US20070026485A1 - Glycopegylation methods and proteins/peptides produced by the methods - Google Patents

Glycopegylation methods and proteins/peptides produced by the methods

Info

Publication number
US20070026485A1
US20070026485A1 US10/552,896 US55289604A US2007026485A1 US 20070026485 A1 US20070026485 A1 US 20070026485A1 US 55289604 A US55289604 A US 55289604A US 2007026485 A1 US2007026485 A1 US 2007026485A1
Authority
US
United States
Prior art keywords
peptide
glycan
glycans
linked
glycosyl
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
US10/552,896
Other languages
English (en)
Inventor
Shawn DeFrees
David Zopf
Robert Bayer
David Hakes
Caryn Bowe
Xi Chen
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
Neose Technologies Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US10/410,997 external-priority patent/US7157277B2/en
Priority claimed from US10/410,980 external-priority patent/US7399613B2/en
Priority claimed from US10/411,012 external-priority patent/US7265084B2/en
Priority claimed from US10/410,945 external-priority patent/US7214660B2/en
Priority claimed from US10/411,049 external-priority patent/US7297511B2/en
Priority claimed from US10/410,930 external-priority patent/US7226903B2/en
Priority claimed from US10/410,897 external-priority patent/US7179617B2/en
Priority claimed from US10/410,913 external-priority patent/US7265085B2/en
Priority claimed from US10/410,962 external-priority patent/US7173003B2/en
Priority claimed from US10/411,044 external-priority patent/US8008252B2/en
Priority claimed from US10/411,026 external-priority patent/US7795210B2/en
Priority claimed from US10/411,043 external-priority patent/US7439043B2/en
Priority claimed from US10/411,037 external-priority patent/US7125843B2/en
Priority to US10/552,896 priority Critical patent/US20070026485A1/en
Application filed by Neose Technologies Inc filed Critical Neose Technologies Inc
Assigned to NEOSE TECHNOLOGIES, INC. reassignment NEOSE TECHNOLOGIES, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BAYER, ROBERT J., CHEN, XI, BOWE, CARYN, DEFREES, SHAWN, HAKES, DAVID JAMES, ZOPF, DAVID A.
Publication of US20070026485A1 publication Critical patent/US20070026485A1/en
Assigned to NOVO NORDISK A/S reassignment NOVO NORDISK A/S ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: NEOSE TECHNOLOGIES, INC.
Priority to US12/496,595 priority patent/US8063015B2/en
Priority to US13/246,512 priority patent/US8853161B2/en
Priority to US13/622,177 priority patent/US20130137157A1/en
Priority to US13/897,529 priority patent/US20130344050A1/en
Priority to US14/052,442 priority patent/US8791070B2/en
Priority to US14/246,519 priority patent/US20140294762A1/en
Priority to US14/721,761 priority patent/US20150343080A1/en
Priority to US15/225,819 priority patent/US20170007712A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/48Hydrolases (3) acting on peptide bonds (3.4)
    • C12N9/50Proteinases, e.g. Endopeptidases (3.4.21-3.4.25)
    • C12N9/64Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue
    • C12N9/6421Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue from mammals
    • C12N9/6424Serine endopeptidases (3.4.21)
    • C12N9/6437Coagulation factor VIIa (3.4.21.21)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/177Receptors; Cell surface antigens; Cell surface determinants
    • A61K38/1793Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/1816Erythropoietin [EPO]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/18Growth factors; Growth regulators
    • A61K38/185Nerve growth factor [NGF]; Brain derived neurotrophic factor [BDNF]; Ciliary neurotrophic factor [CNTF]; Glial derived neurotrophic factor [GDNF]; Neurotrophins, e.g. NT-3
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/193Colony stimulating factors [CSF]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/21Interferons [IFN]
    • A61K38/212IFN-alpha
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/21Interferons [IFN]
    • A61K38/215IFN-beta
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/22Hormones
    • A61K38/24Follicle-stimulating hormone [FSH]; Chorionic gonadotropins, e.g. HCG; Luteinising hormone [LH]; Thyroid-stimulating hormone [TSH]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/36Blood coagulation or fibrinolysis factors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/40Transferrins, e.g. lactoferrins, ovotransferrins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/465Hydrolases (3) acting on ester bonds (3.1), e.g. lipases, ribonucleases
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/47Hydrolases (3) acting on glycosyl compounds (3.2), e.g. cellulases, lactases
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/48Hydrolases (3) acting on peptide bonds (3.4)
    • A61K38/482Serine endopeptidases (3.4.21)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/48Hydrolases (3) acting on peptide bonds (3.4)
    • A61K38/482Serine endopeptidases (3.4.21)
    • A61K38/4846Factor VII (3.4.21.21); Factor IX (3.4.21.22); Factor Xa (3.4.21.6); Factor XI (3.4.21.27); Factor XII (3.4.21.38)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/55Protease inhibitors
    • A61K38/57Protease inhibitors from animals; from humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • 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
    • A61K47/54Medicinal 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 compound
    • A61K47/549Sugars, nucleosides, nucleotides or nucleic acids
    • 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
    • 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
    • A61K47/61Medicinal 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 the organic macromolecular compound being a polysaccharide or a derivative thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/10Drugs for disorders of the endocrine system of the posterior pituitary hormones, e.g. oxytocin, ADH
    • 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
    • A61P7/04Antihaemorrhagics; Procoagulants; Haemostatic agents; Antifibrinolytic agents
    • 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
    • C07K1/107General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length by chemical modification of precursor peptides
    • C07K1/1072General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length by chemical modification of precursor peptides by covalent attachment of residues or functional groups
    • C07K1/1077General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length by chemical modification of precursor peptides by covalent attachment of residues or functional groups by covalent attachment of residues other than amino acids or peptide residues, e.g. sugars, polyols, fatty acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/24Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against cytokines, lymphokines or interferons
    • C07K16/241Tumor Necrosis Factors
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/02Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing at least one abnormal peptide link
    • C07K5/0215Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing at least one abnormal peptide link containing natural amino acids, forming a peptide bond via their side chain functional group, e.g. epsilon-Lys, gamma-Glu
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/10Tetrapeptides
    • C07K5/1002Tetrapeptides with the first amino acid being neutral
    • C07K5/1005Tetrapeptides with the first amino acid being neutral and aliphatic
    • C07K5/1013Tetrapeptides with the first amino acid being neutral and aliphatic the side chain containing O or S as heteroatoms, e.g. Cys, Ser
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/10Tetrapeptides
    • C07K5/1002Tetrapeptides with the first amino acid being neutral
    • C07K5/1016Tetrapeptides with the first amino acid being neutral and aromatic or cycloaliphatic
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K9/00Peptides having up to 20 amino acids, containing saccharide radicals and having a fully defined sequence; Derivatives thereof
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K9/00Peptides having up to 20 amino acids, containing saccharide radicals and having a fully defined sequence; Derivatives thereof
    • C07K9/006Peptides having up to 20 amino acids, containing saccharide radicals and having a fully defined sequence; Derivatives thereof the peptide sequence being part of a ring structure
    • C07K9/008Peptides having up to 20 amino acids, containing saccharide radicals and having a fully defined sequence; Derivatives thereof the peptide sequence being part of a ring structure directly attached to a hetero atom of the saccharide radical, e.g. actaplanin, avoparcin, ristomycin, vancomycin
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/48Hydrolases (3) acting on peptide bonds (3.4)
    • C12N9/50Proteinases, e.g. Endopeptidases (3.4.21-3.4.25)
    • C12N9/64Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue
    • C12N9/6421Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue from mammals
    • C12N9/6424Serine endopeptidases (3.4.21)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/48Hydrolases (3) acting on peptide bonds (3.4)
    • C12N9/50Proteinases, e.g. Endopeptidases (3.4.21-3.4.25)
    • C12N9/64Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue
    • C12N9/6421Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue from mammals
    • C12N9/6424Serine endopeptidases (3.4.21)
    • C12N9/644Coagulation factor IXa (3.4.21.22)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/48Hydrolases (3) acting on peptide bonds (3.4)
    • C12N9/50Proteinases, e.g. Endopeptidases (3.4.21-3.4.25)
    • C12N9/64Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue
    • C12N9/6421Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue from mammals
    • C12N9/6424Serine endopeptidases (3.4.21)
    • C12N9/6456Plasminogen activators
    • C12N9/6459Plasminogen activators t-plasminogen activator (3.4.21.68), i.e. tPA
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/96Stabilising an enzyme by forming an adduct or a composition; Forming enzyme conjugates
    • 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y302/00Hydrolases acting on glycosyl compounds, i.e. glycosylases (3.2)
    • C12Y302/01Glycosidases, i.e. enzymes hydrolysing O- and S-glycosyl compounds (3.2.1)
    • C12Y302/01045Glucosylceramidase (3.2.1.45), i.e. beta-glucocerebrosidase
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y304/00Hydrolases acting on peptide bonds, i.e. peptidases (3.4)
    • C12Y304/21Serine endopeptidases (3.4.21)
    • C12Y304/21021Coagulation factor VIIa (3.4.21.21)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y304/00Hydrolases acting on peptide bonds, i.e. peptidases (3.4)
    • C12Y304/21Serine endopeptidases (3.4.21)
    • C12Y304/21022Coagulation factor IXa (3.4.21.22)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y304/00Hydrolases acting on peptide bonds, i.e. peptidases (3.4)
    • C12Y304/21Serine endopeptidases (3.4.21)
    • C12Y304/21068Tissue plasminogen activator (3.4.21.68), i.e. tPA
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/40Immunoglobulins specific features characterized by post-translational modification
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/40Immunoglobulins specific features characterized by post-translational modification
    • C07K2317/41Glycosylation, sialylation, or fucosylation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/52Constant or Fc region; Isotype
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y301/00Hydrolases acting on ester bonds (3.1)
    • C12Y301/27Endoribonucleases producing 3'-phosphomonoesters (3.1.27)
    • C12Y301/27005Pancreatic ribonuclease (3.1.27.5)

