3 species from EPO-PEG 2 species.
  • HIC in conjunction with cation exchange provided a composition of purified EPO-[PEG(10 kDa)] 3 having a very low residual concentration of EPO-[PEG(10 kDa)] 2 .
  • FIG. 1 An exemplary method of the invention that includes anion exchange and cation exchange chromatography in addition to HIC is outlined in FIG. 1 .
  • the methods of the invention are useful for the separation of different glycoforms of a polypeptide conjugate, especially those glycoforms distinguished by the number of poly(alkylene oxide) moieties that are linked to the polypeptide. Unwanted glycoforms may be formed as by-products under the reaction conditions used to form the desired polypeptide conjugate.
  • the invention provides a method of making a composition that includes a first polypeptide conjugate, the first polypeptide conjugate having a first number of poly(alkylene oxide) moieties covalently linked to the first polypeptide.
  • the method includes: (a) contacting a mixture containing the first polypeptide conjugate with a hydrophobic interaction chromatography (HIC) medium; and (b) eluting the first polypeptide conjugate from the HIC medium.
  • the mixture includes a second polypeptide conjugate, wherein the second polypeptide conjugate has a second number of poly(alkylene oxide) moieties covalently linked to the second polypeptide, wherein the first number and the second number are different.
  • the first polypeptide conjugate includes 3 poly(alkylene oxide) moieties, while the second polypeptide conjugate includes either 0, 1, 2 or 4 poly(alkylene oxide) moieties.
  • the poly(alkylene oxide) is poly(ethylene glycol) (PEG).
  • the invention provides a method of isolating a first polypeptide conjugate including a first number of poly(alkylene oxide) moieties covalently linked to a first polypeptide, from a second polypeptide conjugate that includes a second number of poly(alkylene oxide) moieties covalently linked to a second polypeptide, wherein the first number is selected from 1 to 20 and the second number is selected from 0-20, the first number and the second number being different.
  • the method includes: (a) contacting a mixture containing the first polypeptide conjugate and the second polypeptide conjugate with a hydrophobic interaction chromatography (HIC) medium; and (b) eluting the first polypeptide conjugate from said hydrophobic interaction chromatography medium.
  • the first polypeptide conjugate includes 3 poly(alkylene oxide) moieties
  • the second polypeptide conjugate includes 0, 1, 2, 4, 5, 6 or 7 poly(alkylene oxide) moieties.
  • first polypeptide and the second polypeptide have the same amino acid sequence. In another example according to any of the above embodiments, both the first and the second polypeptide are EPO.
  • the invention provides a method of forming a composition that contains a first erythropoietin (EPO) conjugate, wherein the first EPO conjugate includes a first number of poly(alkylene oxide) moieties covalently linked to an EPO polypeptide.
  • the method includes: (a) contacting a mixture containing the first EPO conjugate with an anion exchange medium; (b) eluting the first EPO conjugate from the anion exchange medium, forming a first eluate including the first EPO conjugate; (c) contacting the first eluate with a hydrophobic interaction chromatography (HIC) medium; and (d) eluting the first EPO conjugate from the hydrophobic interaction chromatography medium.
  • the method may further include (e.g., after step d): (e) eluting the first EPO conjugate from a cation exchange chromatography medium.
  • the method further includes forming the polypeptide conjugate either chemically or through enzymatically catalyzed glycomodification (e.g., glycoPEGylation using a glycosyltransferase and an appropriate glycosyl donor molecule, such as a modified sugar nucleotide).
  • glycoPEGylation methods are art-recognized; see for example, WO 03/031464 to DeFrees et al. or WO 04/99231, the disclosures of which are incorporated herein by reference in their entirety.
  • the invention further provides compositions, which are made by the methods of the invention as well as pharmaceutical formulations including the composition of the invention.
  • the invention provides methods of treatment utilizing the compositions of the invention.
  • FIG. 1 is an overall view of an exemplary polypeptide conjugate purification process according to a method of the invention.
  • the diafiltration/ultrafiltration step following hydrophobic interaction chromatography (HIC) is optional.
  • FIG. 2A is an exemplary chromatogram showing the resolution of tri-PEGylated EPO from mono-, di-, tri- and tetra-PEGylated EPO glycoforms using hydrophobic interaction chromatography with Phenyl 650S as the separation medium.
  • FIG. 2B is an expanded view of the EPO-(SA-PEG-10 kDa) 2 , EPO-(SA-PEG-10 kDa) 3 and EPO-(SA-PEG-10 kDa) 4 elution peaks of the chromatogram in FIG. 2A .
  • the letters (E), (F) and (G) indicate fractions that were pooled, wherein (F) indicates the elution of EPO-(SA-PEG-10 kDa) 3 .
  • FIG. 3A is a scheme of an exemplary EPO polypeptide conjugate of the invention having an insect-specific glycosylation pattern that includes three N-linked, monoantennary glycan residues covalently linked to amino acid residues N24, N38 and N83. Each glycan residue is covalently linked to a 10 kDa PEG moiety via a terminal galactose (Gal) moiety.
  • FIG. 3A also includes an exemplary reaction scheme, which can be used to synthesize the EPO conjugate.
  • the substrate for the enzymatically catalyzed conversions is an EPO polypeptide, which includes at least one glycan residue having a trimannosyl moiety.
  • a first step an N-acetyl glucosamine transferase (GnT-1) is used, which adds a GlcNAc moiety to only one of the terminal mannose moieties.
  • a Gal moiety is linked to the newly added GlcNAc moiety using a galactosyl transferase (GalT-1) forming a terminal -GlcNAc-Gal moiety.
  • the first and the second step maybe performed in the same reaction vessel.
  • a sialic acid moiety that is modified with a PEG moiety is linked to the terminal Gal moiety using a sialyl transferase (ST3Gal3).
  • FIG. 3B is a representation of an exemplary composition of the invention that includes various glycoforms of an exemplary polypeptide conjugate (e.g., EPO conjugate).
  • Each glycoform is distinguished from other glycoforms by the number of PEG moieties that are covalently linked to the polypeptide, or by the structure of the glycans through which the PEG moieties are linked to the polypeptide. Shown percentage values are exemplary.
  • FIG. 4A is a reverse phase (RP) HPLC chromatogram of an exemplary glycoPEGylation reaction mixture containing EPO-(SA-PEG-10 kDa) 1-4 performed at a 25 mg scale.
  • a Zorbax 300SB-C3 (150 ⁇ 2.1 mm, 5 micron) column was used in the analysis.
  • the following eluants were used: 0.1% TFA in water (Buffer A) and 0.09% TFA in CAN (Buffer B).
  • the gradient was 42-55% B in 14 min followed by 55-95% B in 2 min.
  • the flow rate was 0.6 mL/min. Absorption was measured at 214 nm.
  • FIG. 4B is a reverse phase (RP) HPLC chromatogram of an exemplary composition of the invention containing purified EPO-(SA-PEG-10 kDa) 3 as the major component, the composition obtained using a method of the invention.
  • FIG. 5 is a schematic representation of exemplary glycopegylated EPO isoforms isolated from Chinese Hamster Ovary cells.
  • A An exemplary 40 kilodaton O-linked pegylated glycoform.
  • B One of several 30 kilodatton N-linked pegylated glycoforms.
  • the modified sialic acid moiety comprising the PEG molecule may occur on any one or more of any of the branches of the N-linked glycosyl residue.
  • any glycosylated EPO molecule may comprise any mixture of mono-, bi- tri-, or tetra-antennary N-linked glycosyl residues and any one or more of the branches may further comprise a modified sialic acid moiety.
  • PEG poly(ethyleneglycol); PPG, poly(propyleneglycol); Ara, arabinosyl; Fru, fructosyl; Fuc, fucosyl; Gal, galactosyl; GalNAc, N-acetylgalactosaminyl; Glc, glucosyl; GlcNAc, N-acetylglucosaminyl; Man, mannosyl; ManAc, mannosaminyl acetate; Xyl, xylosyl; and NeuAc, sialyl (N-acetylneuraminyl); M6P, mannose-6-phosphate; BEVS, baculovirus expression vector system; CV, column volume; NTU, nominal turbidity units; vvm, volume/volume/min.
  • oligosaccharides described herein are described with the name or abbreviation for the non-reducing saccharide (i.e., Gal), followed by the configuration of the glycosidic bond ( ⁇ or ⁇ ), the ring bond (1 or 2), the ring position of the reducing saccharide involved in the bond (2, 3, 4, 6 or 8), and then the name or abbreviation of the reducing saccharide (i.e., GlcNAc).
  • Each saccharide is preferably a pyranose.
  • Oligosaccharides are considered to have a reducing end and a non-reducing end, whether or not the saccharide at the reducing end is in fact a reducing sugar. In accordance with accepted nomenclature, oligosaccharides are depicted herein with the non-reducing end on the left and the reducing end on the right.
  • substituent groups are specified by their conventional chemical formulae, written from left to right, they equally encompass the chemically identical substituents, which would result from writing the structure from right to left, e.g., —CH 2 O— is intended to also recite —OCH 2 —.
  • alkyl by itself or as part of another substituent, means, unless otherwise stated, a straight or branched chain, or cyclic (i.e., cycloalkyl)hydrocarbon radical, or combination thereof, which may be fully saturated, mono- or polyunsaturated and can include di-(e.g., alkylene) and multivalent radicals, having the number of carbon atoms designated (i.e. C 1 -C 10 means one to ten carbons).
  • saturated hydrocarbon radicals include, but are not limited to, groups such as methyl, ethyl, n-propyl, isopropyl, n-butyl, t-butyl, isobutyl, sec-butyl, cyclohexyl, (cyclohexyl)methyl, cyclopropylmethyl, homologs and isomers of, for example, n-pentyl, n-hexyl, n-heptyl, n-octyl, and the like.
  • An unsaturated alkyl group is one having one or more double bonds or triple bonds.
  • alkyl groups examples include, but are not limited to, vinyl, 2-propenyl, crotyl, 2-isopentenyl, 2-(butadienyl), 2,4-pentadienyl, 3-(1,4-pentadienyl), ethynyl, 1- and 3-propynyl, 3-butynyl, and the higher homologs and isomers.
  • alkyl unless otherwise noted, is also meant to include those derivatives of alkyl defined in more detail below, such as “heteroalkyl.” Alkyl groups that are limited to hydrocarbon groups are termed “homoalkyl”.
  • alkylene by itself or as part of another substituent means a divalent radical derived from an alkane, as exemplified, but not limited, by —CH 2 CH 2 CH 2 CH 2 —, and further includes those groups described below as “heteroalkylene.”
  • an alkyl (or alkylene) group will have from 1 to 24 carbon atoms, with those groups having 10 or fewer carbon atoms being preferred in the present invention.
  • a “lower alkyl” or “lower alkylene” is a shorter chain alkyl or alkylene group, generally having eight or fewer carbon atoms.
  • alkoxy alkylamino and “alkylthio” (or thioalkoxy) are used in their conventional sense, and refer to those alkyl groups attached to the remainder of the molecule via an oxygen atom, an amino group, or a sulfur atom, respectively.
  • heteroalkyl by itself or in combination with another term, means, unless otherwise stated, a stable straight or branched chain, or cyclic hydrocarbon radical, or combinations thereof, consisting of the stated number of carbon atoms and at least one heteroatom selected from the group consisting of O, N, Si and S, and wherein the nitrogen and sulfur atoms may optionally be oxidized and the nitrogen heteroatom may optionally be quaternized.
  • the heteroatom(s) O, N and S and Si may be placed at any interior position of the heteroalkyl group or at the position at which the alkyl group is attached to the remainder of the molecule.
  • Examples include, but are not limited to, —CH 2 —CH 2 —O—CH 3 , —CH 2 —CH 2 —NH—CH 3 , —CH 2 —CH 2 —N(CH 3 )—CH 3 , —CH 2 —S—CH 2 —CH 3 , —CH 2 —CH 2 , —S(O)—CH 3 , —CH 2 —CH 2 —S(O) 2 —CH 3 , —CH ⁇ CH—O—CH 3 , —Si(CH 3 ) 3 , —CH 2 —CH ⁇ N—OCH 3 , and CH ⁇ CH—N(CH 3 )—CH 3 .
  • heteroalkylene by itself or as part of another substituent means a divalent radical derived from heteroalkyl, as exemplified, but not limited by, —CH 2 —CH 2 —S—CH 2 —CH 2 — and CH 2 —S—CH 2 —CH 2 —NH—CH 2 —.
  • heteroatoms can also occupy either or both of the chain termini (e.g., alkyleneoxy, alkylenedioxy, alkyleneamino, alkylenediamino, and the like). Still further, for alkylene and heteroalkylene linking groups, no orientation of the linking group is implied by the direction in which the formula of the linking group is written. For example, the formula —CO 2 R′— represents both —C(O)OR′ and —OC(O)R′.
  • cycloalkyl and “heterocycloalkyl”, by themselves or in combination with other terms, represent, unless otherwise stated, cyclic versions of “alkyl” and “heteroalkyl”, respectively. Additionally, for heterocycloalkyl, a heteroatom can occupy the position at which the heterocycle is attached to the remainder of the molecule. Examples of cycloalkyl include, but are not limited to, cyclopentyl, cyclohexyl, 1-cyclohexenyl, 3-cyclohexenyl, cycloheptyl, and the like.
  • heterocycloalkyl examples include, but are not limited to, 1-(1,2,5,6-tetrahydropyridyl), 1-piperidinyl, 2-piperidinyl, 3-piperidinyl, 4-morpholinyl, 3-morpholinyl, tetrahydrofuran-2-yl, tetrahydrofuran-3-yl, tetrahydrothien-2-yl, tetrahydrothien-3-yl, 1-piperazinyl, 2-piperazinyl, and the like.
  • halo or “halogen,” by themselves or as part of another substituent, mean, unless otherwise stated, a fluorine, chlorine, bromine, or iodine atom. Additionally, terms such as “haloalkyl,” are meant to include monohaloalkyl and polyhaloalkyl.
  • halo(C 1 -C 4 )alkyl is mean to include, but not be limited to, trifluoromethyl, 2,2,2-trifluoroethyl, 4-chlorobutyl, 3-bromopropyl, and the like.
  • aryl means, unless otherwise stated, a polyunsaturated, aromatic, substituent that can be a single ring or multiple rings (preferably from 1 to 3 rings), which are fused together or linked covalently.
  • heteroaryl refers to aryl groups (or rings) that contain from one to four heteroatoms selected from N, O, S, Si and B, wherein the nitrogen and sulfur atoms are optionally oxidized, and the nitrogen atom(s) are optionally quaternized.
  • a heteroaryl group can be attached to the remainder of the molecule through a heteroatom.
  • Non-limiting examples of aryl and heteroaryl groups include phenyl, 1-naphthyl, 2-naphthyl, 4-biphenyl, 1-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 3-pyrazolyl, 2-imidazolyl, 4-imidazolyl, pyrazinyl, 2-oxazolyl, 4-oxazolyl, 2-phenyl-4-oxazolyl, 5-oxazolyl, 3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, 2-furyl, 3-furyl, 2-thienyl, 3-thienyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidyl, 4-pyrimidyl, 5-benzothiazolyl, purinyl, 2-benzimidazolyl, 5-indolyl, 1-isoquinoly
  • aryl when used in combination with other terms (e.g., aryloxy, arylthioxy, arylalkyl) includes both aryl and heteroaryl rings as defined above.
  • arylalkyl is meant to include those radicals in which an aryl group is attached to an alkyl group (e.g., benzyl, phenethyl, pyridylmethyl and the like) including those alkyl groups in which a carbon atom (e.g., a methylene group) has been replaced by, for example, an oxygen atom (e.g., phenoxymethyl, 2-pyridyloxymethyl, 3-(1-naphthyloxy)propyl, and the like).
  • alkyl group e.g., benzyl, phenethyl, pyridylmethyl and the like
  • an oxygen atom e.g., phenoxymethyl, 2-pyridyloxymethyl, 3-(1-naph
  • alkyl e.g., “alkyl,” “heteroalkyl,” “aryl” and “heteroaryl” are meant to include both substituted and unsubstituted forms of the indicated radical.
  • Preferred substituents for each type of radical are provided below.
  • alkyl and heteroalkyl radicals are generically referred to as “alkyl group substituents,” and they can be one or more of a variety of groups selected from, but not limited to: substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted heterocycloalkyl, —OR′, ⁇ O, ⁇ NR′, ⁇ N—OR′, —NR′R′′, —SR′, -halogen, —SiR′R′′R′′′, —OC(O)R′, —C(O)R′, —CO 2 R′, —CONR′R′′, —OC(O)NR′R
  • R′, R′′, R′′′ and R′′′′ each preferably independently refer to hydrogen, substituted or unsubstituted heteroalkyl, substituted or unsubstituted aryl, e.g., aryl substituted with 1-3 halogens, substituted or unsubstituted alkyl, alkoxy or thioalkoxy groups, or arylalkyl groups.
  • each of the R groups is independently selected as are each R′, R′′, R′′′ and R′′′′ groups when more than one of these groups is present.
  • R′ and R′′ are attached to the same nitrogen atom, they can be combined with the nitrogen atom to form a 5-, 6-, or 7-membered ring.
  • —NR′R′′ is meant to include, but not be limited to, 1-pyrrolidinyl and 4-morpholinyl.
  • alkyl is meant to include groups including carbon atoms bound to groups other than hydrogen groups, such as haloalkyl (e.g., —CF 3 and —CH 2 CF 3 ) and acyl (e.g., —C(O)CH 3 , —C(O)CF 3 , —C(O)CH 2 OCH 3 , and the like).
  • haloalkyl e.g., —CF 3 and —CH 2 CF 3
  • acyl e.g., —C(O)CH 3 , —C(O)CF 3 , —C(O)CH 2 OCH 3 , and the like.
  • substituents for the aryl and heteroaryl groups are generically referred to as “aryl group substituents.”
  • the substituents are selected from, for example: substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted heterocycloalkyl, —OR′, ⁇ O, ⁇ NR′, ⁇ N—OR′, —NR′R′′, —SR′, -halogen, —SiR′R′′R′′′, —OC(O)R′, —C(O)R′, —CO 2 R′, —CONR′R′′, —OC(O)NR′R′′, —NR′′C(O)R′, —NR′—C(O)NR′′R′′′, —NR′′C(O) 2 R′,
  • Two of the substituents on adjacent atoms of the aryl or heteroaryl ring may optionally be replaced with a substituent of the formula -T-C(O)—(CRR′) q —U—, wherein T and U are independently —NR—, —O—, —CRR′— or a single bond, and q is an integer of from 0 to 3.
  • two of the substituents on adjacent atoms of the aryl or heteroaryl ring may optionally be replaced with a substituent of the formula -A-(CH 2 ) r —B—, wherein A and B are independently —CRR′—, —O—, —NR—, —S—, —S(O)—, —S(O) 2 —, —S(O) 2 NR′— or a single bond, and r is an integer of from 1 to 4.
  • One of the single bonds of the new ring so formed may optionally be replaced with a double bond.
  • two of the substituents on adjacent atoms of the aryl or heteroaryl ring may optionally be replaced with a substituent of the formula —(CRR′) s —X—(CR′′R′′′) d —, where s and d are independently integers of from 0 to 3, and X is —O—, —NR′—, —S—, —S(O)—, —S(O) 2 —, or S(O) 2 NR′—.
  • the substituents R, R′, R′′ and R′′′ are preferably independently selected from hydrogen or substituted or unsubstituted (C 1 -C 6 )alkyl.
  • acyl describes a substituent containing a carbonyl residue, C(O)R.
  • R exemplary species for R include H, halogen, alkoxy, substituted or unsubstituted alkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, and substituted or unsubstituted heterocycloalkyl.
  • fused ring system means at least two rings, wherein each ring has at least 2 atoms in common with another ring. “Fused ring systems may include aromatic as well as non aromatic rings. Examples of “fused ring systems” are naphthalenes, indoles, quinolines, chromenes and the like.
  • heteroatom includes oxygen (O), nitrogen (N), sulfur (S), silicon (Si) and boron (B).
  • R is a general abbreviation that represents a substituent group.
  • substituent groups include substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, and substituted or unsubstituted heterocycloalkyl groups.
  • salts includes salts of the active compounds which are prepared with relatively nontoxic acids or bases, depending on the particular substituents found on the compounds described herein.
  • base addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired base, either neat or in a suitable inert solvent.
  • pharmaceutically acceptable base addition salts include sodium, potassium, calcium, ammonium, organic amino, or magnesium salt, or a similar salt.
  • acid addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the desired acid, either neat or in a suitable inert solvent.
  • Examples of pharmaceutically acceptable acid addition salts include those derived from inorganic acids like hydrochloric, hydrobromic, nitric, carbonic, monohydrogencarbonic, phosphoric, monohydrogenphosphoric, dihydrogenphosphoric, sulfuric, monohydrogensulfuric, hydriodic, or phosphorous acids and the like, as well as the salts derived from relatively nontoxic organic acids like acetic, propionic, isobutyric, maleic, malonic, benzoic, succinic, suberic, fumaric, lactic, mandelic, phthalic, benzenesulfonic, p-tolylsulfonic, citric, tartaric, methanesulfonic, and the like.
  • inorganic acids like hydrochloric, hydrobromic, nitric, carbonic, monohydrogencarbonic, phosphoric, monohydrogenphosphoric, dihydrogenphosphoric, sulfuric, monohydrogensulfuric, hydriodic, or phosphorous acids and
  • salts of amino acids such as arginate and the like, and salts of organic acids like glucuronic or galactunoric acids and the like (see, for example, Berge et al., Journal of Pharmaceutical Science, 66: 1-19 (1977)).
  • Certain specific compounds of the present invention contain both basic and acidic functionalities that allow the compounds to be converted into either base or acid addition salts.
  • the neutral forms of the compounds are preferably regenerated by contacting the salt with a base or acid and isolating the parent compound in the conventional manner.
  • the parent form of the compound differs from the various salt forms in certain physical properties, such as solubility in polar solvents, but otherwise the salts are equivalent to the parent form of the compound for the purposes of the present invention.
  • the present invention provides compounds, which are in a prodrug form.
  • Prodrugs of the compounds described herein are those compounds that readily undergo chemical changes under physiological conditions to provide the compounds of the present invention.
  • prodrugs can be converted to the compounds of the present invention by chemical or biochemical methods in an ex vivo environment. For example, prodrugs can be slowly converted to the compounds of the present invention when placed in a transdermal patch reservoir with a suitable enzyme or chemical reagent.
  • Certain compounds of the present invention can exist in unsolvated forms as well as solvated forms, including hydrated forms. In general, the solvated forms are equivalent to unsolvated forms and are encompassed within the scope of the present invention. Certain compounds of the present invention may exist in multiple crystalline or amorphous forms. In general, all physical forms are equivalent for the uses contemplated by the present invention and are intended to be within the scope of the present invention.
  • Certain compounds of the present invention possess asymmetric carbon atoms (optical centers) or double bonds; the racemates, diastereomers, geometric isomers and individual isomers are encompassed within the scope of the present invention.
  • the compounds of the invention may be prepared as a single isomer (e.g., enantiomer, cis-trans, positional, diastereomer) or as a mixture of isomers.
  • the compounds are prepared as substantially a single isomer.
  • Methods of preparing substantially isomerically pure compounds are known in the art. For example, enantiomerically enriched mixtures and pure enantiomeric compounds can be prepared by using synthetic intermediates that are enantiomerically pure in combination with reactions that either leave the stereochemistry at a chiral center unchanged or result in its complete inversion. Alternatively, the final product or intermediates along the synthetic route can be resolved into a single stereoisomer.
  • enantiomeric excess and diastereomeric excess are used interchangeably herein.
  • Compounds with a single stereocenter are referred to as being present in “enantiomeric excess,” those with at least two stereocenters are referred to as being present in “diastereomeric excess.”
  • the compounds of the present invention may also contain unnatural proportions of atomic isotopes at one or more of the atoms that constitute such compounds.
  • the compounds may be radiolabeled with radioactive isotopes, such as for example tritium ( 3 H), iodine-125 ( 125 I) or carbon-14 ( 14 C). All isotopic variations of the compounds of the present invention, whether radioactive or not, are intended to be encompassed within the scope of the present invention.
  • Reactive functional group refers to groups including, but not limited to, olefins, acetylenes, alcohols, phenols, ethers, oxides, halides, aldehydes, ketones, carboxylic acids, esters, amides, cyanates, isocyanates, thiocyanates, isothiocyanates, amines, hydrazines, hydrazones, hydrazides, diazo, diazonium, nitro, nitriles, mercaptans, sulfides, disulfides, sulfoxides, sulfones, sulfonic acids, sulfinic acids, acetals, ketals, anhydrides, sulfates, sulfenic acids isonitriles, amidines, imides, imidates, nitrones, hydroxylamines, oximes, hydroxamic acids thiohydroxamic acids, allenes, ortho
  • Reactive functional groups also include those used to prepare bioconjugates, e.g., N-hydroxysuccinimide esters, maleimides and the like. Methods to prepare each of these functional groups are well known in the art and their application or modification for a particular purpose is within the ability of one of skill in the art (see, for example, Sandler and Karo, eds. O RGANIC F UNCTIONAL G ROUP P REPARATIONS , Academic Press, San Diego, 1989).
  • “pharmaceutically acceptable carrier” includes any material, which when combined with the conjugate retains the conjugates' activity and is non-reactive with the subject's immune systems.
  • “Pharmaceutically acceptable carrier” includes solids and liquids, such as vehicles, diluents and solvents. Examples include, but are not limited to, any of the standard pharmaceutical carriers such as a phosphate buffered saline solution, water, emulsions such as oil/water emulsion, and various types of wetting agents. Other carriers may also include sterile solutions, tablets including coated tablets and capsules.
  • Such carriers typically contain excipients such as starch, milk, sugar, certain types of clay, gelatin, stearic acid or salts thereof, magnesium or calcium stearate, talc, vegetable fats or oils, gums, glycols, or other known excipients.
  • excipients such as starch, milk, sugar, certain types of clay, gelatin, stearic acid or salts thereof, magnesium or calcium stearate, talc, vegetable fats or oils, gums, glycols, or other known excipients.
  • Such carriers may also include flavor and color additives or other ingredients.
  • Compositions comprising such carriers are formulated by well known conventional methods.
  • administering means oral administration, administration as a suppository, topical contact, intravenous, intraperitoneal, intramuscular, intralesional, or subcutaneous administration, administration by inhalation, or the implantation of a slow-release device, e.g., a mini-osmotic pump, to the subject.
  • Adminsitration is by any route including parenteral and transmucosal (e.g., oral, nasal, vaginal, rectal, or transdermal), particularly by inhalation.
  • Parenteral administration includes, e.g., intravenous, intramuscular, intra-arteriole, intradermal, subcutaneous, intraperitoneal, intraventricular, and intracranial.
  • injection is to treat a tumor, e.g., induce apoptosis
  • administration may be directly to the tumor and/or into tissues surrounding the tumor.
  • Other modes of delivery include, but are not limited to, the use of liposomal formulations, intravenous infusion, transdermal patches, etc.
  • Ameliorating refers to any indicia of success in the treatment of a pathology or condition, including any objective or subjective parameter such as abatement, remission or diminishing of symptoms or an improvement in a patient's physical or mental well-being. Amelioration of symptoms can be based on objective or subjective parameters; including the results of a physical examination and/or a psychiatric evaluation.
  • therapy refers to “treating” or “treatment” of a disease or condition including preventing the disease or condition from occurring in a subject (e.g., human) that may be predisposed to the disease but does not yet experience or exhibit symptoms of the disease (prophylactic treatment), inhibiting the disease (slowing or arresting its development), providing relief from the symptoms or side-effects of the disease (including palliative treatment), and relieving the disease (causing regression of the disease).
  • an amount effective to or a “therapeutically effective amount” or any gramatically equivalent term means the amount that, when administered to an animal or human for treating a disease, is sufficient to effect treatment for that disease.
  • Insect cell culture refers to the in vitro growth and culturing of cell derived from organisms of the Class Insecta. “Insect cell culture” also refers to a cell culture comprising cells of the Class Insecta which have been grown and cultured in vitro.
  • “Peptide” refers to a polymer in which the monomers are amino acids and are joined together through amide bonds, alternatively referred to as a polypeptide. Additionally, unnatural amino acids, for example, ⁇ -alanine, phenylglycine and homoarginine are also included. Amino acids that are not gene-encoded may also be used in the present invention. Furthermore, amino acids that have been modified to include reactive groups, glycosylation sites, polymers, therapeutic moieties, biomolecules and the like may also be used in the invention. All of the amino acids used in the present invention may be either the D - or L -isomer. The L -isomer is generally preferred. In addition, other peptidomimetics are also useful in the present invention.
  • peptide refers to both glycosylated and unglycosylated peptides. Also included are petides that are incompletely glycosylated by a system that expresses the peptide. For a general review, see, Spatola, A. F., in C HEMISTRY AND B IOCHEMISTRY OF A MINO A CIDS , P EPTIDES AND P ROTEINS , B. Weinstein, eds., Marcel Dekker, New York, p. 267 (1983). The term peptide includes molecules that are commonly referred to as proteins or polypeptides.
  • glycopeptide refers to a peptide having at least one carbohydrate moiety covalently linked thereto. It is understood that a glycopeptide may be a “therapeutic glycopeptide”.
  • glycopeptide is used interchangeably herein with the terms “glycopolypeptide” and “glycoprotein.”
  • peptide conjugate refers to species of the invention in which a peptide is conjugated with a modified sugar as set forth herein.
  • modified sugar refers to a naturally- or non-naturally-occurring carbohydrate that is enzymatically added onto an amino acid or a glycosyl residue of a peptide in a process of the invention.
  • the modified sugar is selected from a number of enzyme substrates including, but not limited to sugar nucleotides (mono-, di-, and tri-phosphates), activated sugars (e.g., glycosyl halides, glycosyl mesylates) and sugars that are neither activated nor nucleotides.
  • the “modified sugar” is covalently functionalized with a “modifying group.”
  • modifying groups include, but are not limited to, PEG moieties, therapeutic moieties, diagnostic moieties, biomolecules and the like.
  • the modifying group is preferably not a naturally occurring, or an unmodified carbohydrate.
  • the locus of functionalization with the modifying group is selected such that it does not prevent the “modified sugar” from being added enzymatically to a peptide.
  • glycoconjugation refers to the enzymatically mediated conjugation of a modified sugar species to an amino acid or glycosyl residue of a polypeptide, e.g., an erythropoietin peptide prepared by the method of the present invention.
  • a subgenus of “glycoconjugation” is “glyco-PEGylation,” in which the modifying group of the modified sugar is poly(ethylene glycol), an alkyl derivative (e.g., m-PEG) or reactive derivative (e.g., H 2 N-PEG, HOOC-PEG) thereof.
  • large-scale and “industrial-scale” are used interchangeably and refer to a reaction cycle or process that produces at least about 250 mg, preferably at least about 500 mg, and more preferably at least about 1 gram of peptide at the completion of a single cycle.
  • glycosyl linking group refers to a glycosyl residue to which a modifying group (e.g., PEG moiety, therapeutic moiety, biomolecule) is covalently attached; the glycosyl linking group joins the modifying group to the remainder of the conjugate.
  • the “glycosyl linking group” becomes covalently attached to a glycosylated or unglycosylated polypeptide, thereby linking the modifying group to an amino acid and/or glycosyl residue of the polypeptide.
  • glycosyl linking group is generally derived from a “modified sugar” by the enzymatic attachment of the “modified sugar” to an amino acid and/or glycosyl residue of the polypeptide.
  • the glycosyl linking group can be a saccharide-derived structure that is degraded during formation of modifying group-modified sugar cassette (e.g., oxidation ⁇ Schiff base formation ⁇ reduction), or the glycosyl linking group may be intact.
  • an “intact glycosyl linking group” refers to a linking group that is derived from a glycosyl moiety in which the saccharide monomer that links the modifying group and to the remainder of the conjugate is not degraded, e.g., oxidized, e.g., by sodium metaperiodate.
  • “Intact glycosyl linking groups” of the invention may be derived from a naturally occurring oligosaccharide by addition of glycosyl unit(s) or removal of one or more glycosyl unit from a parent saccharide structure.
  • a “glycosyl linking group” may include a glycosyl-mimetic moiety.
  • the glycosyl transferase (e.g., sialyl transferase), which is used to add the modified sugar to a glycosylated polypeptide, exhibits tolerance for a glycosyl-mimetic substrate (e.g., a modified sugar in which the sugar moiety is a glycosyl-mimetic moiety—e.g., sialyl-mimetic moiety).
  • a glycosyl-mimetic substrate e.g., a modified sugar in which the sugar moiety is a glycosyl-mimetic moiety—e.g., sialyl-mimetic moiety.
  • the transfer of the modified glycosyl-mimetic sugar results in a conjugate having a glycosyl linking group that is a glycosyl-mimetic moiety.
  • glycosyl-mimetic moiety refers to a moiety, which structurally resembles a glycosyl moiety (e.g., a hexose or a pentose).
  • glycosyl moiety examples include those moieties, wherein the glycosidic oxygen or the ring oxygen of a glycosyl moiety, or both, has been replaced with a bond or another atom (e.g., sulfur), or another moiety, such as a carbon- (e.g., CH 2 ), or nitrogen-containing group (e.g., NH).
  • Examples include substituted or unsubstituted cyclohexyl derivatives, cyclic thioethers, cyclic secondary amines, moieties including a thioglycosidic bond, and the like.
  • the “glycosyl-mimetic moiety” is transferred in an enzymatically catalyzed reaction onto an amino acid residue of a polypeptide or a glycosyl moiety of a glycopeptide. This can, for instance, be accomplished by activating the “glycosyl-mimetic moiety” with a leaving group, such as a halogen.
  • polypeptide glycoform or “glycoform” as used herein refers to two polypeptide conjugates having the same amino acid sequence, but having a different glycosylation pattern with respect to the glycan residues to which the modifying group(s), e.g., poly(alkylene oxide) moieties, are covalently linked.
  • modifying group(s) e.g., poly(alkylene oxide) moieties
  • FIG. 3B shows exemplary glycoforms of an EPO polypeptide conjugate.
  • EPO-PEG conjugates discussed in the Examples, below, are alternatively referred to as EPO-PEG “species”, “forms” or “states”.
  • isolated refers to a material that is essentially free from components, which are used to produce the material.
  • isolated refers to a material that is essentially free from components which normally accompany the material in the mixture used to prepare the peptide conjugate.
  • isolated and pure are used interchangeably.
  • isolated peptide conjugates of the invention have a level of purity expressed as a range.
  • the lower end of the range is about 50%, about 55%, about 60%, about 65%, about 70%, about 75% or about 80% and the upper end of the range is about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, about 95% or more than about 95%.
  • the peptide conjugates are more than about 90% pure, their purities are preferably expressed as a range.
  • the lower end of the range being about 90%, about 92%, about 94%, about 96% or about 98% and the upper end of the range being about 92%, about 94%, about 96%, about 98% or about 100%.
  • Purity of a polypeptide conjugate may be determined by any suitable, art-recognized method of analysis (e.g., band intensity on a silver stained gel, polyacrylamide gel electrophoresis, ELISA, HPLC and the like).
  • An exemplary method is size-exclusion chromatography (SEC) HPLC, described herein below.
  • Purity may be determined using relative “area under the curve” (AUC) values, which can typically be obtained for peaks in a chromatogram, such as an HPLC chromatogram.
  • AUC area under the curve
  • purities are determined by chromatographic or other means using a standard curve generated using a reference material of known purity. Purity may also be determined on a weight-by-weight basis.
  • Methods that are useful for the determination of “purity” are also useful for the determination of the “concentration” of a particular component in a mixture (e.g., a composition of the invention) or relative concentration of one component with respect to one or more other components.
  • SEC HPLC may be used to determine the ratio between different glycoforms or to determine the concentration of a specific glycoform in a composition of the invention.
  • each member of the population speaks to the “homogeneity” of the sites on the peptide and to a population of peptide that share a common structure, e.g., a common glycosylation pattern glycosyl structure.
  • “Homogeneity” refers to the structural consistency across a population of polypeptides. Thus, in a glycopeptide of the invention, in which each glycan residue has the same structure, the glycopeptide is said to be about 100% homogeneous. Similarly, when a in a population of glycopeptides, each glycopeptide has glycan residues of the same structure, such that each peptide of the population is essentially of the same molecular species, the population is said to be about 100% homogeneous. Homogeneity is typically expressed as a range. The lower end of the range of homogeneity for the peptide conjugates is about 60%, about 70% or about 80% and the upper end of the range of purity is about 70%, about 80%, about 90% or more than about 90%.
  • the peptide conjugates are more than or equal to about 90% homogeneous, their homogeneity is also preferably expressed as a range.
  • the lower end of the range of homogeneity is about 90%, about 92%, about 94%, about 96% or about 98%.
  • the upper end of the range of purity is about 92%, about 94%, about 96%, about 98% or about 100%.
  • the homogeneity of the peptide conjugates is typically determined by one or more methods known to those of skill in the art, e.g., gel electrophoresis, liquid chromatography-mass spectrometry (LC-MS), matrix assisted laser desorption mass time of flight spectrometry (MALDI-TOF), capillary electrophoresis, and the like.
  • a polypeptide that includes multiple N-linked or O-linked glycosylation sites may have a glycosyl residue of the same structure present at all comparable glycosylation sites, at about 90% of all comparable sites, about 80% or about 75% of all comparable glycosylation sites. In these instances the polypeptide would be said to have a “substantially uniform glycosylation pattern”.
  • the population may be said to have a “substantially uniform glycosylation pattern” when a majority of the peptides in the population represent essentially a single molecular species.
  • peptides isolated from insect cells have a substantially uniform insect-specific glycosylation pattern. This refers to the fact that the majority of polypeptides, or substantially all of the polypeptides, in the preparation represent one distinct molecular species.
  • substantially in the above definitions of “substantially uniform” generally means at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, at least about 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, at least about 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or even 100% of the acceptor moieties are glycosylated with the expected insect cell specific glycosylation pattern.
  • insect specific glycosylation pattern refers to the glycosylation pattern found on mature glycopeptides produced by insect cells.
  • insect cells generate simple N-linked oligosaccharides terminating in mannose (for review, see e.g., Essentials of Glycobiology A. Varki et al. eds, CSHL Press (1999) pgs: 32-33).
  • N-linked glycans produced by insect cell lines produce glycoproteins that (at maturity) include a Man 3 GlcNAc 2 structure. Fucose units may also be found on the GlcNAc residue that is directly linked to the peptide.
  • a mature peptide emerging from a cell with an “insect specific glycosylation pattern” thus includes one or more glycans having a trimannosyl (Man 3 ) or Man 3 GlcNAc 2 structure.
  • “Insect specific glycosylation pattern” also refers to polypeptide populations, in which essentially all of the polypeptides have glycan structures terminating with a common motive (e.g., the Man 3 or Man 3 GlcNAc 2 motive) and are not degraded, e.g., to expose one of the two GlcNAc residues directly bound to the polypeptide.
  • loading buffer refers to the buffer, in which the polypeptide conjugate being purified is applied to a purification device, e.g. a chromatography column or a filter cartridge.
  • the loading buffer is selected so that separation of the peptide conjugate of interest from unwanted impurities can be accomplished.
  • a purification device e.g. a chromatography column or a filter cartridge.
