US20050238723A1 - Method of producing hydroxyalkyl starch derivatives - Google Patents

Method of producing hydroxyalkyl starch derivatives Download PDF

Info

Publication number
US20050238723A1
US20050238723A1 US11/078,098 US7809805A US2005238723A1 US 20050238723 A1 US20050238723 A1 US 20050238723A1 US 7809805 A US7809805 A US 7809805A US 2005238723 A1 US2005238723 A1 US 2005238723A1
Authority
US
United States
Prior art keywords
compound
group
functional group
hydroxyalkyl starch
reaction
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/078,098
Other languages
English (en)
Inventor
Norbert Zander
Harald Conradt
Wolfram Eichner
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.)
Fresenius Kabi Deutschland GmbH
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=31995520&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20050238723(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority claimed from EP02020425A external-priority patent/EP1400533A1/fr
Application filed by Individual filed Critical Individual
Priority to US11/078,098 priority Critical patent/US20050238723A1/en
Assigned to FRESENIUS KABI DEUTSCHLAND GMBH reassignment FRESENIUS KABI DEUTSCHLAND GMBH ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CONRADT, HARALD, EICHNER, WOLFRAM, ZANDER, NORBERT
Publication of US20050238723A1 publication Critical patent/US20050238723A1/en
Priority to US12/824,618 priority patent/US20120046240A9/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08BPOLYSACCHARIDES; DERIVATIVES THEREOF
    • C08B31/00Preparation of derivatives of starch
    • C08B31/08Ethers
    • C08B31/12Ethers having alkyl or cycloalkyl radicals substituted by heteroatoms, e.g. hydroxyalkyl or carboxyalkyl starch
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08BPOLYSACCHARIDES; DERIVATIVES THEREOF
    • C08B31/00Preparation of derivatives of starch
    • C08B31/08Ethers
    • C08B31/10Alkyl or cycloalkyl ethers
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • A61K9/0021Intradermal administration, e.g. through microneedle arrays, needleless injectors
    • 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
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • C07K14/505Erythropoietin [EPO]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/53Colony-stimulating factor [CSF]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/5412IL-6
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/55IL-2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/555Interferons [IFN]
    • C07K14/56IFN-alpha
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/555Interferons [IFN]
    • C07K14/565IFN-beta
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K17/00Carrier-bound or immobilised peptides; Preparation thereof
    • C07K17/02Peptides being immobilised on, or in, an organic carrier
    • C07K17/10Peptides being immobilised on, or in, an organic carrier the carrier being a carbohydrate
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08BPOLYSACCHARIDES; DERIVATIVES THEREOF
    • C08B31/00Preparation of derivatives of starch
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08BPOLYSACCHARIDES; DERIVATIVES THEREOF
    • C08B31/00Preparation of derivatives of starch
    • C08B31/003Crosslinking of starch
    • C08B31/006Crosslinking of derivatives of starch
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08BPOLYSACCHARIDES; DERIVATIVES THEREOF
    • C08B31/00Preparation of derivatives of starch
    • C08B31/08Ethers
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08BPOLYSACCHARIDES; DERIVATIVES THEREOF
    • C08B31/00Preparation of derivatives of starch
    • C08B31/18Oxidised starch
    • C08B31/185Derivatives of oxidised starch, e.g. crosslinked oxidised starch
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08HDERIVATIVES OF NATURAL MACROMOLECULAR COMPOUNDS
    • C08H1/00Macromolecular products derived from proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the present invention relates to hydroxyalkyl starch derivates, particularly hydroxyalkyl starch derivatives obtainable by a process in which hydroxyalkyl starch is reacted with a primary or secondary amino group of a crosslinking compound or with two crosslinking compounds wherein the resulting hydroxaylkyl starch derivative has at least one functional group X which is capable of being reacted with a functional group Y of a further compound and wherein this group Y is an aldehyd group, a keto group, a hemiacetal group, an acetal group, or a thio group.
  • the present invention relates to hydroxyalkyl starch derivatives obtainable by a process according to which hydroxyalkyl starch is reacted with a primary or secondary amino group of a crosslinking compound, the resulting reaction product optionally being further reacted with a second crosslinking compound, wherein the resulting hydroxaylkyl starch derivative has at least one functional group X which is capable of being reacted with a functional group Y of a further compound and wherein this group Y is an aldehyd group, a keto group, a hemiacetal group, an acetal group, or a thio group, and the resulting reaction product is reacted with a polypeptide, preferably with a polypeptide such as AT III, IFN-beta or erythropoietin and especially preferably with erythropoietin, which comprises at least one of these functional groups Y.
  • a polypeptide such as AT III, IFN-beta or erythro
  • a hydroxyalkyl starch which is especially preferred is hydroxyethyl starch.
  • the hydroxyalkyl starch and preferably the hydroxyl ethyl starch is reacted with the linker compound at its reducing end which is optionally oxidized prior to said reaction.
  • HES Hydroxyethyl starch
  • Amylopectin consists of glucose moieties, wherein in the main chain alpha-1,4-glycosidic bonds are present and at the branching sites alpha-1,6-glycosidic bonds are found.
  • the physical-chemical properties of this molecule are mainly determined by the type of glycosidic bonds. Due to the nicked alpha-1,4-glycosidic bond, helical structures with about six glucose-monomers per turn are produced.
  • the physicochemical as well as the biochemical properties of the polymer can be modified via substitution.
  • the introduction of a hydroxyethyl group can be achieved via alkaline hydroxyethylation.
  • DE 26 16 086 discloses the conjugation of hemoglobin to hydroxyethyl starch wherein, in a first step, a cross-linking agent, e.g. bromocyane, is bound to hydroxyethyl starch and subsequently hemoglobin is linked to the intermediate product.
  • a cross-linking agent e.g. bromocyane
  • HES HES-like styrene-styrene-styrene-styrene-styrene-styrene-styrene-styrene-styrene-styrene-styrene-styrene-styrene-styrene-styrene-styrene-styrene-styl-N-(2-aminoietin)-2-linked glycoprotein
  • red blood cells in the circulation erythropoietin
  • WO 94/28024 discloses that physiologically active polypeptides modified with polyethyleneglycol (PEG) exhibit reduced immunogenicity and antigenicity and circulate in the bloodstream considerably longer than unconjugated proteins, i.e. have a longer clearance rate.
  • PEG-drug conjugates exhibit several disadvantages, e.g. they do not exhibit a natural structure which can be recognized by elements of in vivo degradation pathways. Therefore, apart from PEG-conjugates, other conjugates and protein polymerates have been produced.
  • a plurality of methods for the cross-linking of different proteins and macromolecules such as polymerase have been described in the literature (see e.g. Wong, Chemistry of protein conjugation and cross-linking, 1993, CRCS, Inc.).
  • WO 02/08079 A2 discloses compounds comprising a conjugate of an active agent and a hydroxyalkyl starch wherein active agent and hydroxyalkyl starch are either linked directly or via a linker compound
  • the reaction of active agent and hydroxyalkyl starch is carried out in an aqueous medium which comprises at least 10 wt.-% of water.
  • No examples are given which are directed to a hydroxyalkyl starch derivative which is linked to a carbonyl group comprised in the active reagent, neither an aldehyd or keto group nor a an acetal or a hemiacetal group.
  • hydroxyalkyl starch derivatives which are capable of forming a chemical linkage to a further compound, e.g. a polypeptide, which comprises, as functional group, a thio group or an aldehyd group, a keto group, a hemiacetal group, or an acetal group.
  • a further compound e.g. a polypeptide, which comprises, as functional group, a thio group or an aldehyd group, a keto group, a hemiacetal group, or an acetal group.
  • the aldehyd group, the keto group, the hemiacetal group, or the acetal group are comprised in a carbohydrate moiety of the further compound
  • the present invention relates to a method of producing a hydroxyalkyl starch derivative, said hydroxyalkyl starch having a structure according to formula (I)
  • hydroxyalkyl starch refers to a starch derivative which has been substituted by at least one hydroxyalkyl group. Therefore, the term hydroxyalkyl starch as used in the present invention is not limited to compounds where the terminal carbohydrate moiety comprises hydroxyalkyl groups R 1 , R 2 , and/or R 3 as depicted, for the sake of brevity, in formula (I), but also refers to compounds in which at least one hydroxy group present anywhere, either in the terminal carbohydrate moiety and/or in the remaining part of the starch molecule, HAS′, is substituted by a hydroxyalkyl group R 1 , R 2 , or R 3 .
  • the alkyl group may be a linear or branched alkyl group which may be suitably substituted.
  • the hydroxyalkyl group contains 1 to 10 carbon atoms, more preferably from 1 to 6 carbon atoms, more preferably from 1 to 4 carbon atoms, and even more preferably 2-4 carbon atoms.
  • “Hydroxyalkyl starch” therefore preferably comprises hydroxyethyl starch, hydroxypropyl starch and hydroxybutyl starch, wherein hydroxyethyl starch and hydroxypropyl starch are particularly preferred.
  • Hydroxyalkyl starch comprising two or more different hydroxyalkyl groups is also comprised in the present invention.
  • the at least one hydroxyalkyl group comprised in HAS may contain two or more hydroxy groups. According to a preferred embodiment, the at least one hydroxyalkyl group comprised HAS contains one hydroxy group.
  • hydroxyalkyl starch also includes derivatives wherein the alkyl group is mono- or polysubstituted.
  • the alkyl group is substituted with a halogen, especially fluorine, or with an aryl group, provided that the HAS remains soluble in water.
  • the terminal hydroxy group a of hydroxyalkyl group may be esterified or etherified.
  • alkyl instead of alkyl, also linear or branched substituted or unsubstituted alkene groups may be used.
  • Hydroxyalkyl starch is an ether derivative of starch.
  • ether derivatives also other starch derivatives can be used in the context of the present invention.
  • derivatives are useful which comprise esterified hydroxy groups. These derivatives may be, e.g., derivatives of unsubstituted mono- or dicarboxylic acids with 2-12 carbon atoms or of substituted derivatives thereof.
  • derivatives of unsubstituted monocarboxylic acids with 2-6 carbon atoms especially derivatives of acetic acid.