Definitions

  • peptides Most naturally occurring peptides contain carbohydrate moieties attached to the peptide via specific linkages to a select number of amino acids along the length of the primary peptide chain. Thus, many naturally occurring peptides are termed “glycopeptides.”
  • the variability of the glycosylation pattern on any given peptide has enormous implications for the function of that peptide.
  • the structure of the N-linked glycans on a peptide can impact various characteristics of the peptide, including the protease susceptibility, intracellular trafficking, secretion, tissue targeting, biological half-life and antigenicity of the peptide in a cell or organism. The alteration of one or more of these characteristics greatly affects the efficacy of a peptide in its natural setting, and also affects the efficacy of the peptide as a therapeutic agent in situations where the peptide has been generated for that purpose.
  • the carbohydrate structure attached to the peptide chain is known as a “glycan” molecule.
  • the specific glycan structure present on a peptide affects the solubility and aggregation characteristics of the peptide, the folding of the primary peptide chain and therefore its functional or enzymatic activity, the resistance of the peptide to proteolytic attack and the control of proteolysis leading to the conversion of inactive forms of the peptide to active forms.
  • terminal sialic acid residues present on the glycan molecule affect the length of the half life of the peptide in the mammalian circulatory system. Peptides whose glycans do not contain terminal sialic acid residues are rapidly removed from the circulation by the liver, an event which negates any potential therapeutic benefit of the peptide.
  • the glycan structures found in naturally occurring glycopeptides are typically divided into two classes, N-linked and O-linked glycans.
  • Peptides expressed in eukaryotic cells are typically N-glycosylated on asparagine residues at sites in the peptide primary structure containing the sequence asparagine-X-serine/threonine where X can be any amino acid except proline and aspartic acid.
  • the carbohydrate portion of such peptides is known as an N-linked glyean.
  • the early events of N-glycosylation occur in the endoplasmic reticulum (ER) and are identical in mammals, plants, insects and other higher eukaryotes.
  • ER endoplasmic reticulum
  • an oligosaccharide chain comprising fourteen sugar residues is constructed on a lipid carrier molecule.
  • the entire oligosaccharide chain is transferred to the amide group of the asparagine residue in a reaction catalyzed by a membrane bound glycosyltransferase enzyme.
  • the N-linked glycan is further processed both in the ER and in the Golgi apparatus.
  • the further processing generally entails removal of some of the sugar residues and addition of other sugar residues in reactions catalyzed by glycosidases and glycosyltransferases specific for the sugar residues removed and added.
  • the final structures of the N-linked glycans are dependent upon the organism in which the peptide is produced.
  • peptides produced in bacteria are completely unglycosylated.
  • Peptides expressed in insect cells contain high mannose and paunci-mannose N-linked oligosaccharide chains, among others.
  • Peptides produced in mammalian cell culture are usually glycosylated differently depending, e.g., upon the species and cell culture conditions. Even in the same species and under the same conditions, a certain amount of heterogeneity in the glycosyl chains is sometimes encountered.
  • peptides produced in plant cells comprise glycan structures that differ significantly from those produced in animal cells.
  • O-linked glycans also called mucin-type glycans because of their prevalence on mucinous glycopeptide.
  • O-glycans are linked primarily to serine and threonine residues and are formed by the stepwise addition of sugars from nucleotide sugars (Tanner et al., Biochim. Biophys. Acta. 906:81-91 (1987); and Hounsell et al., Glycoconj. J. 13:19-26 (1996)).
  • Peptide function can be affected by the structure of the O-linked glycans present thereon.
  • the activity of P-selectin ligand is affected by the O-linked glycan structure present thereon.
  • O-linked glycan structures see Schachter and Brockhausen, The Biosynthesis of Branched O-Linked Glycans, 1989, Society for Experimental Biology, pp. 1-26 (Great Britain).
  • Other glycosylation patterns are formed by linking glycosylphosphatidylinositol to the carboxyl-terminal carboxyl group of the protein (Takeda et al., Trends Biochem. Sci. 20:367-371 (1995); and Udenfriend et al., Ann. Rev. Biochem. 64:593-591 (1995).
  • glycosylated and non-glycosylated peptides for engendering a particular physiological response is well known in the medicinal arts.
  • insulin which is used to treat diabetes.
  • Enzymes have also been used for their therapeutic benefits.
  • a major factor, which has limited the use of therapeutic peptides is the immunogenic nature of most peptides. In a patient, an immunogenic response to an administered peptide can neutralize the peptide and/or lead to the development of an allergic response in the patient.
  • Other deficiencies of therapeutic peptides include suboptimal potency and rapid clearance rates.
  • the problems inherent in peptide therapeutics are recognized in the art, and various methods of eliminating the problems have been investigated.
  • synthetic polymers To provide soluble peptide therapeutics, synthetic polymers have been attached to the peptide backbone.
  • PEG Poly(ethylene glycol)
  • PEG Poly(ethylene glycol)
  • the use of PEG to derivatize peptide therapeutics has been demonstrated to reduce the immunogenicity of the peptides and prolong the clearance time from the circulation.
  • U.S. Pat. No. 4,179,337 (Davis et al.) concerns non-immunogenic peptides, such as enzymes and peptide hormones coupled to polyethylene glycol (PEG) or polypropylene glycol. Between 10 and 100 moles of polymer are used per mole peptide and at least 15% of the physiological activity is maintained.
  • WO 93/15189 (Veronese et al.) concerns a method to maintain the activity of polyethylene glycol-modified proteolytic enzymes by linking the proteolytic enzyme to a macromolecularized inhibitor.
  • the conjugates are intended for medical applications.
  • the principal mode of attachment of PEG, and its derivatives, to peptides is a non-specific bonding through a peptide amino acid residue.
  • U.S. Pat. No. 4,088,538 discloses an enzymatically active polymer-enzyme conjugate of an enzyme covalently linked to PEG.
  • U.S. Pat. No. 4,496,689 discloses a covalently attached complex of ⁇ -1 protease inhibitor with a polymer such as PEG or methoxypoly(ethylene glycol) (“mPEG”).
  • mPEG methoxypoly(ethylene glycol)
  • Pat. No. 4,414,147 discloses a method of rendering interferon less hydrophobic by conjugating it to an anhydride of a dicarboxylic acid, such as poly(ethylene succinic anhydride).
  • PCT WO 87/00056 discloses conjugation of PEG and poly(oxyethylated) polyols to such proteins as interferon- ⁇ , interleukin-2 and immunotoxins.
  • EP 154,316 discloses and claims chemically modified lymphokines, such as IL-2 containing PEG bonded directly to at least one primary amino group of the lymphokine.
  • U.S. Pat. No. 4,055,635 discloses pharmaceutical compositions of a water-soluble complex of a proteolytic enzyme linked covalently to a polymeric substance such as a polysaccharide.
  • Another mode of attaching PEG to peptides is through the non-specific oxidation of glycosyl residues on a peptide.
  • the oxidized sugar is utilized as a locus for attaching a PEG moiety to the peptide.
  • M'Timkulu discloses the use of a hydrazine- or amino-PEG to add PEG to a glycoprotein.
  • the glycosyl moieties are randomly oxidized to the corresponding aldehydes, which are subsequently coupled to the amino-PEG.
  • Bona et al. WO 96/40731
  • a PEG is added to an immunoglobulin molecule by enzymatically oxidizing a glycan on the immunoglobulin and then contacting the glycan with an amino-PEG molecule.
  • poly(ethylene glycol) is added in a random, non-specific manner to reactive residues on a peptide backbone.
  • a derivatization strategy that results in the formation of a specifically labeled, readily characterizable, essentially homogeneous product.
  • glycosyltransferases e.g., sialyltransferases, oligosaccharyltransferases, N-acetylglucosaminyltransferases
  • glycosidases are further classified as exoglycosidases (e.g., ⁇ -mannosidase, ⁇ -glucosidase), and endoglycosidases (e.g., Endo-A, Endo-M).
  • exoglycosidases e.g., ⁇ -mannosidase, ⁇ -glucosidase
  • endoglycosidases e.g., Endo-A, Endo-M
  • Glycosyltransferases modify the oligosaccharide structures on peptides. Glycosyltransferases are effective for producing specific products with good stereochemical and regiochemical control. Glycosyltransferases have been used to prepare oligosaccharides and to modify terminal N— and O-linked carbohydrate structures, particularly on peptides produced in mammalian cells. For example, the terminal oligosaccharides of glycopeptides have been completely sialylated and/or fucosylated to provide more consistent sugar structures, which improves glycopeptide pharmacodynamics and a variety of other biological properties.
  • ⁇ -1,4-galactosyltransferase is used to synthesize lactosamine, an illustration of the utility of glycosyltransferases in the synthesis of carbohydrates (see, e.g., Wong et al., J. Org Chem. 47: 5416-5418 (1982)).
  • numerous synthetic procedures have made use of ⁇ -sialyltransferases to transfer sialic acid from cytidine-5′-monophospho-N-acetylneuraminic acid to the 3-OH or 6-OH of galactose (see, e.g., Kevin et al., Chem. Eur. J. 2: 1359-1362 (1996)).
  • Fucosyltransferases are used in synthetic pathways to transfer a fucose unit from guanosine-5′-diphosphofucose to a specific hydroxyl of a saccharide acceptor.
  • Ichikawa prepared sialyl Lewis-X by a method that involves the fucosylation of sialylated lactosamine with a cloned facosyltransferase (Ichikawa et al., J. Am. Chem. Soc. 114: 9283-9298 (1992)).
  • Koeller et al. Nature Biotechnology 18: 835-841 (2000). See also, U.S. Pat. Nos. 5,876,980; 6,030,815; 5,728,554; 5,922,577; and WO/9831826.
  • Glycosidases can also be used to prepare saccharides. Glycosidases normally catalyze the hydrolysis of a glycosidic bond. However, under appropriate conditions, they can be used to form this linkage. Most glycosidases used for carbohydrate synthesis are exoglycosidases; the glycosyl transfer occurs at the non-reducing terminus of the substrate. The glycosidase binds a glycosyl donor in a glycosyl-enzyme intermediate that is either intercepted by water to yield the hydrolysis product, or by an acceptor, to generate a new glycoside or oligosaccharide.
  • An exemplary pathway using an exoglycosidase is the synthesis of the core trisaccharide of all N-linked glycopeptides, including the ⁇ -mannoside linkage, which is formed by the action of ⁇ -mannosidase (Singh et al., Chem. Commun. 993-994 (1996)).
  • a mutant glycosidase has been prepared in which the normal nucleophilic amino acid within the active site is changed to a non-nucleophilic amino acid.
  • the mutant enzyme does not hydrolyze glycosidic linkages, but can still form them.
  • Such a mutant glycosidase is used to prepare oligosaccharides using an ⁇ -glycosyl fluoride donor and a glycoside acceptor molecule (Withers et al., U.S. Pat. No. 5,716,812).
  • endoglycosidases are also utilized to prepare carbohydrates. Methods based on the use of endoglycosidases have the advantage that an oligosaccharide, rather than a monosaccharide, is transferred. Oligosaccharide fragments have been added to substrates using endo- ⁇ -N-acetylglucosamines such as endo-F, endo-M (Wang et al., Tetrahedron Lett. 37: 1975-1978); and Haneda et al., Carbohydr. Res. 292: 61-70 (1996)).
  • Brossmer et al. (U.S. Pat. No. 5,405,753) discloses the formation of a fluorescent-labeled cytidine monophosphate (“CMP”) derivative of sialic acid and the use of the fluorescent glycoside in an assay for sialyl transferase activity and for the fluorescent-labeling of cell surfaces, glycoproteins and peptides.
  • CMP cytidine monophosphate
  • Gross et al. Analyt. Biochem. 186: 127 (1990) describe a similar assay.
  • Bean et al. (U.S. Pat. No.
  • 5,432,059 discloses an assay for glycosylation deficiency disorders utilizing reglycosylation of a deficiently glycosylated protein.
  • the deficient protein is reglycosylated with a fluorescent-labeled CMP glycoside.
  • Each of the fluorescent sialic acid derivatives is substituted with the fluorescent moiety at either the 9-position or at the amine that is normally acetylated in sialic acid.
  • the methods using the fluorescent sialic acid derivatives are assays for the presence of glycosyltransferases or for non-glycosylated or improperly glycosylated glycoproteins.
  • the assays are conducted on small amounts of enzyme or glycoprotein in a sample of biological origin.
  • the enzymatic derivatization of a glycosylated or non-glycosylated peptide on a preparative or industrial scale using a modified sialic acid has not been disclosed or suggested in the prior art.
  • Enzymatic methods have also been used to activate glycosyl residues on a glycopeptide towards subsequent chemical elaboration.
  • the glycosyl residues are typically activated using galactose oxidase, which converts a terminal galactose residue to the corresponding aldehyde.
  • the aldehyde is subsequently coupled to an amine-containing modifying group.
  • Casares et al. ( Nature Biotech. 19: 142 (2001)) have attached doxorubicin to the oxidized galactose residues of a recombinant MHCII-peptide chimera.
  • Glycosyl residues have also been modified to contain ketone groups.
  • Mahal and co-workers Science 276: 1125 (1997) have prepared N-levulinoyl mannosamine (“ManLev”), which has a ketone functionality at the position normally occupied by the acetyl group in the natural substrate. Cells were treated with the ManLev, thereby incorporating a ketone group onto the cell surface. See, also Saxon et al., Science 287: 2007 (2000); Hang et al., J. Am. Chem. Soc. 123: 1242 (2001); Yarema et al., J. Biol. Chem. 273: 31168 (1998); and Charter et al., Glycobiology 10: 1049 (2000).
  • the methods of modifying cell surfaces have not been applied in the absence of a cell to modify a glycosylated or non-glycosylated peptide. Further, the methods of cell surface modification are not utilized for the enzymatic incorporation preformed modified glycosyl donor moiety into a peptide. Moreover, none of the cell surface modification methods are practical for producing glycosyl-modified peptides on an industrial scale.
  • the invention includes a multitude of methods of remodeling a peptide to have a specific glycan structure attached thereto. Although specific glycan structures are described herein, the invention should not be construed to be limited to any one particular structure. In addition, although specific peptides are described herein, the invention should not be limited by the nature of the peptide described, but rather should encompass any and all suitable peptides and variations thereof.
  • the invention includes a cell-free, in vitro method of remodeling a peptide comprising poly(ethylene glycol), the peptide having the formula: wherein
  • AA is a terminal or internal amino acid residue of the peptide
  • X 1 —X 2 is a saccharide covalently linked to the AA, wherein
  • X 1 is a first glycosyl residue
  • X 2 is a second glycosyl residue covalently linked to X 1 , wherein X 1 and
  • the invention further comprises formation of a truncated glycan by removing a Sia residue.
  • a peptide has the formula: wherein
  • an oligosaccharyl residue is a member selected from GlcNAc-Gal-Sia and GlcNAc-Gal.
  • at least one oligosaccharide member is selected from a, b, c, d, e and x is 1 or 2.
  • the removing of step (a) produces a truncated glycan in which at least one of a, b, c, e and x are 0.
  • the invention includes a method of remodeling a peptide wherein X 3 , X 5 and X 7 are members independently selected from (mannose) z and (mannose) z -(X 8 )
  • X 8 is a glycosyl moiety selected from mono- and oligo-saccharides.
  • z is an integer between 1 and 20, wherein
  • each (mannose) z is independently selected from linear and branched structures.
  • X 4 is selected from the group consisting of GlcNAc and xylose.
  • X 3 , X 5 and X 7 are (mannose) u , wherein u is selected from the integers between 1 and 20, and when u is 3 or greater, each (mannose) u is independently selected from linear and branched structures.
  • the invention also includes a method of remodeling a peptide, wherein the peptide has the formula: wherein
  • a peptide has the formula: wherein
  • X 9 and X 10 are independently selected monosaccharyl or oligosaccharyl residues and m, n and f are integers independently selected from 0 and 1.
  • a peptide has the formula: wherein
  • X 16 is a member selected from: wherein
  • s and i are integers independently selected from 0 and 1.
  • a peptide has the formula: wherein
  • X 13 , X 14 , and X 15 are independently selected glycosyl residues.
  • g, h, i, j, k, and p are independently selected from the integers 0 and 1.
  • At least one of g, h, i, j, k and p is 1.
  • X 14 and X 15 are members independently selected from GlcNAc and Sia and i and k are independently selected from the integers 0 and 1.
  • at least one of i and k is 1, and if k is 1, g, h, and j are 0.
  • the invention also includes a method of remodeling a peptide, wherein the method comprises contacting the truncated glycan with at least one glycosyltransferase and at least one glycosyl donor under conditions suitable to transfer the at least one glycosyl donor to the truncated glycan, thereby remodeling the peptide comprising poly(ethylene glycol).
  • a glycosyl donor comprises a modifying group covalently linked thereto.
  • the invention also includes a method of remodeling a peptide, the method comprising removing X 1 , thereby exposing AA.
  • a method includes contacting AA with at least one glycosyltransferase and at least one glycosyl donor under conditions suitable to transfer said at least one glycosyl donor to AA, thereby remodeling said peptide comprising poly(ethylene glycol).
  • At least one glycosyl donor comprises a modifying group covalently linked thereto.
  • a modifying group is poly(ethylene glycol).
  • a poly(ethylene glycol) has a molecular weight distribution that is essentially homodisperse.
  • the invention includes a method of remodeling a peptide, wherein, prior to contacting the truncated glycan with at least one glycosyltransferase and at least one glycosyl donor under conditions suitable to transfer the at least one glycosyl donor to the truncated glycan, thereby remodeling the peptide comprising poly(ethylene glycol), a group added to the saccharide during post-translational modification is removed.
  • a removed group is a member selected from phosphate, sulfate, carboxylate and esters thereof.
  • the invention includes a method of remodeling a peptide wherein a peptide has the formula: wherein
  • Z is a member selected from O, S, NH and a cross-linker.
  • the invention also includes a method of remodeling a peptide, wherein the peptide has the formula: wherein
  • X 11 and X 12 are independently selected glycosyl moieties
  • r and x are integers independently selected from 0 and 1.
  • X 11 and X 12 are (mannose) q , wherein q is selected from the integers between 1 and 20, and when q is three or greater, (mannose) q is selected from linear and branched structures.
  • the invention includes a pharmaceutical composition comprising a pharmaceutically acceptable diluent and a remodeled peptide according to a cell-free, in vitro method of remodeling a peptide comprising poly(ethylene glycol), the peptide having the formula: wherein
  • AA is a terminal or internal amino acid residue of the peptide
  • X 1 —X 2 is a saccharide covalently linked to the AA, wherein
  • X 1 is a first glycosyl residue
  • X 2 is a second glycosyl residue covalently linked to X 1 , wherein X 1 and X 2 are selected from monosaccharyl and oligosaccharyl residues;
  • the invention also includes a cell-free, in vitro method of remodeling a peptide comprising poly(ethylene glycol), the peptide having the formula: wherein
  • At least one glycosyl donor comprises a modifying group covalently linked thereto.
  • the modifying group is poly(ethylene glycol).
  • the poly(ethylene glycol) has a molecular weight distribution that is essentially homodisperse.
  • the invention also includes a pharmaceutical composition comprising a pharmaceutically acceptable diluent and a remodeled peptide according to a cell-free, in vitro method of remodeling a peptide comprising poly(ethylene glycol), the peptide having the formula: wherein
  • FIG. 1 is a scheme depicting a trimannosyl core glycan (left side) and the enzymatic process for the generation of a glycan having a bisecting GlcNAc (right side).
  • FIG. 2 is a scheme depicting an elemental trimannosyl core structure and complex chains in various degrees of completion.
  • the in vitro enzymatic generation of an elemental trimannosyl core structure from a complex carbohydrate glycan structure which does not contain a bisecting GlcNAc residue is shown, as is the generation of a glyean structure therefrom which contains a bisecting GlcNAc.
  • FIG. 3 is a scheme for the enzymatic generation of a sialylated glycan structure (right side) beginning with a glycan having a trimannosyl core and a bisecting GlcNAc (left side).
  • FIG. 4 is a scheme of a typical high mannose containing glycan structure (left side) and the enzymatic process for reduction of this structure to an elemental trimannosyl core structure.
  • X is mannose as a monosaccharide, an oligosaccharide or a polysaccharide.
  • FIG. 5 is a diagram of a fucose and xylose containing N-linked glycan structure typically produced in plant cells.
  • FIG. 6 is a diagram of a fucose containing N-linked glycan structure typically produced in insect cells. Note that the glycan may have no core fucose, it amy have a single core fucose with either linkage, or it may have a single core fucose having a preponderance of one linkage.
  • FIG. 7 is a scheme depicting a variety of pathways for the trimming of a high mannose structure and the synthesis of complex sugar chains therefrom. Symbols: squares: GlcNAc; circles: Man; diamonds: fucose; pentagon: xylose.
  • FIG. 8 is a scheme depicting in vitro strategies for the synthesis of complex structures from an elemental trimannosyl core structure. Symbols: Squares: GlcNAc; light circles: Man; dark circles: Gal; dark triangles: NeuAc; GnT: N-acetyl glucosaminyltransferase; GalT: galactosyltransferase; ST: sialyltransferase.
  • FIG. 9 is a scheme depicting two in vitro strategies for the synthesis of monoantennary glycans, and the optional glycoPEGylation of the same. Dark squares: GlcNAc; dark circles: Man; light circles: Gal; dark triangles: sialic acid.
  • FIG. 10 is a scheme depicting two in vitro strategies for the synthesis of monoantennary glycans, and the optional glycoPEGylation of the same. Dark squares: GlcNAc; dark circles: Man; light circles: Gal; dark triangles: sialic acid.
  • FIG. 11 is a scheme depicting various complex structures, which may be synthesized from an elemental trimannosyl core structure. Symbols: Squares: GlcNAc; light circles: Man; dark circles: Gal; triangles: NeuAc; diamonds: fucose; FT and FucT: fucosyltransferase; GalT: galactosyltransferase; ST: sialyltransferase; Le: Lewis antigen; SLe: sialylated Lewis antigen.
  • FIG. 12 is an exemplary scheme for preparing O-inked glycopeptides originating with serine or threonine.
  • a water soluble polymer such as poly(ethylene glycol) is added to the final glycan structure.
  • FIG. 13 is a series of diagrams depicting the four types of O-glycan structures, termed cores 1 through 4.
  • the core structure is outlined in dotted lines.
  • FIG. 14 is a series of schemes showing an exemplary embodiment of the invention in which carbohydrate residues comprising complex carbohydrate structures and/or high mannose high mannose structures are trimmed back to the first generation biantennary structure.
  • fucose is added only after reaction with GnT I.
  • a modified sugar bearing a water-soluble polymer (WSP) is then conjugated to one or more of the sugar residues exposed by the trimming back process.
  • FIG. 15 is a scheme similar to that shown in FIG. 4 , in which a high mannose or complex structure is “trimmed back” to the mannose beta-linked core and a modified sugar bearing a water soluble polymer is then conjugated to one or more of the sugar residues exposed by the trimming back process.
  • Sugars are added sequentially using glycosyltransferases.
  • FIG. 16 is a scheme similar to that shown in FIG. 4 , in which a high mannose or complex structure is trimmed back to the GlcNAc to which the first mannose is attached, and a modified sugar bearing a water soluble polymer is then conjugated to one or more of the sugar residues exposed by the trimming back process.
  • Sugars are added sequentially using glycosyltransferases.
  • FIG. 17 is a scheme similar to that shown in FIG. 4 , in which a high mannose or cpomplex structure is trimmed back to the first GlcNAc attached to the Asn of the peptide, following which a water soluble polymer is conjugated to one or more sugar residues which have subsequently been added on. Sugars are added sequentially using glycosyltransferases.
  • FIG. 18 is a scheme in which an N-linked carbohydrate is optionally trimmed back from a high mannose or cpmplex structure, and subsequently derivatized with a modified sugar moiety (Gal or GlcNAc) bearing a water-soluble polymer.
  • a modified sugar moiety Gal or GlcNAc
  • FIG. 19 is a scheme in which an N-linked carbohydrate is trimmed back from a high mannose or complex structure and subsequently derivatized with a sialic acid moiety bearing a water-soluble polymer. Sugars are added sequentially using glycosyltransferases.
  • FIG. 20 is a scheme in which an N-linked carbohydrate is optionally trimmed back from a high mannose oor complex structure and subsequently derivatized with one or more sialic acid moieties, and terminated with a sialic acid derivatized with a water-soluble polymer.
  • Sugars are added sequentially using glycosyltransferases.
  • FIG. 21 is a scheme in which an O-inked saccharide is “trimmed back” and subsequently conjugated to a modified sugar bearing a water-soluble polymer.
  • the carbohydrate moiety is “trimmed back” to the first generation of the biantennary structure.
  • FIG. 22 is an exemplary scheme for trimming back the carbohydrate moiety of an O-linked glycopeptide to produce a mannose available for conjugation with a modified sugar having a water-soluble polymer attached thereto.
  • FIG. 23 is a series of exemplary schemes.
  • FIG. 23A is a scheme that illustrates addition of a PEGylated sugar, followed by the addition of a non-modified sugar.
  • FIG. 23B is a scheme that illustrates the addition of more that one kind of modified sugar onto one glycan.
  • FIG. 23C is a scheme that illustrates the addition of different modified sugars onto O-linked glycans and N-linked glycans.
  • FIG. 24 is a diagram of various methods of improving the therapeutic function of a peptide by glycan remodeling, including conjugation.
  • FIG. 25 is a set of schemes for glycan remodeling of a therapeutic peptide to treat Gaucher Disease.
  • FIG. 26 is a scheme for glycan remodeling to generate glycans having a terminal mannose-6-phosphate moiety.
  • FIG. 27 is a diagram illustrating the array of glycan structures found on CHO-produced glucocerebrosidase (CerezymeTM) after sialylation.
  • FIG. 28 is a list of peptides useful in the methods of the invention.
  • FIG. 29 comprising FIGS. 29A to 29 G, provides exemplary schemes for remodeling glycan structures on granulocyte colony stimulating factor (G-CSF).
  • G-CSF granulocyte colony stimulating factor
  • FIG. 29A is a diagram depicting the G-CSF peptide indicating the amino acid residue to which a glycan is bonded, and an exemplary glycan formula linked thereto.
  • FIG. 29B to 29 G are diagrams of contemplated remodeling steps of the glycan of the peptide in FIG. 29A based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 30 comprising FIGS. 30A to 30 EE sets forth exemplary schemes for remodeling glycan structures on interferon-alpha.
  • FIG. 30A is a diagram depicting the interferon-alpha isoform 14c peptide indicating the amino acid residue to which a glycan is bonded, and an exemplary glycan formula linked thereto.
  • FIG. 30B to 30 D are diagrams of contemplated remodeling steps of the glycan of the peptide in FIG. 30A based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 30A is a diagram depicting the interferon-alpha isoform 14c peptide indicating the amino acid residue to which a glycan is bonded, and an exemplary glycan formula linked thereto.
  • FIG. 30B to 30 D are diagrams of contemplated remodeling steps of the glycan of the peptide in FIG. 30A based on the type of cell the peptid
  • FIG. 30E is a diagram depicting the interferon-alpha isoform 14c peptide indicating the amino acid residue to which a glycan is linked, and an exemplary glycan formula linked thereto.
  • FIG. 30F to 30 N are diagrams of contemplated remodeling steps of the glycan of the peptide in FIG. 30E based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 30O is a diagram depicting the interferon-alpha isoform 2a or 2b peptides indicating the amino acid residue to which a glycan is linked, and an exemplary glycan formula linked thereto.
  • FIG. 30P to 30 W are diagrams of contemplated remodeling steps of the glycan of the peptide in FIG. 30O based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 30X is a diagram depicting the interferon-alpha-mucin fusion peptides indicating the residue(s) which is linked to glycans contemplated for remodeling, and exemplary glycan formulas linked thereto.
  • FIG. 30Y to 30 AA are diagrams of contemplated remodeling steps of the glycan of the peptides in FIG. 30X based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 30Y to 30 AA are diagrams of contemplated remodeling steps of the glycan of the peptides in FIG. 30X based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 30B B is a diagram depicting the interferon-alpha-mucin fusion peptides and interferon-alpha peptides indicating the residue(s) which bind to glycans contemplated for remodeling, and formulas for the glycans.
  • FIG. 30C C to 30 EE are diagrams of contemplated remodeling steps of the glycan of the peptides in FIG. 30B B based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 31 comprising FIGS. 31A to 31 S, sets forth exemplary schemes for remodeling glycan structures on interferon-beta.
  • FIG. 31A is a diagram depicting the interferon-beta peptide indicating the amino acid residue to which a glycan is linked, and an exemplary glycan formula linked thereto.
  • FIG. 31B to 31 O are diagrams of contemplated remodeling steps of the glycan of the peptide in FIG. 31A based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 31 is a diagram depicting the interferon-beta peptide indicating the amino acid residue to which a glycan is linked, and an exemplary glycan formula linked thereto.
  • FIG. 31B to 31 O are diagrams of contemplated remodeling steps of the glycan of the peptide in FIG. 31A based on the type of cell the peptide is expressed in and the desired remodeled
  • FIG. 31P is a diagram depicting the interferon-beta peptide indicating the amino acid residue to which a glycan is linked, and an exemplary glycan formula linked thereto.
  • FIG. 31Q to 31 S are diagrams of contemplated remodeling steps of the glycan of the peptide in FIG. 31P based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 32 comprising FIGS. 32A to 32 D, sets forth exemplary schemes for remodeling glycan structures on Factor VII and Factor VIIa.
  • FIG. 32A is a diagram depicting the Factor-VII and Factor-VIIa peptides A (solid line) and B (dotted line) indicating the residues which bind to glycans contemplated for remodeling, and the formulas for the glycans.
  • FIG. 32B to 32 D are diagrams of contemplated remodeling steps of the glycan of the peptide in FIG. 32A based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 33 comprising FIGS. 33A to 33 G, sets forth exemplary schemes for remodeling glycan structures on Factor IX.
  • FIG. 33A is a diagram depicting the Factor-IX peptide indicating residues which bind to glycans contemplated for remodeling, and formulas of the glycans.
  • FIG. 33B to 33 G are diagrams of contemplated remodeling steps of the glycan of the peptide in FIG. 33A based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 34 sets forth exemplary schemes for remodeling glycan structures on follicle stimulating hormone (FSH), comprising ⁇ and ⁇ subunits.
  • FIG. 34A is a diagram depicting the Follicle Stimulating Hormone peptides FSH ⁇ and FSH ⁇ indicating the residues which bind to glycans contemplated for remodeling, and exemplary glycan formulas linked thereto.
  • FIG. 34B to 34 J are diagrams of contemplated remodeling steps of the glycan of the peptides in FIG. 34A based on the type of cell the peptides are expressed in and the desired remodeled glycan structures.
  • FIG. 35 comprising FIGS. 35A to 35 AA, sets forth exemplary schemes for remodeling glycan structures on Erythropoietin (EPO).
  • FIG. 35A is a diagram depicting the EPO peptide indicating the residues which bind to glycans contemplated for remodeling, and formulas for the glycans.
  • FIG. 35B to 35 S are diagrams of contemplated remodeling steps of the glycan of the peptide in FIG. 35A based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 35 is a diagram depicting the EPO peptide indicating the residues which bind to glycans contemplated for remodeling, and formulas for the glycans.
  • FIG. 35B to 35 S are diagrams of contemplated remodeling steps of the glycan of the peptide in FIG. 35A based on the type of cell the peptide is expressed in and the desired remodeled g
  • 35T is a diagram depicting the EPO peptide indicating the residues which bind to glycans contemplated for remodeling, and formulas for the glycans.
  • FIG. 35U to 35 W are diagrams of contemplated remodeling steps of the glycan of the peptide in FIG. 35T based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 35X is a diagram depicting the EPO peptide indicating the residues which bind to glycans contemplated for remodeling, and formulas for the glycans.
  • FIG. 35Y to 35 AA are diagrams of contemplated remodeling steps of the glycan of the peptide in FIG. 35X based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 36 comprising FIGS. 36A to 36 K sets forth exemplary schemes for remodeling glycan structures on Granulocyte-Macrophage Colony Stimulating Factor (GM-CSF).
  • FIG. 36A is a diagram depicting the GM-CSF peptide indicating the residues which bind to glycans contemplated for remodeling, and formulas for the glycans.
  • FIG. 36B to 36 G are diagrams of contemplated remodeling steps of the glycan of the peptide in FIG. 36A based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 36 comprising FIGS. 36A to 36 K sets forth exemplary schemes for remodeling glycan structures on Granulocyte-Macrophage Colony Stimulating Factor (GM-CSF).
  • FIG. 36A is a diagram depicting the GM-CSF peptide indicating the residues which bind to glycans contemplated for remodeling, and
  • FIG. 36H is a diagram depicting the GM-CSF peptide indicating the residues which bind to glycans contemplated for remodeling, and formulas for the glycans.
  • FIG. 36I to 36 K are diagrams of contemplated remodeling steps of the glycan of the peptide in FIG. 36H based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 37 comprising FIGS. 37A to 37 N, sets forth exemplary schemes for remodeling glycan structures on interferon-gamma.
  • FIG. 37A is a diagram depicting an interferon-gamma peptide indicating the residues which bind to glycans contemplated for remodeling, and exemplary glycan formulas linked thereto.
  • FIG. 37B to 37 G are diagrams of contemplated remodeling steps of the peptide in FIG. 37A based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 37 is a diagram depicting an interferon-gamma peptide indicating the residues which bind to glycans contemplated for remodeling, and exemplary glycan formulas linked thereto.
  • FIG. 37B to 37 G are diagrams of contemplated remodeling steps of the peptide in FIG. 37A based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 37H is a diagram depicting an interferon-gamma peptide indicating the residues which bind to glycans contemplated for remodeling, and exemplary glycan formulas linked thereto.
  • FIG. 37I to 37 N are diagrams of contemplated remodeling steps of the peptide in FIG. 37H based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 38 sets forth exemplary schemes for remodeling glycan structures on ⁇ 1 -antitrypsin (ATT, or ⁇ -1 protease inhibitor).
  • FIG. 38A is a diagram depicting an AAT peptide indicating the residues which bind to glycans contemplated for remodeling, and exemplary glycan formulas linked thereto.
  • FIG. 38B to 38 F are diagrams of contemplated remodeling steps of the glycan of the peptide in FIG. 38A based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 38 is a diagram depicting an AAT peptide indicating the residues which bind to glycans contemplated for remodeling, and exemplary glycan formulas linked thereto.
  • FIG. 38B to 38 F are diagrams of contemplated remodeling steps of the glycan of the peptide in FIG. 38A based on the type of cell the peptide is expressed in and the desired remodeled
  • FIG. 38G is a diagram depicting an AAT peptide indicating the residues which bind to glycans contemplated for remodeling, and exemplary glycan formulas linked thereto.
  • FIG. 38H to 38 J are diagrams of contemplated remodeling steps of the peptide in FIG. 38G based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 38K is a diagram depicting an AAT peptide indicating the residues which bind to glycans contemplated for remodeling, and exemplary glycan formulas linked thereto.
  • FIG. 38L to 38 N are diagrams of contemplated remodeling steps of the peptide in FIG. 38K based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 39 comprising FIGS. 39A to 39 J sets forth exemplary schemes for remodeling glycan structures on glucocerebrosidase.
  • FIG. 39A is a diagram depicting the glucocerebrosidase peptide indicating the residues which bind to glyeans contemplated for remodeling, and exemplary glycan formulas linked thereto.
  • FIG. 39B to 39 F are diagrams of contemplated remodeling steps of the glycan of the peptide in FIG. 39A based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 39A is a diagram depicting the glucocerebrosidase peptide indicating the residues which bind to glyeans contemplated for remodeling, and exemplary glycan formulas linked thereto.
  • FIG. 39B to 39 F are diagrams of contemplated remodeling steps of the glycan of the peptide in FIG. 39A based on the type
  • FIG. 39G is a diagram depicting the glucocerebrosidase peptide indicating the residues which bind to glycans contemplated for remodeling, and exemplary glycan formulas linked thereto.
  • FIG. 39H to 39 K are diagrams of contemplated remodeling steps of the glycan of the peptide in FIG. 39G based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 40 comprising FIGS. 40A to 40 W, sets forth exemplary schemes for remodeling glycan structures on Tissue-Type Plasminogen Activator (TPA).
  • FIG. 40A is a diagram depicting the TPA peptide indicating the residues which bind to glyeans contemplated for remodeling, and formulas for the glycans.
  • FIG. 40B to 40 G are diagrams of contemplated remodeling steps of the peptide in FIG. 40A based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 40H is a diagram depicting the TPA peptide indicating the residues which bind to glycans contemplated for remodeling, and formulas for the glycans.
  • FIG. 40A is a diagram depicting the TPA peptide indicating the residues which bind to glycans contemplated for remodeling, and formulas for the glycans.
  • FIG. 40I to 40 K are diagrams of contemplated remodeling steps of the peptide in FIG. 40H based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 40L is a diagram depicting a mutant TPA peptide indicating the residues which bind to glycans contemplated for remodeling, and the formula for the glycans.
  • FIG. 40M to 40 O are diagrams of contemplated remodeling steps of the peptide in FIG. 40L based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 40M to 40 O are diagrams of contemplated remodeling steps of the peptide in FIG. 40L based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 40P is a diagram depicting a mutant TPA peptide indicating the residues which bind to glycans contemplated for remodeling, and formulas for the glycans.
  • FIG. 40Q to 40 S are diagrams of contemplated remodeling steps of the peptide in FIG. 40P based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 40T is a diagram depicting a mutant TPA peptide indicating the residues which links to glycans contemplated for remodeling, and formulas for the glycans.
  • FIG. 40U to 40 W are diagrams of contemplated remodeling steps of the peptide in FIG. 40T based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 41 comprising FIGS. 41A to 41 G, sets forth exemplary schemes for remodeling glycan structures on Interleukin-2 (IL-2).
  • FIG. 41A is a diagram depicting the Interleukin-2 peptide indicating the amino acid residue to which a glycan is linked, and an exemplary glycan formula linked thereto.
  • FIG. 41B to 41 G are diagrams of contemplated remodeling steps of the glycan of the peptide in FIG. 41A based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 42 comprising FIGS. 42A to 42 M, sets forth exemplary schemes for remodeling glycan structures on Factor VIII.
  • FIG. 42A are the formulas for the glycans that bind to the N-linked glycosylation sites (A and A′) and to the O-linked sites (B) of the Factor VIII peptides.
  • FIG. 42B to 42 F are diagrams of contemplated remodeling steps of the peptides in FIG. 42A based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 42A are the formulas for the glycans that bind to the N-linked glycosylation sites (A and A′) and to the O-linked sites (B) of the Factor VIII peptides.
  • FIG. 42B to 42 F are diagrams of contemplated remodeling steps of the peptides in FIG. 42A based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 42G are the formulas for the glycans that bind to the N-linked glycosylation sites (A and A′) and to the O-linked sites (B) of the Factor VIII peptides.
  • FIG. 42H to 42 M are diagrams of contemplated remodeling steps of the peptides in FIG. 42G based on the type of cell the peptide is expressed in and the desired remodeled glycan structures.
  • FIG. 43 comprising FIGS. 43A to 43 M, sets forth exemplary schemes for remodeling glycan structures on urokinase.
  • FIG. 43A is a diagram depicting the urokinase peptide indicating a residue which is linked to a glycan contemplated for remodeling, and an exemplary glycan formula linked thereto.
  • FIG. 43B to 43 F are diagrams of contemplated remodeling steps of the peptide in FIG. 43A based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 43A is a diagram depicting the urokinase peptide indicating a residue which is linked to a glycan contemplated for remodeling, and an exemplary glycan formula linked thereto.
  • FIG. 43B to 43 F are diagrams of contemplated remodeling steps of the peptide in FIG. 43A based on the type of cell the peptide is expressed in and the desired remodeled gly
  • FIG. 43G is a diagram depicting the urokinase peptide indicating a residue which is linked to a glycan contemplated for remodeling, and an exemplary glycan formula linked thereto.
  • FIG. 43H to 43 L are diagrams of contemplated remodeling steps of the peptide in FIG. 43G based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 44 comprising FIGS. 44A to 44 J, sets forth exemplary schemes for remodeling glycan structures on human DNase (hDNase).
  • FIG. 44A is a diagram depicting the human DNase peptide indicating the residues which bind to glycans contemplated for remodeling, and exemplary glycan formulas linked thereto.
  • FIG. 44B to 44 F are diagrams of contemplated remodeling steps of the peptide in FIG. 44A based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 44G is a diagram depicting the human DNase peptide indicating residues which bind to glycans contemplated for remodeling, and exemplary glycan formulas linked thereto.
  • FIG. 44H to 44 J are diagrams of contemplated remodeling steps of the peptide in FIG. 44F based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 45 comprising FIGS. 45A to 45 L, sets forth exemplary schemes for remodeling glycan structures on insulin.
  • FIG. 45A is a diagram depicting the insulin peptide mutated to contain an N glycosylation site and an exemplary glycan formula linked thereto.
  • FIG. 45B to 45 D are diagrams of contemplated remodeling steps of the peptide in FIG. 45A based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 45E is a diagram depicting insulin-mucin fusion peptides indicating a residue(s) which is linked to a glycan contemplated for remodeling, and an exemplary glycan formula linked thereto.
  • FIG. 45A is a diagram depicting the insulin peptide mutated to contain an N glycosylation site and an exemplary glycan formula linked thereto.
  • FIG. 45B to 45 D are diagrams of contemplated remodeling steps of the peptide in FIG. 45
  • FIG. 45F to 45 H are diagrams of contemplated remodeling steps of the peptide in FIG. 45E based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 45I is a diagram depicting the insulin-mucin fusion peptides and insulin peptides indicating a residue(s) which is linked to a glycan contemplated for remodeling, and formulas for the glycan.
  • FIG. 45J to 45 L are diagrams of contemplated remodeling steps of the peptide in FIG. 45I based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 46 comprising FIGS. 46A to 46 K, sets forth exemplary schemes for remodeling glycan structures on the M-antigen (preS and S) of the Hepatitis B surface protein (HbsAg).
  • FIG. 46A is a diagram depicting the M-antigen peptide indicating the residues which bind to glycans contemplated for remodeling, and formulas for the glycans.
  • FIG. 46B to 46 G are diagrams of contemplated remodeling steps of the peptide in FIG. 46A based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 46 is a diagram depicting the M-antigen peptide indicating the residues which bind to glycans contemplated for remodeling, and formulas for the glycans.
  • FIG. 46B to 46 G are diagrams of contemplated remodeling steps of the peptide in FIG. 46A based on the type of cell the peptide is expressed in and the desired
  • FIG. 46H is a diagram depicting the M-antigen peptide indicating the residues which bind to glycans contemplated for remodeling, and formulas for the glycans.
  • FIG. 46I to 46 K are diagrams of contemplated remodeling steps of the peptide in FIG. 46H based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 47 comprising FIGS. 47A to 47 K, sets forth exemplary schemes for remodeling glycan structures on human growth hormone, including N, V and variants thereof.
  • FIG. 47A is a diagram depicting the human growth hormone peptide indicating a residue which is linked to a glycan contemplated for remodeling, and an exemplary glycan formula linked thereto.
  • FIG. 47B to 47 D are diagrams of contemplated remodeling steps of the glycan of the peptide in FIG. 47A based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 47 is a diagram depicting the human growth hormone peptide indicating a residue which is linked to a glycan contemplated for remodeling, and an exemplary glycan formula linked thereto.
  • FIG. 47B to 47 D are diagrams of contemplated remodeling steps of the glycan of the peptide in FIG. 47A based on the type of cell the peptide is expressed in and
  • FIG. 47E is a diagram depicting the three fusion peptides comprising the human growth hormone peptide and part or all of a mucin peptide, and indicating a residue(s) which is linked to a glycan contemplated for remodeling, and exemplary glycan formula(s) linked thereto.
  • FIG. 47F to 47 K are diagrams of contemplated remodeling steps of the glycan of the peptides in FIG. 47E based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 48 comprising FIGS. 48A to 48 G, sets forth exemplary schemes for remodeling glycan structures on a TNF Receptor-IgG Fc region fusion protein (EnbrelTM).
  • FIG. 48A is a diagram depicting a TNF Receptor-IgG Fc region fusion peptide which may be mutated to contain additional N-glycosylation sites indicating the residues which bind to glycans contemplated for remodeling, and formulas for the glycans.
  • the TNF receptor peptide is depicted in bold line, and the IgG Fc regions is depicted in regular line.
  • FIG. 48B to 48 G are diagrams of contemplated remodeling steps of the peptide in FIG. 48A based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 49 comprising FIGS. 49A to 49 D, sets forth exemplary schemes for remodeling glycan structures on an anti-HER2 monoclonal antibody (HerceptinTM).
  • FIG. 49A is a diagram depicting an anti-HER2 monoclonal antibody which has been mutated to contain an N-glycosylation site(s) indicating a residue(s) on the antibody heavy chain which is linked to a glycan contemplated for remodeling, and an exemplary glycan formula linked thereto.
  • FIG. 49B to 49 D are diagrams of contemplated remodeling steps of the glycan of the peptides in FIG. 49A based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 50 comprising FIGS. 50A to 50 D, sets forth exemplary schemes for remodeling glycan structures on a monoclonal antibody to Protein F of Respiratory Syncytial Virus (SynagisTM).
  • FIG. 50A is a diagram depicting a monoclonal antibody to Protein F peptide which is mutated to contain an N-glycosylation site(s) indicating a residue(s) which is linked to a glyean contemplated for remodeling, and an exemplary glycan formula linked thereto.
  • FIG. 50B to 50 D are diagrams of contemplated remodeling steps of the peptide in FIG. 50A based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 51 comprising FIGS. 51A to 51 D, sets forth exemplary schemes for remodeling glycan structures on a monoclonal antibody to TNF- ⁇ (RemicadeTM).
  • FIG. 51A is a diagram depicting a monoclonal antibody to TNF- ⁇ which has an N-glycosylation site(s) indicating a residue which is linked to a glycan contemplated for remodeling, and an exemplary glycan formula linked thereto.
  • FIG. 51B to 51 D are diagrams of contemplated remodeling steps of the peptide in FIG. 51A based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 52 comprising FIGS. 52A to 52 L, sets forth exemplary schemes for remodeling glycan structures on a monoclonal antibody to glycoprotein IIb/IIIa (ReoproTM).
  • FIG. 52A is a diagram depicting a mutant monoclonal antibody to glycoprotein IIb/IIIa peptides which have been mutated to contain an N-glycosylation site(s) indicating the residue(s) which bind to glycans contemplated for remodeling, and exemplary glycan formulas linked thereto.
  • FIG. 52B to 52 D are diagrams of contemplated remodeling steps based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 52A is a diagram depicting a mutant monoclonal antibody to glycoprotein IIb/IIIa peptides which have been mutated to contain an N-glycosylation site(s) indicating the residue(s) which bind to glycans contemplated for remodeling, and exemplary
  • FIG. 52E is a diagram depicting monoclonal antibody to glycoprotein IIb/IIIa-mucin fusion peptides indicating the residues which bind to glycans contemplated for remodeling, and exemplary glycan formulas linked thereto.
  • FIG. 52F to 52 H are diagrams of contemplated remodeling steps based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 52I is a diagram depicting monoclonal antibody to glycoprotein IIb/IIIa-mucin fusion peptides and monoclonal antibody to glycoprotein IIb/IIIa peptides indicating the residues which bind to glycans contemplated for remodeling, and exemplary glycan formulas linked thereto.
  • FIG. 52J to 52 L are diagrams of contemplated remodeling steps based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 53 comprising FIGS. 53A to 53 G, sets forth exemplary schemes for remodeling glycan structures on a monoclonal antibody to CD20 (RituxanTM).
  • FIG. 53A is a diagram depicting monoclonal antibody to CD20 which have been mutated to contain an N-glycosylation site(s) indicating the residue which is linked to glycans contemplated for remodeling, and exemplary glycan formulas linked thereto.
  • FIG. 53B to 53 D are diagrams of contemplated remodeling steps of the glycan of the peptides in FIG. 53A based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 53 is a diagram depicting monoclonal antibody to CD20 which have been mutated to contain an N-glycosylation site(s) indicating the residue which is linked to glycans contemplated for remodeling, and exemplary glycan formulas linked thereto.
  • FIG. 53E is a diagram depicting monoclonal antibody to CD20 which has been mutated to contain an N-glycosylation site(s) indicating the residue(s) which is linked to glycans contemplated for remodeling, and exemplary glycan formulas linked thereto.
  • FIG. 53F to 53 G are diagrams of contemplated remodeling steps of the glycan of the peptides in FIG. 53E based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 54 comprising FIGS. 54A to 54 O, sets forth exemplary schemes for remodeling glycan structures on anti-thrombin III (AT III).
  • FIG. 54A is a diagram depicting the anti-thrombin III peptide indicating the amino acid residues to which an N-linked glycan is linked, and an exemplary glycan formula linked thereto.
  • FIG. 54B to 54 G are diagrams of contemplated remodeling steps of the glycan of the peptide in FIG. 54A based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 54 is a diagram depicting the anti-thrombin III peptide indicating the amino acid residues to which an N-linked glycan is linked, and an exemplary glycan formula linked thereto.
  • FIG. 54B to 54 G are diagrams of contemplated remodeling steps of the glycan of the peptide in FIG. 54A based on the type of cell the peptide is expressed in
  • FIG. 54H is a diagram depicting the anti-thrombin III peptide indicating the amino acid residues to which an N-linked glycan is linked, and an exemplary glycan formula linked thereto.
  • FIG. 54I to 54 K are diagrams of contemplated remodeling steps of the glycan of the peptide in FIG. 54H based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 54L is a diagram depicting the anti-thrombin III peptide indicating the amino acid residues to which an N-linked glycan is linked, and an exemplary glycan formula linked thereto.
  • FIG. 54M to 54 O are diagrams of contemplated remodeling steps of the glycan of the peptide in FIG. 54L based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 55 comprising FIGS. 55A to 55 J, sets forth exemplary schemes for remodeling glycan structures on subunits ⁇ and ⁇ of human Chorionic Gonadotropin (hCG).
  • FIG. 55A is a diagram depicting the hCG ⁇ and hCG ⁇ peptides indicating the residues which bind to N-linked glycans (A) and O-linked glycans (B) contemplated for remodeling, and formulas for the glycans.
  • FIG. 55B to 55 J are diagrams of contemplated remodeling steps based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 56 comprising FIGS. 56A to 56 J, sets forth exemplary schemes for remodeling glycan structures on alpha-galactosidase (FabrazymeTM).
  • FIG. 56A is a diagram depicting the alpha-galactosidase A peptide indicating the amino acid residues which bind to N-linked glycans (A) contemplated for remodeling, and formulas for the glycans.
  • FIG. 56B to 56 J are diagrams of contemplated remodeling steps based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 57 comprising FIGS. 57A to 57 J, sets forth exemplary schemes for remodeling glycan structures on alpha-iduronidase (AldurazymeTM).
  • FIG. 57A is a diagram depicting the alpha-iduronidase peptide indicating the amino acid residues which bind to N-linked glycans (A) contemplated for remodeling, and formulas for the glycans.
  • FIG. 57B to 57 J are diagrams of contemplated remodeling steps based on the type of cell the peptide is expressed in and the desired remodeled glycan structure.
  • FIG. 58 is an exemplary nucleotide and corresponding amino acid sequence of granulocyte colony stimulating factor (G-CSF) (SEQ ID NOS: 1 and 2, respectively).
  • G-CSF granulocyte colony stimulating factor
  • FIG. 59 is an exemplary nucleotide and corresponding amino acid sequence of interferon alpha (IFN-alpha) (SEQ ID NOS: 3 and 4, respectively).
  • IFN-alpha interferon alpha
  • FIG. 60 is an exemplary nucleotide and corresponding amino acid sequence of interferon beta (IFN-beta) (SEQ ID NOS: 5 and 6, respectively).
  • IFN-beta interferon beta
  • FIG. 61 is an exemplary nucleotide and corresponding amino acid sequence of Factor VIIa (SEQ ID NOS: 7 and 8, respectively).
  • FIG. 62 is an exemplary nucleotide and corresponding amino acid sequence of Factor IX (SEQ ID NOS: 9 and 10, respectively).
  • FIG. 63 is an exemplary nucleotide and corresponding amino acid sequence of the alpha and beta chains of follicle stimulating hormone (FSH), respectively (SEQ ID NOS: 11 through 14, respectively).
  • FSH follicle stimulating hormone
  • FIG. 64 is an exemplary nucleotide and corresponding amino acid sequence of erythropoietin (EPO) (SEQ ID NOS: 15 and 16, respectively).
  • EPO erythropoietin
  • FIG. 65 is an amino acid sequence of mature EPO, i.e. 165 amino acids (SEQ ID NO:73).
  • FIG. 66 is an exemplary nucleotide and corresponding amino acid sequence of granulocyte-macrophage colony stimulating factor (GM-CSF) (SEQ ID NOS: 17 and 18, respectively).
  • GM-CSF granulocyte-macrophage colony stimulating factor
  • FIG. 67 is an exemplary nucleotide and corresponding amino acid sequence of interferon gamma (IFN-gamma) (SEQ ID NOS: 19 and 20, respectively).
  • IFN-gamma interferon gamma
  • FIG. 68 is an exemplary nucleotide and corresponding amino acid sequence of ⁇ -1-protease inhibitor (A-1-PI, or ⁇ -antitrypsin) (SEQ ID NOS: 21 and 22, respectively).
  • A-1-PI ⁇ -1-protease inhibitor
  • ⁇ -antitrypsin SEQ ID NOS: 21 and 22, respectively.
  • FIG. 69 is an exemplary nucleotide and corresponding amino acid sequence of glucocerebrosidase (SEQ ID NOS: 23 and 24, respectively).
  • FIG. 70 is an exemplary nucleotide and corresponding amino acid sequence of tissue-type plasminogen activator (TPA) (SEQ ID NOS: 25 and 26, respectively).
  • TPA tissue-type plasminogen activator
  • FIG. 71 is an exemplary nucleotide and corresponding amino acid sequence of Interleukin-2 (IL-2) (SEQ ID NOS: 27 and 28, respectively).
  • IL-2 Interleukin-2
  • FIG. 72 is an exemplary nucleotide and corresponding amino acid sequence of Factor VIII (SEQ ID NOS: 29 and 30, respectively).
  • FIG. 73 is an exemplary nucleotide and corresponding amino acid sequence of urokinase (SEQ ID NOS: 33 and 34, respectively).
  • FIG. 74 is an exemplary nucleotide and corresponding amino acid sequence of human recombinant DNase (hrDNase) (SEQ ID NOS: 39 and 40, respectively).
  • hrDNase human recombinant DNase
  • FIG. 75 is an exemplary nucleotide and corresponding amino acid sequence of an insulin molecule (SEQ ID NOS: 43 and 44, respectively).
  • FIG. 76 is an exemplary nucleotide and corresponding amino acid sequence of S-protein from a Hepatitis B virus (HbsAg) (SEQ ID NOS: 45 and 46, respectively).
  • HbsAg Hepatitis B virus
  • FIG. 77 is an exemplary nucleotide and corresponding amino acid sequence of human growth hormone (hGH) (SEQ ID NOS: 47 and 48, respectively).
  • FIG. 78 comprising FIGS. 78A and 78D , are exemplary nucleotide and corresponding amino acid sequences of anti-thrombin III.
  • FIGS. 78A and 78B are an exemplary nucleotide and corresponding amino acid sequences of “WT” anti-thrombin III (SEQ ID NOS: 63 and 64, respectively).
  • FIG. 79 is exemplary nucleotide and corresponding amino acid sequences of human chorionic gonadotropin (hCG) ⁇ and ⁇ subunits.
  • FIGS. 79A and 79B are an exemplary nucleotide and corresponding amino acid sequence of the ⁇ -subunit of human chorionic gonadotropin (SEQ ID NOS: 69 and 70, respectively).
  • FIGS. 79C and 79D are an exemplary nucleotide and corresponding amino acid sequence of the beta subunit of human chorionic gonadotrophin (SEQ ID NOS: 71 and 72, respectively).
  • FIG. 80 is an exemplary nucleotide and corresponding amino acid sequence of ⁇ -iduronidase (SEQ ID NOS: 65 and 66, respectively).
  • FIG. 81 is an exemplary nucleotide and corresponding amino acid sequence of ⁇ -galactosidase A (SEQ ID NOS: 67 and 68, respectively).
  • FIG. 82 is an exemplary nucleotide and corresponding amino acid sequence of the 75 kDa tumor necrosis factor receptor (INF-R), which comprises a portion of EnbrelTM (tumor necrosis factor receptor (TNF-R)/IgG fusion) (SEQ ID NOS: 31 and 32, respectively).
  • INF-R tumor necrosis factor receptor
  • FIG. 83 is an exemplary amino acid sequence of the light and heavy chains, respectively, of HerceptinTM (monoclonal antibody (MAb) to Her-2, human epidermal growth factor receptor) (SEQ ID NOS: 35 and 36, respectively).
  • HerceptinTM monoclonal antibody (MAb) to Her-2, human epidermal growth factor receptor
  • FIG. 84 is an exemplary amino acid sequence the heavy and light chains, respectively, of SynagisTM (MAb to F peptide of Respiratory Syncytial Virus) (SEQ ID NOS: 37 and 38, respectively).
  • SynagisTM MAb to F peptide of Respiratory Syncytial Virus
  • FIG. 85 is an exemplary nucleotide and corresponding amino acid sequence of the non-human variable regions of RemicadeTM (MAb to TNF ⁇ ) (SEQ ID NOS: 41 and 42, respectively).
  • FIG. 86 is an exemplary nucleotide and corresponding amino acid sequence of the Fc portion of human IgG (SEQ ID NOS: 49 and 50, respectively).
  • FIG. 87 is an exemplary amino acid sequence of the mature variable region light chain of an anti-glycoprotein IIb/IIIa murine antibody (SEQ ID NO: 52).
  • FIG. 88 is an exemplary amino acid sequence of the mature variable region heavy chain of an anti-glycoprotein IIb/IIa murine antibody (SEQ ID NO: 54).
  • FIG. 89 is an exemplary amino acid sequence of variable region light chain of a human IgG (SEQ ID NO: 51).
  • FIG. 90 is an exemplary amino acid sequence of variable region heavy chain of a human IgG (SEQ ID NO:53).
  • FIG. 91 is an exemplary amino acid sequence of a light chain of a human IgG (SEQ ID NO:55).
  • FIG. 92 is an exemplary amino acid sequence of a heavy chain of a human IgG (SEQ ID NO:56).
  • FIG. 93 is an exemplary nucleotide and corresponding amino acid sequence of the mature variable region of the light chain of an anti-CD20 murine antibody (SEQ ID NOS: 59 and 60, respectively).
  • FIG. 94 is an exemplary nucleotide and corresponding amino acid sequence of the mature variable region of the heavy chain of an anti-CD20 murine antibody (SEQ ID NOS: 61 and 62, respectively).
  • FIG. 95 is the nucleotide sequence of the tandem chimeric antibody expression vector TCAE 8 (SEQ ID NO:57).
  • FIG. 96 is the nucleotide sequence of the tandem chimeric antibody expression vector TCAE 8 containing the light and heavy variable domains of the anti-CD20 murine antibody (SEQ ID NO:58).
  • FIG. 97 comprising FIGS. 97A to 97 C, are graphs depicting 2-AA HPLC analysis of glyeans released by PNGaseF from myeloma-expressed Cri-IgG1 antibody.
  • the structure of the glycans is determined by retention time: the G0 glycoform elutes at 30 min., the G1 glycoform elutes at ⁇ 33 min., the G2 glycoform elutes at about approximately 37 min. and the S1-G1 glycoform elutes at ⁇ 70 min.
  • FIG. 97A depicts the analysis of the DEAE antibody sample.
  • FIG. 97B depicts the analysis of the SPA antibody sample.
  • FIG. 97C depicts the analysis of the Fc antibody sample. The percent area under the peaks for these graphs is summarized in Table 14.
  • FIG. 98 comprising FIGS. 98A to 98 C, are graphs depicting the MALDI analysis of glycans released by PNGaseF from myeloma-expressed Cri-IgG1 antibody. The glycans were derivatized with 2-AA and then analyzed by MALDI.
  • FIG. 98A depicts the analysis of the DEAE antibody sample.
  • FIG. 98B depicts the analysis of the SPA antibody sample.
  • FIG. 98C depicts the analysis of the Fc antibody sample.
  • FIG. 99 comprising FIGS. 99A to 99 D, are graphs depicting the capillary electrophoresis analysis of glycans released from Cri-IgG1 antibodies that have been glycoremodeled to contain M3N2 glycoforms.
  • a graph depicting the capillary electrophoresis analysis of glycan standards derivatized with APTS is shown in FIG. 99A .
  • FIG. 99B depicts the analysis of the DEAE antibody sample.
  • FIG. 99C depicts the analysis of the SPA antibody sample.
  • FIG. 99D depicts the analysis of the Fc antibody sample. The percent area under the peaks for these graphs is summarized in Table 15.
  • FIG. 100 comprising FIGS. 100A to 100 D, are graphs depicting the capillary electrophoresis analysis of glycans released from Cri-IgG1 antibodies that have been glycoremodeled to contain G0 glycoforms.
  • a graph depicting the capillary electrophoresis analysis of glycan standards derivatized with APTS is shown in FIG. 100A .
  • FIG. 100B depicts the analysis of the DEAE antibody sample.
  • FIG. 100C depicts the analysis of the SPA antibody sample.
  • FIG. 100D depicts the analysis of the Fc antibody sample. The percent area under the peaks for these graphs is summarized in Table 16.
  • FIG. 101 comprising FIGS. 101A to 101 C, are graphs depicting 2-AA HPLC analysis of glycans released from Cri-IgG1 antibodies that have been glycoremodeled to contain G0 glycoforms. The released glycans were labeled with 2AA and separated by HPLC on a NH2P-50 4D amino column.
  • FIG. 101A depicts the analysis of the DEAE antibody sample.
  • FIG. 101B depicts the analysis of the SPA antibody sample.
  • FIG. 101C depicts the analysis of the Fc antibody sample. The percent area under the peaks for these graphs is summarized in Table 16
  • FIG. 102 comprising FIGS. 102A to 102 C, are graphs depicting the MALDI analysis of glycans released from Cri-IgG1 antibodies that have been glycoremodeled to contain G0 glycoforms. The released glycans were derivatized with 2-AA and then analyzed by MALDI.
  • FIG. 102A depicts the analysis of the DEAE antibody sample.
  • FIG. 102B depicts the analysis of the SPA antibody sample.
  • FIG. 102C depicts the analysis of the Fc antibody sample.
  • FIG. 103 is graphs depicting the capillary electrophoresis analysis of glycans released from Cri-IgG1 antibodies that have been glycoremodeled to contain G2 glycoforms.
  • a graph depicting the capillary electrophoresis analysis of glycan standards derivatized with APTS is shown in FIG. 103A .
  • FIG. 103B depicts the analysis of the DEAE antibody sample.
  • FIG. 103C depicts the analysis of the SPA antibody sample.
  • FIG. 103D depicts the analysis of the Fc antibody sample. The percent area under the peaks for these graphs is summarized in Table 17.
  • FIG. 104 comprising FIGS. 104A to 104 C, are graphs depicting the 2-AA HPLC analysis of glycans released from remodeled Cri-IgG1 antibodies that have been glycoremodeled to contain G2 glycoforms. The released glycans were labeled with 2AA and then separated by HPLC on a NH2P-50 4D amino column.
  • FIG. 104A depicts the analysis of the DEAE antibody sample.
  • FIG. 104B depicts the analysis of the SPA antibody sample.
  • FIG. 104C depicts the analysis of the Fc antibody sample. The percent area under the peaks for these graphs is summarized in Table 17.
  • FIG. 105 comprising FIGS. 105A to 105 C, are graphs depicting MALDI analysis of glycans released from Cri-IgG1 antibodies that have been glycoremodeled to contain G2 glycoforms. The released glycans were derivatized with 2-AA and then analyzed by MALDI.
  • FIG. 105A depicts the analysis of the DEAE antibody sample.
  • FIG. 105B depicts the analysis of the SPA antibody sample.
  • FIG. 105C depicts the analysis of the Fc antibody sample.
  • FIG. 106 comprising FIGS. 106A to 106 D, are graphs depicting capillary electrophoresis analysis of glycans released from Cri-IgG1 antibodies that have been glycoremodeled by GnT-I treatment of M3N2 glycoforms.
  • a graph depicting the capillary electrophoresis analysis of glycan standards derivatized with APTS is shown in FIG. 106A .
  • FIG. 106B depicts the analysis of the DEAE antibody sample.
  • FIG. 106C depicts the analysis of the SPA antibody sample.
  • FIG. 106D depicts the analysis of the Fc antibody sample.
  • FIG. 107 comprising FIGS. 107A to 107 C, are graphs depicting 2-AA HPLC analysis of glycans released from Cri-IgG1 antibodies that have been remodeled by GnT-I treatment of M3N2 glycoforms. The released glycans were labeled with 2-AA and separated by HPLC on a NH2P-50 4D amino column.
  • FIG. 107A depicts the analysis of the DEAE antibody sample.
  • FIG. 107B depicts the analysis of the SPA antibody sample.
  • FIG. 107C depicts the analysis of the Fc antibody sample.
  • FIG. 108 comprising FIGS. 108A to 108 C, are graphs depicting MALDI analysis of glycans released from Cri-IgG1 antibodies that have been glycoremodeled by GnT-I treatment of M3N2 glycoforms. The released glycans were derivatized with 2-AA and then analyzed by MALDI.
  • FIG. 108A depicts the analysis of the DEAE antibody sample.
  • FIG. 108B depicts the analysis of the SPA antibody sample.
  • FIG. 108C depicts the analysis of the Fc antibody sample.
  • FIG. 109 comprising FIGS. 109A to 109 D, are graphs depicting capillary electrophoresis of glycans released from Cri-IgG1 antibodies that have been glycoremodeled by GnT-I, II and III treatment of M3N2 glycoforms.
  • a graph depicting the capillary electrophoresis analysis of glycan standards derivatized with APTS is shown in FIG. 109A .
  • FIG. 109B depicts the analysis of the DEAE antibody sample.
  • FIG. 109C depicts the analysis of the SPA antibody sample.
  • FIG. 109D depicts the analysis of the Fc antibody sample. The percent area under the peaks for these graphs is summarized in Table 18.
  • FIG. 110 comprising FIGS. 110A to 110 C, are graphs depicting 2-AA HPLC analysis of glycans released from Cri-IgG1 antibodies that have been glycoremodeled by GnT-I, II and III treatment of M3N2 glycoforms. The released glycans were labeled with 2AA and then separated by HPLC on a NH2P-50 4D amino column.
  • FIG. 110A depicts the analysis of the DEAE antibody sample.
  • FIG. 110B depicts the analysis of the SPA antibody sample.
  • FIG. 110C depicts the analysis of the Fc antibody sample. The percent area under the peaks for these graphs is summarized in Table 18.
  • FIG. 111 comprising FIGS. 111A to 111 C, are graphs depicting MALDI analysis of glycans released from Cri-IgG1 antibodies that have been glycoremodeled by galactosyltransferase treatment of NGA2F glycoforms. The released glycans were derivatized with 2-AA and then analyzed by MALDI.
  • FIG. 111A depicts the analysis of the DEAE antibody sample.
  • FIG. 111B depicts the analysis of the SPA antibody sample.
  • FIG. 111C depicts the analysis of the Fc antibody sample.
  • FIG. 112 comprising 112 A to 112 D, are graphs depicting 2-AA HPLC analysis of glycans released from Cri-IgG1 antibodies containing NGA2F isoforms before GalT1 treatment ( FIGS. 112A and 112C ) and after GalT1 treatment ( FIGS. 112B and 112D ).
  • FIGS. 112A and 112B depict the analysis of the DEAE sample of antibodies.
  • FIGS. 112C and 112D depict the analysis of the Fc sample of antibodies.
  • the released glycans were labeled with 2AA and separated by HPLC on a NH2P-50 4D amino column.
  • FIG. 113 comprising 113 A to 113 C, are graphs depicting 2-AA HPLC analysis of glycans released from Cri-IgG1 antibodies that have been glycoremodeled by ST3Gal3 treatment of G2 glycoforms. The released glycans are labeled with 2-AA and then separated by HPLC on a NH2P-50 4D amino column.
  • FIG. 113A depicts the analysis of the DEAE antibody sample.
  • FIG. 113B depicts the analysis of the SPA antibody sample.
  • FIG. 113C depicts the analysis of the Fc antibody sample. The percent area under the peaks for these graphs is summarized in Table 19.
  • FIG. 114 comprising FIGS. 114A to 114 C, are graphs depicting MALDI analysis of glycans released from Cri-IgG1 antibodies that had been glycoremodeled by ST3Gal3 treatment of G2 glycoforms. The released glycans were derivatized with 2-AA and then analyzed by MALDI.
  • FIG. 114A depicts the analysis of the DEAE antibody sample.
  • FIG. 114B depicts the analysis of the SPA antibody sample.
  • FIG. 114C depicts the analysis of the Fc antibody sample.
  • FIG. 115 comprising FIGS. 115A to 115 D, are graphs depicting capillary electrophoresis analysis of glycans released from Cri-IgG1 antibodies that had been glycoremodeled by ST6Gal1 treatment of G2 glycoforms.
  • a graph depicting the capillary electrophoresis analysis of glycan standards derivatized with APTS is shown in FIG. 115A .
  • FIG. 115B depicts the analysis of the DEAE antibody sample.
  • FIG. 115C depicts the analysis of the SPA antibody sample.
  • FIG. 115D depicts the analysis of the Fc antibody sample.
  • FIG. 116 comprising FIGS. 116A to 116 C, are graphs depicting 2-AA HPLC analysis of glycans released from Cri-IgG1 antibodies that had been glycoremodeled by ST6Gal1 treatment of G2 glycoforms. The released glycans were labeled with 2-AA and separated by HPLC on a NH2P-50 4D amino column.
  • FIG. 116A depicts the analysis of the DEAE antibody sample.
  • FIG. 116B depicts the analysis of the SPA antibody sample.
  • FIG. 116C depicts the analysis of the Fc antibody sample.
  • FIG. 117 comprising FIGS. 117A to 117 C, are graphs depicting MALDI analysis of glycans released from Cri-IgG1 antibodies that had been glycoremodeled by ST6Gal1 treatment of G2 glycoforms. The released glycans were derivatized with 2-AA and then analyzed by MALDI.
  • FIG. 117A depicts the analysis of the DEAE antibody sample.
  • FIG. 117B depicts the analysis of the SPA antibody sample.
  • FIG. 117C depicts the analysis of the Fc antibody sample.
  • FIG. 118 depicts images of SDS-PAGE analysis of the glycoremodeled of Cri-IgG1 antibodies with different glycoforms under non-reducing conditions.
  • Bovine serum albumin (BSA) was run under reducing conditions as a quantitative standard. Protein molecular weight standards are displayed and their size is indicated in kDa.
  • FIG. 118A depicts SDS-PAGE analysis of the DEAE, SPA and Fc Cri-IgG1 antibodies glycoremodeled to contain G0 and G2 glycoforms.
  • FIG. 118B depicts SDS-PAGE analysis of the DEAE, SPA and Fe Cri-IgG1 antibodies glycoremodeled to contain NGA2F (bisecting) and GnT-I-M3N2 (GnT1) glycoforms.
  • FIG. 118C depicts SDS-PAGE analysis of the DEAE, SPA and Fc Cri-IgG1 antibodies glycoremodeled to contain S2G2 (ST6Gal1) glycoforms.
  • FIG. 118D depicts SDS-PAGE analysis of the DEAE, SPA and Fc Cri-IgG1 antibodies glycoremodeled to contain M3N2 glycoforms, and BSA.
  • FIG. 118E depicts SDS-PAGE analysis of the DEAE, SPA and Fc Cri-IgG1 antibodies glycoremodeled to contain Gal-NGA2F (Gal-bisecting) glycoforms, and BSA.
  • FIG. 119 is an image of an acrylamide gel depicting the results of FACE analysis of the pre- and post-sialylation of TP10.
  • the BiNA 0 species has no sialic acid residues.
  • the BiNA 1 species has one sialic acid residue.
  • the BiNA 2 species has two sialic acid residues.
  • FIG. 120 is a graph depicting the plasma concentration in ⁇ g/ml over time of pre- and post-sialylation TP10 injected into rats.
  • FIG. 121 is a graph depicting the area under the plasma concentration-time curve (AUC) in ⁇ g/hr/ml for pre- and post sialylated TP10.
  • FIG. 122 is an image of an acrylamide gel depicting the results of FACE glycan analysis of the pre- and post-fucosylation of TP10 and FACE glycan analysis of CHO cell produced TP-20.
  • the BiNA 2 F 2 species has two neuraminic acid (NA) residues and two fucose residues (F).
  • FIG. 123 is a graph depicting the in vitro binding of TP20 (sCR1sLe X ) glycosylated in vitro (diamonds) and in vivo in Lec11 CHO cells (squares).
  • FIG. 124 is a graph depicting the analysis by 2-AA HPLC of glycoforms from the GlcNAc-ylation of EPO.
  • FIG. 125 comprising FIGS. 125A and 125B , are graphs depicting the 2-AA HPLC analysis of two lots of EPO to which N-acetylglucosamine was been added.
  • FIG. 125A depicts the analysis of lot A
  • FIG. 125B depicts the analysis of lot B.
  • FIG. 126 is a graph depicting the 2-AA HPLC analysis of the products the reaction introducing a third glycan branch to EPO with GnT-V.
  • FIG. 127 is a graph depicting a MALDI-TOF spectrum of the glycans of the EPO preparation after treatment with GnT-I, GnT-II, GnT-III, GnT-V and GalT1, with appropriate donor groups.
  • FIG. 128 is a graph depicting a MALDI spectrum the glycans of native EPO.
  • FIG. 129 is an image of an SDS-PAGE gel of the products of the PEGylation reactions using CMP-SA-PEG (1 kDa), and CMP-SA-PEG (10 kDa).
  • FIG. 130 is a graph depicting the results of the in vitro bioassay of PEGylated EPO.
  • Diamonds represent the data from sialylated EPO having no PEG molecules. Squares represent the data obtained using EPO with PEG (1 kDa). Triangles represent the data obtained using EPO with PEG (10 kDa).
  • FIG. 131 is a diagram of CHO-expressed EPO.
  • the EPO polypeptide is 165 amino acids in length, with a molecular weight of 18 kDa without glycosylation.
  • the glycosylated forms of EPO produced in CHO cells have a molecular weight of about 33 kDa to 39 kDa.
  • the shapes which represent the sugars in the glycan chains are identified in the box at the lower edge of the drawing.
  • FIG. 132 is a diagram of insect cell expressed EPO.
  • the shapes that represent the sugars in the glycan chains are identified in the box at the lower edge of FIG. 131 .
  • FIG. 133 is a bar graph depicting the molecular weights of the EPO peptides expressed in insect cells which were remodeled to form complete mono-, bi- and tri-antennary glycans, with optional glycoPEGylation with 1 kDa, 10 kDa or 20 kDa PEG.
  • EpoetinTM is EPO expressed in mammalian cells without further glycan modification or PEGylation.
  • NESP (AranespTM, Amgen, Thousand Oaks, Calif.) is a form of EPO having 5 N-linked glycan sites that is also expressed in mammalian cells without further glycan modification or PEGylation.
  • FIG. 134 depicts one scheme for the remodeling and glycoPEGylation of insect cell expressed EPO.
  • FIG. 134A depicts the remodeling and glycoPEGylation steps that remodel the insect expressed glycan to a mono-antennary glycoPEGylated glycan.
  • FIG. 134B depicts the remodeled EPO polypeptide having a completed glycoPEGylated mono-antennary glycan at each N-linked glyean site of the polypeptide.
  • the shapes that represent the sugars in the glycan chains are identified in the box at the lower edge of FIG. 131 , except that the triangle represents sialic acid.
  • FIG. 135 is a graph depicting the in vitro bioactivities of EPO-SA and EPO-SA-PEG constructs.
  • the in vitro assay measured the proliferation of TF-1 erythroleukemia cells which were maintained for 48 hr in RBMI+FBS 10%+GM-CSF (12 ng/ml) after the EPO construct was added at 10.0, 5.0, 2.0, 1.0, 0.5, and 0 ⁇ g/ml.
  • Tri-SA refers to EPO constructs where the glycans are tri-antennary and have SA.
  • Tri-SA 1K PEG refers to EPO constructs where the glycans are tri-antennary and have Gal and are then glycoPEGylated with SA-PEG 1 kDa.
  • Di-SA 10K PEG refers to EPO constructs where the glycans are bi-antennary and have Gal and are then glycoPEGylated with SA-PEG 10 kDa.
  • Di-SA 1K PEG refers to EPO constructs where the glycans are bi-antennary and have Gal and are then glycoPEGylated with SA-PEG 1 kDa.
  • Di-SA refers to EPO constructs where the glycans are bi-antennary and are built out to SA.
  • EpogenTM is EPO expressed in CHO cells with no further glycan modification.
  • FIG. 136 is a graph depicting the pharmacokinetics of the EPO constructs in rat. Rats were bolus injected with [I 125 ]-labeled glycoPEGylated and non-glycoPEGylated EPO. The graph shows the concentration of the radio-labeled EPO in the bloodstream of the rat at 0 to about 72 minutes after injection.
  • Biant-10K refers to EPO with biantennary glycan structures with terminal 10 kDa PEG moieties.
  • “Mono-20K” refers to EPO with monoantennary glycan structures with terminal 20 kDa PEG moieties.
  • NESP refers to the commercially available Aranesp.
  • Biant-1K refers to EPO with biantennary glycan structures with terminal 1 kDa PEG moieties.
  • Biant-SA refers to EPO with biantennary glycan structures with terminal 1 kDa moieties.
  • concentration of the EPO constructs in the bloodstream at 72 hr. is as follows: Biant-10K, 5.1 cpm/ml; Mono-20K, 3.2 cpm/ml; NESP, 1 cpm/ml; and Biant-1K, 0.2 cpm/ml; Biant-SA, 0.1 cpm/ml.
  • the relative area under the curve of the EPO constructs is as follows: Biant-10K, 2.9; Mono-20K, 2.1; NESP, 1; Biant-1K, 0.5; and Biant-SA, 0.2.
  • FIG. 137 is a bar graph depicting the ability of the EPO constructs to stimulate reticulocytosis in vivo.
  • Each treatment group is composed of eight mice. Mice were given a single subcutaneous injection of 10 ⁇ g protein/kg body weight. The percent reticulocytosis was measured at 96 hr.
  • Tri-antennary-SA2,3(6) construct has the SA molecule bonded in a 2,3 or 2,6 linkage (see, Example 18 herein for preparation) wherein the glycan on EPO is tri-antennary N-glycans with SA-PEG 10 K is attached thereon.
  • bi-antennary-10K PEG is EPO having a bi-antennary N-glycan with SA-PEG at 10 K PEG attached thereon.
  • FIG. 138 is a bar graph depicting the ability of EPO constructs to increase the hematocrit of the blood of mice in vivo.
  • CD-1 female mice were injected i.p. with 2.5 ⁇ g protein/kg body weight. The hematocrit of the mice was measured on day 15 after the EPO injection.
  • Bi-1k refers to EPO constructs where the glycans are bi-antennary and are built out to the Gal and then glycoPEGylated with SA-PEG 1 kDa.
  • Mono-20k refers to EPO constructs where the glycans are mono-antennary and are built out to the Gal and then glycoPEGylated with SA-PEG 20 kDa.
  • FIG. 139 depicts the analysis of glycans enzymatically released from EPO expressed in insect cells (Protein Sciences, Lot #060302).
  • FIG. 139A depicts the HPLC analysis of the released glycans.
  • FIG. 139B depicts the MALDI analysis of the released glycans. Diamonds represent fucose, and squares represent GlcNAc, circles represent mannose.
  • FIG. 140 depicts the MALDI analysis of glycans released from EPO after the GnT-I/GalT-1 reaction.
  • the structures of the glycans have been determined by comparison of the peak spectrum with that of standard glycans.
  • the glycan structures are depicted beside the peaks.
  • Diamonds represent fucose, and squares represent GlcNAc, circles represent mannose, stars represent galactose.
  • FIG. 141 depicts the SDS-PAGE analysis of EPO after the GnT-I/GalT-1 reaction, Superdex 75 purification, ST3Gal3 reaction with SA-PEG (10 kDa) and SA-PEG (20 kDa).
  • FIG. 142 depicts the results of the TF-1 cell in vitro bioassay of PEGylated mono-antennary EPO.
  • FIG. 143 depicts the analysis of glycan released from EPO after the GnT-I/GnT-II reaction.
  • FIG. 143A depicts the HPLC analysis of the released glycans, where peak 3 represents the bi-antennary GlcNAc glycan.
  • FIG. 143B depicts the MALDI analysis of the released glycans. The structures of the glycans have been determined by comparison of the peak spectrum with that of standard glycans. The glycan structures are depicted beside the peaks. Diamonds represent fucose, and squares represent GlcNAc, circles represent mannose.
  • FIG. 144 depicts the HPLC analysis of glycans released from EPO after the GalT-1 reaction.
  • FIG. 144A depicts the glycans released after the small scale GalT-1 reaction.
  • FIG. 144B depicts the glycans released after the large scale GalT-1 reaction.
  • Peak 1 is the bi-antennary glycan with terminal galactose moieties and Peak 2 is the bi-antennary glycan without terminal galactose moieties.
  • FIG. 145 depicts the Superdex 75 chromatography separation of EPO species after the GalT-1 reaction. Peak 2 contains EPO with bi-antennary glycans with terminal galactose moieties.
  • FIG. 146 depicts the SDS-PAGE analysis of each of the products of the glycoremodeling process to make bi-antennary glycans with terminal galactose moieties.
  • FIG. 147 depicts the SDS-PAGE analysis of EPO after ST3Gal3 sialylation or PEGylation with SA-PEG (1 kDa) or SA-PEG (10 kDa).
  • FIG. 148 depicts the HPLC analysis of glycans released from EPO after the GnT-I/GnT-II reaction.
  • the structures of the glycans have been determined by comparison of the peak retention with that of standard glycans.
  • the glycan structures are depicted beside the peaks.
  • Diamonds represent fucose, and squares represent GlcNAc, circles represent mannose.
  • FIG. 149 depicts the HPLC analysis of glycans released from EPO after the GnT-V reaction.
  • the structures of the glycans have been determined by comparison of the peak retention with that of standard glycans.
  • the glycan structures are depicted beside the peaks.
  • Diamonds represent fucose, and squares represent GlcNAc, circles represent mannose.
  • FIG. 150 depicts the HPLC analysis of glycans released from EPO after the GalT-1 reaction.
  • the structures of the glycans have been determined by comparison of the peak retention with that of standard glycans.
  • the glycan structures are depicted beside the peaks.
  • Diamonds represent fucose, and squares represent GlcNAc, circles represent mannose, open circles represent galactose and triangles represent sialic acid.
  • FIG. 151 depicts the HPLC analysis of glycans released from EPO after the ST3Gal3 reaction.
  • the structures of the glycans have been determined by comparison of the peak retention with that of standard glycans.
  • the glycan structures are depicted beside the peaks.
  • Diamonds represent fucose, and squares represent GlcNAc, circles represent mannose, open circles represent galactose and triangles represent sialic acid.
  • FIG. 152 depicts the HPLC analysis of glycans released from EPO after the ST6Gal1 reaction.
  • the structures of the glycans have been determined by comparison of the peak retention with that of standard glycans.
  • the glycan structures are depicted beside the peaks.
  • FIG. 153 depicts the results of the TF-1 cells in vitro bioassay of EPO with bi-antennary and triantennary glycans.
  • Di-SA refers to EPO with bi-antennary glycans that terminate in sialic acid.
  • Di-SA 10K PEG refers to EPO with bi-antennary glycans that terminate in sialic acid derivatized with PEG (10 kDa).
  • Di-SA 1K PEG refers to EPO with bi-antennary glycans that terminate in sialic acid derivatized with PEG (1 kDa).
  • Tri-SA ST6 +ST3 refers to EPO with tri-antennary glycans terminating in 2,6-SA capped with 2,3-SA
  • Tri-SA ST3 refers to EPO with tri-antennary glycans terminating in 2,3-SA.
  • FIG. 154 is an image of an IEF gel depicting the pI of the products of the desialylation procedure. Lanes 1 and 5 are IEF standards. Lane 2 is Factor IX protein. Lane 3 is rFactor IX protein. Lane 4 is the desialylation reaction of rFactor IX protein at 20 hr.
  • FIG. 155 is an image of an SDS-PAGE gel depicting the molecular weight of Factor IX conjugated with either SA-PEG (1 kDa) or SA-PEG (10 kDa) after reaction with CMP-SA-PEG.
  • Lanes 1 and 6 are SeeBlue+2 molecular weight standards.
  • Lane 2 is rF-IX.
  • Lane 3 is desialylated rF-IX.
  • Lane 4 is rFactor IX conjugated to SA-PEG (1 kDa).
  • Lane 5 is rFactor IX conjugated to SA-PEG (10 kDa).
  • FIG. 156 is an image of an SDS-PAGE gel depicting the reaction products of direct-sialylation of Factor-IX and sialic acid capping of Factor-IX-SA-PEG.
  • Lane 1 is protein standards, lane 2 is blank; lane 3 is rFactor-IX; lane 4 is SA capped rFactor-IX-SA-PEG (10 kDa); lane 5 is rFactor-IX-SA-PEG (10 kDa); lane 6 is ST3Gal1; lane 7 is ST3Gal3; lanes 8, 9, 10 are rFactor-IX-SA-PEG(10 kDa) with no prior sialidase treatment.
  • FIG. 157 is an image of an isoelectric focusing gel (pH 3-7) of asialo-Factor VIIa.
  • Lane 1 is rFactor VIIa; lanes 2-5 are asialo-Factor VIIa.
  • FIG. 158 is a graph of a MALDI spectra of Factor VIIa.
  • FIG. 159 is a graph of a MALDI spectra of Factor VIIa-PEG (1 kDa).
  • FIG. 160 is a graph depicting a MALDI spectra of Factor VIIa-PEG (10 kDa).
  • FIG. 161 is an image of an SDS-PAGE gel of PEGylated Factor VIIa.
  • Lane 1 is asialo-Factor VIIa.
  • Lane 2 is the product of the reaction of asialo-Factor VIIa and CMP-SA-PEG(1 kDa) with ST3Gal3 after 48 hr.
  • Lane 3 is the product of the reaction of asialo-Factor VIIa and CMP-SA-PEG (1 kDa) with ST3Gal3 after 48 hr.
  • Lane 4 is the product of the reaction of asialo-Factor VIIa and CMP-SA-PEG (10 kDa) with ST3Gal3 at 96 hr.
  • FIG. 162 is an image of an isoelectric focusing (IEF) gel depicting the products of the desialylation reaction of human pituitary FSH.
  • Lanes 1 and 4 are isoelectric focusing (IEF) standards.
  • Lane 2 is native FSH.
  • Lane 3 is desialylated FSH.
  • FIG. 163 is an image of an SDS-PAGE gel of the products of the reactions to make PEG-sialylation of rFSH.
  • Lanes 1 and 8 are SeeBlue+2 molecular weight standards.
  • Lane 2 is 15 ⁇ g of native FSH.
  • Lane 3 is 15 ⁇ g of asialo-FSH (AS-FSH).
  • Lane 4 is 15 ⁇ g of the products of the reaction of AS-FSH with CMP-SA.
  • Lane 5 is 15 ⁇ g of the products of the reaction of AS-FSH with CMP-SA-PEG (1 kDa).
  • Lane 6 is 15 ⁇ g of the products of the reaction of AS-FSH with CMP-SA-PEG (5 kDa).
  • Lane 7 is 15 ⁇ g of the products of the reaction of AS-FSH with CMP-SA-PEG (10 kDa).
  • FIG. 164 is an image of an isoelectric focusing gel of the products of the reactions to make PEG-sialylation of FSH.
  • Lanes 1 and 8 are IEF standards.
  • Lane 2 is 15 ⁇ g of native FSH.
  • Lane 3 is 15 ⁇ g of asialo-FSH (AS-FSH).
  • Lane 4 is 15 ⁇ g of the products of the reaction of AS-FSH with CMP-SA.
  • Lane 5 is 15 ⁇ g of the products of the reaction of AS-FSH with CMP-SA-PEG (1 kDa).
  • Lane 6 is 15 ⁇ g of the products of the reaction of AS-FSH with CMP-SA-PEG (5 kDa).
  • Lane 7 is 15 ⁇ g of the products of the reaction of AS-FSH with CMP-SA-PEG (10 kDa).
  • FIG. 165 is an image of an SDS-PAGE gel of native non-recombinant FSH produced in human pituitary cells. Lanes 1, 2 and 5 are SeeBlueTM+2 molecular weight standards. Lanes 3 and 4 are native FSH at 5 ⁇ g and 25 ⁇ g, respectively.
  • FIG. 166 is an image of an isoelectric focusing gel (pH 3-7) depicting the products of the asialylation reaction of rFSH. Lanes 1 and 4 are IEF standards. Lane 2 is native rFSH. Lane 3 is asialo-rFSH.
  • FIG. 167 is an image of an SDS-PAGE gel depicting the results of the PEG-sialylation of asialo-rFSH.
  • Lane 1 is native rFSH.
  • Lane 2 is asialo-FSH.
  • Lane 3 is the products of the reaction of asialo-FSH and CMP-SA.
  • Lanes 4-7 are the products of the reaction between asialoFSH and 0.5 mM CMP-SA-PEG (10 kDa) at 2 hr, 5 hr, 24 hr, and 48 hr, respectively.
  • Lane 8 is the products of the reaction between asialo-FSH and 1.0 mM CMP-SA-PEG (10 kDa) at 48 hr.
  • Lane 9 is the products of the reaction between asialo-FSH and 1.0 mM CMP-SA-PEG (1 kDa) at 48 hr.
  • FIG. 168 is an image of an isoelectric focusing gel showing the products of PEG-sialylation of asialo-rFSH with a CMP-SA-PEG (1 kDa).