  • the pH of the loading buffer and the salt concentration in the loading buffer may be selected so that the polypeptide conjugate is initially retained on the column while certain impurities are found in the flow through.
  • the loading buffer is selected to retain impurities while the desired polypeptide conjugate is found in the flow-through.
  • the term “elution buffer” refers to the buffer, which is typically used to remove (elute) the polypeptide conjugate from the purification device (e.g. a chromatographic column or filter cartridge) to which it was applied earlier.
  • the elution buffer is selected so that separation of the polypeptide conjugate of interest from unwanted impurities can be accomplished.
  • concentration of a particular ingredient, such as a particular salt (e.g. NaCl) in the elution buffer is varied during the elution procedure (gradient). The gradient may be continuous or stepwise (interrupted by hold periods).
  • controlled room temperature refers to a temperature of at least about 10° C., at least about 15° C., at least about 20° C. or at least about 25° C. Typically, controlled room temperature is between about 20° C. and about 25° C.
  • the present invention provides processes for the isolation (e.g., large-scale isolation) of polypeptide conjugates from a mixture.
  • the polypeptide conjugates isolated by the methods of the invention include at least one modifying group.
  • Exemplary modifying groups include polymers, such as poly(alkylene oxide) moieties (e.g., poly(ethylene glycol) or poly(propylene glycol)). Exemplary modifying groups are described herein, below.
  • the polypeptide conjugate is isolated from a reaction mixture.
  • the reaction mixture is the product of a chemical reaction, such as a chemical PEGylation reaction.
  • the reaction mixture may contain chemicals, such as unreacted polymeric reagents and/or hydrolysis products thereof.
  • the reaction mixture is the product of an enzymatically catalyzed reaction, such as an enzymatically catalyzed glycoPEGylation reaction.
  • the reaction mixture may include enzymes, and may further include reagents, such as unreacted enzyme substrates (e.g., nucleotide sugars and the like).
  • the methods of the invention are suitable for the isolation of a polypeptide conjugate from the above listed reaction mixture components.
  • the methods of the invention are useful to isolate a desired polypeptide conjugate from a mixture that includes other polypeptide conjugates, which are sought to be separated from the desired polypeptide conjugate.
  • Such “unwanted” polypeptide conjugates or side-products may be generated during the same reaction that leads to the formation of the desired polypeptide conjugate.
  • a recombinant polypeptide is subjected to a chemical PEGylation reaction.
  • the reaction product includes different polypeptide conjugates, in which each type of polypeptide conjugate includes a different number of PEG moieties, e.g., the majority of the polypeptide conjugates includes three PEG moieties, while a small percentage of the polypeptide conjugates in the reaction mixture is covalently linked to only one or two PEG moieties.
  • a recombinantly produced polypeptide is subjected to an enzymatically catalyzed glycoPEGylation reaction.
  • the reaction mixture includes different polypeptide conjugates, in which each type of polypeptide conjugate has a different structure with respect to the number of PEG moieties covalently linked to the polypeptide and/or the structure of the glycan residues, to which each PEG moiety is attached to the polypeptide.
  • the mixture may also contain unreacted polypeptide.
  • HIC hydrophobic interaction chromatography
  • HIC is efficient in separating an erythropoietin (EPO) conjugate that includes three poly(ethylene glycol) (PEG) moieties from other EPO conjugates that include 0, 1, 2, 4, 5, 6 or 7 PEG moieties.
  • EPO erythropoietin
  • PEG poly(ethylene glycol)
  • HIC can be used to separate polypeptide conjugates that include the same number of poly(alkylene oxide) moieties, but wherein the polypeptide conjugates have a different glycosylation pattern.
  • the methods of the invention may further employ additional chromatographic steps.
  • the method includes anion exchange or mixed-mode chromatography in addition to HIC.
  • the method includes cation exchange chromatography in addition to HIC.
  • the method includes both, anion exchange or mixed-mode chromatography and cation exchange chromatography in addition to HIC.
  • the method includes hydroxyapatite or fluoroapatite chromatography in addition to HIC.
  • the chromatographic steps employed in the methods of the invention can be performed in any desired order.
  • anion exchange or mixed-mode chromatography is performed prior to hydrophobic interaction chromatography.
  • anion exchange or mixed-mode chromatography is performed after hydrophobic interaction chromatography.
  • cation exchange chromatography is performed prior to hydrophobic interaction chromatography.
  • cation exchange chromatography is performed after hydrophobic interaction chromatography.
  • hydroxyapatite or fluoroapatite chromatography is performed prior to HIC.
  • hydroxyapatite or fluoroapatite chromatography is performed after HIC.
  • the invention provides a method of making a composition that includes a first polypeptide conjugate, wherein the first polypeptide conjugate includes a first number of poly(alkylene oxide) moieties covalently linked to a first polypeptide.
  • the method includes: (a) contacting a mixture containing the first polypeptide conjugate with a hydrophobic interaction chromatography (HIC) medium; and (b) eluting the first polypeptide conjugate from the HIC medium.
  • the method may further include: (c) eluting the first polypeptide conjugate from an anion exchange or mixed-mode chromatography medium.
  • step (c) is performed prior to step (a).
  • step (c) is performed after step (b).
  • the method may further include: (d) eluting the first polypeptide conjugate from a cation exchange chromatography medium.
  • step (d) is performed prior to step (a).
  • step (d) is performed after step (b).
  • the mixture includes additional polypeptide conjugates, from which the first polypeptide conjugate is isolated.
  • the mixture includes a second polypeptide conjugate, wherein the second polypeptide conjugate has a second number of poly(alkylene oxide) moieties covalently linked to a second polypeptide.
  • the first polypeptide and the second polypeptide have the same amino acid sequence.
  • the first polypeptide and the second polypeptide have a different amino acid sequence.
  • the first number and the second number are different, which means that the first polypeptide conjugate and the second polypeptide conjugate are distinguished by the number of poly(alkylene oxide) moieties that are linked to each polypeptide.
  • the first polypeptide conjugate includes 3 poly(alkylene oxide) moieties, while the second polypeptide conjugate includes either 0, 1, 2 or 4 poly(alkylene oxide) moieties.
  • the first polypeptide and the second polypeptide have the same amino acid sequence and the first polypeptide conjugate and the second polypeptide conjugate are distinguished by a different number of poly(alkylene oxide) moieties (first number and second number are different).
  • the method of the invention is useful to provide a composition including a first polypeptide conjugate, wherein the concentration of the second polypeptide conjugate in this composition is less than about 30%, less than about 25%, less than about 20%, less than about 15% and preferably less than about 10%, less than about 9%, less than about 8%, less than about 7%, less than about 6%, less than about 5%, less than about 4%, less than about 3%, less than about 2% or less than about 1%.
  • the mixture includes more than one glycoform of the first polypeptide conjugate and the method provides a composition, in which the combined concentration of all glycoforms having a structure distinct from the first polypeptide conjugate is less than about 30%, less than about 25%, less than about 20%, less than about 15% and preferably less than about 10%, less than about 9%, less than about 8%, less than about 7%, less than about 6%, less than about 5%, less than about 4%, less than about 3%, less than about 2% or less than about 1%.
  • the first polypeptide is a glycopeptide and comprises a first glycosylation pattern that includes at least one glycan residue covalently linked to the first polypeptide.
  • Each glycan residue can be linked to at least one polymeric modifying group, such as a poly(alkylene oxide) moiety.
  • the first polypeptide includes a first number of poly(alkylene oxide) moieties, each of which is covalently linked to the first polypeptide via an N-linked or O-linked glycan.
  • the method of the invention is useful to separate two polypeptide glycoforms that may include the same number of modifying groups, but that have different glycosylation patterns.
  • the mixture from which the first polypeptide is isolated includes a third polypeptide conjugate that includes a third number of poly(alkylene oxide) moieties.
  • the third polypeptide conjugate and the first polypeptide conjugate include the same number of poly(alkylene oxide) moieties, but the third polypeptide has a glycosylation pattern that differs from the glycosylation pattern of the first polypeptide conjugate by at least one glycosyl moiety.
  • the third polypeptide conjugate includes a glycan residue that is not present in the first polypeptide conjugate.
  • the third polypeptide includes an O-linked glycan, while the first polypeptide includes only N-linked glycans (see, e.g., FIG. 3B , tri-PEGylated EPO structures).
  • the third polypeptide includes a truncated glycan residue, while the corresponding glycan residue of the first polypeptide conjugate is intact (i.e., includes a larger number of glycosyl moieties).
  • the invention provides a method of isolating a first polypeptide conjugate including a first number of poly(alkylene oxide) moieties covalently linked to a first polypeptide, from a second polypeptide conjugate that includes a second number of poly(alkylene oxide) moieties covalently linked to a second polypeptide.
  • the method includes: (a) contacting a mixture containing the first polypeptide conjugate and the second polypeptide conjugate with a hydrophobic interaction chromatography (HIC) medium; and (b) eluting the first polypeptide conjugate from said hydrophobic interaction chromatography medium.
  • the method may further include: (c) eluting the first polypeptide conjugate from an anion exchange chromatography medium.
  • step (c) is performed prior to step (a). In another embodiment, step (c) is performed after step (b).
  • the method may further include: (d) eluting the first polypeptide conjugate from a cation exchange chromatography medium. In one embodiment, step (d) is performed prior to step (a). In another embodiment, step (d) is performed after step (b).
  • the first number of poly(alkylene oxide) moieties that are linked to the first polypeptide is selected from 1 to about 40. In another example, the first number is selected from 1 to about 30. In yet another example, the first number is selected from 1 to about 20. In a further example, the first number is selected from 1, 2, 3, 4, 5, 6, 7, 8, 9 and about 10. In another example, the first polypeptide conjugate includes exactly three poly(alkylene oxide) moieties.
  • the second number of poly(alkylene oxide) moieties that are linked to the second polypeptide is selected from 0 to about 40. In another example, the second number is selected from 0 to about 30. In yet another example, the second number is selected from 0 to about 20. In a further example, the second number is selected from 0, 1, 2, 3, 4, 5, 6, 7, 8, 9 and about 10. In another example, the first number and the second number are different.
  • the first polypeptide conjugate includes 3 poly(alkylene oxide) moieties, while the second polypeptide conjugate includes either 0, 1, 2 or 4 poly(alkylene oxide) moieties.
  • the first polypeptide and the second polypeptide have the same amino acid sequence.
  • the first polypeptide is a therapeutic polypeptide. Exemplary therapeutic polypeptides are described herein, below.
  • the first polypeptide is EPO.
  • both the first polypeptide and the second polypeptide are EPO.
  • both the first polypeptide and the third polypeptide are EPO.
  • the first polypeptide conjugate is formed by an enzymatically catalyzed glycomodification reaction, during which a modified glycosyl moiety [e.g., a glycosyl moiety modified with at least one poly(alkylene oxide) moiety] is covalently linked to the first polypeptide.
  • a modified glycosyl moiety e.g., a glycosyl moiety modified with at least one poly(alkylene oxide) moiety
  • the method of the invention may further include: contacting the first polypeptide and a modified glycosyl donor species (e.g., a modified sugar nucleotide) having a glycosyl moiety covalently linked to a polymer (e.g., a poly(alkylene oxide) moiety), in the presence of an enzyme (e.g., a glycosyltransferase), for which the modified glycosyl donor species is a substrate, under conditions sufficient for the enzyme to catalyze the formation of a covalent bond between the glycosyl moiety that is linked to the polymer and the first polypeptide.
  • a modified glycosyl donor species e.g., a modified sugar nucleotide
  • an enzyme e.g., a glycosyltransferase
  • the modified glycosyl moiety is a sialic acid (SA) moiety.
  • the enzyme is a sialyltransferase.
  • the polymer is PEG (e.g., m-PEG). GlycoPEGylation methods are art-recognized; see for example, WO 03/031464 to DeFrees et al. or WO 04/99231, the disclosures of which are incorporated herein by reference in their entirety.
  • the method of the invention may further include: recombinantly expressing the first polypeptide in a host cell, such as an insect cell, a mammalian cell (e.g., a CHO cell) or a fungal cell (e.g., yeast cell).
  • a host cell such as an insect cell, a mammalian cell (e.g., a CHO cell) or a fungal cell (e.g., yeast cell).
  • the first polypeptide is expressed in an insect cell line (e.g., a Spodoptera frugiperda cell, e.g., Sf9).
  • the first polypeptide may be further modified (e.g., through glycan remodeling) to include a substantially uniform (e.g., insect-specific) glycosylation pattern.
  • the glycosylation pattern of the peptides can be elaborated, trimmed back or otherwise modified by methods utilizing enzymes.
  • the methods of remodeling peptides using enzymes that transfer a sugar donor to an acceptor are discussed in detail in WO 03/031464 to De Frees et al. (published Apr. 17, 2003); U.S. Patent Application 20040137557 (filed Nov. 5, 2002); U.S. Patent Application 20050143292 (filed Nov. 24, 2004) and WO 05/051327 (filed Nov. 24, 2004), each of which is incorporated herein by reference in its entirety.
  • the method of the invention may further include: contacting the first polypeptide and a glycosyl donor molecule (e.g., a nucleotide sugar) in the presence of an enzyme for which the glycosyl donor molecule is a substrate, under conditions sufficient for the enzyme to form a covalent bond between a glycosyl moiety of the glycosyl donor molecule and the first polypeptide.
  • a glycosyl donor molecule e.g., a nucleotide sugar
  • the polypeptide used as a substrate in this reaction may be glycosylated or non-glycosylated.
  • the enzyme may be a glycosyltransferase, such as a GlcNAc-transferase, a GalNAc-transferase, a Gal-transferase or a sialyltransferase.
  • a glycosyltransferase such as a GlcNAc-transferase, a GalNAc-transferase, a Gal-transferase or a sialyltransferase.
  • the method of the invention includes: contacting a glycosylated or non-glycosylated first polypeptide and a nucleotide-N-acetylglucosamine (GlcNAc) or a nucleotide-N-acetylgalactosamine (GalNAc) molecule in the presence of a N-acetylglucosamine transferase (e.g., GnT1 or GnT2) or a N-acetylgalactosamine transferase, respectively.
  • the reaction mixture may further include a nucleotide galactose (Gal) molecule, and a galactosyl transferase (e.g., GalT1).
  • the components of the reaction mixture are contacted (e.g., in a single reaction vessel or sequentially) under conditions sufficient for the N-acetylglucosamine transferase and the galactosyl transferase to form a glycosylated first polypeptide having at least one glycan residue with a terminal -GlcNAc-Gal moiety or a GalNAc-Gal moiety.
  • That glycan residue is preferably mono-antennary with respect to the newly added GlcNAc-Gal or -GalNAc-Gal moiety.
  • the GlcNAc-Gal moiety is added to a mannose residue, which is part of a tri-mannosyl motif.
  • the -GalNAc-Gal moiety is added to a serine or threonine residue of the first polypeptide.
  • the invention provides a method of making a composition that contains a first erythropoietin (EPO) conjugate, wherein the first EPO conjugate includes a first number of poly(alkylene oxide) moieties covalently linked to an EPO polypeptide.
  • the method includes: (a) contacting a mixture containing the first EPO conjugate with an anion exchange medium; (b) eluting the first EPO conjugate from the anion exchange medium, forming a first eluate including the first EPO conjugate; (c) contacting the first eluate with a hydrophobic interaction chromatography (HIC) medium; and (d) eluting the first EPO conjugate from the hydrophobic interaction chromatography medium.
  • HIC hydrophobic interaction chromatography
  • the method may further include: (e) eluting the first EPO conjugate from a cation exchange chromatography medium.
  • step (e) is performed after step (d).
  • step (e) is performed prior to step (c).
  • the method may further include one or more dilution or diafiltration steps.
  • diafiltration is used to concentrate and/or exchange the buffer in order to condition the sample for the next process step. For example, the eluate from the HIC step is concentrated and diafiltered into a new buffer system in order to prepare the sample for cation exchange chromatography.
  • the mixture includes additional EPO conjugates, from which the first EPO conjugate is isolated.
  • the mixture includes a second EPO conjugate having a second number of poly(alkylene oxide) moieties covalently linked to an EPO polypeptide.
  • the first number and the second number are different, which means that the first EPO conjugate and the second EPO conjugate are glycoforms distinguished by the number of poly(alkylene oxide) moieties that are linked to each EPO polypeptide.
  • the first EPO conjugate includes 3 poly(alkylene oxide) moieties, while the second EPO conjugate may include 0, 1, 2 or 4 poly(alkylene oxide) moieties.
  • the method is useful to provide a composition including a first EPO conjugate, wherein the concentration of the second EPO conjugate in this composition is less than about 30%, less than about 25%, less than about 20%, less than about 15% and preferably less than about 10%, less than about 9%, less than about 8%, less than about 7%, less than about 6%, less than about 5%, less than about 4%, less than about 3%, less than about 2% or less than about 1%.
  • the mixture includes more than one glycoform of the first EPO conjugate and the method provides a composition, in which the combined concentration of all glycoforms having a structure distinct from the structure of the first EPO conjugate is less than about 30%, less than about 25%, less than about 20%, less than about 15% and preferably less than about 10%, less than about 9%, less than about 8%, less than about 7%, less than about 6%, less than about 5%, less than about 4%, less than about 3%, less than about 2% or less than about 1%.
  • an exemplary EPO sequence useful in conjunction with any of the above embodiments is represented by SEQ ID NO:1, which may include at least one mutation (e.g., Arg 139 to Ala 139 , Arg 143 to Ala 143 and Lys 154 to Ala 154 ).
  • the EPO conjugates of the invention may include at least one N-linked glycan residue.
  • the N-linked glycan residue is covalently linked to an amino acid residue selected from Asn 24 , Asn 38 and Asn 83 of SEQ ID NO:1.
  • the EPO conjugate may further include an O-linked glycan residue.
  • the O-linked glycan residue is covalently linked to a serine (e.g., Ser 126) residue of SEQ ID NO:1. Any of the above described glycan residues can optionally be linked to a poly(alkylene oxide) moiety.
  • EPO is covalently linked to three poly(alkylene oxide) moieties (e.g., three PEG moieties).
  • EPO is covalently linked to three poly(alkylene oxide) moieties (e.g., PEG), wherein at least two of the three poly(alkylene oxide) moieties are covalently linked to the EPO polypeptide via N-linked glycans.
  • at least one N-linked glycan is mono-antennary.
  • all three N-linked glycans are mono-antennary (e.g., as shown in FIG. 3A ).
  • Exemplary tri-PEGylated EPO conjugates are shown in FIG. 3B .
  • the EPO conjugate is formed by an enzymatically catalyzed glycomodification reaction, wherein a modified glycosyl moiety (e.g., a glycosyl moiety modified with at least one poly(alkylene oxide) moiety) is attached to the EPO polypeptide.
  • a modified glycosyl moiety e.g., a glycosyl moiety modified with at least one poly(alkylene oxide) moiety
  • the method of the invention may further include: contacting an EPO polypeptide and a modified glycosyl donor species (e.g., a modified sugar nucleotide) having a glycosyl moiety covalently linked to a polymer (e.g., a poly(alkylene oxide) moiety), in the presence of an enzyme (e.g., a glycosyltransferase), for which the modified glycosyl donor species is a substrate, under conditions sufficient for the enzyme to catalyze the formation of a covalent bond between the glycosyl moiety that is linked to the polymer and the EPO polypeptide.
  • a modified glycosyl donor species e.g., a modified sugar nucleotide
  • an enzyme e.g., a glycosyltransferase
  • the modified glycosyl moiety is a sialic acid (SA) moiety.
  • the enzyme is a sialyltransferase.
  • the method may further include: recombinantly expressing the EPO polypeptide in a host cell, such as a bacterial (e.g., E. coli ), an insect cell, a mammalian cell (e.g., CHO) cell or a fungal cell.
  • the EPO polypeptide is expressed in an insect cell line (e.g., Sf9) and is optionally purified from insect cell culture, e.g., according to the methods outlined in WO 06/105426 to Kang et al.
  • the EPO peptide may be further modified through glycan remodeling to include a substantially uniform (e.g., insect-specific) glycosylation pattern.
  • the method of the invention may further include: contacting (e.g., in a single reaction vessel) a glycosylated EPO polypeptide with a nucleotide-N-acetylglucosamine (GlcNAc) molecule and a nucleotide galactose (Gal) molecule in the presence of a N-acetylglucosamine transferase (e.g., GnT1 or GnT2), and a galactosyl transferase (e.g., GalT1), under conditions sufficient for said N-acetylglucosamine transferase and said galactosyl transferase to form a glycosylated EPO polypeptide having at least one glycan residue with a terminal -GlcNAc-Gal
  • That glycan residue is preferably mono-antennary with respect to the newly added GlcNAc-Gal moiety.
  • the -GlcNAc-Gal moiety is added to a mannose residue, which is part of a tri-mannosyl motive.
  • each poly(alkylene oxide) moiety is a member independently selected from poly(ethylene glycol) (e.g., m-PEG) and poly(propylene glycol) (e.g., m-PPG). Exemplary poly(ethylene glycol) moieties are described herein, below.
  • each poly(alkylene oxide) moiety has an independently selected molecular weight between about 1 kDa and about 200 kDa. Additional molecular weight ranges for poly(alkylene oxide) moieties are given herein, below.
  • the first polypeptide conjugate includes at least one poly(alkylene oxide) moiety that is covalently linked to the first polypeptide via a glycosyl linking group.
  • the glycosyl linking group is covalently linked to an amino acid residue of the first polypeptide.
  • the glycosyl linking group is covalently linked to a glycosyl moiety of said first polypeptide.
  • the glycosyl linking group is an intact glycosyl linking group.
  • glycosyl linking groups are described herein and, for example, in WO 03/031464 to DeFrees et al., WO 04/99231, and PCT/U.S.07/74139 filed Jul. 23, 2007, the disclosures of which are incorporated herein by reference in their entirety.
  • Exemplary intact glycosyl linking groups include sialic acid moieties, GlcNH and GlcNAc moieties, as well as Gal, GalNH and GalNAc moieties.
  • HIC media that are useful in any of the above described embodiments, include butyl and phenyl resins, such as Phenyl 650S (e.g., ToyoPearl). Hydrophobic interaction chromatography and suitable HIC media are described herein below and, for example in Process Scale Bioseparations for the Biopharmaceutical Industry , Ed. Shukla A A, Etzel M R, Gadam S, CRC Press Taylor & Francis Group (2007), pages 197-206, the disclosure of which is incorporated herein by reference.
  • Phenyl 650S e.g., ToyoPearl
  • Hydrophobic interaction chromatography and suitable HIC media are described herein below and, for example in Process Scale Bioseparations for the Biopharmaceutical Industry , Ed. Shukla A A, Etzel M R, Gadam S, CRC Press Taylor & Francis Group (2007), pages 197-206, the disclosure of which is incorporated herein by reference.
  • polypeptide conjugates isolated by the methods of the invention include a polypeptide and at least one modifying group covalently linked to the polypeptide, e.g., via a glycosyl linking group.
  • exemplary polypeptide conjugates are discussed herein below and, for example WO 03/031464 to DeFrees et al., WO 04/99231 to DeFrees et al., and WO 04/33651 to DeFrees et al., the disclosures of which are incorporated herein by reference in their entirety.
  • polypeptide that is part of polypeptide conjugates of the invention can be any glycosylated or non-glycosylated polypeptide.
  • the polypeptide is a recombinant polypeptide.
  • the polypeptide is expressed in a host cell selected from bacterial cells (e.g., E. coli ), insect cells (e.g., Spodoptera frugiperda cells), fungal cells (e.g., yeast cells), mammalian cells (e.g., CHO cells) and bacterial cells (e.g., E. coli cells). Methods for the expression of polypeptides in insect cell lines are discussed herein below.
  • the polypeptide is chemically synthesized and optionally includes non-natural amino acids.
  • the polypeptide can have any number of amino acids.
  • the peptide or glycopeptide has a molecular weight of about 5 kDa to about 500 kDa.
  • the peptide or glycopeptide has a molecular weight of about 10 kDa to about 100 kDa.
  • the polypeptide has a molecular weight of about 10 kDa to about 30 kDa.
  • the polypeptide has a molecular weight of about 20 kDa to about 25 kDa.
  • Exemplary polypeptides include wild-type polypeptides and fragments thereof as well as polypeptides, which are modified from their naturally occurring counterpart (e.g., by mutation or truncation).
  • a polypeptide may also be a fusion protein.
  • Exemplary fusion proteins include those, in which the polypeptide is fused to a fluorescent protein (e.g., GFP), a therapeutic polypeptide, an antibody, a receptor ligand, a proteinaceous toxin, MBP, a Histag and the like.
  • the polypeptide is a therapeutic polypeptide (i.e., authorized drug), such as those currently used as pharmaceutical agents.
  • a therapeutic polypeptide i.e., authorized drug
  • Exemplary polypeptides include growth factors, such as fibroblast growth factors (e.g., FGF-1, FGF-2, FGF-3, FGF-4, FGF-5, FGF-6, FGF-7, FGF-8, FGF-9, FGF-10, FGF-11, FGF-12, FGF-13, FGF-14, FGF-15, FGF-16, FGF-17, FGF-18, FGF-19, FGF-20, FGF-21, FGF-22 and FGF-23), blood coagulation factors (e.g., Factor V, Factor VII, Factor VIII, B-domain deleted Factor VIII, partial B-domain deleted Factor VIII, vWF-Factor VIII fusion (e.g., with full-length or B-domain deleted Factor VIII), Factor IX, Factor X and Factor XIII), hormones, such as human growth hormone (hGH) and follicle stimulating hormone (FSH), as well as cytokines, such as interleukins (e.g., IL-1, IL-2,
  • polypeptides include enzymes, such as glucocerebrosidase, alpha-galactosidase (e.g., FabrazymeTM), acid-alpha-glucosidase (acid maltase), alpha-L-iduronidase (e.g., AldurazymeTM), thyroid peroxidase (TPO), beta-glucosidase (see e.g., enzymes described in U.S. patent application Ser. No. 10/411,044), and alpha-galactosidase A (see e.g., enzymes described in U.S. Pat. No. 7,125,843).
  • enzymes such as glucocerebrosidase, alpha-galactosidase (e.g., FabrazymeTM), acid-alpha-glucosidase (acid maltase), alpha-L-iduronidase (e.g., AldurazymeTM), thyroid
  • exemplary parent polypeptides include bone morphogenetic proteins (e.g., BMP-1, BMP-2, BMP-3, BMP-4, BMP-5, BMP-6, BMP-7, BMP-8, BMP-9, BMP-10, BMP-11, BMP-12, BMP-13, BMP-14, BMP-15), neurotrophins (e.g., NT-3, NT-4, NT-5), erythropoietins (EPO), growth differentiation factors (e.g., GDF-5), glial cell line-derived neurotrophic factor (GDNF), brain derived neurotrophic factor (BDNF), nerve growth factor (NGF), von Willebrand factor (vWF), vWF protease, granulocyte colony stimulating factor (G-CSF), granulocyte-macrophage colony stimulating factor (GM-CSF), ⁇ 1 -antitrypsin (ATT, or ⁇ -1 protease inhibitor), tissue-type plasminogen activator (TPA), hirudi
  • polypeptide is EPO comprising the amino acid sequence of (SEQ ID NO:1), which is shown below:
  • polypeptides that are antibodies.
  • the term antibody is meant to include antibody fragments (e.g., Fc domains), single chain antibodies, Lama antibodies, nano-bodies and the like.
  • antibody-fusion proteins such as Ig chimeras.
  • Preferred antibodies include humanized, monoclonal antibodies or fragments thereof. All known isotypes of such antibodies are within the scope of the invention.
  • Exemplary antibodies include those to growth factors, such as endothelial growth factor (EGF), vascular endothelial growth factors (e.g., monoclonal antibody to VEGF-A, such as ranibizumab (LucentisTM)) and fibroblast growth factors, such as FGF-7, FGF-21 and FGF-23) and antibodies to their respective receptors.
  • growth factors such as endothelial growth factor (EGF)
  • vascular endothelial growth factors e.g., monoclonal antibody to VEGF-A, such as ranibizumab (LucentisTM)
  • fibroblast growth factors such as FGF-7, FGF-21 and FGF-283
  • Other exemplary antibodies include anti-TNF-alpha monoclonal antibodies (see e.g., U.S. patent application Ser. No.
  • TNF receptor-IgG Fc region fusion protein e.g., EnbrelTM
  • anti-HER2 monoclonal antibodies e.g., HerceptinTM
  • monoclonal antibodies to protein F of respiratory syncytial virus e.g., SynagisTM
  • monoclonal antibodies to TNF- ⁇ e.g., RemicadeTM
  • monoclonal antibodies to glycoproteins such as IIb/IIIa (e.g., ReoproTM)
  • monoclonal antibodies to CD20 e.g., RituxanTM
  • CD4 and alpha-CD3 monoclonal antibodies to PSGL-1 and CEA.
  • Any modified (e.g., mutated) version of any of the above listed polypeptides is also within the scope of the invention.
  • the polypeptide is expressed in insect cells.
  • Insect cells suitable for use in the present invention are from any order of the class Insecta which can be hosts to recombinant viruses (e.g. baculovirus) or wild-type viruses, and which can grow and produce recombinant peptide products upon infection with the virus in a medium composition of the invention.
  • the cells are from the Diptera or Lepidoptera orders.
  • Preferred are insect cell lines that can be used to produce polypeptides having a substantially uniform, insect-specific glycosylation pattern.
  • the polypeptide is expressed by a stably transfected cell.
  • NDV nuclear polyhedrosis virus
  • Insect cell lines derived from the following insects are exemplary: Carpocapsa pomonella (preferably cell line CP-128); Trichoplusia ni (preferably cell line TN-368); Autographa californica; Spodoptera frugiperda (preferably cell line Sf9); Lymantria dispar; Mamestra brassicae; Aedes albopictus; Orgyia pseudotsugata; Neodiprion sertifer; Aedes aegypti; Antheraea eucalypti; Gnorimoschema opercullela; Galleria mellonella; Spodoptera littoralis; Drosophila melanogaster, Heliothis zea; Spodoptera exigua; Rachiplusia ou; Plodia interpunctella; Amsacta moorei; Agrotis c - nitrum, Adoxophyes orana, Agrotis segetum, Bombyx mori, Hyponomeu
  • the insect cells are from Spodoptera frugiperda , and in another exemplary embodiment, the cell line is Sf9 (ATCC CRL 1711).
  • Sf9, Sf21, and High-Five insect cells are commonly used for baculovirus expression.
  • Sf9 and Sf21 are ovarian cell lines from Spodoptera frugiperda .
  • High-Five cells are egg cells from Trichoplusia ni .
  • Sf9, Sf21 and High-Five cell lines may be grown at room temperature (e.g. 25 to 27° C.), and do not require CO 2 incubators. Their doubling time is between about 18 and 24 hours.
  • the insect cell lines cultured to produce the peptides and glycopeptides of the invention are preferably those suitable for the reproduction of numerous insect-pathogenic viruses such as picornaviruses, parvoviruses, entomopox viruses, baculoviruses and rhabdoviruses.
  • insect-pathogenic viruses such as picornaviruses, parvoviruses, entomopox viruses, baculoviruses and rhabdoviruses.
  • NPV nucleopolyhedrosis viruses
  • GV granulosis viruses
  • Baculoviruses are characterized by rod-shaped virus particles which are generally occluded in occlusion bodies (also called polyhedra).
  • the family Baculoviridae can be divided in two subfamilies: the Eubaculovirinae comprising two genera of occluded viruses; nuclear polyhedrosis virus (NPV) and granulosis virus (GV), and the subfamily Nudobaculovirinae comprising the nonoccluded viruses.
  • the polypeptide expressed in any suitable expression system is isolated from cell culture before the polypeptide is modified with a modifying group.
  • the polypeptide is first removed from the cell culture medium, cellular debris and other particles and is then further purified to remove contaminants, such as viral particles and unwanted proteins, using a variety of filtration and chromatographic purification devices.
  • Polypeptide purification techniques are known. See, e.g., Protein Purification Methods, A Practical Approach , Ed. Harris E L V, Angal S, IRL Press Oxford, England (1989), Protein Purification , Ed. Janson J C, Ryden L, VCH-Verlag, Weinheim, Germany (1989), Process Scale Bioseparations for the Biopharmaceutical Industry , Ed.
  • the modifying group of the invention can be any chemical moiety. Exemplary modifying groups are discussed below.
  • the modifying group is a linear or branched polymeric modifying group (polymer).
  • a polymeric modifying group includes at least one polymeric moiety, wherein each polymeric moiety is independently selected.
  • the polymeric modifying group is water-soluble.
  • a water-soluble polymeric modifying group includes at least one polar group. Exemplary polar groups, include polyether groups, hydroxyl groups and carboxylic acid groups.
  • water-soluble polymers are known to those of skill in the art and are useful in practicing the present invention.
  • the term water-soluble polymer encompasses species such as saccharides (e.g., dextran, amylose, hyalouronic acid, poly(sialic acid), heparans, heparins, etc.); poly(amino acids), e.g., poly(aspartic acid) and poly(glutamic acid); nucleic acids; synthetic polymers (e.g., poly(acrylic acid), poly(alkylene oxides), peptides, proteins, and the like.
  • the polymer is a poly(alkylene oxide), such as a poly(ethylene glycol) or a polypropylene glycol.
  • the water-soluble polymer is polyethylene glycol (PEG) or a PEG analog, e.g., methoxy-poly(ethylene glycol) (m-PEG).
  • the water-soluble polymer is polypropylene glycol (PPG), e.g., methoxy-polypropylene glycol (m-PPG).
  • PEG is frequently used to modify the properties of polypeptides, such as therapeutic proteins. For example, the in vivo half-life of therapeutic glycopeptides can be enhanced with PEG moieties.
  • PEGylation Chemical modification of polypeptides with PEG increases their molecular size and typically decreases surface- and functional group-accessibility, each of which are dependent on the number and size of the PEG moieties attached to the polypeptide. Frequently, this modification results in an improvement of plasma half-live and in proteolytic-stability, as well as a decrease in immunogenicity and hepatic uptake (Chaffee et al. J. Clin. Invest. 89: 1643-1651 (1992); Pyatak et al. Res. Commun. Chem. Pathol Pharmacol. 29: 113-127 (1980)).
  • the in vivo half-life of a polypeptide derivatized with a PEG moiety by a method of the invention is increased relative to the in vivo half-life of the non-derivatized parent polypeptide.
  • the poly(ethylene glycol) or poly(propylene glycol) is not restricted to any particular form or molecular weight range.
  • the size of these modifying groups may, for example, depend on the nature and size of the polypeptide to which they are attached and the properties desired for the modified polypeptide.
  • the molecular weight is preferably between about 0.5 kDa and about 500 kDa.
  • Branched polymers may be larger than 500 kDa.
  • branched poly(ethylene glycol) or poly(propylene glycol) have a molecular weight from about 0.5 kDa to about 1000 kDa.
  • the PEG or PPG molecule of use in the invention (branched or unbranched) has a molecular weight selected from about 0.5 kDa, 1 kDa, about 2 kDa, about 5 kDa, about 10 kDa, about 15 kDa, about 20 kDa, about 25 kDa, about 30 kDa, about 35 kDa, about 40 kDa, about 45 kDa, about 50 kDa, about 55 kDa, about 60 kDa, about 65 kDa, about 70 kDa, about 75 kDa, about 80 kDa, about 85 kDa, about 90 kDa, about 95 kDa, about 100 kDa, about 105 kDa, about 110 kDa, about 115 kDa, about 120 kDa, about 125 kDa, about 130 kDa, about 135 kDa, about 140 kD
  • the polypeptide is EPO.
  • the EPO peptide has at least two, and preferably three poly(ethylene glycol) moieties covalently linked thereto.
  • each PEG molecule linked to the EPO peptide has a molecular weight from about 2 kDa to about 80 kDa, preferably from about 5 kDa to about 60 kDa and more preferably from about 10 kDa to about 40 kDa.
  • Exemplary water-soluble polymers are those in which a substantial proportion of the polymer molecules in a sample of the polymer are of approximately the same molecular weight; such polymers are “homodisperse.”
  • Exemplary poly(ethylene glycol) molecules of use in the invention include, but are not limited to, those having the formula:
  • R 8 is H, OH, NH 2 , substituted or unsubstituted alkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted heteroalkyl, e.g., acetal, OHC—, H 2 N—(CH 2 ) q —, HS—(CH 2 ) q , or —(CH 2 ) q C(Y)Z 1 .
  • the index “e” represents an integer from 1 to 2500.
  • the indices b, d, and q independently represent integers from 0 to 20.
  • the symbols Z and Z 1 independently represent OH, NH 2 , leaving groups, e.g., imidazole, p-nitrophenyl, HOBT, tetrazole, halide, S—R 9 , the alcohol portion of activated esters; —(CH 2 ) p C(Y 1 )V, or —(CH 2 ) p U(CH 2 ) s C(Y 1 ) v .
  • the symbol Y represents H(2), ⁇ O, ⁇ S, ⁇ N—R 10 .
  • the symbols X, Y, Y 1 , A 1 , and U independently represent the moieties O, S, N—R′′.
  • the symbol V represents OH, NH 2 , halogen, S—R 12 , the alcohol component of activated esters, the amine component of activated amides, sugar-nucleotides, and proteins.
  • the indices p, q, s and v are members independently selected from the integers from 0 to 20.
  • the symbols R 9 , R 10 , R 11 and R 12 independently represent H, substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heterocycloalkyl and substituted or unsubstituted heteroaryl.
  • poly(ethylene glycol) useful in forming the conjugate of the invention is either linear or branched.
  • Branched poly(ethylene glycol) molecules suitable for use in the invention include, but are not limited to those described by the following formula:
  • R 8 and R 8′ are members independently selected from the groups defined for R 8 , above.
  • a 1 and A 2 are members independently selected from the groups defined for A 1 , above.
  • the indices e, f, o, and q are as described above.
  • Z and Y are as described above.
  • X 1 and X 1′ are members independently selected from S, SC(O)NH, HNC(O)S, SC(O)O, O, NH, NHC(O), (O)CNH and NHC(O)O, OC(O)NH.
  • the branched PEG is based upon a cysteine, serine or di-lysine core.
  • the poly(ethylene glycol) molecule is selected from the following structures:
  • the poly(ethylene glycol) is a branched PEG having more than one PEG moiety attached.
  • branched PEGs are described in U.S. Pat. No. 5,932,462; U.S. Pat. No. 5,342,940; U.S. Pat. No. 5,643,575; U.S. Pat. No. 5,919,455; U.S. Pat. No. 6,113,906; U.S. Pat. No. 5,183,660; WO 02/09766; Kodera Y., Bioconjugate Chemistry 5: 283-288 (1994); and Yamasaki et al., Agric. Biol. Chem., 52: 2125-2127, 1998.
  • the molecular weight of each poly(ethylene glycol) of the branched PEG is less than or equal to 40,000 daltons.
  • Representative polymeric modifying moieties include structures that are based on side chain-containing amino acids, e.g., serine, cysteine, lysine, and small peptides, e.g., lys-lys.
  • Exemplary structures include:
  • the free amine in the di-lysine structures can also be pegylated through an amide or urethane bond with a PEG moiety.
  • the polymeric modifying moiety is a branched PEG moiety that is based upon a tri-lysine peptide.
  • the tri-lysine can be mono-, di-, tri-, or tetra-PEG-ylated.
  • Exemplary species according to this embodiment have the formulae:
  • indices e, f and f′ are independently selected integers from 1 to 2500; and the indices q, q′ and q′′ are independently selected integers from 1 to 20.
  • the branched polymers of use in the invention include variations on the themes set forth above.
  • the di-lysine-PEG conjugate shown above can include three polymeric subunits, the third bonded to the ⁇ -amine shown as unmodified in the structure above.
  • the use of a tri-lysine functionalized with three or four polymeric subunits labeled with the polymeric modifying moiety in a desired manner is within the scope of the invention.
  • An exemplary branched modifying group including one or more polymeric moieties includes the formula:
  • the branched polymer species according to this formula are essentially pure water-soluble polymers.
  • C is carbon.
  • X 5 is a non-reactive group.
  • X 5 is selected from H, OH and C 1 -C 6 alkyl (e.g., CH 3 , —CH 2 CH 3 ) optionally substituted with OH.
  • R 16 and R 17 are independently selected from non-reactive groups (e.g., H, unsubstituted alkyl, unsubstituted heteroalkyl) and polymeric arms (e.g., PEG).
  • X 2 and X 4 are linkage fragments that are preferably essentially non-reactive under physiological conditions. X 2 and X 4 are independently selected.
  • An exemplary linker includes neither aromatic nor ester moieties.
  • these linkages can include one or more moiety that is designed to degrade under physiologically relevant conditions, e.g., esters, disulfides, etc.
  • X 2 and X 4 join the polymeric arms R 16 and R 17 to C.
  • Exemplary linkage fragments including X 2 and X 4 are independently selected and include S, SC(O)NH, HNC(O)S, SC(O)O, O, NH, NHC(O), (O)CNH and NHC(O)O, and OC(O)NH, CH 2 S, CH 2 O, CH 2 CH 2 O, CH 2 CH 2 S, (CH 2 ) o O, (CH 2 ) o S or (CH 2 ) o Y′-PEG wherein, Y′ is S, NH, NHC(O), C(O)NH, NHC(O)O, OC(O)NH, or O and o is an integer from 1 to 50.
  • the linkage fragments X 2 and X 4 are different linkage fragments.
  • the modifying group is derived from a natural or unnatural amino acid, amino acid analog or amino acid mimetic, or a small peptide formed from one or more such species.
  • certain branched polymers found in the polypeptide conjugates of the invention have the formula, wherein La is a linker moiety that links the modifying group to the remainder of the polypeptide conjugate.
  • L a is a linking moiety having the structure:
  • X a and X b are independently selected linkage fragments and L 1 is selected from a bond, substituted or unsubstituted alkyl or substituted or unsubstituted heteroalkyl.
  • Exemplary species for X a and X b include S, SC(O)NH, HNC(O)S, SC(O)O, O, NH, NHC(O), C(O)NH and NHC(O)O, and OC(O)NH.
  • X 4 is a peptide bond to R 17 , which is an amino acid, di-peptide (e.g., Lys-Lys) or tri-peptide (e.g., Lys-Lys-Lys) in which the alpha-amine moiety(ies) and/or side chain heteroatom(s) are modified with a polymeric modifying moiety.
  • R 17 is an amino acid, di-peptide (e.g., Lys-Lys) or tri-peptide (e.g., Lys-Lys-Lys) in which the alpha-amine moiety(ies) and/or side chain heteroatom(s) are modified with a polymeric modifying moiety.
  • polypeptide conjugate includes a moiety selected from the group:
  • the indices e and f are independently selected from the integers from 1 to 2500. In further exemplary embodiments, e and f are selected to provide a PEG moiety that is about 1 kDa, 2 kDa, 5 kDa, 10 kDa, 15 kDa, 20 kDa, 25 kDa, 30 kDa, 35 kDa, 40 kDa, 45 kDa, 50 kDa, 55 kDa, 60 kDa, 65 kDa, 70 kDa, 75 kDa and 80 kDa.
  • the symbol Q represents substituted or unsubstituted alkyl (e.g., C 1 -C 6 alkyl, e.g., methyl), substituted or unsubstituted heteroalkyl or H.
  • branched polymers have structures based on di-lysine (Lys-Lys) peptides, e.g.:
  • the indices e, f, f′ and f′′ represent integers independently selected from 1 to 2500.
  • the indices q, q′ and q′′ represent integers independently selected from 1 to 20.
  • the conjugates of the invention include a formula which is a member selected from:
  • Q is a member selected from H and substituted or unsubstituted C 1 -C 6 alkyl.
  • the indices e and f are integers independently selected from 1 to 2500, and the index q is an integer selected from 0 to 20.
  • the conjugates of the invention include a formula which is a member selected from:
  • Q is a member selected from H and substituted or unsubstituted C 1 -C 6 alkyl, preferably Me.
  • the indices e, f and f' are integers independently selected from 1 to 2500, and q and q' are integers independently selected from 1 to 20.
  • the conjugate of the invention includes a structure according to the following formula:
  • indices m and n are integers independently selected from 0 to 5000.
  • the indices j and k are integers independently selected from 0 to 20.
  • a 1 , A 2 , A 3 , A 4 , A 5 , A 6 , A 7 , A 8 , A 9 , A 10 and A 11 are members independently selected from H, substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted aryl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted heteroaryl, —NA 12 A 13 , —OA 12 and —SiA 12 A 13 .
  • a 12 and A 13 are members independently selected from substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, and substituted or unsubstituted heteroaryl.
  • the branched polymer has a structure according to the following formula:
  • a 1 and A 2 are members independently selected from OCH 3 and OH.
  • the linker L a is a member selected from aminoglycine derivatives.
  • Exemplary polymeric modifying groups according to this embodiment have a structure according to the following formulae:
  • a 1 and A 2 are members independently selected from OCH 3 and OH.
  • Exemplary polymeric modifying groups according to this example include:
  • the stereocenter can be either racemic or defined. In one embodiment, in which such stereocenter is defined, it has (S) configuration. In another embodiment, the stereocenter has (R) configuration.
  • one or more of the m-PEG arms of the branched polymer can be replaced by a PEG moiety with a different terminus, e.g., OH, COOH, NH 2 , C 2 -C 10 -alkyl, etc.
  • the structures above are readily modified by inserting alkyl linkers (or removing carbon atoms) between the ⁇ -carbon atom and the functional group of the side chain.
  • “homo” derivatives and higher homologues, as well as lower homologues are within the scope of cores for branched PEGs of use in the present invention.
  • linear and branched PEG conjugates set forth herein may be prepared using art-recognized methods.
  • Several reviews and monographs on the functionalization and conjugation of PEG are available. See, for example, Harris, Cellol. Chem. Phys . C25: 325-373 (1985); Scouten, Methods in Enzymology 135: 30-65 (1987); Wong et al., Enzyme Microb. Technol. 14: 866-874 (1992); Delgado et al., Critical Reviews in Therapeutic Drug Carrier Systems 9: 249-304 (1992); Zalipsky, Bioconjugate Chem. 6: 150-165 (1995); and Bhadra, et al., Pharmazie, 57:5-29 (2002).
  • the modifying group is covalently linked to the polypeptide via a glycosyl linking group.
  • the saccharide component of the modified sugar when interposed between the polypeptide and a modifying group, becomes a “glycosyl linking group.”
  • the glycosyl linking group is formed from a mono- or oligosaccharide that, after modification with a modifying group, is a substrate for an appropriate enzyme, such as a glycosyltransferase.
  • the glycosyl linking group is formed from a glycosyl-mimetic moiety.
  • the polypeptide conjugates of the invention can include glycosyl linking groups that are mono- or multi-valent (e.g., antennary structures).
  • conjugates of the invention include both species in which a modifying group is attached to a polypeptide via a monovalent glycosyl linking group. Also included within the invention are conjugates in which more than one modifying group is attached to a polypeptide via a multivalent linking group. Exemplary linking groups are disclosed in PCT/U.S.07/74139 filed Jul. 23, 2007, the disclosure of which is incorporated by reference herein in its entirety.
  • the invention provides a method for the isolation of a glycopeptide that is conjugated to a polymeric modifying moiety through an intact glycosyl linking group having a formula that is selected from:
  • R 2 is H, CH 2 OR 7 , COOR 7 or OR 7 , in which R 7 represents H, substituted or unsubstituted alkyl or substituted or unsubstituted heteroalkyl.
  • R 7 represents H, substituted or unsubstituted alkyl or substituted or unsubstituted heteroalkyl.
  • COOR 7 is a carboxylic acid or carboxylate
  • both forms are represented by the designation of the single structure COO ⁇ or COOH.
  • the symbols R 3 , R 4 , R 5 , R 6 and R 6′ independently represent H, substituted or unsubstituted alkyl, OR 8 , NHC(O)R 9 .
  • the index d is 0 or 1.
  • R 8 and R 9 are independently selected from H, substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, sialic acid or polysialic acid.
  • At least one of R 3 , R 4 , R 5 , R 6 or R 6′ includes the polymeric modifying moiety e.g., PEG, linked through a bond or a linking group.
  • R 6 and R 6′ together with the carbon to which they are attached are components of the pyruvyl side chain of sialic acid. In a further exemplary embodiment, this side chain is functionalized with the polymeric modifying moiety.
  • R 6 and R 6′ together with the carbon to which they are attached are components of the side chain of sialic acid and the polymeric modifying moiety is a component of R 5 .
  • the polymeric modifying moiety is bound to the sugar core, generally through a heteroatom, e.g, nitrogen, on the core through a linker, L, as shown below:
  • R 1 is the polymeric moiety and L is selected from a bond and a linking group.
  • the index w represents an integer selected from 1-6, preferably 1-3 and more preferably 1-2.
  • Exemplary linking groups include substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl moieties and sialic acid.
  • An exemplary component of the linker is an acyl moiety.
  • An exemplary compound according to the invention has a structure according to Formulae I or II, in which at least one of R 2 , R 3 , R 4 , R 5 , R 6 or R 6′ has the formula:
  • At least one of R 2 , R 3 , R 4 , R 5 , R 6 or R 6′ has the formula:
  • R 1 is a linear polymeric modifying moiety
  • the polymeric modifying moiety-linker construct is a branched structure that includes two or more polymeric chains attached to central moiety.
  • the construct has the formula:
  • R 1 and L are as discussed above and w' is an integer from 2 to 6, preferably from 2 to 4 and more preferably from 2 to 3.
  • L When L is a bond it is formed between a reactive functional group on a precursor of R 1 and a reactive functional group of complementary reactivity on the saccharyl core.
  • a precursor of L can be in place on the glycosyl moiety prior to reaction with the R 1 precursor.
  • the precursors of R 1 and L can be incorporated into a preformed cassette that is subsequently attached to the glycosyl moiety.
  • the selection and preparation of precursors with appropriate reactive functional groups is within the ability of those skilled in the art.
  • coupling the precursors proceeds by chemistry that is well understood in the art.
  • L is a linking group that is formed from an amino acid, or small peptide (e.g., 1-4 amino acid residues) providing a modified sugar in which the polymeric modifying moiety is attached through a substituted alkyl linker.
  • exemplary linkers include glycine, lysine, serine and cysteine.
  • the PEG moiety can be attached to the amine moiety of the linker through an amide or urethane bond.
  • the PEG is linked to the sulfur or oxygen atoms of cysteine and serine through thioether or ether bonds, respectively.
  • R 5 includes the polymeric modifying moiety.
  • R 5 includes both the polymeric modifying moiety and a linker, L, joining the modifying moiety to the remainder of the molecule.
  • L can be a linear or branched structure.
  • the polymeric modifying can be branched or linear.
  • the present invention provides methods for the isolation of an erythropoietin peptide conjugate comprising the moiety:
  • D is a member selected from —OH and R 1 -L-HN—; G is a member selected from H and R 1 -L- and —C(O)(C 1 -C 6 )alkyl; R 1 is a moiety comprising a straight-chain or branched poly(ethylene glycol) residue; and L is a linker, e.g., a bond (“zero order”), substituted or unsubstituted alkyl and substituted or unsubstituted heteroalkyl.
  • D is a member selected from —OH and R 1 -L-HN—
  • G is a member selected from H and R 1 -L- and —C(O)(C 1 -C 6 )alkyl
  • R 1 is a moiety comprising a straight-chain or branched poly(ethylene glycol) residue
  • L is a linker, e.g., a bond (“zero order”), substituted or unsubstituted alkyl and
  • the invention provides a conjugate formed between a modified sugar of the invention and a substrate EPO peptide.
  • the sugar moiety of the modified sugar becomes a glycosyl linking group interposed between the peptide substrate and the modifying group.
  • An exemplary glycosyl linking group is an intact glycosyl linking group, in which the glycosyl moiety or moieties forming the linking group are not degraded by chemical (e.g., sodium metaperiodate) or enzymatic (e.g., oxidase) processes.
  • Selected conjugates of the invention include a modifying group that is attached to the amine moiety of an amino-saccharide, e.g., mannosamine, glucosamine, galactosamine, sialic acid etc.
  • exemplary modifying group-intact glycosyl linking group cassettes according to this motif are based on a sialic acid structure, such as those having the formulae:
  • R 1 and L are as described above. Further detail about the structure of exemplary R 1 groups is provided below.
  • the conjugate is formed between a substrate EPO and a saccharyl moiety in which the modifying group is attached through a linker at the 6-carbon position of the saccharyl moiety.
  • illustrative conjugates according to this embodiment have the formula:
  • saccharyl moieties include, without limitation, glucose, glucosamine, N-acetyl-glucosamine, galactose, galactosamine, N-acetyl-galactosamine, mannose, mannosamine, N-acetyl-mannosamine, and the like.
  • the glycosyl structures on the peptide conjugates of the invention can have substantially any structure.
  • the glycans can be O-linked or N-linked.
  • each of the pyranose and furanose derivatives discussed above can be a component of a glycosyl moiety of a peptide.
  • the invention provides a modified EPO peptide that includes a glycosyl group having the formula:
  • the group has the formula:
  • the group has the formula:
  • the group has the formula:
  • index p represents and integer from 1 to 10; and a is either 0 or 1.
  • a glycoPEGylated EPO peptide of the invention includes at least one N-linked glycosyl residue selected from the glycosyl residues set forth below:
  • R 15′ represents H, OH (e.g., Gal-OH), a sialyl moiety, a polymer modified sialyl moiety (i.e., glycosyl linking group-polymeric modifying moiety (Sia-L-R 1 )) or a sialyl moiety to which is bound a polymer modified sialyl moiety (e.g., Sia-Sia-L-R 1 ) (“Sia-Sia p ”).
  • An exemplary EPO peptide of the invention will include at least one glycan having a R 15′ .
  • the oxygen, with the open valence, of Formulae I and II is preferably attached through a glycosidic linkage to a carbon of a Gal or GalNAc moiety.
  • the oxygen is attached to the carbon at position 3 of a galactose residue.
  • the modified sialic acid is linked ⁇ 2,3- to the galactose residue.
  • the sialic acid is linked ⁇ 2,6- to the galactose residue.
  • the modified glycan is bound to one or more position selected from Asn 24, Asn 38, Asn 83 and/or Ser 126.
  • the EPO is derived from mammalian cells and the modifying group is only on the glycan at Asn 24.
  • the glycosyl linking moiety is linked to a Sia residue through another Sia residue, e.g.:
  • An exemplary species according to this motif is prepared by conjugating Sia-L-R 1 to a terminal sialic acid of the glycan at Asn 24 using an enzyme that forms Sia-Sia bonds, e.g., CST-II, ST8Sia-II, ST8Sia-III and ST8Sia-IV.
  • the glycans have a formula that is selected from the group:
  • the glycans of this group generally correspond to those found on an EPO peptide that is produced by insect cells (e.g., Sf9), followed by remodeling of the glycan and glycoPEGylation according to the methods set forth herein.
  • insect-derived EPO that is expressed with a tri-mannosyl core is subsequently contacted with a GlcNAc donor and a GlcNAc transferase and a Gal donor and a Gal transferase.
  • Appending GlcNAc and Gal to the tri-mannosyl core is accomplished in either two steps or a single step.
  • a modified sialic acid is added to at least one branch of the glycosyl moiety as discussed herein.
  • Those Gal moieties that are not functionalized with the modified sialic acid are optionally “capped” by reaction with a sialic acid donor in the presence of a sialyl transferase.
  • At least 60% of terminal Gal moieties in a population of peptides is capped with sialic acid, preferably at least 70%, more preferably, at least 80%, still more preferably at least 90% and even more preferably at least 95%, 96%, 97%, 98% or 99% are capped with sialic acid.
  • an exemplary modified EPO peptide of the invention will include at least one glycan with an R 15′ moiety having a structure according to Formulae I or II.
  • the EPO is derived from insect cells, which are remodeled by adding GlcNAc and Gal to the mannose core.
  • the remodeled peptide is glycopegylated using a sialic acid bearing a linear PEG moiety, affording an EPO peptide that comprises at least one moiety having the formula:
  • the EPO peptide includes three such moieties, one attached at each of Asn 24, Asn 38 and Asn 83. In another embodiment, the peptide includes two such moieties attached at a combination of two of these Asn moieties.
  • a composition that is a mixture of these two species (i.e., PEG 3 and PEG 2 ). The mixture preferably includes at least 75%, preferably at least 80%, more preferably at least 85%, still more preferably 90% and even more preferably 95%, 96%, 97% or 98% of the species that includes the three modified glycosyl residues. Unmodified terminal Gal residues are optionally capped with Sia as discussed above.
  • the peptide is expressed in insect cells, remodeled and glycopegylated.
  • e for each of the modified glycosyl moieties is an integer that provides as PEG moiety having a molecular weight of approximately 10 kDa.
  • membrane filtration and chromatographic techniques described below are useful steps within the methods of the invention and apply to the isolation of polypeptide conjugates, in which a polypeptide is linked to at least one modifying group, such as a poly(alkylene oxide) moiety. It is to be understood that unless the order of steps is explicitly recited, the exemplary steps can be performed in any desired order.
  • the methods of the invention includes one or more membrane filtration steps.
  • Membrane filtration is a separation technique widely used for clarifying, concentrating, and purifying polypeptides.
  • the polypeptide purification process of the present invention includes at least one diafiltration/ultrafiltration step, e.g., as the final process step to generate a composition suitable for storage.
  • diafiltration/ultrafiltration is performed to condition a mixture for a chromatographic process step.
  • the eluate from a hydrophobic interaction chromatography step is concentrated and the buffer is exchanged to prepare the sample for the next purification step (e.g., cation exchange chromatography).
  • the diafiltration step is employed to concentrate the sample. In another exemplary embodiment the diafiltration step is employed to alter the buffer. In yet another exemplary embodiment, the new buffer is suitable for storage of the purified polypeptide conjugate.
  • the diafiltration/ultrafiltration membrane can have any molecular weight cuttoff (MWCO) specification.
  • the feed is passed through an ultrafiltration membrane with a MWCO suitable to concentrate the purified polypeptide conjugate.
  • the selected MWCO will depend on the combined size of the polypeptide and the modifying group, such as the size of a poly(alkylene oxide) moiety covalently linked to the polypeptide.
  • the membrane is chosen to have a MWCO that is substantially lower than the molecular weight of the purified peptide conjugate.
  • the ultrafiltration membrane is selected to have a MWCO that is 3 to 6 times lower than the molecular weight of the peptide conjugate to be retained by the membrane.
  • the diafiltration membrane has a MWCO of about 2 kDa to about 500 kDa. In another exemplary embodiment, the diafiltration membrane has a MWCO of about 5 kDa to about 400 kDa, about 5 kDa to about 300 kDa or about 5 kDa to about 200 kDa.
  • the diafiltration membrane has a MWCO of about 5 kDa to about 180 kDa, 5 kDa to about 160 kDa, 5 kDa to about 140 kDa, 5 kDa to about 130 kDa, 5 kDa to about 120 kDa, 5 kDa to about 110 kDa, or 5 kDa to 100 kDa.
  • the diafiltration membrane has a MWCO of about 5 kDa to about 80 kDa, 5 kDa to about 60 kDa, 5 kDa to about 40 kDa or 5 kDa to about 20 kDa.
  • the diafiltration membrane has a MWCO of about 8 kDa to about 12 kDa and preferably about 10 kDa.
  • filtration is effected using a transmembrane pressure between about 1 and about 30 psi and a filter membrane with a MWCO of between about 5 kDa to about 15 kDa, and preferably 10 kDa.
  • the filtration step produces a retentate stream and a permeate stream.
  • the retentate may be recycled to a reservoir for the peptide solution feed under conditions of essentially constant peptide concentration in the feed by adding a buffer solution to the retentate.
  • the surface area of the filtration membrane used will generally depend on the amount of peptide conjugate to be purified.
  • the membrane may be made of a low-binding material to minimize adsorptive losses and is preferably durable, cleanable, and chemically compatible with the buffers to be used.
  • a number of suitable membranes are commercially available.
  • the ultrafiltration/diafiltration membrane is a member selected from cellulose acetate, regenerated cellulose and polyethersulfone. Suitable membranes include those, in which the membrane polymer is chemically modified. In a preferred embodiment, the membrane is regenerated cellulose.
  • the flow rate is adjusted to maintain a constant transmembrane pressure.
  • filtration will proceed faster with higher pressures and higher flow rates, but higher flow rates may also result in damage to the peptide or loss of peptide due to passage through the membrane.
  • various devices may have certain pressure limitations on their operation, and the pressure is adjusted according to the manufacturer's specification. In an exemplary embodiment, the pressure is between about 1 to about 30 psi, and in another exemplary embodiment the pressure is between about 8 psi to about 15 psi.
  • the circulation pump is a peristaltic pump or diaphragm pump in the feed channel and the pressure is controlled by adjusting the retentate valve.
  • the retentate is collected.
  • Water or an aqueous buffer e.g. diafiltration buffer
  • the wash liquid may be combined with the original retentate containing the concentrated peptide.
  • the retentate is optionally dialyzed against another buffer, such as TRIS or HEPES.
  • the purified product is stored at a low temperature.
  • the product is stored at about ⁇ 20° C. at a polypeptide concentration of about 1 mg to about 10 mg of peptide conjugate per mL storage buffer.
  • the product solution maybe sterile filtered, e.g., using a membrane filter having a pore size of about 2 ⁇ M (e.g., cellulose acetate filter).
  • chromatographic techniques such as size exclusion chromatography (gel filtration), ion exchange chromatography, hydrophobic interaction chromatography (HIC), affinity chromatography, mixed-mode chromatography, hydroxyapatite and fluoroapatite chromatography are used for the isolation of polypeptides and proteins. These technologies can also be used to isolate polypeptide conjugates.
  • methods of the invention employ a combination of several chromatographic techniques. The order in which these steps are performed is dependent on the nature of the polypeptide conjugate being purified and the nature of the contaminants to be removed.
  • Suitable techniques for the practice of the invention separate the polypeptide conjugate of interest from a variety of contaminants on the basis of charge, degree of hydrophobicity, and/or size. Different chromatographic resins and membranes are available for each of these techniques, allowing accurate tailoring of the purification scheme.
  • the components in a mixture interact differently with the column material and move at different rates along the column length, achieving a physical separation that increases as the components pass through the column.
  • components of the mixture including the peptide conjugate of interest, adhere selectively to the separation medium, while other components are found in the flow-through.
  • the initially retained components are then eluted differentially by varying the composition of the solvent or buffer system.
  • the desired components are found in the flow-through while impurities are retained on the column and thus removed from the mixture.
  • the methods of the invention employ at least one ion exchange chromatography step.
  • Anion and cation exchange chromatography are known in the art. Ion exchange chromatography separates compounds based on their net charge. Ionic molecules are classified as either anions (having a negative charge) or cations (having a positive charge). Some molecules (e.g., proteins) may have both anionic and cationic groups. A positively charged support (anion exchanger) will bind a compound with an overall negative charge. Conversely, a negatively charged support (cation exchanger) will bind a compound with an overall positive charge. Ion exchange matrices can be further categorized as either strong or weak exchangers.
  • Strong ion exchange matrices are charged (ionized) across a wide range of pH levels. Weak ion exchange matrices are ionized within a narrow pH range.
  • the ionic groups of exchange columns are covalently bound to the gel matrix and are compensated by small concentrations of counter ions, which are present in the buffer.
  • the most common ion exchange chemistries include: quaternary ammonium residues (Q) for strong anion exchange, diethylaminoethyl residues (DEAE) for weak anion exchange, sulfopropyl (SP) resins and sulfonic acid (S) resins for strong cation exchange and carboxymethyl residues (CM) for weak cation exchange.
  • the sample components are preferably collected at the top of the column.
  • the gradient is begun with the addition of a stronger eluting mobile phase, the sample components begin their separation. If poor separation is observed, it might be improved by a change in the gradient slope. If the polypeptide conjugate does not bind to the column under the selected conditions, the composition and/or the pH of the starting buffer should be changed.
  • the buffer system can further be optimized by choosing different buffer salts since each buffer composition solvates the ion exchanger and the sample components uniquely.
  • any conventional buffer system with a salt concentration of about 5 mM up to about 50 mM can be used for ion exchange chromatography.
  • positively charged buffering ions are used for anion exchangers and negatively charged ones are used for cation exchangers.
  • Phosphate buffers are generally used on both exchanger types.
  • the highest salt concentration that permits binding of the peptide of interest is used as the starting condition.
  • all buffers are prepared from MilliQ-water and filtered (0.45 or 0.22 ⁇ m filter).
  • a sample containing the peptide conjugate of interest is loaded onto an anion exchanger in a loading buffer comprising a salt concentration below the concentration at which the peptide would elute from the column.
  • the pH of the buffer is selected so that the purified peptide is retained on the anion exchange medium. Changing the pH of the buffer alters the charge of the peptide, and lowering the pH value shortens the retention time with anion exchangers.
  • the isoelectric point (pI) of a protein is the pH at which the charge of a protein is zero.
  • the pH value of the buffer is kept 1.5 to 2 times higher than the pI value of the peptide of interest.
  • the anion exchange conditions are selected to preferentially bind impurities, while the purified peptide is found in the flow-through.
  • the column may be washed with several column volumes (CV) of buffer to remove unbound substances and/or those substances that bind weakly to the resin. Fractions are then eluted from the column using, for example, a saline gradient according to conventional methods. The salt in the solution competes with the protein in binding to the column and the protein is released. Components with weak ionic interactions elute at a lower salt concentration than components with a strong ionic interaction. Sample fractions are collected from the column. Fractions containing high levels of the desired peptide and low levels of impurities are pooled or processed separately.
  • CV column volumes
  • anion exchange used in the process of the current invention is employed to isolate the polypeptide conjugate from contaminants such as particulates, chemicals and proteins/peptides (e.g., enzymes used in a glycoPEGylation reaction).
  • Anion exchange media are known to those of skill in the art. Exemplary anion exchange media are described, e.g., in Protein Purification Methods, A Practical Approach , Ed. Harris E L V, Angal S, IRL Press Oxford, England (1989); Protein Purification , Ed. Janson J C, Ryden L, VCH-Verlag, Weinheim, Germany (1989); Process Scale Bioseparations for the Biopharmaceutical Industry , Ed. Shukla A A, Etzel M R, Gadam S, CRC Press Taylor & Francis Group (2007), pages 188-196 ; Protein Purification Handbook , GE Healthcare 2007 (18-1132-29) and Protein Purification, Principles, High Resolution Methods and Applications (2 nd Edition 1998), Ed.
  • An exemplary anion exchanger of the invention is selected from quaternary ammonium filters and DEAE resins.
  • the anion exchanger is a quaternary ammonium resin (e.g. Mustang Q ion exchange membrane, Pall Corporation).
  • the anion exchanger is Sartobind Q.
  • Other useful resins include QXL, Capto and BigBeads resins.
  • the method of the invention includes at least one cation exchange step.
  • a sample containing the peptide conjugate of interest is loaded onto a cation exchange resin in a loading buffer comprising a salt concentration below the concentration at which the peptide would elute from the column.
  • the pH of the loading buffer is selected so that the peptide conjugate of interest is retained on the cation exchange resin.
  • Changing the pH of the buffer alters the charge of the peptide and increasing the pH of the buffer shortens the retention times with cation exchangers.
  • cation exchanges are performed at 1.5 to 2 pH units below the peptide's pI.
  • the cation exchange conditions are selected to preferentially bind impurities, while the purified peptide is found in the flow-through.
  • the column is washed with several column volumes of buffer to remove unbound substances and those substances that bind weakly to the resin. Fractions are then eluted from the column using a salt gradient according to conventional methods. Sample fractions are collected from the column. One or more fraction containing high levels of the desired peptide and low levels of impurities are collected, and optionally pooled.
  • the cation exchangers used in the process of the current invention provide one of the primary purification steps of the purification process.
  • the cation exchanger removes undesired proteins from the mixture, which contains the peptide conjugate of interest.
  • the cation exchange step is useful to remove unwanted glycoforms of the purified polypeptide conjugate.
  • Cation exchange media are known to those of skill in the art. Exemplary cation exchange media are described, e.g., in Protein Purification Methods, A Practical Approach , Ed. Harris E L V, Angal S, IRL Press Oxford, England (1989); Protein Purification , Ed. Janson J C, Ryden L, VCH-Verlag, Weinheim, Germany (1989); Process Scale Bioseparations for the Biopharmaceutical Industry , Ed. Shukla A A, Etzel M R, Gadam S, CRC Press Taylor & Francis Group (2007), pages 188-196 ; Protein Purification Handbook , GE Healthcare 2007 (18-1132-29) and Protein Purification, Principles, High Resolution Methods and Applications (2 nd Edition 1998), Ed.
  • cation exchange resins of use in the invention are selected from sulfonic acid (S) and carboxymethyl (CM) supports.
  • the cation exchanger is a sulfonic acid support (e.g. UNOsphereS, Bio-Rad Laboratories) or a sulphopropyl (SP) resin.
  • the cation exchange resin is selected from SPFF, SPHP sepharose, BigBeads SP, Capto S and the like.
  • the cation exchanger is Source 15S.
  • the ion exchangers used in the methods of the invention are optionally membrane adsorbers rather than chromatographic resins or supports.
  • the membrane adsorber is a cation exchanger.
  • the membrane adsorber is a sulfonic acid (S) cation exchanger (e.g. Sartobind S, Sartorius AG).
  • S sulfonic acid
  • the membrane adsorber is optionally disposable.
  • the peptide conjugate purification process of the invention includes mixed-mode or pseudo-affinity chromatography.
  • the process involves chromatography performed on ceramic or crystalline apatite media, such as hydroxyapatite (HA) chromatography and fluoroapatite (FA) chromatography.
  • HA and FA chromatography are effective purification mechanisms, providing biomolecule selectivity, complementary to ion exchange and/or hydrophobic interaction techniques. Hydroxyapatite and fluoroapatite chromatography are known in the art.
  • the apatite medium is Adhere MMC.
  • Exemplary hydroxyapatite sorbents of type I and type II are selected from ceramic and crystalline materials. Hydroxyapatite sorbents are available in different particle sizes (e.g. type 1, Bio-Rad Laboratories). In an exemplary embodiment, the particle size of the hydroxyapatite sorbent is between about 20 ⁇ m and about 180 ⁇ m, between about 20 ⁇ m and about 100 ⁇ m or between about 60 ⁇ m and about 100 ⁇ m. In a particular example, the particle size of the hydroxyapatite sorbent is about 80 ⁇ m.
  • the hydroxyapatite sorbent is composed of cross-linked agarose beads with microcrystals of hydroxyapatite entrapped in the agarose mesh.
  • the agarose is chemically stabilized (e.g. with epichlorohydrin under strongly alkaline conditions).
  • the hydroxyapatite sorbent is HA Ultrogel (Pall Corporation).
  • Exemplary type I and type II fluoroapatite sorbents are selected from ceramic (e.g., bead-like particles) and crystalline materials. Ceramic fluoroapatite sorbents are available in different particle sizes (e.g. type 1 and type 2, Bio-Rad Laboratories). In an exemplary embodiment the particle size of the ceramic fluoroapatite sorbent is from about 20 ⁇ m to about 180 ⁇ m, preferably about 20 to about 100 ⁇ m, more preferably about 20 ⁇ m to about 80 ⁇ m. In one example, the particle size of the ceramic fluoroapatite medium is about 40 ⁇ m (e.g., type 1 ceramic fluoroapatite).
  • the fluoroapatite medium includes hydroxyapatite in addition to fluoroapatite.
  • the fluoroapatite medium is Bio-Rad's CFTTM Ceramic Fluoroapatite.
  • the loading flow velocity is selected from about 30 to about 900 cm/h, from about 150 to about 900 cm/h, preferably from about 500 to about 900 cm/h and, more preferably, from about 600 to about 900 cm/h.
  • the pH of the elution buffer is selected from about pH 7 to about pH 9, and preferably from about pH 7.5 to about pH 8.0.
  • the present invention provides a method of purifying a recombinant peptide by hydroxyapatite or fluoroapatite chromatography.
  • the method includes the following steps: (a) desalting a mixture containing the peptide, forming a desalted mixture (e.g.
  • the mixture containing the peptide of interest is desalted prior to subjecting the mixture to HA or fluoroapatite chromatography.
  • the desalting step increases the capacity of the apatite column to bind the peptide of interest.
  • the apatite column capacity increases with decreasing salt conductivity of the load, which contains the peptide.
  • the mass loading of peptide per liter of HA resin is from about 1 to about 25 g/L, from about 1 to about 20 g/L, preferably from about 1 to about 15 g/L and more preferably from about 1 to about 10 g/L.
  • the conductivity of the load can be decreased using a method selected from desalting and diluting.
  • the conductivity of the loading buffer is lowered by desalting and preferred conductivities are from about 0.1 to about 4.0 mS/cm, preferably from about 0.1 to about 1.0 mS/cm, more preferably from about 0.1 to about 0.6 mS/cm and, still more preferably, from about 0.1 to about 0.4 mS/cm.
  • desalting of peptide conjugate solutions is achieved using membrane filters wherein the membrane filter has a MWCO smaller than the peptide/protein of interest.
  • the peptide/protein is found in the retentate and is reconstituted in a buffer of choice.
  • the MWCO of the membrane used for desalting must be relatively small in order to avoid leaking of the peptide through the membrane pores.
  • filtering a large volume of liquid through a small MWCO membrane typically requires large membrane areas and the filtering process is time consuming.
  • desalting of the HA or fluoroapatite chromatography load is accomplished using size-exclusion chromatography (e.g. gel filtration).
  • size-exclusion chromatography e.g. gel filtration
  • the technique separates molecules on the basis of size.
  • high molecular weight components can travel through the column more easily than smaller molecules, since their size prevents them from entering bead pores. Accordingly, low-molecular weight components take longer to pass through the column.
  • low molecular weight materials, such as unwanted salts can be separated from the peptide of interest.
  • the column material is selected from dextran, agarose, and polyacrylamide gels, in which the gels are characterized by different particle sizes.
  • the material is selected from rigid, aqueous-compatible size exclusion materials.
  • An exemplary gel filtration resin of the invention is Sepharose G-25 resin (GE Healthcare).
  • desalting is performed subsequent to cation exchange chromatography (e.g. after Source 15S chromatography).
  • an amino acid is added to the elution buffer, which is used to elute the peptide of interest from the HA or fluoroapatite resin.
  • the amino acid is added to the elution buffer at a final concentration of about 5 mM to about 50 mM, about 10 mM to about 40 mM, preferably about 15 mM to about 30 mM and, more preferably, about 20 mM.
  • the addition of an amino acid (e.g. glycine) to the elution buffer increases the step recovery of peptide from HA chromatography when compared to the recovery obtained without the addition of an amino acid.
  • the recovery of peptide is increased by addition of the amino acid at least about 1% to about 20%, by at least about 1% to about 15%, by at least about 1% to about 10%, preferably by at least about 1% to about 7% and, more preferably, by about 5%.
  • the addition of an amino acid causes the elution peak of the purified peptide to be sharper. Thus, less peptide is recovered in the tail fractions of the peak and more peptide is recovered in the main peak.
  • the addition of an amino acid does not decrease the purity of the product from HA chromatography.
  • the amino acid is glycine.
  • glycine is added to the elution buffer at a final concentration of 20 mM.
  • Hydrophobic interaction chromatography is a liquid chromatography technique that separates biomolecules based on differences in their surface hydrophobicity.
  • hydrophobic amino acid side chains exposed on the surface of a polypeptide can interact with hydrophobic moieties on the HIC matrix.
  • the amount, of exposed hydrophobic amino acids differs between polypeptides and so does the ability of polypeptides to interact with HIC gels.
  • Hydrophobic interaction between a biomolecule and the HIC matrix is typically enhanced by high ionic strength buffers, and of biomolecules is most often performed at high salt concentrations. The elution of the peptide of interest from the column is then initiated by decreasing salt gradients.
  • the HIC resin is selected for optimal resolution of different polypeptide glycoforms.
  • Exemplary HIC resins useful in the methods of the invention are described, e.g., in Protein Purification Methods, A Practical Approach , Ed. Harris ELV, Angal S, IRL Press Oxford, England (1989) page 224 , Protein Purification , Ed. Janson J C, Ryden L, VCH-Verlag, Weinheim, Germany (1989) pages 207-226 , Process Scale Bioseparations for the Biopharmaceutical Industry , Ed.
  • HIC media are distinguished by the hydrophobic moiety that they carry, by the particle size (e.g. bead size), the pore size and the density of the hydrophobic moieties on the HIC matrix (e.g. low substitution or high substitution).
  • the hydrophobic moieties of the column matrix are members selected from alkyl groups, aromatic groups and ethers.
  • Exemplary hydrophobic alkyl groups include lower alkyl groups, such as n-propyl, isopropyl, n-butyl, iso-butyl, and n-octyl.
  • Exemplary aromatic groups include substituted and unsubstituted phenyl.
  • the matrix of the HIC medium is a member selected from agarose, sepharose (GE Healthcare), polystyrene, divinylbenzene, and combinations thereof.
  • Exemplary HIC resins include Butyl Fast Flow and Phenyl Fast Flow (e.g., GE Healthcare) in either low or high substituted versions.
  • the HIC resin is a phenyl resin.
  • the HIC resin is Phenyl650S or Phenyl650M (e.g., Tosohaas, Toyopearl).
  • the HIC medium is selected from the following commercial resins:
  • the amount of polypeptide conjugate loaded onto the HIC medium is between about 0.05 and about 1.0 mg conjugate/mL resin. In one example, the loaded amount of polypeptide conjugate is selected between about 0.05 and 0.3 mg conjugate/mL resin. In another example, the HIC medium is loaded with between about 0.1 and about 0.2 mg conjugate/mL resin (e.g., 0.15-0.18 mg/mL). In another embodiment, the amount of polypeptide conjugate loaded onto the HIC column is optimized for recovery of peptide conjugate and resolution of glycoforms.
  • the HIC loading conditions are selected to create an HIC eluate that includes less than about 8%, preferably less than about 7%, more preferably less than about 6%, even more preferably less than about 5% and most preferably less than about 4% of EPO-PEG 2 .