  • acetyl starch, butyl starch and propyl starch are preferred.
  • derivatives of dicarboxylic acids it is useful that the second carboxy group of the dicarboxylic acid is also esterified. Furthermore, derivatives of monoalkyl esters of dicarboxylic acids are also suitable in the context of the present invention.
  • the substitute groups may be preferably the same as mentioned above for substituted alkyl residues.
  • Hydroxyethyl starch is most preferred for all embodiments of the present invention.
  • the present invention also relates to a method as described above wherein the hydroxyalkyl starch is hydroxyethyl starch.
  • HES is mainly characterized by the molecular weight distribution and the degree of substitution. There are two possibilities of describing the substitution degree:
  • HES solutions are present as polydisperse compositions, wherein each molecule differs from the other with respect to the polymerisation degree, the number and pattern of branching sites, and the substitution pattern. BES is therefore a mixture of compounds with different molecular weight. Consequently, a particular HES solution is determined by average molecular weight with the help of statistical means.
  • M n is calculated as the arithmetic mean depending on the number of molecules.
  • M w the weight mean, represents a unit which depends on the mass of the HES.
  • hydroxyethyl starch may have a mean molecular weight (weight mean) of from 1 to 300 kDa, wherein a mean molecular weight of from 5 to 100 kDa is more preferred. Hydroxyethyl starch can further exhibit a molar degree of substitution of from 0.1 to 0.8 and a ratio between C 2 :C 6 substitution in the range of from 2 to 20 with respect to the hydroxyethyl groups.
  • R 1 , R 2 and R 3 are independently hydrogen or a hydroxyalkyl group, a hydroxyaryl group, a hydroxyaralkly group or a hydroxyalkarly group having of from 1 to 10 carbon atoms. Hydrogen and hydroxyalkyl groups having of from 1 to 6 carbon atoms are preferred.
  • the alkyl, aryl, aralkyl and/or alkaryl group may be linear or branched and suitably substituted.
  • the present invention also related to a method as described above wherein R 1 , R 2 and R 3 are independently hydrogen or a linear or branched hydroxyalkyl group with from 1 to 6 carbon atoms.
  • R 1 , R 2 and R 3 may be hydroxyhexyl, hydroxypentyl, hydroxybutyl, hydroxypropyl such as 1-hydroxypropyl, 2-hydroxypropyl, 3-hydroxypropyl, 1-hydroxyisopropyl, 2-hydroxyisopropyl, hydroxyethyl such as 1-hydroxyethyl, 2-hydroxyethyl, or hydroxymethyl. Hydrogen and hydroxyethyl groups are preferred, hydrogen and the 2-hydroxyethyl group being especially preferred
  • the present invention also relates to a method as described above wherein R 1 , R 2 and R 3 are independently hydrogen or a 2-hydroxyethyl group.
  • compound (D) or compound (L) is reacted with the reducing end of the hydroxyalkyl starch via the reaction of the functional group Z 1 with the reducing end where group Z 1 is comprised in compound (D) or compound (L).
  • compound (D) or compound (L) is reacted with the reducing end of the hydroxyalkyl starch and where the reducing end is oxidized prior to the reaction.
  • This oxidation of the reducing end leads to hydroxyalkyl starch in which the terminal carbohydrate group comprises a lactone group, or in which the terminal carbohydrate group, depending of the chemical reaction conditions and/or the oxidizing agents, has a non-cyclic structure comprising a carboxy group.
  • the hydroxyalkyl starch which is oxidized at its reducing end is present as a mixture of a compound comprising the lactone group and a compound comprising the carboxy group.
  • the respective compounds may be present at any conceivable ratio.
  • the present invention also relates to a method as described above wherein he reducing end of the hydroxyalkyl starch is oxidized prior to the reaction with compound (D) or compound (L), said hydroxyalkyl starch thus having a structure according to formula (IIa) and/or according to formula (IIb)
  • the oxidation of the reducing end of the hydroxyalkyl starch may be carried out according to each method or combination of methods which result compounds having the above-mentioned structures (IIa) and/or (IIb).
  • the oxidation may be carried out according to all suitable method or methods resulting in the oxidized reducing end of hydroxyakyl starch, it is preferably carried out using an alkaline iodine solution as described, e.g., in U.S. Pat. No. 196 28 705 A1.
  • the present invention also relates to a method as mentioned above wherein the reducing end is oxidized by an alkaline iodine solution.
  • compound (D) or compound (L) is reacted with the reducing end of the hydroxyalkyl starch and where the reducing end is not oxidized prior to the reaction.
  • the present invention also relates to a method as mentioned above wherein the reducing end of the hydroxyalkyl starch is not oxidized prior to the reaction with compound (D) or compound (L), said hydroxyalkyl starch thus having a structure according to formula (I)
  • each functional group may be used which is capable of forming a chemical linkage with the optionally oxidized reducing end of the hydroxyalkyl starch.
  • the functional group Z 1 comprises the chemical structure —NH—.
  • the present invention also relates to a method as described above wherein the functional group Z 1 comprises the structure —NH—.
  • the functional group Z 1 is a group having the structure R′—NH— where R′ is hydrogen or a alkyl, cycloalkyl, aryl, aralkyl, arylcycloalkyl, alkaryl or cycloalkylaryl residue where the cycloalkyl, aryl, aralkyl, arylcycloalkyl, alkaryl or cycloalkylaryl residue may be linked directly to the NH group or, according to another embodiment, may be linked by an oxygen bridge to the NH group.
  • alkyl, cycloalkyl, aryl, aralkyl, arylcycloalkyl, alkaryl, or cycloalkylaryl residues may be suitably substituted.
  • halogenes such as F, Cl or Br may be mentioned.
  • Especially preferred residues R′ are hydrogen, allyl and alkoxy groups, and even more preferred are hydrogen and unsubstituted alkyl and alkoxy groups.
  • alkyl and alkoxy groups groups with 1, 2, 3, 4, 5, or 6 C atoms are preferred. More preferred are methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, propoxy, and isopropoxy groups. Especially preferred are methyl, ethyl, methoxy, ethoxy, and particular preference is given to methyl or methoxy.
  • the present invention also relates to a method as described above wherein R′ is hydrogen or a methyl or a methoxy group.
  • the functional group R′′ is selected from the group consisting of where, if G is present twice, it is independently O or S.
  • the present invention also relates to a method as mentioned above wherein the functional group Z 1 is selected from the group consisting of wherein G is O or S and, if present twice, independently O or S, and R′ is methyl.
  • the functional group Y is selected from the group consisting of an aldehyd group, a keto group, a hemiacetal group, and an acetal group
  • the functional group X preferably comprises the chemical structure —NH—.
  • the present invention also relates to a method as described above wherein the functional group Y is selected from the group consisting of an aldehyd group, a keto group, a hemiacetal group, and an acetal group, and the functional group X comprises the structure —NH—.
  • the functional group X is a group having the structure R′—NH— where R′ is hydrogen or a alkyl, cycloalkyl, aryl, aralkyl, arylcycloalkyl, alkaryl or cycloalkylaryl residue where the cycloalkyl, aryl, aralkyl, arylcycloalkyl, alkaryl or cycloalkylaryl residue may be linked directly to the NH group or, according to another embodiment, may be linked by an oxygen bridge to the NH group.
  • alkyl, cycloalkyl, aryl, aralkyl, arylcycloalkyl, alkaryl, or cycloalkylaryl residues may be suitably substituted.
  • halogenes such as F, Cl or Br may be mentioned.
  • Especially preferred residues R′ are hydrogen, alkyl and alkoxy groups, and even more preferred are hydrogen and unsubstituted alkyl and alkoxy groups.
  • alkyl and alkoxy groups groups with 1, 2, 3, 4, 5, or 6 C atoms are preferred. More preferred are methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, propoxy, and isopropoxy groups. Especially preferred are methyl, ethyl, methoxy, ethoxy, and particular preference is given to methyl or methoxy.
  • the present invention also relates to a method as described above wherein R′ is hydrogen or a methyl or a methoxy group.
  • the functional group R′′ is selected from the group consisting of where, if G is present twice, it is independently O or S.
  • the present invention also relates to a method as mentioned above wherein the factional group X is selected from the group consisting of wherein G is O or S and, if present twice, independently O or S, and R′ is methyl.
  • the functional group X is preferably selected from the groups consisting of wherein Hal is Cl, Br or I, preferably Br or I.
  • the present invention also relates to a method as described above whereinwherein the functional group Y is —SH and the functional group X is selected from the group consisting of wherein Hal is Cl, Br or I.
  • hydroxyalkyl starch is reacted with a compound (D) and the resulting reaction product is further reacted with compound (L) where the chemical linkage between compound (L) and the reaction product is formed by reaction of functional group Z 2 comprised in compound (L) and functional group W comprised in compound (D) being part of the reaction product.
  • both W and Z 2 are groups from the list of groups given above.
  • Z 2 or W is a thio group.
  • the functional group W is preferably selected from the group consisting of wherein Hal is Cl, Br, or I, preferably Br or I.
  • the present invention also relates to a method as described above wherein the functional group W or the functional group Z 2 is —SH and the functional group Z 2 or the functional group W is selected from the group consisting of wherein Hal is Cl, Br, or I.
  • Z 2 or W is selected from the group consisting of an activated ester, as described above, or a carboxy group which is optionally transformed into an activated ester.
  • the functional group W or Z 2 respectively, comprises the chemical structure —NH—.
  • the present invention also relates to a method as described above wherein Z 2 or W is selected from the group consisting of an activated ester, as described above, or a carboxy group which is optionally transformed into an activated ester, and the functional group W or Z 2 , respectively, comprises the chemical structure —NH—.
  • the functional group W or Z 2 comprising the structure —NH— is a group having the structure R′—NH— where R′ is hydrogen or a alkyl, cycloalkyl, aryl, aralkyl, arylcycloalkyl, alkaryl or cycloalkylaryl residue where the cycloalkyl, aryl, aralkyl, arylcycloalkyl, alkaryl or cycloalkylaryl residue may be linked directly to the NH group or, according to another embodiment, may be linked by an oxygen bridge to the NH group.