  • Lane 1 is native rFSH.
  • Lane 2 is asialo-rFSH.
  • Lane 3 is the products of the reaction of asialo-rFSH and CMP-SA at 24 hr.
  • Lanes 4-7 are the products of the reaction of asialo-rFSH and 0.5 mM CMP-SA-PEG (1 kDa) at 2 hr, 5 hr, 24 hr, and 48 hr, respectively.
  • Lane 8 is blank.
  • Lanes 9 and 10 are the products of the reaction at 48 hr of asialo-rFSH and CMP-SA-PEG (10 kDa) at 0.5 mM and 1.0 mM, respectively.
  • FIG. 169 is graph of the pharmacokinetics of rFSH and rFSH-SA-PEG (1 kDa and 10 kDa). This graph illustrates the relationship between the time a rFSH compound is in the blood stream of the rat, and the mean concentration of the rFSH compound in the blood for glycoPEGylated rFSH as compared to non-PEGylated rFSH.
  • FIG. 170 is a graph of the results of the FSH bioassay using Sertoli cells. This graph illustrates the relationship between the FSH concentration in the Sertoli cell incubation medium and the amount of 17- ⁇ estradiol released from the Sertoli cells.
  • FIG. 171 is a graph depicting the results of the Steelman-Pohley bioassay of glycoPEGylated and non-glycoPEGylated FSH. Rats were subcutaneously injected with human chorionic gonadotropin and varying amounts of FSH for three days, and the average ovarian weight of the treatment group determined on day 4.
  • rFSH-SA-PEG refers to recombinant FSH that has been glycoPEGylated with PEG (1 kDa). rFSH refers to non-glycoPEGylated FSH.
  • Each treatment group contains 10 rats.
  • FIG. 172 depicts the chromatogram of INF- ⁇ elution from a Superdex-75 column.
  • FIG. 172A depicts the entire chromatogram.
  • FIG. 172B depicts the boxed area of FIG. 172A containing peaks 4 and 5 in greater detail.
  • FIG. 173 depict MALDI analysis of glycans enzymatically released from INF- ⁇ .
  • FIG. 173A depicts the MALDI analysis glycans released from native INF- ⁇ .
  • FIG. 173B depicts the MALDI analysis of glycans released from desialylated INF- ⁇ .
  • the structures of the glyeans have been determined by comparison of the peak spectrum with that of standard glycans. The glycan structures are depicted beside the peaks. Squares represent GlcNAc, triangles represent fucose, circles represent mannose, diamonds represent galactose and stars represent sialic acid.
  • FIG. 174 depicts the lectin blot analysis of the sialylation of the desialylated INF- ⁇ .
  • the blot on the right side is detected with Maackia amurensis agglutinin (MAA) labeled with digoxogenin (DIG) (Roche Applied Science, Indianapolis, Ill.) to detect ⁇ 2,3-sialylation.
  • the blot on the left is detected with Erthrina cristagalli lectin (ECL) labeled with biotin (Vector Laboratories, Burlingame, Calif.) to detect exposed galactose residues.
  • MAA Maackia amurensis agglutinin
  • DIG digoxogenin
  • ECL Erthrina cristagalli lectin
  • FIG. 175 depicts the SDS-PAGE analysis of the products of the PEG (10 kDa) PEGylation reaction of INF- ⁇ .
  • ⁇ PEG refers to INF- ⁇ before the PEGylation reaction.
  • +PEG refers to INF- ⁇ after the PEGylation reaction.
  • FIG. 176 depicts the SDS-PAGE analysis of the products of the PEG (20 kDa) PEGylation reaction of INF- ⁇ .
  • Unmodified refers to INF- ⁇ before the PEGylation reaction.
  • Pegylated refers to INF- ⁇ after the PEGylation reaction.
  • FIG. 177 depicts the chromatogram of PEG (10 kDa) PEGylated INF- ⁇ elution from a Superdex-200 column.
  • FIG. 178 depicts the results of a bioassay of peak fractions of PEG (10 kDa) PEGylated INF- ⁇ shown in the chromatogram depicted Figure INF-PEG 6.
  • FIG. 179 depicts the chromatogram of PEG (20 kDa) PEGylated INF- ⁇ elution from a Superdex-200 column.
  • FIG. 180 is two graphs depicting the MALDI-TOF spectrum of RNaseB ( FIG. 180A ) and the HPLC profile of the oligosaccharides cleaved from RNaseB by N-Glycanase ( FIG. 180B ).
  • the majority of N-glycosylation sites of the peptide are modified with high mannose oligosaccharides consisting of 5 to 9 mannose residues.
  • FIG. 181 is a scheme depicting the conversion of high mannose N-Glycans to hybrid N-Glycans.
  • Enzyme 1 is ⁇ 1,2-mannosidase, from Trichodoma reesei or Aspergillus saitoi.
  • Enzyme 2 is GnT-I ( ⁇ -1,2-N-acetyl glucosaminyl transferase I).
  • Enzyme 3 is GalT-I ( ⁇ 1,4-galactosyltransfease 1).
  • Enzyme 4 is ⁇ 2,3-sialyltransferase or ⁇ 2,6-sialyltransferase.
  • FIG. 182 is two graphs depicting the MALDI-TOF spectrum of RNaseB treated with a recombinant T. reesei ⁇ 1,2-mannosidase ( FIG. 182A ) and the HPLC profile of the oligosaccharides cleaved by N-Glycanase from the modified RNaseB ( FIG. 182B ).
  • FIG. 183 is a graph depicting the MALDI-TOF spectrum of RNaseB treated with a commercially available ⁇ 1,2-mannosidase purified from A. saitoi (Glyko & CalBioChem).
  • FIG. 184 is a graph depicting the MALDI-TOF spectrum of modified RNaseB by treating the product shown in FIG. 182 with a recombinant GnT-I (GlcNAc transferase-I).
  • FIG. 185 is a graph depicting the MALDI-TOF spectrum of modified RNaseB by treating the product shown in FIG. 184 with a recombinant GalT 1 (galactosyltransferase 1).
  • FIG. 186 is a graph depicting the MALDI-TOF spectrum of modified RNaseB by treating the product shown in FIG. 185 with a recombinant ST3Gal III ( ⁇ 2,3-sialyltransferase III) using CMP-SA as the donor for the transferase.
  • ST3Gal III ⁇ 2,3-sialyltransferase III
  • FIG. 187 is a graph depicting the MALDI-TOF spectrum of modified RNaseB by treating the product shown in FIG. 185 with a recombinant ST3Gal III ( ⁇ 2,3-sialyltransferase III) using CMP-SA-PEG (10 kDa) as the donor for the transferase.
  • ST3Gal III ⁇ 2,3-sialyltransferase III
  • FIG. 188 is a series of schemes depicting the conversion of high mannose N-glycans to complex N-glycans.
  • Enzyme 1 is ⁇ 1,2-mannosidase from Trichoderma reesei or Aspergillus saitoi.
  • Enzyme 2 is GnT-I.
  • Enzyme 3 is GalT 1.
  • Enzyme 4 is ⁇ 2,3-sialyltransferase or ⁇ 2,6-sialyltransferase.
  • Enzyme 5 is ⁇ -mannosidase II.
  • Enzyme 6 is ⁇ -mannosidase.
  • Enzyme 7 is GnT-II.
  • Enzyme 8 is ⁇ 1,6-mannosidase.
  • Enzyme 9 is ⁇ 1,3-mannosidase.
  • FIG. 190 is an image of an SDS-PAGE gel: standard (Lane 1); native transferrin (Lane 2); asialotransferrin (Lane 3); asialotransferrin and CMP-SA (Lane 4); Lanes 5 and 6, asialotransferrin and CMP-SA-PEG (1 kDa) at 0.5 mM and 5 mM, respectively; Lanes 7 and 8, asialotransferrin and CMP-SA-PEG, (5 kDa) at 0.5 mM and 5 mM, respectively; Lanes 9 and 10, asialotransferrin and CMP-SA-PEG (10 kDa) at 0.5 mM and 5 mM, respectively.
  • FIG. 191 is an image of an IEF gel: native transferrin (Lane 1); asialotransferrin (Lane 2); asialotransferrin and CMP-SA, 24 hr (Lane 3); asialotransferrin and CMP-SA, 96 hr (Lane 4) Lanes 5 and 6, asialotransferrin and CMP-SA-PEG (1 kDa) at 24 hr and 96 hr, respectively; Lanes 7 and 8, asialotransferrin and CMP-SA-PEG (5 kDa) at 24 hr and 96 hr, respectively; Lanes 9 and 10, asialotransferrin and CMP-SA-PEG (10 kDa) at 24 hr and 96 hr, respectively.
  • the present invention includes methods and compositions for the cell free in vitro addition and/or deletion of sugars to or from a peptide molecule in such a manner as to provide a glycopeptide molecule having a specific customized or desired glycosylation pattern, wherein the glycopeptide is produced at an industrial scale.
  • the glycopeptide so produced has attached thereto a modified sugar that has been added to the peptide via an enzymatic reaction.
  • a key feature of the invention is to take a peptide produced by any cell type and generate a core glycan structure on the peptide, following which the glycan structure is then remodeled in vitro to generate a glycopeptide having a glycosylation pattern suitable for therapeutic use in a mammal.
  • the conjugate molecule is added to the peptide enzymatically because enzyme-based addition of conjugate molecules to peptides has the advantage of regioselectivity and stereoselectivity.
  • the glycoconjugate may be added to the glycan on a peptide before or after glycosylation has been completed. In other words, the order of glycosylation with respect to glycoconjugation may be varied as described elsewhere herein.
  • an element means one element or more than one element.
  • antibody refers to an immunoglobulin molecule which is able to specifically bind to a specific epitope on an antigen.
  • Antibodies can be intact immunoglobulins derived from natural sources or from recombinant sources and can be immunoreactive portions of intact immunoglobulins.
  • Antibodies are typically tetramers of immunoglobulin molecules.
  • the antibodies in the present invention may exist in a variety of forms including, for example, polyclonal antibodies, monoclonal antibodies, Fv, Fab and F(ab) 2 , as well as single chain antibodies and humanized antibodies (Harlow et al., 1999, Using Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, NY; Harlow et al., 1989, Antibodies: A Laboratory Manual, Cold Spring Harbor, N.Y.; Houston et al., 1988, Proc. Natl. Acad. Sci. USA 85:5879-5883; Bird et al., 1988, Science 242:423-426).
  • synthetic antibody as used herein, is meant an antibody which is generated using recombinant DNA technology, such as, for example, an antibody expressed by a bacteriophage as described herein.
  • the term should also be construed to mean an antibody which has been generated by the synthesis of a DNA molecule encoding the antibody and which DNA molecule expresses an antibody protein, or an amino acid sequence specifying the antibody, wherein the DNA or amino acid sequence has been obtained using synthetic DNA or amino acid sequence technology which is available and well known in the art.
  • a “functional” biological molecule is a biological molecule in a form in which it exhibits a property by which it is characterized.
  • a functional enzyme for example, is one which exhibits the characteristic catalytic activity by which the enzyme is characterized.
  • the structure is the point of connection between an amino acid or an amino acid sidechain in the peptide chain and the glycan structure.
  • N-linked oligosaccharides are those oligosaccharides that are linked to a peptide backbone through asparagine, by way of an asparagine-N-acetylglucosamine linkage. N-linked oligosaccharides are also called “N-glycans.” All N-linked oligosaccharides have a common pentasaccharide core of Man 3 GlcNAc 2 . They differ in the presence of, and in the number of branches (also called antennae) of peripheral sugars such as N-acetylglucosamine, galactose, N-acetylgalactosamine, fucose and sialic acid. Optionally, this structure may also contain a core fucose molecule and/or a xylose molecule.
  • an “elemental trimannosyl core structure” refers to a glycan moiety comprising solely a trimannosyl core structure, with no additional sugars attached thereto.
  • the glycan comprises the trirannosyl core structure with additional sugars attached thereto.
  • this structure may also contain a core fucose molecule and/or a xylose molecule.
  • mental trimannosyl core glycopeptide is used herein to refer to a glycopeptide having glycan structures comprised primarily of an elemental trimannosyl core structure.
  • this structure may also contain a core fucose molecule and/or a xylose molecule.
  • O-linked oligosaccharides are those oligosaccharides that are linked to a peptide backbone through threonine, serine, hydroxyproline, tyrosine, or other hydroxy-containing ammo acids.
  • 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.
  • sialic acid refers to any member of a family of nine-carbon carboxylated sugars.
  • the most common member of the sialic acid family is N-acetyl-neuraminic acid (2-keto-5-acetamido-3,5-dideoxy-D-glycero-D-galactononulopyranos-1-onic acid (often abbreviated as Neu5Ac, NeuAc, or NANA).
  • a second member of the family is N-glycolyl-neuraminic acid (Neu5Gc or NeuGc), in which the N-acetyl group of NeuAc is hydroxylated.
  • a third sialic acid family member is 2-keto-3-deoxy-nonulosonic acid (KDN) (Nadano et al. (1986) J. Biol. Chem. 261: 11550-11557;Kanamori et al., J. Biol. Chem. 265:21811-21819 (1990)). Also included are 9-substituted sialic acids such as a 9-O—C 1 -C 6 acyl-Neu5Ac like 9-O-lactyl-Neu5Ac or 9-O-acetyl-Neu5Ac, 9-deoxy-9-fluoro-Neu5Ac and 9-azido-9-deoxy-Neu5Ac.
  • KDN 2-keto-3-deoxy-nonulosonic acid
  • 9-substituted sialic acids such as a 9-O—C 1 -C 6 acyl-Neu5Ac like 9-O-lactyl-Neu5Ac or 9-O
  • sialic acid family see, e.g., Varki, Glycobiology 2: 25-40 (1992); Sialic Acids: Chemistry, Metabolism and Function, R. Schauer, Ed. (Springer-Verlag, New York (1992)).
  • the synthesis and use of sialic acid compounds in a sialylation procedure is disclosed in international application WO 92/16640, published Oct. 1, 1992.
  • a peptide having “desired glycosylation”, as used herein, is a peptide that comprises one or more oligosaccharide molecules which are required for efficient biological activity of the peptide.
  • a “disease” is a state of health of an animal wherein the animal cannot maintain homeostasis, and wherein if the disease is not ameliorated then the animal's health continues to deteriorate.
  • AUC area under the curve
  • half-life or “t 1 ⁇ 2”, as used herein in the context of administering a peptide drug to a patient, is defined as the time required for plasma concentration of a drug in a patient to be reduced by one half. There may be more than one half-life associated with the peptide drug depending on multiple clearance mechanisms, redistribution, and other mechanisms well known in the art. Usually, alpha and beta half-lives are defined such that the alpha phase is associated with redistribution, and the beta phase is associated with clearance. However, with protein drugs that are, for the most part, confined to the bloodstream, there can be at least two clearance half-lives.
  • rapid beta phase clearance may be mediated via receptors on macrophages, or endothelial cells that recognize terminal galactose, N-acetylgalactosamine, N-acetylglucosamine, mannose, or fucose.
  • Slower beta phase clearance may occur via renal glomerular filtration for molecules with an effective radius ⁇ 2 nm (approximately 68 kD) and/or specific or non-specific uptake and metabolism in tissues.
  • GlycoPEGylation may cap terminal sugars (e.g. galactose or N-acetylgalactosamine) and thereby block rapid alpha phase clearance via receptors that recognize these sugars.
  • time is defined as the average time that drug stays in the body of the patient after dosing.
  • isolated nucleic acid refers to a nucleic acid segment or fragment which has been separated from sequences which flank it in a naturally occurring state, e.g., a DNA fragment which has been removed from the sequences which are normally adjacent to the fragment, e.g., the sequences adjacent to the fragment in a genome in which it naturally occurs.
  • the term also applies to nucleic acids which have been substantially purified from other components which naturally accompany the nucleic acid, e.g., RNA or DNA or proteins, which naturally accompany it in the cell.
  • the term therefore includes, for example, a recombinant DNA which is incorporated into a vector, into an autonomously replicating plasmid or virus, or into the genomic DNA of a prokaryote or eukaryote, or which exists as a separate molecule (e.g., as a cDNA or a genomic or cDNA fragment produced by PCR or restriction enzyme digestion) independent of other sequences. It also includes a recombinant DNA which is part of a hybrid nucleic acid encoding additional peptide sequence.
  • a “polynucleotide” means a single strand or parallel and anti-parallel strands of a nucleic acid.
  • a polynucleotide may be either a single-stranded or a double-stranded nucleic acid.
  • nucleic acid typically refers to large polynucleotides.
  • oligonucleotide typically refers to short polynucleotides, generally no greater than about 50 nucleotides.
  • the left-hand end of a single-stranded polynucleotide sequence is the 5′-end; the left-hand direction of a double-stranded polynucleotide sequence is referred to as the 5′-direction.
  • the direction of 5 ′ to 3′ addition of nucleotides to nascent RNA transcripts is referred to as the transcription direction.
  • the DNA strand having the same sequence as an mRNA is referred to as the “coding strand”; sequences on the DNA strand which are located 5′ to a reference point on the DNA are referred to as “upstream sequences”; sequences on the DNA strand which are 3′ to a reference point on the DNA are referred to as “downstream sequences.”
  • Encoding refers to the inherent property of specific sequences of nucleotides in a polynucleotide, such as a gene, a cDNA, or an mRNA, to serve as templates for synthesis of other polymers and macromolecules in biological processes having either a defined sequence of nucleotides (i.e., rRNA, tRNA and mRNA) or a defined sequence of amino acids and the biological properties resulting therefrom.
  • a nucleic acid sequence encodes a protein if transcription and translation of mRNA corresponding to that nucleic acid produces the protein in a cell or other biological system.
  • Both the coding strand, the nucleotide sequence of which is identical to the mRNA sequence and is usually provided in sequence listings, and the non-coding strand, used as the template for transcription of a gene or cDNA, can be referred to as encoding the protein or other product of that nucleic acid or cDNA.
  • nucleotide sequence encoding an amino acid sequence includes all nucleotide sequences that are degenerate versions of each other and that encode the same amino acid sequence. Nucleotide sequences that encode proteins and RNA may include introns.
  • “Homologous” as used herein refers to the subunit sequence similarity between two polymeric molecules, e.g., between two nucleic acid molecules, e.g., two DNA molecules or two RNA molecules, or between two peptide molecules. When a subunit position in both of the two molecules is occupied by the same monomeric subunit, e.g., if a position in each of two DNA molecules is occupied by adenine, then they are homologous at that position.
  • the homology between two sequences is a direct function of the number of matching or homologous positions, e.g., if half (e.g., five positions in a polymer ten subunits in length) of the positions in two compound sequences are homologous then the two sequences are 50% homologous, if 90% of the positions, e.g., 9 of 10, are matched or homologous, the two sequences share 90% homology.
  • the DNA sequences 3′ATTGCC5′ and 3′TATGGC share 50% homology.
  • the determination of percent identity between two nucleotide or amino acid sequences can be accomplished using a mathematical algorithm.
  • a mathematical algorithm useful for comparing two sequences is the algorithm of Karlin and Altschul (1990, Proc. Natl. Acad. Sci. USA 87:2264-2268), modified as in Karlin and Altschul (1993, Proc. Natl. Acad. Sci. USA 90:5873-5877). This algorithm is incorporated into the NBLAST and XBLAST programs of Altschul, et al. (1990, J. Mol. Biol.
  • NCBI National Center for Biotechnology Information
  • BLAST protein searches can be performed with the XBLAST program (designated “blastn” at the NCBI web site) or the NCBI “blastp” program, using the following parameters: expectation value 10.0, BLOSUM62 scoring matrix to obtain amino acid sequences homologous to a protein molecule described herein.
  • Gapped BLAST can be utilized as described in Altschul et al. (1997, Nucleic Acids Res. 25:3389-3402).
  • PSI-Blast or PHI-Blast can be used to perform an iterated search which detects distant relationships between molecules (Id.) and relationships between molecules which share a common pattern.
  • the percent identity between two sequences can be determined using techniques similar to those described above, with or without allowing gaps. In calculating percent identity, typically exact matches are counted.
  • a “heterologous nucleic acid expression unit” encoding a peptide is defined as a nucleic acid having a coding sequence for a peptide of interest operably linked to one or more expression control sequences such as promoters and/or repressor sequences wherein at least one of the sequences is heterologous, i.e., not normally found in the host cell.
  • two polynucleotides as “operably linked” is meant that a single-stranded or double-stranded nucleic acid moiety comprises the two polynucleotides arranged within the nucleic acid moiety in such a manner that at least one of the two polynucleotides is able to exert a physiological effect by which it is characterized upon the other.
  • a promoter operably linked to the coding region of a nucleic acid is able to promote transcription of the coding region.
  • promoter/regulatory sequence means a nucleic acid sequence which is required for expression of a gene product operably linked to the promoter/regulator sequence.
  • this sequence may be the core promoter sequence and in other instances, this sequence may also include an enhancer sequence and other regulatory elements which are required for expression of the gene product.
  • the promoter/regulatory sequence may, for example, be one which expresses the gene product in a tissue specific manner.
  • a “constitutive promoter” is a promoter which drives expression of a gene to which it is operably linked, in a constant manner in a cell.
  • promoters which drive expression of cellular housekeeping genes are considered to be constitutive promoters.
  • an “inducible” promoter is a nucleotide sequence which, when operably linked with a polynucleotide which encodes or specifies a gene product, causes the gene product to be produced in a living cell substantially only when an inducer which corresponds to the promoter is present in the cell.
  • tissue-specific promoter is a nucleotide sequence which, when operably linked with a polynucleotide which encodes or specifies a gene product, causes the gene product to be produced in a living cell substantially only if the cell is a cell of the tissue type corresponding to the promoter.
  • a “vector” is a composition of matter which comprises an isolated nucleic acid and which can be used to deliver the isolated nucleic acid to the interior of a cell.
  • vectors are known in the art including, but not limited to, linear polynucleotides, polynucleotides associated with ionic or amphiphilic compounds, plasmids, and viruses.
  • the term “vector” includes an autonomously replicating plasmid or a virus.
  • the term should also be construed to include non-plasmid and non-viral compounds which facilitate transfer of nucleic acid into cells, such as, for example, polylysine compounds, liposomes, and the like.
  • viral vectors include, but are not limited to, adenoviral vectors, adeno-associated virus vectors, retroviral vectors, and the like.
  • “Expression vector” refers to a vector comprising a recombinant polynucleotide comprising expression control sequences operatively linked to a nucleotide sequence to be expressed.
  • An expression vector comprises sufficient cis-acting elements for expression; other elements for expression can be supplied by the host cell or in an in vitro expression system.
  • Expression vectors include all those known in the art, such as cosmids, plasmids (e.g., naked or contained in liposomes) and viruses that incorporate the recombinant polynucleotide.
  • a “genetically engineered” or “recombinant” cell is a cell having one or more modifications to the genetic material of the cell. Such modifications are seen to include, but are not limited to, insertions of genetic material, deletions of genetic material and insertion of genetic material that is extrachromasomal whether such material is stably maintained or not.
  • a “peptide” is an oligopeptide, polypeptide, peptide, protein or glycoprotein.
  • the use of the term “peptide” herein includes a peptide having a sugar molecule attached thereto when a sugar molecule is attached thereto.
  • “native form” means the form of the peptide when produced by the cells and/or organisms in which it is found in nature. When the peptide is produced by a plurality of cells and/or organisms, the peptide may have a variety of native forms.
  • “Peptide” refers to a polymer in which the monomers are amino acids and are joined together through amide bonds, alternatively referred to as a peptide. Additionally, unnatural amino acids, for example, ⁇ -alanine, phenylglycine and homoarginine are also included. Amino acids that are not nucleic acid-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 thereof. 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 peptides that are incompletely glycosylated by a system that expresses the peptide.
  • 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).
  • peptide conjugate refers to species of the invention in which a peptide is conjugated with a modified sugar as set forth herein.
  • amino acid refers to naturally occurring and synthetic amino acids, as well as amino acid analogs and amino acid mimetics that function in a manner similar to the naturally occurring amino acids.
  • Naturally occurring amino acids are those encoded by the genetic code, as well as those amino acids that are later modified, e.g., hydroxyproline, ⁇ -carboxyglutamate, and O-phosphoserine.
  • Amino acid analogs refers to compounds that have the same basic chemical structure as a naturally occurring amino acid, i.e., an ⁇ carbon that is linked to a hydrogen, a carboxyl group, an amino group, and an R group, e.g., homoserine, norleucine, methionine sulfoxide, methionine methyl sulfonium. Such analogs have modified R groups (e.g., norleucine) or modified peptide backbones, but retain the same basic chemical structure as a naturally occurring amino acid.
  • Amino acid mimetics refers to chemical compounds that have a structure that is different from the general chemical structure of an amino acid, but that function in a manner similar to a naturally occurring amino acid.
  • amino acids are represented by the full name thereof, by the three letter code corresponding thereto, or by the one-letter code corresponding thereto, as indicated in the following Table 1: TABLE 1 Amino acids, and the three letter and one letter codes.
  • Full Name Three-Letter Code One-Letter Code Aspartic Acid Asp D Glutamic Acid Glu E Lysine Lys K Arginine Arg R Histidine His H Tyrosine Tyr Y Cysteine Cys C Asparagine Asn N Glutamine Gln Q Serine Ser S Threonine Thr T Glycine Gly G Alanine Ala A Valine Val V Leucine Leu L Isoleucine Ile I Methionine Met M Proline Pro P Phenylalanine Phe F Tryptophan Trp W
  • the present invention also provides for analogs of proteins or peptides which comprise a protein as identified above.
  • Analogs may differ from naturally occurring proteins or peptides by conservative amino acid sequence differences or by modifications which do not affect sequence, or by both. For example, conservative amino acid changes may be made, which although they alter the primary sequence of the protein or peptide, do not normally alter its function.
  • Conservative amino acid substitutions typically include substitutions within the following groups:
  • Modifications include in vivo, or in vitro, chemical derivatization of peptides, e.g., acetylation, or carboxylation. Also included are modifications of glycosylation, e.g., those made by modifying the glycosylation patterns of a peptide during its synthesis and processing or in further processing steps; e.g., by exposing the peptide to enzymes which affect glycosylation, e.g., mammalian glycosylating or deglycosylating enzymes. Also embraced are sequences which have phosphorylated amino acid residues, e.g., phosphotyrosine, phosphoserine, or phosphothreonine.
  • the peptides may incorporate amino acid residues which are modified without affecting activity.
  • the termini may be derivatized to include blocking groups, i.e. chemical substituents suitable to protect and/or stabilize the N— and C-termini from “undesirable degradation”, a term meant to encompass any type of enzymatic, chemical or biochemical breakdown of the compound at its termini which is likely to affect the function of the compound, i.e. sequential degradation of the compound at a terminal end thereof.
  • Blocking groups include protecting groups conventionally used in the art of peptide chemistry which will not adversely affect the in vivo activities of the peptide.
  • suitable N-terminal blocking groups can be introduced by alkylation or acylation of the N-terminus.
  • suitable N-terminal blocking groups include C 1 -C 5 branched or unbranched alkyl groups, acyl groups such as formyl and acetyl groups, as well as substituted forms thereof, such as the acetamidomethyl (Acm), Fmoc or Boc groups.
  • Desamino analogs of amino acids are also useful N-terminal blocking groups, and can either be coupled to the N-terminus of the peptide or used in place of the N-terminal reside.
  • Suitable C-terminal blocking groups include esters, ketones or amides.
  • Ester or ketone-forming alkyl groups particularly lower alkyl groups such as methyl, ethyl and propyl, and amide-forming amino groups such as primary amines (—NH 2 ), and mono- and di-alkylamino groups such as methylamino, ethylamino, dimethylamino, diethylamino, methylethylamino and the like are examples of C-terminal blocking groups.
  • Descarboxylated amino acid analogues such as agmatine are also useful C-terminal blocking groups and can be either coupled to the peptide's C-terminal residue or used in place of it. Further, it will be appreciated that the free amino and carboxyl groups at the termini can be removed altogether from the peptide to yield desamino and descarboxylated forms thereof without affect on peptide activity.
  • the peptide may include one or more D-amino acid resides, or may comprise amino acids which are all in the D-form.
  • Retro-inverso forms of peptides in accordance with the present invention are also contemplated, for example, inverted peptides in which all amino acids are substituted with D-amino acid forms.
  • Acid addition salts of the present invention are also contemplated as functional equivalents.
  • an inorganic acid such as hydrochloric, hydrobromic, sulfuric, nitric, phosphoric, and the like
  • an organic acid such as an acetic, propionic, glycolic, pyruvic, oxalic
  • peptides which have been modified using ordinary molecular biological techniques so as to improve their resistance to proteolytic degradation or to optimize solubility properties or to render them more suitable as a therapeutic agent.
  • Analogs of such peptides include those containing residues other than naturally occurring L-amino acids, e.g., D-amino acids or non-naturally occurring synthetic amino acids.
  • the peptides of the invention are not limited to products of any of the specific exemplary processes listed herein.
  • MALDI is an abbreviation for Matrix Assisted Laser Desorption Ionization.
  • SA-PEG sialic acid-poly(ethylene glycol)
  • N-glyean structure of the glycoprotein can be partially eliminated from the N-glyean structure of the glycoprotein.
  • glycosyltransferase refers to any enzyme/protein that has the ability to transfer a donor sugar to an acceptor moiety.
  • 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, water-soluble polymers, therapeutic moieties, diagnostic moieties, biomolecules and the like.
  • 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.
  • water-soluble refers to moieties that have some detectable degree of solubility in water. Methods to detect and/or quantify water solubility are well known in the art.
  • Exemplary water-soluble polymers include peptides, saccharides, poly(ethers), poly(amines), poly(carboxylic acids) and the like.
  • Peptides can have mixed sequences or be composed of a single amino acid, e.g. poly(lysine).
  • saccharides can be of mixed sequence or composed of a single saccharide subunit, e.g., dextran, amylose, chitosan, and poly(sialic acid).
  • An exemplary poly(ether) is poly(ethylene glycol).
  • Poly(ethylene imine) is an exemplary polyamine
  • poly(aspartic) acid is a representative poly(carboxylic acid).
  • Poly(alkylene oxide) refers to a genus of compounds having a polyether backbone.
  • Poly(alkylene oxide) species of use in the present invention include, for example, straight- and branched-chain species.
  • exemplary poly(alkylene oxide) species can terminate in one or more reactive, activatable, or inert groups.
  • poly(ethylene glycol) is a poly(alkylene oxide) consisting of repeating ethylene oxide subunits, which may or may not include additional reactive, activatable or inert moieties at either terminus.
  • Useful poly(alkylene oxide) species include those in which one terminus is “capped” by an inert group, e.g., monomethoxy-poly(alkylene oxide).
  • the molecule When the molecule is a branched species, it may include multiple reactive, activatable or inert groups at the termini of the alkylene oxide chains and the reactive groups may be either the same or different. Derivatives of straight-chain poly(alkylene oxide) species that are heterobifunctional are also known in the art.
  • glycosyl linking group refers to a glycosyl residue to which an agent (e.g., water-soluble polymer, therapeutic moiety, biomolecule) is covalently attached.
  • agent e.g., water-soluble polymer, therapeutic moiety, biomolecule
  • the “glycosyl linking group” becomes covalently attached to a glycosylated or unglycosylated peptide, thereby linking the agent to an amino acid and/or glycosyl residue on the peptide.
  • a “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 peptide.
  • a “glycosyl linking group,” as used herein, refers to a moiety that covalently joins a “modifying group,” as discussed herein, and an amino acid residue of a peptide.
  • the glycosyl linking group-modifying group adduct has a structure that is a substrate for an enzyme.
  • the enzymes for which the glycosyl linking group-modifying group adduct are substrates are generally those capable of transferring a saccharyl moiety onto an amino acid residue of a peptide, e.g, a glycosyltransferase, amidase, glycosidase, trans-sialidase, etc.
  • the “glycosyl linking group” is interposed between, and covalently joins a “modifying group” and an amino acid residue of a peptide.
  • an “intact glycosyl linking group” refers to a linking group that is derived from a glycosyl moiety in which the individual saccharide monomer that links 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.
  • An exemplary “intact glycosyl linking group” includes at least one intact, e.g., non-degraded, saccharyl moiety that is covalently attached to an amino acid residue on a peptide.
  • the remainder of the “linking group” can have substantially any structure.
  • the modifying group is optionally linked directly to the intact saccharyl moiety.
  • the modifying group is linked to the intact saccharyl moiety via a linker arm.
  • the linker arm can have substantially any structure determined to be useful in the selected embodiment.
  • the linker arm is one or more intact saccharyl moieties, i.e. “the intact glycosyl linking group” resembles an oligosaccharide.
  • Another exemplary intact glycosyl linking group is one in which a saccharyl moiety attached, directly or indirectly, to the intact saccharyl moiety is degraded and derivatized (e.g., periodate oxidation followed by reductive amination).
  • Still a further linker arm includes the modifying group attached to the intact saccharyl moiety, directly or indirectly, via a cross-linker, such as those described herein or analogues thereof.
  • Degradation refers to the removal of one or more carbon atoms from a saccharyl moiety.
  • targeting moiety and “targeting agent”, as used herein, refer to species that will selectively localize in a particular tissue or region of the body. The localization is mediated by specific recognition of molecular determinants, molecular size of the targeting agent or conjugate, ionic interactions, hydrophobic interactions and the like. Other mechanisms of targeting an agent to a particular tissue or region are known to those of skill in the art.
  • therapeutic moiety means any agent useful for therapy including, but not limited to, antibiotics, anti-inflammatory agents, anti-tumor drugs, cytotoxins, and radioactive agents.
  • therapeutic moiety includes prodrugs of bioactive agents, constructs in which more than one therapeutic moiety is linked to a carrier, e.g., multivalent agents.
  • Therapeutic moiety also includes peptides, and constructs that include peptides. Exemplary peptides include those disclosed in FIG. 28 and Tables 6 and 7, herein.
  • “Therapeutic moiety” thus means any agent useful for therapy including, but not limited to, antibiotics, anti-inflammatory agents, anti-tumor drugs, cytotoxins, and radioactive agents.
  • therapeutic moiety includes prodrugs of bioactive agents, constructs in which more than one therapeutic moiety is linked to a carrier, e.g., multivalent agents.
  • anti-tumor drug means any agent useful to combat cancer including, but not limited to, cytotoxins and agents such as antimetabolites, alkylating agents, anthracyclines, antibiotics, antimitotic agents, procarbazine, hydroxyurea, asparaginase, corticosteroids, interferons and radioactive agents.
  • conjugates of peptides with anti-tumor activity e.g. TNF- ⁇ . Conjugates include, but are not limited to those formed between a therapeutic protein and a glycoprotein of the invention. A representative conjugate is that formed between PSGL-1 and TNF- ⁇ .
  • a cytotoxin or cytotoxic agent means any agent that is detrimental to cells. Examples include taxol, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicin, doxorubicin, daunorubicin, dihydroxy anthracinedione, mitoxantrone, mithramycin, actinomycin D, 1-dehydrótestosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, and puromycin and analogs or homologs thereof.
  • Other toxins include, for example, ricin, CC-1065 and analogues, the duocarmycins. Still other toxins include diphtheria toxin, and snake venom (e.g., cobra venom).
  • a radioactive agent includes any radioisotope that is effective in diagnosing or destroying a tumor. Examples include, but are not limited to, indium-111, cobalt-60 and technetium. Additionally, naturally occurring radioactive elements such as uranium, radium, and thorium, which typically represent mixtures of radioisotopes, are suitable examples of a radioactive agent. The metal ions are typically chelated with an organic chelating moiety.
  • “pharmaceutically acceptable carrier” includes any material, which when combined with the conjugate retains the activity of the conjugate activity and is non-reactive with the subject's immune system.
  • 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 ay also include sterile solutions, tablets including coated tablets and capsules.
  • Such carriers 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.
  • 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, intranasal or subcutaneous administration, intrathecal administration, or the implantation of a slow-release device e.g., a mini-osmotic pump, to the subject.
  • a slow-release device e.g., a mini-osmotic pump
  • isolated refers to a material that is substantially or essentially free from components, which are used to produce the material.
  • isolated refers to material that is substantially or 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 preferably expressed as a range. The lower end of the range of purity 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 about 90% pure, their purities are also preferably expressed as a range.
  • the lower end of the range of purity 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% purity.
  • Purity is determined by any art-recognized method of analysis (e.g., band intensity on a silver stained gel, polyacrylamide gel electrophoresis, HPLC, or a similar means).
  • “Commercial scale” as used herein means about one or more gram of final product produced in the method.
  • Essentially each member of the population describes a characteristic of a population of peptide conjugates of the invention in which a selected percentage of the modified sugars added to a peptide are added to multiple, identical acceptor sites on the peptide. “Essentially each member of the population” speaks to the “homogeneity” of the sites on the peptide conjugated to a modified sugar and refers to conjugates of the invention, which are at least about 80%, preferably at least about 90% and more preferably at least about 95% homogenous.
  • “Homogeneity,” refers to the structural consistency across a population of acceptor moieties to which the modified sugars are conjugated. Thus, in a peptide conjugate of the invention in which each modified sugar moiety is conjugated to an acceptor site having the same structure as the acceptor site to which every other modified sugar is conjugated, the peptide conjugate 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% homogeneity.
  • the purity of the peptide conjugates is typically determined by one or more methods known to those of skill in the art, e.g., liquid chromatography-mass spectrometry (LC-MS), matrix assisted laser desorption time of flight mass spectrometry (MALDI-TOF), capillary electrophoresis, and the like.
  • substantially uniform glycoform or a “substantially uniform glycosylation pattern,” when referring to a glycopeptide species, refers to the percentage of acceptor moieties that are glycosylated by the glycosyltransferase of interest (e.g., facosyltransferase). For example, in the case of a ⁇ 1,2 fucosyltransferase, a substantially uniform fucosylation pattern exists if substantially all (as defined below) of the Gal ⁇ 1,4-GlcNAc-R and sialylated analogues thereof are fucosylated in a peptide conjugate of the invention.
  • the starting material may contain glycosylated acceptor moieties (e.g., fucosylated Gal ⁇ 1,4-GlcNAc-R moieties).
  • glycosylated acceptor moieties e.g., fucosylated Gal ⁇ 1,4-GlcNAc-R moieties.
  • the calculated percent glycosylation will include acceptor moieties that are glycosylated by the methods of the invention, as well as those acceptor moieties already glycosylated in the starting material.
  • substantially in the above definitions of “substantially uniform” generally means at least about 40%, at least about 70%, at least about 80%, or more preferably at least about 90%, and still more preferably at least about 95% of the acceptor moieties for a articular glycosyltransferase are glycosylated.
  • the present invention includes methods and compositions for the in vitro addition and/or deletion of sugars to or from a glycopeptide molecule in such a manner as to provide a peptide molecule having a specific customized or desired glycosylation pattern, preferably including the addition of a modified sugar thereto.
  • a key feature of the invention therefore is to take a peptide produced by any cell type and generate a core glycan structure on the peptide, following which the glycan structure is then remodeled in vitro to generate a peptide having a glycosylation pattern suitable for therapeutic use in a mammal.
  • glycosylation pattern of a peptide is well known in the art as are the limitations of present in vivo methods for the production of properly glycosylated peptides, particularly when these peptides are produced using recombinant DNA methodology. Moreover, until the present invention, it has not been possible to generate glycopeptides having a desired glycan structure thereon, wherein the peptide can be produced at industrial scale.
  • a peptide produced by a cell is enzymatically treated in vitro by the systematic addition of the appropriate enzymes and substrates therefor, such that sugar moieties that should not be present on the peptide are removed, and sugar moieties, optionally including modified sugars, that should be added to the peptide are added in a manner to provide a glycopeptide having “desired glycosylation”, as defined elsewhere herein.
  • the present invention takes advantage of the fact that most peptides of commercial or pharmaceutical interest comprise a common five sugar structure referred to herein as the trimannosyl core, which is N-linked to asparagine at the sequence Asn-X-Ser/Thr on a peptide chain.
  • the elemental trimannosyl core consists essentially of two N-acetylglucosamine (GlcNAc) residues and three mannose (Man) residues attached to a peptide, i.e., it comprises these five sugar residues and no additional sugars, except that it may optionally include a fucose residue.
  • the first GlcNAc is attached to the amide group of the asparagine and the second GlcNAc is attached to the first via a ⁇ 1,4 linkage.
  • a mannose residue is attached to the second GlcNAc via a ⁇ 1,4 linkage and two mannose residues are attached to this mannose via an ⁇ 1,3 and an ⁇ 1,6 linkage respectively.
  • a schematic depiction of a trimannosyl core structure is shown in FIG. 1 , left side. While it is the case that glycan structures on most peptides comprise other sugars in addition to the trimannosyl core, the trimannosyl core structure represents an essential feature of N-linked glycans on mammalian peptides.
  • the present invention includes the generation of a peptide having a trimannosyl core structure as a fundamental element of the structure of the glycan molecules contained thereon.
  • the present invention provides methods whereby a glycan molecule on a peptide produced in any cell type can be reduced to an elemental trimannosyl core structure.
  • the elemental trimannosyl core structure Once the elemental trimannosyl core structure has been generated then it is possible using the methods described herein, to generate in vitro, a desired glycan structure on the peptide which confers on the peptide one or more properties that enhances the therapeutic effectiveness of the peptide.
  • trimannosyl core is used to describe the glycan structure shown in FIG. 1 , left side. Glycopeptides having a trimannosyl core structure may also have additional sugars added thereto, and for the most part, do have additional structures added thereto irrespective of whether the sugars give rise to a peptide having a desired glycan structure.
  • the term “elemental trimannosyl core structure” is defined elsewhere herein. When the term “elemental” is not included in the description of the “trimannosyl core structure,” then the glycan comprises the trimannosyl core structure with additional sugars attached to the mannose sugars.
  • elemental trimannosyl core glycopeptide is used herein to refer to a glycopeptide having glycan structures comprised primarily of an elemental trimannosyl core structure. However, it may also optionally contain a fucose residue attached thereto. As discussed herein, elemental trimannosyl core glycopeptides are one optimal, and therefore preferred, starting material for the glycan remodeling processes of the invention.
  • Another optimal starting material for the glycan remodeling process of the invention is a glycan structure having a trimannosyl core wherein one or two additional GlcNAc residues are added to each of the ⁇ 1,3 and the ⁇ 1,6 mannose residues (see for example, the structure on the second line of FIG. 2 , second structure in from the left of the figure).
  • This structure is referred to herein as “Man3GlcNAc4.”
  • the structure is monoantenary, the structure is referred to herein as “Man3GlcNAc3.”
  • this structure may also contain a core fucose molecule.
  • Man3GlcNAc3 or Man3GlcNAc4 structure has been generated then it is possible using the methods described herein, to generate in vitro, a desired glycan structure on the glycopeptide which confers on the glycopeptide one or more properties that enhances the therapeutic effectiveness of the peptide.
  • the N-linked glycopeptides of the invention are N-linked glycosylated with a trimannosyl core structure and one or more sugars attached thereto.
  • glycopeptide and “glycopolypeptide” are used synonymously herein to refer to peptide chains having sugar moieties attached thereto. No distinction is made herein to differentiate small glycopolypeptides or glycopeptides from large glycopolypeptides or glycopeptides. Thus, hormone molecules having very few amino acids in their peptide chain (e.g., often as few as three amino acids) and other much larger peptides are included in the general terms “glycopolypeptide” and “glycopeptide,” provided they have sugar moieties attached thereto. However, the use of the term “peptide” does not preclude that peptide from being a glycopeptide.
  • N-linked glycopeptide having desired glycosylation is a peptide having an N-linked glycan having a trimannosyl core with at least one GlcNAc residue attached thereto. This residue is added to the trimannosyl core using N-acetyl glucosaminyltransferase I (GnT-I). If a second GlcNAc residue is added, N-acetyl glucosaminyltransferase II (GnT-II) is used.
  • GnT-III N-acetyl glucosaminyltransferase III
  • this structure may be extended by treatment with ⁇ 1,4 galactosyltransferase to add a galactose residue to each non-bisecting GlcNAc, and even further optionally, using ⁇ 2,3 or ⁇ 2,6-sialyltransferase enzymes, sialic acid residues may be added to each galactose residue.
  • a bisecting GlcNAc to the glycan is not required for the subsequent addition of galactose and sialic acid residues; however, with respect to the substrate affinity of the rat and human GnT-III enzymes, the presence of one or more of the galactose residues on the glycan precludes the addition of the bisecting GlcNAc in that the galactose-containing glycan is not a substrate for these forms of GnT-III.
  • GnT-III it is important should the glycan contain added galactose and/or sialic residues, that they are removed prior to the addition of the bisecting GlcNAc.
  • Other forms of GnT-III may not require this specific order of substrates for their activity.
  • a mixture of GNT-I, GnT-II and GNT-III is added to the reaction mixture so that the GlcNAc residues can be added in any order.
  • glycan structures which represent the various aspects of peptides having “desired glycosylation” are shown in the drawings provided herein. The precise procedures for the in vitro generation of a peptide having “desired glycosylation” are described elsewhere herein. However, the invention should in no way be construed to be limited solely to any one glycan structure disclosed herein. Rather, the invention should be construed to include any and all glycan structures which can be made using the methodology provided herein.
  • an elemental trimannosyl core alone may constitute the desired glycosylation of a peptide.
  • a peptide having only a trimannosyl core has been shown to be a useful component of an enzyme employed to treat Gaucher disease (Mistry et al., 1966, Lancet 348: 1555-1559; Bijsterbosch et al., 1996, Eur. J. Biochem. 237:344-349).
  • glycopeptide having one or more glycan molecules which have as a common feature a trimannosyl core structure and at least one or more of a heterogeneous or homogeneous mixture of one or more sugars added thereto, it is possible to increase the proportion of glycopeptides having an elemental trimannosyl core structure as the sole glycan structure or which have Man3GlcNAc3 or Man3GlcNAc4 as the sole glycan structure.
  • a peptide having an elemental trimannosyl core structure as the sole glyean structure on the peptide by isolating a naturally occurring cell whose glycosylation machinery produces such a peptide.
  • DNA encoding a peptide of choice is then transfected into the cell wherein the DNA is transcribed, translated and glycosylated such that the peptide of choice has an elemental trimannosyl core structure as the sole glycan structure thereon.
  • a cell lacking a functional GnT-I enzyme will produce several types of glycopeptides. In some instances, these will be glycopeptides having no additional sugars attached to the trimannosyl core.
  • the peptides produced may have two additional mannose residues attached to the trimannosyl core, resulting in a Man5 glycan.
  • This is also a desired starting material for the remodeling process of the present invention. Specific examples of the generation of such glycan structures are described herein.
  • a cell it is possible to genetically engineer a cell to confer upon it a specific glycosylation machinery such that a peptide having an elemental trimannosyl core or Man3GlcNAc3 or Man3GlcNAc4 structure as the sole glycan structure on the peptide is produced.
  • DNA encoding a peptide of choice is then transfected into the cell wherein the DNA is transcribed, translated and glycosylated such that the peptide of choice has an increased number of glycans comprising solely an elemental trimannosyl core structure.
  • certain types of cells that are genetically engineered to lack GnT-I may produce a glycan having an elemental trimannosyl core structure, or, depending on the cell, may produce a glycan having a trimannosyl core plus two additional mannose residues attached thereto (Man5).
  • the cell may be further genetically engineered to express mannosidase 3 which cleaves off the two additional mannose residues to generate the trimannosyl core.
  • the Man5 glycan may be incubated in vitro with mannosidase 3 to have the same effect.
  • the glycan on the peptide comprises a partially complex chain.
  • Insect cells also express hexosaminidase in the cells which trims the partially complex chain back to a trimannosyl core structure which can then be remodeled as described herein.
  • the cells described in b) and c) produce peptides having 100% elemental trirnannosyl core structures (i.e., having no additional sugars attached thereto) or 100% of Man3GlcNAc3 or Man3GlcNAc4 structures
  • the cells in fact produce a heterogeneous mixture of peptides having, in combination, elemental trimannosyl core structures, or Man3GlcNAc3 or Man3GlcNAc4 structures, as the sole glycan structure in addition to these structures having additional sugars attached thereto.
  • the proportion of peptides having a trimannosyl core or Man3 GlcNAc3 or Man3 GlcNAc4 structures having additional sugars attached thereto, as opposed to those having one structure, will vary depending on the cell which produces them.
  • the complexity of the glycans i.e. which and how many sugars are attached to the trimannosyl core) will also vary depending on the cell which produces them.
  • a glycopeptide having an elemental trimannosyl core or a trimannosyl core with one or two GlcNAc residues attached thereto is produced by following a), b) or c) above, according to the present invention, additional sugar molecules are added in vitro to the trimannosyl core structure to generate a peptide having desired glycosylation (i.e., a peptide having an in vitro customized glycan structure).
  • a peptide having an elemental trimannosyl core or Man3GlcNAc4 structure with some but not all of the desired sugars attached thereto is produced, then it is only necessary to add any remaining desired sugars without reducing the glycan structure to the elemental trimannosyl core or Man3GlcNAc4 structure. Therefore, in some cases, a peptide having a glycan structure having a trimannosyl core structure with additional sugars attached thereto, will be a suitable substrate for remodeling.
  • the elemental trimannosyl core or Man3GlcNAc3 or Man3GlcNAc4 glycopeptides of the invention may be isolated and purified, if necessary, using techniques well known in the art of peptide purification. Suitable techniques include chromatographic techniques, isoelectric focusing techniques, ultrafiltration techniques and the like. Using any such techniques, a composition of the invention can be prepared in which the glycopeptides of the invention are isolated from other peptides and from other components normally found within cell culture media. The degree of purification can be, for example, 90% with respect to other peptides or 95%, or even higher, e.g., 98%. See, e.g., Deutscher et al. (ed., 1990, Guide to Protein Purification, Harcourt Brace Jovanovich, San Diego).
  • heterogeneity of N-linked glycans present in the glycopeptides produced by the prior art methodology generally only permits the isolation of a small portion of the target glycopeptides which can be modified to produce desired glycopeptides.
  • large quantities of elemental trimannosyl core glycopeptides and other desired glycopeptides, including Man3GlcNAc3 or Man3GlcNAc4 glycans can be produced which can then be further modified to generate large quantities of peptides having desired glycosylation.
  • a key feature of the invention which is described in more detail below, is that once a core glycan structure is generated on any peptide, the glycan structure is then remodeled in vitro to generate a peptide having desired glycosylation that has improved therapeutic use in a mammal.
  • the mammal may be any type of suitable mammal, and is preferably a human.
  • peptides produced in cells may be treated in vitro with a variety of enzymes which catalyze the cleavage of sugars that should not be present on the glycan and the addition of sugars which should be present on the glycan such that a peptide having desired glycosylation and thus suitable for therapeutic use in mammals is generated.
  • the generation of different glycoforms of peptides in cells is described above.
  • the starting material i.e., the peptide produced by a cell may differ from one cell type to another.
  • the starting material it is not necessary that the starting material be uniform with respect to its glycan composition.
  • the starting material it is preferable that the starting material be enriched for certain glycoforms in order that large quantities of end product, i.e., correctly glycosylated peptides are produced.
  • the degradation and synthesis events that result in a peptide having desired glycosylation involve at some point, the generation of an elemental trimannosyl core structure or a Man3GlcNAc3 or Man3GlcNAc4 structure on the peptide.
  • the present invention also provides means of adding one or more selected glycosyl residues to a peptide, after which a modified sugar is conjugated to at least one of the selected glycosyl residues of the peptide.
  • the present embodiment is useful, for example, when it is desired to conjugate the modified sugar to a selected glycosyl residue that is either not present on a peptide or is not present in a desired amount.
  • the selected glycosyl residue prior to coupling a modified sugar to a peptide, the selected glycosyl residue is conjugated to the peptide by enzymatic or chemical coupling.
  • the glycosylation pattern of a peptide is altered prior to the conjugation of the modified sugar by the removal of a carbohydrate residue from the peptide. See for example WO 98/31826.
  • Enzymatic cleavage of carbohydrate moieties on peptide variants can be achieved by the use of a variety of endo- and exo-glycosidases as described by Thotakura et al., 1987, Meth. Enzymol. 138: 350.
  • glycosyl moieties Chemical addition of glycosyl moieties is carried out by any art-recognized method. Enzymatic addition of sugar moieties is preferably achieved using a modification of the methods set forth herein, substituting native glycosyl units for the modified sugars used in the invention. Other methods of adding sugar moieties are disclosed in U.S. Pat. Nos. 5,876,980, 6,030,815, 5,728,554, and 5,922,577.
  • Exemplary attachment points for selected glycosyl residue include, but are not limited to: (a) sites for N— and O-glycosylation; (b) terminal glycosyl moieties that are acceptors for a glycosyltransferase; (c) arginine, asparagine and histidine; (d) free carboxyl groups; (e) free sulfhydryl groups such as those of cysteine; (f) free hydroxyl groups such as those of serine, threonine, or hydroxyproline; (g) aromatic residues such as those of phenylalanine, tyrosine, or tryptophan; or (h) the amide group of glutamine. Exemplary methods of use in the present invention are described in WO 87/05330 published Sep. 11, 1987, and in Aplin and Wriston, CRC Crit. Rev. Biochem., pp. 259-306 (1981).
  • FIG. 1 there is shown the structure of an elemental trimannosyl core glycan on the left side. It is possible to convert this structure to a complete glycan structure having a bisecting GlcNAc by incubating the elemental trimannosyl core structure in the presence of GnT-I, followed by GnT-II, and further followed by GnT-III, and a sugar donor comprising UDP-GlcNAc, wherein GlcNAc is sequentially added to the elemental trimannosyl core structure to generate a trimannosyl core having a bisecting GlcNAc.
  • the bisecting GlcNAc structure may be produced by adding a mixture of GnT-I, GnT-II and GnT-III and UDP-GlcNAc to the reaction mixture
  • FIG. 3 there is shown the conversion of a bisecting GlcNAc containing trimannosyl core glycan to a complex glycan structure comprising galactose and N-acetyl neuraminic acid.
  • the bisecting GlcNAc containing trimannosyl core glycan is first incubated with galactosyltransferase and UDP-Gal as a donor molecule, wherein two galactose residues are added to the peripheral GlcNAc residues on the molecule.
  • the enzyme NeuAc-transferase is then used to add two NeuAc residues one to each of the galactose residues.
  • FIG. 4 there is shown the conversion of a high mannose glycan structure to an elemental trimannosyl core glycan.
  • the high mannose glycan (Man9) is incubated sequentially in the presence of the mannosidase 1 to generate a Man5 structure and then in the presence of mannosidase 3, wherein all but three mannose residues are removed from the glycan.
  • incubation of the Man9 structure may be trimmed back to the trimannosyl core structure solely by incubation in the presence of mannosidase 3.
  • conversion of this elemental trimannosyl core glycan to a complex glycan molecule is then possible.
  • FIG. 5 there is shown a typical complex N-linked glycan structure produced in plant cells. It is important to note that when plant cells are deficient in GnT-I enzymatic activity, xylose and fucose cannot be added to the glycan. Thus, the use of GnT-I knock-out cells provides a particular advantage in the present invention in that these cells produce peptides having an elemental trimannosyl core onto which additional sugars can be added without performing any “trimming back” reactions. Similarly, in instances where the structure produced in a plant cell may be of the Man5 variety of glycan, if GnT-I is absent in these cells, xylose and fucose cannot be added to the structure.
  • Man5 structure may be trimmed back to an elemental trimannosyl core (Man3) using mannosidase 3.
  • Man3 elemental trimannosyl core
  • mannosidase 3 mannosidase 3.
  • FIG. 6 there is shown a typical complex N-linked glycan structure produced in insect cells.
  • additional sugars such as, for example, fucose may also be present.
  • insect cells may produce high mannose glycans having as many as nine mannose residues and may have additional sugars attached thereto.
  • GnT-I knock out cells prevent the addition of fucose residues to the glycan.
  • production of a peptide in insect cells may preferably be accomplished in a GnT-I knock out cell.
  • the glycan thus produced may then be trimmed back in vitro if necessary using any of the methods and schemes described herein, and additional sugars may be added in vitro thereto also using the methods and schemes provided herein.
  • FIG. 2 there is shown glycan structures in various stages of completion. Specifically, the in vitro enzymatic generation of an elemental trimannosyl core structure from a complex carbohydrate glycan structure which does not contain a bisecting GlcNAc residue is shown. Also shown is the generation of a glycan structure therefrom which contains a bisecting GlcNAc. Several intermediate glycan structures which can be produced are shown. These structures can be produced by cells, or can be produced in the in vitro trimming back reactions described herein. Sugar moieties may be added in vitro to the elemental trirannosyl core structure, or to any suitable intermediate structure in order that a desired glycan is produced.
  • FIG. 7 there is shown a series of possible in vitro reactions which can be performed to trim back and add onto glycans beginning with a high mannose structure.
  • a Man9 glycan may be trimmed using mannosidase 1 to generate a Man5 glycan, or it may be trimmed to a trimannosyl core using mannosidase 3 or one or more microbial mannosidases.
  • GnT-1 and or GnT-II may then be used to transfer additional GlcNAc residues onto the glycan.
  • the situation which would not occur when the glycan molecule is produced in a cell that does not have GNT-I see shaded box).
  • fucose and xylose may be added to a glycan only when GNT-I is active and facilitates the transfer of a GlcNAc to the molecule.
  • FIG. 8 depicts well known strategies for the synthesis of biantennary, triantennary and even tetraantennary glycan structures beginning with the trimannosyl core structure. According to the methods of the invention, it is possible to synthesize each of these structures in vitro using the appropriate enzymes and reaction conditions well known in the art of glycobiology.
  • FIG. 9 depicts two methods for synthesis of a monoantennary glycan structure beginning from a high mannose (6 to 9 mannose moieties) glycan structures.
  • a terminal sialic acid-PEG moiety may be added in place of the sialic acid moiety in accordance with glycoPEGylation methodology described herein.
  • endo-H is used to cleave the glycan structure on the peptide back to the first GlcNAc residue.
  • Galactose is then added using galactosyltransferase and sialylated-PEG is added as described elsewhere herein.
  • mannosidase I is used to cleave mannose residues from the glycan structure in the peptide.
  • a galactose residue is added to one arm of the remaining mannose residues which were cleaved off the glycan using Jack Bean ⁇ -mannosidase.
  • Sialylated-PEG is then added to this structure as directed.
  • FIG. 10 depicts two additional methods for synthesis of a monoantennary glycan structures beginning from high mannose (6 to 9 mannose moieties) glycan structure.
  • a terminal sialic acid-PEG moiety may be added in place of the sialic acid moiety in accordance with the glycoPEGylation methodology described herein.
  • some of the mannose residues from the arm to which sialylated-PEG is not added, are removed.
  • FIG. 11 there is shown a scheme for the synthesis of yet more complex carbohydrate structures beginning with a trimannosyl core structure.
  • a scheme for the in vitro production of Lewis x and Lewis a antigen structures which may or may not be sialylated is shown.
  • Such structures when present on a peptide may confer on the peptide immunological advantages for upregulating or downregulating the immune response.
  • such structures are useful for targeting the peptide to specific cells, in that these types of structures are involved in binding to cell adhesion peptides and the like.
  • FIG. 12 is an exemplary scheme for preparing an array of O-linked peptides originating with serine or threonine.
  • FIG. 13 is a series of diagrams depicting the four types of O-linked glycan structure termed cores 1 through 4.
  • the core structure is outlined in dotted lines.
  • Sugars which may also be included in this structure include sialic acid residues added to the galactose residues, and fucose residues added to the GlcNAc residues.
  • the present invention provides a method of making an N-linked glycosylated glycopeptide by providing an isolated and purified glycopeptide to which is attached an elemental trimannosyl core or a Man3GlcNAc4 structure, contacting the glycopeptide with a glycosyltransferase enzyme and a donor molecule having a glycosyl moiety under conditions suitable to transfer the glycosyl moiety to the glycopeptide.
  • Customization of a trimannosyl core glycopeptide or Man3GlcNAc4 glycopeptide to produce a peptide having a desired glycosylation pattern is then accomplished by the sequential addition of the desired sugar moieties, using techniques well known in the art.
  • an N-linked glycopeptide When an N-linked glycopeptide is produced by a cell, as noted elsewhere herein, it may comprise a heterogeneous mixture of glycan structures which must be reduced to a common, generally elemental trimannosyl core or Man3GlcNAc4 structure, prior to adding other sugar moieties thereto. In order to determine exactly which sugars should be removed from any particular glycan structure, it is sometimes necessary that the primary glycan structure be identified. Techniques for the determination of glycan primary structure are well know in the art and are described in detail, for example, in Montreuil, “Structure and Biosynthesis of Glycopeptides” In Polysaccharides in Medicinal Applications, pp. 273-327, 1996, Eds.
  • efficient methods are available for (i) the splitting of glycosidic bonds either by chemical cleavage such as hydrolysis, acetolysis, hydrazinolysis, or by nitrous deamination; (ii) complete methylation followed by hydrolysis or methanolysis and by gas-liquid chromatography and mass spectroscopy of the partially methylated monosaccharides; and (iii) the definition of anomeric linkages between monosaccharides using exoglycosidases, which also provide insight into the primary glycan structure by sequential degradation.
  • the techniques of mass spectroscopy and nuclear magnetic resonance (NMR) spectrometry, especially high field NMR have been successfully used to determine glycan primary structure.
  • Kits and equipment for carbohydrate analysis are also commercially available.
  • Fluorophore Assisted Carbohydrate Electrophoresis FACE® is available from Glyko, Inc. (Novato, Calif.).
  • FACE Fluorophore Assisted Carbohydrate Electrophoresis
  • glycoconjugates are released from the peptide with either Endo H or N-glycanase (PNGase F) for N-linked glycans, or hydrazine for Ser/Thr linked glycans.
  • PNGase F N-glycanase
  • hydrazine for Ser/Thr linked glycans.
  • the glycan is then labeled at the reducing end with a fluorophore in a non-structure discriminating manner.
  • the fluorophore labeled glycans are then separated in polyacrylamide gels based on the charge/mass ratio of the saccharide as well as the hydrodynamic volume. Images are taken of the gel under UV light and the composition of the glycans are determined by the migration distance as compared with the standards. Oligosaccharides can be sequenced in this manner by analyzing migration shifts due to the sequential removal of saccharides by exoglycosidase digestion.
  • X 3 , X 4 , X 5 , X 6 , X 7 and X 17 are (independently selected) monosaccharide or oligosaccharide residues;
  • a, b, c, d, e and x are (independently selected) 0, 1 or 2, with the proviso that at least one member selected from a, b, c, d, e and x are 1 or 2.
  • Formula 1 describes glycan structure comprising the tri-mannosyl core, which is preferably covalently linked to an asparagine residue on a peptide backbone.
  • Preferred expression systems will express and secrete exogenous peptides with N-linked glycans comprising the tri-mannosyl core.
  • the remodeling method of the invention the glycan structures on these peptides can be conveniently remodeled to any glycan structure desired. Exemplary reaction conditions are found throughout the examples and in the literature.
  • the glycan structures are remodeled so that the structure described in Formula 1 has specific determinates.
  • the structure of the glycan can be chosen to enhance the biological activity of the peptide, give the peptide a new biological activity, remove the biological activity of peptide, or better approximate the glycosylation pattern of the native peptide, among others.
  • the peptide N-linked glycans are remodeled to better approximate the glycosylation pattern of native human proteins.
  • This embodiment is particularly advantageous for human peptides expressed in heterologous cellular expression systems.
  • the peptide can be made less immunogenic in a human patient, and/or more stable, among others.
  • the peptide N-linked glycans are remodeled to have a bisecting GlcNAc residue on the tri-mannosyl core.
  • the glycan structure described in Formula 1 is remodeled to have the following moieties: X 3 and X 5 are
  • This embodiment is particularly advantageous for recombinant antibody molecules expressed in heterologous cellular systems.
  • the antibody molecule includes a Fc-mediated cellular cytotoxicity, it is known that the presence of bisected oligosaccharides linked the Fc domain dramatically increased antibody-dependent cellular cytotoxicity.
  • the peptide N-linked glycans are remodeled to have a sialylated Lewis X moiety.
  • the peptide N-linked glycans are remodeled to have a conjugated moiety.
  • the conjugated moiety may be a PEG molecule, another peptide, a small molecule such as a drug, among others.
  • the glycan structure described in Formula 1 is remodeled to have the following moieties: X 3 and X 5 are
  • the conjugated moiety may be a PEG molecule, another peptide, a small molecule such as a drug, among others.
  • This embodiment therefore is useful for conjugating the peptide to PEG molecules that will slow the clearance of the peptide from the patient's bloodstream, to peptides that will target both peptides to a specific tissue or cell, or to another peptide of complementary therapeutic use.
  • the invention is not limited to the preferred glycan molecules described above.
  • the preferred embodiments are only a few of the many useful glycan molecules that can be made by the remodeling method of the invention. Those skilled in the art will know how to design other useful glycans.
  • the peptide is expressed in a CHO (Chinese hamster ovarian cell line) according to methods well known in the art.
  • a peptide with N-linked glycan consensus sites is expressed and secreted from CHO cells, the N-linked glycans will have the structures depicted in top row of FIG. 2 , but also comprising a core fucose. While all of these structures may be present, by far the most common structures are the two at the right side.
  • X 3 and X 5 are
  • the N-linked glycans of peptides expressed in CHO cells are remodeled to the preferred humanized glycan by contacting the peptides with a glycosyltransferase that is specific for a galactose acceptor molecule and a sialic acid donor molecule. This process is illustrated in FIG. 2 and Example 17.
  • the N-linked glycans of a peptide expressed and secreted from CHO cells are remodeled to be the preferred PEGylated structures.
  • the peptide is first contacted with a glycosidase specific for sialic acid to remove the terminal SA moiety, and then contacted with a glycosyltransferase specific for a galactose acceptor moiety and an sialic acid acceptor moiety, in the presence of PEG-sialic acid-nucleotide donor molecules.
  • the peptide may then be contacted with a glycosyltransferase specific for a galactose acceptor moiety and an sialic acid acceptor moiety, in the presence of sialic acid-nucleotide donor molecules to ensure complete the SA capping of all of the glycan molecules.
  • the peptide is expressed in insect cells, such as the sf9 cell line, according to methods well known in the art.
  • insect cells such as the sf9 cell line
  • the N-linked glycans will often have the structures depicted in top row of FIG. 6 .
  • X 3 and X 5 are
  • trimannose core is present in the vast majority of the N-linked glycans made by insect cells, and sometimes an antennary GlcNAc and/or fucose residue(s) are also present.
  • the glycan may have no core fucose, it may have a single core fucose having either linkage, or it may have a single core fucose with a perponderance of a single linkage.
  • the N-linked glycans of a peptide expressed and secreted from insect cells is remodeled to the preferred humanized glycan by first contacting the glycans with a glycosidase specific to fucose molecules, then contacting the glycans with a glycosyltransferases specific to the mannose acceptor molecule on each antennary of the trimannose core, a GlcNAc donor molecule in the presence of nucleotide-GlcNAc molecules; then contacting the glycans with a glycosyltransferase specific to a GlcNAc acceptor molecule, a Gal donor molecule in the presence of nucleotide-Gal molecules; and then contacting the glyeans with a glycosyltransferase specific to a galactose acceptor molecule, a sialic acid donor molecule in the presence of nucleotide-SA molecules.
  • the fucose molecules can be removed at any time during the procedure, and if the core fucose is of the same alpha 1,6 linkage as found in human glycans, it may be left intact.
  • the humanized glycan of the previous example is remodeled further to the sialylated Lewis X glycan by contacting the glycan further with a glycosyltransferase specific to a GlcNAc acceptor molecule, a fucose donor molecule in the presence of nucleotide-fucose molecules. This process is illustrated in FIG. 11 and Example 39.
  • the peptide is expressed in yeast, such as Saccharomyces cerevisiae, according to methods well known in the art.
  • yeast such as Saccharomyces cerevisiae
  • the N-linked glycans will have the structures depicted at the left in FIG. 4 .
  • the N-linked glycans will always have the trimannosyl core, which will often be elaborated with mannose or related polysaccharides of up to 1000 residues.
  • X 3 and X 5
  • the N-linked glycans of a peptide expressed and secreted from yeast cells are remodeled to the elemental trimannose core by first contacting the glycans with a glycosidase specific to ⁇ 2 mannose molecules, then contacting the glycans with a glycosidase specific to ⁇ 6 mannose molecules. This process is illustrated in FIG. 4 and Example 38.
  • the N-linked glycans are further remodeled to make a glycan suitable for an recombinant antibody with Fc-mediated cellular toxicity function by contacting the elemental trimannose core glycans with a glycosyltransferase specific to the mannose acceptor molecule on each antennary of the trimannose core and a GlcNAc donor molecule in the presence of nucleotide-GlcNAc molecules.
  • the glycans are contacted with a glycosyltransferase specific to the acceptor mannose molecule in the middle of the trimannose core, a GlcNAc donor molecule in the presence of nucleotide-GlcNAc molecules and further contacting the glycans with a glycosyltransferase specific to a GlcNAc acceptor molecule, a Gal donor molecule in the presence of nucleotide-Gal molecules; and then optionally contacting the glycans with a glycosyltransferase specific to a galactose acceptor molecule and further optionally a sialic acid donor molecule in the presence of nucleotide-SA molecules.
  • This process is illustrated in FIGS. 1, 2 and 3 .
  • the peptide is expressed in bacterial cells, in particular E. coli cells, according to methods well known in the art.
  • E. coli cells When a peptide with N-linked glycans consensus sites is expressed in E. coli cells, the N-linked consensus sites will not be glycosylated.
  • a humanized glycan molecule is built out from the peptide backbone by contacting the peptides with a glycosyltransferase specific for a N-linked consensus site and a GlcNAc donor molecule in the presence of nucleotide-GlcNAc; and further sequentially contacting the growing glyeans with glycosyltransferases specific for the acceptor and donor moieties in the present of the required donor moiety until the desired glycan structure is completed.
  • the mature peptide is likely not to be glycosylated.
  • the peptide may be given N-linked glycosylation by building out from the peptide N-linked consensus site as aforementioned.
  • a protein is chemically modified with a sugar moiety, it can be built out as aforementioned.
  • O-glycosylation is characterized by the attachment of a variety of monosaccharides in an O-glycosidic linkage to hydroxy amino acids. O-glycosylation is a widespread post-translational modification in the animal and plant kingdoms. The structural complexity of glycans O-linked to proteins vastly exceeds that of N-linked glycans. Serine or threonine residues of a newly translated peptide become modified by virtue of a peptidyl GalNAc transferase in the cis to trans compartments of the Golgi.
  • the site of O-glycosylation is determined not only by the sequence specificity of the glycosyltransferase, but also epigenetic regulation mediated by competition between different substrate sites and competition with other glycosyltransferases responsible for forming the glycan.
  • the O-linked glycan has been arbitrarily defined as having three regions: the core, the backbone region and the peripheral region.
  • the “core” region of an O-linked glycan is the inner most two or three sugars of the glycan chain proximal to the peptide.
  • the backbone region mainly contributes to the length of the glycan chain formed by uniform elongation.
  • the peripheral region exhibits a high degree of structural complexity.
  • the structural complexity of the O-linked glycans begins with the core structure. In most cases, the first sugar residue added at the O-linked glycan consensus site is GalNAc; however the sugar may also be GlcNAc, glucose, mannose, galactose or fucose, among others.
  • FIG. 12 is a diagram of some of the known O-linked glycan core structures and the enzymes responsible for their in vivo synthesis.
  • O-linked core structures In mammalian cells, at least eight different O-linked core structures are found, all based on a core- ⁇ -GalNAc residue. The four core structures depicted in FIG. 13 are the most common. Core 1 and core 2 are the most abundant structures in mammalian cells, and core 3 and core 4 are found in more restricted, organ-characteristic expression systems.
  • O-linked glycans are reviewed in Montreuil, Structure and Synthesis of Glycopeptides, In Polysaccharides in Medicinal Applications, pp. 273-327, 1996, Eds. Severian Damitriu, Marcel Dekker, NY, and in Schachter and Brockhausen, The Biosynthesis of Branched O-Linked Glycans, 1989, Society for Experimental Biology, pp. 1-26 (Great Britain).
  • O-glycans differ from N-glycans in that they are linked to a serine or threonine residue rather than an asparagine residue.
  • hydrolytic enzymes can be used to cleave unwanted sugar moieties in an O-linked glycan and additional desired sugars can then be added thereto, to build a customized O-glycan structure on the peptide (See FIGS. 12 and 13 ).
  • the initial step in O-glycosylation in mammalian cells is the attachment of N-acetylgalactosamine (GalNAc) using any of a family of at least eleven known ⁇ -N-acetylgalactosaminyltransferases, each of which has a restricted acceptor peptide specificity.
  • the acceptor peptide recognized by each enzyme constitutes a sequence of at least ten amino acids.
  • Peptides that contain the amino acid sequence recognized by one particular GalNAc-transferase become O-glycosylated at the acceptor site if they are expressed in a cell expressing the enzyme and if they are appropriately localized to the Golgi apparatus where UDP-GalNAc is also present.
  • the ⁇ -N-acetylgalactosaminyltransferase enzyme native to the expressing cell may have a consensus sequence specificity which differs from that of the recombinant peptide being expressed.
  • the desired recombinant peptide may be expressed in a bacterial cell, such as E. coli, that does not synthesize glycan chains.
  • a bacterial cell such as E. coli
  • the GalNAc moiety can be introduced in vitro onto the peptide once the recombinant peptide has been recovered in a soluble form, by contacting the peptide with the appropriate GalNAc transferase in the presence of UDP-GalNAc.
  • an additional sequence of amino acids that constitute an effective acceptor for transfer of an O-linked sugar may be present.
  • Such an amino acid sequence is encoded by a DNA sequence fused in frame to the coding sequence of the peptide, or alternatively, may be introduced by chemical means.
  • the peptide may be otherwise lacking glycan chains.
  • the peptide may have N— and/or O-linked glycan chains but require an additional glycosylation site, for example, when an additional glycan substituent is desired.
  • the amino acid sequence PTTTK-COOH which is the natural GalNAc acceptor sequence in the human mucin MUC-1, is added as a fusion tag.
  • the fusion protein is then expressed in E. coli and purified.
  • the peptide is then contacted with recombinant human GalNAc-transferases T3 or T6 in the presence of UDP-GalNAc to transfer a GalNAc residue onto the peptide in vitro.
  • This glycan chain on the peptide may then be further elongated using the methods described in reference to the N-linked or O-linked glycans herein.
  • the GalNAc transferase reaction can be carried out in the presence of UDP-GalNAc to which PEG is covalently substituted in the O-3, 4, or 6 positions or the N-2 position. Glycoconjugation is described in detail elswhere herein. Any antigenicity introduced into the peptide by the new peptide sequence can be conveniently masked by PEGylation of the associated glycan.
  • the acceptor site fusion technique can be used to introduce not only a PEG moiety, but to introduce other glycan and non-glycan moieties, including, but not limited to, toxins, anti-infectives, cytotoxic agents, chelators for radionucleotides, and glycans with other functionalities, such as tissue targeting.
  • O-linked glycosylation is best illustrated with reference to Formula Formula 2 describes a glycan structure comprising a GalNAc which is covalently linked preferably to a serine or threonine residue on a peptide backbone. While this structure is used to illustrate the most common forms of O-linked glycans, it should not be construed to limit the invention solely to these O-linked glycans. Other forms of O-linked glycans are illustrated in FIG. 12 . Preferred expression systems useful in the present invention express and secrete exogenous peptides having O-linked glycans comprising the GalNAc residue.
  • the glycan structures on these peptides can be conveniently remodeled to generate any desired glycan structure.
  • O-linked glycans can be remodeled using the same principles, enzymes and reaction conditions as those available in the art once armed with the present disclosure. Exemplary reaction conditions are found throughout the Examples.
  • the glycan structures are remodeled so that the structure described in Formula 2 has specific moieties.
  • the structure of the glycan may be chosen to enhance the biological activity of the peptide, confer upon the peptide a new biological activity, remove or alter a biological activity of peptide, or better approximate the glycosylation pattern of the native peptide, among others.
  • the peptide O-linked glycans are remodeled to better approximate the glycosylation pattern of native human proteins.
  • the glycan structure described in Formula 2 is remodeled to have the following moieties: X 2 is
  • This embodiment is particularly advantageous for human peptides expressed in heterologous cellular expression systems.
  • the peptide O-linked glycans are remodeled to display a sialylated Lewis X antigen.
  • the glycan structure described in Formula 2 is remodeled to have the following moieties: X 2 is
  • This embodiment is particularly advantageous when the peptide which is being remodeled is most effective when targeted to a selectin molecule and cells exhibiting the same.
  • the peptide O-linked glycans are remodeled to contain a conjugated moiety.
  • the conjugated moiety may be a PEG molecule, another peptide, a small molecule such as a drug, among others.
  • the glycan structure described in Formula 2 is remodeled to have the following moieties: X 2 is
  • This embodiment is useful for conjugating the peptide to PEG molecules that will slow the clearance of the peptide from the patient's bloodstream, to peptides that will target both peptides to a specific tissue or cell or to another peptide of complementary therapeutic use.
  • the invention is not limited to the preferred glycan molecules described above.
  • the preferred embodiments are only a few of the many useful glycan molecules that can be made using the remodeling methods of the invention. Those skilled in the art will know how to design other useful glycans once armed with the present invention.
  • the peptide is expressed in a CHO (Chinese hamster cell line) according to methods well known in the art.
  • CHO Choinese hamster cell line
  • X 2
  • f 1
  • the O-linked glycans of a peptide expressed and secreted from a CHO cell are remodeled to contain a sialylated Lewis X structure by contacting the glycans with a glycosyltransferase specific for the GalNAc acceptor moiety and the fucose donor moiety in the presence of nucleotide-fucose. This process is illustrated on N-linked glycans in FIG. 11 and Example 39.
  • the peptide is expressed in insect cells such as sf9 according to methods well known in the art.
  • insect cells such as sf9
  • the O-linked glycan on a peptide expressed in an insect cell is remodeled to a humanized glycan by contacting the glycans with a glycosyltransferase specific for a GalNAc acceptor molecule and a galactose donor molecule in the presence of nucleotide-Gal; and then contacting the glycans with a glycosyltransferase specific for a Gal acceptor molecule and a SA donor molecule in the presence of nucleotide-SA.
  • the O-linked glycans are remodeled further from the humanized form to the sialylated Lewis X form by further contacting the glycans with a glycosyltransferase specific for a GalNAc acceptor molecule and a fucose donor molecule in the presence of nucleotide-fucose.
  • the peptide is expressed in fungal cells, in particular S. cerevisiae cells, according to methods well known in the art.
  • S. cerevisiae cells When a peptide with O-linked glycans consensus sites is expressed and secreted from S. cerevisiae cells, the majority of the O-linked glycans have the structure:
  • the glycan be cleaved at the amino acid level and rebuilt from there.
  • the glycan is the O-linked glycan on a peptide expressed in a fungal cell and is remodeled to a humanized glycan by contacting the glycan with an endoglycosylase specific for an amino acid-GalNAc bond; and then contacting the glycan with a glycosyltransferase specific for a O-linked consensus site and a GalNAc donor molecule in the presence of nucleotide-GalNAc; contacting the glycan with a glycosyltransferase specific for a GalNAc acceptor molecule and a galactose donor molecule in the presence of nucleotide-Gal; and then contacting the glycans with a glycosyltransferase specific for a Gal acceptor molecule and a SA donor molecule in the presence of nucleotide-SA.
  • the glycan is the O-linked glycan on a peptide expressed in a fungal cell and is remodeled to a humanized glycan by contacting the glycan with an protein O-mannose ⁇ -1,2-N-acetylglucosaminyltransferase (POMGnTI) in the presence of GlcNAc-nucleotide; then contacting the glycan with an galactosyltransferase in the presence of nucleotide-Gal; and then contracting the glycan with an sialyltransferase in the presence of nucleotide-SA.
  • POMGnTI protein O-mannose ⁇ -1,2-N-acetylglucosaminyltransferase
  • the peptide is expressed in bacterial cells, in particular E. coli cells, according to methods well known in the art.
  • E. coli cells When a peptide with an O-linked glycan consensus site is expressed in E. coli cells, the O-linked consensus site will not be glycosylated. In this case, the desired glycan molecule must be built out from the peptide backbone in a manner similar to that describe for S. cerevisiae expression above. Further, when a peptide having an O-linked glycan is expressed in a eukaryotic cell without the proper leader sequences to direct the nascent peptide to the golgi apparatus, the mature peptide is likely not to be glycosylated.
  • an O-linked glycosyl structure may be added to the peptide by building out the glycan directly from the peptide O-linked consensus site.