  • the loading buffer (the buffer in which the purified polypeptide conjugate is applied to the HIC column) is selected to bind the purified polypeptide conjugate to the HIC medium. Unbound impurities are then washed off the column using a HIC wash buffer. Consequently, polypeptide conjugates are eluted using an HIC elution buffer.
  • the HIC loading buffer, the HIC wash buffer and the HIC elution buffer each contain one or more salts, such as sodium acetate (NaOAc), sodium chloride (NaCl), sodium sulfate (Na 2 SO 4 ) and sodium phosphate.
  • salts such as sodium acetate (NaOAc), sodium chloride (NaCl), sodium sulfate (Na 2 SO 4 ) and sodium phosphate.
  • concentration ranges for these and other salts are generally optimized for each type of HIC resin to affect optimal binding of the polypeptide conjugate being purified.
  • the HIC loading buffer includes sodium sulfate (Na 2 SO 4 ) or ammonium sulfate, (NH 4 ) 2 SO 4 .
  • the concentration of sodium- or ammonium sulfate in the loading buffer is about 100 mM to about 1200 mM.
  • the concentration of sodium sulfate in the HIC loading buffer is about 300 mM to about 1100 mM, about 300 mM to about 1000 mM, about 300 mM to about 900 mM, about 300 mM to about 800 mM, about 300 mM to about 700 mM, about 300 mM to about 600 mM or about 300 mM to about 500 mM.
  • the concentration of sodium sulfate in the HIC loading buffer is about 400 mM to about 800 mM. In a further exemplary embodiment, the concentration of sodium sulfate in the HIC loading buffer is about 500 mM to about 700 mM, and preferably about 600 mM.
  • the HIC loading buffer, HIC wash buffer and HIC elution buffer include sodium phosphate.
  • the concentration of sodium phosphate in any of these buffers is selected between about 5 mM and about 70 mM.
  • the concentration of sodium phosphate in the HIC wash buffer is selected between about 10 mM and about 50 mM, between about 10 mM and about 30 mM or between about 20 mM and about 30 mM.
  • the sodium phosphate concentration in the HIC wash buffer and elution buffer is about 25 mM.
  • the HIC wash buffer has a pH of about 4.0 to about 8.0.
  • the pH of the HIC wash buffer is selected from about 5.0 to about 8.0.
  • the pH is selected from about 6 to about 8.
  • the pH is selected from about 6.5 to about 8.0.
  • the pH of the HIC wash buffer is selected from about 7.0 to about 8.0, from about 7.0 to about 7.9, from about 7.0 to about 7.8, from about 7.0 to about 7.7, from about 7.0 to about 7.6 or from about 7.0 to about 7.5.
  • the pH of the HIC wash buffer is about 7.5.
  • the purified polypeptide conjugate is eluted from the HIC resin using a gradient of decreasing sodium sulfate concentration.
  • the elution buffer does not contain any sodium sulfate.
  • HIC is employed as a method to separate polypeptide glycoforms, each covalently linked to at least one poly(alkylene oxide) moiety.
  • the elution gradient profile is selected to affect optimal resolution of different polypeptide glycoforms contained in the purified mixture.
  • the HIC elution buffer includes 25 mM sodium phosphate and a combination of gradient and hold periods spanning a range of about 600 mM sodium sulfate to no sodium sulfate in the phosphate buffer is employed to elute polypeptide conjugates from the HIC medium.
  • HIC is performed subsequent to anion exchange chromatography.
  • the flow-through from the anion exchanger, which contains the partially purified polypeptide conjugate is conditioned for hydrophobic interaction chromatography.
  • the anion exchange flow-through may be diluted with a buffer suitable as a loading buffer for HIC.
  • the anion exchange flow-through can be diluted with a buffer containing sodium sulfate to adjust the sodium sulfate concentration in the HIC load (e.g., a sodium sulfate concentration suitable to bind the polypeptide conjugate to the HIC medium, e.g., about 600 mM).
  • the anion exchange flow-through is concentrated before dilution.
  • the anion exchange flow-through is subjected to diafiltration/ultrafiltration for concentration and/or buffer exchange.
  • the polypeptide conjugate of interest is purified from a mixture (e.g., a reaction mixture, such as a glycoPEGylation reaction) using the exemplary purification process outlined in FIG. 1 .
  • a mixture e.g., a reaction mixture, such as a glycoPEGylation reaction
  • the product of the glycoPEGylation reaction is subjected to anion exchange chromatography/filtration (e.g., using a Sartobind Q resin).
  • impurities are bound by the anion exchange medium, while the purified polypeptide conjugate is found in the flow-through.
  • this anion exchange step is useful to remove catalytic enzymes used in a glycan remodeling and/or glycomodification (e.g., glycoPEGylation) reaction performed prior to the anion exchange procedure.
  • the anion exchange step is useful to isolate the polypeptide conjugate from at least one glycosyltransferase contained in the glycoPEGylation reaction mixture.
  • the flow-through of the anion exchange step containing the partially purified polypeptide conjugate is conditioned and then loaded onto a hydrophobic interaction chromatography resin.
  • the HIC medium is Phenyl 650S.
  • the anion exchange flow-through is conditioned to generate a HIC loading sample that includes a sufficient salt concentration to affect binding of the polypeptide conjugate to the HIC medium.
  • the anion exchange flow-trough is diluted with a buffer containing sodium sulfate.
  • the dilution buffer contains sufficient sodium sulfate to generate a HIC loading sample having a sodium sulfate concentration between about 500 mM and about 700 mM.
  • the anion exchange flow-through is diluted so that the HIC loading sample includes about 600 mM of sodium sulfate.
  • the HIC resin is washed with a wash buffer to elute unbound impurities.
  • the HIC wash buffer is a phosphate buffer.
  • the HIC wash buffer contains about 25 mM sodium phosphate at a pH of about 7.5.
  • the polypeptide conjugate is eluted from the HIC medium using an elution buffer.
  • the polypeptide conjugate is eluted from the HIC medium using a phosphate buffer (e.g., 25 mM sodium phosphate at pH 7.5) and a gradient of decreasing sodium sulfate in the phosphate buffer.
  • the conjugate is eluted using a gradient from about 600 mM to about 0 mM sodium sulfate.
  • Eluate fractions are collected and optionally analyzed for product content.
  • Product containing fractions are pooled and the resulting HIC pool is optionally conditioned for loading onto a cation exchange medium.
  • the HIC pool is concentrated and the buffer is exchanged using diafiltration.
  • the diafiltration membrane has a MWCO of 10 kDa.
  • the volume of the HIC pool is reduced to between about 1/30 and about 1/10 of the original volume. In a particular example, the HIC pool volume is reduced to about 1/20 of the original volume.
  • the buffer may then be exchanged, for example, by diluting the sample with the new buffer and subsequently re-concentrating the sample. The dilution and re-concentration steps may be repeated (e.g., 2-6 times) until the new buffer has the desired composition (e.g., the desired salt conductivity).
  • the partially purified polypeptide conjugate may be transferred into the desired buffer using a two step buffer exchange.
  • the buffer In the first buffer exchange step, the buffer may be changed to a phosphate buffer that does not include sodium sulfate.
  • the pH of the resulting conjugate solution may optionally be adjusted (e.g., using sodium acetate.
  • the buffer In a second buffer exchange step, the buffer may be changed to a buffer system suitable for loading onto a cation exchanger.
  • the second buffer may include about 10 mM sodium acetate at a pH of about 5.4.
  • the loading buffer for the cation exchange step has a salt conductivity between about 1.0 and about 3.0 mS/cm (e.g., about 1.5 mS/cm).
  • the diafiltered HIC pool is subjected to cation exchange.
  • the cation exchanger is Source 15S.
  • the cation exchange medium is useful to further reduce the content of unwanted glycoforms of the purified polypeptide conjugate.
  • the partially purified polypeptide conjugate is applied to the cation exchange medium and unbound impurities are eluted using a cation exchange wash buffer (e.g., 10 mM sodium acetate, pH 5.4).
  • the bound polypeptide conjugate is then eluted using a cation exchange elution buffer.
  • the conjugate is eluted using increasing NaCl concentrations in the above wash buffer.
  • the conjugate is eluted using a gradient of 0-0.5 M NaCl.
  • the gradient elution profile which may include a combination of gradient and hold periods, is selected for optimized glycoform resolution. Eluate fractions are collected and optionally analyzed (e.g., for product content and purity). Selected product containing fractions are pooled to form a cation exchange pool.
  • the cation exchange pool is concentrated and diafiltered into a storage buffer. In one example, this diafiltration step also uses a 10 kDa MWCO membrane. In one embodiment, the volume of the cation exchange pool is reduced to about 1/100 to about 1/25 of its original volume (e.g., about 1/50 of the original volume).
  • the concentrated cation exchange pool is then subjected to buffer exchange, for example, by diluting the sample with the new buffer and subsequently re-concentrating the sample. The dilution and re-concentration step may be repeated (e.g., 2-6 times) until the new buffer has the desired composition.
  • the final retentate is reconstituted into a storage buffer.
  • Exemplary storage buffers include those having a sodium chloride concentration that is in the physiological range.
  • the storage buffer may be a sodium acetate buffer including about 150 mM NaCl.
  • the concentrated product pool is reconstituted in the storage buffer to reach a desired peptide concentration.
  • the final conjugate concentration is selected between about 0.5 and about 2 mg/mL.
  • the final solution is optionally sterile filtered, for example through a cellulose acetate membrane.
  • the purification process outlined in FIG. 1 may optionally include an additional chromatography step.
  • the process includes a hydroxyapatite (HA) or fluoroapatite chromatography step.
  • the apatite chromatography is performed after anion exchange chromatography.
  • the apatite chromatography is performed after HIC.
  • the apatite chromatography is performed after cation exchange chromatography.
  • the partially purified polypeptide conjugate solution may be desalted, for example, using a size exclusion column (e.g. G25) to lower the salt conductivity of the conjugate solution in preparation for apatite chromatography.
  • a size exclusion column e.g. G25
  • the polypeptide conjugate purified by the above described process is an EPO-conjugate.
  • the invention provides a composition made by a method of the invention.
  • the invention provides a composition including a first polypeptide conjugate, said first polypeptide conjugate having a first number of poly(alkylene oxide) moieties, each of the poly(alkylene oxide) moieties covalently linked to the first polypeptide via an intact glycosyl linking group.
  • the composition is made by a method including: (a) contacting a mixture comprising the first polypeptide conjugate with a hydrophobic interaction chromatography (HIC) medium; and (b) eluting the first polypeptide conjugate from the hydrophobic interaction chromatography medium.
  • HIC hydrophobic interaction chromatography
  • the invention provides an isolated first polypeptide conjugate made by a method comprising: separating the first polypeptide conjugate including a first number of poly(alkylene oxide) moieties covalently linked to a first polypeptide, from a second polypeptide conjugate comprising a second number of poly(alkylene oxide) moieties covalently linked to a second polypeptide.
  • the first number is selected from 1 to 20 and the second number is selected from 0-20. In another embodiment, the first number and the second number are different.
  • the two polypeptide conjugates are separated by: (a) contacting a mixture comprising the first polypeptide conjugate and the second polypeptide conjugate with a hydrophobic interaction chromatography (HIC) medium; and (b) eluting the first polypeptide conjugate from the hydrophobic interaction chromatography medium.
  • HIC hydrophobic interaction chromatography
  • the first polypeptide is a member selected from erythropoietin (EPO), bone morphogenetic protein 2 (BMP-2), bone morphogenetic protein 7 (BMP-7), bone morphogenetic protein 15 (BMP-15), neurotrophin-3 (NT-3), von Willebrand factor (vWF) protease, granulocyte colony stimulating factor (G-CSF), granulocyte-macrophage colony stimulating factor (GM-CSF), interferon alpha, interferon beta, interferon gamma, ⁇ 1 -antitrypsin ( ⁇ -1 protease inhibitor), glucocerebrosidase, tissue-type plasminogen activator (TPA), interleukin-2 (IL-2), leptin, hirudin, urokinase, human DNase, insulin, hepatitis B surface protein (HbsAg), chimeric diphtheria toxin-IL-2, human
  • EPO erythropoiet
  • the invention provides a composition including a first erythropoietin (EPO) conjugate, the first EPO conjugate having a first number of poly(alkylene oxide) moieties covalently linked to an EPO polypeptide via a glycosyl linking group (e.g., an intact glycosyl linking group).
  • EPO erythropoietin
  • the composition is made by a method including: (a) contacting a mixture including the first EPO conjugate with an anion exchange medium; (b) eluting the first EPO conjugate from the anion exchange medium, forming a first eluate comprising the first EPO conjugate; (c) contacting the first eluate with a hydrophobic interaction chromatography (HIC) medium; and (d) eluting the first EPO conjugate from the hydrophobic interaction chromatography medium.
  • HIC hydrophobic interaction chromatography
  • the invention provides a pharmaceutical formulation including a composition made by a method of the invention and a pharmaceutically acceptable carrier.
  • the pharmaceutical formulation includes an isolated polypeptide conjugate made by a method of the invention and a pharmaceutically acceptable carrier.
  • the isolated polypeptide conjugate is an EPO-conjugate.
  • the invention provides methods of treatment utilizing a composition (e.g., an isolated polypeptide conjugate) made by a method of the invention.
  • a composition e.g., an isolated polypeptide conjugate
  • the invention provides a method of treating a condition in a subject in need thereof, the condition characterized by compromised red blood cell production in the subject, the method comprising: administering to the subject an amount of a composition of the invention, effective to ameliorate the condition in the subject.
  • the subject is a mammal, such as a human.
  • the composition includes an EPO conjugate made by a method of the invention.
  • the invention provides a method of treating a tissue injury in a subject in need thereof.
  • the injury is caused by a member selected from ischemia, trauma, inflammation and contact with a toxic substance.
  • the method includes: administering to a subject an amount of a composition made by a method of the invention (e.g., an isolated polypeptide conjugate) that is effective in ameliorating the damage associated with the tissue injury.
  • a composition made by a method of the invention e.g., an isolated polypeptide conjugate
  • the subject is a mammal, such as a human.
  • the composition includes an EPO conjugate made by a method of the invention.
  • the invention provides a method of enhancing red blood cell production in a mammal, said method comprising administering to said mammal a composition made by a method of the invention.
  • the subject is a mammal, such as a human.
  • the composition includes an EPO conjugate made by a method of the invention.
  • the protein concentration was determined using either a UV method (280 nm) or was determined using BCA Protein Assay kit according to manufacture's instructions (Pierce).
  • the ratio of each EPO-PEG form [EPO-(SA-PEG-10 kDa) 1-4 ] was determined using C 3 RP HPLC chromatography (Zorbax 300SB-C3, 150 ⁇ 2.1 mm, 5 micron, 45° C.).
  • the HPLC was performed using the following solutions: A, 0.1% TFA in water, and B, 0.09% TFA in ACN.
  • the mobile phase was performed as a gradient from 42-55% B over 14 min, 55-95% B over 2 min, a 2 min wash at 95% B and then 95-42% B over 2 min.
  • the total time conduct the chromatography was 30 min using a flow rate of 0.6 mL/min. Mixture 1 was used as a reference standard to test system suitability.
  • the injection volume was varied to give a standard injection concentration of 5 to 10 ⁇ g of EPO-(SA-PEG-10 kDa) 3 based on protein. Protein absorbance was detected at 214 nm and the peak areas of the EPO species were used to determine the protein purity. All peaks were integrated using 32 karat software. The EPO-(SA-PEG-10 kDa) 4 peak could typically not be integrated accurately due to its small area.
  • the EPO-(SA-PEG-10 kDa) 3 isoform purity and aggregation were determined by SEC HPLC chromatography (TSK-gel G5000PW ⁇ L, 7.8 ⁇ 300 mm, 10 micron, 4° C.).
  • the isocratic mobile phase 100 mM sodium phosphate, 150 mM sodium chloride, pH 7.0 was used to perform the method at a flow rate of 0.5 mL/min.
  • This example describes the development of an isolation process for the isolation of EPO conjugates from a glycoPEGylation reaction mixture.
  • the resulting process is characterized by high overall EPO conjugate recovery and produces the desired EPO conjugate [EPO-(SA-PEG-10 kDa) 3 ] in high purity.
  • the composition produced by the process is essentially free of other EPO-PEG glycoforms, such as mono-, di- and tetra-PEGylated EPO conjugates.
  • the desired EPO-PEG conjugate is a glycoPEGylated erythropoietin protein that contains three 10 kDa mPEG groups attached to each of the three monoantennary N-linked glycans.
  • the EPO polypeptide is produced by expression of the protein from Sf9 cells using a Baculovirus infection protocol.
  • the insect cell expression system produces EPO with three N-linked glycans, at Asn 24 , Asn 38 and Asn 83 , each containing a trimannosyl core as the predominant species.
  • a variety of other glycan structures are present in small amounts that vary with fermentation conditions.
  • the other structures include trimannosyl core with an additional GlcNAc, higher mannose forms (Man 4 , Man 5 ), missing glycans, and GlcNAc-(Fuc) stub arising as a result of an endoglycosidase-type (Endo-H) activity.
  • GlcNAc-(Fuc) stub arising as a result of an endoglycosidase-type (Endo-H) activity.
  • a small percentage of the EPO molecules contain O-linked glycans at Ser 126 .
  • a very low level ( ⁇ 1%) of phosphorylcholine-linked glycans (PC-glycan) has also been identified in the insect cell-derived EPO.
  • EPO-(SA-PEG-10 kDa) 3 which contains three PEGylated mono-antennary N-linked glycans.
  • EPO-(SA-PEG-10 kDa) 1-2 are also present in lesser amounts, arising from the EPO forms missing one or more glycans or containing one or more GlcNAc-stub glycans.
  • EPO-(SA-PEG-10 kDa) 4 (and higher) are produced at very low levels.
  • EPO glycoforms which contain the tri-mannosyl core with an additional GlcNAc on the Man ⁇ 1,6 branch. Both branches of these glycans can be built out and PEGylated under the reaction conditions, resulting in biantennary PEGylated glycans on a tetra-PEG (or higher) EPO species. Exemplary EPO glycoforms are depicted in FIG. 3 .
  • GlycoPEGylated EPO isolated using reversed-phase chromatography and cation exchange chromatography provided a composition containing greater than or equal to about 85% EPO-(SA-PEG-10 kDa) 3 , about 3-14% EPO-(SA-PEG-10 kDa) 2 , about 1-8% EPO-(SA-PEG-10 kDa) 4-6 and less than or equal to about 1% EPO-(SA-PEG-10 kDa) i .
  • the above composition is referred to as Mixture 1.
  • HIC was capable of separating different PEG states (glycoforms) contained in a mixture, which results from a glycoPEGylation process (e.g., fractionation of isoforms EPO-(SA-PEG-10 kDa) 1-4 ).
  • the HIC purification method was optimized by evaluating a variety of HIC resins and process parameters.
  • a process based on Phenyl 650S resin e.g., Tosohaas, Toyopearl was selected for incorporation into the new isolation process.
  • an anion exchange step which is useful to remove enzyme components of the glycoPEGylation reaction. Enzymes are bound by the anion exchange medium, while the EPO-PEG conjugates EPO-(SA-PEG-10 kDa) 1-6 are found in the flow through.
  • Tricorn 5 column packed with 1 mL (0.5 cm ⁇ 5 cm) of the appropriate resin (Table 1) or a prepacked HiTrap column (1 mL, 0.7 cm ⁇ 2.5 cm) was attached to an AKTA FPLC system (GE Healthcare). Product elution was monitored by absorbance at 280 nm. Each column was equilibrated with 5 column volumes (CV) of Buffer B (as indicated in Table 1). Mixture 1 (100 mcg, 250 mcL) was diluted to 1.25 mL with 1 mL of Buffer B and injected using a 2 mL sample loop. The unbound material was washed with 5 CV of Buffer B.
  • the EPO-(SA-PEG-10 kDa) 3 was eluted with the following gradient elution using Buffer A (as indicated in Table 1): 100-0% Buffer B over 20 CV followed by 0% Buffer B for 5 CV. All steps were performed at 0.64 mL/min (196 cm/hr). The resulting chromatograms were compared and selected fractions were analysed by SDS-PAGE. Resins were selected for their capability to retain the EPO-PEG conjugates, their capability to resolve different glycoforms and peak shapes.
  • Phenyl-FF Low sub was compared to Phenyl 650M and Butyl 650M using either 25 mM Na phosphate, 1 M ammonium Sulfate, pH 7.0 or 25 mM Na phosphate, 4 M ammonium Sulfate, pH 7.0 as binding buffer (loading buffer). Butyl 650M was only tested with ammonium sulfate and resulted in a very wide and tailing elution peak. Both Phenyl 650M and Phenyl-FF low sub had a more symmetric peak shape when using sodium sulfate. The use of sodium chloride resulted in extremely broad elution peaks.
  • a third round of HIC testing compares the Phenyl 650M purification of Mixture 1 with different buffers.
  • Buffer B 1 M Na Sulfate+0.5 M NaCl+20 mM NaOAc, pH 5.0 was tested as this condition had worked for purification of the EPO intermediate prior to PEGylation. This condition resulted in a broad elution peak with multiply bumps, as did the same buffer without 0.5M NaCl (1 M Na Sulfate+20 mM NaOAc, pH 5.0).
  • Phenyl 650M was tested using Mixture 1 bound to the column using 4 M NaCl+buffer A, pH 7.0 and the elution was performed with 25 mM Na phosphate, 20% ethylene glycol, pH 7.0.
  • a Tricorn 5 column packed with a 1 mL (0.5 cm ⁇ 5 cm) Phenyl 650M resin was attached to an AKTA FPLC system continuously monitoring absorbance at A280.
  • the column was equilibrated with 5 column volumes (CV) of Buffer B (1 M sodium sulfate, 25 mM sodium phosphate, pH 7.0). Product elution was monitored by absorbance at 280 nm.
  • the glycoPEGylation enzymes were individually injected on Phenyl 650M to compare the elutio profiles to Mixture 1 using the following condition: Buffer A: 25 mM Na phosphate, pH 7.0. Buffer B: 1 M Na Sulfate+buffer A, pH 7.0. Elution gradient: 100 ⁇ 0% B over 20 cv. The MBP-SBD-ST3Gal3, MBP-GnT1 and MBP-GalT1 all eluted in the same portion of the gradient. Mixture 1 elutes just prior to the enzymes. Although Mixture 1 and glycoPEGylation enzymes do not completely co-elute, Mixture 1 and the leading portion of the enzyme elution peaks seemed to overlap.
  • the concentration of sodium sulfate required to bind Mixture 1 bind Phenyl 650M was investigated.
  • a Tricorn 5 column was packed with Phenyl 650M resin (1 mL, 0.5 cm 5 cm) as described above and attached to a Varian HPLC system. Each column was equilibrated with 5 column volumes (CV) of one of the buffers listed below. Product elution was monitored by absorbance at 280 nm.
  • EPO-(SA-PEG-10 kDa) 3 was eluted using a gradient of Buffer A (25 mM sodium phosphate, pH 7.0): 100-0% Buffer B over 20 CV (gradient change 5% Buffer B/CV), and then 0% Buffer B for 5 CV. Chromatography operations were performed at a flow rate of 0.64 mL/min (196 cm/hr). EPO-(SA-PEG-10 kDa) 3 peak fractions were stored at 4° C. until analysis by SDS-PAGE.
  • Tricorn 5 column was packed with Phenyl 650S resin (1 mL, 0.5 cm ⁇ 5 cm) as described above and attached to an AKTA FPLC system. Product elution was monitored by absorbance at 280 nm. Three separate purifications were performed each using a different elution gradient. The columns from each experiment were equilibrated with 5 column volumes (CV) of Buffer B (25 mM sodium phosphate, 0.6 M sodium sulfate, pH 7.5).
  • Buffer B 25 mM sodium phosphate, 0.6 M sodium sulfate, pH 7.5.
  • Gradient 3 100-60% Buffer B over 2 CV, hold for 1 CV, 60-20% Buffer B over 20 CV (gradient change 2% Buffer B/CV), 20-0% Buffer B over 2 CV and then 0% Buffer B for 5 CV. Chromatography operations were performed at a flow rate of 0.64 mL/min. Chromatography elution profiles were compared and selected fractions were analyzed by SDS PAGE. Out of the three gradient profiles tested, Gradient 3 provided the best separation of the EPO-PEG species.
  • the EPO-PEG x mixture (0.8 mg EPO protein conjugate; 1 mL) solution was adjusted to a sodium sulfate concentration of 0.6 M by addition of 0.4 mL of Buffer A (25 mM sodium phosphate, selected pH) and 0.6 mL of buffer (2 M sodium sulfate, 25 mM sodium phosphate, selected pH). This solution was then diluted with an equal volume of Buffer B and the entire sample (4 mL) was injected onto the column. The column was washed with 5 CV of Buffer B and the product eluted using a gradient using Buffer A.
  • Buffer A 25 mM sodium phosphate, selected pH
  • buffer 2 M sodium sulfate, 25 mM sodium phosphate, selected pH
  • the step yield was calculated as the ratio of EPO-PEG protein recovered after HIC chromatography (combined fractions) versus the EPO-PEG injected onto the column 4
  • Standard purification conditions (Buffer A: 25 mM sodium phosphate, pH 7.5.
  • Buffer B 25 mM sodium phosphate, 0.6M sodium sulfate, pH 7.5 (with 0.05 mg EPO-(SA-PEG-10 kDa) 1-4 /mL resin loaded) were compared to purification runs with buffer A and B prepared at pH 6.5, 7.0 and 8.0.
  • the elution peaks (A280) are shifted slightly to the left and right of the standard elution condition.
  • the X-axis of these A280 traces were shifted to overlay the peaks.
  • Comparison of the X-axis shifted peaks shows similar peak traces with respect to the elution of EPO-(SA-PEG-10 kDa) 2 , EPO-(SA-PEG-10 kDa) 3 , and EPO-(SA-PEG-10 kDa) 4 . This is especially true for pH 7.0, 7.5 and 8.0. The pH 6.5 run had a slightly lower peak max and a little less resolution was noted between the EPO-(SA-PEG-10 kDa) 3 and EPO-(SA-PEG-10 kDa) 4 peaks.
  • EPO-(SA-PEG-10 kDa) 4 Integration of EPO-(SA-PEG-10 kDa) 4 is difficult at low levels but is detected by silver stain SDS-PAGE. No aggregation of EPO-(SA-PEG-10 kDa) 3 is detected by SEC analysis. SDS-PAGE analysis shows that the purity of each EPO-(SA-PEG-10 kDa) 3 pool is similar. However, the pH 6.5 purified material contained a higher amount of a approximate 50 kDa proteolyses band. A minor amount of this proteolysis product was also detected for the pH 7.0 purified material.
  • the EPO-PEG x mixture (0.8 mg EPO protein; 1 mL) solution was adjusted to a sodium sulfate concentration of 0.6 M by addition of 0.4 mL of Buffer A (various concentrations of sodium phosphate, pH 7.5) and 0.6 mL of buffer (2 M sodium sulfate, various concentrations of sodium phosphate, pH 7.5). This solution was then diluted with an equal volume of Buffer B and the entire sample (4 mL) was injected onto the column. The column was washed with 5 CV of Buffer B and the product eluted using a gradient using Buffer A.
  • the step yield was calculated as the ratio of EPO-PEG protein recovered after HIC chromatography (combined fractions) versus the amount EPO-PEG injected onto the column 4
  • Standard purification conditions (Buffer A: 25 mM sodium phosphate, pH 7.5.
  • Buffer B 25 mM sodium phosphate, 0.6M sodium sulfate, pH 7.5 (0.05 mg EPO-(SA-PEG-10 kDa) 1-4 /mL resin loaded) were compared to purification runs with buffer A and B prepared with 15 and 50 mM sodium phosphate. Resulting A280 elution profiles were compared. The 15 mM Na phosphate eluted material elutes earliest, followed by the 25 mM Na phosphate and finally 50 mM Na phosphate. The shift in elution profile is due to the relative changes in buffer conductivity (see table 3).
  • Table 3 compares EPO-(SA-PEG-10 kDa) 3 recovery, MBP-ELISA, RP-HPLC, and SEC analysis data. No significant difference was observed between the EPO-(SA-PEG-10 kDa) 3 purified within this 15 to 50 mM sodium phosphate concentration range. Recovery is between 60.2 and 61.9%.
  • MBP-ELISA indicates ⁇ 0.083-0.085 mcg MBP/mg EPO-(SA-PEG-10 kDa) 3 remaining RP-HPLC analysis showed that the amount of EPO-(SA-PEG-10 kDa) 2 remaining in the EPO-(SA-PEG-10 kDa) 3 peak is between 3.7 and 4.5%.
  • the EPO-PEG x mixture (0.8 mg EPO protein; 1 mL) solution was adjusted to a sodium sulfate concentration by the addition of 0.3-0.5 mL of Buffer A (25 mM sodium phosphate, pH 7.5) and 0.5-0.7 mL of buffer (2 M sodium sulfate, 25 mM sodium phosphate, pH 7.5) depending on the sodium sulfate concentration used in the experiment.
  • Buffer A 25 mM sodium phosphate, pH 7.5
  • buffer 2 M sodium sulfate, 25 mM sodium phosphate, pH 7.5
  • the step yield was calculated as the ratio of EPO-PEG protein recovered after HIC chromatography (combined fractions) versus the amount of EPO-PEG injected onto the column. 4 The percent of EPO-(SA-PEG-10 kDa) 3 in the main product peak. 5 The percent of EPO-(SA-PEG-10 kDa) 2 in the main product peak. 6 The amount of MBP protein in the main product peak as determine by the ELISA versus the amount of EPO-PEG protein. 7 The amount of aggregate EPO-PEG in the product peak observed by SEC.
  • Standard purification conditions (Buffer A: 25 mM sodium phosphate, pH 7.5. Buffer B: 25 mM sodium phosphate, 0.6M sodium sulfate, pH 7.5. With 0.05 mg EPO-(SA-PEG-10 kDa) 1-4 /mL resin loaded) were compared to purification runs with buffer B prepared with 0.5 and 0.7 M sodium sodium sulfate. Elution profiles (A280) were compared. The run with buffer B containing 0.5 M Na sulfate elutes earliest, followed by the 0.6 M Na sulfate and finally 0.7 M Na sulfate run. The X-axis of these A280 traces were shifted to overlay the peaks.
  • Table 4 compares EPO-(SA-PEG-10 kDa) 3 recovery, MBP-ELISA, RP-HPLC, and SEC analysis data. No significant difference was noted between the EPO-(SA-PEG-10 kDa) 3 purified within the 0.5 to 0.7 M sodium sulfate concentration range. Recovery is between 61.5 and 64.2%.
  • ELISA indicates ⁇ 0.04-0.083 mcg MBP/mg EPO-(SA-PEG-10 kDa) 3 remaining RP-HPLC analysis showed that the amount of EPO-(SA-PEG-10 kDa) 2 remaining in the EPO-(SA-PEG-10 kDa) 3 peak is between 3.4 and 4.6%.
  • the EPO-PEG x mixture was adjusted to a sodium sulfate concentration of 0.6 M and a final volume of 4 mL. This solution was injected onto the column. The column was washed with 5 CV of Buffer B and the product eluted using a gradient using Buffer A. A gradient of 100-60% Buffer B over 2 CV, hold for 1 CV, 60-35% Buffer B over 13 CV, hold for 1 CV and then 0% Buffer B for 5 CV. The chromatography flow rate was 2.0 mL/min (150 cm/hr) and the column elution monitored by the absorbance at 280 nm. The chromatography elution profiles were compared. Fractions were pooled and analyzed by SDS-PAGE. Analysis of the main product peak is summarized in Table 5, below:
  • Comparison of the resulting elution profiles shows a decrease in resolution between the EPO-(SA-PEG-10 kDa) 2 and EPO-(SA-PEG-10 kDa) 3 peaks as the amount of protein loaded onto the column is increased.
  • RP-HPLC analysis was used to compare the amount of EPO-(SA-PEG-10 kDa) 2 remaining in the EPO-(SA-PEG-10 kDa) 3 pooled peak.
  • Table 5 compares EPO-(SA-PEG-10 kDa) 3 recovery, MBP-ELISA, RP-HPLC, and SEC analysis data. No significant difference was noted between the EPO-(SA-PEG-10 kDa) 3 purified within the investigated column loading range.
  • EPO-(SA-PEG-10 kDa) 2 in the EPO-(SA-PEG-10 kDa) 3 peak since the peaks were pooled in the same manner. Integration of the EPO-(SA-PEG-10 kDa) 4 was difficult at low levels but the glycoform was detected by silver stain SDS-PAGE. No aggregation of EPO-(SA-PEG-10 kDa) 3 was detected by SEC analysis.
  • Phenyl 650M and Phenyl 650S chromatography resins were examined.
  • a Tricorn 10 column was packed with 15.7 mL (1.0 cm id ⁇ 20 cm) of either Phenyl 650S or Phenyl 650M.
  • Each column was equilibrated with 5 column volumes (CV) of buffer B (25 mM sodium phosphate, 0.6 M sodium sulfate, pH 7.5).
  • the EPO-PEG x (0.8 mg EPO protein; 1 mL) solution was adjusted to a sodium sulfate concentration of 0.6 M by the addition of 0.4 mL of Buffer A (25 mM sodium phosphate, pH 7.5) and 0.6 mL of buffer (2 M sodium sulfate, 25 mM sodium phosphate, pH 7.5). This solution was then diluted with an equal volume of Buffer B and the entire sample (4 mL) was injected onto the column. The column was washed with 5 CV of Buffer B and the product eluted using a gradient using Buffer A.
  • the step yield was calculated as the ratio of EPO-PEG protein recovered after HIC chromatography (combined fractions) versus the amount of EPO-PEG injected onto the column. 4 The percent of EPO-(SA-PEG-10 kDa) 3 in the main product peak. 5 The percent of EPO-(SA-PEG-10 kDa) 2 in the main product peak. 6 The amount of MBP protein contained in the main product peak as determine by ELISA versus the amount of EPO-PEG protein. 7 The amount of aggregate EPO-PEG in the product peak observed by SEC.
  • Phenyl 650S 35 micron bead size
  • Phenyl 650M 65 micron bead size
  • Buffer A 25 mM sodium phosphate, pH 7.5.
  • Buffer B 25 mM sodium phosphate, 0.6M sodium sulfate, pH 7.5 (0.05 mg EPO-(SA-PEG-10 kDa) 1-4 /mL resin loaded).
  • Comparison of the resulting elution profiles showed that Phenyl 650S has a better resolution between EPO-(SA-PEG-10 kDa) 2 and EPO-(SA-PEG-10 kDa) 3 .
  • the total system and column pressure measurements show the average pressure throughout the Phenyl 650M purification was approximately 0.12 mPa, while the Phenyl 650S pressure was approximately 0.29 mPa.
  • EPO-(SA-PEG-10 kDa) 3 purification using Phenyl Sepharose HP 34 micron beads size
  • Buffer A 25 mM sodium phosphate, pH 7.5.
  • Buffer B 25 mM sodium phosphate, 0.6M sodium sulfate, pH 7.5, with 0.05 mg EPO-(SA-PEG-10 kDa) 1-4 /mL resin loaded.
  • the elution profile showed no actual resolution between EPO-(SA-PEG-10 kDa) i , EPO-(SA-PEG-10 kDa) 2 , EPO-(SA-PEG-10 kDa) 3 and EPO-(SA-PEG-10 kDa) 4 (chromatogram not shown). Since there was no resolution between the various PEG states no analysis was performed on the eluted material.
  • the mixture e.g., flowthrough from an anion chromatography medium
  • HIC hydrophobic interaction chromatography
  • An exemplary HIC procedure is outlined below: An XK26 column was packed with Phenyl 650S resin (106 mL, 2.6 cm ⁇ 20 cm) and attached to an AKTA Explorer 100 system.
  • the EPO-(SA-PEG-10 kDa) 3 was eluted from the column using the following gradient elution procedure using Buffer A (25 mM sodium phosphate, pH 7.5). Gradient: 100-60% Buffer B over 2 CV, hold for 1 CV, 60-35% Buffer B over 13 CV (gradient change 1.92% Buffer B/CV), hold 1 CV and then 0% Buffer B for 5 CV. The equilibration, load and wash steps were performed at a flow rate of 8 mL/min (90 cm/hr).
  • Buffer A 25 mM sodium phosphate, pH 7.5
  • Gradient 100-60% Buffer B over 2 CV, hold for 1 CV, 60-35% Buffer B over 13 CV (gradient change 1.92% Buffer B/CV), hold 1 CV and then 0% Buffer B for 5 CV.
  • the equilibration, load and wash steps were performed at a flow rate of 8 mL/min (90 cm/hr).
  • Phenyl 650S Chromatography was scaled up from a 0.8 mg EPO-(SA-PEG-10 kDa) 1-4 load (15.7 mL column) to a 22.4 mg load (106 mL column). The standard conditions were used except the amount of material loaded on the column was increased from 0.05 to 0.2 mg EPO-(SA-PEG-10 kDa) 1-4 /mL resin. The EPO-(SA-PEG-10 kDa) 3 peak was pooled as shown in FIG. 2B resulting in a 408 mL peak pool (3.85 CV).
  • EPO-(SA-PEG-10 kDa) 3 Prior to concentration 16.1 mg EPO-(SA-PEG-10 kDa) 3 was recovered (72%) with 5.7% EPO-(SA-PEG-10 kDa) 2 remaining from the original 14.9% EPO-(SA-PEG-10 kDa) 2 in the load material. Concentration and diafiltration into 10 mM sodium acetate, 150 mM NaCl, pH 5.4 using a Pellicon-2 XL 50 cm 2 yielded a 95% recovery.
  • the purification process began with a Sartobind Q membrane used in a negative binding mode which allowed the PEG-EPO conjugates to flow through while capturing glycoPEGylation enzymes, such as MBP-GnT1, MBP-GalT1, MBP-SBD-ST3Gal3 and other enzyme contaminants.
  • glycoPEGylation enzymes such as MBP-GnT1, MBP-GalT1, MBP-SBD-ST3Gal3 and other enzyme contaminants.
  • the various PEG species generated in the glycoPEGylation reaction were then fractionated using HIC on a Phenyl 650S resin (also compare Example 1), which enriched the EPO-(SA-PEG-10 kDa) 3 to a concentration of >96% (step yield approximately 75%).
  • EPO-(SA-PEG-10 kDa) 3 product was subjected to available drug product release tests. The purity was found to be greater than 99% by HPLC (combined glycoforms).
  • the concentration of EPO-(SA-PEG-10 kDa) 3 in the final composition was 96.9%.
  • the concentration of EPO-(SA-PEG-10 kDa) 2 was 2.5% and the concentration of EPO-(SA-PEG-10 kDa) 4 was 0.6%.
  • Other EPO-PEG glycoforms were not detected in the final composition. There was less than 1% aggregate by SEC.
  • Human EPO intermediate protein (produced by Baculovirus fermentation and purified) was stored frozen in 20 mM HEPES, pH 7.5 at ⁇ 20° C. at a concentration of 1.29 mg/mL as determined by BCA assay.
  • MBP-GnT1 was stored frozen in 50 mM Tris, pH 7.0, 138 mM NaCl at ⁇ 20° C. The reported activity assay value of 0.5 U/mL was used.
  • RP-HPLC analysis determined the protein concentration to be 0.3 mg/mL.
  • MBP-GalT1 was stored frozen in 20 mM HEPES, pH 7.5, 200 mM NaCl at ⁇ 20° C.
  • the activity was reported to be 15 U/mL, with a protein concentration of 1.0 mg/mL as determined by RP-HPLC.
  • MBP-SBD-ST3Gal3 was stored frozen in 20 mM HEPES, pH 7.0 at ⁇ 20° C. The reported activity was 2.05 U/mL and the protein concentration was measured to be 1.06 mg/mL by BCA assay.
  • UDP-GlcNAc and UDP-Gal were prepared as 60 mg/mL stock solutions in 100 mM HEPES, pH 7.0, 20 mM NaCl, 0.02% NaN 3 immediately prior to use.
  • CMP-SA-PEG-10 kDa was prepared as a 200 mg/mL stock solution in 100 mM HEPES, pH 7.0, 20 mM NaCl, 0.02% NaN 3 immediately prior to use.
  • EPO intermediate protein (25 mg, 19.4 mL, 1.25 micromoles by BCA assay) was concentrated to a volume of 1.0 mL in a Centricon Plus-20 centrifugal filter (5 kDa MWCO) and then diluted with 15 mL of 100 mM HEPES, pH 7.0, 20 mM NaCl, 0.02% NaN 3 .
  • the EPO solution was concentrated to 0.94 mL (26.7 mg/mL).
  • MBP-SBD-ST3Gal3 (3.05 mL, 6.25 U) was diluted with 13 mL of 100 mM HEPES, pH 7.0, 20 mM NaCl, 0.02% NaN 3 in another Centricon Plus-20 centrifugal filter (5 kDa MWCO) and was concentrated to 0.51 mL (12.3 U/mL).
  • UDP-GlcNAc 25 micromoles, 0.27 mL
  • UDP-Gal 25 micromoles, 0.25 mL
  • stock solutions both at 60 mg/mL in 100 mM HEPES, pH 7.0, 20 mM NaCl, 0.02% NaN 3
  • MBP-GnT1 0.5 mL, 0.25 U
  • MBP-GalT1 (0.33 mL, 5 U)
  • the concentrated MBP-SBD-ST3Gal3 (0.51 mL of 12.3 U/mL, 6.25 U, from above
  • CMP-SA-PEG-10 kDa 25 micromole, 1.25 mL of 200 mg/mL solution in 100 mM HEPES, pH 7.0, 20 mM NaCl, 0.02% NaN 3
  • MnCl 2 5 mM, 0.104 mL of 200 mM solution in water
  • the cartridge was flushed with 20 mM HEPES, 1 M NaCl, pH 7.0 (approximately 100 mL), followed by 20 mM HEPES, 20 mM NaCl, pH 7.0 (approximately 100 mL) both at a flow rate of 15 mL/min.
  • the PEG-EPO reaction mixture (approximately 25 mg, 6 mg/mL, 4.2 mL) was diluted with 20 mM HEPES, 20 mM NaCl, pH 7.0 (22.8 mL) to a final volume of 27 mL.
  • the PEG-EPO pool was analyzed by MBP-ELISA for residual enzyme contaminants (Table 8) and then divided into two portions: The majority fraction (62 mL, 15.25 mg) was purified by HIC chromatography on Phenyl 650S resin as described below. A small sample (18 mL, 4.4 mg) was purified by an alternate process including Fluoroapatite chromatography.
  • Bound impurities were eluted from the column with 20 mM HEPES, 1 M NaCl, pH 7.0 (13 mL) at a flow rate of 15 mL/min.
  • An XK26 column was packed with Phenyl 650S resin (106 mL, 2.6 cm ⁇ 20 cm) as described herein above and was attached to an AKTA Explorer 100 system continuously monitoring absorbance at 214, 254 and 280 nm.
  • the column was equilibrated with 5 column volumes (CV) 25 mM sodium phosphate, 0.6M sodium sulfate, pH 7.5 (212 mL).
  • the Sartobind Q PEG-EPO product pool (62 mL, 18 mg, 0.17 mg PEG-EPO/mL resin, pH 6.94, 2.59 mS/cm) was diluted 1:1 with 1.2 M sodium sulfate, 25 mM sodium phosphate pH 7.5 (62 mL) to adjust the sodium sulfate concentration to 0.6 M.
  • the conditioned sample (124 mL) was applied to the column. Unbound material was washed from the column using 5 CV of 25 mM sodium phosphate, 0.6M sodium sulfate, pH 7.5 (212 mL) at a flow rate of 8 mL/min (90 cm/hr).
  • the PEG-EPO species were fractionated and eluted with the following gradient using Buffer A (25 mM sodium phosphate, pH 7.5) and Buffer B (25 mM sodium phosphate, 0.6M sodium sulfate, pH 7.5) at 8 mL/min (90 cm/hr): 100-60% B over 2 CV, isocratic hold at 60% B for 1 CV, 60-35% B over 13 CV (gradient change 1.92% B/CV), isocratic hold at 35% B for 1 CV, 35-0% B over 1 CV and 0% B for 5 CV.
  • the flow through and wash fraction were collected in the bottle from a 1 L Nalgene filter unit and 12 mL elution fractions were collected in 14 mL Falcon tubes.
  • Tricorn 10 column was packed with Source 15S (15.7 mL, 1 cm ⁇ 20 cm) and attached to an AKTA Explorer 100 system continuously monitoring absorbance at 214, 254 and 280 nm.
  • the column was equilibrated with 5 column volumes (CV) 10 mM Na acetate, pH 5.4 (Buffer A).
US12/444,380 2006-10-03 2007-10-03 Methods for the purification of polypeptide conjugates Abandoned US20100075375A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/444,380 US20100075375A1 (en) 2006-10-03 2007-10-03 Methods for the purification of polypeptide conjugates