  • alkyl, cycloalkyl, aryl, aralkyl, arylcycloalkyl, alkaryl, or cycloalkylaryl residues may be suitably substituted.
  • halogenes such as F, Cl or Br may be mentioned.
  • Especially preferred residues R′ are hydrogen, alkyl and alkoxy groups, and even more preferred are hydrogen and unsubstituted alkyl and alkoxy groups.
  • alkyl and alkoxy groups groups with 1, 2, 3, 4, 5, or 6 C atoms are preferred. More preferred are methyl, ethyl, propyl, isopropyl, methoxy, ethoxy, propoxy, and isopropoxy groups. Especially preferred are methyl, ethyl, methoxy, ethoxy, and particular preference is given to methyl or methoxy.
  • the present invention also relates to a method as described above wherein W or Z 2 is selected from the group consisting of an activated ester, as described above, or a carboxy group which is optionally transformed into an activated ester, and the functional group W or Z 2 , respectively, is R′—NH— wherein R′ is hydrogen or a methyl or a methoxy group.
  • the functional group R′′ is selected from the group consisting of where, if G is present twice, it is independently O or S.
  • the present invention also relates to a method as mentioned above wherein the functional group W or Z 2 is selected from the group consisting of wherein G is O or S and, if present twice, independently O or S, and R′ is methyl.
  • the at least one functional group X, Z 2 and/or W may be a group which is not capable of reacting directly with a given further compound, but which may be chemically modified in order to be capable of reacting in the desired way.
  • a 1,2-amino alcohol or a 1,2-diol may be mentioned which is modified, e.g., by oxidation to form an aldehyd or a keto group.
  • a functional group to be modified prior to the reaction with a further compound is a —NH 2 group which is modified by the reaction with, e.g., a compound according to the following formula to give a structure of the following formula which is, e.g., reactive towards a thio group.
  • a functional group to be modified prior to the reaction with a fatter compound is a —NH 2 group which is modified by the reaction with, e.g., a compound according to the following formula to give a structure of the following formula which is, e.g., reactive towards a thio group.
  • a functional group to be modified prior to the reaction with a further compound is an amino group which is reacted with bromoacetic anhydride or N-succinimidyl iodo acetate.
  • a compound (L) has the structure Z 1 -L′-X or Z 2 -L′-X, L′ being an organic residue separating the functional groups and being optionally absent, the structure depending on whether a compound (D) is reacted with the hydroxyalkyl starch or not.
  • no compound (D) is involved and Y is selected from the group consisting of an aldehyd group, a keto group, a hemiacetal group, and an acetal group.
  • L′ may be a linear or branched alkyl or cycloalkyl or aryl or or aralkyl or arylcycloalkyl or alkaryl or cycloalkylaryl group, wherein L′ may comprise at least one heteroatom such as N, O, S, and wherein L′ may be suitably substituted.
  • the size of the group L′ may be adapted to the specific needs.
  • the separating group L′ generally has from 1 to 60, preferably from 1 to 40, more preferably from 1 to 20, more preferably from 1 to 10, more preferably from 1 to 6 and especially preferably from 1 to 4 carbon atoms.
  • the separating group comprises generally from 1 to 20, preferably from 1 to 8 and especially preferably from 1 to 4 heteroatoms.
  • the separating group L′ comprises 1 to 4 oxygen atoms.
  • the separating group L′ may comprise an optionally branched alkyl chain or an aryl group or a cycloalkyl group having, e.g., from 5 to 7 carbon atoms, or be a aralkyl group, an alkaryl group where the alkyl part may be a linear and/or cyclic alkyl group.
  • the separating group is an alkyl chain of from 1 to 20, preferably from 1 to 8, more preferably from 1 to 6, more preferably from 1 to 4 and especially preferably from 2 to 4 carbon atoms.
  • a chain comprising 1 to 4 oxygen atoms is particularly preferred.
  • a compound (D) is involved.
  • a compound (D) has the structure Z 1 -D′-W, D′ being an organic residue separating the functional groups and being optionally absent.
  • compound (D) having the structure Z 1 -D′-W where D′ is absent:
  • a specific example of a compound D where D′ is absent is NH 3 .
  • D′ may be a linear or branched alkyl or cycloalkyl or aryl or or aralkyl or arylcycloalkyl or alkaryl or cycloalkylaryl group, wherein D′ may comprise at least one heteroatom such as N, O, S, and wherein D′ may be suitably substituted
  • the size of the group D′ may be adapted to the specific needs.
  • the separating group D′ generally has from 1 to 60, preferably from 1 to 40, more preferably from 1 to 20, more preferably from 1 to 10, more preferably from 1 to 6 and especially preferably from 1 to 4 carbon atoms.
  • the separating group comprises generally from 1 to 20, preferably from 1 to 8 and especially preferably from 1 to 4 heteroatoms.
  • the separating group D′ comprises 1 to 4 oxygen atoms.
  • the separating group D′ may comprise an optionally branched alkyl chain or an aryl group or a cycloalkyl group having, e.g., from 5 to 7 carbon atoms, or be an aralkyl group, an alkaryl group where the alkyl part may be a linear and/or cyclic alkyl group.
  • the separating group is an alkyl chain of from 1 to 20, preferably from 1 to 8, more preferably from 1 to 6, more preferably from 1 to 4 and especially preferably from 2 to 4 carbon atoms.
  • a chain comprising 1 to 4 oxygen atoms is particularly preferred.
  • preferred compounds (D) having the structure Z 1 -D′-W where D′ is not absent are:
  • the functional group Y is a thio group and the functional group W comprises the structure —NH—
  • the following types of compounds (L) are, among others, preferred: Type of compound (L) Functional group X Functional group Z 2 C Iodoalkyl N-succinimide ester D Bromoalkyl N-succinimide ester E Maleimido N-succinimide ester F Pydridyldithio N-succinimide ester G Vinylsulfone N-succinimide ester
  • the functional group Y is selected from the group consisting of an aldehyd group, a keto group, a hemiacetal group, and an acetal group
  • the functional group W is a thio group
  • the following types of compounds (L) are, among others, preferred: Type of compound (L) Functional group X Functional group Z 2 A Hydrazide Maleimido B Hydrazide Pyridyldithio
  • the separating groups L′ and/or D′ may be suitably substituted.
  • Preferred substituents are, e.g, halides such as F, Cl, Br or I.
  • the separating groups L′ and/or D′ may comprise one or more cleavage sites such as which allow for an easy cleavage of a resulting compound at a pre-determined site.
  • hydroxyalkyl starch derivative comprises the functional group X capable of being reacted with a functional group Y comprised in a further compound (M) and wherein said functional group Y is selected from the group consisting of an aldehyd group, a keto group, a hemiacetal group, an acetal group, are being particularly preferred.
  • compounds (D) which may be linked to hydroxyalkyl starch wherein the resulting hydroxyalkyl starch derivative comprises the functional group W capable of being reacted with a functional group Z 2 comprised in a compound (L) wherein the resulting hydroxyalkyl starch derivative which comprises hydroxyalkyl starch, compound (D) and compound (L), is capable of being reacted with the functional group Y of a further compound (M) and wherein said functional group Y is a thio group, are the compounds (D) being particularly preferred.
  • a compound (D) or a compound (L) is reacted with the reducing end of the hydroxyalkyl starch which is not oxidised.
  • the reaction product of a compound (D) or a compound (L) is reacted with the reducing end of the hydroxyalkyl starch which is not oxidised may have different constitutions.
  • this reaction is carried out in an aqueous system.
  • aqueous system refers to a solvent or a a mixture of solvents comprising water in the range of from at least 10% per weight, preferably at least 50% per weight, more preferably at least 80% per weight, even more preferably at least 90% per weight or up to 100% per weight, based on the weight of the solvents involved.
  • solvents such as DMSO, DMF, ethanol or methanol may be mentioned.
  • the hydroxyalkyl starch derivative may have a constitution according to formula (IIIa)
  • the hydroxyalkyl starch derivative may have a constitution according to formula (IIIa) or formula (IIIb) or be a mixture of compounds according to formulae (IIIa) and (IIIb)
  • the compounds according to formula (IIIa) may be present with the N atom in equatorial or axial position where also a mixture of both forms may be present having a certain equilibrium distribution.
  • the compounds according to formula (IIIb) may be present with the C—N double bond in E or Z conformation where also a mixture of both forms may be present having a certain equilibrium distribution.
  • the present invention also relates to a hydroxyalkyl starch derivative as described above having a constitution according to formula (IIIb) or according to formula (IIIb) or according to formulae (IIIa) and (IIIb).
  • acylation of the compound according to formula (IIIa) is particularly preferred, especially in the case where R′ is hydrogen.
  • acylation reagent all suitable reagents may be used which result in the desired hydroxyalkyl starch derivative according to formula (IVa)
  • the residue Ra being part of the acylation reagent is methyl.
  • acylation reagents carboxylic acid anhydrides, carboxylic acid halides, and carboxylic acid active esters are preferably used.
  • the acylation is carried at a temperature in the range of from 0 to 30° C., preferably in the range of from 2 to 20° C. and especially preferably in the range of from 4 to 10° C.
  • the present invention also relates to a hydroxyalkyl starch derivate obtainable by a method as described above wherein said derivative has a constitution according to formula (IVa).
  • boro hydrides such as NaCNBH 3 or NaBH 4 are used.
  • the reduction is carried at a temperature in the range of from 4 to 100° C., preferably in the range of from 10 to 90° C. and especially preferably in the range of from 25 to 80° C.
  • the present invention also relates to a hydroxyalkyl starch derivate obtainable by a method as described above wherein said derivative has a constitution according to formula (IVb).
  • the present invention further relates to mixtures of compounds having constitutions according to formulae (IIIa) and (IIIb), (IVa) and (IVb), (IIIa) and (IVa), (IIIa) and (IVb), (IIIb) and (IVa), (IIIb) and (IVb), (IIIa) and (IIIb) and (IVa), (IIIa) and (IIIb) and (IVb), (IIIa) and (IIIb) and (IVa), (IIIa) and (IIIb) and (IVb), (IVa) and (IVb) and (IIIa), and (IVa) and (IVb) and (IIIb) wherein (IIIa) and/or (IVa) may be independently present in a conformation where the N atom in equatorial or axial position and/or wherein (IIIb) may be present with the C—N double bond in E or Z conformation.