  • a protein when chemically modified with a sugar moiety, it can also be remodeled as described herein.
  • the invention provides methods of preparing a conjugate of a glycosylated or an unglycosylated peptide.
  • the conjugates of the invention are formed between peptides and diverse species such as water-soluble polymers, therapeutic moieties, diagnostic moieties, targeting moieties and the like.
  • conjugates that include two or more peptides linked together through a linker arm i.e., multifunctional conjugates.
  • the multi-functional conjugates of the invention can include two or more copies of the same peptide or a collection of diverse peptides with different structures, and/or properties.
  • the conjugates of the invention are formed by the enzymatic attachment of a modified sugar to the glycosylated or unglycosylated peptide.
  • the modified sugar when interposed between the peptide and the modifying group on the sugar becomes what is referred to herein as “an intact glycosyl linking group.”
  • the present method provides peptides that bear a desired group at one or more specific locations.
  • a modified sugar is attached directly to a selected locus on the peptide chain or, alternatively, the modified sugar is appended onto a carbohydrate moiety of a peptide.
  • Peptides in which modified sugars are linked to both a peptide carbohydrate and directly to an amino acid residue of the peptide backbone are also within the scope of the present invention.
  • the methods of the invention make it possible to assemble peptides and glycopeptides that have a substantially homogeneous derivatization pattern; the enzymes used in the invention are generally selective for a particular amino acid residue or combination of amino acid residues of the peptide or particular glycan structure.
  • the methods are also practical for large-scale production of modified peptides and glycopeptides.
  • the methods of the invention provide a practical means for large-scale preparation of peptides having preselected substantially uniform derivatization patterns.
  • the methods are particularly well suited for modification of therapeutic peptides, including but not limited to, peptides that are incompletely glycosylated during production in cell culture cells (e.g., mammalian cells, insect cells, plant cells, fungal cells, yeast cells, or prokaryotic cells) or transgenic plants or animals.
  • cell culture cells e.g., mammalian cells, insect cells, plant cells, fungal cells, yeast cells, or prokaryotic cells
  • transgenic plants or animals e.g., transgenic plants or animals.
  • the methods of the invention also provide conjugates of glycosylated and unglycosylated peptides with increased therapeutic half-life due to, for example, reduced clearance rate, or reduced rate of uptake by the immune or reticuloendothelial system (RES). Moreover, the methods of the invention provide a means for masking antigenic determinants on peptides, thus reducing or eliminating a host immune response against the peptide. Selective attachment of targeting agents can also be used to target a peptide to a particular tissue or cell surface receptor that is specific for the particular targeting agent. Moreover, there is provided a class of peptides that are specifically modified with a therapeutic moiety.
  • the present invention provides a conjugate between a peptide and a selected moiety.
  • the link between the peptide and the selected moiety includes an intact glycosyl linking group interposed between the peptide and the selected moiety.
  • the selected moiety is essentially any species that can be attached to a saccharide unit, resulting in a “modified sugar” that is recognized by an appropriate transferase enzyme, which appends the modified sugar onto the peptide.
  • the saccharide component of the modified sugar when interposed between the peptide and a selected moiety, becomes an “intact glycosyl linking group.”
  • the glycosyl linking group is formed from any mono- or oligo-saccharide that, after modification with a selected moiety, is a substrate for an appropriate transferase.
  • the “agent” is a therapeutic agent, a bioactive agent, a detectable label, water-soluble moiety or the like.
  • the “agent” can be a peptide, e.g., enzyme, antibody, antigen, etc.
  • the linker can be any of a wide array of linking groups, infra. Alternatively, the linker may be a single bond or a “zero order linker.” The identity of the peptide is without limitation. Exemplary peptides are provided in FIG. 28 .
  • the selected moiety is a water-soluble polymer.
  • the water-soluble polymer is covalently attached to the peptide via an intact glycosyl linking group.
  • the glycosyl linking group is covalently attached to either an amino acid residue or a glycosyl residue of the peptide.
  • the glycosyl linking group is attached to one or more glycosyl units of a glycopeptide.
  • the invention also provides conjugates in which the glycosyl linking group is attached to both an amino acid residue and a glycosyl residue.
  • the present invention provides conjugates that are highly homogenous in their substitution patterns. Using the methods of the invention, it is possible to form peptide conjugates in which essentially all of the modified sugar moieties across a population of conjugates of the invention are attached to multiple copies of a structurally identical amino acid or glycosyl residue.
  • the invention provides a peptide conjugate having a population of water-soluble polymer moieties, which are covalently linked to the peptide through an intact glycosyl linking group.
  • each member of the population is linked via the glycosyl linking group to a glycosyl residue of the peptide, and each glycosyl residue of the peptide to which the glycosyl linking group is attached has the same structure.
  • a peptide conjugate having a population of water-soluble polymer moieties covalently linked thereto through an intact glycosyl linking group.
  • essentially every member of the population of water soluble polymer moieties is linked to an amino acid residue of the peptide via an intact glycosyl linking group, and each amino acid residue having an intact glycosyl linking group attached thereto has the same structure.
  • the present invention also provides conjugates analogous to those described above in which the peptide is conjugated to a therapeutic moiety, diagnostic moiety, targeting moiety, toxin moiety or the like via an intact glycosyl linking group.
  • a therapeutic moiety e.g., peptide
  • a targeting moiety e.g., targeting moiety
  • toxin moiety e.g., toxin moiety or the like via an intact glycosyl linking group.
  • Each of the above-recited moieties can be a small molecule, natural polymer (e.g., peptide) or synthetic polymer.
  • interleukin-2 is conjugated to transferrin via a bifunctional linker that includes an intact glycosyl linking group at each terminus of the PEG moiety (Scheme 1).
  • one terminus of the PEG linker is fuctionalized with an intact sialic acid linker that is attached to transferrin and the other is functionalized with an intact GalNAc linker that is attached to IL-2.
  • EPO is conjugated to transferrin.
  • EPO is conjugated to glial derived neurotropic growth factor (GDNF).
  • GDNF glial derived neurotropic growth factor
  • each conjugation is accomplished via a bifunctional linker that includes an intact glycosyl linking group at each terminus of the PEG moiety, as aforementioned. Transferrin transfers the protein across the blood brain barrier.
  • the conjugates of the invention can include intact glycosyl linking groups that are mono- or multi-valent (e.g., antennary structures), see, FIGS. 14-22 .
  • the conjugates of the invention also include glycosyl linking groups that are O-linked glycans originating from serine or threonine ( FIG. 11 ).
  • conjugates of the invention include both Species in which a selected moiety is attached to a peptide via a monovalent glycosyl linking group.
  • conjugates in which more than one selected moiety is attached to a peptide via a multivalent linking group.
  • One or more proteins can be conjugated together to take advantage of their biophysical and biological properties.
  • the invention provides conjugates that localize selectively in a particular tissue due to the presence of a targeting agent as a component of the conjugate.
  • the targeting agent is a protein.
  • Exemplary proteins include transferrin (brain, blood pool), human serum (HS)-glycoprotein (bone, brain, blood pool), antibodies (brain, tissue with antibody-specific antigen, blood pool), coagulation Factors V-XII (damaged tissue, clots, cancer, blood pool), serum proteins, e.g., ⁇ -acid glycoprotein, fetuin, ⁇ -fetal protein (brain, blood pool), ⁇ 2-glycoprotein (liver, atherosclerosis plaques, brain, blood pool), G-CSF, GM-CSF, M-CSF, and EPO (immune stimulation, cancers, blood pool, red blood cell overproduction, neuroprotection), and albumin (increase in half-life).
  • the present invention provides methods for preparing these and other conjugates.
  • the invention provides a method of forming a covalent conjugate between a selected moiety and a peptide.
  • the invention provides methods for targeting conjugates of the invention to a particular tissue or region of the body.
  • the conjugate is formed between a water-soluble polymer, a therapeutic moiety, targeting moiety or a biomolecule, and a glycosylated or non-glycosylated peptide.
  • the polymer, therapeutic moiety or biomolecule is conjugated to the peptide via an intact glycosyl linking group, which is interposed between, and covalently linked to both the peptide and the modifying group (e.g., water-soluble polymer).
  • the method includes contacting the peptide with a mixture containing a modified sugar and a glycosyltransferase for which the modified sugar is a substrate.
  • the reaction is conducted under conditions sufficient to form a covalent bond between the modified sugar and the peptide.
  • the sugar moiety of the modified sugar is preferably selected from nucleotide sugars, activated sugars and sugars, which are neither nucleotides nor activated.
  • the invention provides a method for linking two or more peptides through a linking group.
  • the linking group is of any useful structure and may be selected from straight-chain and branched chain structures.
  • each terminus of the linker, which is attached to a peptide includes a modified sugar (i.e., a nascent intact glycosyl linking group).
  • two peptides are linked together via a linker moiety that includes a PEG linker.
  • a PEG moiety is functionalized at a first terminus with a first glycosyl unit and at a second terminus with a second glycosyl unit.
  • the first and second glycosyl units are preferably substrates for different transferases, allowing orthogonal attachment of the first and second peptides to the first and second glycosyl units, respectively.
  • the (glycosyl) 1 -PEG-(glycosyl) 2 linker is contacted with the first peptide and a first transferase for which the first glycosyl unit is a substrate, thereby forming (peptide) 1 -(glycosyl) 1 -PEG-(glycosyl) 2 .
  • the first transferase and/or unreacted peptide is then optionally removed from the reaction mixture.
  • the second peptide and a second transferase for which the second glycosyl unit is a substrate are added to the (peptide) 1 -(glycosyl) 1 -PEG-(glycosyl) 2 conjugate, forming (peptide) 1 -(glycosyl) 1 -PEG-(glycosyl) 2 -(peptide) 2 .
  • Those of skill in the art will appreciate that the method outlined above is also applicable to forming conjugates between more than two peptides by, for example, the use of a branched PEG, dendrimer, poly(amino acid), polysaccharide or the like.
  • interleukin-2 is conjugated to transferrin via a bifunctional linker that includes an intact glycosyl linking group at each terminus of the PEG moiety (Scheme 1).
  • the IL-2 conjugate has an in vivo half-life that is increased over that of IL-2 alone by virtue of the greater molecular size of the conjugate.
  • the conjugation of IL-2 to transferrin serves to selectively target the conjugate to the brain.
  • one terminus of the PEG linker is functionalized with a CMP-sialic acid and the other is functionalized with an UDP-GalNAc.
  • the linker is combined with IL-2 in the presence of a GalNAc transferase, resulting in the attachment of the GalNAc of the linker arm to a serine and/or threonine residue on the IL-2.
  • transferrin is conjugated to a nucleic acid for use in gene therapy.
  • the processes described above can be carried through as many cycles as desired, and is not limited to forming a conjugate between two peptides with a single linker.
  • the reactions functionalizing the intact glycosyl linking groups at the termini of the PEG (or other) linker with the peptide can occur simultaneously in the same reaction vessel, or they can be carried out in a step-wise fashion.
  • the conjugate produced at each step is optionally purified from one or more reaction components (e.g., enzymes, peptides).
  • Scheme 2 shows a method of preparing a conjugate that targets a selected protein, e.g., EPO, to bone and increases the circulatory half-life of the selected protein.
  • a selected protein e.g., EPO
  • reactive derivatives of PEG or other linkers
  • the use of reactive derivatives of PEG (or other linkers) to attach one or more peptide moieties to the linker is within the scope of the present invention.
  • the invention is not limited by the identity of the reactive PEG analogue.
  • Many activated derivatives of poly(ethylene glycol) are available commercially and in the literature. It is well within the abilities of one of skill to choose, and synthesize if necessary, an appropriate activated PEG derivative with which to prepare a substrate useful in the present invention. See, Abuchowski et al. Cancer Biochem. Biophys., 7: 175-186 (1984); Abuchowski et al., J. Biol. Chem., 252: 3582-3586 (1977); Jackson et al., Anal.
  • linking groups include the urethane linkage between amino groups and activated PEG. See, Veronese, et al., Appl. Biochem. Biotechnol., 11: 141-152 (1985).
  • the invention provides a method for extending the blood-circulation half-life of a selected peptide, in essence targeting the peptide to the blood pool, by conjugating the peptide to a synthetic or natural polymer of a size sufficient to retard the filtration of the protein by the glomerulus (e.g., albumin).
  • a synthetic or natural polymer of a size sufficient to retard the filtration of the protein by the glomerulus e.g., albumin.
  • This embodiment of the invention is illustrated in Scheme 3 in which erythropoietin (EPO) is conjugated to albumin via a PEG linker using a combination of chemical and enzymatic modification.
  • an amino acid residue of albumin is modified with a reactive PEG derivative, such as X-PEG-(CMP-sialic acid), in which X is an activating group (e.g., active ester, isothiocyanate, etc).
  • X is an activating group (e.g., active ester, isothiocyanate, etc).
  • the PEG derivative and EPO are combined and contacted with a transferase for which CMP-sialic acid is a substrate.
  • an ⁇ -amine of lysine is reacted with the N-hydroxysuccinimide ester of the PEG-linker to form the albumin conjugate.
  • the CMP-sialic acid of the linker is enzymatically conjugated to an appropriate residue on EPO, e.g., Gal, thereby forming the conjugate.
  • EPO e.g., Gal
  • Those of skill will appreciate that the above-described method is not limited to the reaction partners set forth. Moreover, the method can be practiced to form conjugates that include more than two protein moieties by, for example, utilizing a branched linker having more than two termini.
  • Modified glycosyl donor species are preferably selected from modified sugar nucleotides, activated modified sugars and modified sugars that are simple saccharides that are neither nucleotides nor activated. Any desired carbohydrate structure can be added to a peptide using the methods of the invention. Typically, the structure will be a monosaccharide, but the present invention is not limited to the use of modified monosaccharide sugars; oligosaccharides and polysaccharides are useful as well.
  • the modifying group is attached to a sugar moiety by enzymatic means, chemical means or a combination thereof, thereby producing a modified sugar.
  • the sugars are substituted at any position that allows for the attachment of the modifying moiety, yet which still allows the sugar to function as a substrate for the enzyme used to ligate the modified sugar to the peptide.
  • sialic acid when sialic acid is the sugar, the sialic acid is substituted with the modifying group at either the 9-position on the pyruvyl side chain or at the 5-position on the amine moiety that is normally acetylated in sialic acid.
  • a modified sugar nucleotide is utilized to add the modified sugar to the peptide.
  • exemplary sugar nucleotides that are used in the present invention in their modified form include nucleotide mono-, di- or triphosphates or analogs thereof.
  • the modified sugar nucleotide is selected from a UDP-glycoside, CMP-glycoside, or a GDP-glycoside.
  • the modified sugar nucleotide is selected from an UDP-galactose, UDP-galactosamine, UDP-glucose, UDP-glucosamine, GDP-mannose, GDP-fucose, CMP-sialic acid, or CMP-NeuAc.
  • N-acetylamine derivatives of the sugar nucleotides are also of use in the method of the invention.
  • the invention also provides methods for synthesizing a modified peptide using a modified sugar, e.g., modified-galactose, -fucose, and -sialic acid.
  • a modified sialic acid either a sialyltransferase or a trans-sialidase (for ⁇ 2,3-linked sialic acid only) can be used in these methods.
  • the modified sugar is an activated sugar.
  • Activated modified sugars, which are useful in the present invention are typically glycosides which have been synthetically altered to include an activated leaving group.
  • the term “activated leaving group” refers to those moieties, which are easily displaced in enzyme-regulated nucleophilic substitution reactions.
  • Many activated sugars are known in the art. See, for example, Vocadlo et al., In C ARBOHYDRATE C HEMISTRY AND B IOLOGY, Vol. 2, Ernst et al. Ed., Wiley-VCH Verlag: Weinheim, Germany, 2000; Kodama et al., Tetrahedron Lett. 34: 6419 (1993); Lougheed, et al., J. Biol. Chem. 274: 37717 (1999)).
  • activating groups include fluoro, chloro, bromo, tosylate ester, mesylate ester, triflate ester and the like.
  • Preferred activated leaving groups, for use in the present invention are those that do not significantly sterically encumber the enzymatic transfer of the glycoside to the acceptor. Accordingly, preferred embodiments of activated glycoside derivatives include glycosyl fluorides and glycosyl mesylates, with glycosyl fluorides being particularly preferred.
  • glycosyl fluorides ⁇ -galactosyl fluoride, ⁇ -mannosyl fluoride, ⁇ -glucosyl fluoride, ⁇ -fucosyl fluoride, ⁇ -xylosyl fluoride, ⁇ -sialyl fluoride, ⁇ -N-acetylglucosaminyl fluoride, ⁇ -N-acetylgalactosaminyl fluoride, ⁇ -galactosyl fluoride, ⁇ -mannosyl fluoride, ⁇ -glucosyl fluoride, ⁇ -fucosyl fluoride, ⁇ -xylosyl fluoride, ⁇ -sialyl fluoride, ⁇ -N-acetylglucosaminyl fluoride and ⁇ -N-acetylgalactosaminyl fluoride are most preferred.
  • glycosyl fluorides can be prepared from the free sugar by first acetylating the sugar and then treating it with HF/pyridine. This generates the thermodynamically most stable anomer of the protected (acetylated) glycosyl fluoride (i. e., the ⁇ -glycosyl fluoride). If the less stable anomer (i.e., the ⁇ -glycosyl fluoride) is desired, it can be prepared by converting the peracetylated sugar with HBr/HOAc or with HCl to generate the anomeric bromide or chloride. This intermediate is reacted with a fluoride salt such as silver fluoride to generate the glycosyl fluoride.
  • Acetylated glycosyl fluorides may be deprotected by reaction with mild (catalytic) base in methanol (e.g. NaOMe/MeOH). In addition, many glycosyl fluorides are commercially available.
  • glycosyl mesylates can be prepared by treatment of the fully benzylated hemiacetal form of the sugar with mesyl chloride, followed by catalytic hydrogenation to remove the benzyl groups.
  • the modified sugar is an oligosaccharide having an antennary structure.
  • one or more of the termini of the antennae bear the modifying moiety.
  • the oligosaccharide is useful to “amplify” the modifying moiety; each oligosaccharide unit conjugated to the peptide attaches multiple copies of the modifying group to the peptide.
  • the general structure of a typical chelate of the invention as set forth in the drawing above, encompasses multivalent species resulting from preparing a conjugate of the invention utilizing an antennary structure. Many antennary saccharide structures are known in the art, and the present method can be practiced with them without limitation.
  • modifying groups are discussed below.
  • the modifying groups can be selected for one or more desirable property.
  • Exemplary properties include, but are not limited to, enhanced pharmacokinetics, enhanced pharmacodynamics, improved biodistribution, providing a polyvalent species, improved water solubility, enhanced or diminished lipophilicity, and tissue targeting.
  • hydrophilicity of a selected peptide is enhanced by conjugation with polar molecules such as amine-, ester-, hydroxyl- and polyhydroxyl-containing molecules.
  • polar molecules such as amine-, ester-, hydroxyl- and polyhydroxyl-containing molecules.
  • Representative examples include, but are not limited to, polylysine, polyethyleneimine, poly(ethylene glycol) and poly(propyleneglycol).
  • Preferred water-soluble polymers are essentially non-fluorescent, or emit such a minimal amount of fluorescence that they are inappropriate for use as a fluorescent marker in an assay. Polymers that are not naturally occurring sugars may be used.
  • a therapeutic sugar moiety is conjugated to a linker arm and the sugar-linker arm is subsequently conjugated to a peptide via a method of the invention.
  • U.S. Pat. No. 5,672,662 discloses a water soluble and isolatable conjugate of an active ester of a polymer acid selected from linear or branched poly(alkylene oxides), poly(oxyethylated polyols), poly(olefinic alcohols), and poly(acrylomorpholine), wherein the polymer has about 44 or more recurring units.
  • U.S. Pat. No. 6,376,604 sets forth a method for preparing a water-soluble 1-benzotriazolylcarbonate ester of a water-soluble and non-peptidic polymer by reacting a terminal hydroxyl of the polymer with di(1-benzotriazoyl)carbonate in an organic solvent.
  • the active ester is used to form conjugates with a biologically active agent such as a protein or peptide.
  • WO 99/45964 describes a conjugate comprising a biologically active agent and an activated water soluble polymer comprising a polymer backbone having at least one terminus linked to the polymer backbone through a stable linkage, wherein at least one terminus comprises a branching moiety having proximal reactive groups linked to the branching moiety, in which the biologically active agent is linked to at least one of the proximal reactive groups.
  • Other branched poly(ethylene glycols) are described in WO 96/21469, U.S. Pat. No. 5,932,462 describes a conjugate formed with a branched PEG molecule that includes a branched terminus that includes reactive functional groups.
  • the free reactive groups are available to react with a biologically active species, such as a protein or peptide, forming conjugates between the poly(ethylene glycol) and the biologically active species.
  • a biologically active species such as a protein or peptide
  • U.S. Pat. No. 5,446,090 describes a bifunctional PEG linker and its use in forming conjugates having a peptide at each of the PEG linker termini.
  • Conjugates that include degradable PEG linkages are described in WO 99/34833; and WO 99/14259, as well as in U.S. Pat. No. 6,348,558. Such degradable linkages are applicable in the present invention.
  • 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(glutamic acid); nucleic acids; synthetic polymers (e.g., poly(acrylic acid), poly(ethers), e.g., poly(ethylene glycol); peptides, proteins, and the like.
  • the present invention may be practiced with any water-soluble polymer with the sole limitation that the polymer must include a point at which the remainder of the conjugate can be attached.
  • Preferred 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.”
  • the present invention is further illustrated by reference to a poly(ethylene glycol) conjugate.
  • a poly(ethylene glycol) conjugate 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).
  • Poly(ethylene glycol) molecules suitable for use in the invention include, but are not limited to, those described by the following Formula 3: R ⁇ H, alkyl, benzyl, aryl, acetal, OHC—, H 2 N—CH 2 CH 2 —, HS—CH 2 CH 2 —, -sugar-nucleotide, protein, methyl, ethyl;
  • poly(ethylene glycol) molecule is selected from the following:
  • the 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:
  • the in vivo half-life, area under the curve, and/or residence time of therapeutic peptides can also be enhanced with water-soluble polymers such as polyethylene glycol (PEG) and polypropylene glycol (PPG).
  • PEG polyethylene glycol
  • PPG polypropylene glycol
  • chemical modification of proteins with PEG increases their molecular size and decreases their surface- and functional group-accessibility, each of which are dependent on the size of the PEG attached to the protein. This results in an improvement of plasma half-lives and in proteolytic-stability, and a decrease in immunogenicity and hepatic uptake (Chaffee et al. J. Clin. Invest. 89: 1643-1651 (1992); Pyatak et al. Res. Commun. Chem.
  • PEGylation of interleukin-2 has been reported to increase its antitumor potency in vivo (Katre et al. Proc. Natl. Acad. Sci. USA. 84: 1487-1491 (1987)) and PEGylation of a F(ab′)2 derived from the monoclonal antibody A7 has improved its tumor localization (Kitamura et al. Biochem. Biophys. Res. Commun. 28:1387-1394 (1990)).
  • the in vivo half-life of a peptide derivatized with a water-soluble polymer by a method of the invention is increased relevant to the in vivo half-life of the non-derivatized peptide.
  • the area under the curve of a peptide derivatized with a water-soluble polymer using a method of the invention is increased relevant to the area under the curve of the non-derivatized peptide.
  • the residence time of a peptide derivatized with a water-soluble polymer using a method of the invention is increased relevant to the residence time of the non-derivatized peptide.
  • the increase in peptide in vivo half-life is best expressed as a range of percent increase in this quantity.
  • the lower end of the range of percent increase is about 40%, about 60%, about 80%, about 100%, about 150% or about 200%.
  • the upper end of the range is about 60%, about 80%, about 100%, about 150%, or more than about 250%.
  • the present invention provides a PEGylated follicle stimulating hormone (Examples 23 and 24). In a further exemplary embodiment, the invention provides a PEGylated transferrin (Example 42).
  • exemplary water-soluble polymers of use in the invention include, but are not limited to linear or branched poly(alkylene oxides), poly(oxyethylated polyols), poly(olefinic alcohols), and poly(acrylomorpholine), dextran, starch, poly(amino acids), etc.
  • the conjugates of the invention may also include one or more water-insoluble polymers.
  • This embodiment of the invention is illustrated by the use of the conjugate as a vehicle with which to deliver a therapeutic peptide in a controlled manner.
  • Polymeric drug delivery systems are known in the art. See, for example, Dunn et al., Eds. P OLYMERIC D RUGS AND D RUG D ELIVERY S YSTEMS, ACS Symposium Series Vol. 469, American Chemical Society, Washington, D.C. 1991.
  • substantially any known drug delivery system is applicable to the conjugates of the present invention.
  • Representative water-insoluble polymers include, but are not limited to, polyphosphazines, poly(vinyl alcohols), polyamides, polycarbonates, polyalkylenes, polyacrylamides, polyalkylene glycols, polyalkylene oxides, polyalkylene terephthalates, polyvinyl ethers, polyvinyl esters, polyvinyl halides, polyvinylpyrrolidone, polyglycolides, polysiloxanes, polyurethanes, poly(methyl methacrylate), poly(ethyl methacrylate), poly(butyl methacrylate), poly(isobutyl methacrylate), poly(hexyl methacrylate), poly(isodecyl methacrylate), poly(lauryl methacrylate), poly(phenyl methacrylate), poly(methyl acrylate), poly(isopropyl acrylate), poly(isobutyl acrylate), poly(octadecyl acrylate)
  • Synthetically modified natural polymers of use in conjugates of the invention include, but are not limited to, alkyl celluloses, hydroxyalkyl celluloses, cellulose ethers, cellulose esters, and nitrocelluloses.
  • Particularly preferred members of the broad classes of synthetically modified natural polymers include, but are not limited to, methyl cellulose, ethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methyl cellulose, hydroxybutyl methyl cellulose, cellulose acetate, cellulose propionate, cellulose acetate butyrate, cellulose acetate phthalate, carboxymethyl cellulose, cellulose triacetate, cellulose sulfate sodium salt, and polymers of acrylic and methacrylic esters and alginic acid.
  • biodegradable polymers of use in the conjugates of the invention include, but are not limited to, polylactides, polyglycolides and copolymers thereof, poly(ethylene terephthalate), poly(butyric acid), poly(valeric acid), poly(lactide-co-caprolactone), poly(lactide-co-glycolide), polyanhydrides, polyorthoesters, blends and copolymers thereof.
  • compositions that form gels such as those including collagen, pluronics and the like.
  • the polymers of use in the invention include “hybrid” polymers that include water-insoluble materials having within at least a portion of their structure, a bioresorbable molecule.
  • An example of such a polymer is one that includes a water-insoluble copolymer, which has a bioresorbable region, a hydrophilic region and a plurality of crosslinkable functional groups per polymer chain.
  • water-insoluble materials includes materials that are substantially insoluble in water or water-containing environments. Thus, although certain regions or segments of the copolymer may be hydrophilic or even water-soluble, the polymer molecule, as a whole, does not to any substantial measure dissolve in water.
  • bioresorbable molecule includes a region that is capable of being metabolized or broken down and resorbed and/or eliminated through normal excretory routes by the body. Such metabolites or break down products are preferably substantially non-toxic to the body.
  • the bioresorbable region may be either hydrophobic or hydrophilic, so long as the copolymer composition as a whole is not rendered water-soluble.
  • the bioresorbable region is selected based on the preference that the polymer, as a whole, remains water-insoluble. Accordingly, the relative properties, i.e., the kinds of functional groups contained by, and the relative proportions of the bioresorbable region, and the hydrophilic region are selected to ensure that useful bioresorbable compositions remain water-insoluble.
  • Exemplary resorbable polymers include, for example, synthetically produced resorbable block copolymers of poly(a-hydroxy-carboxylic acid)/poly(oxyalkylene, (see, ohn et al., U.S. Pat. No. 4,826,945). These copolymers are not crosslinked and are water-soluble so that the body can excrete the degraded block copolymer compositions. See, Younes et al., J Biomed. Mater. Res. 21: 1301-1316 (1987); and Cohn et al., J Biomed. Mater. Res. 22: 993-1009 (1988).
  • bioresorbable polymers include one or more components selected from poly(esters), poly(hydroxy acids), poly(lactones), poly(amides), poly(ester-amides), poly (amino acids), poly(anhydrides), poly(orthoesters), poly(carbonates), poly(phosphazines), poly(phosphoesters), poly(thioesters), polysaccharides and mixtures thereof. More preferably still, the biosresorbable polymer includes a poly(hydroxy) acid component. Of the poly(hydroxy) acids, polylactic acid, polyglycolic acid, polycaproic acid, polybutyric acid, polyvaleric acid and copolymers and mixtures thereof are preferred.
  • preferred polymeric coatings for use in the methods of the invention can also form an excretable and/or metabolizable fragment.
  • Bioresorbable regions of coatings useful in the present invention can be designed to be hydrolytically and/or enzymatically cleavable.
  • hydrolytically cleavable refers to the susceptibility of the copolymer, especially the bioresorbable region, to hydrolysis in water or a water-containing environment.
  • enzymatically cleavable refers to the susceptibility of the copolymer, especially the bioresorbable region, to cleavage by endogenous or exogenous enzymes.
  • the hydrophilic region When placed within the body, the hydrophilic region can be processed into excretable and/or metabolizable fragments.
  • the hydrophilic region can include, for example, polyethers, polyalkylene oxides, polyols, poly(vinyl pyrrolidine), poly(vinyl alcohol), poly(alkyl oxazolines), polysaccharides, carbohydrates, peptides, proteins and copolymers and mixtures thereof.
  • the hydrophilic region can also be, for example, a poly(alkylene) oxide.
  • Such poly(alkylene) oxides can include, for example, poly(ethylene) oxide, poly(propylene) oxide and mixtures and copolymers thereof.
  • Hydrogels are polymeric materials that are capable of absorbing relatively large quantities of water.
  • hydrogel forming compounds include, but are not limited to, polyacrylic acids, sodium carboxymethylcellulose, polyvinyl alcohol, polyvinyl pyrrolidine, gelatin, carrageenan and other polysaccharides, hydroxyethylenemethacrylic acid (HEMA), as well as derivatives thereof, and the like.
  • Hydrogels can be produced that are stable, biodegradable and bioresorbable.
  • hydrogel compositions can include subunits that exhibit one or more of these properties.
  • Bio-compatible hydrogel compositions whose integrity can be controlled through crosslinking are known and are presently preferred for use in the methods of the invention.
  • Hubbell et al. U.S. Pat. No. 5,410,016, which issued on Apr. 25, 1995 and U.S. Pat. No. 5,529,914, which issued on Jun. 25, 1996, disclose water-soluble systems, which are crosslinked block copolymers having a water-soluble central block segment sandwiched between two hydrolytically labile extensions. Such copolymers are further end-capped with photopolymerizable acrylate functionalities. When crosslinked, these systems become hydrogels.
  • the water soluble central block of such copolymers can include poly(ethylene glycol); whereas, the hydrolytically labile extensions can be a poly( ⁇ -hydroxy acid), such as polyglycolic acid or polylactic acid. See, Sawhney et al., Macromolecules 26: 581-587 (1993).
  • the gel is a thermoreversible gel.
  • Thermoreversible gels including components, such as pluronics, collagen, gelatin, hyalouronic acid, polysaccharides, polyurethane hydrogel, polyurethane-urea hydrogel and combinations thereof are presently preferred.
  • the conjugate of the invention includes a component of a liposome.
  • Liposomes can be prepared according to methods known to those skilled in the art, for example, as described in Eppstein et al., U.S. Pat. No. 4,522,811, which issued on Jun. 11, 1985.
  • liposome formulations may be prepared by dissolving appropriate lipid(s) (such as stearoyl phosphatidyl ethanolamine, stearoyl phosphatidyl choline, arachadoyl phosphatidyl choline, and cholesterol) in an inorganic solvent that is then evaporated, leaving behind a thin film of dried lipid on the surface of the container.
  • appropriate lipid(s) such as stearoyl phosphatidyl ethanolamine, stearoyl phosphatidyl choline, arachadoyl phosphatidyl choline, and cholesterol
  • aqueous solution of the active compound or its pharmaceutically acceptable salt is then introduced into the container.
  • the container is then swirled by hand to free lipid material from the sides of the container and to disperse lipid aggregates, thereby forming the liposomal suspension.
  • microparticles and methods of preparing the microparticles are offered by way of example and they are not intended to define the scope of microparticles of use in the present invention. It will be apparent to those of skill in the art that an array of microparticles, fabricated by different methods, are of use in the present invention.
  • the modified sugar bears a biomolecule.
  • the biomolecule is a functional protein, enzyme, antigen, antibody, peptide, nucleic acid (e.g., single nucleotides or nucleosides, oligonucleotides, polynucleotides and single- and higher-stranded nucleic acids), lectin, receptor or a combination thereof.
  • biomolecules are essentially non-fluorescent, or emit such a minimal amount of fluorescence that they are inappropriate for use as a fluorescent marker in an assay.
  • Other biomolecules may be fluorescent.
  • the use of an otherwise naturally occurring sugar that is modified by covalent attachment of another entity e.g., PEG, biomolecule, therapeutic moiety, diagnostic moiety, etc.
  • a sugar moiety which is a biomolecule, is conjugated to a linker arm and the sugar-linker arm cassette is subsequently conjugated to a peptide via a method of the invention.
  • Biomolecules useful in practicing the present invention can be derived from any source.
  • the biomolecules can be isolated from natural sources or they can be produced by synthetic methods.
  • Peptides can be natural peptides or mutated peptides. Mutations can be effected by chemical mutagenesis, site-directed mutagenesis or other means of inducing mutations known to those of skill in the art.
  • Peptides useful in practicing the instant invention include, for example, enzymes, antigens, antibodies and receptors.
  • Antibodies can be either polyclonal or monoclonal; either intact or fragments.
  • the peptides are optionally the products of a program of directed evolution.
  • Both naturally derived and synthetic peptides and nucleic acids are of use in conjunction with the present invention; these molecules can be attached to a sugar residue component or a crosslinking agent by any available reactive group.
  • peptides can be attached through a reactive amine, carboxyl, sulfhydryl, or hydroxyl group.
  • the reactive group can reside at a peptide terminus or at a site internal to the peptide chain.
  • Nucleic acids can be attached through a reactive group on a base (e.g., exocyclic amine) or an available hydroxyl group on a sugar moiety (e.g., 3′- or 5′-hydroxyl).
  • the peptide and nucleic acid chains can be further derivatized at one or more sites to allow for the attachment of appropriate reactive groups onto the chain. See, Chrisey et al. Nucleic Acids Res. 24: 3031-3039 (1996).
  • the biomolecule is selected to direct the peptide modified by the methods of the invention to a specific tissue, thereby enhancing the delivery of the peptide to that tissue relative to the amount of underivatized peptide that is delivered to the tissue.
  • the amount of derivatized peptide delivered to a specific tissue within a selected time period is enhanced by derivatization by at least about 20%, more preferably, at least about 40%, and more preferably still, at least about 100%.
  • preferred biomolecules for targeting applications include antibodies, hormones and ligands for cell-surface receptors.
  • Exemplary targeting biomolecules include, but are not limited to, an antibody specific for the transferrin receptor for delivery of the molecule to the brain (Penichet et al., 1999, J. Immunol. 163:4421-4426; Pardridge, 2002, Adv. Exp. Med. Biol. 513:397-430), a peptide that recognizes the vasculature of the prostate (Arap et al., 2002, PNAS 99:1527-1531), and an antibody specific for lung caveolae (McIntosh et al., 2002, PNAS 99:1996-2001).
  • the modifying group is a protein.
  • the protein is an interferon.
  • the interferons are antiviral glycoproteins that, in humans, are secreted by human primary fibroblasts after induction with virus or double-stranded RNA. Interferons are of interest as therapeutics, e.g., antivirals and treatment of multiple sclerosis.
  • interferon- ⁇ see, e.g., Yu, et al., J. Neuroimmunol., 64(1):91-100 (1996); Schmidt, J., J. Neurosci.
  • interferon ⁇ is conjugated to a second peptide via a linker arm.
  • the linker arm includes an intact glycosyl linking group through which it is attached to the second peptide via a method of the invention.
  • the linker arm also optionally includes a second intact glycosyl linking group, through which it is attached to the interferon.
  • the invention provides a conjugate of follicle stimulating hormone (FSH).
  • FSH is a glycoprotein hormone. See, for example, Saneyoshi, et al., Biol. Reprod., 65:1686-1690 (2001); Hakola, et al., J. Endocrinol., 158:441-448 (1998); Stanton, et al., Mol. Cell. Endocrinol., 125:133-141 (1996); Walton, et al., J. Clin. Endocrinol.
  • the FSH conjugate can be formed in a manner similar to that described for interferon.
  • the conjugate includes erythropoietin (EPO).
  • EPO erythropoietin
  • An exemplary EPO conjugate is formed analogously to the conjugate of interferon.
  • the invention provides a conjugate of human granulocyte colony stimulating factor (G-CSF).
  • G-CSF is a glycoprotein that stimulates proliferation, differentiation and activation of neutropoietic progenitor cells into functionally mature neutrophils. Injected G-CSF is known to be rapidly cleared from the body. See, for example, Nohynek, et al., Cancer Chemother.
  • G-CSF G-CSF
  • a conjugate with biotin there is provided a conjugate with biotin.
  • a selectively biotinylated peptide is elaborated by the attachment of an avidin or streptavidin moiety bearing one or more modifying groups.
  • the biomolecule is selected to direct the peptide modified by the methods of the invention to a specific intracellular compartment, thereby enhancing the delivery of the peptide to that intracellular compartment relative to the amount of underivatized peptide that is delivered to the tissue.
  • the amount of derivatized peptide delivered to a specific intracellular compartment within a selected time period is enhanced by derivatization by at least about 20%, more preferably, at least about 40%, and more preferably still, at least about 100%.
  • the biomolecule is linked to the peptide by a cleavable linker that can hydrolyze once internalized.
  • biomolecules for intracellular targeting applications include transferrin, lactotransferrin (lactoferrin), melanotransferrin (p97), ceruloplasmin, and divalent cation transporter, as well as antibodies directed against specific vascular targets.
  • Contemplated linkages include, but are not limited to, protein-sugar-linker-sugar-protein, protein-sugar-linker-protein and multivalent forms thereof, and protein-sugar-linker-drug where the drug includes small molecules, peptides, lipids, among others.
  • Site-specific and target-oriented delivery of therapeutic agents is desirable for the purpose of treating a wide variety of human diseases, such as different types of malignancies and certain neurological disorders. Such procedures are accompanied by fewer side effects and a higher efficiacy of drug.
  • Various principles have been relied on in designing these delivery systems. For a review, see Garnett, Advanced Drug Delivery Reviews 53:171-216 (2001).
  • the MAbs can be connected to a toxin, which may be obtained from a plant, bacterial, or fungal source, to form chimeric proteins called immunotoxins.
  • a toxin which may be obtained from a plant, bacterial, or fungal source, to form chimeric proteins called immunotoxins.
  • plant toxins are divided into two classes: (1) holotoxins (or class II ribosome inactivating proteins), such as ricin, abrin, mistletoe lectin, and modeccin, and (2) hemitoxins (class I ribosome inactivating proteins), such as pokeweed antiviral protein (PAP), saporin, Bryodin 1, bouganin, and gelonin.
  • PAP pokeweed antiviral protein
  • bacterial toxins include diphtheria toxin (DT) and Pseudomonas exotoxin (PE). Kreitman, Current Pharmaceutical Biotechnology 2:313-325 (2001). Other toxins contemplated for use with the present invention include, but are not limited to, those in Table 2. TABLE 2 Toxins.
  • BMS-310705 Discodermolide/ 127943-53-7/ solid tumors/ tubulin Broad activity (A549- synthetic; orginally analogs less not reported; 100-fold stabilizing nsclung, prostate, P388, isolated from Discodermia potent increase in water agent ovarian with IC50's dissoluta (deep water solubility over taxol (similar to about 10 nM) including sponge); rare compound taxol) multi-drug resistant cell (7 mg per 0.5 Kg sponge/ lines; XAA-296 Chondramide D/ 172430-63-6 cancer/ tubulin 5 nM A-549 not reported not reported binding (epidermoid carcinoma) agent; actin 15 nM A-498 (kidney) polymeriza- 14 nM A549 (lung) tion inhibitor 5 nM SK-OV-3 (ovary) 3 nM U-937 (lymphoma) Cryptophycin analogs 204990-60-3 solid tumors, colon tubulin broad activity (
  • NCI NSC-667642 tumor panel GI50's from 100 nM to 10 pM; LC50's from 100 nM to 25 pM.
  • Cryptophycin 8/ 168482-36-8 solid tumors/ tubulin broad spectrum semi-synthetic; starting 168482-40-4; not reported polymeriza- anticancer activity (cell material from Nostoc sp.
  • NCI tumor panel cell Streptomyces natural not reported chaperone culture
  • most active in Geldanus/ function colon, lung and NSC-212518 Antibiotic leukemia.
  • NCI tumor U 29135; NSC-122750 panel GI50's from 10 ⁇ M to 0.1 nM
  • LC50's from 100 ⁇ M to 100 nM. Two assays with very different potencies.
  • Pr(—X—S—S—W) m m 0.5-15
  • Pr proteinaceous center
  • Y anitbody P76.6 Calicheamicin- 113440-58-7
  • AML/ DNA Kills CD33+ cells HL- IgG(CD33 antigen)- 220578-59-6/ mild toxicity cleaving 60, NOMO-1, and conjugate 15 / several agent NKM-1) at 100 ng/mL
  • semi-synthetic reported in MDR cell lines are not Micromnonospora patents effected by the drug.
  • TES-23-NCS TES-23 antibody agent and (without anticancer immunostlin- agent) was as effective at ulator eliminating tumors as the drug conjugated protein Kedarcidin 18 / 128512-40-3; cancer/ DNA cell culture (IC50's in Streptoalloteichus sp 1285 12-39-0/ not reported cleaving ng/mL), 0.4 HCT116; NOV strain L5856, ATCC chromophore agent 0.3 HCT116NP35; 53650/ and protein 0.3 HCT116/VM46; NSC-646276 conjugate 0.2 A2780; 1.3 A2780/DDP. animal models in P388 and B-16 melanoma.
  • NCI tumor panel GI50's from 50 ⁇ M to 5 ⁇ M. Eleutherobins/ 174545-76-7/ cancer/ tubulin similar potency to taxol; marine coral sarcodictyins not reported binding not effective against (marine coral) agent MDR cell lines Bryostatin-1/ 83314-01-6 leukemia, melanoma, immunostim- not reported Bugula neritina (marine lung, cancer/ ulant (TNF, bryosoan)/ myalgia; accumulated GMCSF, GMY-45618; NSC- toxicity; poor water etc); 339555 solubility; dose limiting enhances cell toxicity kill by current anticancer agents FR-901228/ 128517-07-7 leukemia, T-cell histone In vitro cell lines (NCI Chromobacterium lymphoma, cancer/ deacetylase tumor panel); violaceum strain 968/ toxic doses (LD50) 6.4 inhiibitor
  • the 165883-76-1/ inhibition values analogs (clonogenic growth of prepared human cancer cells) at 10 aM ranged from 6.2 to >99.9% against NALM-6 human B- lineage acute lymophoblastic leukemia cells, BT-20 breast cancer cells and U373 glioblastoma cells, with the specified compound showing inhibition values in the range of 42.4 to > 99.9% against these cell lines.; IC50's are aM for MDR cell lines.
  • the increase in lifespan (ILS) for CDF1 mice after ip injection of P388 tumor cells was in the range of ⁇ 45 to +70% over the dose range of 0.13 to 0.006 mg/kg.
  • Conventional immunotoxins contain an MAb chemically conjugated to a toxin that is mutated or chemically modified to minimized binding to normal cells. Examples include anti-B4-blocked ricin, targeting CD5; and RFB4-deglycosylated ricin A chain, targeting CD22.
  • Recombinant immunotoxins developed more recently are chimeric proteins consisting of the variable region of an antibody directed against a tumor antigen fused to a protein toxin using recombinant DNA technology. The toxin is also frequently genetically modified to remove normal tissue binding sites but retain its cytotoxicity.
  • a large number of differentiation antigens, overexpressed receptors, or cancer-specific antigens have been identified as targets for immunotoxins, e.g., CD19, CD22, CD20, IL-2 receptor (CD25), CD33, IL-4 receptor, EGF receptor and its mutants, ErB2, Lewis carbohydrate, mesothelin, transferrin receptor, GM-CSF receptor, Ras, Bcr-Abl, and c-Kit, for the treatment of a variety of malignancies including hematopoietic cancers, glioma, and breast, colon, ovarian, bladder, and gastrointestinal cancers. See e.g., Brinkmann et al., Expert Opin. Biol. Ther. 1:693-702 (2001); Perentesis and Sievers, Hematology/Oncology Clinics of North America 15:677-701 (2001).
  • MAbs conjugated with radioisotope are used as another means of treating human malignancies, particularly hematopoietic malignancies, with a high level of specificity and effectiveness.
  • the most commonly used isotopes for therapy are the high-energy-emitters, such as 131 I and 90 Y.
  • 213 Bi-labeled anti-CD33 humanized MAb has also been tested in phase I human clinical trials. Reff et al., supra.
  • rituximab a recombinant chimeric anti-CD20 MAb
  • rituximab a recombinant chimeric anti-CD20 MAb
  • Other MAbs that have since been approved for therapeutic uses in treating human cancers include: alemtuzumab (Campath-1HTM), a humanized rat antibody against CD52; and gemtuzumab ozogamicin (MylotargTM), a calicheamicin-conjugated humanized mouse antCD33 MAb.
  • the FDA is also currently examining the safety and efficacy of several other MAbs for the purpose of site-specific delivery of cytotoxic agents or radiation, e.g., radiolabeled ZevalinTM and BexxarTM. Romb al., supra.
  • a second important consideration in designing a drug delivery system is the accessibility of a target tissue to a therapeutic agent. This is an issue of particular concern in the case of treating a disease of the central nervous system (CNS), where the blood-brain barrier prevents the diffusion of macromolecules.
  • CNS central nervous system
  • Several approaches have been developed to bypass the blood-brain barrier for effective delivery of therapeutic agents to the CNS.
  • BBB blood-brain barrier
  • peptides can be coupled with a Mab directed against the transferrin receptor to achieve greater uptake by the brain, Moos and Morgan, Supra.
  • a Mab directed against the transferrin receptor when coupled with an MAb directed against the transferrin receptor, the transportation of basic fibroblast growth factor (bFGF) across the blood-brain barrier is enhanced.
  • bFGF basic fibroblast growth factor
  • a liposomal delivery system for effective transport of the chemotherapy drug, doxorubicin, into C6 glioma has been reported, where transferrin was attached to the distal ends of liposomal PEG chains.
  • U.S. Pat. Nos. 5,672,683, 5,977,307 and WO 95/02421 relate to a method of delivering a neuropharmaceutical agent across the blood-brain barrier, where the agent is administered in the form of a fusion protein with a ligand that is reactive with a brain capillary endothelial cell receptor;
  • WO 99/00150 describes a drug delivery system in which the transportation of a drug across the blood-brain barrier is facilitated by conjugation with an MAb directed against human insulin receptor;
  • WO 89/10134 describes a chimeric peptide, which includes a peptide capable of crossing the blood brain barrier at a relatively high rate and a hydrophilic neuropeptide incapable of transcytosis, as a means of introducing hydrophilic neuropeptides into the brain;
  • WO 01/60411 A1 provides a pharmaceutical composition that can easily transport a pharmaceutically active ingredient into the brain.
  • the active ingredient is bound to a hibernation-specific protein that is used as a conjugate, and administered with a thyroid hormone or a substance promoting thyroid hormone production.
  • a hibernation-specific protein that is used as a conjugate
  • a thyroid hormone or a substance promoting thyroid hormone production is explored.
  • an alternative route of drug delivery for bypassing the blood-brain barrier has been explored.
  • intranasal delivery of therapeutic agents without the need for conjugation has been shown to be a promising alternative delivery method (Frey, 2002, Drug Delivery Technology, 2(5):46-49).
  • transferrin-transferrin receptor interaction is also useful for specific targeting of certain tumor cells, as many tumor cells overexpress transferrin receptor on their surface.
  • This strategy has been used for delivering bioactive macromolecules into K562 cells via a transferrin conjugate (Wellhoner et al., The Journal of Biological Chemistry 266:4309-4314 (1991)), and for delivering insulin into enterocyte-like Caco-2 cells via a transferrin conjugate (Shah and Shen, Journal of Pharmaceutical Sciences 85:1306-1311 (1996)).
  • iron transport proteins such as lactotransferrin receptor, melanotransferrin, ceruloplasmin, and Divalent Cation Transporter and their expression pattern
  • some of the proteins involved in iron transport mechanism(e.g., melanotransferrin), or their fragments have been found to be similarly effective in assisting therapeutic agents transport across the blood-brain barrier or targeting specific tissues (WO 02/13843 A2, WO 02/13873 A2).
  • transferrin and related proteins involved in iron uptake as conjugates in drug delivery see Li and Qian, Medical Research Reviews 22:225-250 (2002).
  • tissue-specific delivery of therapeutic agents goes beyond the interaction between transferrin and transferrin receptor or their related proteins.
  • a bone-specific delivery system has been described in which proteins are conjugated with a bone-seeking aminobisphosphate for improved delivery of proteins to mineralized tissue. Uludag and Yang, Biotechnol. Prog. 18:604-611 (2002). For a review on this topic, see Vyas et al., Critical Reviews in Therapeutic Drug Carrier System 18:1-76 (2001).
  • linkers may be used in the process of generating bioconjugates for the purpose of specific delivery of therapeutic agents,.
  • Suitable linkers include homo- and heterobifunctional cross-linking reagents, which may be cleavable by, e.g., acid-catalyzed dissociation, or non-cleavable (see, e.g., Srinivasachar and Neville, Biochemistry 28:2501-2509 (1989); Wellhoner et al., The Journal of Biological Chemistry 266:4309-4314 (1991)).
  • proteins may be used to deliver molecules to intracellular compartments as conjugates. Proteins, peptides, hormones, cytokines, small molecules or the like that bind to specific cell surface receptors that are internalized after ligand binding may be used for intracellular targeting of conjugated therapeutic compounds.
  • the receptor-ligand complex is internalized into intracellular vesicles that are delivered to specific cell compartments, including, but not limited to, the nucleus, mitochondria, golgi, ER, lysosome, and endosome, depending on the intracellular location targeted by the receptor.
  • the drug By conjugating the receptor ligand with the desired molecule, the drug will be carried with the receptor-ligand complex and be delivered to the intracellular compartments normally targeted by the receptor. The drug can therefore be delivered to a specific intracellular location in the cell where it is needed to treat a disease.
  • Targeting proteins include, but are not limited to, growth factors (EPO, HGH, EGF, nerve growth factor, FGF, among others), cytokines (GM-CSF, G-CSF, the interferon family, interleukins, among others), hormones (FSH, LH, the steroid families, estrogen, corticosteroids, insulin, among others), serum proteins (albumin, lipoproteins, fetoprotein, human serum proteins, antibodies and fragments of antibodies, among others), and vitamins (folate, vitamin C, vitamin A, among others).
  • EPO growth factors
  • Contemplated linkage configurations include, but are not limited to, protein-sugar-linker-sugar-protein and multivalent forms thereof, protein-sugar-linker-protein and multivalent forms thereof, protein-sugar-linker-therapeutic agent, where the therapeutic agent includes, but are not limited to, small molecules, peptides and lipids.
  • a hydrolysable linker is used that can be hydrolyzed once internalized.
  • An acid labile linker can be used to advantage where the protein conjugate is internalized into the endosomes or lysosomes which have an acidic pH. Once internalized into the endosome or lysosome, the linker is hydrolyzed and the therapeutic agent is released from the targeting agent.
  • transferrin is conjugated via a linker to an enzyme or a nucleic acid vector that encoded the enzyme desired to be targeted to a cell that presents transferrin receptors in a patient.
  • the patient could, for example, require enzyme replacement therapy for that particular enzyme.
  • the enzyme is one that is lacking in a patient with a lysosomal storage disease (see Table 5).
  • the transferrin-enzyme conjugate is linked to transferrin receptors and is internalized in early endosomes (Xing et al., 1998, Biochem. J. 336:667; Li et al., 2002, Trends in Pharmcol. Sci. 23:206; Suhaila et al., 1998, J.
  • contemplated targeting agents that are related to transferrin include, but are not limited to, lactotransferrin (lactoferrin), melanotransferrin (p97), ceruloplasmin, and divalent cation transporter.
  • transferrin-dystrophin conjugates would enter endosomes by the transferrin pathway. Once there, the dystrophin is released due to a hydrolysable linker which can then be taken to the intracellular compartment where it is required.
  • This embodiment may be used to treat a patient with muscular dystrophy by supplementing a genetically defective dystrophin gene and/or protein with the functional dystrophin peptide connected to the transferrin.
  • the modified sugar includes a therapeutic moiety.
  • a therapeutic moiety Those of skill in the art will appreciate that there is overlap between the category of therapeutic moieties and biomolecules; many biomolecules have therapeutic properties or potential.
  • the therapeutic moieties can be agents already accepted for clinical use or they can be drugs whose use is experimental, or whose activity or mechanism of action is under investigation.
  • the therapeutic moieties can have a proven action in a given disease state or can be only hypothesized to show desirable action in a given disease state.
  • the therapeutic moieties are compounds, which are being screened for their ability to interact with a tissue of choice.
  • Therapeutic moieties, which are useful in practicing the instant invention include drugs from a broad range of drug classes having a variety of pharmacological activities. In some embodiments, it is preferred to use therapeutic moieties that are not sugars.
  • a sugar that is modified by covalent attachment of another entity, such as a PEG, biomolecule, therapeutic moiety, diagnostic moiety and the like.
  • an antisense nucleic acid moeity is conjugated to a linker arm which is attached to the targeting moiety.
  • a therapeutic sugar moiety is conjugated to a linker arm and the sugar-linker arm cassette is subsequently conjugated to a peptide via a method of the invention.
  • the therapeutic moiety is attached to the modified sugar via a linkage that is cleaved under selected conditions.
  • exemplary conditions include, but are not limited to, a selected pH (e.g., stomach, intestine, endocytotic vacuole), the presence of an active enzyme (e.g., esterase, protease, reductase, oxidase), light, heat and the like.
  • an active enzyme e.g., esterase, protease, reductase, oxidase
  • light heat and the like.
  • cleavable groups are known in the art. See, for example, Jung et al., Biochem. Biophys. Acta, 761: 152-162 (1983); Joshi et al., J. Biol. Chem., 265: 14518-14525 (1990); Zarling et al., J.
  • Classes of useful therapeutic moieties include, for example, non-steroidal anti-inflammatory drugs (NSAIDS).
  • NSAIDS non-steroidal anti-inflammatory drugs
  • the NSAIDS can, for example, be selected from the following categories: (e.g., propionic acid derivatives, acetic acid derivatives, fenamic acid derivatives, biphenylcarboxylic acid derivatives and oxicams); steroidal anti-inflammatory drugs including hydrocortisone and the like; adjuvants; antihistaminic drugs (e.g., chlorpheniramine, triprolidine); antitussive drugs (e.g., dextromethorphan, codeine, caramiphen and carbetapentane); antipruritic drugs (e.g., methdilazine and trimeprazine); anticholinergic drugs (e.g., scopolamine, atropine, homatropine, levodopa); anti-emetic and antinauseant drugs (e.g.,
  • Classes of useful therapeutic moieties include adjuvants.
  • the adjuvants can, for example, be selected from keyhole lymphet hemocyanin conjugates, monophosphoryl lipid A, mycoplasma-derived lipopeptide MALP-2, cholera toxin B subunit, Escherichia coli heat-labile toxin, universal T helper epitope from tetanus toxoid, interleukin-12, CpG oligodeoxynucleotides, dimethyldioctadecylammonium bromide, cyclodextrin, squalene, aluminum salts, meningococcal outer membrane vesicle (OMV), montanide ISA, TiterMaxTM (available from Sigma, St.
  • OMV meningococcal outer membrane vesicle
  • TiterMaxTM available from Sigma, St.
  • Antimicrobial drugs which are preferred for incorporation into the present composition include, for example, pharmaceutically acceptable salts of ⁇ -lactam drugs, quinolone drugs, ciprofloxacin, norfloxacin, tetracycline, erythromycin, amikacin, triclosan, doxycycline, capreomycin, chlorhexidine, chlortetracycline, oxytetracycline, clindamycin, ethambutol, hexamidine isothionate, metronidazole, pentamidine, gentamycin, kanamycin, lineomycin, methacycline, methenamine, minocycline, neomycin, netilmycin, paromomycin, streptomycin, tobramycin, miconazole and amantadine.
  • drugs e.g., antiandrogens (e.g., leuprolide or flutamide), cytocidal agents (e.g., adriamycin, doxorubicin, taxol, cyclophosphamide, busulfan, cisplatin, ⁇ -2-interferon) anti-estrogens (e.g., tamoxifen), antimetabolites (e.g., fluorouracil, methotrexate, mercaptopurine, thioguanine).
  • antiandrogens e.g., leuprolide or flutamide
  • cytocidal agents e.g., adriamycin, doxorubicin, taxol, cyclophosphamide, busulfan, cisplatin, ⁇ -2-interferon
  • anti-estrogens e.g., tamoxifen
  • antimetabolites e.g., fluorouracil,
  • radioisotope-based agents for both diagnosis and therapy and conjugated toxins, such as ricin, geldanamycin, mytansin, CC-1065, C-1027, the duocarmycins, calicheamycin and related structures and analogues thereof, and the toxins listed in Table 2.
  • the therapeutic moiety can also be a hormone (e.g., medroxyprogesterone, estradiol, leuprolide, megestrol, octreotide or somatostatin); muscle relaxant drugs (e.g., cinnamedrine, cyclobenzaprine, flavoxate, orphenadrine, papaverine, mebeverine, idaverine, ritodrine, diphenoxylate, dantrolene and azumolen); antispasmodic drugs; bone-active drugs (e.g., diphosphonate and phosphonoalkylphosphinate drug compounds); endocrine modulating drugs (e.g., contraceptives (e.g., ethinodiol, ethinyl estradiol, norethindrone, mestranol, desogestrel, medroxyprogesterone), modulators of diabetes (e.g., glyburide or
  • estrogens e.g., diethylstilbesterol
  • glucocorticoids e.g., triamcinolone, betamethasone, etc.
  • progesterones such as norethindrone, ethynodiol, norethindrone, levonorgestrel
  • thyroid agents e.g., liothyronine or levothyroxine
  • anti-thyroid agents e.g., methimazole
  • antihyperprolactinemic drugs e.g., cabergoline
  • hormone suppressors e.g., danazol or goserelin
  • oxytocics e.g., methylergonovine or oxytocin
  • prostaglandins such as mioprostol, alprostadil or dinoprostone
  • immunomodulating drugs e.g., antihistamines, mast cell stabilizers, such as lodoxamide and/or cromolyn
  • steroids e.g., triamcinolone, beclomethazone, cortisone, dexamethasone, prednisolone, methylprednisolone, beclomethasone, or clobetasol
  • histamine H2 antagonists e.g., famotidine, cimetidine, ranitidine
  • immunosuppressants e.g., azathioprine, cyclosporin
  • Groups with anti-inflammatory activity such as sulindac, etodolac, ketoprofen and ketorolac, are also of use.
  • Other drugs of use in conjunction with the present invention will be apparent to those of skill in the art.
  • Classes of useful therapeutic moieties include, for example, antisense drugs and also naked DNA.
  • the antisense drugs can be selected from for example Affinitak (ISIS, Carlsbad, Calif.) and GenasenseTM (from Genta, Berkeley Heights, N.J.). Naked DNA can be delivered as a gene therapy therapeutic for example with the DNA encoding for example factors VIII and IX for treatment of hemophilia disorders.
  • Modified sugars useful in forming the conjugates of the invention are discussed herein.
  • the discussion focuses on preparing a sugar modified with a water-soluble polymer for clarity of illustration.
  • the discussion focuses on the preparation of modified sugars that include a poly(ethylene glycol) moiety.
  • Those of skill will appreciate that the methods set forth herein are broadly applicable to the preparation of modified sugars, therefore, the discussion should not be interpreted as limiting the scope of the invention.
  • the sugar moiety and the modifying group are linked together through the use of reactive groups, which are typically transformed by the linking process into a new organic functional group or unreactive species.
  • the sugar reactive functional group(s) is located at any position on the sugar moiety.
  • Reactive groups and classes of reactions useful in practicing the present invention are generally those that are well known in the art of bioconjugate chemistry. Currently favored classes of reactions available with reactive sugar moieties are those, which proceed under relatively mild conditions.
  • nucleophilic substitutions e.g., reactions of amines and alcohols with acyl halides, active esters
  • electrophilic substitutions e.g., enamine reactions
  • additions to carbon-carbon and carbon-heteroatom multiple bonds e.g., Michael reaction, Diels-Alder addition.
  • Useful reactive functional groups pendent from a sugar nucleus or modifying group include, but are not limited to:
  • haloalkyl groups wherein the halide can be later displaced with a nucleophilic group such as, for example, an amine, a carboxylate anion, thiol anion, carbanion, or an alkoxide ion, thereby resulting in the covalent attachment of a new group at the functional group of the halogen atom;
  • a nucleophilic group such as, for example, an amine, a carboxylate anion, thiol anion, carbanion, or an alkoxide ion
  • dienophile groups which are capable of participating in Diels-Alder reactions such as, for example, maleimido groups;
  • aldehyde or ketone groups such that subsequent derivatization is possible via formation of carbonyl derivatives such as, for example, imines, hydrazones, semicarbazones or oximes, or via such mechanisms as Grignard addition or alkyllithium addition;
  • amine or sulfhydryl groups which can be, for example, acylated, alkylated or oxidized;
  • alkenes which can undergo, for example, cycloadditions, acylation, Michael addition, etc.
  • the reactive functional groups can be chosen such that they do not participate in, or interfere with, the reactions necessary to assemble the reactive sugar nucleus or modifying group.
  • a reactive functional group can be protected from participating in the reaction by the presence of a protecting group.
  • protecting groups see, for example, Greene et al., P ROTECTIVE G ROUPS IN O RGANIC S YNTHESIS, John Wiley & Sons, New York, 1991.
  • a sialic acid derivative is utilized as the sugar nucleus to which the modifying group is attached.
  • the focus of the discussion on sialic acid derivatives is for clarity of illustration only and should not be construed to limit the scope of the invention.
  • Those of skill in the art will appreciate that a variety of other sugar moieties can be activated and derivatized in a manner analogous to that set forth using sialic acid as an example.
  • numerous methods are available for modifying galactose, glucose, N-acetylgalactosamine and fucose to name a few sugar substrates, which are readily modified by art recognized methods. See, for example, Elhalabi et al., Curr. Med. Chem. 6: 93 (1999); and Schafer et al., J. Org. Chem. 65: 24 (2000).
  • the peptide that is modified by a method of the invention is a peptide that is produced in mammalian cells (e.g., CHO cells) or in a transgenic animal and thus, contains N- and/or O-linked oligosaccharide chains, which are incompletely sialylated.
  • the oligosaccharide chains of the glycopeptide lacking a sialic acid and containing a terminal galactose residue can be PEGylated, PPGylated or otherwise modified with a modified sialic acid.
  • the mannosamine glycoside 1 is treated with the active ester of a protected amino acid (e.g., glycine) derivative, converting the sugar amine residue into the corresponding protected amino acid amide adduct.
  • the adduct is treated with an aldolase to form the sialic acid 2.
  • Compound 2 is converted to the corresponding CMP derivative by the action of CMP-SA synthetase, followed by catalytic hydrogenation of the CMP derivative to produce compound 3.
  • the amine introduced via formation of the glycine adduct is utilized as a locus of PEG or PPG attachment by reacting compound 3 with an activated PEG or PPG derivative (e.g., PEG-C(O)NHS, PPG-C(O)NHS), producing 4 or 5, respectively.
  • an activated PEG or PPG derivative e.g., PEG-C(O)NHS, PPG-C(O)NHS
  • Table 3 sets forth representative examples of sugar monophosphates that are derivatized with a PEG or PPG moiety. Certain of the compounds of Table 3 are prepared by the method of Scheme 1. Other derivatives are prepared by art-recognized methods. See, for example, Keppler et al., Glycobiology 11: 11R (2001); and Charter et al., Glycobiology 10: 1049 (2000)). Other amine reactive PEG and PPG analogues are commercially available, or they can be prepared by methods readily accessible to those of skill in the art. TABLE 3 Examples of sugar monophosphates that are derivatized with a PEG or PPG moiety
  • modified sugar phosphates of use in practicing the present invention can be substituted in other positions as well as those set forth above.
  • “i” may be Na or another salt and “i” may be interchangeable with Na.
  • Presently preferred substitutions of sialic acid are set forth in Formula 5.
  • X is a linking group, which is preferably selected from —O—, —N(H)—, —S, CH 2 —, and N(R) 2 , in which each R is a member independently selected from R 1 -R 5 .
  • “i” may be Na or another salt, and Na may be interchangeable with “i:The symbols Y, Z, A and B each represent a group that is selected from the group set forth above for the identity of X, X, Y, Z, A and B are each independently selected and, therefore, they can be the same or different.
  • the symbols R 1 , R 2 , R 3 , R 4 and R 5 represent H, polymers, a water-soluble polymer, therapeutic moiety, biomolecule or other moiety.
  • the symbol R6 represents H, OH, or a polymer. Alternatively, these symbols represent a linker that is linked to a polymer, water-soluble polymer, therapeutic moiety, biomolecule or other moiety.
  • a mannosamine is simultaneously acylated and activated for a nucleophilic substitution by the use of chloroacetic anhydride as set forth in Scheme 5.
  • i + or Na + can be interchangeable, wherein the salt can be sodium, or can be any other suitable salt.
  • the resulting chloro-derivatized glycan is contacted with pyruvate in the presence of an aldolase, forming a chloro-derivatized sialic acid.
  • the corresponding nucleotide sugar is prepared by contacted the sialic acid derivative with an appropriate nucleotide triphosphates and a synthetase.
  • the chloro group on the sialic acid moiety is then displaced with a nucleophilic PEG derivative, such as thio-PEG.
  • a mannosamine is acylated with a bis-HOBT dicarboxylate, producing the corresponding amido-alkyl-carboxylic acid, which is subsequently converted to a sialic acid derivative.
  • the sialic acid derivative is converted to a nucleotide sugar, and the carboxylic acid is activated and reacted with a nucleophilic PEG derivative, such as amino-PEG.
  • amine- and carboxyl-protected neuraminic acid is activated by converting the primary hydroxyl group to the corresponding p-toluenesulfonate ester, and the methyl ester is cleaved.
  • the activated neuraminic acid is converted to the corresponding nucleotide sugar, and the activating group is displaced by a nucleophilic PEG species, such as thio-PEG.
  • the primary hydroxyl moiety of an amine- and carboxyl-protected neuraminic acid derivative is alkylated using an electrophilic PEG, such as chloro-PEG.
  • the methyl ester is subsequently cleaved and the PEG-sugar is converted to a nucleotide sugar.
  • Glycans other than sialic acid can be derivatized with PEG using the methods set forth herein.
  • the derivatized glycans, themselves, are also within the scope of the invention.
  • Scheme 9 provides an exemplary synthetic route to a PEGylated galactose nucleotide sugar.
  • the primary hydroxyl group of galactose is activated as the corresponding toluenesulfonate ester, which is subsequently converted to a nucleotide sugar.
  • Scheme 10 sets forth an exemplary route for preparing a galactose-PEG derivative that is based upon a galactose-6-amine moiety.
  • galactosamine is converted to a nucleotide sugar, and the amine moiety of galactosamine is functionalized with an active PEG derivative.
  • Scheme 11 provides another exemplary route to galactose derivatives.
  • the starting point for Scheme 11 is galactose-2-amine, which is converted to a nucleotide sugar.
  • the amine moiety of the nucleotide sugar is the locus for attaching a PEG derivative, such as Methoxy-PEG (MPEG) carboxylic acid.
  • MPEG Methoxy-PEG
  • moieties attached to the conjugates disclosed herein include, but are not limited to, PEG derivatives (e.g., acyl-PEG, acyl-alkyl-PEG, alkyl-acyl-PEG carbamoyl-PEG, aryl-PEG, alkyl-PEG), PPG derivatives (e.g., acyl-PPG, acyl-alkyl-PPG, alkyl-acyl-PPG carbamoyl-PPG, aryl-PPG), polyapartic acid, polyglutamate, polylysine, therapeutic moieties, diagnostic moieties, mannose-6-phosphate, heparin, heparan, SLeX, mannose, mannose-6-phosphate, Sialyl Lewis X, FGF, VFGF, proteins (e.g., transferrin), chondroitin, keratan, dermatan, dextran, modified dextran, amylose, bisphosphate, poly-SA, h
  • the nucleotide sugars and derivatives produced by the above processes can be used without purification. However, it is usually preferred to recover the product. Standard, well-known techniques for recovery of glycosylated saccharides such as thin or thick layer chromatography, column chromatography, ion exchange chromatography, or membrane filtration can be used. It is preferred to use membrane filtration, more preferably utilizing a reverse osmotic membrane, or one or more column chromatographic techniques for the recovery as is discussed hereinafter and in the literature cited herein. For instance, membrane filtration wherein the membranes have molecular weight cutoff of about 3000 to about 10,000 can be used to remove proteins for reagents having a molecular weight of less than 10,000 Da.
  • Nanofilter membranes are a class of reverse osmosis membranes that pass monovalent salts but retain polyvalent salts and uncharged solutes larger than about 100 to about 2,000 Daltons, depending upon the membrane used. Thus, in a typical application, saccharides prepared by the methods of the present invention will be retained in the membrane and contaminating salts will pass through.
  • Preparation of the modified sugar for use in the methods of the present invention includes attachment of a modifying group to a sugar residue and forming a stable adduct, which is a substrate for a glycosyltransferase.
  • a cross-linking agent to conjugate the modifying group and the sugar.
  • Exemplary bifunctional compounds which can be used for attaching modifying groups to carbohydrate moieties include, but are not limited to, bifunctional poly(ethylene glycols), polyamides, polyethers, polyesters and the like.
  • General approaches for linking carbohydrates to other molecules are known in the literature. See, for example, Lee et al., Biochemistry 28: 1856 (1989); Bhatia et al., Anal. Biochem.
  • An exemplary strategy involves incorporation of a protected sulfhydryl onto the sugar using the heterobifunctional crosslinker SPDP (n-succinimidyl-3-(2-pyridyldithio)propionate and then deprotecting the sulfhydryl for formation of a disulfide bond with another sulfhydryl on the modifying group.
  • SPDP heterobifunctional crosslinker
  • one of an array of other crosslinkers such as 2-iminothiolane or N-succinimidyl S-acetylthioacetate (SATA) is used to form a disulfide bond.
  • 2-iminothiolane reacts with primary amines, instantly incorporating an unprotected sulfhydryl onto the amine-containing molecule.
  • SATA also reacts with primary amines, but incorporates a protected sulfhydryl, which is later deacetylated using hydroxylamine to produce a free sulfhydryl. In each case, the incorporated sulfhydryl is free to react with other sulfhydryls or protected sulfhydryl, like SPDP, forming the required disulfide bond.
  • TPCH(S-(2-thiopyridyl)-L-cysteine hydrazide and TPMPH (S-(2-thiopyridyl) mercapto-propionohydrazide) react with carbohydrate moieties that have been previously oxidized by mild periodate treatment, thus forming a hydrazone bond between the hydrazide portion of the crosslinker and the periodate generated aldehydes.
  • TPCH and TPMPH introduce a 2-pyridylthione protected sulfhydryl group onto the sugar, which can be deprotected with DTT and then subsequently used for conjugation, such as forming disulfide bonds between components.
  • crosslinkers may be used that incorporate more stable bonds between components.
  • the heterobifunctional crosslinkers GMBS (N-gama-malimidobutyryloxy)succinimide) and SMCC (succinimidyl 4-(N-maleimido-methyl)cyclohexane) react with primary amines, thus introducing a maleimide group onto the component.
  • the maleimide group can subsequently react with sulfhydryls on the other component, which can be introduced by previously mentioned crosslinkers, thus forming a stable thioether bond between the components.
  • crosslinkers can be used which introduce long spacer arms between components and include derivatives of some of the previously mentioned crosslinkers (i.e., SPDP).
  • SPDP derivatives of some of the previously mentioned crosslinkers
  • a variety of reagents are used to modify the components of the modified sugar with intramolecular chemical crosslinks (for reviews of crosslinking reagents and crosslinking procedures see: Wold, F., Meth. Enzymol. 25: 623-651, 1972; Weetall, H. H., and Cooney, D. A., In: E NZYMES AS D RUGS . (Holcenberg, and Roberts, eds.) pp. 395-442, Wiley, N.Y., 1981; Ji, T. H., Meth. Enzymol. 91: 580-609, 1983; Mattson et al., Mol. Biol. Rep. 17: 167-183, 1993, all of which are incorporated herein by reference).
  • Preferred crosslinking reagents are derived from various zero-length, homo-bifunctional, and hetero-bifunctional crosslinking reagents.
  • Zero-length crosslinking reagents include direct conjugation of two intrinsic chemical groups with no introduction of extrinsic material. Agents that catalyze formation of a disulfide bond belong to this category.
  • Another example is reagents that induce condensation of a carboxyl and a primary amino group to form an amide bond such as carbodiimides, ethylchloroformate, Woodward's reagent K (2-ethyl-5-phenylisoxazolium-3′-sulfonate), and carbonyldiimidazole.
  • transglutaminase (glutamyl-peptide ⁇ -glutamyltransferase; EC 2.3.2.13) may be used as zero-length crosslinking reagent.
  • This enzyme catalyzes acyl transfer reactions at carboxamide groups of protein-linked glutaminyl residues, usually with a primary amino group as substrate.
  • Preferred homo- and hetero-bifunctional reagents contain two identical or two dissimilar sites, respectively, which may be reactive for amino, sulfhydryl, guanidino, indole, or nonspecific groups.
  • the sites on the cross-linker are amino-reactive groups.
  • amino-reactive groups include N-hydroxysuccinimide (NHS) esters, imidoesters, isocyanates, acylhalides, arylazides, p-nitrophenyl esters, aldehydes, and sulfonyl chlorides.
  • NHS esters react preferentially with the primary (including aromatic) amino groups of a modified sugar component.
  • the imidazole groups of histidines are known to compete with primary amines for reaction, but the reaction products are unstable and readily hydrolyzed.
  • the reaction involves the nucleophilic attack of an amine on the acid carboxyl of an NHS ester to form an amide, releasing the N-hydroxysuccinimide. Thus, the positive charge of the original amino group is lost.
  • Imidoesters are the most specific acylating reagents for reaction with the amine groups of the modified sugar components. At a pH between 7 and 10, imidoesters react only with primary amines. Primary amines attack imidates nucleophilically to produce an intermediate that breaks down to amidine at high pH or to a new imidate at low pH. The new imidate can react with another primary amine, thus crosslinking two amino groups, a case of a putatively monofunctional imidate reacting bifunctionally. The principal product of reaction with primary amines is an amidine that is a stronger base than the original amine. The positive charge of the original amino group is therefore retained.
  • Isocyanates (and isothiocyanates) react with the primary amines of the modified sugar components to form stable bonds. Their reactions with sulfhydryl, imidazole, and tyrosyl groups give relatively unstable products.
  • Acylazides are also used as amino-specific reagents in which nucleophilic amines of the affinity component attack acidic carboxyl groups under slightly alkaline conditions, e.g. pH8.5.
  • Arylhalides such as 1,5-difluoro-2,4-dinitrobenzene react preferentially with the amino groups and tyrosine phenolic groups of modified sugar components, but also with sulfhydryl and imidazole groups.
  • p-Nitrophenyl esters of mono- and dicarboxylic acids are also useful amino-reactive groups. Although the reagent specificity is not very high, ⁇ - and ⁇ -amino groups appear to react most rapidly.
  • Aldehydes such as glutaraldehyde react with primary amines of modified sugar.
  • glutaraldehyde is capable of modifying the modified sugar with stable crosslinks.
  • the cyclic polymers undergo a dehydration to form ⁇ - ⁇ unsaturated aldehyde polymers.
  • Schiff bases are stable, when conjugated to another double bond. The resonant interaction of both double bonds prevents hydrolysis of the Schiff linkage.
  • amines at high local concentrations can attack the ethylenic double bond to form a stable Michael addition product.
  • Aromatic sulfonyl chlorides react with a variety of sites of the modified sugar components, but reaction with the amino groups is the most important, resulting in a stable sulfonamide linkage.
  • the sites are sulfhydryl-reactive groups.
  • sulfhydryl-reactive groups include maleimides, alkyl halides, pyridyl disulfides, and thiophthalimides.
  • Maleimides react preferentially with the sulfhydryl group of the modified sugar components to form stable thioether bonds. They also react at a much slower rate with primary amino groups and the imidazole groups of histidines. However, at pH 7 the maleimide group can be considered a sulfhydryl-specific group, since at this pH the reaction rate of simple thiols is 1000-fold greater than that of the corresponding amine.
  • Alkyl halides react with sulfhydryl groups, sulfides, imidazoles, and amino groups. At neutral to slightly alkaline pH, however, alkyl halides react primarily with sulfhydryl groups to form stable thioether bonds. At higher pH, reaction with amino groups is favored.
  • Pyridyl disulfides react with free sulfhydryls via disulfide exchange to give mixed disulfides.
  • pyridyl disulfides are the most specific sulfhydryl-reactive groups.
  • Thiophthalimides react with free sulfhydryl groups to form disulfides.
  • carbodiimides soluble in both water and organic solvent are used as carboxyl-reactive reagents. These compounds react with free carboxyl groups forming a pseudourea that can then coupled to available amines yielding an amide linkage. Procedures to modify a carboxyl group with carbodiimide is well know in the art (see, Yamada et al., Biochemistry 20: 4836-4842, 1981).
  • the present invention contemplates the use of non-specific reactive groups to link the sugar to the modifying group.
  • Exemplary non-specific cross-linkers include photoactivatable groups, completely inert in the dark, which are converted to reactive species upon absorption of a photon of appropriate energy.
  • photoactivatable groups are selected from precursors of nitrenes generated upon heating or photolysis of azides. Electron-deficient nitrenes are extremely reactive and can react with a variety of chemical bonds including N—H, O—H, C—H, and C ⁇ C. Although three types of azides (aryl, alkyl, and acyl derivatives) may be employed, arylazides are presently preferred. The reactivity of arylazides upon photolysis is better with N—H and O—H than C—H bonds.
  • Electron-deficient arylnitrenes rapidly ring-expand to form dehydroazepines, which tend to react with nucleophiles, rather than form C—H insertion products.
  • the reactivity of arylazides can be increased by the presence of electron-withdrawing substituents such as nitro or hydroxyl groups in the ring. Such substituents push the absorption maximum of arylazides to longer wavelength.
  • Unsubstituted arylazides have an absorption maximum in the range of 260-280 nm, while hydroxy and nitroarylazides absorb significant light beyond 305 nm. Therefore, hydroxy and nitroarylazides are most preferable since they allow to employ less harmful photolysis conditions for the affinity component than unsubstituted arylazides.
  • photoactivatable groups are selected from fluorinated arylazides.
  • the photolysis products of fluorinated arylazides are arylnitrenes, all of which undergo the characteristic reactions of this group, including C—H bond insertion, with high efficiency (Keana et al., J. Org. Chem. 55: 3640-3647, 1990).
  • photoactivatable groups are selected from benzophenone residues.
  • Benzophenone reagents generally give higher crosslinking yields than arylazide reagents.
  • photoactivatable groups are selected from diazo compounds, which form an electron-deficient carbene upon photolysis. These carbenes undergo a variety of reactions including insertion into C—H bonds, addition to double bonds (including aromatic systems), hydrogen attraction and coordination to nucleophilic centers to give carbon ions.
  • photoactivatable groups are selected from diazopyruvates.
  • diazopyruvates the p-nitrophenyl ester of p-nitrophenyl diazopyruvate reacts with aliphatic amines to give diazopyruvic acid amides that undergo ultraviolet photolysis to form aldehydes.
  • the photolyzed diazopyruvate-modified affinity component will react like formaldehyde or glutaraldehyde forming crosslinks.
  • homobifunctional NHS esters include disuccinimidyl glutarate (DSG), disuccinimidyl suberate (DSS), bis(sulfosuccinimidyl) suberate (BS), disuccinimidyl tartarate (DST), disulfosuccinimidyl tartarate (sulfo-DST), bis-2-(succinimidooxycarbonyloxy)ethylsulfone (BSOCOES), bis-2-(sulfosuccinimidooxy-carbonyloxy)ethylsulfone (sulfo-BSOCOES), ethylene glycolbis(succinimidylsuccinate) (EGS), ethylene glycolbis(sulfosuccinimidylsuccinate) (sulfo-EGS), dithiobis(succinimidyl-propionate (DSP), and dithiobis(sulfosuccin
  • homobifunctional imidoesters include dimethyl malonimidate (DMM), dimethyl succinimidate (DMSC), dimethyl adipimidate (DMA), dimethyl pimelimidate (DMP), dimethyl suberimidate (DMS), dimethyl-3,3′-oxydipropionimidate (DODP), dimethyl-3,3′-(methylenedioxy)dipropionimidate (DMDP), dimethyl-,3′-(dimethylenedioxy)dipropionimidate (DDDP), dimethyl-3,3′-(tetramethylenedioxy)-dipropionimidate (DTDP), and dimethyl-3,3′-dithiobispropionimidate (DTBP).
  • DM malonimidate
  • DMSC dimethyl succinimidate
  • DMA dimethyl adipimidate
  • DMP dimethyl pimelimidate
  • DMS dimethyl suberimidate
  • DODP dimethyl-3,3′-oxydipropionimidate
  • DMDP dimethyl-3,3′
  • homobifunctional isothiocyanates include: p-phenylenediisothiocyanate (DITC), and 4,4′-diisothiocyano-2,2′-disulfonic acid stilbene (DIDS).
  • DITC p-phenylenediisothiocyanate
  • DIDS 4,4′-diisothiocyano-2,2′-disulfonic acid stilbene
  • homobifunctional isocyanates include xylene-diisocyanate, toluene-2,4-diisocyanate, toluene-2-isocyanate4-isothiocyanate, 3-methoxydiphenylmethane-4,4′-diisocyanate, 2,2′-dicarboxy-4,4′-azophenyldiisocyanate, and hexamethylenediisocyanate.
  • homobifunctional arylhalides include 1,5-difluoro-2,4-dinitrobenzene (DFDNB), and 4,4′-difluoro-3,3′-dinitrophenyl-sulfone.
  • homobifunctional aliphatic aldehyde reagents include glyoxal, malondialdehyde, and glutaraldehyde.
  • homobifunctional acylating reagents include nitrophenyl esters of dicarboxylic acids.
  • Preferred, non-limiting examples of homobifunctional aromatic sulfonyl chlorides include phenol-2,4-disulfonyl chloride, and ⁇ -naphthol-2,4-disulfonyl chloride.
  • additional amino-reactive homobifunctional reagents include erythritolbiscarbonate which reacts with amines to give biscarbamates.
  • homobifunctional maleimides include bismaleimidohexane (BMH), N,N′-(1,3-phenylene)bismaleimide, N,N′-(1,2-phenylene)bismaleimide, azophenyldimaleimide, and bis(N-maleimidomethyl)ether.
  • homobifunctional pyridyl disulfides include 1,4-di-3′-(2′-pyridyldithio)propionamidobutane (DPDPB).
  • homobifunctional alkyl halides include 2,2′-dicarboxy-4,4′-diiodoacetamidoazobenzene, ⁇ , ⁇ ′-diiodo-p-xylenesulfonic acid, ⁇ , ⁇ ′-dibromo-p-xylenesulfonic acid, N,N′-bis(b-bromoethyl)benzylamine, N,N′-di(bromoacetyl)phenylthydrazine, and 1,2-di(bromoacetyl)amino-3-phenylpropane.
  • homobifunctional photoactivatable crosslinker examples include bis- ⁇ -(4-azidosalicylamido)ethyldisulfide (BASED), di-N-(2-nitro-4-azidophenyl)-cystamine-S,S-dioxide (DNCO), and 4,4′-dithiobisphenylazide.
  • hetero-bifunctional reagents with a pyridyl disulfide moiety and an amino-reactive NHS ester include N-succinimidyl-3-(2-pyridyldithio)propionate (SPDP), succinimidyl 6-3-(2-pyridyldithio)propionamidohexanoate (LC-SPDP), sulfosuccinimidyl 6-3-(2-pyridyldithio)propionamidohexanoate (sulfo-LCSPDP), 4-succinimidyloxycarbonyl- ⁇ -methyl- ⁇ -(2-pyridyldithio)toluene (SMPT), and sulfosuccinimidyl 6- ⁇ -methyl- ⁇ -(2-pyridyldithio)toluamidohexanoate (sulfo-LC-SMPT).
  • SPDP N-succinimidyl
  • hetero-bifunctional reagents with a maleimide moiety and an amino-reactive NHS ester include succinimidyl maleimidylacetate (AMAS), succinimidyl 3-maleimidylpropionate (BMPS), N- ⁇ -maleimidobutyryloxysuccinimide ester (GMBS)N- ⁇ -maleimidobutyryloxysulfo succinimide ester (sulfo-GMBS) succinimidyl 6-maleimidylhexanoate (EMCS), succinimidyl 3-maleimidylbenzoate (SMB), m-maleimidobenzoyl-N-hydroxysuccinimide ester (MBS), m-maleimidobenzoyl-N-hydroxysulfosuccinimide ester (sulfo-MBS), succinimidyl 4-(N-maleimidomethyl)
  • hetero-bifunctional reagents with an alkyl halide moiety and an amino-reactive NHS ester include N-succinimidyl-(4-iodoacetyl)aminobenzoate (SIAB), sulfosuccinimidyl-(4-iodoacetyl)aminobenzoate (sulfo-SIAB), succinimidyl-6-(iodoacetyl)aminohexanoate (SIAX), succinimidyl-6-(6-((iodoacetyl)-amino)hexanoylamino)hexanoate (SIAXX), succinimidyl-6-(((4-(iodoacetyl)-amino)-methyl)-cyclohexane-1-carbonyl)aminohexanoate (SIAB) N-succinimidyl-(4-iodoacetyl)amin