Applications Claiming Priority (9)

Application Number Priority Date Filing Date Title
US84896906P 2006-10-03 2006-10-03
US86411706P 2006-11-02 2006-11-02
US86805706P 2006-11-30 2006-11-30
US88751707P 2007-01-31 2007-01-31
US95115907P 2007-07-20 2007-07-20
US95500107P 2007-08-09 2007-08-09
US95646807P 2007-08-17 2007-08-17
PCT/US2007/080353 WO2008057683A2 (fr) 2006-10-03 2007-10-03 Méthodes de purification de conjugués de polypeptides
US12/444,380 US20100075375A1 (en) 2006-10-03 2007-10-03 Methods for the purification of polypeptide conjugates

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2007/080353 A-371-Of-International WO2008057683A2 (fr) 2006-10-03 2007-10-03 Méthodes de purification de conjugués de polypeptides

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/418,530 Continuation US20100035299A1 (en) 2006-10-03 2009-04-03 Methods for the purification of polypeptide conjugates

Publications (1)

Publication Number Publication Date
US20100075375A1 true US20100075375A1 (en) 2010-03-25

Family

ID=39365174

Family Applications (3)

Application Number Title Priority Date Filing Date
US11/866,969 Expired - Fee Related US8969532B2 (en) 2006-10-03 2007-10-03 Methods for the purification of polypeptide conjugates comprising polyalkylene oxide using hydrophobic interaction chromatography
US12/444,380 Abandoned US20100075375A1 (en) 2006-10-03 2007-10-03 Methods for the purification of polypeptide conjugates
US12/418,530 Abandoned US20100035299A1 (en) 2006-10-03 2009-04-03 Methods for the purification of polypeptide conjugates

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US11/866,969 Expired - Fee Related US8969532B2 (en) 2006-10-03 2007-10-03 Methods for the purification of polypeptide conjugates comprising polyalkylene oxide using hydrophobic interaction chromatography

Family Applications After (1)

Application Number Title Priority Date Filing Date
US12/418,530 Abandoned US20100035299A1 (en) 2006-10-03 2009-04-03 Methods for the purification of polypeptide conjugates

Country Status (5)

Country Link
US (3) US8969532B2 (fr)
EP (1) EP2054521A4 (fr)
JP (1) JP2010505874A (fr)
IL (1) IL197765A0 (fr)
WO (1) WO2008057683A2 (fr)