  • a compound (D) or a compound (L) is reacted with the reducing end of the hydroxyalkyl starch which is oxidised.
  • polar aprotic solvents are used which may also contain a certain amount of water, such as up to 10 wt.-%.
  • Preferred aprotic solvents are, among others, DMSO or DMF.
  • An example of a preferred reaction temperature range is from room 20 to 65° C., and the reaction times are generally in the range of 1 minute to several hours and up to several days, depending on the chemical nature of the functional group which is reacted with the oxidized reducing end og the hydroxyalkyl starch and the other reaction conditions.
  • the hydroxyalkyl starch derivative may have a constitution according to formula (Va)
  • the present invention also relates to a hydroxyalkyl starch derivate obtainable by a method as described above wherein said derivative has a constitution according to formula (Va).
  • a preferred embodiment of the present invention relates to a method as described above wherein hydroxyalkyl starch is reacted with a compound (L) via the reaction of functional group Z 1 with the optionally oxidized reducing end of the hydroxyalkyl starch and the resulting reaction product is reacted with a further compound (M) via the reaction of the functional group X comprised in compound (L) with the functional group Y comprised in compound (M).
  • Another embodiment of the present invention relates to a method as described above wherein hydroxyalkyl starch is reacted with a compound (L) via the reaction of functional group Z 1 with the optionally oxidized reducing end of the hydroxyalkyl starch, where compound (L), prior to the reaction with hydroxyalkyl starch, is reacted with a further compound (M) via the reaction of functional group X comprised in compound (L) with the functional group Y comprised in compound (M).
  • Still another embodiment of the present invention relates to a method as described above wherein hydroxyalkyl starch is reacted with a compound (D) via the reaction of the functional group Z 1 comprised in compound (D), with the optionally oxidized reducing end of the hydroxyalkyl starch to give a first hydroxyalkyl starch derivative, and where the first hydroxyalkyl starch derivative is reacted with a compound (L) via the reaction of functional group Z 2 comprised in compound (L) with the functional group W comprised in compound (D) to give a second hydroxyalkyl starch derivative.
  • Yet another embodiment of the present invention relates to the latter method wherein the second hydroxyalkyl starch derivative is reacted with a further compound (M) via the reaction of functional group X comprised in compound (L) with the functional group Y comprised in compound (M).
  • Still yet another embodiment of the present invention relates to a method as described above wherein hydroxyalkyl starch is reacted with a compound (D) via the reaction of functional group Z 1 comprised in compound (D) with the optionally oxidized reducing end of the hydroxyalkyl starch to give a first hydroxyalkyl starch derivative, and where the first hydroxyalkyl starch derivative is reacted, via the reaction of the functional group W, comprised in compound (D), and the functional group Z 2 , comprised in compound (L), with compound (L), where compound (L), prior to the reaction with the first hydroxyalkyl starch derivative, is reacted with a further compound (M) via the reaction of functional group X comprised in compound (L) with the functional group Y comprised in compound (M).
  • water is used as solvent, either alone or in combination with at least one other solvent.
  • DMSO, DMF, methanol and ethanol may be mentioned.
  • Preferred solvents other than water are DMSO, DMF, methanol and ethanol.
  • hydroxylalkyl starch is preferably reacted via the non-oxidized reducing end.
  • the temperature of the reaction is preferably in the range of from 5 to 45° C., more preferably in the range of from 10 to 30° C. and especially preferably in the range of from 15 to 25° C.
  • the temperature is preferably in the range of up to 100° C., more preferably in the range of from 70 to 90° C. and especially preferably in the range of from 75 to 85° C.
  • the temperature may be varied, preferably in the above-given ranges, or held essentially constant.
  • reaction time for the reaction of hydroxyalkyl starch with compound (D) or compound (L) may be adapted to the specific needs and is generally in the range of from 1 h to 7 d.
  • reaction time is preferably in the range of from 1 h to 3 d and more preferably of from 2 h to 48 h.
  • reaction time is preferably in tie range of from 2 h to 7 d.
  • the pH value for the reaction of hydroxyalkyl starch with compound (D) or compound (L) may be adapted to the specific needs such as the chemical nature of the reactants.
  • the pH value is preferably in the range of from 4.5 to 6.5.
  • the pH value is preferably in the range of from 8 to 12.
  • the suitable pH value of the reaction mixture may be adjusted, for each reaction step, by adding at least one suitable buffer.
  • suitable buffers sodium acetate buffer, phosphate or borate buffers may be mentioned.
  • the at least one functional group X may be protected with at least one suitable protecting group prior to the reaction of hydroxyalkyl starch with compound (L) or prior to the reaction of compound (D) with compound (L) or prior to the reaction of compound (L) with the reaction product of the reaction of hydroxyalkyl starch with compound (D).
  • the protecting group may be chosen depending from the chemical nature of the functional group X to be protected, from, e.g., the solvent the reaction is carried out in or the pH of the reaction mixture.
  • Preferred protecting groups are, among others, the benzyloxycarbonyl group, the tert-butoxycarbonyl group, the methoxyphenyl group, the 2,4-dimethoxyphenyl group, triarly methyl groups, trityl, the monomethoxytrityl group, the dimethoxytrityl group, the monomethyltrityl group, the dimethyltrityl group, the trifluoracetyl group, phthalimin compounds, 2-(trialkylsilyl)ethoxy carbonyl compounds, Fmoc, the tert-butyl group, or trialkyl silyl groups.
  • the at least one protecting group may be left in the reaction product or removed by suitable methods such as conventional methods known to the person skilled in the art. If two different functional groups X are protected by suitable protecting groups, it is possible to remove at least one protecting group so as to make at least one functional group X available for further reaction with at least one further compound (M), and leave at least one other functional group protected until the reaction product comprising compound (L) is reacted with the further compound (M). Afterwards, the protecting group of the functional group still protected may be removed to make the remaining functional group X available for reaction with yet a further compound (M).
  • the use of at least one protecting group may be important for preventing the reaction from resulting in a hydroxyalkyl starch derivative comprising a compound (L) or compound (D) which has been reacted with two or more hydroxyalkyl starch molecules, i.e. a multiple HAS substituted compound (L) or (D).
  • a hydroxyalkyl starch derivative comprising a compound (L) or compound (D) which has been reacted with two or more hydroxyalkyl starch molecules, i.e. a multiple HAS substituted compound (L) or (D).
  • the same result may be achieved by reacting hydroxyalkyl starch with an excess of compound (L) or (D). If an excess amount of compound (L) or (D) is used in the process of the present invention, the molar ratio of compound (L) or (D) to hydroxyalkyl starch is preferably in the range of from 2 to 100.
  • reaction product of the respective reaction step as described above, it may be isolated from the reaction mixture by at least one suitable method. If necessary, the reaction product may be precipitated prior to the isolation by at least one suitable method.
  • reaction product is precipitated first, it is possible, e.g., to contact the reaction mixture with at least one solvent or solvent mixture other than the solvent or solvent mixture present in the reaction mixture at suitable temperatures.
  • the reaction mixture is contacted with a mixture of ethanol and acetone, preferably a 1:1 mixture, indicating equal volumes of said compounds, at a temperature, preferably in the range of from ⁇ 20 to +50° C. and especially preferably in the range of from 0 to 25° C.
  • Isolation of the reaction product may be carried out by a suitable process which may comprise one or more steps.
  • the reaction product is first separated off the reaction mixture or the mixture of the reaction mixture with, e.g., the ethanol-acetone mixture, by a suitable method such as centrifugation or filtration.
  • the separated reaction product may be subjected to a further treatment such as an after-treatment like dialysis, centrifugal filtration or pressure filtration, ion exchange chromatography, HPLC, MPLC, gel filtration and/or lyophilisation.
  • the separated reaction product is first dialysed, preferably against water, and then lyophilized until the solvent content of the reaction product is sufficiently low according to the desired specifications of the product. Lyophilisation may be carried out at temperature of from 20 to 35° C., preferably of from 25 to 30° C.
  • the hydroxyalkyl starch derivative comprising hydroxyalkyl starch and compound (L) or comprising hydroxyalkyl starch, compound (D) and compound (L) is further reacted with the further compound (M) which comprises at least one functional group Y.
  • compound (M) there are no limitations regarding compound (M).
  • a polypeptide is used as compound (M) in the context of the present invention.
  • other compounds M) are also possible, either polymers or oligomers or monomolecular compounds or mixtures of two or more thereof.
  • polypeptide refers to a compound which comprises at least 2 amino acids which are linked via a peptide bond, i.e. a bond with structure —(C ⁇ O)—NH—.
  • the polypeptide may be a naturally occuring compound or a polypeptide which does not occur naturally, the latter comprising naturally occuring amino acids and/or at least one amino acid which does not naturally occur.
  • the backbone of the polypeptide, the polypeptide chain may be further substituted with at least one suitable substituent thus having at least one side-chain.
  • the at least one functional group Y may be part of the polypeptide backbone or of at least one substituent of the backbone wherein embodiments are possible comprising at least one functional group being part of the polypeptide backbone and at least one functional group being part of at least one substituent of the polypeptide backbone.
  • the polypeptide comprises at least one functional group Y.
  • Said functional group Y may be linked directly to the polypeptide backbone or be part of a side-chain of the backbone. Either side-chain or functional group Y or both may be part of a naturally occuring polypeptide or may be introduced into a naturally occuring polypeptide or into a polypeptide which, at least partially, does not occur naturally, prior to the reaction with the functional group X.
  • polypeptide can be, at least partly, of any human or animal source.
  • polypeptide is of human source.
  • the polypeptide may be a cytokine, especially erythropoietin, an antithrombin (AT) such as AT III, an interleukin, especially interleukin-2, IFN-beta, IFN-alpha, G-CSF, CSF, interleukin-6 and therapeutic antibodies.