  • a preferred example of a hetero-bifunctional reagent with an amino-reactive NHS ester and an alkyl dihalide moiety is N-hydroxysuccinimidyl 2,3-dibromopropionate (SDBP).
  • SDBP N-hydroxysuccinimidyl 2,3-dibromopropionate introduces intramolecular crosslinks to the affinity component by conjugating its amino groups.
  • the reactivity of the dibromopropionyl moiety towards primary amine groups is controlled by the reaction temperature (McKenzie et al., Protein Chem. 7: 581-592 (1988)).
  • hetero-bifunctional reagents with an alkyl halide moiety and an amino-reactive p-nitrophenyl ester moiety include p-nitrophenyl iodoacetate (NPIA).
  • cross-linking agents are known to those of skill in the art. See, for example, Pomato et al., U.S. Pat. No. 5,965,106. It is within the abilities of one of skill in the art to choose an appropriate cross-linking agent for a particular application.
  • the linker group is provided with a group that can be cleaved to release the modifying group from the sugar residue.
  • cleavable groups are known in the art. See, for example, Jung et al., Biochem. Biophys. Acta 761: 152-162 (1983); Joshi et al., J. Biol. Chem. 265: 14518-14525 (1990); Zarling et al., J. Immunol. 124: 913-920 (1980); Bouizar et al., Eur. J. Biochem. 155: 141-147 (1986); Park et al., J Biol. Chem. 261: 205-210 (1986); Browning et al., J. Immunol. 143: 1859-1867 (1989).
  • cleavable, bifunctional (both homo- and hetero-bifunctional) linker groups is commercially available from suppliers such as Pierce.
  • cleavable moieties can be cleaved using light, heat or reagents such as thiols, hydroxylamine, bases, periodate and the like. Moreover, certain preferred groups are cleaved in vivo in response to being endocytosed (e.g., cis-aconityl; see, Shen et al., Biochem. Biophys. Res. Commun. 102: 1048 (1991)). Preferred cleavable groups comprise a cleavable moiety which is a member selected from the group consisting of disulfide, ester, imide, carbonate, nitrobenzyl, phenacyl and benzoin groups.
  • the modified sugars are conjugated to a glycosylated or non-glycosylated peptide using an appropriate enzyme to mediate the conjugation.
  • concentrations of the modified donor sugar(s), enzyme(s) and acceptor peptide(s) are selected such that glycosylation proceeds until the acceptor is consumed.
  • the present invention is practiced using a single glycosyltransferase or a combination of glycosyltransferases.
  • a single glycosyltransferase or a combination of glycosyltransferases For example, one can use a combination of a sialyltransferase and a galactosyltransferase.
  • the enzymes and substrates are preferably combined in an initial reaction mixture, or the enzymes and reagents for a second enzymatic reaction are added to the reaction medium once the first enzymatic reaction is complete or nearly complete.
  • each of the first and second enzyme is a glycosyltransferase.
  • one enzyme is an endoglycosidase.
  • one enzyme is an exoglycosidase.
  • more than two enzymes are used to assemble the modified glycoprotein of the invention. The enzymes are used to alter a saccharide structure on the peptide at any point either before or after the addition of the modified sugar to the peptide.
  • At least two of the enzymes are glycosyltransferases and the last sugar added to the saccharide structure of the peptide is a non-modified sugar. Instead, the modified sugar is internal to the glycan structure and therefore need not be the ultimate sugar on the glycan.
  • galactosyltransferase may catalyze the transfer of Gal-PEG from UDP-Gal-PEG onto the glycan, followed by incubation in the presence of ST3Gal3 and CMP-SA, which serves to add a “capping” unmodified sialic acid onto the glycan ( FIG. 23A ).
  • At least two of the enzymes used are glycosyltransferases, and at least two modified sugars are added to the glycan structures on the peptide.
  • two or more different glycoconjugates may be added to one or more glycans on a peptide. This process generates glycan structures having two or more functionally different modified sugars.
  • incubation of the peptide with GnT-I, II and UDP-GlcNAc-PEG serves to add a GlcNAc-PEG molecule to the glycan; incubation with galactosyltransferase and UDP-Gal then serves to add a Gal residue thereto; and, incubation with ST3Gal3 and CMP-SA-Man-6-Phosphate serves to add a SA-mannose-6-phosphate molecule to the glycan.
  • This series of reactions results in a glycan chain having the functional characteristics of a PEGylated glycan as well as mannose-6-phosphate targeting activity ( FIG. 23B ).
  • At least two of the enzymes used in the reaction are glycosyltransferases, and again, different modified sugars are added to N-linked and O-linked glycans on the peptide.
  • This embodiment is useful when two different modified sugars are to be added to the glycans of a peptide, but when it is important to spatially separate the modified sugars on the peptide from each other.
  • the modified sugars comprise bulky molecules, including but not limited to, PEG and other molecules such as a linker molecule, this method may be preferable.
  • the modified sugars may be added simultaneously to the glycan structures on a peptide, or they may be added sequentially.
  • incubation with ST3Gal3 and CMP-SA-PEG serves to add sialic acid-PEG to the N-linked glycans
  • incubation with ST3 Gal1 and CMP-SA-bisPhosphonate serves to add sialic acid-BisPhosphonate to the O-linked glycans ( FIG. 23C ).
  • the method makes use of one or more exo- or endoglycosidase.
  • the glycosidase is typically a mutant, which is engineered to form glycosyl bonds rather than rupture them.
  • the mutant glycanase sometimes called a glycosynthase, typically includes a substitution of an amino acid residue for an active site acidic amino acid residue.
  • the substituted active site residues will typically be Asp at position 130, Glu at position 132 or a combination thereof.
  • the amino acids are generally replaced with serine, alanine, asparagine, or glutamine. Exoglycosidases such as transialylidase are also useful.
  • the glycosyl donor molecule e.g., a desired oligo- or mono-saccharide structure
  • the reaction proceeds with the addition of the donor molecule to a GlcNAc residue on the protein.
  • the leaving group can be a halogen, such as fluoride.
  • the leaving group is a Asn, or a Asn-peptide moiety.
  • the GlcNAc residue on the glycosyl donor molecule is modified.
  • the GlcNAc residue may comprise a 1,2 oxazoline moiety.
  • each of the enzymes utilized to produce a conjugate of the invention are present in a catalytic amount.
  • the catalytic amount of a particular enzyme varies according to the concentration of that enzyme's substrate as well as to reaction conditions such as temperature, time and pH value. Means for determining the catalytic amount for a given enzyme under preselected substrate concentrations and reaction conditions are well known to those of skill in the art.
  • the temperature at which an above-described process is carried out can range from just above freezing to the temperature at which the most sensitive enzyme denatures. Preferred temperature ranges are about 0° C. to about 55° C., and more preferably about 20° C. to about 37° C.
  • one or more components of the present method are conducted at an elevated temperature using a thermophilic enzyme.
  • the reaction mixture is maintained for a period of time sufficient for the acceptor to be glycosylated, thereby forming the desired conjugate. Some of the conjugate can often be detected after a few hours, with recoverable amounts usually being obtained within 24 hours or less.
  • rate of reaction is dependent on a number of variable factors (e.g., enzyme concentration, donor concentration, acceptor concentration, temperature, solvent volume), which are optimized for a selected system.
  • the present invention also provides for the industrial-scale production of modified peptides.
  • an industrial scale generally produces at least one gram of finished, purified conjugate.
  • the invention is exemplified by the conjugation of modified sialic acid moieties to a glycosylated peptide.
  • the exemplary modified sialic acid is labeled with PEG.
  • PEG-modified sialic acid and glycosylated peptides are for clarity of illustration and is not intended to imply that the invention is limited to the conjugation of these two partners.
  • One of skill understands that the discussion is generally applicable to the additions of modified glycosyl moieties other than sialic acid.
  • the discussion is equally applicable to the modification of a glycosyl unit with agents other than PEG including other water-soluble polymers, therapeutic moieties, and biomolecules.
  • An enzymatic approach can be used for the selective introduction of PEGylated or PPGylated carbohydrates onto a peptide or glycopeptide.
  • the method utilizes modified sugars containing PEG, PPG, or a masked reactive functional group, and is combined with the appropriate glycosyltransferase or glycosynthase.
  • the PEG or PPG can be introduced directly onto the peptide backbone, onto existing sugar residues of a glycopeptide or onto sugar residues that have been added to a peptide.
  • acceptor for the sialyltransferase is present on the peptide to be modified by the methods of the present invention either as a naturally occurring structure or one placed there recombinantly, enzymatically or chemically.
  • Suitable acceptors include, for example, galactosyl acceptors such as Gal ⁇ 1,4GlcNAc, Gal ⁇ 1,4GalNAc, Gal ⁇ 1,3GalNAc, lacto-N-tetraose, Gal ⁇ 1,3GlcNAc, Gal ⁇ 1,3Ara, Gal ⁇ 1,6GlcNAc, Gal ⁇ 1,4Glc (lactose), and other acceptors known to those of skill in the art (see, e.g., Paulson et al., J. Biol. Chem. 253: 5617-5624 (1978)).
  • an acceptor for the sialyltransferase is present on the peptide to be modified upon in vivo synthesis of the peptide.
  • Such peptides can be sialylated using the claimed methods without prior modification of the glycosylation pattern of the peptide.
  • the methods of the invention can be used to sialylate a peptide that does not include a suitable acceptor; one first modifies the peptide to include an acceptor by methods known to those of skill in the art.
  • a GalNAc residue is added by the action of a GalNAc transferase.
  • the galactosyl acceptor is assembled by attaching a galactose residue to an appropriate acceptor linked to the peptide, e.g., a GlcNAc.
  • the method includes incubating the peptide to be modified with a reaction mixture that contains a suitable amount of a galactosyltransferase (e.g., gal ⁇ 1,3 or gal ⁇ 1,4), and a suitable galactosyl donor (e.g., UDP-galactose).
  • a galactosyltransferase e.g., gal ⁇ 1,3 or gal ⁇ 1,4
  • a suitable galactosyl donor e.g., UDP-galactose
  • peptide-linked oligosaccharides are first “trimmed,” either in whole or in part, to expose either an acceptor for the sialyltransferase or a moiety to which one or more appropriate residues can be added to obtain a suitable acceptor.
  • Enzymes such as glycosyltransferases and endoglycosidases (see, for example U.S. Pat. No. 5,716,812) are useful for the attaching and trimming reactions.
  • FIG. 14 An exemplary embodiment of the invention in which a carbohydrate residue is “trimmed” prior to the addition of the modified sugar is set forth in FIG. 14 , which sets forth a scheme in which high mannose is trimmed back to the first generation biantennary structure.
  • a modified sugar bearing a water-soluble polymer is conjugated to one or more of the sugar residues exposed by the “trimming back.”
  • a water-soluble polymer is added via a GlcNAc moiety conjugated to the water-soluble polymer.
  • the modified GlcNAc is attached to one or both of the terminal mannose residues of the biantennary structure.
  • an unmodified GlcNAc can be added to one or both of the termini of the branched species.
  • a water-soluble polymer is added to one or both of the terminal mannose residues of the biantennary structure via a modified sugar having a galactose residue, which is conjugated to a GlcNAc residue added onto the terminal mannose residues.
  • an unmodified Gal can be added to one or both terminal GlcNAc residues.
  • a water-soluble polymer is added onto a Gal residue using a modified sialic acid.
  • FIG. 15 Another exemplary embodiment is set forth in FIG. 15 , which displays a scheme similar to that shown in FIG. 14 , in which the high mannose structure is “trimmed back” to the mannose from which the biantennary structure branches.
  • a water-soluble polymer is added via a GlcNAc modified with the polymer.
  • an unmodified GlcNAc is added to the mannose, followed by a Gal with an attached water-soluble polymer.
  • unmodified GlcNAc and Gal residues are sequentially added to the mannose, followed by a sialic acid moiety modified with a water-soluble polymer.
  • FIG. 16 sets forth a further exemplary embodiment using a scheme similar to that shown in FIG. 14 , in which high mannose is “trimmed back” to the GlcNAc to which the first mannose is attached.
  • the GlcNAc is conjugated to a Gal residue bearing a water-soluble polymer.
  • an unmodified Gal is added to the GlcNAc, followed by the addition of a sialic acid modified with a water-soluble sugar.
  • the terminal GlcNAc is conjugated with Gal and the GlcNAc is subsequently fucosylated with a modified fucose bearing a water-soluble polymer.
  • FIG. 17 is a scheme similar to that shown in FIG. 14 , in which high mannose is trimmed back to the first GlcNAc attached to the Asn of the peptide.
  • the GlcNAc of the GlcNAc-(Fuc)a residue is conjugated with a GlcNAc bearing a water soluble polymer.
  • the GlcNAc of the GlcNAc-(Fuc)a residue is modified with Gal, which bears a water soluble polymer.
  • the GlcNAc is modified with Gal, followed by conjugation to the Gal of a sialic acid modified with a water-soluble polymer.
  • FIGS. 18-22 Other exemplary embodiments are set forth in FIGS. 18-22 .
  • An illustration of the array of reaction types with which the present invention may be practiced is provided in each of the aforementioned figures.
  • the Examples set forth above provide an illustration of the power of the methods set forth herein. Using the methods of the invention, it is possible to “trim back” and build up a carbohydrate residue of substantially any desired structure.
  • the modified sugar can be added to the termini of the carbohydrate moiety as set forth above, or it can be intermediate between the peptide core and the terminus of the carbohydrate.
  • an existing sialic acid is removed from a glycopeptide using a sialidase, thereby unmasking all or most of the underlying galactosyl residues.
  • a peptide or glycopeptide is labeled with galactose residues, or an oligosaccharide residue that terminates in a galactose unit.
  • an appropriate sialyltransferase is used to add a modified sialic acid. The approach is summarized in Scheme 12.
  • a masked reactive functionality is present on the sialic acid.
  • the masked reactive group is preferably unaffected by the conditions used to attach the modified sialic acid to the peptide.
  • the mask is removed and the peptide is conjugated with an agent such as PEG, PPG, a therapeutic moiety, biomolecule or other agent.
  • the agent is conjugated to the peptide in a specific manner by its reaction with the unmasked reactive group on the modified sugar residue.
  • Any modified sugar can be used with its appropriate glycosyltransferase, depending on the terminal sugars of the oligosaccharide side chains of the glycopeptide (Table 4).
  • the terminal sugar of the glycopeptide required for introduction of the PEGylated or PPGylated structure can be introduced naturally during expression or it can be produced post expression using the appropriate glycosidase(s), glycosyltransferase(s) or mix of glycosidase(s) and glycosyltransferase(s).
  • X O, NH, S, CH 2 , N—(R 1-5 ) 2 .
  • M Ligand of interest
  • Ligand of interest acyl-PEG, acyl-PPG, alkyl-PEG, acyl-alkyl-PEG, acyl-alkyl-PEG, carbamoyl-PEG, carbamoyl-PPG, PEG, PPG, acyl-aryl-PEG, acyl-aryl-PPG, aryl-PEG, aryl-PPG, Mannose- 6 -phosphate, heparin, heparan, SLex, Mannose, FGF, VFGF, protein, chondroitin, keratan, dermatan, albumin, integrins, peptides, etc.
  • UDP-galactose-PEG is reacted with bovine milk ⁇ 1,4-galactosyltransferase, thereby transferring the modified galactose to the appropriate terminal N-acetylglucosamine structure.
  • the terminal GlcNAc residues on the glycopeptide may be produced during expression, as may occur in such expression systems as mammalian, insect, plant or fungus, but also can be produced by treating the glycopeptide with a sialidase and/or glycosidase and/or glycosyltransferase, as required.
  • a GlcNAc transferase such as GnT-I-IV, is utilized to transfer PEGylated-GlcNc to a mannose residue on a glycopeptide.
  • the N- and/or O-linked glycan structures are enzymatically removed from a glycopeptide to expose an amino acid or a terminal glycosyl residue that is subsequently conjugated with the modified sugar.
  • an endoglycanase is used to remove the N-inked structures of a glycopeptide to expose a terminal GlcNAc as a GlcNAc-linked-Asn on the glycopeptide.
  • UDP-Gal-PEG and the appropriate galactosyltransferase is used to introduce the PEG- or PPG-galactose functionality onto the exposed GlcNAc.
  • the modified sugar is added directly to the peptide backbone using a glycosyltransferase known to transfer sugar residues to the peptide backbone.
  • a glycosyltransferase known to transfer sugar residues to the peptide backbone.
  • This exemplary embodiment is set forth in Scheme 14.
  • Exemplary glycosyltransferases useful in practicing the present invention include, but are not limited to, GalNAc transferases (GalNAc T1-14), GlcNAc transferases, fucosyltransferases, glucosyltransferases, xylosyltransferases, mannosyltransferases and the like. Use of this approach allows the direct addition of modified sugars onto peptides that lack any carbohydrates or, alternatively, onto existing glycopeptides.
  • the addition of the modified sugar occurs at specific positions on the peptide backbone as defined by the substrate specificity of the glycosyltransferase and not in a random manner as occurs during modification of a protein's peptide backbone using chemical methods.
  • An array of agents can be introduced into proteins or glycopeptides that lack the glycosyltransferase substrate peptide sequence by engineering the appropriate amino acid sequence into the peptide chain.
  • one or more additional chemical or enzymatic modification steps can be utilized following the conjugation of the modified sugar to the peptide.
  • an enzyme e.g., fucosyltransferase
  • a glycosyl unit e.g., fucose
  • an enzymatic reaction is utilized to “cap” sites to which the modified sugar failed to conjugate.
  • a chemical reaction is utilized to alter the structure of the conjugated modified sugar. For example, the conjugated modified sugar is reacted with agents that stabilize or destabilize its linkage with the peptide component to which the modified sugar is attached.
  • a component of the modified sugar is deprotected following its conjugation to the peptide.
  • One of skill will appreciate that there is an array of enzymatic and chemical procedures that are useful in the methods of the invention at a stage after the modified sugar is conjugated to the peptide. Further elaboration of the modified sugar-peptide conjugate is within the scope of the invention.
  • the peptide is derivatized with at least one mannose-6-phosphate moiety.
  • the mannose-6-phosphate moiety targets the peptide to a lysosome of a cell, and is useful, for example, to target therapeutic proteins to lysosomes for therapy of lysosomal storage diseases.
  • Lysosomal storage diseases are a group of over 40 disorders which are the result of defects in genes encoding enzymes that break down glycolipid or polysaccharide waste products within the lysosomes of cells. The enzymatic products, e.g., sugars and lipids, are then recycled into new products. Each of these disorders results from an inherited autosomal or X-linked recessive trait which affects the levels of enzymes in the lysosome. Generally, there is no biological or functional activity of the affected enzymes in the cells and tissues of affected individuals. Table 5 provides a list of representative storage diseases and the enzymatic defect associated with the diseases.
  • enzyme replacement may also be beneficial for treating Fabry's disease, as well as other lysosomal storage diseases. See, for example, Dawson et al., Ped. Res. 7(8): 684-690 (1973) (in vitro) and Mapes et al., Science 169: 987 (1970) (in vivo). Clinical trials of enzyme replacement therapy have been reported for Fabry patients using infusions of normal plasma (Mapes et al., Science 169: 987-989 (1970)), ⁇ -galactosidase A purified from placenta (Brady et al., N. Eng. J. Med.
  • the present invention provides compositions and methods for delivering sufficient quantities of biologically active lysosomal peptides to deficient cells.
  • the present invention provides a peptide according to Table 7 that is derivatized with mannose-6-phosphate ( FIG. 24 and FIG. 25 ).
  • the peptide may be recombinantly or chemically prepared.
  • the peptide can be the full, natural sequence, or it may be modified by, for example, truncation, extension, or it may include substitutions or deletions.
  • Exemplary proteins that are remodeled using a method of the present invention include glucocerebrosidase, ⁇ -glucosidase, ⁇ -galactosidase A, acid- ⁇ -glucosidase (acid maltase).
  • modified peptides that are in clinical use include, but are not limited to, CeredaseTM, CerezymeTM, and FabryzymeTM.
  • a glycosyl group on modified and clinically relevant peptides may also be altered utilizing a method of the invention.
  • the mannose-6-phosphate is attached to the peptide via a glycosyl linking group.
  • the glycosyl linking group is derived from sialic acid.
  • Exemplary sialic acid-derived glycosyl linking groups are set forth in Table 3, in which one or more of the “R” moieties is mannose-6-phosphate or a spacer group having one or more mannose-6-phosphate moieties attached thereto.
  • the modified sialic acid moiety is preferably the terminal residue of an oligosaccharide linked to the surface of the peptide ( FIG. 26 )
  • the peptides of the invention may be further derivatized with a moiety such as a water-soluble polymer, a therapeutic moiety, or an additional targeting moiety.
  • a moiety such as a water-soluble polymer, a therapeutic moiety, or an additional targeting moiety.
  • Methods for attaching these and other groups are set forth herein.
  • the group other than mannose-6-phosphate is attached to the peptide via a derivatized sialic acid derivative according to Table 3, in which one or more of the “R” moieties is a group other than mannose-6-phosphate.
  • a sialic acid moiety modified with a Cbz-protected glycine-based linker arm is prepared.
  • the corresponding nucleotide sugar is prepared and the Cbz group is removed by catalytic hydrogenation.
  • the resulting nucleotide sugar has an available, reactive amine that is contacted with an activated mannose-6-phosphate derivative, providing a mannose-6-phosphate derivatized nucleotide sugar that is useful in practicing the methods of the invention.
  • an exemplary activated mannose-6-phosphate derivative is formed by converting a 2-bromo-benzyl-protected phosphotriester into the corresponding triflate, in situ, and reacting the triflate with a linker having a reactive oxygen-containing moiety, forming an ether linkage between the sugar and the linker.
  • the benzyl protecting groups are removed by catalytic hydrogenation, and the methyl ester of the linker is hydrolyzed, providing the corresponding carboxylic acid.
  • the carboxylic acid is activated by any method known in the art.
  • An exemplary activation procedure relies upon the conversion of the carboxylic acid to the N-hydroxysuccinimide ester.
  • a N-acetylated sialic acid is converted to an amine by manipulation of the pyruvyl moiety.
  • the primary hydroxyl is converted to a sulfonate ester and reacted with sodium azide.
  • the azide is catalytically reduced to the corresponding amine.
  • the sugar is subsequently converted to its nucleotide analogue and coupled, through the amine group, to the linker arm-derivatized mannose-6-phosphate prepared as discussed above.
  • Peptides useful to treat lysosomal storage disease can be derivatized with other targeting moieties including, but not limited to, transferrin (to deliver the peptide across the blood-brain barrier, and to endosomes), carnitine (to deliver the peptide to muscle cells), and phosphonates, e.g., bisphosphonate (to target the peptide to bone and other calciferous tissues).
  • targeting moieties including, but not limited to, transferrin (to deliver the peptide across the blood-brain barrier, and to endosomes), carnitine (to deliver the peptide to muscle cells), and phosphonates, e.g., bisphosphonate (to target the peptide to bone and other calciferous tissues).
  • transferrin to deliver the peptide across the blood-brain barrier, and to endosomes
  • carnitine to deliver the peptide to muscle cells
  • phosphonates e.g., bisphosphonate
  • the targeting agent and the therapeutic peptide are coupled via a linker moiety.
  • at least one of the therapeutic peptide or the targeting agent is coupled to the linker moiety via an intact glycosyl linking group according to a method of the invention.
  • the linker moiety includes a poly(ether) such as poly(ethylene glycol).
  • the linker moiety includes at least one bond that is degraded in vivo, releasing the therapeutic peptide from the targeting agent, following delivery of the conjugate to the targeted tissue or region of the body.
  • the in vivo distribution of the therapeutic moiety is altered via altering a glycoform on the therapeutic moiety without conjugating the therapeutic peptide to a targeting moiety.
  • the therapeutic peptide can be shunted away from uptake by the reticuloendothelial system by capping a terminal galactose moiety of a glycosyl group with sialic acid (or a derivative thereof) ( FIGS. 24 and 27 ).
  • Sialylation to cover terminal Gal avoids uptake of the peptide by hepatic asialoglycoprotein (ASGP) receptors, and may extend the half life of the peptide as compared with peptides having only complex glycan chains, in the absence of sialylation.
  • ASGP hepatic asialoglycoprotein
  • the present invention provides a composition comprising multiple copies of a single peptide having an elemental trimannosyl core as the primary glycan structure attached thereto.
  • the peptide may be a therapeutic molecule.
  • the natural form of the peptide may comprise complex N-linked glycans or may be a high mannose glycan.
  • the peptide may be a mammalian peptide, and is preferably a human peptide.
  • the peptide is selected from the group consisting of an immunoglobulin, erythropoietin, tissue-type activator peptide, and others (See FIG. 28 ).
  • Exemplary peptides whose glycans can be remodeled using the methods of the invention are set forth in FIG. 28 .
  • FIG. 28 A more detailed list of peptides useful in the invention and their source is provided in FIG. 28 .
  • exemplary peptides that are modified by the methods of the invention include members of the immunoglobulin family (e.g., antibodies, MHC molecules, T cell receptors, and the like), intercellular receptors (e.g., integrins, receptors for hormones or growth factors and the like) lectins, and cytokines (e.g., interleukins).
  • members of the immunoglobulin family e.g., antibodies, MHC molecules, T cell receptors, and the like
  • intercellular receptors e.g., integrins, receptors for hormones or growth factors and the like
  • lectins e.g., cytokines
  • tissue-type plasminogen activator renin
  • clotting factors such as Factor VIII and Factor IX
  • bombesin thrombin
  • hematopoietic growth factor colony stimulating factors
  • viral antigens complement peptides
  • ⁇ 1-antitrypsin erythropoietin
  • P-selectin glycopeptide ligand-1 PSGL-1
  • granulocyte-macrophage colony stimulating factor anti-thrombin III, interleukins, interferons, peptides A and C
  • fibrinogen herceptinTM
  • leptin glycosidases
  • the methods of the invention are also useful for modifying chimeric peptides, including, but not limited to, chimeric peptides that include a moiety derived from an immunoglobulin, such as IgG.
  • Peptides modified by the methods of the invention can be synthetic or wild-type peptides or they can be mutated peptides, produced by methods known in the art, such as site-directed mutagenesis. Glycosylation of peptides is typically either N-linked or O-linked. An exemplary N-linkage is the attachment of the modified sugar to the side chain of an asparagine residue.
  • the tripeptide sequences asparagine-X-serine and asparagine-X-threonine, where X is any amino acid except proline, are the recognition sequences for enzymatic attachment of a carbohydrate moiety to the asparagine side chain.
  • 0-linked glycosylation refers to the attachment of one sugar (e.g., N-acetylgalactosamine, galactose, mannose, GlcNAc, glucose, fucose or xylose) to a hydroxy side chain of a hydroxyamino acid, preferably serine or threonine, although 5-hydroxyproline or 5-hydroxylysine may also be used.
  • one sugar e.g., N-acetylgalactosamine, galactose, mannose, GlcNAc, glucose, fucose or xylose
  • the present invention provides methods for modifying Granulocyte Colony Stimulating Factor (G-CSF).
  • G-CSF Granulocyte Colony Stimulating Factor
  • FIGS. 29A to 29 G set forth some examples of how this is accomplished using the methodology disclosed herein.
  • FIG. 29B a G-CSF peptide that is expressed in a mammalian cell system is tried back using a sialidase. The residues thus exposed are modified by the addition of a sialic acid-poly(ethylene glycol) moiety (PEG moiety), using an appropriate donor therefor and ST3Gal1.
  • FIG. 29C sets forth an exemplary scheme for modifying a G-CSF peptide that is expressed in an insect cell.
  • the peptide is modified by adding a galactose moiety using an appropriate donor thereof and a galactosyltransferase.
  • the galactose residues are functionalized with PEG via a sialic acid-PEG derivative, through the action of ST3 Gal1.
  • bacterially expressed G-CSF is contacted with an N-acetylgalactosamine donor and N-acetylgalactosamine transferase.
  • the peptide is functionalized with PEG, using a PEGylated sialic acid donor and a sialyltransferase.
  • mammalian cell expressed G-CSF is contacted with a sialic acid donor that is modified with levulinic acid, adding a reactive ketone to the sialic acid donor. After addition to a glycosyl residue on the glycan on the peptide, the ketone is derivatized with a moiety such as a hydrazine- or amine-PEG.
  • bacterially expressed G-CSF is remodeled by contacting the peptide with an endo-GalNAc enzyme under conditions where it functions in a synthetic, rather than a hydrolytic manner, thereby adding a PEG-Gal-GalNAc molecule from an activated derivative thereof.
  • FIG. 29E mammalian cell expressed G-CSF is contacted with a sialic acid donor that is modified with levulinic acid, adding a reactive ketone to the sialic acid donor. After addition to a glycosyl residue on the glycan on the peptide, the ketone is derivat
  • 29G provides another route for remodeling bacterially expressed G-CSF.
  • the polypeptide is derivatized with a PEGylated N-acetylgalactosamine residue by contacting the polypeptide with an N-acetylgalactosamine transferase and an appropriate donor of PEGylated N-acetylgalactosamine.
  • the invention provides methods for modifying Interferon ⁇ -14C (IFN ⁇ 14C), as shown in FIGS. 30A to 30 N.
  • IFN ⁇ 14C Interferon ⁇ -14C
  • FIGS. 30A to 30 N The various forms of IFN ⁇ are disclosed elsewhere herein.
  • IFN ⁇ 14C expressed in mammalian cells is first treated with sialidase to trim back the sialic acid units thereon, and then the molecule is PEGylated using ST3Gal3 and a PEGylated sialic acid donor.
  • N-acetylglucosamine is first added to IFN ⁇ 14C which has been expressed in insect or fungal cells, where the reaction is conducted via the action of GnT-I and/or II using an N-acetylglucosamine donor.
  • the polypeptide is then PEGylated using a galactosyltransferase and a donor of PEG-galactose.
  • IFN ⁇ 14C expressed in yeast is first treated with Endo-H to trim back the glycosyl units thereon.
  • the molecules is galactosylated using a galactosyltransferase and a galactose donor, and it is then PEGylated using ST3Gal3 and a donor of PEG-sialic acid.
  • FIG. 30F IFN ⁇ 14C produced by mammalian cells is modified to inched a PEG moiety using ST3Gal3 and a donor of PEG-sialic acid.
  • IFN ⁇ 14C expressed in insect of fungal cells first has N-acetylglucosamine added using one or more of GnT-I, II, IV, and V, and an N-acetylglucosamine donor. The protein is subsequently galactosylated using an appropriate donor and a galactosyltransferase. Then, IFN ⁇ 14C is PEGylated using ST3Gal3 and a donor of PEG-sialic acid.
  • yeast produced IFN ⁇ 14C is first treated with mannosidases to trim back the mannosyl groups.
  • N-acetylglucosamine is then added using a donor of N-acetylglucosamine and one or more of GnT-I, II, IV, and V.
  • IFN ⁇ 14C is further galactosylated using an appropriate donor and a galactosyltransferase.
  • the polypeptide is PEGylated using ST3Gal3 and a donor of PEG-sialic acid.
  • FIG. 30I NSO cell expressed IFN ⁇ 14C is modified by capping appropriate terminal residues with a sialic acid donor that is modified with levulinic acid, thereby adding a reactive ketone to the sialic acid donor.
  • the ketone is derivatized with a moiety such as a hydrazine- or amine-PEG.
  • IFN ⁇ 14C expressed by mammalian cells is PEGylated using a donor of PEG-sialic acid and ⁇ 2,8-sialyltransferase.
  • IFN ⁇ 14C produced by mammalian cells is first treated with sialidase to trim back the terminal sialic acid residues, and then the molecule is PEGylated using trans-sialidase and PEGylated sialic acid-lactose complex.
  • sialidase to trim back the terminal sialic acid residues
  • IFN ⁇ 14C expressed in a mammalian system is sialylated using a donor of sialic acid and ⁇ 2,8-sialyltransferase.
  • IFN ⁇ 14C expressed in insect or fungal cells first has N-acetylglucosamine added using an appropriate donor and GnT-I and/or II. The molecule is then contacted with a galactosyltransferase and a galactose donor that is derivatized with a reactive sialic acid via a linker, so that the polypeptide is attached to the reactive sialic acid via the linker and the galactose residue.
  • IFN ⁇ 14C expressed in either insect or fungal cells is first treated with endoglycanase to trim back the glycosyl groups, and is then contacted with a galactosyltransferase and a galactose donor that is derivatized with a reactive sialic acid via a linker, so that the polypeptide is attached to the reactive sialic acid via the linker and the galactose residue.
  • the molecule is then contacted with ST3Gal3 and transferrin, and thus becomes connected with transferrin via the sialic acid residue.
  • the invention provides methods for modifying Interferon a-2a or 2b (IFN ⁇ ), as shown in FIGS. 30O to 30 EE.
  • IFN ⁇ Interferon a-2a or 2b
  • FIGS. 30O to 30 EE Interferon a-2a or 2b
  • FIG. 30P IFN ⁇ produced in mammalian cells is first treated with sialidase to trim back the glycosyl units, and is then PEGylated using ST3Gal3 and a PEGylated sialic acid donor.
  • IFN ⁇ expressed in insect cells is first galactosylated using an appropriate donor and a galactosyltransferase, and is then PEGylated using ST3Gal1 and a PEGylated sialic acid donor.
  • FIG. 30P IFN ⁇ produced in mammalian cells is first treated with sialidase to trim back the glycosyl units, and is then PEGylated using ST3Gal3 and a PEGylated
  • FIG. 30R offers another method for remodeling IFN ⁇ expressed in bacteria: PEGylated N-acetylgalactosamine is added to the protein using an appropriate donor and N-acetylgalactosamine transferase.
  • IFN ⁇ expressed in mammalian cells is modified by capping appropriate terminal residues with a sialic acid donor that is modified with levulinic acid, adding a reactive ketone to the sialic acid donor. After addition to a glycosyl residue of the peptide, the ketone is derivatized with a moiety such as a hydrazine- or amine- PEG.
  • IFN ⁇ expressed in bacteria is PEGylated using a modified enzyme Endo-N-acetylgalactosamidase, which functions in a synthetic instead of a hydrolytic manner, and using a N-acetylgalactosamine donor derivatized with a PEG moiety.
  • N-acetylgalactosamine is first added IFN ⁇ using an appropriate donor and N-acetylgalactosamine transferase, and then is PEGylated using a sialyltransferase and a PEGylated sialic acid donor.
  • FIG. 30U N-acetylgalactosamine is first added IFN ⁇ using an appropriate donor and N-acetylgalactosamine transferase, and then is PEGylated using a sialyltransferase and a PEGylated sialic acid donor.
  • IFN ⁇ expressed in a mammalian system is first treated with sialidase to trim back the sialic acid residues, and is then PEGylated using a suitable donor and ST3Gal1 and/or ST3Gal3.
  • IFNA expressed in mammalian cells is first treated with sialidase to trim back the sialic acid residues.
  • the polypeptide is then contacted with ST3Gal1 and two reactive sialic acid residues that are connect via a linker, so that the polypeptide is attached to one reactive sialic acid via the linker and the second sialic acid residue.
  • IFN ⁇ expressed in mammalian cells is first treated with sialidase to trim back the sialic acid residues, and is then PEGylated using ST3Gal1 and a donor of PEG-sialic acid.
  • IFN ⁇ produced by insect cells is PEGylated using a galactosyltransferase and a donor of PEGylated galactose.
  • FIG. 30Y IFN ⁇ expressed in mammalian cells is first treated with sialidase to trim back the sialic acid residues, and is then PEGylated using ST3Gal1 and a donor of PEG-sialic acid.
  • IFN ⁇ produced by insect cells is PEGylated using a galactosyltransferase and a donor of PEGylated galactose.
  • IFN ⁇ expressed in bacteria is remodeled in yet another scheme.
  • the polypeptide is first contacted with N-acetylgalactosamine transferase and a donor of N-acetylgalactosamine that is derivatized with a reactive sialic acid via a linker, so that IFN ⁇ is attached to the reactive sialic acid via the linker and the N-acetylgalactosamine.
  • IFN ⁇ is then contacted with ST3Gal3 and asialo-transferrin so that it becomes connected with transferrin via the sialic acid residue.
  • IFNA is capped with sialic acid residues using ST3Gal3 and a sialic acid donor.
  • An additional method for modifying bacterially expressed IFN ⁇ is disclosed in FIG. 30E E, where IFN ⁇ is first exposed to NHS-CO-linker-SA-CMP and is then connected to a reactive sialic acid via the linker. It is subsequently conjugated with transferrin using ST3Gal3 and transferrin.
  • the methods for remodeling INN omega are essentially identical to those presented here for IFN alpha except that the attachment of the glycan to the IFN omega peptide occurs at amino acid residue 101 in SEQ ID NO:75.
  • the nucleotide and amino acid sequences for IFN omega are presented herein as SEQ ID NOS:74 and 75. Methods of making and using IFN omega are found in U.S. Pat. Nos. 4,917,887 and 5,317,089, and in EP Pat. No. 0170204-A.
  • the invention provides methods for modifying Interferon ⁇ (IFN- ⁇ ), as shown in FIGS. 31A to 31 S.
  • IFN- ⁇ Interferon ⁇
  • FIGS. 31A to 31 S In FIGS. 31A to 31 S.
  • IFN- ⁇ expressed in a mammalian system is first treated with sialidase to trim back the terminal sialic acid residues.
  • the protein is then PEGylated using ST3Gal3 and a donor of PEGylated sialic acid.
  • FIG. 31C is a scheme for modifying IFN- ⁇ produced by insect cells. First, N-acetylglucosamine is added to IFN- ⁇ using an appropriate donor and GnT-I and/or -II.
  • IFN- ⁇ expressed in yeast is first treated with Endo-H to trim back its glycosyl chains, and is then galactosylated using a galactose donor and a galactosyltransferase, and is then PEGylated using ST3Gal3 and a donor of PEGylated sialic acid.
  • IFN- ⁇ expressed in yeast is first treated with Endo-H to trim back its glycosyl chains, and is then galactosylated using a galactose donor and a galactosyltransferase, and is then PEGylated using ST3Gal3 and a donor of PEGylated sialic acid.
  • FIG. 31D IFN- ⁇ expressed in yeast is first treated with Endo-H to trim back its glycosyl chains, and is then galactosylated using a galactose donor and a galactosyltransferase, and is then PEGylated using ST3
  • IFN- ⁇ produced by mammalian cells is modified by PEGylation using ST3Gal3 and a donor of sialic acid already derivatized with a PEG moiety.
  • IFN- ⁇ expressed in insect cells first has N-acetylglucosamine added by one or more of GnT-I, II, IV, and V using a N-acetylglucosamine donor, and then is galactosylated using a galactose donor and a galactosyltransferase, and is then PEGylated using ST3Gal3 and a donor of PEG-sialic acid.
  • FIG. 31E IFN- ⁇ produced by mammalian cells is modified by PEGylation using ST3Gal3 and a donor of sialic acid already derivatized with a PEG moiety.
  • IFN- ⁇ expressed in insect cells first has N-acetylglucosamine added by one or more of GnT-I, II, IV, and
  • IFN- ⁇ expressed in yeast is first treated with mannosidases to trim back the mannosyl units, then has N-acetylglucosamine added using a N-acetylglucosamine donor and one or more of GnT-I, II, IV, and V.
  • the protein is further galactosylated using a galactose donor and a galactosyltransferase, and then PEGylated using ST3Gal3 and a PEG-sialic acid donor.
  • mammalian cell expressed IFN- ⁇ is modified by capping appropriate terminal residues with a sialic acid donor that is modified with levulinic acid, adding a reactive ketone to the sialic acid donor. After addition to a glycosyl residue of the peptide, the ketone is derivatized with a moiety such as a hydrazine- or amine-PEG.
  • IFN- ⁇ expressed in a mammalian system is PEGylated using a donor of PEG-sialic acid and ⁇ 2,8-sialyltransferase.
  • FIG. 31H mammalian cell expressed IFN- ⁇ is modified by capping appropriate terminal residues with a sialic acid donor that is modified with levulinic acid, adding a reactive ketone to the sialic acid donor. After addition to a glycosyl residue of the peptide, the ketone is derivatized with a moiety such as a hydrazine- or amine-PEG
  • IFN- ⁇ expressed by mammalian cells is first treated with sialidase to trim back its terminal sialic acid residues, and then PEGylated using trans-sialidase and a donor of PEGylated sialic acid.
  • IFN- ⁇ expressed in mammalian cells is first treated with sialidase to trim back terminal sialic acid residues, then PEGylated using ST3Gal3 and a donor of PEG-sialic acid, and then sialylated using ST3Gal3 and a sialic acid donor.
  • FIG. 31J IFN- ⁇ expressed by mammalian cells is first treated with sialidase to trim back its terminal sialic acid residues, and then PEGylated using trans-sialidase and a donor of PEGylated sialic acid.
  • FIG. 31K IFN- ⁇ expressed in mammalian cells is first treated with sialidase to trim back terminal sialic acid residues, then PEGylated using ST3G
  • IFN- ⁇ expressed in mammalian cells is first treated with sialidase and galactosidase to trim back the glycosyl chains, then galactosylated using a galactose donor and an ⁇ -galactosyltransferase, and then PEGylated using ST3Gal3 or a sialyltransferase and a donor of PEG-sialic acid.
  • IFN- ⁇ expressed in mammalian cells is first treated with sialidase to trim back the glycosyl units.
  • IFN- ⁇ expressed in mammalian cells is modified by capping appropriate terminal residues with a sialic acid donor that is modified with levulinic acid, adding a reactive ketone to the sialic acid donor. After addition to a glycosyl residue of the peptide, the ketone is derivatized with a moiety such as a hydrazine- or amine-PEG.
  • a sialic acid donor that is modified with levulinic acid
  • ketone is derivatized with a moiety such as a hydrazine- or amine-PEG.
  • IFN- ⁇ expressed in mammalian cells is sialylated using a sialic acid donor and ⁇ 2,8-sialyltransferase.
  • IFN- ⁇ produced by insect cells first has N-acetylglucosamine added using a donor of N-acetylglucosamine and one or more of GnT-I, II, IV, and V, and is further PEGylated using a donor of PEG-galactose and a galactosyltransferase.
  • IFN- ⁇ expressed in yeast is first treated with endoglycanase to trim back the glycosyl groups, then galactosylated using a galactose donor and a galactosyltransferase, and then PEGylated using ST3Gal3 and a donor of PEG-sialic acid.
  • IFN- ⁇ expressed in a mammalian system is first contacted with ST3Gal3 and two reactive sialic acid residues connected via a linker, so that the polypeptide is attached to one reactive sialic acid via the linker and the second sialic acid residue.
  • polypeptide is then contacted with ST3Gal3 and desialylated transferrin, and thus becomes connected with transferrin via the sialic acid residue. Then, IFN- ⁇ is further sialylated using a sialic acid donor and ST3Gal3.
  • the invention provides methods for modifying Factor VII or VIIa, as shown in FIGS. 32 A to 32 D.
  • FIG. 32B Factor VII or VIIa produced by a mammalian system is first treated with sialidase to trim back the terminal sialic acid residues, and then PEGylated using ST3Gal3 and a donor of PEGylated sialic acid.
  • FIG. 32C Factor VII or VIIa expressed by mammalian cells is first treated with sialidase to trim back the terminal sialic acid residues, and then PEGylated using ST3Gal3 and a donor of PEGylated sialic acid.
  • FIG. 32D offers another modification scheme for Factor VII or VIIa produced by mammalian cells: the polypeptide is first treated with sialidase and galactosidase to trim back its sialic acid and galactose residues, then galactosylated using a galactosyltransferase and a galactose donor, and then PEGylated using ST3Gal3 and a donor of PEGylated sialic acid.
  • the invention provides methods for modifying Factor IX, some examples of which are included in FIGS. 33A to 33 G.
  • Factor IX produced by mammalian cells is first treated with sialidase to trim back the terminal sialic acid residues, and is then PEGylated with ST3Gal3 using a PEG-sialic acid donor.
  • FIG. 33C Factor IX expressed by mammalian cells is first treated with sialidase to trim back the terminal sialic acid residues, it is then PEGylated using ST3Gal3 and a PEG-sialic acid donor, and further sialylated using ST3Gal1 and a sialic acid donor.
  • FIG. 33D Another scheme for remodeling mammalian cell produced Factor IX can be found in FIG. 33D .
  • the polypeptide is first treated with sialidase to trim back the terminal sialic acid residues, then galactosylated using a galactose donor and a galactosyltransferase, further sialylated using a sialic acid donor and ST3Gal3, and then PEGylated using a donor of PEGylated sialic acid and ST3Gal1.
  • FIG. 33E Factor IX that is expressed in a mammalian system is PEGylated through the process of sialylation catalyzed by ST3 Gal3 using a donor of PEG-sialic acid.
  • FIG. 33E Another scheme for remodeling mammalian cell produced Factor IX can be found in FIG. 33D .
  • the polypeptide is first treated with sialidase to trim back the terminal sialic acid residues, then galactosy
  • Factor IX expressed in mammalian cells is modified by capping appropriate terminal residues with a sialic acid donor that is modified with levulinic acid, adding a reactive ketone to the sialic acid donor. After addition to a glycosyl residue of the peptide, the ketone is derivatized with a moiety such as a hydrazine- or amine-PEG.
  • FIG. 33G provides an additional method of modifying Factor IX.
  • the polypeptide, produced by mammalian cells is PEGylated using a donor of PEG-sialic acid and ⁇ 2,8-sialyltransferase.
  • FIGS. 34A to 34 J present some examples.
  • FSH is expressed in a mammalian system and modified by treatment of sialidase to trim back terminal sialic acid residues, followed by PEGylation using ST3Gal3 and a donor of PEG-sialic acid.
  • FIG. 34C FSH expressed in mammalian cells is first treated with sialidase to trim back terminal sialic acid residues, then PEGylated using ST3Gal3 and a donor of PEG-sialic acid, and then sialylated using ST3Gal3 and a sialic acid donor.
  • FIG. 34A to 34 J present some examples.
  • FIG. 34B FSH is expressed in a mammalian system and modified by treatment of sialidase to trim back terminal sialic acid residues, followed by PEGylation using ST3Gal3 and a donor of PEG-sialic acid.
  • FIG. 34C FSH expressed in mammalian cells is first treated with sialidase
  • 34D provides a scheme for modifying FSH expressed in a mammalian system.
  • the polypeptide is treated with sialidase and galactosidase to trim back its sialic acid and galactose residues, then galactosylated using a galactose donor and a galactosyltransferase, and then PEGylated using ST3Gal3 and a donor of PEG-sialic acid.
  • FSH expressed in mammalian cells is modified in the following procedure: FSH is first treated with sialidase to trim back the sialic acid residues, then PEGylated using ST3Gal3 and a donor of PEG-sialic acid, and is then sialylated using ST3Gal3 and a sialic acid donor.
  • FIG. 34F offers another example of modifying FSH produced by mammalian cells: The polypeptide is modified by capping appropriate terminal residues with a sialic acid donor that is modified with levulinic acid, adding a reactive ketone to the sialic acid donor.
  • the ketone is derivatized with a moiety such as a hydrazine- or amine-PEG.
  • FSH expressed in a mammalian system is modified in another procedure: the polypeptide is remodeled with addition of sialic acid using a sialic acid donor and an ⁇ 2,8-sialyltransferase.
  • FSH is expressed in insect cells and modified in the following procedure: N-acetylglucosamine is first added to FSH using an appropriate N-acetylglucosamine donor and one or more of GnT-I, II, IV, and V; FSH is then PEGylated using a donor of PEG-galactose and a galactosyltransferase.
  • FIG. 34I depicts a scheme of modifying FSH produced by yeast.
  • FSH is first treated with endoglycanase to trim back the glycosyl groups, galactosylated using a galactose donor and a galactosyltransferase, and is then PEGylated with ST3Gal3 and a donor of PEG-sialic acid.
  • FSH expressed by mammalian cells is first contacted with ST3Gal3 and two reactive sialic acid residues via a linker, so that the polypeptide is attached to a reactive sialic acid via the linker and a second sialic acid residue.
  • the polypeptide is then contacted with ST3Gal1 and desialylated chorionic gonadotrophin (CG) produced in CHO, and thus becomes connected with CG via the second sialic acid residue.
  • FSH is sialylated using a sialic acid donor and ST3Gal3 and/or ST3Gal1.
  • FIGS. 35A to 35 AA set forth some examples which are relevant to the remodeling of both wild-type and mutant EPO peptides.
  • FIG. 35B EPO expressed in various mammalian systems is remodeled by contacting the expressed protein with a sialidase to remove terminal sialic acid residues. The resulting peptide is contacted with a sialyltransferase and a CMP-sialic acid that is derivatized with a PEG moiety.
  • FIG. 35B EPO expressed in various mammalian systems is remodeled by contacting the expressed protein with a sialidase to remove terminal sialic acid residues. The resulting peptide is contacted with a sialyltransferase and a CMP-sialic acid that is derivatized with a PEG moiety.
  • EPO that is expressed in insect cells is remodeled with N-acetylglucosamine, using GnT-I and/or GnT-II.
  • Galactose is then added to the peptide, using galactosyltransferase.
  • PEG group is added to the remodeled peptide by contacting it with a sialyltransferase and a CMP-sialic acid that is derivatized with a PEG moiety.
  • FIG. 35D EPO that is expressed in a mammalian cell system is remodeled by removing terminal sialic acid moieties via the action of a sialidase.
  • the terminal galactose residues of the N-linked glycosyl units are “capped” with sialic acid, using ST3Gal3 and a sialic acid donor.
  • the terminal galactose residues on the O-linked glycan are functionalized with a sialic acid bearing a PEG moiety, using an appropriate sialic acid donor and ST3Gal1.
  • EPO that is expressed in a mammalian cell system is remodeled by functionalizing the N-linked glycosyl residues with a PEG-derivatized sialic acid moiety.
  • the peptide is contacted with ST3Gal3 and an appropriately modified sialic acid donor.
  • EPO that is expressed in an insect cell system, yeast or fungi is remodeled by adding at least one N-acetylglucosamine residues by contacting the peptide with a N-acetylglucosamine donor and one or more of GnT-I, GnT-II, and GnT-V.
  • the peptide is then PEGylated by contacting it with a PEGylated galactose donor and a galactosyltransferase.
  • EPO that is expressed in an insect cell system, yeast or fungi is remodeled by the addition of at least one N-acetylglucosamine residues, using an appropriate N-acetylglucosamine donor and one or more of GnT-, GnT-II, and GnT-V.
  • a galactosidase that is altered to operate in a synthetic, rather than a hydrolytic manner is used to add an activated PEGylated galactose donor to the N-acetylglucosamine residues.
  • FIG. 35H EPO that is expressed in an insect cell system, yeast or fungi, is remodeled by the addition of at least one terminal N-acetylglucosamine-PEG residue.
  • the peptide is contacted with GnT-I and an appropriate N-acetlyglucosamine donor that is derivatized with a PEG moiety.
  • EPO that is expressed in an insect cell system, yeast or fungi, is remodeled by adding one or more terminal galactose-PEG residues.
  • the peptide is contacted with GnT-I and an appropriate N-acetylglucosamine donor that is derivatized with a PEG moiety.
  • the peptide is then contacted with galactosyltransferase and an appropriate galactose donor that is modified with a PEG moiety.
  • FIG. 35I EPO that is expressed in an insect cell system, yeast or fungi, is remodeled by adding one or more terminal galactose-PEG residues.
  • the peptide is contacted with GnT-I and an appropriate N-acetylglucosamine donor that is derivatized with a PEG moiety.
  • 35J EPO expressed in an insect cell system, yeast or fungi, is remodeled by the addition of one more terminal sialic acid-PEG residues.
  • the peptide is contacted with an appropriate N-acetylglucosamine donor and GnT-I.
  • the peptide is further contacted with galactosyltransferase and an appropriate galactose donor.
  • the peptide is then contacted with ST3Gal3 and an appropriate sialic acid donor that is derivatized with a PEG moiety.
  • FIG. 35K EPO expressed in an insect cell system, yeast or fungi, is remodeled by the addition of terminal sialic acid-PEG residues.
  • the peptide is contacted with an appropriate N-acetylglucosamine donor and one or more of GnT-I, GnT-II, and GnT-V.
  • the peptide is then contacted with galactosyltransferase and an appropriate galactose donor.
  • the peptide is flurther contacted with ST3Gal3 and an appropriate sialic acid donor that is derivatized with a PEG moiety.
  • EPO expressed in an insect cell system, yeast or fungi is remodeled by the addition of one or more terminal ⁇ 2,6-sialic acid-PEG residues.
  • the peptide is contacted with an appropriate N-acetylglucosamine donor and one or more of GnT-I, GnT-II, and GnT-V.
  • the peptide is further contacted with galactosyltransferase and an appropriate galactose donor.
  • the peptide is then contacted with ⁇ 2,6-sialyltransferase and an appropriately modified sialic acid donor.
  • EPO expressed in a mammalian cell system is remodeled by addition of one or more terminal sialic acid-PEG residues.
  • the peptide is contacted with a sialidase to remove terminal sialic acid residues.
  • the peptide is further contacted with a sialyltransferase and an appropriate sialic acid donor.
  • the peptide is further contacted with a sialyltransferase and an appropriate sialic acid donor that is derivatized with a PEG moiety.
  • EPO expressed in a mammalian cell system is remodeled by the addition of one or more terminal sialic acid-PEG residues.
  • the peptide is contacted with a sialyltransferase and an appropriate sialic acid donor that is derivatized with a PEG moiety.
  • FIG. 35N EPO expressed in a mammalian cell system is remodeled by the addition of one or more terminal sialic acid-PEG residues.
  • the peptide is contacted with a sialyltransferase and an appropriate sialic acid donor that is derivatized with a PEG moiety.
  • EPO expressed in a mammalian cell system is remodeled by the addition of one or more terminal ⁇ 2,8-sialic acid-PEG residues to primarily O-linked glycans.
  • the peptide is contacted with ⁇ 2,8-sialyltransferase and an appropriate sialic acid donor that is derivatized with a PEG moiety.
  • EPO expressed in a mammalian cell is remodeled by the addition of one or more terminal ⁇ 2,8-sialic acid-PEG residues to O-linked and N-linked glycans.
  • the peptide is contacted with ⁇ 2,8-sialyltransferase and an appropriate sialic acid donor that is derivatized with a PEG moiety.
  • EPO expressed in yeast or fungi is remodeled by the addition of one or more terminal sialic acid-PEG residues.
  • the peptide is contacted with mannosidases to remove terminal mannose residues.
  • the peptide is contacted with GnT-I and an appropriate N-acetylglucosamine donor.
  • the peptide is further contacted with galactosyltransferase and an appropriate galactose donor.
  • EPO expressed in yeast or fungi is remodeled by the addition of at least one terminal N-acetylglucosamine-PEG residues.
  • the peptide is contacted with mannosidases to remove terminal mannose residue.
  • the peptide is then contacted with GnT-I and an appropriate N-acetylglucosamine donor that is derivatized with a PEG moiety.
  • EPO expressed in yeast or fungi is remodeled by the addition of one or more terminal sialic acid-PEG residues.
  • the peptide is contacted with mannosidase-I to remove ⁇ 2 mannose residues.
  • the peptide is further contacted with GnT-I and an appropriate N-acetylglucosamine donor.
  • the peptide is then contacted with galactosyltransferase and an appropriate galacose donor.
  • the peptide is then contacted with a sialyltransferase and an appropriate sialic acid donor that is derivatized with a PEG moiety.
  • EPO expressed in yeast or fungi is remodeled by addition of one or more galactose-PEG residues.
  • the peptide is contacted with endo-H to trim back glycosyl groups.
  • the peptide is then contacted with galactosyltransferase and an appropriate galactose donor that is derivatized with a PEG moiety.
  • EPO expressed in yeast or fungi is remodeled by the addition of one or more terminal sialic acid-PEG residues.
  • the peptide is contacted with endo-H to trim back glycosyl groups.
  • the peptide is further contacted with galactosyltransferase and an appropriate galactose donor.
  • the peptide is then contacted with a sialyltransferase and an appropriate sialic acid donor that is derivatized with a PEG moiety.
  • FIG. 35V EPO expressed in yeast or fungi is remodeled by the addition of one or more terminal sialic acid-PEG residues.
  • the peptide is contacted with endo-H to trim back glycosyl groups.
  • the peptide is further contacted with galactosyltransfer
  • EPO expressed in an insect cell system is remodeled by the addition of terminal galactose-PEG residues.
  • the peptide is contacted with mannosidases to remove terminal mannose residues.
  • the peptide is then contacted with galactosyltransferase and an appropriate galactose donor that is derivatized with a PEG moiety.
  • a mutant EPO called “novel erythropoiesis-stimulating protein” or NESP, expressed in NSO murine myeloma cells is remodeled by capping appropriate terminal residues with a sialic acid donor that is modified with levulinic acid, adding a reactive ketone to the sialic acid donor.
  • mutant EPO i.e. NESP
  • NESP expressed in a mammalian cell system
  • PEG is added to the glycosyl residue on the glycan using a PEG-modified sialic acid and an ⁇ 2,8-sialyltransferase.
  • NESP expressed in a mammalian cell system is remodeled by the addition of terminal sialic acid residues.
  • the sialic acid is added to the glycosyl residue using a sialic acid donor and an ⁇ 2,8-sialyltransferase.
  • the invention provides methods for modifying granulocyte-macrophage colony-stimulating factor (GM-CSF), as shown in FIGS. 36A to 36 K.
  • GM-CSF granulocyte-macrophage colony-stimulating factor expressed in mammalian cells
  • FIG. 36B GM-CSF expressed in mammalian cells is first treated with sialidase to trim back the sialic acid residues, and then PEGylated using ST3Gal3 and a donor of PEG-sialic acid.
  • FIG. 36B GM-CSF expressed in mammalian cells is first treated with sialidase to trim back the sialic acid residues, and then PEGylated using ST3Gal3 and a donor of PEG-sialic acid.
  • GM-CSF expressed in mammalian cells is first treated with sialidase to trim back the sialic acid residues, then PEGylated using ST3Gal3 and a donor of PEG-sialic acid, and then is further sialylated using a sialic acid donor and ST3Gal1 and/or ST3Gal3.
  • FIG. 36D GM-CSF expressed in NSO cells is first treated with sialidase and ⁇ -galactosidase to trim back the glycosyl groups, then sialylated using a sialic acid donor and ST3Gal3, and is then PEGylated using ST3Gal1 and a donor of PEG-sialic acid.
  • FIG. 36C GM-CSF expressed in mammalian cells is first treated with sialidase to trim back the sialic acid residues, then PEGylated using ST3Gal3 and a donor of PEG-sialic acid, and then is further sialylated using
  • GM-CSF expressed in mammalian cells is first treated with sialidase to trim back sialic acid residues, then PEGylated using ST3Gal3 and a donor of PEG-sialic acid, and then is further sialylated using ST3Gal3 and a sialic acid donor.
  • FIG. 36F GM-CSF expressed in mammalian cells is modified by capping appropriate terminal residues with a sialic acid donor that is modified with levulinic acid, adding a reactive ketone to the sialic acid donor. After addition to a glycosyl residue of the peptide, the ketone is derivatized with a moiety such as a hydrazine- or amine-PEG.
  • GM-CSF expressed in mammalian cells is sialylated using a sialic acid donor and ⁇ 2,8-sialyltransferase.
  • GM-CSF expressed in insect cells is modified by addition of N-acetylglucosamine using a suitable donor and one or more of GnT-I, II, IV, and V, followed by addition of PEGylated galactose using a suitable donor and a galactosyltransferase.
  • FIG. 36G GM-CSF expressed in mammalian cells is sialylated using a sialic acid donor and ⁇ 2,8-sialyltransferase.
  • FIG. 36I GM-CSF expressed in insect cells is modified by addition of N-acetylglucosamine using a suitable donor and one or more of GnT-I, II, IV, and V, followed by addition of PEGylated galactose using a suitable donor and a galactos
  • yeast expressed GM-CSF is first treated with endoglycanase and/or mannosidase to trim back the glycosyl units, and subsequently PEGylated using a galactosyltransferase and a donor of PEG-galactose.
  • GM-CSF expressed in mammalian cells is first treated with sialidase to trim back sialic acid residues, and is subsequently sialylated using ST3Gal3 and a sialic acid donor.
  • the polypeptide is then contacted with ST3Gal1 and two reactive sialic acid residues connected via a linker, so that the polypeptide is attached to one reactive sialic acid via the linker and second sialic acid residue.
  • the polypeptide is further contacted with ST3Gal3 and transferrin, and thus becomes connected with transferrin.
  • the invention provides methods for modification of Interferon gamma (IFN ⁇ ).
  • FIGS. 37A to 37 N contain some examples.
  • IFN ⁇ expressed in a variety of mammalian cells is first treated with sialidase to trim back terminal sialic acid residues, and is subsequently PEGylated using ST3Gal3 and a donor of PEG-sialic acid.
  • FIG. 37C IFN ⁇ expressed in a mammalian system is first treated with sialidase to trim back terminal sialic acid residues.
  • the polypeptide is then PEGylated using ST3Gal3 and a donor of PEG-sialic acid, and is further sialylated with ST3Gal3 and a donor of sialic acid.
  • mammalian cell expressed IFN ⁇ is first treated with sialidase and ⁇ -galactosidase to trim back sialic acid and galactose residues.
  • the polypeptide is then galactosylated using a galactose donor and a galactosyltransferase. Then, IFN ⁇ is PEGylated using a donor of PEG-sialic acid and ST3Gal3.
  • FIG. 37D mammalian cell expressed IFN ⁇ is first treated with sialidase and ⁇ -galactosidase to trim back sialic acid and galactose residues.
  • the polypeptide is then galactosylated using a galactose donor and a galactosyl
  • IFN ⁇ that is expressed in a mammalian system is first treated with sialidase to trim back terminal sialic acid residues.
  • the polypeptide is then PEGylated using ST3Gal3 and a donor of PEG-sialic acid, and is further sialylated with ST3Gal3 and a sialic acid donor.
  • FIG. 37F describes another method for modifying IFN ⁇ expressed in a mammalian system.
  • the protein is modified by capping appropriate terminal residues with a sialic acid donor that is modified with levulinic acid, adding a reactive ketone to the sialic acid donor.
  • IFN ⁇ expressed in mammalian cells is remodeled by addition of sialic acid using a sialic acid donor and an ⁇ 2,8-sialyltransferase.
  • IFN ⁇ expressed in insect or fungal cells is modified by addition of N-acetylglucosamine using an appropriate donor and one or more of GnT-I, II, IV, and V.
  • FIG. 37J offers a method for modifying IFN ⁇ expressed in yeast.
  • the polypeptide is first treated with endoglycanase to trim back the saccharide chains, and then galactosylated using a galactose donor and a galactosyltransferase. Then, IFN ⁇ is PEGylated using a donor of PEGylated sialic acid and ST3Gal3.
  • FIG. 37J offers a method for modifying IFN ⁇ expressed in yeast.
  • the polypeptide is first treated with endoglycanase to trim back the saccharide chains, and then galactosylated using a galactose donor and a galactosyltransferase.
  • IFN ⁇ is PEGylated using a donor of PEGylated sialic acid and ST3Gal3.
  • IFN ⁇ produced by mammalian cells is modified as follows: the polypeptide is first contacted with ST3Gal3 and a donor of sialic acid that is derivatized with a reactive galactose via a linker, so that the polypeptide is attached to the reactive galactose via the linker and sialic acid residue. The polypeptide is then contacted with a galactosyltransferase and transferrin pre-treated with endoglycanase, and thus becomes connected with transferrin via the galactose residue. In the scheme illustrated by FIG.
  • FIG. 37L IFN ⁇ , which is expressed in a mammalian system, is modified via the action of ST3Gal3: PEGylated sialic acid is transferred from a suitable donor to IFN ⁇ .
  • FIG. 37M is an example of modifying IFN ⁇ expressed in insect or fungal cells, where PEGylation of the polypeptide is achieved by transferring PEGylated N-acetylglucosamine from a donor to IFN ⁇ using GnT-I and/or II.
  • FIG. 37N IFN ⁇ expressed in a mammalian system is remodeled with addition of PEGylated sialic acid using a suitable donor and an ⁇ 2,8-sialyltransferase.
  • the invention provides methods for modifying ⁇ 1 anti-trypsin ( ⁇ 1-protease inhibitor). Some such examples can be found in FIGS. 38A to 38 N.
  • ⁇ 1 anti-trypsin expressed in a variety of mammalian cells is first treated with sialidase to trim back sialic acid residues. PEGylated sialic acid residues are then added using an appropriate donor, such as CMP-SA-PEG, and a sialyltransferase, such as ST3Gal3.
  • FIG. 38C demonstrates another scheme of ⁇ 1 anti-trypsin modification.
  • ⁇ 1 anti-trypsin expressed in a mammalian system is first treated with sialidase to trim back sialic acid residues.
  • Sialic acid residues derivatized with PEG are then added using an appropriate donor and a sialyltransferase, such as ST3Gal3.
  • the molecule is further modified by the addition of sialic acid residues using a sialic acid donor and ST3Gal3.
  • mammalian cell expressed ⁇ 1 anti-trypsin is first treated with sialidase and ⁇ -galactosidase to trim back terminal sialic acid and ⁇ -linkage galactose residues.
  • the polypeptide is then galactosylated using galactosyltransferase and a suitable galactose donor. Further, sialic acid derivatized with PEG is added by the action of ST3Gal3 using a PEGylated sialic acid donor.
  • ⁇ 1 anti-trypsin expressed in a mammalian system first has the terminal sialic acid residues trimmed back using sialidase. PEG is then added to N-linked glycosyl residues via the action of ST3Gal3, which mediates the transfer of PEGylated sialic acid from a donor, such as CMP-SA-PEG, to ⁇ 1 anti-trypsin.
  • FIG. 38E illustrates another process through which ⁇ 1 anti-trypsin is remodeled.
  • ⁇ 1 anti-trypsin expressed in mammalian cells is modified by capping appropriate terminal residues with a sialic acid donor that is modified with levulinic acid, adding a reactive ketone to the sialic acid donor. After addition to a glycosyl residue of the peptide, the ketone is derivatized with a moiety such as a hydrazine- or amine-PEG.
  • FIG. 38F yet another method of ⁇ 1 anti-trypsin modification is disclosed.
  • ⁇ 1 anti-trypsin obtained from a mammalian expression system is remodeled with addition of sialic acid using a sialic acid donor and an ⁇ 2,8-sialyltransferase.
  • ⁇ 1 anti-trypsin is expressed in insect or yeast cells, and remodeled by the addition of terminal N-acetylglucosamine residues by way of contacting the polypeptide with UDP-N-acetylglucosamine and one or more of GnT-I, II, IV, or V. Then, the polypeptide is modified with PEG moieties using a donor of PEGylated galactose and a galactosyltransferase.
  • FIG. 38H ⁇ 1 anti-trypsin obtained from a mammalian expression system is remodeled with addition of sialic acid using a sialic acid donor and an ⁇ 2,8-sialyltransferase.
  • ⁇ 1 anti-trypsin expressed in yeast cells is treated first with endoglycanase to trim back glycosyl chains. It is then galactosylated with a galactosyltransferase and a galactose donor. Then, the polypeptide is PEGylated using ST3Gal3 and a donor of PEG-sialic acid.
  • ⁇ 1 anti-trypsin is expressed in a mammalian system.
  • the polypeptide is first contacted with ST3Gal3 and a donor of sialic acid that is derivatized with a reactive galactose via a linker, so that the polypeptide is attached to the reactive galactose via the linker and sialic acid residue.
  • the polypeptide is then contacted with a galactosyltransferase and transferrin pre-treated with endoglycanase, and thus becomes connected with transferrin via the galactose residue.
  • ⁇ 1 anti-trypsin expressed in yeast is first treated with endoglycanase to trim back its glycosyl groups.
  • ⁇ 1 anti-trypsin expressed in plant cells is treated with hexosaminidase, mannosidase, and xylosidase to trim back its glycosyl chains, and subsequently modified with N-acetylglucosamine derivatized with a PEG moiety, using N-acetylglucosamine transferase and a suitable donor.
  • ⁇ 1 anti-trypsin expressed in mammalian cells is modified by adding PEGylated sialic acid residues using ST3Gal3 and a donor of sialic acid derivatized with PEG.
  • the invention provides methods for modifying glucocerebrosidase ( ⁇ -glucosidase, CerezymeTM or CeredaseTM), as shown in FIGS. 39A to 39 K.
  • CerezymeTM expressed in a mammalian system is first treated with sialidase to trim back terminal sialic acid residues, and is then PEGylated using ST3Gal3 and a donor of PEG-sialic acid.
  • FIG. 39B CerezymeTM expressed in a mammalian system is first treated with sialidase to trim back terminal sialic acid residues, and is then PEGylated using ST3Gal3 and a donor of PEG-sialic acid.
  • CerezymeTM expressed in mammalian cells is first treated with sialidase to trim back the sialic acid residues, then has mannose-6-phosphate group attached using ST3Gal3 and a reactive sialic acid derivatized with mannose-6-phosphate, and then is sialylated using ST3Gal3 and a sialic acid donor.
  • NSO cell expressed CerezymeTM is first treated with sialidase and galactosidase to trim back the glycosyl groups, and is then galactosylated using a galactose donor and an ⁇ -galactosyltransferase.
  • mannose-6-phosphate moiety is added to the molecule using ST3Gal3 and a reactive sialic acid derivatized with mannose-6-phosphate.
  • CerezymeTM expressed in mammalian cells is first treated with sialidase to trim back the sialic acid residues, it is then PEGylated using ST3Gal3 and a donor of PEG-sialic acid, and is then sialylated using ST3Gal3 and a sialic acid donor.
  • FIG. 39D CerezymeTM expressed in mammalian cells is first treated with sialidase to trim back the sialic acid residues, it is then PEGylated using ST3Gal3 and a donor of PEG-sialic acid, and is then sialylated using ST3Gal3 and a sialic acid donor.
  • CerezymeTM expressed in mammalian cells is modified by capping appropriate terminal residues with a sialic acid donor that is modified with levulinic acid, adding a reactive ketone to the sialic acid donor. After addition to a glycosyl residue of the peptide, the ketone is derivatized with a moiety such as one or more mannose-6-phosphate groups.
  • CerezymeTM expressed in mammalian cells is sialylated using a sialic acid donor and ⁇ 2,8-sialyltransferase.
  • FIG. 39E CerezymeTM expressed in mammalian cells is modified by capping appropriate terminal residues with a sialic acid donor that is modified with levulinic acid, adding a reactive ketone to the sialic acid donor. After addition to a glycosyl residue of the peptide, the ketone is derivatized with a moiety such as one or more mannose-6-phosphate groups.
  • CerezymeTM expressed in insect cells first has N-acetylglucosamine added using a suitable donor and one or more of GnT-I, II, IV, and V, and then is PEGylated using a galactosyltransferase and a donor of PEG-galactose.
  • Cerezymem expressed in yeast is first treated with endoglycanase to trim back the glycosyl groups, then galactosylated using a galactose donor and a galactosyltransferase, and then PEGylated using ST3Gal3 and a donor of PEG-sialic acid.
  • FIG. 39H CerezymeTM expressed in insect cells first has N-acetylglucosamine added using a suitable donor and one or more of GnT-I, II, IV, and V, and then is PEGylated using a galactosyltransferase and a donor of PEG-gal
  • CerezymeTM expressed in mammalian cells is first contacted with ST3Gal3 and two reactive sialic acid residues connected via a linker, so that the polypeptide is attached to one reactive sialic acid via the linker and the second sialic acid residue.
  • the polypeptide is then contacted with ST3Gal3 and desialylated transferrin, and thus becomes connected with transferrin. Then, the polypeptide is sialylated using a sialic acid donor and ST3Gal3.
  • the invention provides methods for modifying Tissue-Type Plasminogen Activator (TPA) and its mutant.
  • TPA Tissue-Type Plasminogen Activator
  • FIGS. 40A to 40 W Several specific modification schemes are presented in FIGS. 40A to 40 W.
  • FIG. 40B illustrates one modification procedure: after TPA is expressed by mammalian cells, it is treated with one or more of mannosidase(s) and sialidase to trim back mannosyl and/or sialic acid residues. Terminal N-acetylglucosamine is then added by contacting the polypeptide with a suitable donor of N-acetylglucosamine and one or more of GnT-I, II, IV, and V.
  • TPA is further galactosylated using a galactose donor and a galactosyltransferase. Then, PEG is attached to the molecule by way of sialylation catalyzed by ST3Gal3 and using a donor of sialic acid derivatized with a PEG moiety. In FIG. 40C , TPA is expressed in insect or fungal cells.
  • the modification includes the steps of addition of N-acetylglucosamine using an appropriate donor of N-acetylglucosamine and GnT-I and/or II; galactosylation using a galactose donor and a galactosyltransferase; and attachment of PEG by way of sialylation using ST3Gal3 and a donor of sialic acid derivatized with PEG.
  • TPA is expressed in yeast and subsequently treated with endoglycanase to trim back the saccharide chains.
  • the polypeptide is further PEGylated via the action of a galactosyltransferase, which catalyzes the transfer of a PEG-galactose from a donor to TPA.
  • TPA is expressed in insect or yeast cells.
  • the polypeptide is then treated with ⁇ - and ⁇ -mannosidases to trim back terminal mannosyl residues.
  • PEG moieties are attached to the molecule via transfer of PEG-galactose from a suitable donor to TPA, which is mediated by a galactosyltransferase.
  • FIG. 40F provides a different method for modification of TPA obtained from an insect or yeast system: the polypeptide is remodeled by addition of N-acetylglucosamine using a donor of N-acetylglucosamine and GnT-I and/or II, followed by PEGylation using a galactosyltransferase and a donor of PEGylated galactose.
  • FIG. 40G offers another scheme for remodeling TPA expressed in insect or yeast cells. Terminal N-acetylglucosamine is added using a donor of N-acetylglucosamine and GnT-I and/or II.
  • a galactosidase that is modified to operate in a synthetic, rather than a hydrolytic manner, is utilized to add PEGylated galactose from a proper donor to the N-acetylglucosamine residues.
  • TPA expressed in a mammalian system is first treated with sialidase and galactosidase to trim back sialic acid and galactose residues.
  • the polypeptide is further modified by capping appropriate terminal residues with a sialic acid donor that is modified with levulinic acid, adding a reactive ketone to the sialic acid donor.
  • TPA which is expressed in a mammalian system, is remodeled following this scheme: first, the polypeptide is treated with ⁇ - and ⁇ -mannosidases to trim back the terminal mannosyl residues; sialic acid residues are then attached to terminal galactosyl residues using a sialic acid donor and ST3Gal3; further, TPA is PEGylated via the transfer of PEGylated galactose from a donor to a N-acetylglucosaminyl residue catalyzed by a galactosyltransferase.
  • TPA is expressed in a plant system.
  • the modification procedure in this example is as follows: TPA is first treated with hexosaminidase, mannosidase, and xylosidase to trim back its glycosyl groups; PEGylated N-acetylglucosamine is then added to TPA using a proper donor and N-acetylglucosamine transferase.
  • a TPA mutant (TNK TPA), expressed in mammalian cells, is remodeled.
  • Terminal sialic acid residues are first trimed back using sialidase; ST3Gal3 is then used to transfer PEGylated sialic acid from a donor to TNK TPA, such that the polypeptide is PEGylated.
  • TNK TPA expressed in a mammalian system is first treated with sialidase to trim back terminal sialic acid residues.
  • the protein is then PEGylated using CMP-SA-PEG as a donor and ST3Gal3, and further sialylated using a sialic acid donor and ST3Gal3.
  • CMP-SA-PEG CMP-SA-PEG
  • NSO cell expressed TNK TPA is first treated with sialidase and a-galactosidase to trim back terminal sialic acid and galactose residues.
  • TNK TPA is then galactosylated using a galactose donor and a galactosyltransferase.
  • the last step in this remodeling scheme is transfer of sialic acid derivatized with PEG moiety from a donor to TNK TPA using a sialyltransferase such as ST3Gal3.
  • TNK TPA is expressed in a mammalian system and is first treated with sialidase to trim back terminal sialic acid residues.
  • TNK TPA expressed in a mammalian system is modified by capping appropriate terminal residues with a sialic acid donor that is modified with levulinic acid, adding a reactive ketone to the sialic acid donor. After addition to a glycosyl residue of the peptide, the ketone is derivatized with a moiety such as a hydrazine- or amine- PEG.
  • a sialic acid donor that is modified with levulinic acid
  • ketone is derivatized with a moiety such as a hydrazine- or amine- PEG.
  • TNK TPA expressed in mammalian cells is modified via a different method: the polypeptide is remodeled with addition of sialic acid using a sialic acid donor and ⁇ 2,8-sialyltransferase.
  • TNK TPA expressed in insect cells is remodeled by addition of N-acetylglucosamine using an appropriate donor and one or more of GnT-I, II, IV, and V.
  • the protein is further modified by addition of PEG moieties using a donor of PEGylated galactose and a galactosyltransferase.
  • TNK TPA is expressed in yeast.
  • the polypeptide is first treated with endoglycanase to trim back its glycosyl chains and then PEGylated using a galactose donor derivatized with PEG and a galactosyltransferase.
  • TNK TPA is produced in a mammalian system.
  • the polypeptide is first contacted with ST3Gal3 and a donor of sialic acid that is derivatized with a reactive galactose via a linker, so that the polypeptide is attached to the reactive galactose via the linker and sialic acid residue.
  • the polypeptide is then contacted with a galactosyltransferase and anti-TNF IG chimera produced in CHO, and thus becomes connected with the chimera via the galactose residue.
  • FIGS. 41A to 41 G provide some examples.
  • FIG. 41B provides a two-step modification scheme: IL-2 produced by mammalian cells is first treated with sialidase to trim back its terminal sialic acid residues, and is then PEGylated using ST3Gal3 and a donor of PEGylated sialic acid.
  • FIG. 41C insect cell expressed IL-2 is modified first by galactosylation using a galactose donor and a galactosyltransferase. Subsequently, IL-2 is PEGylated using ST3Gal3 and a donor of PEGylated sialic acid.
  • FIG. 41B provides a two-step modification scheme: IL-2 produced by mammalian cells is first treated with sialidase to trim back its terminal sialic acid residues, and is then PEGylated using ST3Gal3 and a donor of PEGylated sialic acid.
  • FIG. 41C insect cell expressed IL-2
  • IL-2 expressed in bacteria is modified with N-acetylgalactosamine using a proper donor and N-acetylgalactosamine transferase, followed by a step of PEGylation with a PEG-sialic acid donor and a sialyltransferase.
  • FIG. 41E offers another scheme of modifying IL-2 produced by a mammalian system. The polypeptide is modified by capping appropriate terminal residues with a sialic acid donor that is modified with levulinic acid, adding a reactive ketone to the sialic acid donor.
  • FIG. 41F illustrates an example of remodeling IL-2 expressed by E. coli.
  • the polypeptide is PEGylated using a reactive N-acetylgalactosamine complex derivatized with a PEG group and an enzyme that is modified so that it functions as a synthetic enzyme rather than a hydrolytic one.
  • FIG. 41G IL-2 expressed by bacteria is modified by addition of PEGylated N-acetylgalactosamine using a proper donor and N-acetylgalactosamine transferase.
  • the invention provides methods for modifying Factor VIII, as shown in FIGS. 42A to 42 N.
  • Factor VIII expressed in mammalian cells is first treated with sialidase to trim back the sialic acid residues, and is then PEGylated using ST3Gal3 and a donor of PEG-sialic acid.
  • FIG. 42C Factor VIII expressed in mammalian cells is first treated with sialidase to trim back the sialic acid residues, then PEGylated using ST3Gal3 and a proper donor, and is then further sialylated using ST3 Gal1 and a sialic acid donor.
  • mammalian cell produced Factor VIII is modified by the single step of PEGylation, using ST3Gal3 and a donor of PEGylated sialic acid.
  • FIG. 42F offers another example of modification of Factor VIII that is expressed by mammalian cells. The protein is PEGylated using ST3Gal1 and a donor of PEGylated sialic acid.
  • mammalian cell expressed Factor VII is remodeled following another scheme: it is PEGylated using a 2,8-sialyltransferase and a donor of PEG-sialic acid.
  • FIG. 42E mammalian cell produced Factor VIII is modified by the single step of PEGylation, using ST3Gal3 and a donor of PEGylated sialic acid.
  • FIG. 42F offers another example of modification of Factor VIII that is expressed by mammalian cells. The protein is PEGylated using ST3Gal1 and a donor of PEGylated sialic acid.
  • Factor VIII produce by mammalian cells is modified by capping appropriate terminal residues with a sialic acid donor that is modified with levulinic acid, adding a reactive ketone to the sialic acid donor. After addition to a glycosyl residue of the peptide, the ketone is derivatized with a moiety such as a hydrazine- or amine-PEG.
  • FIG. 42J Factor VIII expressed by mammalian cells is first treated with Endo-H to trim back glycosyl groups. It is then PEGylated using a galactosyltransferase and a donor of PEG-galactose.
  • FIG. 42I Factor VIII produce by mammalian cells is modified by capping appropriate terminal residues with a sialic acid donor that is modified with levulinic acid, adding a reactive ketone to the sialic acid donor. After addition to a glycosyl residue of the peptide, the ketone is derivatized with a
  • Factor VIII expressed in a mammalian system is first sialylated using ST3Gal3 and a sialic acid donor, then treated with Endo-H to trim back the glycosyl groups, and then PEGylated with a galactosyltransferase and a donor of PEG-galactose.
  • ST3Gal3 and a sialic acid donor then treated with Endo-H to trim back the glycosyl groups
  • PEGylated with a galactosyltransferase and a donor of PEG-galactose is first sialylated using ST3Gal3 and a sialic acid donor, then treated with Endo-H to trim back the glycosyl groups, and then PEGylated with a galactosyltransferase and a donor of PEG-galactose.
  • Factor VIII expressed in a mammalian system is first treated with mannosidases to trim back terminal mannosyl residues, then has an N-acetylglucosamine group added using a suitable donor and GnT-I and/or II, and then is PEGylated using a galactosyltransferase and a donor of PEG-galactose.
  • FIG. 42M Factor VIII expressed in mammalian cells is first treated with mannosidases to trim back mannosyl units, then has N-acetylglucosamine group added using N-acetylglucosamine transferase and a suitable donor.
  • Factor VIII is produced by mammalian cells and modified as follows: it is first treated with mannosidases to trim back the terminal mannosyl groups. A PEGylated N-acetylglucosamine group is then added using GnT-I and a suitable donor of PEGylated N-acetylglucosamine.
  • the invention provides methods for modifying urokinase, as shown in FIGS. 43A to 43 M.
  • urokinase expressed in mammalian cells is first treated with sialidase to trim back sialic acid residues, and is then PEGylated using ST3Gal3 and a donor of PEGylated sialic acid.
  • FIG. 43C urokinase expressed in mammalian cells is first treated with sialidase to trim back sialic acid residues, then PEGylated using ST3Gal3 and a donor of PEGylated sialic acid, and then sialylated using ST3Gal3 and a sialic acid donor.
  • urokinase expressed in a mammalian system is first treated with sialidase and galactosidase to trim back glycosyl chains, then galactosylated using a galactose donor and an a-galactosyltransferase, and then PEGylated using ST3Gal3 or sialyltransferase and a donor of PEG-sialic acid.
  • urokinase expressed in mammalian cells is first treated with sialidase to trim back sialic acid residues, then PEGylated using ST3Gal3 and a donor of PEG-sialic acid, and then further sialylated using ST3Gal3 and a sialic acid donor.
  • urokinase expressed in mammalian cells is modified by capping appropriate terminal residues with a sialic acid donor that is modified with levulinic acid, adding a reactive ketone to the sialic acid donor.
  • urokinase expressed in mammalian cells is sialylated using a sialic acid donor and ⁇ 2,8-sialyltransferase.
  • urokinase expressed in insect cells is modified in the following steps: first, N-acetylglucosamine is added to the polypeptide using a suitable donor of N-acetylglucosamine and one or more of GnT-I, II, IV, and V; then PEGylated galactose is added, using a galactosyltransferase and a donor of PEG-galactose.
  • N-acetylglucosamine is added to the polypeptide using a suitable donor of N-acetylglucosamine and one or more of GnT-I, II, IV, and V
  • PEGylated galactose is added, using a galactosyltransferase and a donor of PEG-galactose.
  • urokinase expressed in yeast is first treated with endoglycanase to trim back glycosyl groups, then galactosylated using a galactose donor and a galactosyltransferase, and then PEGylated using ST3Gal3 and a donor of PEG-sialic acid.
  • urokinase expressed in mammalian cells is first contacted with ST3Gal3 and two reactive sialic acid residues that are connected via a linker, so that the polypeptide is attached to one reactive sialic acid via the linker and second sialic acid residue.
  • the polypeptide is then contacted with ST3Gal1 and desialylated urokinase produced in mammalian cells, and thus becomes connected with a second molecule of urokinase. Then, the whole molecule is further sialylated using a sialic donor and ST3Gal1 and/or ST3Gal3.
  • isolated urokinase is first treated with sulfohydrolase to remove sulfate groups, and is then PEGylated using a sialyltransferase and a donor of PEG-sialic acid.
  • isolated urokinase is first treated with sulfohydrolase and hexosaminidase to remove sulfate groups and hexosamine groups, and then PEGylated using a galactosyltransferase and a donor of PEG-galactose.
  • the invention provides methods for modifying DNase I, as shown in FIGS. 44A to 44 J.
  • DNase I is expressed in a mammalian system and modified in the following steps: first, the protein is treated with sialidase to trim back the sialic acid residues; then the protein is PEGylated with ST3Gal3 using a donor of PEG-sialic acid.
  • FIG. 44C DNase I expressed in mammalian cells is first treated with sialidase to trim back the sialic acid residues, then PEGylated with ST3Gal3 using a PEG-sialic acid donor, and is then sialylated using ST3Gal3 and a sialic acid donor.
  • DNase I expressed in a mammalian system is first exposed to sialidase and galactosidase to trim back the glycosyl groups, then galactosylated using a galactose donor and an a-galactosyltransferase, and then PEGylated using ST3Gal3 or sialyltransferase and a donor of PEG-sialic acid.
  • sialidase and galactosidase to trim back the glycosyl groups
  • galactosylated using a galactose donor and an a-galactosyltransferase is then PEGylated using ST3Gal3 or sialyltransferase and a donor of PEG-sialic acid.
US10/552,896 2003-04-09 2004-04-09 Glycopegylation methods and proteins/peptides produced by the methods Abandoned US20070026485A1 (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
US10/552,896 US20070026485A1 (en) 2003-04-09 2004-04-09 Glycopegylation methods and proteins/peptides produced by the methods
US12/496,595 US8063015B2 (en) 2003-04-09 2009-07-01 Glycopegylation methods and proteins/peptides produced by the methods
US13/246,512 US8853161B2 (en) 2003-04-09 2011-09-27 Glycopegylation methods and proteins/peptides produced by the methods
US13/622,177 US20130137157A1 (en) 2003-04-09 2012-09-18 Glycopegylated factor vii and factor viia
US13/897,529 US20130344050A1 (en) 2003-04-09 2013-05-20 Glycopegylated Factor IX
US14/052,442 US8791070B2 (en) 2003-04-09 2013-10-11 Glycopegylated factor IX
US14/246,519 US20140294762A1 (en) 2003-04-09 2014-04-07 Glycopegylation methods and proteins/peptides produced by the methods
US14/721,761 US20150343080A1 (en) 2003-04-09 2015-05-26 Glycopegylation methods and proteins/peptides produced by the methods
US15/225,819 US20170007712A1 (en) 2003-04-09 2016-08-01 Glycopegylation methods and proteins/peptides produced by the methods