Cited By (44)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040063911A1 (en) * 2001-10-10 2004-04-01 Neose Technologies, Inc. Protein remodeling methods and proteins/peptides produced by the methods
US20060287224A1 (en) * 2003-11-24 2006-12-21 Neose Technologies, Inc. Glycopegylated erythropoietin formulations
US20070032405A1 (en) * 2003-03-14 2007-02-08 Neose Technologies, Inc. Branched water-soluble polymers and their conjugates
US20070059275A1 (en) * 2003-07-25 2007-03-15 Defrees Shawn Antibody toxin conjugates
US20070254836A1 (en) * 2003-12-03 2007-11-01 Defrees Shawn Glycopegylated Granulocyte Colony Stimulating Factor
US20080050772A1 (en) * 2001-10-10 2008-02-28 Neose Technologies, Inc. Granulocyte colony stimulating factor: remodeling and glycoconjugation of G-CSF
US20080102083A1 (en) * 2003-05-09 2008-05-01 Neose Technologies, Inc. Compositions and Methods for the Preparation of Human Growth Hormone Glycosylation Mutants
US20080146782A1 (en) * 2006-10-04 2008-06-19 Neose Technologies, Inc. Glycerol linked pegylated sugars and glycopeptides
US20080176790A1 (en) * 2004-10-29 2008-07-24 Defrees Shawn Remodeling and Glycopegylation of Fibroblast Growth Factor (Fgf)
US20080242846A1 (en) * 2004-01-08 2008-10-02 Neose Technologies, Inc. O-linked glycosylation of peptides
US20080255040A1 (en) * 2006-07-21 2008-10-16 Neose Technologies, Inc. Glycosylation of peptides via o-linked glycosylation sequences
US20080300175A1 (en) * 2003-11-24 2008-12-04 Neose Technologies, Inc. Glycopegylated erythropoietin
US20080305992A1 (en) * 2003-11-24 2008-12-11 Neose Technologies, Inc. Glycopegylated erythropoietin
US20090000924A1 (en) * 2007-06-29 2009-01-01 Harley-Davidson Motor Company Group, Inc. Integrated ignition and key switch
US20090028822A1 (en) * 2004-09-10 2009-01-29 Neose Technologies, Inc. Glycopegylated Interferon Alpha
US20090048440A1 (en) * 2005-11-03 2009-02-19 Neose Technologies, Inc. Nucleotide Sugar Purification Using Membranes
US20090143292A1 (en) * 2007-08-29 2009-06-04 Neose Technologies, Inc. Liquid Formulation of G-CSF Conjugate
US20090203579A1 (en) * 2005-01-10 2009-08-13 Defrees Shawn Glycopegylated Granulocyte Colony Stimulating Factor
US20090227504A1 (en) * 2002-06-21 2009-09-10 Novo Nordisk A/S Pegylated Factor VII Glycoforms
US20090292110A1 (en) * 2004-07-23 2009-11-26 Defrees Shawn Enzymatic modification of glycopeptides
US20100009902A1 (en) * 2005-01-06 2010-01-14 Neose Technologies, Inc. Glycoconjugation Using Saccharyl Fragments
US20100120666A1 (en) * 2007-04-03 2010-05-13 Biogenerix Ag Methods of treatment using glycopegylated g-csf
US20100261872A1 (en) * 2001-10-10 2010-10-14 Neose Technologies, Inc. Factor VIII: remodeling and glycoconjugation of factor VIII
US20100286067A1 (en) * 2008-01-08 2010-11-11 Biogenerix Ag Glycoconjugation of polypeptides using oligosaccharyltransferases
US20110003744A1 (en) * 2005-05-25 2011-01-06 Novo Nordisk A/S Glycopegylated Erythropoietin Formulations
US7956032B2 (en) 2003-12-03 2011-06-07 Novo Nordisk A/S Glycopegylated granulocyte colony stimulating factor
US20110166332A1 (en) * 2008-09-12 2011-07-07 Ge Healthcare Bio-Sciences Ab Enhanced antibody aggregate removal with capto adhere in the presence of protein-excluded zwitterions
US20110177029A1 (en) * 2007-06-04 2011-07-21 Novo Nordisk A/S O-linked glycosylation using n-acetylglucosaminyl transferases
US8008252B2 (en) 2001-10-10 2011-08-30 Novo Nordisk A/S Factor VII: remodeling and glycoconjugation of Factor VII
US8063015B2 (en) 2003-04-09 2011-11-22 Novo Nordisk A/S Glycopegylation methods and proteins/peptides produced by the methods
US8404809B2 (en) 2005-05-25 2013-03-26 Novo Nordisk A/S Glycopegylated factor IX
US8632770B2 (en) 2003-12-03 2014-01-21 Novo Nordisk A/S Glycopegylated factor IX
US8633157B2 (en) 2003-11-24 2014-01-21 Novo Nordisk A/S Glycopegylated erythropoietin
US8716240B2 (en) 2001-10-10 2014-05-06 Novo Nordisk A/S Erythropoietin: remodeling and glycoconjugation of erythropoietin
US8791066B2 (en) 2004-07-13 2014-07-29 Novo Nordisk A/S Branched PEG remodeling and glycosylation of glucagon-like peptide-1 [GLP-1]
US8791070B2 (en) 2003-04-09 2014-07-29 Novo Nordisk A/S Glycopegylated factor IX
US8911967B2 (en) 2005-08-19 2014-12-16 Novo Nordisk A/S One pot desialylation and glycopegylation of therapeutic peptides
US8969532B2 (en) 2006-10-03 2015-03-03 Novo Nordisk A/S Methods for the purification of polypeptide conjugates comprising polyalkylene oxide using hydrophobic interaction chromatography
US9150848B2 (en) 2008-02-27 2015-10-06 Novo Nordisk A/S Conjugated factor VIII molecules
US9169331B2 (en) 2012-12-21 2015-10-27 Dionex Corporation Separation of glycans by mixed-mode liquid chromatography
US9187546B2 (en) 2005-04-08 2015-11-17 Novo Nordisk A/S Compositions and methods for the preparation of protease resistant human growth hormone glycosylation mutants
US9310344B2 (en) 2013-06-14 2016-04-12 Dionex Corporation HILIC/anion-exchange/cation-exchange multimodal media
US9488625B2 (en) 2010-12-15 2016-11-08 Baxalta GmbH Purification of factor VIII using a conductivity gradient
US9493499B2 (en) 2007-06-12 2016-11-15 Novo Nordisk A/S Process for the production of purified cytidinemonophosphate-sialic acid-polyalkylene oxide (CMP-SA-PEG) as modified nucleotide sugars via anion exchange chromatography

Families Citing this family (53)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN100387713C (zh) 2000-08-11 2008-05-14 麒麟医药株式会社 调节磷酸代谢,钙代谢,钙化和维生素d代谢的多肽及其编码dna
DE602005023429D1 (de) 2004-10-19 2010-10-21 Lonza Ag Verfahren zur festphasen-peptidsynthese
ES2533992T3 (es) 2005-08-24 2015-04-16 Immunogen, Inc. Procedimiento para preparar conjugados de anticuerpo maitansinoide
CA2670618C (fr) 2006-12-15 2016-10-04 Baxter International Inc. Conjugue du facteur viia et de l'acide (poly)sialique presentant une demi-vie in vivo prolongee
US7883705B2 (en) 2007-02-14 2011-02-08 Kyowa Hakko Kirin Co., Ltd. Anti FGF23 antibody and a pharmaceutical composition comprising the same
FR2920429B1 (fr) * 2007-08-30 2012-10-05 Lfb Biotechnologies Procede de purification du facteur viii et du facteur von willebrand
JOP20190083A1 (ar) 2008-06-04 2017-06-16 Amgen Inc بولي ببتيدات اندماجية طافرة لـfgf21 واستخداماتها
PT2848625T (pt) * 2008-08-14 2019-10-25 Genentech Inc Métodos para remover um contaminante com a utilização de cromatografia de membrana de permuta iónica de deslocação de proteína indígena.
US9279013B2 (en) 2008-10-10 2016-03-08 Amgen Inc. FGF-21 mutants comprising polyethylene glycol and uses thereof
US9150894B2 (en) * 2009-02-19 2015-10-06 Xellia Pharmaceuticals Aps Process for purifying lipopeptides
US20120052069A1 (en) 2009-05-05 2012-03-01 Amgen Inc Fgf21 mutants and uses thereof
CN107188950B (zh) 2009-05-05 2021-09-28 安姆根有限公司 Fgf21突变体及其用途
KR102444399B1 (ko) 2009-06-03 2022-09-16 이뮤노젠 아이엔씨 메이탄시노이드의 제조방법
CA2764835A1 (fr) * 2009-06-17 2010-12-23 Amgen Inc. Polypeptides fgf19 chimeriques et leurs utilisations
US8642737B2 (en) 2010-07-26 2014-02-04 Baxter International Inc. Nucleophilic catalysts for oxime linkage
ES2597954T3 (es) 2009-07-27 2017-01-24 Baxalta GmbH Conjugados de proteína de la coagulación sanguínea
NZ623810A (en) 2009-07-27 2015-10-30 Lipoxen Technologies Ltd Glycopolysialylation of non-blood coagulation proteins
KR101912335B1 (ko) 2009-07-27 2018-10-26 리폭센 테크놀로지즈 리미티드 비혈액 응고 단백질의 글리코폴리시알화
US8809501B2 (en) 2009-07-27 2014-08-19 Baxter International Inc. Nucleophilic catalysts for oxime linkage
TWI532495B (zh) * 2009-10-05 2016-05-11 菲瑞茵國際中心股份有限公司 藥學製劑
US8372952B2 (en) * 2009-12-02 2013-02-12 Amgen Inc. Binding proteins that bind to human FGFR1C, human β-klotho and both human FGFR1C and human β-klotho
UA109888C2 (uk) 2009-12-07 2015-10-26 ІЗОЛЬОВАНЕ АНТИТІЛО АБО ЙОГО ФРАГМЕНТ, ЩО ЗВ'ЯЗУЄТЬСЯ З β-КЛОТО, РЕЦЕПТОРАМИ FGF І ЇХНІМИ КОМПЛЕКСАМИ
EP2558497A2 (fr) 2010-04-15 2013-02-20 Amgen Inc. Protéines de liaison au récepteur de fgf humain et à beta-klotho
ES2534936T3 (es) * 2010-08-02 2015-04-30 Ratiopharm Gmbh Método para producir y purificar una sialiltransferasa soluble activa
BR112013015898A2 (pt) 2010-12-22 2018-06-26 Baxter International Inc. derivado de ácido graxo solúvel em água, e, métodos para preparar um derivado de ácido graxo e uma proteína terapêutica conjugada.
UA116524C2 (uk) 2011-03-29 2018-04-10 Іммуноджен, Інк. Спосіб одержання кон'югата антитіло-майтансиноїд
DE102011101995A1 (de) 2011-05-19 2012-11-22 Sartorius Stedim Biotech Gmbh Verfahren zur Auftrennung eines Gemisches aus einem Protein und seinem Reaktionsprodukt mit einem Polyalkylenglykol
AU2012283858C1 (en) 2011-07-20 2020-01-23 Zepteon, Incorporated Polypeptide separation methods
NZ626945A (en) 2012-01-12 2016-10-28 Biogen Ma Inc Chimeric factor viii polypeptides and uses thereof
CN111548418A (zh) 2012-02-15 2020-08-18 比奥贝拉蒂治疗公司 因子viii组合物及其制备和使用方法
KR20190094480A (ko) 2012-02-15 2019-08-13 바이오버라티브 테라퓨틱스 인크. 재조합 인자 viii 단백질
PT2882450T (pt) * 2012-07-11 2020-02-19 Bioverativ Therapeutics Inc Complexo de fator viii com xten e a proteína fator de von willebrand, e utilizações do mesmo
RU2661083C2 (ru) 2012-10-04 2018-07-11 Иммуноджен, Инк. Использование пвдф-мембраны для очистки конъюгатов клеточно-связывающий агент - цитотоксический агент
WO2014129876A1 (fr) * 2013-02-25 2014-08-28 에스케이케미칼주식회사 Procédé d'isolement et de purification de protéine de fusion comprenant le facteur vii
US11229789B2 (en) 2013-05-30 2022-01-25 Neurostim Oab, Inc. Neuro activator with controller
PL3003473T3 (pl) 2013-05-30 2019-05-31 Neurostim Solutions LLC Miejscowa stymulacja neurologiczna
EP3013365B1 (fr) * 2013-06-26 2019-06-05 Academia Sinica Antigènes rm2 et leur utilisation
TW202003554A (zh) 2013-08-14 2020-01-16 美商百歐維拉提夫治療公司 因子viii-xten融合物及其用途
SI3091997T1 (sl) 2014-01-10 2022-10-28 Bioverativ Therapeutics Inc. Himerni proteini faktorja VIII in njihova uporaba
US11077301B2 (en) 2015-02-21 2021-08-03 NeurostimOAB, Inc. Topical nerve stimulator and sensor for bladder control
CN108348604B (zh) * 2015-09-08 2022-04-29 沃特世科技公司 用于分析抗体-药物缀合物的多维色谱方法
SG11201900201YA (en) 2016-08-16 2019-02-27 Regeneron Pharma Methods for quantitating individual antibodies from a mixture
SG11201902667UA (en) 2016-10-25 2019-05-30 Regeneron Pharma Methods and systems for chromatography data analysis
WO2018172219A1 (fr) 2017-03-20 2018-09-27 F. Hoffmann-La Roche Ag Procédé de glyco-ingénierie in vitro d'une protéine stimulant l'érythropoïèse
EP3706856A4 (fr) 2017-11-07 2021-08-18 Neurostim Oab, Inc. Activateur de nerf non invasif à circuit adaptatif
ES2905105T3 (es) * 2017-12-29 2022-04-07 Hoffmann La Roche Procedimiento para proporcionar una composición de proteína PEGilada
US20200323993A1 (en) * 2017-12-29 2020-10-15 Hoffmann-La Roche Inc. Process for providing pegylated protein composition
SG11202005952TA (en) * 2017-12-29 2020-07-29 Hoffmann La Roche Process for providing pegylated protein composition
TW202005694A (zh) 2018-07-02 2020-02-01 美商里珍納龍藥品有限公司 自混合物製備多肽之系統及方法
EP3990100A4 (fr) 2019-06-26 2023-07-19 Neurostim Technologies LLC Activateur de nerf non invasif à circuit adaptatif
US20220251137A1 (en) * 2019-07-09 2022-08-11 Jennifer Renee HOPP Temperature-controlled purification of granulocyte-colony stimulating factor
JP2023506713A (ja) 2019-12-16 2023-02-20 ニューロスティム テクノロジーズ エルエルシー 昇圧電荷送達を用いた非侵襲性神経アクティベータ
KR20230125179A (ko) * 2020-12-23 2023-08-29 재즈 파마슈티칼즈 아일랜드 리미티드 전하-차폐 융합 단백질의 정제 방법

Citations (92)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4088538A (en) * 1975-05-30 1978-05-09 Battelle Memorial Institute Reversibly precipitable immobilized enzyme complex and a method for its use
US4385260A (en) * 1975-09-09 1983-05-24 Beckman Instruments, Inc. Bargraph display
US4438253A (en) * 1982-11-12 1984-03-20 American Cyanamid Company Poly(glycolic acid)/poly(alkylene glycol) block copolymers and method of manufacturing the same
US4496689A (en) * 1983-12-27 1985-01-29 Miles Laboratories, Inc. Covalently attached complex of alpha-1-proteinase inhibitor with a water soluble polymer
US4565653A (en) * 1984-03-30 1986-01-21 Pfizer Inc. Acyltripeptide immunostimulants
US4806595A (en) * 1985-08-12 1989-02-21 Koken Co., Ltd. Method of preparing antithrombogenic medical materials
US4826945A (en) * 1987-06-09 1989-05-02 Yissum Research Development Company Biodegradable polymeric materials based on polyether glycols, processes for the preparation thereof and surgical articles made therefrom
US4925796A (en) * 1986-03-07 1990-05-15 Massachusetts Institute Of Technology Method for enhancing glycoprotein stability
US5122614A (en) * 1989-04-19 1992-06-16 Enzon, Inc. Active carbonates of polyalkylene oxides for modification of polypeptides
US5182107A (en) * 1989-09-07 1993-01-26 Alkermes, Inc. Transferrin receptor specific antibody-neuropharmaceutical or diagnostic agent conjugates
US5194376A (en) * 1989-02-28 1993-03-16 University Of Ottawa Baculovirus expression system capable of producing foreign gene proteins at high levels
US5202413A (en) * 1993-02-16 1993-04-13 E. I. Du Pont De Nemours And Company Alternating (ABA)N polylactide block copolymers
US5219564A (en) * 1990-07-06 1993-06-15 Enzon, Inc. Poly(alkylene oxide) amino acid copolymers and drug carriers and charged copolymers based thereon
US5281698A (en) * 1991-07-23 1994-01-25 Cetus Oncology Corporation Preparation of an activated polymer ester for protein conjugation
US5324844A (en) * 1989-04-19 1994-06-28 Enzon, Inc. Active carbonates of polyalkylene oxides for modification of polypeptides
US5405753A (en) * 1990-03-26 1995-04-11 Brossmer; Reinhard CMP-activated, fluorescing sialic acids, as well as processes for their preparation
US5410016A (en) * 1990-10-15 1995-04-25 Board Of Regents, The University Of Texas System Photopolymerizable biodegradable hydrogels as tissue contacting materials and controlled-release carriers
US5492841A (en) * 1994-02-18 1996-02-20 E. I. Du Pont De Nemours And Company Quaternary ammonium immunogenic conjugates and immunoassay reagents
US5527527A (en) * 1989-09-07 1996-06-18 Alkermes, Inc. Transferrin receptor specific antibody-neuropharmaceutical agent conjugates
US5529914A (en) * 1990-10-15 1996-06-25 The Board Of Regents The Univeristy Of Texas System Gels for encapsulation of biological materials
US5605793A (en) * 1994-02-17 1997-02-25 Affymax Technologies N.V. Methods for in vitro recombination
US5614184A (en) * 1992-07-28 1997-03-25 New England Deaconess Hospital Recombinant human erythropoietin mutants and therapeutic methods employing them
US5621039A (en) * 1993-06-08 1997-04-15 Hallahan; Terrence W. Factor IX- polymeric conjugates
US5629384A (en) * 1994-05-17 1997-05-13 Consiglio Nazionale Delle Ricerche Polymers of N-acryloylmorpholine activated at one end and conjugates with bioactive materials and surfaces
US5635603A (en) * 1993-12-08 1997-06-03 Immunomedics, Inc. Preparation and use of immunoconjugates
US5705367A (en) * 1994-09-26 1998-01-06 The Rockefeller University Glycosyltransferases for biosynthesis of oligosaccharides, and genes encoding them
US5716812A (en) * 1995-12-12 1998-02-10 The University Of British Columbia Methods and compositions for synthesis of oligosaccharides, and the products formed thereby
US5728554A (en) * 1995-04-11 1998-03-17 Cytel Corporation Enzymatic synthesis of glycosidic linkages
US5770420A (en) * 1995-09-08 1998-06-23 The Regents Of The University Of Michigan Methods and products for the synthesis of oligosaccharide structures on glycoproteins, glycolipids, or as free molecules, and for the isolation of cloned genetic sequences that determine these structures
US5858751A (en) * 1992-03-09 1999-01-12 The Regents Of The University Of California Compositions and methods for producing sialyltransferases
US5876980A (en) * 1995-04-11 1999-03-02 Cytel Corporation Enzymatic synthesis of oligosaccharides
US6010999A (en) * 1990-05-04 2000-01-04 American Cyanamid Company Stabilization of fibroblast growth factors by modification of cysteine residues
US6015555A (en) * 1995-05-19 2000-01-18 Alkermes, Inc. Transferrin receptor specific antibody-neuropharmaceutical or diagnostic agent conjugates
US6030815A (en) * 1995-04-11 2000-02-29 Neose Technologies, Inc. Enzymatic synthesis of oligosaccharides
US6037452A (en) * 1992-04-10 2000-03-14 Alpha Therapeutic Corporation Poly(alkylene oxide)-Factor VIII or Factor IX conjugate
US6057292A (en) * 1995-09-21 2000-05-02 Genentech, Inc. Method for inhibiting growth hormone action
US6183738B1 (en) * 1997-05-12 2001-02-06 Phoenix Pharamacologics, Inc. Modified arginine deiminase
US20020016003A1 (en) * 2000-03-16 2002-02-07 Eliana Saxon Chemoselective ligation
US6348558B1 (en) * 1999-12-10 2002-02-19 Shearwater Corporation Hydrolytically degradable polymers and hydrogels made therefrom
US6362254B2 (en) * 1998-03-12 2002-03-26 Shearwater Corporation Poly(ethylene glycol) derivatives with proximal reactive groups
US20020037841A1 (en) * 2000-05-15 2002-03-28 Apollon Papadimitriou Erythropoietin composition
US6376604B2 (en) * 1999-12-22 2002-04-23 Shearwater Corporation Method for the preparation of 1-benzotriazolylcarbonate esters of poly(ethylene glycol)
US6399336B1 (en) * 1997-01-16 2002-06-04 Neose Technologies, Inc. Practical in vitro sialylation of recombinant glycoproteins
US6399337B1 (en) * 1997-06-06 2002-06-04 The Governors Of The University Of Alberta α1,3-fucosyltransferase
US20030027257A1 (en) * 1997-08-21 2003-02-06 University Technologies International, Inc. Sequences for improving the efficiency of secretion of non-secreted protein from mammalian and insect cells
US20030040037A1 (en) * 2000-05-12 2003-02-27 Neose Technologies, Inc. In vitro modification of glycosylation patterns of recombinant glycopeptides
US6531121B2 (en) * 2000-12-29 2003-03-11 The Kenneth S. Warren Institute, Inc. Protection and enhancement of erythropoietin-responsive cells, tissues and organs
US6555346B1 (en) * 1997-12-18 2003-04-29 Stichting Instituut Voor Dierhouderij En Diergezondheid Protein expression in baculovirus vector expression systems
US6692931B1 (en) * 1998-11-16 2004-02-17 Werner Reutter Recombinant glycoproteins, method for the production thereof, medicaments containing said glycoproteins and use thereof
US20040043446A1 (en) * 2001-10-19 2004-03-04 Neose Technologies, Inc. Alpha galalctosidase a: remodeling and glycoconjugation of alpha galactosidase A
US20040063911A1 (en) * 2001-10-10 2004-04-01 Neose Technologies, Inc. Protein remodeling methods and proteins/peptides produced by the methods
US6716626B1 (en) * 1999-11-18 2004-04-06 Chiron Corporation Human FGF-21 nucleic acids
US20040077836A1 (en) * 2001-10-10 2004-04-22 Neose Technologies, Inc. Granulocyte colony stimulating factor: remodeling and glycoconjugation of G-CSF
US20040082026A1 (en) * 2001-10-10 2004-04-29 Neose Technologies, Inc. Interferon alpha: remodeling and glycoconjugation of interferon alpha
US20040102607A1 (en) * 1998-03-25 2004-05-27 Danishefsky Samuel J. Trimeric antigenic O-linked glycopeptide conjugates, methods of preparation and uses thereof
US20040115168A1 (en) * 2001-10-10 2004-06-17 Neose Technologies, Inc. Interferon beta: remodeling and glycoconjugation of interferon beta
US20050026266A1 (en) * 2002-11-08 2005-02-03 Glycozym Aps Methods to identify agents modulating functions of polypeptide galnac-transferases, pharmaceutical compositions comprising such agents and the use of such agents for preparing medicaments
US20050031584A1 (en) * 2001-10-10 2005-02-10 Neose Technologies, Inc. Interleukin-2:remodeling and glycoconjugation of interleukin-2
US20050064540A1 (en) * 2002-11-27 2005-03-24 Defrees Shawn Ph.D Glycoprotein remodeling using endoglycanases
US20050100982A1 (en) * 2001-10-10 2005-05-12 Neose Technologies, Inc. Factor IX: remodeling and glycoconjugation of factor IX
US20050106658A1 (en) * 2001-10-10 2005-05-19 Defrees Shawn Remodeling and glycoconjugation of peptides
US20060024286A1 (en) * 2004-08-02 2006-02-02 Paul Glidden Variants of tRNA synthetase fragments and uses thereof
US20060030521A1 (en) * 2001-11-28 2006-02-09 Neose Technologies, Inc. Remodeling and glycoconjugation of peptides
US20060035224A1 (en) * 2002-03-21 2006-02-16 Johansen Jack T Purification methods for oligonucleotides and their analogs
US20060111279A1 (en) * 2003-11-24 2006-05-25 Neose Technologies, Inc. Glycopegylated erythropoietin
US7157277B2 (en) * 2001-11-28 2007-01-02 Neose Technologies, Inc. Factor VIII remodeling and glycoconjugation of Factor VIII
US20070027068A1 (en) * 2001-10-10 2007-02-01 Defrees Shawn Erythropoietin: remodeling and glycoconjugation of erythropoietin
US20070032405A1 (en) * 2003-03-14 2007-02-08 Neose Technologies, Inc. Branched water-soluble polymers and their conjugates
US20070059275A1 (en) * 2003-07-25 2007-03-15 Defrees Shawn Antibody toxin conjugates
US7214660B2 (en) * 2001-10-10 2007-05-08 Neose Technologies, Inc. Erythropoietin: remodeling and glycoconjugation of erythropoietin
US20070105755A1 (en) * 2005-10-26 2007-05-10 Neose Technologies, Inc. One pot desialylation and glycopegylation of therapeutic peptides
US20080015142A1 (en) * 2003-12-03 2008-01-17 Defrees Shawn Glycopegylated Follicle Stimulating Hormone
US7338933B2 (en) * 2004-01-08 2008-03-04 Neose Technologies, Inc. O-linked glycosylation of peptides
US20080102083A1 (en) * 2003-05-09 2008-05-01 Neose Technologies, Inc. Compositions and Methods for the Preparation of Human Growth Hormone Glycosylation Mutants
US7368108B2 (en) * 2001-11-28 2008-05-06 Neose Technologies, Inc. Glycopeptide remodeling using amidases
US20080108557A1 (en) * 2004-09-29 2008-05-08 Novo Nordisk Healthcare A/G Modified Proteins
US20090028822A1 (en) * 2004-09-10 2009-01-29 Neose Technologies, Inc. Glycopegylated Interferon Alpha
US20090048440A1 (en) * 2005-11-03 2009-02-19 Neose Technologies, Inc. Nucleotide Sugar Purification Using Membranes
US20090053167A1 (en) * 2007-05-14 2009-02-26 Neose Technologies, Inc. C-, S- and N-glycosylation of peptides
US20090055942A1 (en) * 2005-09-14 2009-02-26 Novo Nordisk Healthcare A/G Human Coagulation Factor VII Polypeptides
US20090054623A1 (en) * 2004-12-17 2009-02-26 Neose Technologies, Inc. Lipo-Conjugation of Peptides
US20090093399A1 (en) * 2001-10-10 2009-04-09 Neose Technologies, Inc. Glycopegylation methods and proteins/peptides produced by the methods
US20090124544A1 (en) * 2005-04-08 2009-05-14 Neose Technologies ,Inc. A Delaware Corporation Compositions and methods for the preparation of protease resistant human growth hormone glycosylation mutants
US20100009902A1 (en) * 2005-01-06 2010-01-14 Neose Technologies, Inc. Glycoconjugation Using Saccharyl Fragments
US20100015684A1 (en) * 2001-10-10 2010-01-21 Neose Technologies, Inc. Factor vii: remodeling and glycoconjugation of factor vii
US20100028939A1 (en) * 2003-08-08 2010-02-04 Novo Nordisk Healthcare A/G Use of Galactose Oxidase for Selective Chemical Conjugation of Protractor Molecules to Proteins of Therapeutic Interest
US20100035299A1 (en) * 2006-10-03 2010-02-11 Novo Nordisk A/S Methods for the purification of polypeptide conjugates
US20100041872A1 (en) * 2006-10-04 2010-02-18 Defrees Shawn Glycerol linked pegylated sugars and glycopeptides
US20100048456A1 (en) * 2003-04-09 2010-02-25 Novo Nordisk A/S Glycopegylation methods and proteins/peptides produced by the methods
US20100056428A1 (en) * 2006-09-01 2010-03-04 Novo Nordisk Health Care Ag Modified proteins
US20100081791A1 (en) * 2005-05-25 2010-04-01 Novo Nordisk A/S Glycopegylated factor ix
US7691603B2 (en) * 2003-04-09 2010-04-06 Novo Nordisk A/S Intracellular formation of peptide conjugates