  • AT antithrombin
  • interleukin especially interleukin-2, IFN-beta, IFN-alpha, G-CSF, CSF, interleukin-6 and therapeutic antibodies.
  • the polypeptide is an antithrombin (AT), preferably AT III (Levy J H, Weisinger A, Ziomek C A, Echelard Y, Recombinant Antithrombin: Production and Role in Cardiovascular Disorder, Seminars in Thrombosis and Hemostasis 27, 4 (2001) 405-416; Edmunds T, Van Patten S M, Pollock J, Hanson E, Bernasconi R, Higgins E, Manavalan P, Ziomek C, Meade H, McPherson J, Cole E S, Transgenically Produced Human Antithrombin: Structural and Functional Comparison to Human Plasma-Derived Antithrombin, Blood 91, 12 (1998) 4661-4671; Minnema M C, Chang A C K, Jansen P M, Lubbers Y T P, Pratt B M, Whittaker B G, Taylor F B, hack C E, Friedman B.
  • AT antithrombin
  • Recombinant human antithrombin III improves survival and attenuates inflammatory responses in baboons lethally challenged with Escherichia coli, Blood 95, 4 (2000) 1117-1123; Van Patten S M, Hanson E H, Bernasconi R, Zhang K, Manavaln P, Cole E S, McPherson J M, Edmunds T, Oxidation of Methionine Residues in Antithrombin, J. Biol. Chemistry 274, 15 (1999) 10268-10276).
  • the polypeptide is human IFN-beta, in particular IFN-beta 1a (cf. Avonex®, REBIFO) and IFN-beta 1b (cf. BETASERON®).
  • a further preferred polypeptide is human G-CSF (granulocyte colony stimulating factor).
  • G-CSF granulocyte colony stimulating factor
  • the at least two polypeptides may differ, e.g., in the molecular mass, the number and/or sequence of amino acids, the number and/or chemical nature of the substituents or the number of polypeptide chains linked by suitable chemical bonds such as disulfide bridges.
  • the reaction product of hydroxyalkyl starch and compound (L) or the reaction product of hydroxyalkyl starch and compound (D) which is further reacted with compound (L) is isolated, preferably according to at least one of the above-mentioned processes, and then reacted with a polypeptide having at least one functional group Y.
  • the functional group Y is comprised in a carbohydrate moiety of the polypeptide.
  • carbohydrate moiety refers to hydroxyaldehydes or hydroxyketones as well as to chemical modifications thereof (see Römpp Chemielexikon, Thieme Verlag Stuttgart, Germany, 9 th edition 1990, Volume 9, pages 2281-2285 and the literature cited therein). Furthermore, it also refers to derivatives of naturally occuring carbohydrate moieties like glucose, galactose, mannose, sialic acid and the like. The term also includes chemically oxidized, naturally occuring carbohydrate moieties. The structure of the oxidized carbohydrate moiety may be cyclic or linear.
  • the carbohydrate moiety may be linked directly to the polypeptide backbone.
  • the carbohydrate moiety is part of a carbohydrate side chain. More preferably, the carbohydrate moiety is the terminal moiety of the carbohydrate side chain.
  • the carbohydrate moiety is a galactose residue of the carbohydrate side chain, preferably the terminal galactose residue of the carbohydrate side chain.
  • This galactose residue can be made available for reaction with the functional group X comprised in the reaction product of hydroxyalkyl starch and compound (L) or the reaction product of hydroxyalkyl starch and compound (I) which is further reacted with compound (L), by removal of terminal sialic acids, followed by oxidation, as described hereinunder.
  • reaction product of hydroxyalkyl starch and compound (L) or the reaction product of hydroxyalkyl starch and compound (D) which is further reacted with compound (L) is linked to a sialic acid residue of the carbohydrate side chains, preferably the terminal sialic acid residue of the carbohydrate side chain.
  • Oxidation of terminal carbohydrate moieties can be performed either chemically or enzymatically.
  • the carbohydrate moiety may be oxidized enzymatically.
  • Enzymes for the oxidation of the individual carbohydrate moieties are known in the art, e.g. in the case of galactose the enzyme is galactose oxidase. If it is intended to oxidize terminal galactose moieties, it will be eventually necessary to remove terminal sialic acids (partially or completely) if the polypeptide has been produced in cells capable of attaching sialic acids to carbohydrate chains, e.g. in mammalian cells or in cells which have been genetically modified to be capable of attaching sialic acids to carbohydrate chains.
  • the functional group of the polypeptide is the thio group. Therefore, the reaction product of hydroxyalkyl starch and compound (L) or the reaction product of hydroxyalkyl starch and compound (D) which is further reacted with compound (L) may be linked to the polypeptide via a thioether group wherein the S atom can be derived from any thio group comprised in the polypeptide.
  • the present invention also relates to a method as described above wherein the reaction product of hydroxyalkyl starch and compound (D) is further reacted with compound (L) is reacted with the polypeptide via a thio group comprised in the polypeptide.
  • the present invention also relates to a method as described above wherein the reaction product of hydroxyalkyl starch and compound (D) which is further reacted with compound (L) is reacted with the polypeptide via an oxidized carbohydrate moiety and a thio group comprised in the polypeptide.
  • the thio group may be present in the polypeptide as such. Moreover, it is possible to introduce a thio group into the polyeptide according to a suitbale method. Among others, chemical methods may be mentioned. If a disulfide bridge is present in the polypeptide, it is possible to reduce the —S—S— structure to get a thio group. It is also possible to transform an amino group present in the polypeptide into a SH group by reaction the polypeptide via the amino group with a compound which has at least two different functional groups, one of which is capable of being reacted with the amino group and the other is an SH group or a precursor of an SH group.
  • This modification of an amino group may be regarded as an example where the protein is first reacted with a compound (L) which has at least two different functional groups, one of which is capable of being reacted with the amino group and the other is an SH group, and the resulting reaction product is then reacted with, e.g., a HAS derivative comprising HAS and a compound (D), said derivative comprising a functional group being capable of reacting with the SH group.
  • a HAS derivative comprising HAS and a compound (D)
  • said derivative comprising a functional group being capable of reacting with the SH group.
  • an SH group by mutation of the polypeptide such as by introducing a cystein or a suitable SH functional amino acid into the polypeptide or such as removing a cystein from the polypeptide so as to disable another cystein in the polypeptide to form a disulfide bridge.
  • EPO erythropoietin
  • the present invention also relates to a method as described above wherein the polypeptide is erythropoietin.
  • the EPO can be of any human (see e.g. Inoue, Wada, Takeuchi, 1994, An improved method for the purification of human erythropoietin with high in vivo activity from the urine of anemic patients, Biol. Pharm. Bull. 17(2), 180-4; Miyake, Kung, Goldwasser, 1977, Purification of human erythropoietin., J. Biol. Chem., 252(15), 5558-64) or another mammalian source and can be obtained by purification from naturally occurring sources like human kidney, embryonic human liver or animal, preferably monkey kidney.
  • erytliropoietin or “EPO” encompasses also an EPO variant wherein one or more amino acids (e.g. 1 to 25, preferably 1 to 10, more preferred 1 to 5, most preferred 1 or 2) have been exchanged by another amino acid and which exhibits erythropoietic activity (see e.g. EP 640 619 B1).
  • the measurement of erythropoietic activity is described in the art (for measurement of activity in vitro see e.g. Fibi et al.,1991, Blood, 77, 1203 ff; Kitamura et al, 1989, J. Cell Phys., 140, 323-334; for measurement of EPO activity in vivo see Ph. Eur.
  • the EPO is recombinantly produced.
  • the EPO may be expressed in transgenic animals (e.g. in body fluids like milk, blood, etc.), in eggs of transgenic birds, especially poultry, preferred chicken, or in transgenic plants.
  • the recombinant production of a polypeptide is known in the art. In general, this includes the transfection of host cells with an appropriate expression vector, the cultivation of the host cells under conditions which enable the production of the polypeptide and the purification of the polypeptide from the host cells. For detained information see e.g.
  • the EPO has the amino acid sequence of human EPO (see EP 148 605 B2).
  • the EPO may comprise one or more carbohydrate side chains, preferably 1 to 12, more preferably 1 to 9, even more preferably 1 to 6 and particularly 1 to 4, especially preferably 4 carbohydrate side chains, attached to the EPO via N- and/ or O-linked glycosylation, i.e. the EPO is glycosylated.
  • carbohydrate side chains may have been attached to the EPO during biosynthesis in mammalian, especially human, insect or yeast cells.
  • the hydroxyalkyl starch derivative according to the present invention may comprise at least one, preferably 1 to 12, more preferably 1 to 9, even more preferably 1 to 6 and particularly preferably 1 to 4 HAS molecules per EPO molecule.
  • the number of HAS-molecules per EPO molecule can be determined by quanatitative carbohydrate compositional analysis using GC-MS after hydrolysis of the product and derivatisation of the resulting monosaccharides (see Chaplin and Kennedy (eds.), 1986, Carbohydrate Analysis: a practical approach, IRL Press Practical approach series (ISBN 0-947946-44-3), especially Chapter 1, Monosaccharides, page 1-36; Chapter 2, Oligosaccharides, page 37-53, Chapter 3, Neutral Polysaccharides, page 55-96).
  • the carbohydrate moiety linked to EPO is part of a carbohydrate side chain. More preferably, the carbohydrate moiety is the terminal moiety of the carbohydrate side chain. In an even more preferred embodiment, the carbohydrate moiety is a galactose residue of the carbohydrate side chain, preferably the terminal galactose residue of the carbohydrate side chain. This galactose residue can be made available for reaction with the reaction product of compound (I) and compound (II) by removal of terminal sialic acids, followed by oxidation, as described hereinunder.
  • reaction product of compound (I) and (II) is linked to a sialic acid residue of the carbohydrate side chains, preferably the terminal sialic acid residue of the carbohydrate side chain.
  • the sialic acid is oxidized as described hereinunder.
  • this galactose residue is made available for reaction with the reaction product of hydroxyalkyl starch and compound (L) or the reaction product of hydroxyalkyl starch and compound (D) which is further reacted with compound (L) via the functional group X by removal of terminal sialic acid followed by oxidation.