Applications Claiming Priority (28)

Application Number Priority Date Filing Date Title
US10/411,043 US7439043B2 (en) 2001-10-10 2003-04-09 Galactosyl nucleotide sugars
US10/411,026 2003-04-09
US10/411,037 2003-04-09
US10/411,012 US7265084B2 (en) 2001-10-10 2003-04-09 Glycopegylation methods and proteins/peptides produced by the methods
US10/410,980 2003-04-09
US10/410,897 2003-04-09
US10/411,043 2003-04-09
US10/410,962 US7173003B2 (en) 2001-10-10 2003-04-09 Granulocyte colony stimulating factor: remodeling and glycoconjugation of G-CSF
US10/410,962 2003-04-09
US10/410,930 2003-04-09
US10/411,049 US7297511B2 (en) 2001-10-10 2003-04-09 Interferon alpha: remodeling and glycoconjugation of interferon alpha
US10/411,044 2003-04-09
US10/411,026 US7795210B2 (en) 2001-10-10 2003-04-09 Protein remodeling methods and proteins/peptides produced by the methods
US10/410,980 US7399613B2 (en) 2001-10-10 2003-04-09 Sialic acid nucleotide sugars
US10/410,913 US7265085B2 (en) 2001-10-10 2003-04-09 Glycoconjugation methods and proteins/peptides produced by the methods
US10/410,997 US7157277B2 (en) 2001-11-28 2003-04-09 Factor VIII remodeling and glycoconjugation of Factor VIII
US10/411,049 2003-04-09
US10/410,913 2003-04-09
US10/410,897 US7179617B2 (en) 2001-10-10 2003-04-09 Factor IX: remolding and glycoconjugation of Factor IX
US10/410,930 US7226903B2 (en) 2001-10-10 2003-04-09 Interferon beta: remodeling and glycoconjugation of interferon beta
US10/411,037 US7125843B2 (en) 2001-10-19 2003-04-09 Glycoconjugates including more than one peptide
US10/410,945 US7214660B2 (en) 2001-10-10 2003-04-09 Erythropoietin: remodeling and glycoconjugation of erythropoietin
US10/411,012 2003-04-09
US10/410,997 2003-04-09
US10/410,945 2003-04-09
US10/411,044 US8008252B2 (en) 2001-10-10 2003-04-09 Factor VII: remodeling and glycoconjugation of Factor VII
PCT/US2004/011494 WO2004099231A2 (en) 2003-04-09 2004-04-09 Glycopegylation methods and proteins/peptides produced by the methods
US10/552,896 US20070026485A1 (en) 2003-04-09 2004-04-09 Glycopegylation methods and proteins/peptides produced by the methods