Family Cites Families (308)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB1479268A (en) 1973-07-05 1977-07-13 Beecham Group Ltd Pharmaceutical compositions
US4179337A (en) 1973-07-20 1979-12-18 Davis Frank F Non-immunogenic polypeptides
GB1451798A (en) 1973-08-02 1976-10-06 Ici Ltd Prostanoic acid derivatives
US4414147A (en) 1981-04-17 1983-11-08 Massachusetts Institute Of Technology Methods of decreasing the hydrophobicity of fibroblast and other interferons
JPS57206622A (en) 1981-06-10 1982-12-18 Ajinomoto Co Inc Blood substitute
US4451566A (en) * 1981-12-04 1984-05-29 Spencer Donald B Methods and apparatus for enzymatically producing ethanol
ATE37984T1 (de) 1983-03-01 1988-11-15 Crc Ricerca Chim Pharmazeutische zusammensetzung welche 5acetamido-3,5-dideoxy-d-glycero-dgalactononulosamins|ure cytidin monophosphat enthaelt.
DE3308806A1 (de) 1983-03-12 1984-09-13 Basf Ag, 6700 Ludwigshafen Fungizide mittel, substituierte glucopyranosylamine und verfahren zur bekaempfung von pilzen
JPS59172425A (ja) 1983-03-18 1984-09-29 Nippon Chemiphar Co Ltd 新規な血液凝固因子誘導体およびその製造法ならびにそれを含有する血液凝固促進剤
US4879236A (en) 1984-05-16 1989-11-07 The Texas A&M University System Method for producing a recombinant baculovirus expression vector
US4675414A (en) * 1985-03-08 1987-06-23 The United States Of America As Represented By The Secretary Of The Navy Maleimidomethyl-carbonate polyethers
GR860984B (en) 1985-04-17 1986-08-18 Zymogenetics Inc Expression of factor vii and ix activities in mammalian cells
EP0229108B1 (fr) 1985-06-26 1990-12-27 Cetus Corporation Solubilisation de proteines pour des compositions pharmaceutiques utilisant une conjugaison de polymeres
US5206344A (en) * 1985-06-26 1993-04-27 Cetus Oncology Corporation Interleukin-2 muteins and polymer conjugation thereof
SE451849B (sv) * 1985-12-11 1987-11-02 Svenska Sockerfabriks Ab Sett att syntetisera glykosidiska bindningar samt anvendning av pa detta sett erhallna produkter
IT1213029B (it) 1986-01-30 1989-12-07 Bracco Ind Chimica Spa Chelati di ioni metallici paramagnetici.
US4767702A (en) 1986-02-06 1988-08-30 Cohenford Menashi A Paper strip assay for neisseria species
EP0272253A4 (fr) 1986-03-07 1990-02-05 Massachusetts Inst Technology Procede pour ameliorer la stabilite des glycoproteines.
US5272066A (en) 1986-03-07 1993-12-21 Massachusetts Institute Of Technology Synthetic method for enhancing glycoprotein stability
US4902505A (en) 1986-07-30 1990-02-20 Alkermes Chimeric peptides for neuropeptide delivery through the blood-brain barrier
US5153265A (en) 1988-01-20 1992-10-06 Cetus Corporation Conjugation of polymer to colony stimulating factor-1
US4847325A (en) 1988-01-20 1989-07-11 Cetus Corporation Conjugation of polymer to colony stimulating factor-1
GB8810808D0 (en) 1988-05-06 1988-06-08 Wellcome Found Vectors
US5169933A (en) 1988-08-15 1992-12-08 Neorx Corporation Covalently-linked complexes and methods for enhanced cytotoxicity and imaging
US5874261A (en) 1988-09-02 1999-02-23 The Trustees Of The University Of Pennsylvania Method for the purification of glycosyltransferases
US5218092A (en) 1988-09-29 1993-06-08 Kyowa Hakko Kogyo Co., Ltd. Modified granulocyte-colony stimulating factor polypeptide with added carbohydrate chains
US5104651A (en) 1988-12-16 1992-04-14 Amgen Inc. Stabilized hydrophobic protein formulations of g-csf
US5047335A (en) 1988-12-21 1991-09-10 The Regents Of The University Of Calif. Process for controlling intracellular glycosylation of proteins
US6166183A (en) 1992-11-30 2000-12-26 Kirin-Amgen, Inc. Chemically-modified G-CSF
EP0401384B1 (fr) 1988-12-22 1996-03-13 Kirin-Amgen, Inc. Facteur de stimulation de colonies de granulocytes modifies chimiquement
WO1990007572A1 (fr) 1988-12-23 1990-07-12 Genentech, Inc. DNase HUMAINE
AU634517B2 (en) 1989-01-19 1993-02-25 Pharmacia & Upjohn Company Somatotropin analogs
JP3207416B2 (ja) 1989-01-31 2001-09-10 ファルマシア・アンド・アップジョン・カンパニー ソマトトロピン・アナログ類
US5059535A (en) 1989-04-12 1991-10-22 Chembiomed, Ltd. Process for the separation and purification of sialyl transferases
DE69033192T3 (de) 1989-04-19 2004-05-19 Enzon, Inc. Aktive karbonate von polyalkylenoxyden zur modifizierung von polypeptiden
US5166322A (en) 1989-04-21 1992-11-24 Genetics Institute Cysteine added variants of interleukin-3 and chemical modifications thereof
US5342940A (en) 1989-05-27 1994-08-30 Sumitomo Pharmaceuticals Company, Limited Polyethylene glycol derivatives, process for preparing the same
US5977307A (en) 1989-09-07 1999-11-02 Alkermes, Inc. Transferrin receptor specific ligand-neuropharmaceutical agent fusion proteins
US5672683A (en) 1989-09-07 1997-09-30 Alkermes, Inc. Transferrin neuropharmaceutical agent fusion protein
US5154924A (en) 1989-09-07 1992-10-13 Alkermes, Inc. Transferrin receptor specific antibody-neuropharmaceutical agent conjugates
US5032519A (en) * 1989-10-24 1991-07-16 The Regents Of The Univ. Of California Method for producing secretable glycosyltransferases and other Golgi processing enzymes
US5312808A (en) 1989-11-22 1994-05-17 Enzon, Inc. Fractionation of polyalkylene oxide-conjugated hemoglobin solutions
IL96477A0 (en) * 1989-12-01 1991-08-16 Amgen Inc Megakaryocyte production
SE465222C5 (sv) 1989-12-15 1998-02-10 Pharmacia & Upjohn Ab Ett rekombinant, humant faktor VIII-derivat och förfarande för dess framställning
US5324663A (en) * 1990-02-14 1994-06-28 The Regents Of The University Of Michigan Methods and products for the synthesis of oligosaccharide structures on glycoproteins, glycolipids, or as free molecules, and for the isolation of cloned genetic sequences that determine these structures
US5595900A (en) 1990-02-14 1997-01-21 The Regents Of The University Of Michigan Methods and products for the synthesis of oligosaccharide structures on glycoproteins, glycolipids, or as free molecules, and for the isolation of cloned genetic sequences that determine these structures
WO1991016449A1 (fr) * 1990-04-16 1991-10-31 The Trustees Of The University Of Pennsylvania Compositions saccharides et procedes et appareil servant a les synthetiser
US5583042A (en) 1990-04-16 1996-12-10 Neose Pharmaceuticals, Inc. Apparatus for the synthesis of saccharide compositions
GB9107846D0 (en) 1990-04-30 1991-05-29 Ici Plc Polypeptides
US5399345A (en) * 1990-05-08 1995-03-21 Boehringer Mannheim, Gmbh Muteins of the granulocyte colony stimulating factor
CU22302A1 (es) 1990-09-07 1995-01-31 Cigb Secuencia nucleotidica codificante para una proteina de la membrana externa de neisseria meningitidis y uso de dicha proteina en preparados vacunales
DK0538300T3 (da) 1990-07-10 1994-10-10 Boehringer Ingelheim Int O-Glycosyleret IFN-alfa
DE4028800A1 (de) 1990-09-11 1992-03-12 Behringwerke Ag Gentechnische sialylierung von glykoproteinen
EP0513332A4 (en) * 1990-11-14 1993-03-17 Cargill, Incorporated Conjugates of poly(vinylsaccharide) with proteins for the stabilization of proteins
US5164374A (en) 1990-12-17 1992-11-17 Monsanto Company Use of oligosaccharides for treatment of arthritis
US5861374A (en) * 1991-02-28 1999-01-19 Novo Nordisk A/S Modified Factor VII
US5788965A (en) 1991-02-28 1998-08-04 Novo Nordisk A/S Modified factor VII
US5833982A (en) 1991-02-28 1998-11-10 Zymogenetics, Inc. Modified factor VII
DE69233398D1 (de) 1991-02-28 2004-09-16 Novo Nordisk As Modifizierter faktor-vii
US5278299A (en) * 1991-03-18 1994-01-11 Scripps Clinic And Research Foundation Method and composition for synthesizing sialylated glycosyl compounds
DE69231127D1 (de) 1991-03-18 2000-07-06 Scripps Research Inst La Jolla Oligosaccharide als enzymsubstrate und -inhibitoren: verfahren und zusammensetzungen
CA2101918A1 (fr) 1991-03-18 1992-09-19 Samuel Zalipsky Conjugues de polypeptides et de glycopolypeptides avec des polymeres contenant de l'hydrazine
US5212075A (en) 1991-04-15 1993-05-18 The Regents Of The University Of California Compositions and methods for introducing effectors to pathogens and cells
JP3417558B2 (ja) 1991-05-10 2003-06-16 ジェネンテク,インコーポレイテッド 配位子作用薬および拮抗薬の選択
SE9201544L (sv) 1991-05-31 1992-12-01 Ciba Geigy Ag Saett att framstaella glykosyltransferaser
GB2256197B (en) 1991-05-31 1995-11-22 Ciba Geigy Ag Yeast as host for expression of heterologous glycosyl transferase enzymes
US5352670A (en) 1991-06-10 1994-10-04 Alberta Research Council Methods for the enzymatic synthesis of alpha-sialylated oligosaccharide glycosides
US5374655A (en) 1991-06-10 1994-12-20 Alberta Research Council Methods for the synthesis of monofucosylated oligosaccharides terminating in di-N-acetyllactosaminyl structures
KR950014915B1 (ko) 1991-06-19 1995-12-18 주식회사녹십자 탈시알로당단백-포함화합물
EP0863154A1 (fr) 1991-10-12 1998-09-09 The Regents Of The University Of California Utilisation des protéines redox des groupes thiol pour la réduction des liaisonsdisulfide intramoléculaires, pour augmenter la qualité des produits céréaliers, des pâtes et des produits de four
US6319695B1 (en) 1991-10-15 2001-11-20 The Scripps Research Insitute Production of fucosylated carbohydrates by enzymatic fucosylation synthesis of sugar nucleotides; and in situ regeneration of GDP-fucose
ATE168014T1 (de) 1991-11-08 1998-07-15 Somatogen Inc Hämoglobine als arzneimittelabgabesystem
US5384249A (en) * 1991-12-17 1995-01-24 Kyowa Hakko Kogyo Co., Ltd. α2→3 sialyltransferase
IT1260468B (it) 1992-01-29 1996-04-09 Metodo per mantenere l'attivita' di enzimi proteolitici modificati con polietilenglicole
US5962294A (en) 1992-03-09 1999-10-05 The Regents Of The University Of California Compositions and methods for the identification and synthesis of sialyltransferases
WO1993018157A1 (fr) 1992-03-09 1993-09-16 The Regents Of The University Of California Compositions et procedes d'identification et de synthese de sialyltransferases
WO1993018787A1 (fr) 1992-03-25 1993-09-30 New York University Trans-sialidase et ses procedes d'utilisation et de fabrication
JPH0686684A (ja) 1992-05-26 1994-03-29 Monsanto Co シアロ抱合体の合成
JPH08503125A (ja) * 1992-08-07 1996-04-09 プロジェニクス・ファーマスーティカルス・インコーポレーテッド 非ペプチジル成分と複合化されたCD4−ガンマ2およびCD4−IgG2免疫複合体、並びにその使用
AU5006993A (en) 1992-08-21 1994-03-15 Enzon, Inc. Novel attachment of polyalkylene oxides to bio-effecting substances
JP3979678B2 (ja) 1992-08-24 2007-09-19 サントリー株式会社 新規糖転移酵素及びそれをコードする遺伝子並びに該酵素の製造方法
WO1994005332A2 (fr) 1992-09-01 1994-03-17 Berlex Laboratories, Inc. Glycolation de macromolecules glycosylees
US5308460A (en) * 1992-10-30 1994-05-03 Glyko, Incorporated Rapid synthesis and analysis of carbohydrates
US6361977B1 (en) * 1992-11-24 2002-03-26 S. Christopher Bauer Methods of using multivariant IL-3 hematopoiesis fusion protein
JP2845698B2 (ja) 1992-11-25 1999-01-13 オルガノ株式会社 復水循環系の復水に含まれる陰イオンの測定方法及び装置
JP3202365B2 (ja) 1992-12-04 2001-08-27 株式会社紀文フードケミファ オリゴマンヌロン酸を重合度によって分離する方法
NZ250375A (en) 1992-12-09 1995-07-26 Ortho Pharma Corp Peg hydrazone and peg oxime linkage forming reagents and protein derivatives
CA2110543A1 (fr) 1992-12-09 1994-06-10 David E. Wright Reactifs formant des chainons peg-hydrazone et peg-oxyme et derives proteiniques ainsi obtenus
FI935485A (fi) 1992-12-09 1994-06-10 Ortho Pharma Corp PEG-hydratsoni- ja PEG-oksiimisidoksen muodostavat reagenssit ja niiden proteiinijohdannaiset
AU6029594A (en) 1993-01-15 1994-08-15 Enzon, Inc. Factor viii - polymeric conjugates
US5349001A (en) 1993-01-19 1994-09-20 Enzon, Inc. Cyclic imide thione activated polyalkylene oxides
US5321095A (en) 1993-02-02 1994-06-14 Enzon, Inc. Azlactone activated polyalkylene oxides
US6180134B1 (en) 1993-03-23 2001-01-30 Sequus Pharmaceuticals, Inc. Enhanced ciruclation effector composition and method
EP0643132B1 (fr) 1993-03-29 2006-05-31 Kyowa Hakko Kogyo Co., Ltd. Alpha-1-3-fucosyltransferase
US5374541A (en) 1993-05-04 1994-12-20 The Scripps Research Institute Combined use of β-galactosidase and sialyltransferase coupled with in situ regeneration of CMP-sialic acid for one pot synthesis of oligosaccharides
US5409817A (en) * 1993-05-04 1995-04-25 Cytel, Inc. Use of trans-sialidase and sialyltransferase for synthesis of sialylα2→3βgalactosides
JPH09501044A (ja) 1993-05-14 1997-02-04 ジ・アップジョン・カンパニー UDP−GALNAc:ポリペプチド、N−アセチルガラクトサミニルトランスフェラーゼをコードするクローン化されたDNA
RO117533B1 (ro) 1993-05-14 2002-04-30 Cytel Corp San Diego DERIVATI DE ANALOGI SIALIL LeX INHIBITORI AI ADEZIUNII CELULARE SI COMPOZITIE FARMACEUTICA CU ACESTIA
HU219682B (hu) 1993-05-21 2001-06-28 Novo Nordisk A/S. Módosított VII faktor
WO1994028024A1 (fr) 1993-06-01 1994-12-08 Enzon, Inc. Polymere modifie aux glucides presentant une activite erythropoïetique
WO1995002683A1 (fr) 1993-07-15 1995-01-26 Neose Pharmaceuticals Procede de synthese de compositions de saccharide
DE4325317C2 (de) 1993-07-29 1998-05-20 Univ Dresden Tech Verfahren zur radioaktiven Markierung von Immunglobulinen
CN1057534C (zh) 1993-08-17 2000-10-18 柯瑞英-艾格公司 促红细胞生成素类似物
JPH0770195A (ja) * 1993-08-23 1995-03-14 Yutaka Mizushima 糖修飾インターフェロン
US6485930B1 (en) * 1993-09-15 2002-11-26 The Scripps Research Institute Mannosyl transfer with regeneration of GDP-mannose
CN1057775C (zh) 1993-09-22 2000-10-25 味之素株式会社 具有抗血栓活性的肽及其制法
US5874075A (en) * 1993-10-06 1999-02-23 Amgen Inc. Stable protein: phospholipid compositions and methods
US5643575A (en) * 1993-10-27 1997-07-01 Enzon, Inc. Non-antigenic branched polymer conjugates
US5919455A (en) 1993-10-27 1999-07-06 Enzon, Inc. Non-antigenic branched polymer conjugates
US5446090A (en) * 1993-11-12 1995-08-29 Shearwater Polymers, Inc. Isolatable, water soluble, and hydrolytically stable active sulfones of poly(ethylene glycol) and related polymers for modification of surfaces and molecules
US5369017A (en) 1994-02-04 1994-11-29 The Scripps Research Institute Process for solid phase glycopeptide synthesis
JP3516272B2 (ja) 1994-02-10 2004-04-05 株式会社成和化成 化粧品基材
US5837458A (en) 1994-02-17 1998-11-17 Maxygen, Inc. Methods and compositions for cellular and metabolic engineering
MX9504664A (es) * 1994-03-07 1997-05-31 Dow Chemical Co Conjugados de dendrimeros bioactivos y/o dirigidos.
IL113010A (en) 1994-03-31 1999-10-28 Pharmacia & Upjohn Ab Pharmaceutical formulation comprising factor viii with an activity of at least 500iu/ml and an enhancer for improved subcutaneous intramuscular or intradermal administration
US5432059A (en) * 1994-04-01 1995-07-11 Specialty Laboratories, Inc. Assay for glycosylation deficiency disorders
US5646113A (en) 1994-04-07 1997-07-08 Genentech, Inc. Treatment of partial growth hormone insensitivity syndrome
WO1996010089A1 (fr) 1994-09-29 1996-04-04 Ajinomoto Co., Inc. Modification d'un peptide et d'une proteine
US5824784A (en) 1994-10-12 1998-10-20 Amgen Inc. N-terminally chemically modified protein compositions and methods
US5834251A (en) 1994-12-30 1998-11-10 Alko Group Ltd. Methods of modifying carbohydrate moieties
US5932462A (en) 1995-01-10 1999-08-03 Shearwater Polymers, Inc. Multiarmed, monofunctional, polymer for coupling to molecules and surfaces
IL116730A0 (en) 1995-01-13 1996-05-14 Amgen Inc Chemically modified interferon
SE9501285L (sv) 1995-04-06 1996-10-07 Johanna Ljung Förfarande för framställning av biologiskt aktiva proteiner
US5922577A (en) 1995-04-11 1999-07-13 Cytel Corporation Enzymatic synthesis of glycosidic linkages
ATE222294T1 (de) 1995-04-11 2002-08-15 Neose Technologies Inc Verbesserte verfahren zur enzymatischen synthese von oligosacchariden
US5695760A (en) * 1995-04-24 1997-12-09 Boehringer Inglehiem Pharmaceuticals, Inc. Modified anti-ICAM-1 antibodies and their use in the treatment of inflammation
AU5920096A (en) 1995-05-15 1996-11-29 Constantin A. Bona Carbohydrate-mediated coupling of peptides to immunoglobulins
US5824864A (en) 1995-05-25 1998-10-20 Pioneer Hi-Bred International, Inc. Maize gene and protein for insect control
US5858752A (en) 1995-06-07 1999-01-12 The General Hospital Corporation Fucosyltransferase genes and uses thereof
US6127153A (en) 1995-06-07 2000-10-03 Neose Technologies, Inc. Method of transferring at least two saccharide units with a polyglycosyltransferase, a polyglycosyltransferase and gene encoding a polyglycosyltransferase
US6251864B1 (en) * 1995-06-07 2001-06-26 Glaxo Group Limited Peptides and compounds that bind to a receptor
WO1996040731A1 (fr) 1995-06-07 1996-12-19 Mount Sinai School Of Medicine Of The City University Of New York Proteines modifiees conjuguees a du polyethylene-glycol (peg)
US5672662A (en) 1995-07-07 1997-09-30 Shearwater Polymers, Inc. Poly(ethylene glycol) and related polymers monosubstituted with propionic or butanoic acids and functional derivatives thereof for biotechnical applications
AU6597996A (en) 1995-07-27 1997-02-26 Cytec Technology Corp. Synthetic cationic polymers as promoters for ASA sizing
SE9503380D0 (sv) * 1995-09-29 1995-09-29 Pharmacia Ab Protein derivatives
CA2165041C (fr) 1995-12-12 2005-07-05 The University Of British Columbia Methodes et compositions pour la synthese d'oligosaccharides; produits ainsi obtenus
WO1997021822A2 (fr) 1995-12-12 1997-06-19 The University Of British Columbia Procedes et compositions pour la synthese d'oligosaccharides a l'aide d'enzymes mutantes de type glycosidase
JP3065925B2 (ja) 1996-01-30 2000-07-17 日清製油株式会社 活性酸素種消去剤及び退色防止剤
DE69738368T2 (de) * 1996-03-08 2008-12-04 The Regents Of The University Of Michigan, Ann Arbor MURINE alpha(1,3)-FUCOSYLTRANSFERASE (Fuc-TVII)
IL128229A0 (en) 1996-08-02 1999-11-30 Ortho Mcneil Pharm Inc Polypeptides having a single covalently bound n-terminal water-soluble polymer
EP0953354A4 (fr) 1996-08-13 2002-10-23 Fujisawa Pharmaceutical Co Agents de proliferation des cellules souches hematopoietiques
US20020064546A1 (en) 1996-09-13 2002-05-30 J. Milton Harris Degradable poly(ethylene glycol) hydrogels with controlled half-life and precursors therefor
AU735695B2 (en) 1996-10-10 2001-07-12 Neose Technologies, Inc. Carbohydrate purification using ultrafiltration, reverse osmosis and nanofiltration
DE69723473T2 (de) 1996-10-15 2004-05-27 The Liposome Co., Inc. Peptid-lipid konjugate, liposomen und liposomale arzneimittelverabreichung
ATE386126T1 (de) 1996-11-08 2008-03-15 Neose Technologies Inc Verbesserte expressionsvektoren
JP2001508783A (ja) 1997-01-29 2001-07-03 ポリマスク・ファーマシューティカルズ・パブリック・リミテッド・カンパニー Peg化法
DE19709787A1 (de) 1997-03-11 1998-09-17 Bayer Ag Oligosaccaride und deren Derivate sowie ein chemo-enzymatisches Verfahren zu deren Herstellung
US5945314A (en) 1997-03-31 1999-08-31 Abbott Laboratories Process for synthesizing oligosaccharides
EP0981548A4 (fr) 1997-04-30 2005-11-23 Enzon Inc Proteines a chaine unique fixant les antigenes capables de glycosylation, production et utilisations de ces dernieres
JPH10307356A (ja) 1997-05-08 1998-11-17 Konica Corp ハロゲン化銀乳剤およびそれを用いたハロゲン化銀写真感光材料
US6075134A (en) * 1997-05-15 2000-06-13 The Regents Of The University Of California Glycoconjugates and methods
CA2293829C (fr) 1997-06-24 2011-06-14 Genentech, Inc. Procedes et compositions concernant des glycoproteines galactosylees
AU8269898A (en) 1997-06-27 1999-01-19 Regents Of The University Of California, The Drug targeting of a peptide radiopharmaceutical through the primate blood-brain barrier in vivo with a monoclonal antibody to the human insulin receptor
WO1999013063A1 (fr) 1997-09-09 1999-03-18 Nycomed Imaging As Fragments du facteur vii et leurs analogues, utilisation dans le traitement des troubles dus aux caillots sanguins
AU759779B2 (en) 1997-10-31 2003-05-01 Genentech Inc. Methods and compositions comprising glycoprotein glycoforms
AU744303B2 (en) 1997-12-01 2002-02-21 Neose Technologies, Inc. Enzymatic synthesis of gangliosides
US6362276B1 (en) 1998-01-07 2002-03-26 Debio Recherche Pharmaceutique S.A. Degradable heterobifunctional poly(ethylene glycol) acrylates and gels and conjugates derived therefrom
AU2559799A (en) 1998-01-22 1999-08-09 Genentech Inc. Antibody fragment-polymer conjugates and humanized anti-il-8 monoclonal antibodies and uses of same
PT1071700E (pt) 1998-04-20 2010-04-23 Glycart Biotechnology Ag Modificação por glicosilação de anticorpos para melhorar a citotoxicidade celular dependente de anticorpos
ATE399567T1 (de) 1998-04-28 2008-07-15 Serono Lab Peg konjugate von lhrh analogen
US20030166525A1 (en) 1998-07-23 2003-09-04 Hoffmann James Arthur FSH Formulation
AU762616B2 (en) 1998-10-16 2003-07-03 Biogen Ma Inc. Polymer conjugates of interferon beta-1a and uses
US7304150B1 (en) 1998-10-23 2007-12-04 Amgen Inc. Methods and compositions for the prevention and treatment of anemia
ES2289824T3 (es) 1998-10-30 2008-02-01 Novozymes A/S Proteinas glicosiladas con alergenicidad reducida.
EP1129179B1 (fr) 1998-11-13 2008-09-10 Henrick Clausen Udp-galactose: beta1(n)-acetyle-glucosamine beta1,3galactosyle-transferases, beta3gal-t5
AU773845B2 (en) 1998-11-18 2004-06-10 Neose Technologies, Inc. Low cost manufacture of oligosaccharides
US6465220B1 (en) 1998-12-21 2002-10-15 Glycozym Aps Glycosylation using GalNac-T4 transferase
ATE246202T1 (de) 1999-01-29 2003-08-15 Hoffmann La Roche Gcsf konjugate
US6503744B1 (en) 1999-02-01 2003-01-07 National Research Council Of Canada Campylobacter glycosyltransferases for biosynthesis of gangliosides and ganglioside mimics
US6949372B2 (en) 1999-03-02 2005-09-27 The Johns Hopkins University Engineering intracellular sialylation pathways
AU769842B2 (en) 1999-04-22 2004-02-05 Astrazeneca Ab Assay for detecting phospho-N-acetylmuramyl-pentapeptide translocase activity
PE20010288A1 (es) 1999-07-02 2001-03-07 Hoffmann La Roche Derivados de eritropoyetina
US6261805B1 (en) 1999-07-15 2001-07-17 Boyce Thompson Institute For Plant Research, Inc. Sialyiation of N-linked glycoproteins in the baculovirus expression vector system
WO2001005434A2 (fr) 1999-07-20 2001-01-25 Amgen Inc. Conjugues hyaluroniques acide-proteine, compositions pharmaceutiques et procedes correspondants
US6534300B1 (en) 1999-09-14 2003-03-18 Genzyme Glycobiology Research Institute, Inc. Methods for producing highly phosphorylated lysosomal hydrolases
CA2393200C (fr) 1999-12-02 2011-07-12 Zymogenetics, Inc. Procedes ciblant les cellules exprimant les recepteurs 2 et 3 de croissance des fibroblastes
CA2395254C (fr) 1999-12-24 2010-05-11 Kyowa Hakko Kogyo Co., Ltd. Glycols de polyalkylene ramifies
IL150314A0 (en) 1999-12-30 2002-12-01 Maxygen Aps Improved lysosomal enzymes and lysosomal enzyme activators
US6555660B2 (en) 2000-01-10 2003-04-29 Maxygen Holdings Ltd. G-CSF conjugates
US6646110B2 (en) * 2000-01-10 2003-11-11 Maxygen Holdings Ltd. G-CSF polypeptides and conjugates
CN1188172C (zh) 2000-01-10 2005-02-09 马克西根控股公司 G-csf偶联物
RU2278123C2 (ru) 2000-02-11 2006-06-20 Максиджен Холдингз Лтд. Молекулы, подобные фактору vii или viia
WO2001058493A1 (fr) 2000-02-11 2001-08-16 Maxygen Aps Conjugates of follicle stimulating hormones
WO2001060411A1 (fr) 2000-02-18 2001-08-23 Kanagawa Academy Of Science And Technology Composition pharmaceutique, reactif et procede d'administration intracerebrale d'ingredients a activite pharmaceutique ou de substances de marquage
US20010041683A1 (en) 2000-03-09 2001-11-15 Schmitz Harold H. Cocoa sphingolipids, cocoa extracts containing sphingolipids and methods of making and using same
US6586398B1 (en) 2000-04-07 2003-07-01 Amgen, Inc. Chemically modified novel erythropoietin stimulating protein compositions and methods
US6905683B2 (en) 2000-05-03 2005-06-14 Novo Nordisk Healthcare A/G Human coagulation factor VII variants
EP1282693B1 (fr) 2000-05-03 2010-10-20 Novo Nordisk Health Care AG Variants du facteur vii humain de coagulation
US7338932B2 (en) 2000-05-11 2008-03-04 Glycozym Aps Methods of modulating functions of polypeptide GalNAc-transferases and of screening test substances to find agents herefor, pharmaceutical compositions comprising such agents and the use of such agents for preparing medicaments
EP1284987B1 (fr) 2000-05-16 2007-07-18 Bolder Biotechnology, Inc. Procede de repliement de proteines renfermant des residus de cysteine libre
DE60139720D1 (de) 2000-06-28 2009-10-08 Glycofi Inc Verfahren für die Herstellung modifizierter Glykoproteine
CA2412882A1 (fr) 2000-06-30 2002-01-10 Maxygen Aps Polypeptides glycosyles a extension peptidique
US6423826B1 (en) 2000-06-30 2002-07-23 Regents Of The University Of Minnesota High molecular weight derivatives of vitamin K-dependent polypeptides
KR100396983B1 (ko) 2000-07-29 2003-09-02 이강춘 고반응성의 가지 달린 고분자 유도체 및 고분자와 단백질또는 펩타이드의 접합체
AU2001283740A1 (en) 2000-08-17 2002-02-25 University Of British Columbia Chemotherapeutic agents conjugated to p97 and their methods of use in treating neurological tumours
WO2002013873A2 (fr) 2000-08-17 2002-02-21 Synapse Technologies, Inc. Compositions de composes conjugues a p97 et leurs methodes d'utilisation
US6903069B2 (en) 2000-10-02 2005-06-07 Novo Nordisk Health Care A/S Factor VII glycoforms
US20020142964A1 (en) 2000-11-02 2002-10-03 Nissen Torben Lauesgaard Single-chain polypeptides
MXPA03003576A (es) 2000-11-27 2003-10-14 Rmf Dictagene Sa Procedimiento para plegar polipeptidos sintetizados quimicamente.
WO2002044196A1 (fr) 2000-11-28 2002-06-06 University Of Massachusetts Methodes et reactifs pour l'introduction d'un groupe sulfhydryle dans l'extremite 5' de l'arn
MXPA03005406A (es) 2000-12-20 2003-09-25 Hoffmann La Roche Conjugados de eritropoyetina.
ATE505204T1 (de) 2000-12-20 2011-04-15 Hoffmann La Roche Konjugate von erythropoietin (epo) mit polyethylenglykol (peg)
US7892730B2 (en) 2000-12-22 2011-02-22 Sagres Discovery, Inc. Compositions and methods for cancer
PA8536201A1 (es) 2000-12-29 2002-08-29 Kenneth S Warren Inst Inc Protección y mejoramiento de células, tejidos y órganos que responden a la eritropoyetina
SE0004932D0 (sv) * 2000-12-31 2000-12-31 Apbiotech Ab A method for mixed mode adsorption and mixed mode adsorbents
MXPA03007619A (es) 2001-02-27 2003-12-04 Maxygen Aps Nuevas moleculas similares a interferon beta.
KR100399357B1 (ko) * 2001-03-19 2003-09-26 삼성전자주식회사 코발트 실리사이드를 이용한 반도체 장치 및 그 형성 방법
ES2432967T3 (es) 2001-03-22 2013-12-05 Novo Nordisk Health Care Ag Derivados del Factor VII de coagulación
US7235638B2 (en) 2001-03-22 2007-06-26 Novo Nordisk Healthcare A/G Coagulation factor VII derivatives
BR0209177A (pt) 2001-05-03 2004-10-05 Merck Patent Gmbh Anticorpo especìfico a tumor recombinante e uso do mesmo
US20020168323A1 (en) 2001-05-11 2002-11-14 Igor Gonda Optimization of the molecular properties and formulation of proteins delivered by inhalation
WO2002092619A2 (fr) 2001-05-14 2002-11-21 The Gouvernment Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services Hormone de croissance modifiee
NZ530545A (en) 2001-07-11 2006-10-27 Maxygen Holdings Ltd Specific conjugates comprising a polypeptide exhibiting G-CSF activity and a non-polypeptide moiety
KR100453877B1 (ko) 2001-07-26 2004-10-20 메덱스젠 주식회사 연쇄체화에 의한 면역 글로블린 융합 단백질의 제조 방법 및 이 방법에 의해 제조된 TNFR/Fc 융합 단백질, 상기 단백질을 코딩하는 DNA, 상기 DNA를 포함하는벡터, 및 상기 벡터에 의한 형질전환체
WO2003011879A1 (fr) 2001-08-01 2003-02-13 Neose Technologies, Inc. Glycosyle-sphingosines et glycosphingolipides neutres et procedes d'isolement de ces composes
NZ531139A (en) 2001-08-29 2007-09-28 Neose Technologies Inc Synthetic ganglioside derivatives and compositions thereof
US6930086B2 (en) 2001-09-25 2005-08-16 Hoffmann-La Roche Inc. Diglycosylated erythropoietin
US7052868B2 (en) * 2001-09-27 2006-05-30 Novo Nordisk Healthcare A/G Human coagulation factor VII polypeptides
US7439043B2 (en) 2001-10-10 2008-10-21 Neose Technologies, Inc. Galactosyl nucleotide sugars
US7265085B2 (en) 2001-10-10 2007-09-04 Neose Technologies, Inc. Glycoconjugation methods and proteins/peptides produced by the methods
US6784154B2 (en) * 2001-11-01 2004-08-31 University Of Utah Research Foundation Method of use of erythropoietin to treat ischemic acute renal failure
EP1461445B1 (fr) 2001-11-28 2008-08-20 Neose Technologies, Inc. Remodelage glycoproteinique au moyen d'endoglycanases
EP1504023A4 (fr) 2002-05-03 2006-03-22 Neose Technologies Inc Proteines de fusion recombinantes de glycosyltransferase
MXPA04012496A (es) 2002-06-21 2005-09-12 Novo Nordisk Healthcare Ag Glicoformos del factor vii pegilados.
AU2003240439B2 (en) 2002-06-21 2009-05-21 Novo Nordisk Health Care Ag Pegylated factor VII glycoforms
DE10232916B4 (de) 2002-07-19 2008-08-07 Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. Vorrichtung und Verfahren zum Charakterisieren eines Informationssignals
WO2004009838A2 (fr) 2002-07-23 2004-01-29 Neose Technologies, Inc. Fucosyltransferases de h. pylori
AU2003254665C1 (en) 2002-08-01 2010-08-19 National Research Council Of Canada Campylobacter glycans and glycopeptides
PT2255826E (pt) 2002-08-02 2016-06-08 Glaxosmithkline Biologicals Sa Composições de vacina de neisseria compreendendo uma combinação de antigénios
JP2006508918A (ja) 2002-09-05 2006-03-16 ザ ジェネラル ホスピタル コーポレーション 修飾されたアシアロインターフェロンおよびその使用
AU2003273300A1 (en) 2002-09-06 2004-03-29 Bayer Pharmaceuticals Corporation Modified glp-1 receptor agonists and their pharmacological methods of use
WO2005025606A1 (fr) 2003-09-09 2005-03-24 Warren Pharmaceuticals, Inc. Erythropoietines a action prolongee pouvant maintenir une activite de protection tissulaire d'une erythropoietine endogene
CN1703421B (zh) * 2002-09-20 2010-06-23 法玛西亚公司 降低peg化蛋白的聚集体水平的方法
DE60315847T2 (de) 2002-09-25 2008-05-15 Novo Nordisk Health Care Ag Varianten des menschliche koagulationsfaktors vii
US20040062748A1 (en) 2002-09-30 2004-04-01 Mountain View Pharmaceuticals, Inc. Polymer conjugates with decreased antigenicity, methods of preparation and uses thereof
DK1549677T3 (da) 2002-09-30 2011-07-18 Bayer Healthcare Llc FVII- eller FVIIa-varianter med forøget koagulationsaktivitet
BR0315178A (pt) 2002-10-09 2005-08-16 Neose Technologies Inc Eritropoietina: remodelagem e glicoconjugação de eritropoietina
JP4412461B2 (ja) 2002-11-20 2010-02-10 日油株式会社 修飾された生体関連物質、その製造方法および中間体
US7459436B2 (en) 2002-11-22 2008-12-02 Hoffmann-La Roche Inc. Treatment of disturbances of iron distribution
EP1424344A1 (fr) 2002-11-29 2004-06-02 Aventis Behring Gesellschaft mit beschränkter Haftung ADNc modifié du facteur VIII et dérivés
DE60228460D1 (de) * 2002-12-13 2008-10-02 Bioceuticals Arzneimittel Ag Verfahren zur Herstellung und Reinigung von Erythropoietin
WO2004060299A2 (fr) 2002-12-26 2004-07-22 Mountain View Pharmaceuticals, Inc. Conjugues polymeres d'interferon-beta presentant une activite biologique accrue
US7041635B2 (en) 2003-01-28 2006-05-09 In2Gen Co., Ltd. Factor VIII polypeptide
HUE058897T2 (hu) * 2003-02-26 2022-09-28 Nektar Therapeutics Polimer VIII-as faktor egység konjugátumok
WO2004083259A2 (fr) 2003-03-18 2004-09-30 Neose Technologies Inc. Formes activees de polymeres hydrosolubles
PL1605897T3 (pl) 2003-03-19 2012-12-31 Lilly Co Eli Związki będące połączeniem GLP-1 z poli(glikolem etylenowym)
WO2007022512A2 (fr) * 2005-08-19 2007-02-22 Neose Technologies, Inc. Facteur vii et facteur viia glycopegyle
US8791070B2 (en) * 2003-04-09 2014-07-29 Novo Nordisk A/S Glycopegylated factor IX
US7718363B2 (en) 2003-04-25 2010-05-18 The Kenneth S. Warren Institute, Inc. Tissue protective cytokine receptor complex and assays for identifying tissue protective compounds
TWI353991B (en) 2003-05-06 2011-12-11 Syntonix Pharmaceuticals Inc Immunoglobulin chimeric monomer-dimer hybrids
DK2298347T3 (en) 2003-05-06 2016-01-11 Biogen Hemophilia Inc COAGULATION FACTOR CHEMICAL PROTEINS FOR TREATING A HEMOSTATIC DISORDER
WO2004101597A2 (fr) 2003-05-13 2004-11-25 Frutarom Ltd. Procedes de reduction de liaisons disulfure
US7074755B2 (en) 2003-05-17 2006-07-11 Centocor, Inc. Erythropoietin conjugate compounds with extended half-lives
EP1481985A1 (fr) 2003-05-28 2004-12-01 Innogenetics N.V. NS3 du virus l'hépatite C (VHC) modifié pour traitement médical
GB0315457D0 (en) 2003-07-01 2003-08-06 Celltech R&D Ltd Biological products
JP2007501811A (ja) 2003-08-08 2007-02-01 ノボ ノルディスク ヘルス ケア アクチェンゲゼルシャフト 治療的な関心対象のタンパク質に対する遅延分子の選択的な化学物質接合のためのガラクトースオキシダーゼの使用。
EP1653991A2 (fr) 2003-08-08 2006-05-10 Fresenius Kabi Deutschland GmbH Conjugues de polymere et de proteine lies au moyen d'un groupe de liaison oxime
US7524813B2 (en) * 2003-10-10 2009-04-28 Novo Nordisk Health Care Ag Selectively conjugated peptides and methods of making the same
US20080305992A1 (en) 2003-11-24 2008-12-11 Neose Technologies, Inc. Glycopegylated erythropoietin
KR20060120141A (ko) 2003-11-24 2006-11-24 네오스 테크놀로지스, 인크. 글리코페질화 에리트로포이에틴
BRPI0417342A (pt) 2003-12-03 2007-04-17 Neose Technologies Inc fator estimulante de colÈnia de granulócitos glicopeguilado
US7956032B2 (en) * 2003-12-03 2011-06-07 Novo Nordisk A/S Glycopegylated granulocyte colony stimulating factor
JP4738346B2 (ja) 2003-12-03 2011-08-03 ノヴォ ノルディスク アー/エス GlycoPEG化された第IX因子
US20060040856A1 (en) * 2003-12-03 2006-02-23 Neose Technologies, Inc. Glycopegylated factor IX
US20080318850A1 (en) 2003-12-03 2008-12-25 Neose Technologies, Inc. Glycopegylated Factor Ix
WO2005067601A2 (fr) 2004-01-09 2005-07-28 Neose Technologies, Inc. Vecteurs pour l'expression proteique recombinante dans e. coli
US20070105770A1 (en) 2004-01-21 2007-05-10 Novo Nordisk A/S Transglutaminase mediated conjugation of peptides
AU2005208897B2 (en) 2004-01-26 2011-05-19 Ratiopharm Gmbh Branched polymeric sugars and nucleotides thereof
DK2860251T3 (en) 2004-02-12 2018-06-06 Archemix Llc APTAPMER PHARMACEUTICALS USEFUL IN TREATMENT OF COMPLEMENT-RELATED DISEASES
CA2557782A1 (fr) 2004-03-17 2005-10-06 Eli Lilly And Company Composes fgf-21 lies au glycol
US20070037966A1 (en) * 2004-05-04 2007-02-15 Novo Nordisk A/S Hydrophobic interaction chromatography purification of factor VII polypeptides
ATE455861T1 (de) 2004-05-04 2010-02-15 Novo Nordisk Healthcare Ag O-verknüpfte glykoformen von faktor vii und verfahren zu deren herstellung
JP4251399B2 (ja) 2004-05-21 2009-04-08 独立行政法人産業技術総合研究所 O結合型糖鎖が付加されたペプチドのスクリーニング方法
EP1765992A4 (fr) 2004-06-03 2009-01-07 Neose Technologies Inc Polypeptides galnact2 tronques et acides nucleiques
US20080206810A1 (en) 2004-06-03 2008-08-28 Neose Technologies, Inc. Truncated St6galnaci Polypeptides and Nucleic Acids
MXPA06015234A (es) 2004-06-30 2007-11-22 Nektar Therapeutics Al Corp Conjugados de fraccion polimero-factor ix.
JP2008509889A (ja) * 2004-06-30 2008-04-03 イージェン コーポレーション ペグ化インターフェロンα−1b
WO2006014466A2 (fr) 2004-07-02 2006-02-09 The Kenneth S. Warren Institute, Inc. Nouvelle epo carbamylee et procede permettant de la fabriquer
EP1771190A4 (fr) 2004-07-02 2009-07-22 Kenneth S Warren Inst Inc Procédé de production d'érythropoiétine entièrement carbamylée
EP1771066A2 (fr) 2004-07-13 2007-04-11 Neose Technologies, Inc. Remodelage de peg ramifie et glycosylation de peptide-1 semblable a glucagon glp-1
US20090292110A1 (en) 2004-07-23 2009-11-26 Defrees Shawn Enzymatic modification of glycopeptides
SE0401951D0 (sv) * 2004-07-29 2004-07-29 Amersham Biosciences Ab Chromatography method
JP2008507990A (ja) 2004-08-02 2008-03-21 ノボ ノルディスク ヘルス ケア アクチェンゲゼルシャフト Fviiの抱合体
DE602005021509D1 (de) 2004-08-17 2010-07-08 Csl Behring Gmbh Modifizierte vitamin-k-abhängige polypeptide
WO2006050247A2 (fr) 2004-10-29 2006-05-11 Neose Technologies, Inc. Remodelage et glycopegylation du facteur de croissance des fibroblastes (fgf)
EP1824988B1 (fr) 2004-11-12 2017-04-19 Bayer HealthCare LLC Modification du facteur fviii en fonction du site
MX2007007591A (es) * 2004-12-22 2007-07-25 Ambrx Inc Metodos para expresion y purificacion de hormona de crecimiento humano recombinante.
NZ556436A (en) 2005-01-10 2010-11-26 Biogenerix Ag Glycopegylated granulocyte colony stimulating factor
WO2006078645A2 (fr) 2005-01-19 2006-07-27 Neose Technologies, Inc. Expression de polypeptides heterologues associee a une faible multiplicite d'infection de virus
PA8660701A1 (es) 2005-02-04 2006-09-22 Pfizer Prod Inc Agonistas de pyy y sus usos
KR20080021590A (ko) 2005-03-24 2008-03-07 네오스 테크놀로지스, 인크. 가용성이고 활성인 진핵생물 글리코실트랜스퍼라제의원핵생물 유기체 내에서의 발현
WO2006105426A2 (fr) 2005-03-30 2006-10-05 Neose Technologies, Inc. Procede de fabrication pour la production de peptides cultives dans des lignees cellulaires d'insectes
US20080227691A1 (en) 2005-04-01 2008-09-18 Novo Nordisk Health Care Ag Blood Coagulation FVIII Analogues
EP2853600B1 (fr) 2005-05-11 2018-09-19 ETH Zürich Proteines N-glycosylées de recombinaison produites a partir de cellules procaryotes
US20110003744A1 (en) * 2005-05-25 2011-01-06 Novo Nordisk A/S Glycopegylated Erythropoietin Formulations
WO2006127910A2 (fr) 2005-05-25 2006-11-30 Neose Technologies, Inc. Formulations d'erythropoietine glycopegylees
WO2006134173A2 (fr) 2005-06-17 2006-12-21 Novo Nordisk Health Care Ag Reduction et derivation selectives de proteines conçues par le genie genetique comprenant au moins une cysteine non native
DE202006020194U1 (de) 2006-03-01 2007-12-06 Bioceuticals Arzneimittel Ag G-CSF-Flüssigformulierung
US7645860B2 (en) * 2006-03-31 2010-01-12 Baxter Healthcare S.A. Factor VIII polymer conjugates
CN104593348A (zh) 2006-05-24 2015-05-06 诺沃—诺迪斯克保健股份有限公司 具有延长的体内半寿期的因子ix类似物
ES2516694T3 (es) 2006-07-21 2014-10-31 Ratiopharm Gmbh Glicosilación de péptidos a través de secuencias de glicosilación con unión en O
ITMI20061624A1 (it) 2006-08-11 2008-02-12 Bioker Srl Mono-coniugati sito-specifici di g-csf
WO2008073620A2 (fr) 2006-11-02 2008-06-19 Neose Technologies, Inc. Procédé de fabrication pour la production de polypeptides exprimés dans des lignées cellulaires d'insectes
BRPI0809670A8 (pt) 2007-04-03 2018-12-18 Biogenerix Ag métodos para aumentar a produção de célula tronco, para aumentar o número de granulócitos em um indivíduo, para prevenir, tratar e aliviar a mielossupressão que resulta de uma terapia contra o câncer, para tratar uma condição em um indivíduo, para tratar neutropenia e trombocitopenia em um mamífero, para expandir células tronco hematopoiéticas em cultura, para aumentar a hematopoiese em um indivíduo, para aumentar o número de célulars progenitoras hematopoiéticas em um indivíduo, e para fornecer enxerto estável da medula óssea, e, forma de dosagem oral.
US20110177029A1 (en) 2007-06-04 2011-07-21 Novo Nordisk A/S O-linked glycosylation using n-acetylglucosaminyl transferases
CN101778859B (zh) 2007-06-12 2014-03-26 诺和诺德公司 改良的用于生产核苷酸糖的方法
US8207112B2 (en) * 2007-08-29 2012-06-26 Biogenerix Ag Liquid formulation of G-CSF conjugate
US20100286067A1 (en) 2008-01-08 2010-11-11 Biogenerix Ag Glycoconjugation of polypeptides using oligosaccharyltransferases