  • this galactose residue is made available for reaction with the reaction product of hydroxyalkyl starch and compound (L) or the reaction product of hydroxyalkyl starch and compound (I) which is further reacted with compound (L) via the functional group X by oxidation wherein terminal sialic acid is not removed.
  • reaction product of hydroxyalkyl starch and compound (L) or the reaction product of hydroxyalkyl starch and compound (D) which is further reacted with compound (L) be reacted with a thio group comprised in EPO.
  • reaction product of hydroxyalkyl starch and compound (L) or the reaction product of hydroxyalkyl starch and compound (I) which is further reacted with compound (L) with a thio group as well as with a carbohydrate moiety each of them comprised in the at least one further compound, preferably a polypeptide, more preferably erythropoietin.
  • this SH group may be linked to a preferably oxidized carbohydrate moiety, e.g. by using a hydroxylamine derivative, e.g. 2-(aminooxy)ethylmercaptan hydrochloride (Bauer L. et al., 1965, J. Org. Chem., 30, 949) or by using a hydrazide derivative, e.g. thioglycolic acid hydrazide (Whitesides et al., 1977, J. Org. Chem., 42, 332.)
  • a hydroxylamine derivative e.g. 2-(aminooxy)ethylmercaptan hydrochloride (Bauer L. et al., 1965, J. Org. Chem., 30, 949)
  • a hydrazide derivative e.g. thioglycolic acid hydrazide (Whitesides et al., 1977, J. Org. Chem., 42, 332
  • the thio group is preferably introduced in an oxidized carbohydrate moiety of EPO, more preferably an oxidized carbohydrate moiety which is part of a carbohydrate side chain of EPO.
  • the thio group is derived from a naturally occurring cysteine or from an added cysteine. More preferably, the EPO has the amino acid sequence of human EPO and the naturally occurring cysteines are cysteine 29 and/or 33. In a more preferred embodiment, t the reaction product of hydroxyalkyl starch and compound (L) or the reaction product of hydroxyalkyl starch and compound (D) which is further reacted with compound (S) is reacted with cysteine 29 whereas cysteine 33 is replaced by another amino acid.
  • reaction product of hydroxyalkyl starch and compound (L) or the reaction product of hydroxyalkyl starch and compound (D) which is further reacted with compound (L) is reacted with cysteine 33 whereas cysteine 29 is replaced by another amino acid.
  • the term “added cysteines” indicates that the polypeptides, preferably EPO, comprise a cysteine residue which is not present in the wild-type polypeptide.
  • cysteine may be an additional amino acid added at the N- or C-terminal end of EPO.
  • the added cysteine may have been added by replacing a naturally occuring amino acid by cysteine or a suitably substituted cysteine.
  • the EPO is human EPO and the replaced amino acid residue is serine 126.
  • reaction conditions regarding the reaction of the reaction product of hydroxyalkyl starch and compound (I), optionally with compound (D), with the further compound (M) no specific limitations exist, and the reaction conditions may be adjusted to the specific needs.
  • water is used as solvent, either alone or in combination with at least one other solvent.
  • DMSO, DMF, methanol or ethanol may be mentioned.
  • Preferred solvents other than water are methanol and ethanol.
  • DMSO or DMF or methanol or ethanol or a mixture of two or more thereof is used as solvent.
  • reaction temperatur is preferably preferably in the range of from 4 to 37° C., more preferably of from 10 to 30° C. and especially preferably of from 15 to 25° C.
  • Isolation of the reaction product comprising the further compound (M), preferably the polypeptide and especially preferably erythropoietin can be performed by using known procedures for the purification of natural and recombinant EPO (e.g. size exclusion chromatography, ion-exchange chromatography, RP-HPLC, hydroxyapatite chromatography, hydrophobic interaction chromatography or combinations thereof). Isolation of the reaction product may be carried out by a suitable process which may comprise one or more steps. According to a preferred embodiment of the present invention, the reaction product is first separated off the reaction mixture or the mixture of the reaction mixture with, e.g., the ethanol-acetone mixture, by a suitable method such as centrifugation or filtration.
  • a suitable method such as centrifugation or filtration.
  • the separated reaction product may be subjected to a further treatment such as an after-treatment like dialysis, centrifugal filtration or pressure filtration, ion exchange chromatography such as, e.g., by a column containing Q-sepharose, HPLC, MPLC, gel filtration and/or lyophilisation.
  • a further treatment such as an after-treatment like dialysis, centrifugal filtration or pressure filtration, ion exchange chromatography such as, e.g., by a column containing Q-sepharose, HPLC, MPLC, gel filtration and/or lyophilisation.
  • the separated reaction product is first dialysed, preferably against water, and then lyophilized until the solvent content of the reaction product is sufficiently low according to the desired specifications of the product. Lyophilisation may be carried out at temperature of from 20 to 35° C., preferably of from 25 to 30° C.
  • the reaction mixture comprising the reaction product is applied to a column containing Q-Sepharose to give an
  • the present invention relates to a hydroxyalkyl starch derivative obtainable by a method of producing a hydroxyalkyl starch derivative, said hydroxyalkyl starch having a structure according to formula (I)
  • the present invention relates to a hydroxyalkyl starch derivative as described above wherein R 1 , R 2 and R 3 are independently hydrogen or a linear or branched hydroxyalkyl group.
  • the present invention relates to a hydroxyalkyl starch derivative as described above wherein R 1 , R 2 and R 3 are independently hydrogen or a 2-hydroxyethyl group.
  • the present invention relates to a hydroxyalkyl starch derivative as described wherein the hydroxyalkyl starch is hydroxyethyl starch.
  • the present invention relates to a hydroxyalkyl starch derivative as described above wherein the functional group Z 1 comprises the structure —NH—.
  • the present invention relates to a hydroxyalkyl starch derivative as described above wherein Z 1 is selected from the group consisting of wherein G is O or S and, if present twice, independently O or S, and R′ is methyl.
  • the present invention relates to a hydroxyalkyl starch derivative as described above wherein the functional group Y is selected from the group consisting of an aldehyd group, a keto group, a hemiacetal group, and an acetal group, and the functional group X comprises the structure —NH—.
  • the present invention relates to a hydroxyalkyl starch derivative as described above X is selected from the group consisting of wherein G is O or S and, if present twice; independently O or S, and R is methyl.
  • the present invention relates to a hydroxyalkyl starch derivative as described above wherein the functional group Y is —SH and the functional group X is selected from the group consisting of wherein Hal is Cl, Br or I.
  • the present invention relates to a hydroxyalkyl starch derivative as described above wherein the functional group W or the functional group Z 2 is —SH and the functional group Z 2 or the functional group W is selected from the group consisting of wherein Hal is Cl, Br, or I.
  • the present invention relates to a hydroxyalkyl starch derivative as described above wherein the functional group W or the functional group Z 2 is selected from the group consisting of an activated ester, as described above, or a carboxy group which is optionally transformed into an activated ester and the functional group Z 2 or the functional group W is selected from the group consisting of wherein G is O or S and, if present twice, independently O or S, and R is methyl.
  • the present invention relates to a hydroxyalkyl starch derivative as described above wherein the reducing end of the hydroxyalkyl starch is not oxidized prior to the reaction with compound (D) or compound (L), said hydroxyalkyl starch thus having a structure according to formula (I)
  • the present invention relates to a hydroxyalkyl starch derivative as described above wherein the reducing end of the hydroxyalkyl starch is oxidized prior to the reaction with compound (D) or compound (L), said hydroxyalkyl starch thus having a structure according to formula (IIa)
  • the present invention relates to a hydroxyalkyl starch derivative as described above wherein the reducing end is oxidized by an alkaline iodine solution.
  • the present invention relates to a hydroxyalkyl starch derivative as described above wherein hydroxyalkyl starch is reacted with a compound (L) via the reaction of functional group Z 1 with the optionally oxidized reducing end of the hydroxyalkyl starch and the resulting reaction product is reacted with a further compound (M) via the reaction of the functional group X comprised in compound (L) with the functional group Y comprised in compound (M).
  • the present invention relates to a hydroxyalkyl starch derivative as described above hydroxyalkyl starch is reacted with a compound (L) via the reaction of functional group Z 1 with the optionally oxidized reducing end of the hydroxyalkyl starch, where compound (L), prior to the reaction with hydroxyalkyl starch, is reacted with a further compound (M) via the reaction of functional group X comprised in compound (L) with the functional group Y comprised in compound (M).
  • the present invention relates to a hydroxyalkyl starch derivative as described above wherein hydroxyalkyl starch is reacted with a compound (D) via the reaction of the functional group Z 1 comprised in compound (D), with the optionally oxidized reducing end of the hydroxyalkyl starch to give a first hydroxyalkyl starch derivative, and where the first hydroxyalkyl starch derivative is reacted with a compound (L) via the reaction of functional group Z 2 comprised in compound (L) with the functional group W comprised in compound (D) to give a second hydroxyalkyl starch derivative.
  • the present invention relates to the aforesaid hydroxyalkyl starch derivative wherein the second hydroxyalkyl starch derivative is reacted with a further compound (M) via the reaction of functional group X comprised in compound (L) with the functional group Y comprised in compound (M).
  • the present invention relates to a hydroxyalkyl starch derivative as described above wherein hydroxyalkyl starch is reacted with a compound (D) via the reaction of functional group Z 1 comprised in compound (D) with the optionally oxidized reducing end of the hydroxyalkyl starch to give a first hydroxyalkyl starch derivative, and where the first hydroxyalkyl starch derivative is reacted, via the reaction of the functional group W, comprised in compound (D), and the functional group Z 2 , comprised in compound (L), with compound (L), where compound (L), prior to the reaction with the first hydroxyalkyl starch derivative, is reacted with a further compound (M) via the reaction of functional group X comprised in compound (L) with the functional group Y comprised in compound (M).
  • the present invention relates to a hydroxyalkyl starch derivative as described above wherein the at least one further compound (M) is a polypeptide.
  • the present invention relates to a hydroxyalkyl starch derivative as described above wherein the polypeptide is erythropoietin.