Related Parent Applications (3)

Application Number Title Priority Date Filing Date
US10/411,012 Continuation US7265084B2 (en) 2001-10-10 2003-04-09 Glycopegylation methods and proteins/peptides produced by the methods
US10/411,026 Continuation US7795210B2 (en) 2001-10-10 2003-04-09 Protein remodeling methods and proteins/peptides produced by the methods
PCT/US2004/011494 A-371-Of-International WO2004099231A2 (en) 2001-10-10 2004-04-09 Glycopegylation methods and proteins/peptides produced by the methods

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/496,595 Continuation US8063015B2 (en) 2003-04-09 2009-07-01 Glycopegylation methods and proteins/peptides produced by the methods

Publications (1)

Publication Number Publication Date
US20070026485A1 true US20070026485A1 (en) 2007-02-01

Family

ID=33437331

Family Applications (6)

Application Number Title Priority Date Filing Date
US10/552,896 Abandoned US20070026485A1 (en) 2003-04-09 2004-04-09 Glycopegylation methods and proteins/peptides produced by the methods
US12/496,595 Expired - Fee Related US8063015B2 (en) 2003-04-09 2009-07-01 Glycopegylation methods and proteins/peptides produced by the methods
US13/246,512 Expired - Lifetime US8853161B2 (en) 2003-04-09 2011-09-27 Glycopegylation methods and proteins/peptides produced by the methods
US14/246,519 Abandoned US20140294762A1 (en) 2003-04-09 2014-04-07 Glycopegylation methods and proteins/peptides produced by the methods
US14/721,761 Abandoned US20150343080A1 (en) 2003-04-09 2015-05-26 Glycopegylation methods and proteins/peptides produced by the methods
US15/225,819 Abandoned US20170007712A1 (en) 2003-04-09 2016-08-01 Glycopegylation methods and proteins/peptides produced by the methods

Family Applications After (5)

Application Number Title Priority Date Filing Date
US12/496,595 Expired - Fee Related US8063015B2 (en) 2003-04-09 2009-07-01 Glycopegylation methods and proteins/peptides produced by the methods
US13/246,512 Expired - Lifetime US8853161B2 (en) 2003-04-09 2011-09-27 Glycopegylation methods and proteins/peptides produced by the methods
US14/246,519 Abandoned US20140294762A1 (en) 2003-04-09 2014-04-07 Glycopegylation methods and proteins/peptides produced by the methods
US14/721,761 Abandoned US20150343080A1 (en) 2003-04-09 2015-05-26 Glycopegylation methods and proteins/peptides produced by the methods
US15/225,819 Abandoned US20170007712A1 (en) 2003-04-09 2016-08-01 Glycopegylation methods and proteins/peptides produced by the methods

Country Status (14)

Country Link
US (6) US20070026485A1 (de)
EP (3) EP1615945B1 (de)
JP (1) JP4674702B2 (de)
AU (1) AU2004236174B2 (de)
BE (1) BE2014C003I2 (de)
CA (1) CA2522345A1 (de)
CY (1) CY1114626T1 (de)
IL (1) IL171109A (de)
LU (1) LU92358I2 (de)
MX (1) MXPA05010773A (de)
NZ (1) NZ542586A (de)
PL (1) PL1615945T3 (de)
SG (1) SG155777A1 (de)
WO (1) WO2004099231A2 (de)

Cited By (48)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060111279A1 (en) * 2003-11-24 2006-05-25 Neose Technologies, Inc. Glycopegylated erythropoietin
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
US20080015142A1 (en) * 2003-12-03 2008-01-17 Defrees Shawn Glycopegylated Follicle Stimulating Hormone
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)
US20080187955A1 (en) * 2001-10-10 2008-08-07 Neose Technologies, Inc. Erythropoietin: remodeling and glycoconjugation of erythropoietin
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
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
WO2008151258A3 (en) * 2007-06-04 2009-02-12 Neose Technologies Inc O-linked glycosylation using n-acetylglucosaminyl transferases
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
WO2009089154A3 (en) * 2008-01-03 2010-01-14 Cornell Research Foundation, Inc. Glycosylated protein expression in prokaryotes
US20100048456A1 (en) * 2003-04-09 2010-02-25 Novo Nordisk A/S Glycopegylation methods and proteins/peptides produced by the methods
US20100120666A1 (en) * 2007-04-03 2010-05-13 Biogenerix Ag Methods of treatment using glycopegylated g-csf
US20100174059A1 (en) * 2007-06-12 2010-07-08 Novo Nordisk A/S Process for the production of nucleotide sugars
US20100286067A1 (en) * 2008-01-08 2010-11-11 Biogenerix Ag Glycoconjugation of polypeptides using oligosaccharyltransferases
US20100330645A1 (en) * 2005-08-19 2010-12-30 Novo Nordisk A/S One pot desialylation and glycopegylation of therapeutic peptides
US20110003744A1 (en) * 2005-05-25 2011-01-06 Novo Nordisk A/S Glycopegylated Erythropoietin Formulations
US8008252B2 (en) 2001-10-10 2011-08-30 Novo Nordisk A/S Factor VII: remodeling and glycoconjugation of Factor VII
US8076292B2 (en) 2001-10-10 2011-12-13 Novo Nordisk A/S Factor VIII: remodeling and glycoconjugation of factor VIII
WO2012016131A1 (en) 2010-07-30 2012-02-02 Baxter International Inc. Nucleophilic catalysts for oxime linkage
WO2012087838A1 (en) 2010-12-22 2012-06-28 Baxter International Inc. Materials and methods for conjugating a water soluble fatty acid derivative to a protein
US8247381B2 (en) 2003-03-14 2012-08-21 Biogenerix Ag Branched water-soluble polymers and their conjugates
WO2012166622A1 (en) 2011-05-27 2012-12-06 Baxter International Inc. Therapeutic proteins with increased half-life and methods of preparing same
US8404809B2 (en) 2005-05-25 2013-03-26 Novo Nordisk A/S Glycopegylated factor IX
WO2013173557A1 (en) 2012-05-16 2013-11-21 Baxter International Inc. Nucleophilic catalysts for oxime linkage and use of nmr analyses of the same
US8632770B2 (en) 2003-12-03 2014-01-21 Novo Nordisk A/S Glycopegylated factor IX
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
US8916360B2 (en) 2003-11-24 2014-12-23 Novo Nordisk A/S Glycopegylated erythropoietin
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
US20150087811A1 (en) * 2006-03-27 2015-03-26 University Of Maryland, Baltimore Glycoprotein synthesis and remodeling by enzymatic transglycosylation
US9150848B2 (en) 2008-02-27 2015-10-06 Novo Nordisk A/S Conjugated factor VIII molecules
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
US10322192B2 (en) 2016-03-02 2019-06-18 Eisai R&D Management Co., Ltd. Eribulin-based antibody-drug conjugates and methods of use
US11291680B2 (en) 2016-12-15 2022-04-05 Société des Produits Nestlé S.A. Compositions and methods that modulate white blood cells or neutrophils in a companion animal
US11530432B2 (en) 2018-03-19 2022-12-20 Northwestern University Compositions and methods for rapid in vitro synthesis of bioconjugate vaccines in vitro via production and N-glycosylation of protein carriers in detoxified prokaryotic cell lysates
US11725224B2 (en) 2018-04-16 2023-08-15 Northwestern University Methods for co-activating in vitro non-standard amino acid (nsAA) incorporation and glycosylation in crude cell lysates
US11898187B2 (en) 2017-08-15 2024-02-13 Northwestern University Protein glycosylation sites by rapid expression and characterization of N-glycosyltransferases

Families Citing this family (61)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7795210B2 (en) 2001-10-10 2010-09-14 Novo Nordisk A/S Protein remodeling methods and proteins/peptides produced by the methods
US7696163B2 (en) 2001-10-10 2010-04-13 Novo Nordisk A/S Erythropoietin: remodeling and glycoconjugation of erythropoietin
EP2572732A1 (de) 2003-02-26 2013-03-27 Nektar Therapeutics Konjugate mit Polymer-Faktor-VIII-Anteil
US7691603B2 (en) 2003-04-09 2010-04-06 Novo Nordisk A/S Intracellular formation of peptide conjugates
US7842661B2 (en) 2003-11-24 2010-11-30 Novo Nordisk A/S Glycopegylated erythropoietin formulations
US7956032B2 (en) 2003-12-03 2011-06-07 Novo Nordisk A/S Glycopegylated granulocyte colony stimulating factor
AU2014280936B2 (en) * 2004-06-30 2016-12-15 Nektar Therapeutics Polymer-factor ix moiety conjugates
EP1781700B1 (de) 2004-08-03 2014-03-19 TransTech Pharma, LLC Rage-fusionsproteine und anwendungsverfahren
US20100009902A1 (en) * 2005-01-06 2010-01-14 Neose Technologies, Inc. Glycoconjugation Using Saccharyl Fragments
AU2005325768A1 (en) * 2005-01-25 2006-08-03 Apollo Life Sciences Limited Molecules and chimeric molecules thereof
CA2597795A1 (en) * 2005-02-15 2006-08-24 Apollo Life Sciences Limited A molecule and chimeric molecules thereof
CN102719508A (zh) * 2005-08-19 2012-10-10 诺和诺德公司 糖基聚乙二醇化的因子vii和因子viia
WO2007081031A1 (ja) * 2006-01-16 2007-07-19 Hokkaido University 糖転移酵素阻害剤
US20150038413A1 (en) 2006-02-03 2015-02-05 Opko Biologics Ltd. Long-acting polypeptides and methods of producing and administering same
US9249407B2 (en) 2006-02-03 2016-02-02 Opko Biologics Ltd. Long-acting coagulation factors and methods of producing same
US20140113860A1 (en) 2006-02-03 2014-04-24 Prolor Biotech Ltd. Long-acting polypeptides and methods of producing and administering same
US10221228B2 (en) 2006-02-03 2019-03-05 Opko Biologics Ltd. Long-acting polypeptides and methods of producing and administering same
US8946155B2 (en) 2006-02-03 2015-02-03 Opko Biologics Ltd. Long-acting polypeptides and methods of producing and administering same
US9458444B2 (en) 2006-02-03 2016-10-04 Opko Biologics Ltd. Long-acting coagulation factors and methods of producing same
US10351615B2 (en) 2006-02-03 2019-07-16 Opko Biologics Ltd. Methods of treatment with long-acting growth hormone
US8048849B2 (en) 2006-02-03 2011-11-01 Modigene, Inc. Long-acting polypeptides and methods of producing same
US7998486B2 (en) * 2006-10-25 2011-08-16 Newlink Genetics Corporation Enhanced immunogenicity of tumor associated antigens by addition of alphaGal epitopes
US20100113294A1 (en) * 2007-04-16 2010-05-06 Momenta Pharmaceuticals, Inc. Defined glycoprotein products and related methods
US20110137011A1 (en) * 2008-04-21 2011-06-09 Novo Nordisk A/S Hyperglycosylated human coagulation factor ix
CA2723197C (en) 2008-05-02 2017-09-19 Seattle Genetics, Inc. Methods and compositions for making antibodies and antibody derivatives with reduced core fucosylation
AU2009202778B2 (en) * 2008-07-11 2014-05-08 Commonwealth Of Australia As Represented By And Acting Through The Department Of The Environment, Water, Heritage And The Arts Improved baiting method and composition
EP2421892A1 (de) 2009-04-20 2012-02-29 Pfizer Inc. Steuerung der proteinglycosylierung und damit in zusammenhang stehende zusammensetzungen und verfahren
US9663778B2 (en) 2009-07-09 2017-05-30 OPKO Biologies Ltd. Long-acting coagulation factors and methods of producing same
TWI604850B (zh) * 2009-10-05 2017-11-11 菲瑞茵國際中心股份有限公司 藥學製劑
US8697844B2 (en) 2009-11-24 2014-04-15 Novo Nordisk A/S Method of purifying pegylated proteins
MX2012011648A (es) 2010-04-07 2012-11-29 Momenta Pharmaceuticals Inc Glicanos de alta manosa.
US20130123126A1 (en) * 2010-04-07 2013-05-16 Momenta Pharmaceuticals, Inc. Selection and use of host cells for production of glycoproteins
WO2012001087A1 (en) 2010-06-30 2012-01-05 Novo Nordisk A/S Antibodies that are capable of specifically binding tissue factor pathway inhibitor
MX356671B (es) 2010-08-05 2018-06-08 Seattle Genetics Inc Métodos para inhibir fucosilación de proteína in vivo utilizando análogos de fucosa.
EP2491952A1 (de) * 2011-02-22 2012-08-29 Cepep III AB System zur Frachtlieferung in Zellen
AU2012222366A1 (en) 2011-03-02 2013-08-01 Novo Nordisk A/S Coagulation factor-targeting to TLT-1 on activated platelets
US9170249B2 (en) 2011-03-12 2015-10-27 Momenta Pharmaceuticals, Inc. N-acetylhexosamine-containing N-glycans in glycoprotein products
US9765158B2 (en) 2011-05-31 2017-09-19 Probiogen Ag Methods for preparation of fucose-linked site specific conjugates of proteins with toxins, adjuvants, detection labels and pharmacokinetic half life extenders
PE20142405A1 (es) 2012-04-19 2015-01-25 Opko Biolog Ltd Variantes de oxintomodulina de accion prolongada y metodos de produccion de las mismas
US9695244B2 (en) 2012-06-01 2017-07-04 Momenta Pharmaceuticals, Inc. Methods related to denosumab
CA2882725C (en) 2012-08-23 2021-03-09 Regents Of The University Of Minnesota Treatment of sickle cell disease and inflammatory conditions
US20150273027A1 (en) 2012-10-10 2015-10-01 Novo Nordisk Healthcare Ag Liquid pharmaceutical composition of factor vii polypeptide
US9808534B2 (en) 2012-11-20 2017-11-07 Opko Biologics Ltd. Method of increasing the hydrodynamic volume of polypeptides by attaching to gonadotrophin carboxy terminal peptides
EP2983701A2 (de) * 2013-03-11 2016-02-17 Genzyme Corporation Ortsspezifische antikörper-arzneimittel-konjugation durch glycoengineering
EP2855533A4 (de) 2013-03-15 2015-11-25 Momenta Pharmaceuticals Inc Verfahren in zusammenhang mit ctla4-fc-fusionsproteinen
ES2926773T3 (es) 2013-03-15 2022-10-28 Novo Nordisk As Anticuerpos capaces de unirse específicamente a dos epítopos en el inhibidor de la ruta del factor tisular
WO2014186310A1 (en) 2013-05-13 2014-11-20 Momenta Pharmaceuticals, Inc. Methods for the treatment of neurodegeneration
WO2015057622A1 (en) 2013-10-16 2015-04-23 Momenta Pharmaceuticals, Inc. Sialylated glycoproteins
US20150158926A1 (en) 2013-10-21 2015-06-11 Opko Biologics, Ltd. Long-acting polypeptides and methods of producing and administering same
SG10201808158UA (en) * 2014-03-19 2018-10-30 Genzyme Corp Site-specific glycoengineering of targeting moieties
JP6355842B2 (ja) * 2014-07-10 2018-07-11 アカデミア シニカAcademia Sinica マルチドラッグ送達システム及びその使用
KR102473014B1 (ko) 2015-06-19 2022-11-30 옵코 바이오로직스 리미티드 장기-작용성 응고 인자 및 그의 제조 방법
KR20180050340A (ko) 2015-09-03 2018-05-14 에그리메티스, 엘엘씨 살곤충제로서의 스피노신 유도체
CN110191888A (zh) 2017-01-13 2019-08-30 阿格里麦蒂斯有限责任公司 氮丙啶多杀菌素衍生物和制备方法
EP3570871B1 (de) 2017-03-20 2020-11-18 H. Hoffnabb-La Roche Ag Verfahren für in-vitro-glyco-engineering eines erythropoiesestimulierenden proteins
SG11202000167SA (en) 2017-07-11 2020-02-27 Synthorx Inc Incorporation of unnatural nucleotides and methods thereof
CA3071016A1 (en) 2017-08-03 2019-02-07 Synthorx, Inc. Cytokine conjugates for the treatment of autoimmune diseases
MA54952A (fr) 2019-02-06 2022-05-11 Synthorx Inc Conjugués d'il-2 et méthodes d'utilisation de ceux-ci
WO2021009669A2 (en) * 2019-07-14 2021-01-21 Kashiv Biosciences, Llc A process for separation and quantitation of proteins using capillary electrophoresis
EP4301775A1 (de) * 2021-03-04 2024-01-10 Children's Hospital Medical Center Modulation der antikörperfunktion durch sialinsäuremodifikation
WO2023114812A1 (en) * 2021-12-15 2023-06-22 BioLegend, Inc. Compositions and methods for enhanced sample multiplexing

Citations (71)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4055635A (en) * 1973-07-05 1977-10-25 Beecham Group Limited Fibrinolytic compositions
US4088538A (en) * 1975-05-30 1978-05-09 Battelle Memorial Institute Reversibly precipitable immobilized enzyme complex and a method for its use
US4179337A (en) * 1973-07-20 1979-12-18 Davis Frank F Non-immunogenic polypeptides
US4385260A (en) * 1975-09-09 1983-05-24 Beckman Instruments, Inc. Bargraph display
US4412989A (en) * 1981-06-10 1983-11-01 Ajinomoto Company Incorporated Oxygen carrier
US4414147A (en) * 1981-04-17 1983-11-08 Massachusetts Institute Of Technology Methods of decreasing the hydrophobicity of fibroblast and other interferons
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
US4879236A (en) * 1984-05-16 1989-11-07 The Texas A&M University System Method for producing a recombinant baculovirus expression vector
US4925796A (en) * 1986-03-07 1990-05-15 Massachusetts Institute Of Technology Method for enhancing glycoprotein stability
US5147788A (en) * 1988-05-06 1992-09-15 Burroughs Wellcome Co. Baculovirus vectors and methods of use
US5153265A (en) * 1988-01-20 1992-10-06 Cetus Corporation Conjugation of polymer to colony stimulating factor-1
US5154924A (en) * 1989-09-07 1992-10-13 Alkermes, Inc. Transferrin receptor specific antibody-neuropharmaceutical agent conjugates
US5169933A (en) * 1988-08-15 1992-12-08 Neorx Corporation Covalently-linked complexes and methods for enhanced cytotoxicity and imaging
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
US5346696A (en) * 1991-06-19 1994-09-13 Korea Green Cross Corporation Asialoglycoprotein - conjugated medicinal agent
US5352670A (en) * 1991-06-10 1994-10-04 Alberta Research Council Methods for the enzymatic synthesis of alpha-sialylated oligosaccharide glycosides
US5369017A (en) * 1994-02-04 1994-11-29 The Scripps Research Institute Process for solid phase glycopeptide synthesis
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
US5405753A (en) * 1990-03-26 1995-04-11 Brossmer; Reinhard CMP-activated, fluorescing sialic acids, as well as processes for their preparation
US5432059A (en) * 1994-04-01 1995-07-11 Specialty Laboratories, Inc. Assay for glycosylation deficiency disorders
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
US5545553A (en) * 1994-09-26 1996-08-13 The Rockefeller University Glycosyltransferases for biosynthesis of oligosaccharides, and genes encoding them
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
US5635603A (en) * 1993-12-08 1997-06-03 Immunomedics, Inc. Preparation and use of immunoconjugates
US5646113A (en) * 1994-04-07 1997-07-08 Genentech, Inc. Treatment of partial growth hormone insensitivity syndrome
US5672683A (en) * 1989-09-07 1997-09-30 Alkermes, Inc. Transferrin neuropharmaceutical agent fusion protein
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
US5834251A (en) * 1994-12-30 1998-11-10 Alko Group Ltd. Methods of modifying carbohydrate moieties
US5849535A (en) * 1995-09-21 1998-12-15 Genentech, Inc. Human growth hormone variants
US5876980A (en) * 1995-04-11 1999-03-02 Cytel Corporation Enzymatic synthesis of oligosaccharides
US5922577A (en) * 1995-04-11 1999-07-13 Cytel Corporation Enzymatic synthesis of glycosidic linkages
US5932462A (en) * 1995-01-10 1999-08-03 Shearwater Polymers, Inc. Multiarmed, monofunctional, polymer for coupling to molecules and surfaces
US5945314A (en) * 1997-03-31 1999-08-31 Abbott Laboratories Process for synthesizing oligosaccharides
US5977307A (en) * 1989-09-07 1999-11-02 Alkermes, Inc. Transferrin receptor specific ligand-neuropharmaceutical agent fusion proteins
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
US6087325A (en) * 1996-10-15 2000-07-11 The Liposome Company, Inc. Peptide-lipid conjugates
US6096512A (en) * 1993-05-14 2000-08-01 Pharmacia & Upjohn Company Cloned DNA encoding a UDP-GalNAc: Polypeptide, N-acetylgalactosaminyltransferase
US6166183A (en) * 1992-11-30 2000-12-26 Kirin-Amgen, Inc. Chemically-modified G-CSF
US6183738B1 (en) * 1997-05-12 2001-02-06 Phoenix Pharamacologics, Inc. Modified arginine deiminase
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
US6362254B2 (en) * 1998-03-12 2002-03-26 Shearwater Corporation Poly(ethylene glycol) derivatives with proximal reactive groups
US6440703B1 (en) * 1997-12-01 2002-08-27 Neose Technologies, Inc. Enzymatic synthesis of gangliosides
US20020137134A1 (en) * 2000-06-28 2002-09-26 Gerngross Tillman U. Methods for producing modified glycoproteins
US20020150981A1 (en) * 1999-09-14 2002-10-17 Novazyme Pharmaceuticals, Inc. Methods for producing highly phosphorylated lysosomal hydrolases
US20020168323A1 (en) * 2001-05-11 2002-11-14 Igor Gonda Optimization of the molecular properties and formulation of proteins delivered by inhalation
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
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
US20040063911A1 (en) * 2001-10-10 2004-04-01 Neose Technologies, Inc. Protein remodeling methods and proteins/peptides produced by the methods
US20040137557A1 (en) * 2001-10-10 2004-07-15 Neose Technologies, Inc. Remodeling and glycoconjugation of peptides
US20040136955A1 (en) * 2002-09-05 2004-07-15 Barker Nicholas P Modified asialo-interferons and uses thereof
US20050118672A1 (en) * 2001-11-28 2005-06-02 Defrees Shawn Glycopeptide remodeling using amidases
US20050143292A1 (en) * 2003-11-24 2005-06-30 Defrees Shawn Glycopegylated erythropoietin
US20060024286A1 (en) * 2004-08-02 2006-02-02 Paul Glidden Variants of tRNA synthetase fragments and uses thereof
US20060111279A1 (en) * 2003-11-24 2006-05-25 Neose Technologies, Inc. Glycopegylated erythropoietin
US7125843B2 (en) * 2001-10-19 2006-10-24 Neose Technologies, Inc. Glycoconjugates including more than one peptide
US20070027068A1 (en) * 2001-10-10 2007-02-01 Defrees Shawn Erythropoietin: remodeling and glycoconjugation of erythropoietin
US7202208B2 (en) * 2000-05-15 2007-04-10 Hoffman-La Roche Inc. Erythropoietin composition
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