Patent Citations (98)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4088538A (en) * 1975-05-30 1978-05-09 Battelle Memorial Institute Reversibly precipitable immobilized enzyme complex and a method for its use
US4385260A (en) * 1975-09-09 1983-05-24 Beckman Instruments, Inc. Bargraph display
US4438253A (en) * 1982-11-12 1984-03-20 American Cyanamid Company Poly(glycolic acid)/poly(alkylene glycol) block copolymers and method of manufacturing the same
US4496689A (en) * 1983-12-27 1985-01-29 Miles Laboratories, Inc. Covalently attached complex of alpha-1-proteinase inhibitor with a water soluble polymer
US4565653A (en) * 1984-03-30 1986-01-21 Pfizer Inc. Acyltripeptide immunostimulants
US4806595A (en) * 1985-08-12 1989-02-21 Koken Co., Ltd. Method of preparing antithrombogenic medical materials
US4925796A (en) * 1986-03-07 1990-05-15 Massachusetts Institute Of Technology Method for enhancing glycoprotein stability
US4826945A (en) * 1987-06-09 1989-05-02 Yissum Research Development Company Biodegradable polymeric materials based on polyether glycols, processes for the preparation thereof and surgical articles made therefrom
US5194376A (en) * 1989-02-28 1993-03-16 University Of Ottawa Baculovirus expression system capable of producing foreign gene proteins at high levels
US5324844A (en) * 1989-04-19 1994-06-28 Enzon, Inc. Active carbonates of polyalkylene oxides for modification of polypeptides
US5122614A (en) * 1989-04-19 1992-06-16 Enzon, Inc. Active carbonates of polyalkylene oxides for modification of polypeptides
US5527527A (en) * 1989-09-07 1996-06-18 Alkermes, Inc. Transferrin receptor specific antibody-neuropharmaceutical agent conjugates
US5182107A (en) * 1989-09-07 1993-01-26 Alkermes, Inc. Transferrin receptor specific antibody-neuropharmaceutical or diagnostic agent conjugates
US5405753A (en) * 1990-03-26 1995-04-11 Brossmer; Reinhard CMP-activated, fluorescing sialic acids, as well as processes for their preparation
US6010999A (en) * 1990-05-04 2000-01-04 American Cyanamid Company Stabilization of fibroblast growth factors by modification of cysteine residues
US5219564A (en) * 1990-07-06 1993-06-15 Enzon, Inc. Poly(alkylene oxide) amino acid copolymers and drug carriers and charged copolymers based thereon
US5410016A (en) * 1990-10-15 1995-04-25 Board Of Regents, The University Of Texas System Photopolymerizable biodegradable hydrogels as tissue contacting materials and controlled-release carriers
US5529914A (en) * 1990-10-15 1996-06-25 The Board Of Regents The Univeristy Of Texas System Gels for encapsulation of biological materials
US5281698A (en) * 1991-07-23 1994-01-25 Cetus Oncology Corporation Preparation of an activated polymer ester for protein conjugation
US5858751A (en) * 1992-03-09 1999-01-12 The Regents Of The University Of California Compositions and methods for producing sialyltransferases
US6037452A (en) * 1992-04-10 2000-03-14 Alpha Therapeutic Corporation Poly(alkylene oxide)-Factor VIII or Factor IX conjugate
US5614184A (en) * 1992-07-28 1997-03-25 New England Deaconess Hospital Recombinant human erythropoietin mutants and therapeutic methods employing them
US5202413A (en) * 1993-02-16 1993-04-13 E. I. Du Pont De Nemours And Company Alternating (ABA)N polylactide block copolymers
US5621039A (en) * 1993-06-08 1997-04-15 Hallahan; Terrence W. Factor IX- polymeric conjugates
US5635603A (en) * 1993-12-08 1997-06-03 Immunomedics, Inc. Preparation and use of immunoconjugates
US5605793A (en) * 1994-02-17 1997-02-25 Affymax Technologies N.V. Methods for in vitro recombination
US5492841A (en) * 1994-02-18 1996-02-20 E. I. Du Pont De Nemours And Company Quaternary ammonium immunogenic conjugates and immunoassay reagents
US5629384A (en) * 1994-05-17 1997-05-13 Consiglio Nazionale Delle Ricerche Polymers of N-acryloylmorpholine activated at one end and conjugates with bioactive materials and surfaces
US5705367A (en) * 1994-09-26 1998-01-06 The Rockefeller University Glycosyltransferases for biosynthesis of oligosaccharides, and genes encoding them
US6342382B1 (en) * 1994-09-26 2002-01-29 The Rockefeller University Glycosyltransferases for biosynthesis of oligosaccharides, and genes encoding them
US5728554A (en) * 1995-04-11 1998-03-17 Cytel Corporation Enzymatic synthesis of glycosidic linkages
US5876980A (en) * 1995-04-11 1999-03-02 Cytel Corporation Enzymatic synthesis of oligosaccharides
US6030815A (en) * 1995-04-11 2000-02-29 Neose Technologies, Inc. Enzymatic synthesis of oligosaccharides
US6015555A (en) * 1995-05-19 2000-01-18 Alkermes, Inc. Transferrin receptor specific antibody-neuropharmaceutical or diagnostic agent conjugates
US5770420A (en) * 1995-09-08 1998-06-23 The Regents Of The University Of Michigan Methods and products for the synthesis of oligosaccharide structures on glycoproteins, glycolipids, or as free molecules, and for the isolation of cloned genetic sequences that determine these structures
US6057292A (en) * 1995-09-21 2000-05-02 Genentech, Inc. Method for inhibiting growth hormone action
US5716812A (en) * 1995-12-12 1998-02-10 The University Of British Columbia Methods and compositions for synthesis of oligosaccharides, and the products formed thereby
US6399336B1 (en) * 1997-01-16 2002-06-04 Neose Technologies, Inc. Practical in vitro sialylation of recombinant glycoproteins
US6183738B1 (en) * 1997-05-12 2001-02-06 Phoenix Pharamacologics, Inc. Modified arginine deiminase
US6399337B1 (en) * 1997-06-06 2002-06-04 The Governors Of The University Of Alberta α1,3-fucosyltransferase
US20030027257A1 (en) * 1997-08-21 2003-02-06 University Technologies International, Inc. Sequences for improving the efficiency of secretion of non-secreted protein from mammalian and insect cells
US6555346B1 (en) * 1997-12-18 2003-04-29 Stichting Instituut Voor Dierhouderij En Diergezondheid Protein expression in baculovirus vector expression systems
US6362254B2 (en) * 1998-03-12 2002-03-26 Shearwater Corporation Poly(ethylene glycol) derivatives with proximal reactive groups
US20040102607A1 (en) * 1998-03-25 2004-05-27 Danishefsky Samuel J. Trimeric antigenic O-linked glycopeptide conjugates, methods of preparation and uses thereof
US6692931B1 (en) * 1998-11-16 2004-02-17 Werner Reutter Recombinant glycoproteins, method for the production thereof, medicaments containing said glycoproteins and use thereof
US6716626B1 (en) * 1999-11-18 2004-04-06 Chiron Corporation Human FGF-21 nucleic acids
US6348558B1 (en) * 1999-12-10 2002-02-19 Shearwater Corporation Hydrolytically degradable polymers and hydrogels made therefrom
US6376604B2 (en) * 1999-12-22 2002-04-23 Shearwater Corporation Method for the preparation of 1-benzotriazolylcarbonate esters of poly(ethylene glycol)
US20020016003A1 (en) * 2000-03-16 2002-02-07 Eliana Saxon Chemoselective ligation
US20030040037A1 (en) * 2000-05-12 2003-02-27 Neose Technologies, Inc. In vitro modification of glycosylation patterns of recombinant glycopeptides
US20020037841A1 (en) * 2000-05-15 2002-03-28 Apollon Papadimitriou Erythropoietin composition
US7202208B2 (en) * 2000-05-15 2007-04-10 Hoffman-La Roche Inc. Erythropoietin composition
US6531121B2 (en) * 2000-12-29 2003-03-11 The Kenneth S. Warren Institute, Inc. Protection and enhancement of erythropoietin-responsive cells, tissues and organs
US7696163B2 (en) * 2001-10-10 2010-04-13 Novo Nordisk A/S Erythropoietin: remodeling and glycoconjugation of erythropoietin
US20100015684A1 (en) * 2001-10-10 2010-01-21 Neose Technologies, Inc. Factor vii: remodeling and glycoconjugation of factor vii
US20040077836A1 (en) * 2001-10-10 2004-04-22 Neose Technologies, Inc. Granulocyte colony stimulating factor: remodeling and glycoconjugation of G-CSF
US20040115168A1 (en) * 2001-10-10 2004-06-17 Neose Technologies, Inc. Interferon beta: remodeling and glycoconjugation of interferon beta
US20040063911A1 (en) * 2001-10-10 2004-04-01 Neose Technologies, Inc. Protein remodeling methods and proteins/peptides produced by the methods
US20050031584A1 (en) * 2001-10-10 2005-02-10 Neose Technologies, Inc. Interleukin-2:remodeling and glycoconjugation of interleukin-2
US20080050772A1 (en) * 2001-10-10 2008-02-28 Neose Technologies, Inc. Granulocyte colony stimulating factor: remodeling and glycoconjugation of G-CSF
US20050100982A1 (en) * 2001-10-10 2005-05-12 Neose Technologies, Inc. Factor IX: remodeling and glycoconjugation of factor IX
US20050106658A1 (en) * 2001-10-10 2005-05-19 Defrees Shawn Remodeling and glycoconjugation of peptides
US20040082026A1 (en) * 2001-10-10 2004-04-29 Neose Technologies, Inc. Interferon alpha: remodeling and glycoconjugation of interferon alpha
US7214660B2 (en) * 2001-10-10 2007-05-08 Neose Technologies, Inc. Erythropoietin: remodeling and glycoconjugation of erythropoietin
US7179617B2 (en) * 2001-10-10 2007-02-20 Neose Technologies, Inc. Factor IX: remolding and glycoconjugation of Factor IX
US20090137763A1 (en) * 2001-10-10 2009-05-28 Neose Technologies, Inc. Glucosamine nucleotide sugars
US20090093399A1 (en) * 2001-10-10 2009-04-09 Neose Technologies, Inc. Glycopegylation methods and proteins/peptides produced by the methods
US20070027068A1 (en) * 2001-10-10 2007-02-01 Defrees Shawn Erythropoietin: remodeling and glycoconjugation of erythropoietin
US20040043446A1 (en) * 2001-10-19 2004-03-04 Neose Technologies, Inc. Alpha galalctosidase a: remodeling and glycoconjugation of alpha galactosidase A
US7157277B2 (en) * 2001-11-28 2007-01-02 Neose Technologies, Inc. Factor VIII remodeling and glycoconjugation of Factor VIII
US7368108B2 (en) * 2001-11-28 2008-05-06 Neose Technologies, Inc. Glycopeptide remodeling using amidases
US20060030521A1 (en) * 2001-11-28 2006-02-09 Neose Technologies, Inc. Remodeling and glycoconjugation of peptides
US20060035224A1 (en) * 2002-03-21 2006-02-16 Johansen Jack T Purification methods for oligonucleotides and their analogs
US20050026266A1 (en) * 2002-11-08 2005-02-03 Glycozym Aps Methods to identify agents modulating functions of polypeptide galnac-transferases, pharmaceutical compositions comprising such agents and the use of such agents for preparing medicaments
US20050064540A1 (en) * 2002-11-27 2005-03-24 Defrees Shawn Ph.D Glycoprotein remodeling using endoglycanases
US20070032405A1 (en) * 2003-03-14 2007-02-08 Neose Technologies, Inc. Branched water-soluble polymers and their conjugates
US7691603B2 (en) * 2003-04-09 2010-04-06 Novo Nordisk A/S Intracellular formation of peptide conjugates
US20100048456A1 (en) * 2003-04-09 2010-02-25 Novo Nordisk A/S Glycopegylation methods and proteins/peptides produced by the methods
US20080102083A1 (en) * 2003-05-09 2008-05-01 Neose Technologies, Inc. Compositions and Methods for the Preparation of Human Growth Hormone Glycosylation Mutants
US20070059275A1 (en) * 2003-07-25 2007-03-15 Defrees Shawn Antibody toxin conjugates
US20100028939A1 (en) * 2003-08-08 2010-02-04 Novo Nordisk Healthcare A/G Use of Galactose Oxidase for Selective Chemical Conjugation of Protractor Molecules to Proteins of Therapeutic Interest
US20060111279A1 (en) * 2003-11-24 2006-05-25 Neose Technologies, Inc. Glycopegylated erythropoietin
US20080015142A1 (en) * 2003-12-03 2008-01-17 Defrees Shawn Glycopegylated Follicle Stimulating Hormone
US7338933B2 (en) * 2004-01-08 2008-03-04 Neose Technologies, Inc. O-linked glycosylation of peptides
US20060024286A1 (en) * 2004-08-02 2006-02-02 Paul Glidden Variants of tRNA synthetase fragments and uses thereof
US20090028822A1 (en) * 2004-09-10 2009-01-29 Neose Technologies, Inc. Glycopegylated Interferon Alpha
US20080108557A1 (en) * 2004-09-29 2008-05-08 Novo Nordisk Healthcare A/G Modified Proteins
US20090054623A1 (en) * 2004-12-17 2009-02-26 Neose Technologies, Inc. Lipo-Conjugation of Peptides
US20100009902A1 (en) * 2005-01-06 2010-01-14 Neose Technologies, Inc. Glycoconjugation Using Saccharyl Fragments
US20090124544A1 (en) * 2005-04-08 2009-05-14 Neose Technologies ,Inc. A Delaware Corporation Compositions and methods for the preparation of protease resistant human growth hormone glycosylation mutants
US20100081791A1 (en) * 2005-05-25 2010-04-01 Novo Nordisk A/S Glycopegylated factor ix
US20090055942A1 (en) * 2005-09-14 2009-02-26 Novo Nordisk Healthcare A/G Human Coagulation Factor VII Polypeptides
US20070105755A1 (en) * 2005-10-26 2007-05-10 Neose Technologies, Inc. One pot desialylation and glycopegylation of therapeutic peptides
US20090048440A1 (en) * 2005-11-03 2009-02-19 Neose Technologies, Inc. Nucleotide Sugar Purification Using Membranes
US20100056428A1 (en) * 2006-09-01 2010-03-04 Novo Nordisk Health Care Ag Modified proteins
US20100035299A1 (en) * 2006-10-03 2010-02-11 Novo Nordisk A/S Methods for the purification of polypeptide conjugates
US20100041872A1 (en) * 2006-10-04 2010-02-18 Defrees Shawn Glycerol linked pegylated sugars and glycopeptides
US20090053167A1 (en) * 2007-05-14 2009-02-26 Neose Technologies, Inc. C-, S- and N-glycosylation of peptides

Cited By (67)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8716240B2 (en) 2001-10-10 2014-05-06 Novo Nordisk A/S Erythropoietin: remodeling and glycoconjugation of erythropoietin
US20100261872A1 (en) * 2001-10-10 2010-10-14 Neose Technologies, Inc. Factor VIII: remodeling and glycoconjugation of factor VIII
US8076292B2 (en) 2001-10-10 2011-12-13 Novo Nordisk A/S Factor VIII: remodeling and glycoconjugation of factor VIII
US8716239B2 (en) 2001-10-10 2014-05-06 Novo Nordisk A/S Granulocyte colony stimulating factor: remodeling and glycoconjugation G-CSF
US7795210B2 (en) 2001-10-10 2010-09-14 Novo Nordisk A/S Protein remodeling methods and proteins/peptides produced by the methods
US20080050772A1 (en) * 2001-10-10 2008-02-28 Neose Technologies, Inc. Granulocyte colony stimulating factor: remodeling and glycoconjugation of G-CSF
US8008252B2 (en) 2001-10-10 2011-08-30 Novo Nordisk A/S Factor VII: remodeling and glycoconjugation of Factor VII
US20040063911A1 (en) * 2001-10-10 2004-04-01 Neose Technologies, Inc. Protein remodeling methods and proteins/peptides produced by the methods
US20090227504A1 (en) * 2002-06-21 2009-09-10 Novo Nordisk A/S Pegylated Factor VII Glycoforms
US8053410B2 (en) 2002-06-21 2011-11-08 Novo Nordisk Health Care A/G Pegylated factor VII glycoforms
US20100331489A1 (en) * 2003-03-14 2010-12-30 Biogenerix Ag Branched water-soluble polymers and their conjugates
US7803777B2 (en) 2003-03-14 2010-09-28 Biogenerix Ag Branched water-soluble polymers and their conjugates
US8247381B2 (en) 2003-03-14 2012-08-21 Biogenerix Ag Branched water-soluble polymers and their conjugates
US20070032405A1 (en) * 2003-03-14 2007-02-08 Neose Technologies, Inc. Branched water-soluble polymers and their conjugates
US8791070B2 (en) 2003-04-09 2014-07-29 Novo Nordisk A/S Glycopegylated factor IX
US8853161B2 (en) 2003-04-09 2014-10-07 Novo Nordisk A/S Glycopegylation methods and proteins/peptides produced by the methods
US8063015B2 (en) 2003-04-09 2011-11-22 Novo Nordisk A/S Glycopegylation methods and proteins/peptides produced by the methods
US20080102083A1 (en) * 2003-05-09 2008-05-01 Neose Technologies, Inc. Compositions and Methods for the Preparation of Human Growth Hormone Glycosylation Mutants
US7932364B2 (en) 2003-05-09 2011-04-26 Novo Nordisk A/S Compositions and methods for the preparation of human growth hormone glycosylation mutants
US9005625B2 (en) 2003-07-25 2015-04-14 Novo Nordisk A/S Antibody toxin conjugates
US20070059275A1 (en) * 2003-07-25 2007-03-15 Defrees Shawn Antibody toxin conjugates
US8633157B2 (en) 2003-11-24 2014-01-21 Novo Nordisk A/S Glycopegylated erythropoietin
US20080305992A1 (en) * 2003-11-24 2008-12-11 Neose Technologies, Inc. Glycopegylated erythropoietin
US20080300175A1 (en) * 2003-11-24 2008-12-04 Neose Technologies, Inc. Glycopegylated erythropoietin
US8916360B2 (en) 2003-11-24 2014-12-23 Novo Nordisk A/S Glycopegylated erythropoietin
US7842661B2 (en) 2003-11-24 2010-11-30 Novo Nordisk A/S Glycopegylated erythropoietin formulations
US20060287224A1 (en) * 2003-11-24 2006-12-21 Neose Technologies, Inc. Glycopegylated erythropoietin formulations
US7956032B2 (en) 2003-12-03 2011-06-07 Novo Nordisk A/S Glycopegylated granulocyte colony stimulating factor
US8632770B2 (en) 2003-12-03 2014-01-21 Novo Nordisk A/S Glycopegylated factor IX
US20070254836A1 (en) * 2003-12-03 2007-11-01 Defrees Shawn Glycopegylated Granulocyte Colony Stimulating Factor
US20090169509A1 (en) * 2004-01-08 2009-07-02 Defrees Shawn O-linked glycosylation of peptides
US20080242846A1 (en) * 2004-01-08 2008-10-02 Neose Technologies, Inc. O-linked glycosylation of peptides
US8361961B2 (en) 2004-01-08 2013-01-29 Biogenerix Ag O-linked glycosylation of peptides
US8791066B2 (en) 2004-07-13 2014-07-29 Novo Nordisk A/S Branched PEG remodeling and glycosylation of glucagon-like peptide-1 [GLP-1]
US20090292110A1 (en) * 2004-07-23 2009-11-26 Defrees Shawn Enzymatic modification of glycopeptides
US8268967B2 (en) 2004-09-10 2012-09-18 Novo Nordisk A/S Glycopegylated interferon α
US20090028822A1 (en) * 2004-09-10 2009-01-29 Neose Technologies, Inc. Glycopegylated Interferon Alpha
US20080176790A1 (en) * 2004-10-29 2008-07-24 Defrees Shawn Remodeling and Glycopegylation of Fibroblast Growth Factor (Fgf)
US10874714B2 (en) 2004-10-29 2020-12-29 89Bio Ltd. Method of treating fibroblast growth factor 21 (FGF-21) deficiency
US9200049B2 (en) 2004-10-29 2015-12-01 Novo Nordisk A/S Remodeling and glycopegylation of fibroblast growth factor (FGF)
US20100009902A1 (en) * 2005-01-06 2010-01-14 Neose Technologies, Inc. Glycoconjugation Using Saccharyl Fragments
US20090203579A1 (en) * 2005-01-10 2009-08-13 Defrees Shawn Glycopegylated Granulocyte Colony Stimulating Factor
US9029331B2 (en) 2005-01-10 2015-05-12 Novo Nordisk A/S Glycopegylated granulocyte colony stimulating factor
US9187546B2 (en) 2005-04-08 2015-11-17 Novo Nordisk A/S Compositions and methods for the preparation of protease resistant human growth hormone glycosylation mutants
US8404809B2 (en) 2005-05-25 2013-03-26 Novo Nordisk A/S Glycopegylated factor IX
US20110003744A1 (en) * 2005-05-25 2011-01-06 Novo Nordisk A/S Glycopegylated Erythropoietin Formulations
US8911967B2 (en) 2005-08-19 2014-12-16 Novo Nordisk A/S One pot desialylation and glycopegylation of therapeutic peptides
US20090048440A1 (en) * 2005-11-03 2009-02-19 Neose Technologies, Inc. Nucleotide Sugar Purification Using Membranes
US8841439B2 (en) 2005-11-03 2014-09-23 Novo Nordisk A/S Nucleotide sugar purification using membranes
US9187532B2 (en) 2006-07-21 2015-11-17 Novo Nordisk A/S Glycosylation of peptides via O-linked glycosylation sequences
US20080255040A1 (en) * 2006-07-21 2008-10-16 Neose Technologies, Inc. Glycosylation of peptides via o-linked glycosylation sequences
US8969532B2 (en) 2006-10-03 2015-03-03 Novo Nordisk A/S Methods for the purification of polypeptide conjugates comprising polyalkylene oxide using hydrophobic interaction chromatography
US20100041872A1 (en) * 2006-10-04 2010-02-18 Defrees Shawn Glycerol linked pegylated sugars and glycopeptides
US20080146782A1 (en) * 2006-10-04 2008-06-19 Neose Technologies, Inc. Glycerol linked pegylated sugars and glycopeptides
US9050304B2 (en) 2007-04-03 2015-06-09 Ratiopharm Gmbh Methods of treatment using glycopegylated G-CSF
US20100120666A1 (en) * 2007-04-03 2010-05-13 Biogenerix Ag Methods of treatment using glycopegylated g-csf
US20110177029A1 (en) * 2007-06-04 2011-07-21 Novo Nordisk A/S O-linked glycosylation using n-acetylglucosaminyl transferases
US9493499B2 (en) 2007-06-12 2016-11-15 Novo Nordisk A/S Process for the production of purified cytidinemonophosphate-sialic acid-polyalkylene oxide (CMP-SA-PEG) as modified nucleotide sugars via anion exchange chromatography
US20090000924A1 (en) * 2007-06-29 2009-01-01 Harley-Davidson Motor Company Group, Inc. Integrated ignition and key switch
US8207112B2 (en) 2007-08-29 2012-06-26 Biogenerix Ag Liquid formulation of G-CSF conjugate
US20090143292A1 (en) * 2007-08-29 2009-06-04 Neose Technologies, Inc. Liquid Formulation of G-CSF Conjugate
US20100286067A1 (en) * 2008-01-08 2010-11-11 Biogenerix Ag Glycoconjugation of polypeptides using oligosaccharyltransferases
US9150848B2 (en) 2008-02-27 2015-10-06 Novo Nordisk A/S Conjugated factor VIII molecules
US20110166332A1 (en) * 2008-09-12 2011-07-07 Ge Healthcare Bio-Sciences Ab Enhanced antibody aggregate removal with capto adhere in the presence of protein-excluded zwitterions
US9488625B2 (en) 2010-12-15 2016-11-08 Baxalta GmbH Purification of factor VIII using a conductivity gradient
US9169331B2 (en) 2012-12-21 2015-10-27 Dionex Corporation Separation of glycans by mixed-mode liquid chromatography
US9310344B2 (en) 2013-06-14 2016-04-12 Dionex Corporation HILIC/anion-exchange/cation-exchange multimodal media

Also Published As

Publication number Publication date
US8969532B2 (en) 2015-03-03
JP2010505874A (ja) 2010-02-25
IL197765A0 (en) 2009-12-24
EP2054521A2 (fr) 2009-05-06
US20100035299A1 (en) 2010-02-11
EP2054521A4 (fr) 2012-12-19
WO2008057683A2 (fr) 2008-05-15
US20080253992A1 (en) 2008-10-16
WO2008057683A3 (fr) 2009-01-15

Similar Documents

Publication Publication Date Title
US8969532B2 (en) Methods for the purification of polypeptide conjugates comprising polyalkylene oxide using hydrophobic interaction chromatography
US20080207487A1 (en) Manufacturing process for the production of polypeptides expressed in insect cell-lines
KR101556248B1 (ko) 글리세롤이 연결된 페길화된 당 및 글리코펩티드
US7368108B2 (en) Glycopeptide remodeling using amidases
EP1461445B1 (fr) Remodelage glycoproteinique au moyen d'endoglycanases
EP1694274B1 (fr) Facteur de stimulation de colonies de granulocytes glycopegylé
EP1428878B1 (fr) Procédé pour la préparation et la purification de l'érythropoiétine
US9029331B2 (en) Glycopegylated granulocyte colony stimulating factor
US20060246544A1 (en) Manufacturing process for the production of peptides grown in insect cell lines
US20050064540A1 (en) Glycoprotein remodeling using endoglycanases
US20070105755A1 (en) One pot desialylation and glycopegylation of therapeutic peptides
CA2607844C (fr) Polypeptides g-csf pegyles et procedes de production correspondants
JP6151640B2 (ja) たん白質の精製方法
JP2010531135A (ja) N−アセチルグルコサミニルトランスフェラーゼを使用したo結合型グリコシル化
EP1613261A2 (fr) Formation intracellulaire de conjugues de peptides
KR20090016596A (ko) 에리트로포이에틴 조성물
KR101847169B1 (ko) 지속형 에리트로포이에틴 함유 조성물
WO2023040792A1 (fr) Procédé de préparation d'érythropoïétine
KR20180026688A (ko) 지속형 에리트로포이에틴 함유 조성물

Legal Events

Date Code Title Description
AS Assignment

Owner name: NOVO NORDISK A/S,DENMARK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:NEOSE TECHNOLOGIES, INC.;REEL/FRAME:022441/0937

Effective date: 20090127

Owner name: NOVO NORDISK A/S, DENMARK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:NEOSE TECHNOLOGIES, INC.;REEL/FRAME:022441/0937

Effective date: 20090127

AS Assignment

Owner name: NOVO NORDISK A/S,DENMARK

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:DEFREES, SHAWN;KINEALY, KYLE;SIGNING DATES FROM 20090420 TO 20090422;REEL/FRAME:023013/0551

STCB Information on status: application discontinuation

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