  • the hydroxyalkyl starch derivative which in the following is referred to as HAS-EPO conjugate and which is formed by reaction of hydroxyalkyl starch with compound (L) and optionally compound (D) and erythrpoietin, has the advantage that it exhibits an improved biological stability when compared to the erythropoietin before conjugation. Furthermore, it exhibits a higher biological activity than standard BRP EPO. This is mainly due to the fact that this hydroxyalkyl starch derivative is less or even not recognized by the removal systems of the liver and kidney and therefore persists in the circulatory system for a longer period of time. Furthermore, since the HAS is attached site-specifically, the risk of destroying the in-vivo biological activity of EPO by conjugation of HAS to EPO is minimized.
  • the HAS-EPO conjugate of the invention may exhibit essentially the same in-vitro biological activity as recombinant native EPO, since the in-vitro biological activity only measures binding affinity to the EPO receptor. Methods for determining the in-vitro biological activity are known in the art.
  • the HAS-EPO exhibits a greater in-vivo activity than the EPO used as a starting material for conjugation (unconjugated EPO).
  • Methods for determining the in vivo biological activity are known in the art.
  • the HAS-EPO conjugate may exhibit an in vivo activity of from 110% to 500%, preferably of from 300 to 400%, or preferably of from 110 to 300%, more preferably from 110% to 200%, more preferably from 110% to 180% or from 110 to 150%, most preferably from 110% to 140%, if the in-vivo activity of the unconjugated EPO is set as 100%.
  • the HAS-EPO Compared to the highly sialylated EPO of Amgen (see EP 428 267 B1), the HAS-EPO exhibits preferably at least 50%, more preferably at least 70%, even more preferably at least 85% or at least 95%, at least 150%, at least 200% or at least 300% of the in vivo activity of the highly sialylated EPO if the in-vivo activity of highly sialylated EPO is set as 100%. Most preferably, it exhibits at least 95% of the in vivo activity of the highly sialylated EPO.
  • the high in-vivo biological activity of the HAS-EPO conjugate of the invention mainly results from the fact that the HAS-EPO conjugate remains longer in the circulation than the unconjugated EPO because it is less recognized by the removal systems of the liver and because renal clearance is reduced due to the higher molecular weight.
  • Methods for the determination of the in-vivo half life time of EPO in the circulation are known in the art (Sytkowski, Lurn, Davis, Feldman, Siekman, 1998, Human erythropoietin dhiers with markedly enhanced in vivo activity, Proc. Natl. Acad. Sci. USA, 95(3), 1184-8).
  • HAS-EPO conjugate which may be administered less frequently than the EPO preparations commercially available at present. While standard EPO preparations have to be administered at least every 3 days, the HAS-EPO conjugate of the invention is preferable administered twice a week, more preferably once a week.
  • the method of the invention has the advantage that an effective EPO derivative can be produced at reduced costs since the method does not comprise extensive and time consuming purification steps resulting in low final yield, e.g. it is not necessary to purify away under-sialylated EPO forms which are known to exhibit low or no in-vivo biological activity.
  • Example 8.11(d) demonstrates that a HES-EPO produced with few modifications steps exhibits a 3-fold activity over standard BRP EPO.
  • the present invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising, in a therapeutically effective amount, the HAS-polypeptide conjugate, preferably the HAS-EPO conjugate, more preferably the HES-EPO conjugate of the present invention.
  • the pharmaceutical composition comprises further at least one pharmaceutically acceptable diluent, adjuvant and/or carrier useful in erythropoietin therapy.
  • the present invention also relates to a pharmaceutical composition
  • a pharmaceutical composition comprising, in a therapeutically effective amount, a hydroxyalkyl starch derivative obtainable by a method of producing a hydroxyalkyl starch derivative, said hydroxyalkyl starch having a structure according to formula (I)
  • the present invention relates to the use of a hydroxyalkyl starch derivative as described for the preparation of a medicament for the treatment of anemic disorders or hematopoietic dysfunction disorders or diseases related thereto.
  • the present invention relates to a pharmaceutical composition as described above wherein the polypeptide is an antithrombin (AT), preferably AT III (Levy J H, Weisinger A, Ziomek C A, Echelard Y, Recombinant Antithrombin: Production and Role in Cardiovascular Disorder, Seminars in Thrombosis and Hemostasis 27, 4 (2001) 405-416; Edmunds T, Van Patten S M, Pollock J, Hanson E, Bemasconi R, Higgins E, Manavalan P, Ziomek C, Meade H, McPherson J, Cole E S, Transgenically Produced Human Antithrombin: Structural and Functional Comparison to Human Plasma-Derived Antithrombin, Blood 91, 12 (1998) 4661-4671; Minnema M C, Chang A C K, Jansen P M, Lubbers Y T P, Pratt B M, Whittaker B G, Taylor F B, hack C E, Friedrnan B
  • AT antithrom
  • the present invention relates to pharmaceutical compositions wherein the polypeptide is G-CSF or IFN-beta.
  • the present invention relates to a pharmaceutical composition as described above wherein the polypeptide is erythropoietin.
  • the present invention relates to a pharmaceutical composition as described above wherein the functional group Y is —SH and compound (L) is a compound of general formula Z 1 -L′-X where the functional group Z 1 is selected from the group consisting of wherein G is O or S and, if present twice, independently O or S, and R′ is methyl, and where the functional group X is selected from the group consisting of wherein Hal is Cl, Br or I, and where L′ is an organic chain bridging Z 1 and X or where L′ is absent.
  • the present invention relates to a pharmaceutical composition as described above wherein the functional group Y is selected from the group consisting of an aldehyd group, a keto group, a hemiacetal group, and an acetal group, and compound (L) is a compound of general formula Z 1 -L′-X where the functional group Z 1 is selected from the group consisting of wherein G is O or S and, if present twice, independently O or S, and R′ is methyl, and where the functional group X is selected from the group consisting of wherein G is O or S and, if present twice, independently O or S, and R′ is methyl, and where L′ is an organic chain bridging Z 1 and X or where L′ is absent.
  • the present invention relates to a pharmaceutical composition as described above wherein the functional group Y is —SH, compound (D) is a compound of general formula Z 1 -D′-W, and compound (L) is a compound of general formula Z 2 -L′-X, where the functional group Z 1 is selected from the group consisting of wherein G is O or S and, if present twice, independently O or S, and R′ is methyl, where the functional group X is selected from the group consisting of wherein Hal is Cl, Br or I, where the functional group W or the functional group Z 2 is —SH and the functional group Z 2 or the functional group W is selected from the group consisting of wherein Hal is Cl, Br, or I, or where the functional group W or the functional group Z 2 is selected from the group consisting of an activated ester, as described above, or a carboxy group which is optionally transformed into an activated ester and the functional group Z 2 or the functional group W is selected from the group consisting of wherein G is O or S and,
  • the present invention relates to a pharmaceutical composition as described above wherein the functional group Y is selected from the group consisting of an aldehyd group, a keto group, a hemiacetal group, and an acetal group, compound (D) is a compound of general formula Z 1 -D′-W, and compound (L) is a compound of general formula Z 2 -L′-X, where the functional group Z 1 is selected from the group consisting of wherein G is O or S and, if present twice, independently O or S, and R′ is methyl, where the functional group X is selected from the group consisting of wherein G is O or S and, if present twice, independently O or S, and R′ is methyl, the functional group W or the functional group Z 2 is —SH and the functional group Z 2 or the functional group W is selected from the group consisting of wherein Hal is Cl, Br, or I., or where the functional group W or the functional group Z 2 is selected from the group consisting of an activated ester,
  • the present invention relates to a pharmaceutical composition as described above wherein hydroxyethyl starch is reacted in an aqueous medium with a compound according to the following formula and the reaction product is reacted with erythropoietin.
  • the present invention relates to the aformentioned pharmaceutical composition wherein the erythropoietin is oxidised with sodium periodate prior to the reaction.
  • the present invention relates to pharmaceutical composition as described above wherein the erythropoietin is partially desialylated and subsequently oxidised with sodium periodate prior to the reaction.
  • compositions comprising a hydroxyalkyl starch derivative which are produced on the basis of a completely reduced Thio-EPO according to Example 6 are excluded.
  • the above-mentioned pharmaceutical composition is especially suitable for the treatment of anemic disorders or hematopoietic dysfunction disorders or diseases related thereto.
  • a “therapeutically effective amount” as used herein refers to that amount which provides therapeutic effect for a given condition and administration regimen.
  • the administration of erythropoietin isoforms is preferably by parenteral routes. The specific route chosen will depend upon the condition being treated.
  • the administration of erythropoietin isoforms is preferably done as part of a formulation containing a suitable carrier, such as human serum albumin, a suitable diluent, such as a buffered saline solution, and/or a suitable adjuvant.
  • the required dosage will be in amounts sufficient to raise the hematocrit of patients and will vary depending upon the severity of the condition being treated, the method of administration used and the like.
  • the object of the treatment with the pharmaceutical composition of the invention is preferably an increase of the hemoglobin value of more than 6.8 mmol/l in the blood.
  • the pharmaceutical composition may be administered in a way that the hemoglobin value increases between from 0.6 mmol/l and 1.6 mmol/l per week. If the hemoglobin value exceeds 8.7 mmol/l, the therapy should be preferably interrupted until the hemoglobin value is below 8.1 mmol/l.
  • composition of the invention is preferably used in a formulation suitable for subcutaneous or intravenous or parenteral injection.
  • suitable excipients and carriers are e.g. sodium dihydrogen phosphate, disodium hydrogen phosphate, sodium chlorate, polysorbate 80, HSA and water for injection.
  • the composition may be administered three times a week, preferably two times a week, more preferably once a week, and most preferably every two weeks.
  • the pharmaceutical composition is administered in an amount of 0.01-10 ⁇ g/kg body weight of the patient, more preferably 0,1 to 5 ⁇ g/kg, 0,1 to 1 ⁇ g/kg, or 0.2-0.9 ⁇ g/kg, most preferably 0.3-0.7 ⁇ g/kg, and most preferred 0.4-0.6 ⁇ g/kg body weight.