Family Cites Families (475)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US1088538A (en) * 1913-07-28 1914-02-24 Chester H Clark Grease-cup.
US2042369A (en) 1935-04-23 1936-05-26 Kilian Albert Von One-button switch
US3691016A (en) 1970-04-17 1972-09-12 Monsanto Co Process for the preparation of insoluble enzymes
CA1023287A (en) 1972-12-08 1977-12-27 Boehringer Mannheim G.M.B.H. Process for the preparation of carrier-bound proteins
GB1451798A (en) 1973-08-02 1976-10-06 Ici Ltd Prostanoic acid derivatives
US4235871A (en) 1978-02-24 1980-11-25 Papahadjopoulos Demetrios P Method of encapsulating biologically active materials in lipid vesicles
IL63270A0 (en) 1980-08-05 1981-10-30 Univ Leland Stanford Junior Eukaryotic autonomously replicating segment
US4579821A (en) 1981-11-23 1986-04-01 University Patents, Inc. Control of DNA sequence transcription
US4656134A (en) 1982-01-11 1987-04-07 Board Of Trustees Of Leland Stanford Jr. University Gene amplification in eukaryotic cells
US4975276A (en) 1982-01-15 1990-12-04 Cetus Corporation Interferon-alpha 54
US4748233A (en) 1982-03-23 1988-05-31 Bristol-Myers Company Alpha-interferon Gx-1
US4695543A (en) 1982-03-23 1987-09-22 Bristol-Myers Company Alpha Interferon GX-1
US6936694B1 (en) 1982-05-06 2005-08-30 Intermune, Inc. Manufacture and expression of large structural genes
DE3381090D1 (de) 1982-05-19 1990-02-15 Gist Brocades Nv Klonierungssystem fuer kluyveromyce spezies.
US4486533A (en) 1982-09-02 1984-12-04 St. Louis University Filamentous fungi functional replicating extrachromosomal element
US4522811A (en) 1982-07-08 1985-06-11 Syntex (U.S.A.) Inc. Serial injection of muramyldipeptides and liposomes enhances the anti-infective activity of muramyldipeptides
US4599311A (en) 1982-08-13 1986-07-08 Kawasaki Glenn H Glycolytic promotersfor regulated protein expression: protease inhibitor
US5151511A (en) 1982-09-16 1992-09-29 Amgen Inc. DNA encoding avian growth hormones
US4588585A (en) 1982-10-19 1986-05-13 Cetus Corporation Human recombinant cysteine depleted interferon-β muteins
US4737462A (en) 1982-10-19 1988-04-12 Cetus Corporation Structural genes, plasmids and transformed cells for producing cysteine depleted muteins of interferon-β
US4966843A (en) 1982-11-01 1990-10-30 Cetus Corporation Expression of interferon genes in Chinese hamster ovary cells
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
US4501728A (en) 1983-01-06 1985-02-26 Technology Unlimited, Inc. Masking of liposomes from RES recognition
US4603044A (en) 1983-01-06 1986-07-29 Technology Unlimited, Inc. Hepatocyte Directed Vesicle delivery system
US4713339A (en) 1983-01-19 1987-12-15 Genentech, Inc. Polycistronic expression vector construction
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
US4518584A (en) 1983-04-15 1985-05-21 Cetus Corporation Human recombinant interleukin-2 muteins
US4745051A (en) 1983-05-27 1988-05-17 The Texas A&M University System Method for producing a recombinant baculovirus expression vector
US4923805A (en) 1983-11-02 1990-05-08 Integrated Genetics, Inc. Fsh
US5156957A (en) 1983-11-02 1992-10-20 Genzyme Corporation Follicle stimulating hormone
US5639639A (en) 1983-11-02 1997-06-17 Genzyme Corporation Recombinant heterodimeric human fertility hormones, and methods, cells, vectors and DNA for the production thereof
US4840896A (en) 1983-11-02 1989-06-20 Integrated Genetics, Inc. Heteropolymeric protein
US4703008A (en) 1983-12-13 1987-10-27 Kiren-Amgen, Inc. DNA sequences encoding erythropoietin
DE3572982D1 (en) 1984-03-06 1989-10-19 Takeda Chemical Industries Ltd Chemically modified lymphokine and production thereof
US4931373A (en) 1984-05-25 1990-06-05 Zymogenetics, Inc. Stable DNA constructs for expression of α-1 antitrypsin
US4589402A (en) 1984-07-26 1986-05-20 Serono Laboratories, Inc. Method of in vitro fertilization
EP0170204B1 (de) 1984-08-01 1991-09-25 BOEHRINGER INGELHEIM INTERNATIONAL GmbH Neue genetische Sequenzen, die durch sie codierten Interferon-Peptide vom Typ I und diese sie produzierende Organismen
US4761371A (en) 1985-02-12 1988-08-02 Genentech, Inc. Insulin receptor
US4675414A (en) 1985-03-08 1987-06-23 The United States Of America As Represented By The Secretary Of The Navy Maleimidomethyl-carbonate polyethers
US5034506A (en) 1985-03-15 1991-07-23 Anti-Gene Development Group Uncharged morpholino-based polymers having achiral intersubunit linkages
US4683195A (en) 1986-01-30 1987-07-28 Cetus Corporation Process for amplifying, detecting, and/or-cloning nucleic acid sequences
US4683202A (en) 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
GR860984B (en) 1985-04-17 1986-08-18 Zymogenetics Inc Expression of factor vii and ix activities in mammalian cells
DE3684546D1 (de) 1985-04-22 1992-04-30 Genetics Inst Herstellung mit hoher leistung des aktivfaktors ix.
DE3676670D1 (de) 1985-06-26 1991-02-07 Cetus Corp Solubilisierung von proteinen fuer pharmazeutische zusammensetzungen mittels polymerkonjugierung.
US5206344A (en) 1985-06-26 1993-04-27 Cetus Oncology Corporation Interleukin-2 muteins and polymer conjugation thereof
US4810643A (en) 1985-08-23 1989-03-07 Kirin- Amgen Inc. Production of pluripotent granulocyte colony-stimulating factor
US6004548A (en) 1985-08-23 1999-12-21 Amgen, Inc. Analogs of pluripotent granulocyte colony-stimulating factor
IL80529A0 (en) 1985-11-14 1987-02-27 Daiichi Seiyaku Co Method of producing peptides
SE451849B (sv) 1985-12-11 1987-11-02 Svenska Sockerfabriks Ab Sett att syntetisera glykosidiska bindningar samt anvendning av pa detta sett erhallna produkter
US4935349A (en) 1986-01-17 1990-06-19 Zymogenetics, Inc. Expression of higher eucaryotic genes in aspergillus
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
US5272066A (en) 1986-03-07 1993-12-21 Massachusetts Institute Of Technology Synthetic method for enhancing glycoprotein stability
AU597574B2 (en) 1986-03-07 1990-06-07 Massachusetts Institute Of Technology Method for enhancing glycoprotein stability
DE3607835A1 (de) 1986-03-10 1987-09-24 Boehringer Ingelheim Int Hybridinterferone, deren verwendung als arzneimittel und als zwischenprodukte zur herstellung von antikoerpern und deren verwendung sowie verfahren zu ihrer herstellung
IT1203758B (it) 1986-03-27 1989-02-23 Univ Roma Vettori di clonazione e di espressione di geni eterologhi in lieviti e lieviti trasformati con tali vettori
GB8610600D0 (en) 1986-04-30 1986-06-04 Novo Industri As Transformation of trichoderma
DK173067B1 (da) 1986-06-27 1999-12-13 Univ Washington Humant erythropoietin-gen, fremgangsmåde til ekspression deraf i transficerede cellelinier, de transficerede cellelinier sa
US4902505A (en) 1986-07-30 1990-02-20 Alkermes Chimeric peptides for neuropeptide delivery through the blood-brain barrier
DE3633323A1 (de) 1986-10-01 1988-04-07 Boehringer Ingelheim Int Neue monoklonale antikoerper gegen ifn-omega, verfahren zu ihrer herstellung und deren verwendung zur reinigung sowie zum nachweis von ifn-omega
US5075109A (en) 1986-10-24 1991-12-24 Southern Research Institute Method of potentiating an immune response
US4894330A (en) 1986-12-23 1990-01-16 Cetus Corporation Purification of recombinant beta-interferon incorporating RP-HPLC
US4837028A (en) 1986-12-24 1989-06-06 Liposome Technology, Inc. Liposomes with enhanced circulation time
DK175243B1 (da) 1987-03-23 2004-07-19 Zymogenetics Inc Ekspressionsvektor, der er i stand til at styre ekspression af heterologe gener eller cDNA i gær, gærværtscelle samt fremgangsmåde til öget produktion af proteiner i gærværtsceller
IL82834A (en) 1987-06-09 1990-11-05 Yissum Res Dev Co Biodegradable polymeric materials based on polyether glycols,processes for the preparation thereof and surgical artiicles made therefrom
CA1323850C (en) 1987-07-28 1993-11-02 Johannes A. Van Den Berg Kluyveromyces as a host strain
US4897268A (en) 1987-08-03 1990-01-30 Southern Research Institute Drug delivery system and method of making the same
US5252726A (en) 1987-09-04 1993-10-12 Novo Nordisk A/S Promoters for use in aspergillus
GB8725529D0 (en) 1987-10-30 1987-12-02 Delta Biotechnology Ltd Polypeptides
US4847325A (en) 1988-01-20 1989-07-11 Cetus Corporation Conjugation of polymer to colony stimulating factor-1
JP2511494B2 (ja) 1988-05-12 1996-06-26 善治 松浦 日本脳炎ウイルス表面抗原蛋白質の製造法
FR2631974B1 (fr) 1988-05-31 1992-12-11 Agronomique Inst Nat Rech Baculovirus modifie, son procede de preparation et son application en tant que vecteur d'expression de genes
FR2649991B2 (fr) 1988-08-05 1994-03-04 Rhone Poulenc Sante Utilisation de derives stables du plasmide pkd1 pour l'expression et la secretion de proteines heterologues dans les levures du genre kluyveromyces
US5874261A (en) 1988-09-02 1999-02-23 The Trustees Of The University Of Pennsylvania Method for the purification of glycosyltransferases
US5223409A (en) 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
US5162215A (en) 1988-09-22 1992-11-10 Amgen Inc. Method of gene transfer into chickens and other avian species
US5218092A (en) 1988-09-29 1993-06-08 Kyowa Hakko Kogyo Co., Ltd. Modified granulocyte-colony stimulating factor polypeptide with added carbohydrate chains
FR2638643B1 (fr) 1988-11-09 1991-04-12 Transgene Sa Sequence d'adn codant pour le facteur ix humain ou une proteine analogue, vecteur d'expression, cellules transformees, procede de preparation du facteur ix et produits obtenus correspondants
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
JP2989002B2 (ja) 1988-12-22 1999-12-13 キリン―アムジエン・インコーポレーテツド 化学修飾顆粒球コロニー刺激因子
DE68929551T2 (de) 1988-12-23 2008-03-06 Genentech, Inc., South San Francisco Menschliche DNase
US5162228A (en) 1988-12-28 1992-11-10 Takeda Chemical Industries, Ltd. Gylceraldehyde-3-phosphate dehydrogenase gene and promoter
US5198346A (en) 1989-01-06 1993-03-30 Protein Engineering Corp. Generation and selection of novel DNA-binding proteins and polypeptides
US5096815A (en) 1989-01-06 1992-03-17 Protein Engineering Corporation Generation and selection of novel dna-binding proteins and polypeptides
EP0454730A1 (de) 1989-01-19 1991-11-06 The Upjohn Company Somatotropinanaloga
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
US4957773A (en) 1989-02-13 1990-09-18 Syracuse University Deposition of boron-containing films from decaborane
US5475090A (en) 1989-02-21 1995-12-12 Boyce Thompson Institute For Plant Research Gene coded for a polypeptide which enhances virus infection of host insects
US5338835A (en) 1989-02-21 1994-08-16 Washington University CTP-extended form of FSH
DE3906540A1 (de) 1989-03-02 1990-09-13 Behringwerke Ag Expressionsvektoren zur synthese von proteinen in der spalthefe schizosaccharomyces pombe
US5179023A (en) 1989-03-24 1993-01-12 Research Corporation Technologies, Inc. Recombinant α-galactosidase a therapy for Fabry disease
US5059535A (en) 1989-04-12 1991-10-22 Chembiomed, Ltd. Process for the separation and purification of sialyl transferases
US5122614A (en) 1989-04-19 1992-06-16 Enzon, Inc. Active carbonates of polyalkylene oxides for modification of polypeptides
ATE300519T1 (de) 1989-04-19 2005-08-15 Enzon Inc Verfahren zum herstellen von modifizierten polypeptiden die eine polypeptid und ein polyalkylenoxid enthalten
US5324844A (en) 1989-04-19 1994-06-28 Enzon, Inc. Active carbonates of polyalkylene oxides for modification of polypeptides
US5166322A (en) 1989-04-21 1992-11-24 Genetics Institute Cysteine added variants of interleukin-3 and chemical modifications thereof
DE394538T1 (de) 1989-04-28 1991-04-11 Rhein Biotech Ges. Fuer Biotechnologische Prozesse Und Produkte Mbh, 4000 Duesseldorf, De Hefezellen der schwanniomyces-gattung.
US5766883A (en) 1989-04-29 1998-06-16 Delta Biotechnology Limited Polypeptides
US5244805A (en) 1989-05-17 1993-09-14 University Of Georgia Research Foundation, Inc. Baculovirus expression vectors
US5342940A (en) 1989-05-27 1994-08-30 Sumitomo Pharmaceuticals Company, Limited Polyethylene glycol derivatives, process for preparing the same
FR2649565B1 (fr) 1989-07-05 1996-06-14 Lem Dispositif de couplage acoustique destine a effectuer des mesures et/ou des controles telephonometriques sur des combines telephoniques
US5162222A (en) 1989-07-07 1992-11-10 Guarino Linda A Use of baculovirus early promoters for expression of foreign genes in stably transformed insect cells or recombinant baculoviruses
US5077214A (en) 1989-07-07 1991-12-31 The Texas A&M University System Use of baculovirus early promoters for expression of foreign genes in stably transformed insect cells
US5179007A (en) 1989-07-07 1993-01-12 The Texas A & M University System Method and vector for the purification of foreign proteins
DE3924746A1 (de) 1989-07-26 1991-01-31 Behringwerke Ag Erythropoietin (epo)-peptide und dagegen gerichtete antikoerper
FR2650598B1 (fr) 1989-08-03 1994-06-03 Rhone Poulenc Sante Derives de l'albumine a fonction therapeutique
US5155037A (en) 1989-08-04 1992-10-13 The Texas A&M University System Insect signal sequences useful to improve the efficiency of processing and secretion of foreign genes in insect systems
US5023328A (en) 1989-08-04 1991-06-11 The Texas A&M University System Lepidopteran AKH signal sequence
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
US5580560A (en) 1989-11-13 1996-12-03 Novo Nordisk A/S Modified factor VII/VIIa
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
US5104654A (en) * 1989-12-05 1992-04-14 University Of California Ovipositional disruption of the navel orangeworm with fatty acids
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
US5606031A (en) 1990-04-06 1997-02-25 Lile; Jack Production and purification of biologically active recombinant neurotrophic protein in bacteria
US5583042A (en) 1990-04-16 1996-12-10 Neose Pharmaceuticals, Inc. Apparatus for the synthesis of saccharide compositions
US5180674A (en) 1990-04-16 1993-01-19 The Trustees Of The University Of Pennsylvania Saccharide compositions, methods and apparatus for their synthesis
GB9107846D0 (en) 1990-04-30 1991-05-29 Ici Plc Polypeptides
US5951972A (en) * 1990-05-04 1999-09-14 American Cyanamid Company Stabilization of somatotropins and other proteins by modification of cysteine residues
US5399345A (en) 1990-05-08 1995-03-21 Boehringer Mannheim, Gmbh Muteins of the granulocyte colony stimulating factor
US5219564A (en) 1990-07-06 1993-06-15 Enzon, Inc. Poly(alkylene oxide) amino acid copolymers and drug carriers and charged copolymers based thereon
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
CA2084514A1 (en) 1990-07-10 1992-01-11 Guenther Adolf O-glycosylated ifn-alpha
FR2664905B1 (fr) 1990-07-18 1994-08-12 Agronomique Inst Nat Rech Baculovirus modifie, son procede d'obtention, et vecteurs d'expression obtenus a partir dudit baculovirus.
DE4028800A1 (de) 1990-09-11 1992-03-12 Behringwerke Ag Gentechnische sialylierung von glykoproteinen
US5169784A (en) 1990-09-17 1992-12-08 The Texas A & M University System Baculovirus dual promoter expression vector
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
US5401650A (en) 1990-10-24 1995-03-28 The Mount Sinai School Of Medicine Of The City University Of New York Cloning and expression of biologically active α-galactosidase A
GB9023111D0 (en) 1990-10-24 1990-12-05 Wellcome Found Expression system
CA2073511A1 (en) 1990-11-14 1992-05-29 Matthew R. Callstrom 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
US5420027A (en) 1991-01-10 1995-05-30 Board Of Regents, The University Of Texas System Methods and compositions for the expression of biologically active fusion proteins comprising a eukaryotic cytochrome P450 fused to a reductase in bacteria
US5948682A (en) 1991-02-22 1999-09-07 Sembiosys Genetics Inc. Preparation of heterologous proteins on oil bodies
US5650554A (en) 1991-02-22 1997-07-22 Sembiosys Genetics Inc. Oil-body proteins as carriers of high-value peptides in plants
US6288304B1 (en) 1991-02-22 2001-09-11 Sembiosys Genetics Inc. Expression of somatotropin in plant seeds
US5997864A (en) 1995-06-07 1999-12-07 Novo Nordisk A/S Modified factor VII
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
US5817788A (en) 1991-02-28 1998-10-06 Zymogenetics, Inc. Modified factor VII
CA2103546C (en) 1991-02-28 2002-10-01 Kathleen L. Berkner Modified factor vii
WO1994027631A1 (en) 1993-05-21 1994-12-08 Zymogenetics, Inc. Modified factor vii
US5833982A (en) 1991-02-28 1998-11-10 Zymogenetics, Inc. Modified factor VII
US5278299A (en) 1991-03-18 1994-01-11 Scripps Clinic And Research Foundation Method and composition for synthesizing sialylated glycosyl compounds
AU668505B2 (en) 1991-03-18 1996-05-09 Scripps Research Institute, The Oligosaccharide enzyme substrates and inhibitors: methods and compositions
JPH06506217A (ja) 1991-03-18 1994-07-14 エンゾン,インコーポレーテッド ポリペプチドまたはグリコポリペプチドとポリマーとのヒドラジン含有結合体
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
GB2256197B (en) 1991-05-31 1995-11-22 Ciba Geigy Ag Yeast as host for expression of heterologous glycosyl transferase enzymes
SE9201544L (sv) 1991-05-31 1992-12-01 Ciba Geigy Ag Saett att framstaella glykosyltransferaser
US5472858A (en) 1991-06-04 1995-12-05 Wisconsin Alumni Research Foundation Production of recombinant proteins in insect larvae
US5374655A (en) 1991-06-10 1994-12-20 Alberta Research Council Methods for the synthesis of monofucosylated oligosaccharides terminating in di-N-acetyllactosaminyl structures
SE9101853D0 (sv) * 1991-06-17 1991-06-17 Jonas Wadstroem Improved tissue ashesive
US5633146A (en) 1991-07-02 1997-05-27 Rhone-Poulenc Rorer S.A. Method for producing recombinant proteins and host cells used therein
US5281698A (en) 1991-07-23 1994-01-25 Cetus Oncology Corporation Preparation of an activated polymer ester for protein conjugation
EP0863154A1 (de) 1991-10-12 1998-09-09 The Regents Of The University Of California Verwendung von Thiol-Redox-Proteinen zur Reduktion des intramolekularen Disulfid-Bindung bei Proteinen, zur Besserung der Qualität der Getreidenprodukte, des Teigs und der gebackenen Ware
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
AU665599B2 (en) 1991-11-08 1996-01-11 Hemosol Inc. Hemoglobins as drug delivery agents
US5384249A (en) 1991-12-17 1995-01-24 Kyowa Hakko Kogyo Co., Ltd. α2→3 sialyltransferase
DK8892D0 (da) 1992-01-23 1992-01-23 Symbicom Ab Humant proteingen
IT1260468B (it) 1992-01-29 1996-04-09 Metodo per mantenere l'attivita' di enzimi proteolitici modificati con polietilenglicole
US6039944A (en) 1992-02-28 2000-03-21 Zymogenetics, Inc. Modified Factor VII
US5965106A (en) 1992-03-04 1999-10-12 Perimmune Holdings, Inc. In vivo binding pair pretargeting
US5858751A (en) * 1992-03-09 1999-01-12 The Regents Of The University Of California Compositions and methods for producing sialyltransferases
JPH07505771A (ja) 1992-03-09 1995-06-29 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア シアリルトランスフェラーゼの同定と合成のための構成物と方法
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
AU3937593A (en) 1992-03-25 1993-10-21 New York University Trans-sialidase and methods of use and making thereof
DE69332538T2 (de) 1992-04-02 2003-11-06 Sembiosys Genetics Inc Cis elemente des ölkörperproteins als regulationische signale
US5516657A (en) 1992-05-11 1996-05-14 Cambridge Biotech Corporation Baculovirus vectors for expression of secretory and membrane-bound proteins
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 (en) 1992-09-01 1994-03-17 Berlex Laboratories, Inc. Glycolation of glycosylated macromolecules
US5348886A (en) 1992-09-04 1994-09-20 Monsanto Company Method of producing recombinant eukaryotic viruses in bacteria
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
KR950704497A (ko) 1992-11-27 1995-11-20 베르너 발데크 글리코실트랜스퍼라제 활성을 가진 단백질(Proteins Having Glycosyltransferase Activity)
CA2110543A1 (en) 1992-12-09 1994-06-10 David E. Wright Peg hydrazone and peg oxime linkage forming reagents and protein derivatives thereof
NZ250375A (en) 1992-12-09 1995-07-26 Ortho Pharma Corp Peg hydrazone and peg oxime linkage forming reagents and protein derivatives
NO934477L (no) 1992-12-09 1994-06-10 Ortho Pharma Corp PEG hydrazon- og PEG oksim-bindingdannende reagenser og proteinderivater derav
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
US5202413A (en) 1993-02-16 1993-04-13 E. I. Du Pont De Nemours And Company Alternating (ABA)N polylactide block copolymers
US6180134B1 (en) 1993-03-23 2001-01-30 Sequus Pharmaceuticals, Inc. Enhanced ciruclation effector composition and method
EP0643132B1 (de) 1993-03-29 2006-05-31 Kyowa Hakko Kogyo Co., Ltd. Alpha-1,3-Fucosyltransferase
CN1125401A (zh) 1993-04-29 1996-06-26 艾博特公司 促红细胞生成素类似物组合物和方法
US5409817A (en) 1993-05-04 1995-04-25 Cytel, Inc. Use of trans-sialidase and sialyltransferase for synthesis of sialylα2→3βgalactosides
US6174530B1 (en) 1993-05-05 2001-01-16 Gryphon Sciences Homogeneous polyoxime compositions and their preparation by parallel assembly
US6001364A (en) 1993-05-05 1999-12-14 Gryphon Sciences Hetero-polyoxime compounds and their preparation by parallel assembly
PL176272B1 (pl) 1993-05-14 1999-05-31 Cytel Corp Analogi sialilowe Le jako inhibitory adhezji komórkowej
AU7097094A (en) 1993-06-01 1994-12-20 Enzon, Inc. Carbohydrate-modified polymer conjugates with erythropoietic activity
AU691510B2 (en) 1993-07-15 1998-05-21 Neose Pharmaceuticals Method of synthesizing saccharide compositions
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
US5919455A (en) 1993-10-27 1999-07-06 Enzon, Inc. Non-antigenic branched polymer conjugates
US5643575A (en) 1993-10-27 1997-07-01 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
US5827690A (en) 1993-12-20 1998-10-27 Genzyme Transgenics Corporatiion Transgenic production of antibodies in milk
AU1377395A (en) 1993-12-23 1995-07-10 University Technologies International Inc. Methods of expressing proteins in insect cells and methods of killing insects
JP3516272B2 (ja) 1994-02-10 2004-04-05 株式会社成和化成 化粧品基材
US5605793A (en) 1994-02-17 1997-02-25 Affymax Technologies N.V. Methods for in vitro recombination
US6117679A (en) 1994-02-17 2000-09-12 Maxygen, Inc. Methods for generating polynucleotides having desired characteristics by iterative selection and recombination
US5834252A (en) 1995-04-18 1998-11-10 Glaxo Group Limited End-complementary polymerase reaction
US6165793A (en) 1996-03-25 2000-12-26 Maxygen, Inc. Methods for generating polynucleotides having desired characteristics by iterative selection and recombination
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.
US5545723A (en) 1994-03-15 1996-08-13 Biogen Inc. Muteins of IFN-β
JP3090586B2 (ja) 1994-03-15 2000-09-25 片倉工業株式会社 システインプロテアーゼ遺伝子欠損バキュロウイルスおよびその製造法並びにこれを利用する有用タンパク質の製造法
IL113010A0 (en) 1994-03-31 1995-10-31 Pharmacia Ab Pharmaceutical formulation comprising factor VIII or factor ix with an activity of at least 200 IU/ml and an enhancer for improved subcutaneous intramuscular or intradermal administration
GB9408717D0 (en) 1994-05-03 1994-06-22 Biotech & Biolog Scien Res DNA sequences
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
JP3729210B2 (ja) * 1994-07-26 2005-12-21 富士通株式会社 半導体装置の製造方法
US5871986A (en) 1994-09-23 1999-02-16 The General Hospital Corporation Use of a baculovirus to express and exogenous gene in a mammalian cell
EP0785276A4 (de) 1994-09-29 2002-01-09 Ajinomoto Kk Modifizierung eines peptids und eines proteins
US5824784A (en) 1994-10-12 1998-10-20 Amgen Inc. N-terminally chemically modified protein compositions and methods
US5521299A (en) 1994-11-22 1996-05-28 National Science Council Oligonucleotides for detection of baculovirus infection
IL116730A0 (en) 1995-01-13 1996-05-14 Amgen Inc Chemically modified interferon
US5843705A (en) 1995-02-21 1998-12-01 Genzyme Transgenic Corporation Transgenically produced antithrombin III
GB9506249D0 (en) 1995-03-27 1995-05-17 Karobio Ab Media for insect cell cultures
SE9501285L (sv) 1995-04-06 1996-10-07 Johanna Ljung Förfarande för framställning av biologiskt aktiva proteiner
AU5445396A (en) 1995-04-11 1996-10-30 Cytel Corporation Improved enzymatic synthesis of oligosaccharides
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
AU6255096A (en) 1995-06-07 1996-12-30 Mount Sinai School Of Medicine Of The City University Of New York, The Pegylated modified proteins
US6251864B1 (en) 1995-06-07 2001-06-26 Glaxo Group Limited Peptides and compounds that bind to a receptor
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
US5858752A (en) 1995-06-07 1999-01-12 The General Hospital Corporation Fucosyltransferase genes and uses thereof
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
WO1997005330A1 (en) 1995-07-27 1997-02-13 Cytec Technology Corp. Synthetic cationic polymers as promoters for asa sizing
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
US5811634A (en) 1995-09-12 1998-09-22 Thomas G. O'Brien Transgenic mammal encoding ornithine decarboxylase
SE9503380D0 (sv) 1995-09-29 1995-09-29 Pharmacia Ab Protein derivatives
US5767379A (en) 1995-11-06 1998-06-16 John Howard Commercial production of avidin in plants
EP0862640A2 (de) 1995-11-09 1998-09-09 ZymoGenetics, Inc. Herstellung von gad65 in methylotrophen hefen
US5965408A (en) 1996-07-09 1999-10-12 Diversa Corporation Method of DNA reassembly by interrupting synthesis
US6171820B1 (en) 1995-12-07 2001-01-09 Diversa Corporation Saturation mutagenesis in directed evolution
US6361974B1 (en) 1995-12-07 2002-03-26 Diversa Corporation Exonuclease-mediated nucleic acid reassembly in directed evolution
CA2165041C (en) 1995-12-12 2005-07-05 The University Of British Columbia Methods and compositions for synthesis of oligosaccharides, and the products formed thereby
WO1997021822A2 (en) 1995-12-12 1997-06-19 The University Of British Columbia Methods and compositions for synthesis of oligosaccharides using mutant glycosidase enzymes
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
US5728580A (en) 1996-02-20 1998-03-17 Cornell Research Foundation, Inc. Methods and culture media for inducing single cell suspension in insect cell lines
DE69738368T2 (de) * 1996-03-08 2008-12-04 The Regents Of The University Of Michigan, Ann Arbor MURINE alpha(1,3)-FUCOSYLTRANSFERASE (Fuc-TVII)
US6096548A (en) 1996-03-25 2000-08-01 Maxygen, Inc. Method for directing evolution of a virus
US5750383A (en) 1996-05-14 1998-05-12 Boyce Thompson Institute For Plant Research, Inc. Baculovirus cloning system
US6096529A (en) 1996-06-10 2000-08-01 National Research Council Of Canada Recombinant α-2,3-sialyltransferases and their uses
CA2261151C (en) 1996-07-17 2003-09-16 Zymogenetics, Inc. Transformation of pichia methanolica
WO1998002536A2 (en) 1996-07-17 1998-01-22 Zymogenetics, Inc. Preparation of pichia methanolica auxotrophic mutants
SI0964702T1 (sl) 1996-08-02 2007-02-28 Ortho Mcneil Pharm Inc Polipeptidi, ki imajo posamezen kovalentno vezan N-terminalni vodotopni polimer
EP0953354A4 (de) 1996-08-13 2002-10-23 Fujisawa Pharmaceutical Co Hämatopoietische stammzellen proliferierende wirkstoffe
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
US6117651A (en) 1996-11-08 2000-09-12 Neose Technologies, Inc. Expression vectors
US5856452A (en) 1996-12-16 1999-01-05 Sembiosys Genetics Inc. Oil bodies and associated proteins as affinity matrices
BR9606270A (pt) 1996-12-18 1998-09-22 Univ Minas Gerais Processo para a produção da proteína do interferon beta-cis humano recombinante e proteína de interferon beta-cis humano recombinante
ATE263841T1 (de) 1997-01-16 2004-04-15 Neose Technologies Inc Praktische in vitro sialylierung von rekombinanten glykpproteinen
ATE200030T1 (de) 1997-01-29 2001-04-15 Polymasc Pharmaceuticals Plc Pegylationsverfahren
DE19709787A1 (de) 1997-03-11 1998-09-17 Bayer Ag Oligosaccaride und deren Derivate sowie ein chemo-enzymatisches Verfahren zu deren Herstellung
WO1998049198A1 (en) 1997-04-30 1998-11-05 Enzon, Inc. Single-chain antigen-binding proteins capable of glycosylation, production and uses thereof
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
US7585645B2 (en) 1997-05-27 2009-09-08 Sembiosys Genetics Inc. Thioredoxin and thioredoxin reductase containing oil body based products
WO1998055630A2 (en) 1997-06-06 1998-12-10 The Governors Of The University Of Alberta Alpha-1,3-fucosyltransferase of helicobacter pylori
EP1001968B1 (de) 1997-06-13 2004-12-22 Gryphon Therapeutics, Inc. Festphasige native chemische ligation von ungeschützten oder n-cystein-geschützten peptiden in wässrigen lösungen
US6210736B1 (en) 1997-06-17 2001-04-03 Genzyme Transgenics Corporation Transgenically produced prolactin
EP0994903B1 (de) 1997-06-24 2005-05-25 Genentech, Inc. Galactosylierte glykoproteine enthaltende zusammensetzungen und verfahren zur deren herstellung
WO1999000150A2 (en) 1997-06-27 1999-01-07 Regents Of The University Of California Drug targeting of a peptide radiopharmaceutical through the primate blood-brain barrier in vivo with a monoclonal antibody to the human insulin receptor
US6187564B1 (en) 1997-07-10 2001-02-13 Beth Israel Deaconess Medical Center DNA encoding erythropoietin multimers having modified 5′ and 3′ sequences and its use to prepare EPO therapeutics
US6090584A (en) 1997-08-21 2000-07-18 University Technologies International Inc. Baculovirus artificial chromosomes and methods of use
WO1999013063A1 (en) 1997-09-09 1999-03-18 Nycomed Imaging As Factor vii fragments and analogs thereof and their use in the treatment of blood clottng disorders
EP1028751B1 (de) 1997-10-31 2008-12-31 Genentech, Inc. Methoden und zusammensetzungen bestehend aus glykoprotein-glykoformen
AU1808699A (en) 1997-12-08 1999-06-28 Board Of Trustees Of The University Of Arkansas, The Purified beta1,2-xylosyltransferase and uses thereof
US7244601B2 (en) 1997-12-15 2007-07-17 National Research Council Of Canada Fusion proteins for use in enzymatic synthesis of oligosaccharides
TW445295B (en) 1997-12-31 2001-07-11 Shiu Li Wei Expression vector pcDNA3.1-HC for human erythropoietin, BHK-21 host cell line transformed therewith, and production of human erythropoietin using the transformed cell
ATE255422T1 (de) 1998-01-07 2003-12-15 Debio Rech Pharma Sa Abbaubare, heterobifunktionelle polyethylenglykolacrylate, sowie damit herstellbare gele und konjugate
WO1999037779A1 (en) 1998-01-22 1999-07-29 Genentech, Inc. Antibody fragment-polymer conjugates and humanized anti-il-8 monoclonal antibodies and uses of same
AR014491A1 (es) 1998-01-29 2001-02-28 Dow Agrosciences Llc Metodo para obtener plantas transgenicas fertiles de gossypium hirsutum.
US6689604B1 (en) 1998-03-20 2004-02-10 National Research Council Of Canada Lipopolysaccharide α-2,3 sialyltransferase of Campylobacter jejuni and its uses
AU758097B2 (en) * 1998-03-25 2003-03-13 Sloan-Kettering Institute For Cancer Research Trimeric antigenic O-linked glycopeptide conjugates, methods of preparation and uses thereof
EP2261229A3 (de) 1998-04-20 2011-03-23 GlycArt Biotechnology AG Glykosylierungs-Engineering von Antikörpern zur Verbesserung der antikörperabhängigen zellvermittelten Zytotoxizität
CA2330448A1 (en) 1998-04-28 1999-11-04 Applied Research Systems Ars Holding N.V. Peg-lhrh analog conjugates
US20030166525A1 (en) 1998-07-23 2003-09-04 Hoffmann James Arthur FSH Formulation
KR100618495B1 (ko) 1998-10-06 2006-08-31 마크 아론 에말파브 섬유상의 진균성 숙주 영역에서의 형질전환 시스템:크리소스포륨속에서
US6245539B1 (en) 1998-10-06 2001-06-12 Board Of Trustees Operating Michigan State University Human asparaginyl-tRNA synthetase DNA
EE04967B1 (et) 1998-10-16 2008-02-15 Biogen, Incorporated Glkoslitud interferoon-beeta, selle kasutamineja seda sisaldav farmatseutiline kompositsioon, meetod interferoon-beeta-1a aktiivsuse pikendamiseks ja leiutisekohase valgu valmistamiseks
US7304150B1 (en) 1998-10-23 2007-12-04 Amgen Inc. Methods and compositions for the prevention and treatment of anemia
CN1325443A (zh) 1998-10-30 2001-12-05 诺沃奇梅兹有限公司 具有减弱的变应原性的糖基化蛋白
EP1129179B1 (de) 1998-11-13 2008-09-10 Henrick Clausen Udp-galaktose: beta-i(n)-acetyl-glukosamin beta1,3 galaktosyltransferasen, beta3gal-t5
EP1131415A4 (de) 1998-11-18 2002-09-11 Neose Technologies Inc Kostengünstige herstellung von oligosacchariden
US6465220B1 (en) 1998-12-21 2002-10-15 Glycozym Aps Glycosylation using GalNac-T4 transferase
DK1157037T3 (da) 1999-01-29 2003-11-24 Hoffmann La Roche GCSF-konjugater
US6503744B1 (en) 1999-02-01 2003-01-07 National Research Council Of Canada Campylobacter glycosyltransferases for biosynthesis of gangliosides and ganglioside mimics
US6699705B2 (en) 1999-02-01 2004-03-02 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
CN1157484C (zh) 1999-04-22 2004-07-14 阿斯特拉曾尼卡有限公司 用于检测磷酸基-n-乙酰胞壁酰五肽移位酶活性的测定方法
US6365410B1 (en) 1999-05-19 2002-04-02 Genencor International, Inc. Directed evolution of microorganisms
JO2291B1 (en) * 1999-07-02 2005-09-12 اف . هوفمان لاروش ايه جي Erythropoietin derivatives
WO2001005434A2 (en) 1999-07-20 2001-01-25 Amgen Inc. Hyaluronic acid-protein conjugates
US6716626B1 (en) * 1999-11-18 2004-04-06 Chiron Corporation Human FGF-21 nucleic acids
ES2377935T3 (es) 1999-12-02 2012-04-03 Zymogenetics, Inc. Procedimiento para dirigir células que expresan receptor -3 o -2 del factor de crecimiento de fibroblastos
US6348558B1 (en) 1999-12-10 2002-02-19 Shearwater Corporation Hydrolytically degradable polymers and hydrogels made therefrom
DK1259563T4 (en) 1999-12-22 2016-11-21 Nektar Therapeutics PROCEDURE FOR PREPARING 1-BENZOTRIAZOLYL CARBONATE ESTERS OF WATER SOLUBLE POLYMERS
WO2001048052A1 (fr) 1999-12-24 2001-07-05 Kyowa Hakko Kogyo Co., Ltd. Glycols de polyalkylene ramifies
EP1246915A2 (de) 1999-12-30 2002-10-09 Maxygen Aps Verbesserte lysosomale enzyme und lysosomale enzymaktivatoren
US6646110B2 (en) 2000-01-10 2003-11-11 Maxygen Holdings Ltd. G-CSF polypeptides and conjugates
US6555660B2 (en) * 2000-01-10 2003-04-29 Maxygen Holdings Ltd. G-CSF conjugates
AU782580B2 (en) 2000-01-10 2005-08-11 Maxygen, Inc. G-CSF conjugates
US6423488B1 (en) 2000-01-15 2002-07-23 Avigenics, Inc High throughput screening assay for detecting a DNA sequence
DK1257295T3 (da) 2000-02-11 2009-08-10 Bayer Healthcare Llc Faktor VII eller VIIA-lignende molekyler
AU2001231531A1 (en) 2000-02-11 2001-08-20 Maxygen Aps Follicle stimulating hormones
AU2001232337A1 (en) 2000-02-18 2001-08-27 Kanagawa Academy Of Science And Technology Pharmaceutical composition, reagent and method for intracerebral delivery of pharmaceutically active ingredient or labeling substance
US20010041683A1 (en) 2000-03-09 2001-11-15 Schmitz Harold H. Cocoa sphingolipids, cocoa extracts containing sphingolipids and methods of making and using same
EP1263771B1 (de) * 2000-03-16 2006-06-14 The Regents Of The University Of California Chemoselektive anknüpfung durch verwendung eines phosphins
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
JP4455802B2 (ja) 2000-05-03 2010-04-21 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 ヒト第vii凝固因子変異型
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
US20020019342A1 (en) 2000-05-12 2002-02-14 Robert Bayer In vitro modification of glycosylation patterns of recombinant glycopeptides
ATE367398T1 (de) 2000-05-16 2007-08-15 Bolder Biotechnology Inc Verfahren zur rückfaltung von proteinen mit freien cysteinresten
JP2004504016A (ja) 2000-06-30 2004-02-12 マキシゲン・エイピーエス ペプチド拡張されたグリコシル化ポリペプチド
US6423826B1 (en) 2000-06-30 2002-07-23 Regents Of The University Of Minnesota High molecular weight derivatives of vitamin K-dependent polypeptides
WO2002004512A2 (en) 2000-07-10 2002-01-17 Diosynth Rtp, Inc. Purification of human troponin i
KR100396983B1 (ko) 2000-07-29 2003-09-02 이강춘 고반응성의 가지 달린 고분자 유도체 및 고분자와 단백질또는 펩타이드의 접합체
WO2002013843A2 (en) 2000-08-17 2002-02-21 University Of British Columbia Chemotherapeutic agents conjugated to p97 and their methods of use in treating neurological tumours
AU2001285020A1 (en) 2000-08-17 2002-02-25 Synapse Technologies, Inc. P97-active agent conjugates and their methods of use
BR0114373A (pt) 2000-10-02 2004-02-17 Novo Nordisk As Célula hospedeira eucariótica, métodos para a produção de fvii e de fviia ou de suas variantes, de uma proteìna dependente de vitamina k e de uma célula hospedeira eucariótica produtora de fvii ou de seu análogo, vetor recombinante, fvii e fviia recombinante ou suas variantes, e, proteìna dependente de vitamina k recombinante
US20020142964A1 (en) 2000-11-02 2002-10-03 Nissen Torben Lauesgaard Single-chain polypeptides
AU2002229603B2 (en) 2000-11-27 2007-11-08 Rmf Dictagene S.A. Process for folding chemically synthesized polypeptides
AU2002217944A1 (en) 2000-11-28 2002-06-11 University Of Massachusetts Methods and reagents for introducing a sulfhydryl group into the 5'-terminus of rna
AU3323002A (en) 2000-12-20 2002-07-01 Hoffmann La Roche Erythropoietin conjugates
EP1345628B1 (de) * 2000-12-20 2011-04-13 F. Hoffmann-La Roche AG 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
EP1366075B1 (de) 2001-02-27 2009-05-27 Maxygen Aps Neue interferon-beta-ähnliche moleküle
CZ20032454A3 (en) 2001-03-22 2004-03-17 Novo Nordisk Health Care Ag Coagulation factor vii derivative
US7235638B2 (en) 2001-03-22 2007-06-26 Novo Nordisk Healthcare A/G Coagulation factor VII derivatives
DK1383785T3 (da) 2001-05-03 2011-05-23 Merck Patent Gmbh Rekombinant tumorspecifikt antistof og anvendelse deraf
US7271150B2 (en) 2001-05-14 2007-09-18 United States Of America, Represented By The Secretary, Department Of Health And Human Services Modified growth hormone
ATE468862T1 (de) 2001-07-11 2010-06-15 Maxygen Inc G-csf konjugate
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
CA2455347A1 (en) 2001-08-01 2003-02-13 Neose Technologies, Inc. Neutral glycosphingolipids and glycosyl-sphingosines and methods for isolating the same
JP2005527467A (ja) 2001-08-29 2005-09-15 ネオーズ テクノロジーズ, インコーポレイテッド 新規な合成ガングリオシド誘導体およびその組成物
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
US7265085B2 (en) 2001-10-10 2007-09-04 Neose Technologies, Inc. Glycoconjugation methods and proteins/peptides produced by the methods
US7226903B2 (en) 2001-10-10 2007-06-05 Neose Technologies, Inc. Interferon beta: remodeling and glycoconjugation of interferon beta
US7173003B2 (en) * 2001-10-10 2007-02-06 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
US7157277B2 (en) 2001-11-28 2007-01-02 Neose Technologies, Inc. Factor VIII remodeling and glycoconjugation of Factor VIII
US7265084B2 (en) * 2001-10-10 2007-09-04 Neose Technologies, Inc. Glycopegylation methods and proteins/peptides produced by the methods
US7439043B2 (en) 2001-10-10 2008-10-21 Neose Technologies, Inc. Galactosyl nucleotide sugars
US7399613B2 (en) * 2001-10-10 2008-07-15 Neose Technologies, Inc. Sialic acid nucleotide sugars
US7179617B2 (en) 2001-10-10 2007-02-20 Neose Technologies, Inc. Factor IX: remolding and glycoconjugation of Factor IX
US7297511B2 (en) * 2001-10-10 2007-11-20 Neose Technologies, Inc. Interferon alpha: remodeling and glycoconjugation of interferon alpha
US6784154B2 (en) 2001-11-01 2004-08-31 University Of Utah Research Foundation Method of use of erythropoietin to treat ischemic acute renal failure
US7473680B2 (en) * 2001-11-28 2009-01-06 Neose Technologies, Inc. Remodeling and glycoconjugation of peptides
CA2468295C (en) 2001-11-28 2010-08-03 Neose Technologies, Inc. Glycoprotein remodeling using endoglycanases
US20060035224A1 (en) * 2002-03-21 2006-02-16 Johansen Jack T Purification methods for oligonucleotides and their analogs
AU2003228380A1 (en) 2002-03-27 2003-10-13 Molex Incorporated Differential signal connector assembly with improved retention capabilities
AU2003239365A1 (en) 2002-05-03 2003-11-17 Neose Technologies, Inc. Recombinant glycosyltransferase fusion proteins
MXPA04012496A (es) 2002-06-21 2005-09-12 Novo Nordisk Healthcare Ag Glicoformos del factor vii pegilados.
CN101301475B (zh) 2002-06-21 2012-12-19 诺和诺德医疗保健公司 Peg化因子vii糖型
DE10232916B4 (de) 2002-07-19 2008-08-07 Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. Vorrichtung und Verfahren zum Charakterisieren eines Informationssignals
JP2005533525A (ja) 2002-07-23 2005-11-10 ニオズ テクノロジーズ インコーポレイテッド 細菌グリコシルトランスフェラーゼを用いたオリゴ糖、糖脂質、及び糖タンパク質の合成
WO2004013151A2 (en) 2002-08-01 2004-02-12 National Research Council Of Canada Campylobacter glycans and glycopeptides
SI1524993T1 (sl) 2002-08-02 2013-07-31 Glaxosmithkline Biologicals S.A. Neisserijski vakcinski sestavki, ki obsegajo kombinacijo antigenov
JP2006501820A (ja) 2002-09-06 2006-01-19 バイエル・フアーマシユーチカルズ・コーポレーシヨン 修飾glp−1受容体アゴニストおよびそれらの薬理学的使用法
WO2005025606A1 (en) 2003-09-09 2005-03-24 Warren Pharmaceuticals, Inc. Long acting erythropoietins that maintain tissue protective activity of endogenous erythropoietin
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
EP1546202B1 (de) 2002-09-25 2007-08-22 Novo Nordisk Health Care AG Varianten des menschliche koagulationsfaktors vii
AU2003266931B2 (en) 2002-09-30 2010-01-21 Bayer Healthcare Llc FVII or FVIIa variants having increased clotting activity
US20040062748A1 (en) 2002-09-30 2004-04-01 Mountain View Pharmaceuticals, Inc. Polymer conjugates with decreased antigenicity, methods of preparation and uses thereof
NZ539415A (en) 2002-10-09 2008-12-24 Neose Technologies Inc Remodelling and glycoconjugation of erythropoietin
DE60314658D1 (de) * 2002-11-08 2007-08-09 Glycozym Aps Verfahren zur identifizierung von die funktionen von polypeptid-galnac-transferasen modulierenden agentien, solche agentien umfassende pharmazeutische zusammensetzungen und verwendung solcher agentien zur herstellung von arzneimitteln
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
US20050064540A1 (en) * 2002-11-27 2005-03-24 Defrees Shawn Ph.D Glycoprotein remodeling using endoglycanases
EP1424344A1 (de) 2002-11-29 2004-06-02 Aventis Behring Gesellschaft mit beschränkter Haftung Modifizierte cDNA des Factors VIII und Derivate davon
ES2312523T3 (es) 2002-12-13 2009-03-01 Bioceuticals Arzneimittel Ag Procedimiento para la produccion y purificacion de eritropoyetina.
EP1667708B9 (de) 2002-12-26 2012-10-31 Mountain View Pharmaceuticals, Inc. INTERFERON-BETA-1b POLYETHYLENEGLYKOL-KONJUGATE MIT ERHÖHTER IN VITRO BIOLOGISCHER WIRKSAMKEIT
US7041635B2 (en) 2003-01-28 2006-05-09 In2Gen Co., Ltd. Factor VIII polypeptide
EP2572732A1 (de) 2003-02-26 2013-03-27 Nektar Therapeutics Konjugate mit Polymer-Faktor-VIII-Anteil
CA2519092C (en) 2003-03-14 2014-08-05 Neose Technologies, Inc. Branched water-soluble polymers and their conjugates
WO2004083259A2 (en) 2003-03-18 2004-09-30 Neose Technologies Inc. Activated forms of water-soluble polymers
AU2004231461B2 (en) 2003-03-19 2009-11-12 Eli Lilly And Company Polyethelene glycol linked GLP-1 compounds
US20070026485A1 (en) 2003-04-09 2007-02-01 Neose Technologies, Inc. Glycopegylation methods and proteins/peptides produced by the methods
US7691603B2 (en) * 2003-04-09 2010-04-06 Novo Nordisk A/S Intracellular formation of peptide conjugates
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
ATE497783T1 (de) 2003-05-06 2011-02-15 Syntonix Pharmaceuticals Inc Gerinnungsfaktor vii-fc chimäre proteine zur behandlung von hämostatischen krankheiten
TWI353991B (en) 2003-05-06 2011-12-11 Syntonix Pharmaceuticals Inc Immunoglobulin chimeric monomer-dimer hybrids
BRPI0410164A (pt) * 2003-05-09 2006-05-16 Neose Technologies Inc composições e métodos para preparação de mutantes de glicosilação de hormÈnio do crescimento humano
WO2004101597A2 (en) 2003-05-13 2004-11-25 Frutarom Ltd. Methods for the reduction of disulfide bonds
US7074755B2 (en) 2003-05-17 2006-07-11 Centocor, Inc. Erythropoietin conjugate compounds with extended half-lives
EP1481985A1 (de) 2003-05-28 2004-12-01 Innogenetics N.V. Modifiziertes Hepatitis C Virus (HCV) NS3 Protein zur medizinischen Behandlung
GB0315457D0 (en) 2003-07-01 2003-08-06 Celltech R&D Ltd Biological products
US9005625B2 (en) * 2003-07-25 2015-04-14 Novo Nordisk A/S Antibody toxin conjugates
JP4337457B2 (ja) * 2003-07-30 2009-09-30 日本電気株式会社 アンテナ装置及びそれを用いた無線通信装置
US20060198819A1 (en) 2003-08-08 2006-09-07 Novo Nordisk Healthcare A/G Use of galactose oxidase for selective chemical conjugation of protractor molecules to proteins of therapeutic interest
WO2005014024A2 (en) 2003-08-08 2005-02-17 Fresenius Kabi Deutschland Gmbh Conjugates of a polymer and a protein linked by an oxime linking group
EP1654004A2 (de) 2003-08-08 2006-05-10 Novo Nordisk A/S Synthese und applikation von neuen strukturellen gut definierten verzweigten polymeren als konjugatmittel für peptide
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
US7842661B2 (en) 2003-11-24 2010-11-30 Novo Nordisk A/S Glycopegylated erythropoietin formulations
US20080318850A1 (en) 2003-12-03 2008-12-25 Neose Technologies, Inc. Glycopegylated Factor Ix
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
US7956032B2 (en) * 2003-12-03 2011-06-07 Novo Nordisk A/S Glycopegylated granulocyte colony stimulating factor
US20060040856A1 (en) 2003-12-03 2006-02-23 Neose Technologies, Inc. Glycopegylated factor IX
NZ547554A (en) 2003-12-03 2009-09-25 Biogenerix Ag Glycopegylated granulocyte colony stimulating factor
US20080058245A1 (en) 2004-01-09 2008-03-06 Johnson Karl F Vectors for Recombinant Protein Expression in 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
EP1727567B1 (de) 2004-02-12 2013-07-03 Archemix LLC Zur Behandlung von Erkrankungen in Verbindung mit dem Komplementsystem geeignete Aptamer-Therapeutika
EP1735340A2 (de) 2004-03-17 2006-12-27 Eli Lilly And Company Glykolgebundene fgf-21-verbindungen
US20070037966A1 (en) 2004-05-04 2007-02-15 Novo Nordisk A/S Hydrophobic interaction chromatography purification of factor VII polypeptides
RU2006138181A (ru) 2004-05-04 2008-06-10 Ново Нордиск Хелс Кеа Аг (Ch) О-связанные гликоформы полипептидов и способ их изготовления
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
WO2005121332A2 (en) 2004-06-03 2005-12-22 Neose Technologies, Inc. Truncated st6galnaci polypeptides and nucleic acids
JP2008512085A (ja) 2004-06-03 2008-04-24 ネオス テクノロジーズ インコーポレイティッド 切断型GalNAcT2ポリペプチドおよび核酸
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
JP2008505119A (ja) 2004-06-30 2008-02-21 ネクター セラピューティクス アラバマ,コーポレイション 高分子−第ix因子部分の抱合体
AU2005270092A1 (en) 2004-07-02 2006-02-09 Cerami, Anthony Method of producing fully carbamylated erythropoietin
WO2006014466A2 (en) 2004-07-02 2006-02-09 The Kenneth S. Warren Institute, Inc. Novel carbamylated epo and method for its production
US20080300173A1 (en) 2004-07-13 2008-12-04 Defrees Shawn Branched Peg Remodeling and Glycosylation of Glucagon-Like Peptides-1 [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
US20090176967A1 (en) 2004-08-02 2009-07-09 Novo Nordisk Healthcare A/G Conjugation of FVII
DK1781782T3 (da) 2004-08-17 2010-08-23 Csl Behring Gmbh Modificerede vitamin K-afhængige polypeptider
EP1799249A2 (de) 2004-09-10 2007-06-27 Neose Technologies, Inc. Glycopegyliertes interferon alpha
JP2008514215A (ja) * 2004-09-29 2008-05-08 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 改変タンパク質
US20080176790A1 (en) 2004-10-29 2008-07-24 Defrees Shawn Remodeling and Glycopegylation of Fibroblast Growth Factor (Fgf)
NZ555032A (en) 2004-11-12 2010-02-26 Bayer Healthcare Llc Site-directed modification of FVIII
US20090054623A1 (en) * 2004-12-17 2009-02-26 Neose Technologies, Inc. Lipo-Conjugation of Peptides
JP2008525032A (ja) 2004-12-22 2008-07-17 アンブレツクス・インコーポレイテツド 組換えヒト成長ホルモンを発現及び精製するための方法
US20100009902A1 (en) * 2005-01-06 2010-01-14 Neose Technologies, Inc. Glycoconjugation Using Saccharyl Fragments
EP1858543B1 (de) 2005-01-10 2013-11-27 BioGeneriX AG Glycopegylierten Granulocyten-Kolonie-stimulierender Faktor
WO2006078645A2 (en) 2005-01-19 2006-07-27 Neose Technologies, Inc. Heterologous polypeptide expression using low multiplicity of infection of viruses
PA8660701A1 (es) 2005-02-04 2006-09-22 Pfizer Prod Inc Agonistas de pyy y sus usos
US20100143969A1 (en) 2005-03-24 2010-06-10 Neose Technologies, Inc. Expression of soluble, active eukaryotic glycosyltransferases in prokaryotic organisms
EP1866427A4 (de) 2005-03-30 2010-09-01 Novo Nordisk As Herstellungsverfahren zur produktion von in insektenzelllinien entstandenen peptiden
EP1871801A2 (de) 2005-04-01 2008-01-02 Novo Nordisk Health Care AG Analoga des blutgerinnungsfaktors fviii
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
DK2311972T3 (en) 2005-05-11 2015-04-27 Eth Zuerich Recombinant N-glycosylated proteins from prokaryotic cells
US20110003744A1 (en) 2005-05-25 2011-01-06 Novo Nordisk A/S Glycopegylated Erythropoietin Formulations
US20080255026A1 (en) 2005-05-25 2008-10-16 Glycopegylated Factor 1X Glycopegylated Factor Ix
WO2006134173A2 (en) 2005-06-17 2006-12-21 Novo Nordisk Health Care Ag Selective reduction and derivatization of engineered proteins comprising at least one non-native cysteine
CN102719508A (zh) 2005-08-19 2012-10-10 诺和诺德公司 糖基聚乙二醇化的因子vii和因子viia
WO2007031559A2 (en) * 2005-09-14 2007-03-22 Novo Nordisk Health Care Ag Human coagulation factor vii polypeptides
US20090048440A1 (en) * 2005-11-03 2009-02-19 Neose Technologies, Inc. Nucleotide Sugar Purification Using Membranes
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
RU2008145084A (ru) 2006-05-24 2010-06-27 Ново Нордиск Хелс Кеа Аг (Ch) Аналоги фактора ix, имеющие пролонгированное время полужизни in vivo
CN101516388B (zh) 2006-07-21 2012-10-31 诺和诺德公司 通过o-联糖基化序列的肽的糖基化
ITMI20061624A1 (it) * 2006-08-11 2008-02-12 Bioker Srl Mono-coniugati sito-specifici di g-csf
JP5570809B2 (ja) * 2006-09-01 2014-08-13 ノボ ノルディスク ヘルス ケア アーゲー 修飾タンパク質
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
CA2665480C (en) 2006-10-04 2019-11-12 Shawn Defrees Glycerol linked pegylated sugars and glycopeptides
WO2008073620A2 (en) 2006-11-02 2008-06-19 Neose Technologies, Inc. Manufacturing process for the production of polypeptides expressed in insect cell-lines
DK2144923T3 (da) 2007-04-03 2013-05-13 Biogenerix Ag Behandlingsfremgangsmåder under anvendelse af glycopegyleret g-csf
US20090053167A1 (en) * 2007-05-14 2009-02-26 Neose Technologies, Inc. C-, S- and N-glycosylation of peptides
JP2010531135A (ja) 2007-06-04 2010-09-24 ノボ ノルディスク アクティーゼルスカブ 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
ES2551123T3 (es) 2007-06-12 2015-11-16 Ratiopharm Gmbh Proceso mejorado para la producción de azúcares de nucleótido
US8207112B2 (en) 2007-08-29 2012-06-26 Biogenerix Ag Liquid formulation of G-CSF conjugate
JP5647899B2 (ja) 2008-01-08 2015-01-07 ラツィオファルム ゲーエムベーハーratiopharm GmbH オリゴサッカリルトランスフェラーゼを使用するポリペプチドの複合糖質化

Patent Citations (76)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4055635A (en) * 1973-07-05 1977-10-25 Beecham Group Limited Fibrinolytic compositions
US4179337A (en) * 1973-07-20 1979-12-18 Davis Frank F Non-immunogenic polypeptides
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
US4414147A (en) * 1981-04-17 1983-11-08 Massachusetts Institute Of Technology Methods of decreasing the hydrophobicity of fibroblast and other interferons
US4412989A (en) * 1981-06-10 1983-11-01 Ajinomoto Company Incorporated Oxygen carrier
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
US4879236A (en) * 1984-05-16 1989-11-07 The Texas A&M University System Method for producing a recombinant baculovirus expression vector
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
US5153265A (en) * 1988-01-20 1992-10-06 Cetus Corporation Conjugation of polymer to colony stimulating factor-1
US5147788A (en) * 1988-05-06 1992-09-15 Burroughs Wellcome Co. Baculovirus vectors and methods of use
US5169933A (en) * 1988-08-15 1992-12-08 Neorx Corporation Covalently-linked complexes and methods for enhanced cytotoxicity and imaging
US5194376A (en) * 1989-02-28 1993-03-16 University Of Ottawa Baculovirus expression system capable of producing foreign gene proteins at high levels
US5154924A (en) * 1989-09-07 1992-10-13 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
US5527527A (en) * 1989-09-07 1996-06-18 Alkermes, Inc. Transferrin receptor specific antibody-neuropharmaceutical agent conjugates
US5977307A (en) * 1989-09-07 1999-11-02 Alkermes, Inc. Transferrin receptor specific ligand-neuropharmaceutical agent fusion proteins
US5833988A (en) * 1989-09-07 1998-11-10 Alkermes, Inc. Transferrin receptor specific antibody-neuropharmaceutical or diagnostic agent conjugates
US5672683A (en) * 1989-09-07 1997-09-30 Alkermes, Inc. Transferrin neuropharmaceutical agent fusion protein
US5405753A (en) * 1990-03-26 1995-04-11 Brossmer; Reinhard CMP-activated, fluorescing sialic acids, as well as processes for their preparation
US5352670A (en) * 1991-06-10 1994-10-04 Alberta Research Council Methods for the enzymatic synthesis of alpha-sialylated oligosaccharide glycosides
US5346696A (en) * 1991-06-19 1994-09-13 Korea Green Cross Corporation Asialoglycoprotein - conjugated medicinal agent
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
US6166183A (en) * 1992-11-30 2000-12-26 Kirin-Amgen, Inc. Chemically-modified G-CSF
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
US6096512A (en) * 1993-05-14 2000-08-01 Pharmacia & Upjohn Company Cloned DNA encoding a UDP-GalNAc: Polypeptide, N-acetylgalactosaminyltransferase
US5621039A (en) * 1993-06-08 1997-04-15 Hallahan; Terrence W. Factor IX- polymeric conjugates
US5969040A (en) * 1993-06-08 1999-10-19 Hallahan; Terrence W. Factor IX -- polymeric conjugates
US5635603A (en) * 1993-12-08 1997-06-03 Immunomedics, Inc. Preparation and use of immunoconjugates
US5369017A (en) * 1994-02-04 1994-11-29 The Scripps Research Institute Process for solid phase glycopeptide synthesis
US5492841A (en) * 1994-02-18 1996-02-20 E. I. Du Pont De Nemours And Company Quaternary ammonium immunogenic conjugates and immunoassay reagents
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
US5545553A (en) * 1994-09-26 1996-08-13 The Rockefeller University Glycosyltransferases for biosynthesis of oligosaccharides, and genes encoding them
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
US5922577A (en) * 1995-04-11 1999-07-13 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
US5728554A (en) * 1995-04-11 1998-03-17 Cytel Corporation Enzymatic synthesis of glycosidic linkages
US6015555A (en) * 1995-05-19 2000-01-18 Alkermes, Inc. Transferrin receptor specific antibody-neuropharmaceutical or diagnostic agent conjugates
US5849535A (en) * 1995-09-21 1998-12-15 Genentech, Inc. Human growth hormone variants
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
US6087325A (en) * 1996-10-15 2000-07-11 The Liposome Company, Inc. Peptide-lipid conjugates
US5945314A (en) * 1997-03-31 1999-08-31 Abbott Laboratories Process for synthesizing oligosaccharides
US6183738B1 (en) * 1997-05-12 2001-02-06 Phoenix Pharamacologics, Inc. Modified arginine deiminase
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
US6440703B1 (en) * 1997-12-01 2002-08-27 Neose Technologies, Inc. Enzymatic synthesis of gangliosides
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
US6692931B1 (en) * 1998-11-16 2004-02-17 Werner Reutter Recombinant glycoproteins, method for the production thereof, medicaments containing said glycoproteins and use thereof
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
US20020150981A1 (en) * 1999-09-14 2002-10-17 Novazyme Pharmaceuticals, Inc. Methods for producing highly phosphorylated lysosomal hydrolases
US7202208B2 (en) * 2000-05-15 2007-04-10 Hoffman-La Roche Inc. Erythropoietin composition
US20020137134A1 (en) * 2000-06-28 2002-09-26 Gerngross Tillman U. Methods for producing modified glycoproteins
US6531121B2 (en) * 2000-12-29 2003-03-11 The Kenneth S. Warren Institute, Inc. Protection and enhancement of erythropoietin-responsive cells, tissues and organs
US20020168323A1 (en) * 2001-05-11 2002-11-14 Igor Gonda Optimization of the molecular properties and formulation of proteins delivered by inhalation
US7138371B2 (en) * 2001-10-10 2006-11-21 Neose Technologies, Inc Remodeling and glycoconjugation of peptides
US7214660B2 (en) * 2001-10-10 2007-05-08 Neose Technologies, Inc. Erythropoietin: remodeling and glycoconjugation of erythropoietin
US20040063911A1 (en) * 2001-10-10 2004-04-01 Neose Technologies, Inc. Protein remodeling methods and proteins/peptides produced by the methods
US20070027068A1 (en) * 2001-10-10 2007-02-01 Defrees Shawn Erythropoietin: remodeling and glycoconjugation of erythropoietin
US20040137557A1 (en) * 2001-10-10 2004-07-15 Neose Technologies, Inc. Remodeling and glycoconjugation of peptides
US7125843B2 (en) * 2001-10-19 2006-10-24 Neose Technologies, Inc. Glycoconjugates including more than one peptide
US20050118672A1 (en) * 2001-11-28 2005-06-02 Defrees Shawn Glycopeptide remodeling using amidases
US20040136955A1 (en) * 2002-09-05 2004-07-15 Barker Nicholas P Modified asialo-interferons and uses thereof
US20060111279A1 (en) * 2003-11-24 2006-05-25 Neose Technologies, Inc. Glycopegylated erythropoietin
US20050143292A1 (en) * 2003-11-24 2005-06-30 Defrees Shawn Glycopegylated erythropoietin
US7405198B2 (en) * 2003-11-24 2008-07-29 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
US20070105755A1 (en) * 2005-10-26 2007-05-10 Neose Technologies, Inc. One pot desialylation and glycopegylation of therapeutic peptides

Cited By (86)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080187955A1 (en) * 2001-10-10 2008-08-07 Neose Technologies, Inc. Erythropoietin: remodeling and glycoconjugation of erythropoietin
US8008252B2 (en) 2001-10-10 2011-08-30 Novo Nordisk A/S Factor VII: remodeling and glycoconjugation of Factor VII
US8716239B2 (en) 2001-10-10 2014-05-06 Novo Nordisk A/S Granulocyte colony stimulating factor: remodeling and glycoconjugation G-CSF
US8716240B2 (en) 2001-10-10 2014-05-06 Novo Nordisk A/S Erythropoietin: remodeling and glycoconjugation of erythropoietin
US8076292B2 (en) 2001-10-10 2011-12-13 Novo Nordisk A/S Factor VIII: remodeling and glycoconjugation of factor VIII
US20080050772A1 (en) * 2001-10-10 2008-02-28 Neose Technologies, Inc. Granulocyte colony stimulating factor: remodeling and glycoconjugation of G-CSF
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
US8247381B2 (en) 2003-03-14 2012-08-21 Biogenerix Ag Branched water-soluble polymers and their conjugates
US8853161B2 (en) 2003-04-09 2014-10-07 Novo Nordisk A/S Glycopegylation methods and proteins/peptides produced by the methods
US8791070B2 (en) 2003-04-09 2014-07-29 Novo Nordisk A/S Glycopegylated factor IX
US8063015B2 (en) 2003-04-09 2011-11-22 Novo Nordisk A/S Glycopegylation methods and proteins/peptides produced by the methods
US20100048456A1 (en) * 2003-04-09 2010-02-25 Novo Nordisk A/S Glycopegylation methods and proteins/peptides produced by the methods
US7932364B2 (en) 2003-05-09 2011-04-26 Novo Nordisk A/S Compositions and methods for the preparation of human growth hormone glycosylation mutants
US20080102083A1 (en) * 2003-05-09 2008-05-01 Neose Technologies, Inc. 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
US8916360B2 (en) 2003-11-24 2014-12-23 Novo Nordisk A/S Glycopegylated erythropoietin
US8633157B2 (en) 2003-11-24 2014-01-21 Novo Nordisk A/S Glycopegylated erythropoietin
US20060111279A1 (en) * 2003-11-24 2006-05-25 Neose Technologies, Inc. Glycopegylated erythropoietin
US20100210507A9 (en) * 2003-11-24 2010-08-19 Novo Nordisk A/S Glycopegylated erythropoietin
US20080300175A1 (en) * 2003-11-24 2008-12-04 Neose Technologies, Inc. Glycopegylated erythropoietin
US8632770B2 (en) 2003-12-03 2014-01-21 Novo Nordisk A/S Glycopegylated factor IX
US20080015142A1 (en) * 2003-12-03 2008-01-17 Defrees Shawn Glycopegylated Follicle Stimulating Hormone
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
US8361961B2 (en) 2004-01-08 2013-01-29 Biogenerix Ag O-linked glycosylation of peptides
US20080242846A1 (en) * 2004-01-08 2008-10-02 Neose Technologies, Inc. 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
US9200049B2 (en) 2004-10-29 2015-12-01 Novo Nordisk A/S 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
US20080176790A1 (en) * 2004-10-29 2008-07-24 Defrees Shawn Remodeling and Glycopegylation of Fibroblast Growth Factor (Fgf)
US9029331B2 (en) 2005-01-10 2015-05-12 Novo Nordisk A/S Glycopegylated granulocyte colony stimulating factor
US20090203579A1 (en) * 2005-01-10 2009-08-13 Defrees Shawn 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
US20110003744A1 (en) * 2005-05-25 2011-01-06 Novo Nordisk A/S Glycopegylated Erythropoietin Formulations
US8404809B2 (en) 2005-05-25 2013-03-26 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
US20100330645A1 (en) * 2005-08-19 2010-12-30 Novo Nordisk A/S One pot desialylation and glycopegylation of therapeutic peptides
US8841439B2 (en) 2005-11-03 2014-09-23 Novo Nordisk A/S Nucleotide sugar purification using membranes
US20090048440A1 (en) * 2005-11-03 2009-02-19 Neose Technologies, Inc. Nucleotide Sugar Purification Using Membranes
US9605050B2 (en) * 2006-03-27 2017-03-28 University Of Maryland, Baltimore Glycoprotein synthesis and remodeling by enzymatic transglycosylation
US20150087811A1 (en) * 2006-03-27 2015-03-26 University Of Maryland, Baltimore Glycoprotein synthesis and remodeling by enzymatic transglycosylation
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
US20080146782A1 (en) * 2006-10-04 2008-06-19 Neose Technologies, Inc. Glycerol linked pegylated sugars and glycopeptides
US20100120666A1 (en) * 2007-04-03 2010-05-13 Biogenerix Ag Methods of treatment using glycopegylated g-csf
US9050304B2 (en) 2007-04-03 2015-06-09 Ratiopharm Gmbh Methods of treatment using glycopegylated G-CSF
WO2008151258A3 (en) * 2007-06-04 2009-02-12 Neose Technologies Inc O-linked glycosylation using n-acetylglucosaminyl transferases
US20110177029A1 (en) * 2007-06-04 2011-07-21 Novo Nordisk A/S O-linked glycosylation using n-acetylglucosaminyl transferases
US20100174059A1 (en) * 2007-06-12 2010-07-08 Novo Nordisk A/S Process for the production of nucleotide sugars
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
US20090143292A1 (en) * 2007-08-29 2009-06-04 Neose Technologies, Inc. Liquid Formulation of G-CSF Conjugate
US8207112B2 (en) 2007-08-29 2012-06-26 Biogenerix Ag Liquid formulation of G-CSF conjugate
WO2009089154A3 (en) * 2008-01-03 2010-01-14 Cornell Research Foundation, Inc. Glycosylated protein expression in prokaryotes
KR101589554B1 (ko) 2008-01-03 2016-02-01 코넬 리서치 파운데이션 인코포레이티드 원핵생물에서의 글리코실화 단백질 발현
US8999668B2 (en) 2008-01-03 2015-04-07 Cornell Research Foundation, Inc. Glycosylated protein expression in prokaryotes
CN101960017A (zh) * 2008-01-03 2011-01-26 康乃尔研究基金会有限公司 原核生物中的糖基化蛋白表达
US9512434B2 (en) 2008-01-03 2016-12-06 Cornell Research Foundation, Inc. Glycosylated protein expression in prokaryotes
US20110039729A1 (en) * 2008-01-03 2011-02-17 Cornell Research Foundation, Inc. Glycosylated protein expression in prokaryotes
KR20100108420A (ko) * 2008-01-03 2010-10-06 코넬 리서치 파운데이션 인코포레이티드 원핵생물에서의 글리코실화 단백질 발현
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
US8945897B2 (en) 2010-07-26 2015-02-03 Baxter International Inc. Materials and methods for conjugating a water soluble fatty acid derivative to a protein
EP3505186A1 (de) 2010-07-30 2019-07-03 Baxalta GmbH Nukleophile katalysatoren für oxim-verbindung
WO2012016131A1 (en) 2010-07-30 2012-02-02 Baxter International Inc. Nucleophilic catalysts for oxime linkage
EP4023247A1 (de) 2010-07-30 2022-07-06 Takeda Pharmaceutical Company Limited Nukleophile katalysatoren für oxim-verbindung
WO2012087838A1 (en) 2010-12-22 2012-06-28 Baxter International Inc. Materials and methods for conjugating a water soluble fatty acid derivative to a protein
EP3673921A1 (de) 2010-12-22 2020-07-01 Baxalta GmbH Materialien und verfahren zum konjugieren eines wasserlöslichen fettsäurederivats mit einem protein
EP3808378A1 (de) 2011-05-27 2021-04-21 Baxalta GmbH Therapeutische proteine mit erhöhter halbwertzeit und verfahren zur herstellung davon
EP3412314A1 (de) 2011-05-27 2018-12-12 Baxalta GmbH Mit polysialsäure konjugierte therapeutische proteine und verfahren zur herstellung davon
WO2012166622A1 (en) 2011-05-27 2012-12-06 Baxter International Inc. Therapeutic proteins with increased half-life and methods of preparing same
WO2013173543A1 (en) 2012-05-16 2013-11-21 Baxter International Inc. Nucleophilic catalysts for oxime linkage
WO2013173557A1 (en) 2012-05-16 2013-11-21 Baxter International Inc. Nucleophilic catalysts for oxime linkage and use of nmr analyses of the same
EP4019049A1 (de) 2012-05-16 2022-06-29 Takeda Pharmaceutical Company Limited Verfahren zur herstellung von therapeutisches protein-polymer-konjugate
US10322192B2 (en) 2016-03-02 2019-06-18 Eisai R&D Management Co., Ltd. Eribulin-based antibody-drug conjugates and methods of use
US10548986B2 (en) 2016-03-02 2020-02-04 Eisai R&D Management Co., Ltd. Eribulin-based antibody-drug conjugates and methods of use
US11291680B2 (en) 2016-12-15 2022-04-05 Société des Produits Nestlé S.A. Compositions and methods that modulate white blood cells or neutrophils in a companion animal
US11898187B2 (en) 2017-08-15 2024-02-13 Northwestern University Protein glycosylation sites by rapid expression and characterization of N-glycosyltransferases
US11530432B2 (en) 2018-03-19 2022-12-20 Northwestern University Compositions and methods for rapid in vitro synthesis of bioconjugate vaccines in vitro via production and N-glycosylation of protein carriers in detoxified prokaryotic cell lysates
US11725224B2 (en) 2018-04-16 2023-08-15 Northwestern University Methods for co-activating in vitro non-standard amino acid (nsAA) incorporation and glycosylation in crude cell lysates

Also Published As

Publication number Publication date
EP1615945A4 (de) 2007-10-17
WO2004099231A3 (en) 2006-03-16
CY1114626T1 (el) 2016-10-05
US20170007712A1 (en) 2017-01-12
US20140294762A1 (en) 2014-10-02
US20150343080A1 (en) 2015-12-03
BE2014C003I2 (de) 2023-08-09
NZ542586A (en) 2009-11-27
IL171109A (en) 2015-09-24
US8063015B2 (en) 2011-11-22
EP2338333A3 (de) 2011-07-27
AU2004236174B2 (en) 2011-06-02
EP2338333A2 (de) 2011-06-29
SG155777A1 (en) 2009-10-29
US20120220517A1 (en) 2012-08-30
AU2004236174A1 (en) 2004-11-18
EP2055189A1 (de) 2009-05-06
EP2338333B1 (de) 2017-09-06
JP2007525941A (ja) 2007-09-13
CA2522345A1 (en) 2004-11-18
PL1615945T3 (pl) 2012-03-30
WO2004099231A2 (en) 2004-11-18
JP4674702B2 (ja) 2011-04-20
US20100048456A1 (en) 2010-02-25
EP1615945B1 (de) 2011-09-28
LU92358I2 (fr) 2014-03-24
EP1615945A2 (de) 2006-01-18
US8853161B2 (en) 2014-10-07
MXPA05010773A (es) 2005-12-12

Similar Documents

Publication Publication Date Title
US8853161B2 (en) Glycopegylation methods and proteins/peptides produced by the methods
US7265084B2 (en) Glycopegylation methods and proteins/peptides produced by the methods
US8716239B2 (en) Granulocyte colony stimulating factor: remodeling and glycoconjugation G-CSF
US7265085B2 (en) Glycoconjugation methods and proteins/peptides produced by the methods
US8008252B2 (en) Factor VII: remodeling and glycoconjugation of Factor VII
US8076292B2 (en) Factor VIII: remodeling and glycoconjugation of factor VIII
US8716240B2 (en) Erythropoietin: remodeling and glycoconjugation of erythropoietin
US7795210B2 (en) Protein remodeling methods and proteins/peptides produced by the methods
US7399613B2 (en) Sialic acid nucleotide sugars
US7439043B2 (en) Galactosyl nucleotide sugars
US7696163B2 (en) Erythropoietin: remodeling and glycoconjugation of erythropoietin
US7179617B2 (en) Factor IX: remolding and glycoconjugation of Factor IX
US7125843B2 (en) Glycoconjugates including more than one peptide
US7297511B2 (en) Interferon alpha: remodeling and glycoconjugation of interferon alpha
US7226903B2 (en) Interferon beta: remodeling and glycoconjugation of interferon beta
NZ539415A (en) Remodelling and glycoconjugation of erythropoietin

Legal Events

Date Code Title Description
AS Assignment

Owner name: NEOSE TECHNOLOGIES, INC., PENNSYLVANIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:DEFREES, SHAWN;ZOPF, DAVID A.;BAYER, ROBERT J.;AND OTHERS;REEL/FRAME:017754/0551;SIGNING DATES FROM 20060501 TO 20060506

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

STCB Information on status: application discontinuation

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