  • the invention further relates to a HAS-polypeptide according to the present invention for use in method for treatment of the human or animal body.
  • the invention further relates to the use of a HAS-EPO conjugate of the present invention for the preparation of a medicament for the treatment of anemic disorders or hematopoietic dysfunction disorders or diseases related hereto.
  • compound (II) may be present in R conformation or in S conformation or as racemic compound with respect to each chiral center.
  • compound (D) optionally used in the present invention comprises one or more chiral centers
  • compound (D) may be present in R conformation or in S conformation or as racemic compound with respect to each chiral center.
  • FIG. 1 A first figure.
  • FIG. 1 shows an SDS page analysis of the HES-EPO conjugate, produced according to example 5.1.
  • Lane A Protein marker Roti®-Mark PRESTAINED (Carl Roth GmbH+Co, Düsseldorf, D); molecular weights (in kD) of the protein marker from top to bottom: 245, 123, 77, 42, 30, 25.4, and 17.
  • Lane B Crude product after conjugation according to example 5.1.
  • Lane C EPO starting material.
  • FIG. 2 shows an SDS page analysis of the HES-EPO conjugate, produced according to example 5.3.
  • Lane A Crude product after conjugation according to example 5.3.
  • Lane B EPO starting material.
  • Lane C Protein marker Roti®-Mark PRESTAINED (Carl Roth GmbH+Co, Düsseldorf, D); molecular weights (in kD) of the protein marker from top to bottom: 245, 123, 77, 42, 30, 25.4, and 17.
  • FIG. 3 shows an SDS page analysis of the HES-EPO conjugate, produced according to example 5.4 and 5.5.
  • Lane A Protein marker Roti®-Mark PRESTAINED (Carl Roth GmbH+Co, Düsseldorf, D); molecular weights (in kD) of the protein marker from top to bottom: 245, 123, 77, 42, 30, 25.4, and 17.
  • Lane B Crude product after conjugation according to example 5.4.
  • Lane C Crude product after conjugation according to example 5.5.
  • Lane D EPO starting material.
  • FIG. 4 shows an SDS page analysis of HES-EPO conjugates, produced according to examples 7.1 and 7.4.
  • Lane A Protein marker Roti®-Mark PRESTADMD (Carl Roth GmbH+Co, Karisruhe, D); molecular weights (in kD) of the protein marker from top to bottom: 245, 123, 77, 42, 30, 25.4, and 17.
  • Lane B Crude product after conjugation according to example 7.4.
  • Lane C Crude product after conjugation according to example 7.1.
  • Lane D EPO starting material.
  • FIG. 5 shows an SDS page analysis of HES-EPO conjugates, produced according to examples 7.2, 7.3, 7.5, and 7.6.
  • Lane A Protein marker Roti®-Mark PRESTAINED (Carl Roth GmbH+Co, Düsseldorf, D); molecular weights (in kD) of the protein marker from top to bottom: 245, 123, 77, 42, 30, 25.4, and 17.
  • Lane B Crude product after conjugation according to example 7.6, based on Example 1.3 b).
  • Lane C Crude product after conjugation according to example 7.5, based on Example 1.1 b).
  • Lane D Crude product after conjugation according to example 7.6, based on Example 1.3 a).
  • Lane E Crude product after conjugation according to example 7.5, based on Example 1.1 a).
  • Lane F Crude product after conjugation according to example 7.2.
  • Lane G Crude product after conjugation according to example 7.3.
  • FIG. 6 shows an SDS page analysis of HES-EPO conjugates, produced according to examples 7.7, 7.8, 7.9, 7.10, 7.11, and 7.12.
  • Lane B Crude product after conjugation according to example 7.11.
  • Lane D Crude product after conjugation according to example 7.7.
  • Lane E Crude product after conjugation according to example 7.8.
  • Lane F Crude product after conjugation according to example 7.12.
  • Lane G EPO starting material.
  • Lane K Crude product after conjugation according to example 7.9.
  • HPAEC-PAD pattern of oligosaccharides isolated from untreated EPO and from EPO incubated for 5 min., 10 min. and 60 min. under mild acid hydrolysis conditions.
  • the elution area of oligosaccharides structures without, with 1-4 sialic acid is indicated by brackets.
  • HPAEC-PAD HPAEC-PAD pattern of native oligosaccharides isolated from untreated EPO and from EPO incubated for 5 min and 10 min under mild acid hydrolysis conditions and subsequent periodate treatment.
  • the elution area of oligosaccharides structures without and with 1-4 sialic acid is indicated by brackets 1-5.
  • FIGS. 17 to 23 represent MALDI/TOF mass spectra of the enzymatically liberated and chemically desialylated N-glycans isolated from HES-modified EPO and control EPO preparations.
  • Major signals at m/z 1809.7, 2174.8, 2539.9, 2905.0 and 3270.1 correspond to di- to tetraantennary complex-type N-glycan structures with no, one or two N-acetyllactosamine repeats accompanied by weak signals due to loss of fucose or galactose which are due to acid hydrolysis conditions employed for the desialylation of samples for MS analysis.
  • MALDI/TOF spectrum desialylated oligosaccharides of HES-modified EPO A2.
  • MALDI/TOF spectrum desialylated oligosaccharides of EPO GT-1-A.
  • MALDI/TOF spectum desialylated oligosaccharides of EPO K2.
  • MALDI/ROF spectum desialylated oligosaccharides of EPO-GT-1.
  • MALDI/TOF spectrum desialylated oligosaccharides of EPO-GT-1 subjected to acid hydrolysis for 5 min.
  • MALDI/TOF spectrum desialylated oligosaccharides of EPO-GT-1 subjected to acid hydrolysis for 10 min.
  • MALDI/TOF spectrum desialylated oligosaccharides of EPO-GT-1 subjected to acid hydrolysis for 60 min.
  • FIG. 24 shows an SDS page analysis of two HES-EPO conjugates
  • Lane 1 HES-EPO produced according to example protocol 8B: EPO is conjugated to hydrazido-HES 12KD L
  • Lane 2 HES-EPO produced according to example protocol 9B: EPO is conjugated to hydroxylamino HES 12 KD K
  • control unconjugated EPO
  • the upper band represents EPO dimer
  • FIG. 25 demonstrates that the HES is conjugated to a carbohydrate moiety of a carbohydrate side chain by showing a digestion of HAS modified EPO forms with polypeptide N-glycosidase
  • Lane 1 HES-EPO produced according to example protocol 8B after digestion with N-glycosidase
  • Lane 2 HES-EPO produced according to example protocol 9B after digestion with N-glycosidase
  • mw marker (Bio-Rad SDS-PAGE Standards Low range Catalog No 161-0305, Bio-Rad Laboratories, Hercules, Calif., USA)
  • the degree of substitution relates to the molar substitution, as described above (see also Sommermeyer et al., 1987, Whypharmazie, 8(8), 271-278, in particular p. 273).
  • the DS of the HES18/04 when measured according to Sommermeyer et al., 1987, Rohpharmazie, 8(8), 271-278 was 0,5.
  • the precipitated product was collected by centrifugation, re-dissolved in 40 ml water,.and dialysed for 3 d against water (SnakeSkin dialysis tubing, 3.5 KD cut off, Perbio Science GmbH, Bonn, D), and lyophilized.
  • the precipitated product was collected by centrifugation, re-dissolved in 40 ml water, and centrifugated for 15 min at 4,500 rpm.
  • the clear supernatant was dialysed for 3 d against water (SnakeSkin dialysis tubing, 3.5 KD cut off, Perbio Science GmbH, Bonn, D), and lyophilized.
  • DMSO absolute dimethyl sulfoxide
  • DMSO dimethyl sulfoxide
  • the precipitated product was collected by centrifugation, redissolved in 40 mL of water and dialysed for 2 days against a 0.5% (v/v) triethylamine in water solution and for 2 days against water (SnakeSkin dialysis tubing, 3.5 KD cut off, Perbio Science GmbH, Bonn, Germany) and lyophilized.
  • the precipitated product was collected by centrifugation, redissolved in 40 ml of water and dialysed for 4 days against water (SnakeSkin dialysis tubing, 3.5 KD cut off, Perbio Science GmbH, Bonn, Germany) and lyophilized.
  • reaction mixture was added to 200 ml of water
  • solution containing the reaction product was dialysed for 2 days against a 0.5% (v/v) triethylamine in water solution and for 2 days against water (SnakeSkin dialysis tubing, 3.5 KD cut off, Perbio Science GmbH, Bonn, Germany) and lyophilized.
  • the precipitate Amino-HESIOKD/0.4 was collected by centrifugation, redissolved in 40 ml of water and dialysed for 4 days against water (SnakeSkin dialysis tubing, 3.5 KD cut off, Perbio Science GmbH, Bonn, Germany) and lyophilized.
  • Oxidized erythropoietin was produced as described in Example 8.
  • EPO-GT-1-A as described in Example 8.11(c) was used (EPO-GT-1 without acid hydroylsis, treated with mild periodate oxidation).
  • Oxidized EPO (1.055 ⁇ g/ ⁇ l) in 20 mM PBS buffer was adjusted to pH 5.3 with 5 M sodium acetate buffer, pH 5.2.
  • FIG. 1 The experimental result is shown in FIG. 1 .
  • a successful conjugation is indicated by the migration of the protein band to higher molecular weights.
  • the increased bandwidth is due to the molecular weight distribution of the HES derivatives used and the number of HES derivatives linked to the protein.
  • Oxidized EPO (1.055 ⁇ g/ ⁇ L) in 20 mM PBS buffer was adjusted to pH 5.3 with 5 M sodium acetate buffer, pH 5.2.
  • Oxidized EPO (1.055 ⁇ g/ ⁇ l) in 20 mM PBS buffer was adjusted to pH 5.3 with 5 M sodium acetate buffer, pH 5.2.
  • 18 ⁇ l of a solution of the HES derivate as produced according to example 2.4 was added, and the mixture was incubated for 16 h at 25° C.
  • the crude product was analyzed by SDS-Page with NUPAGE 10% Bis-Tris Gels/MOPS buffer (Invitrogen, Carlsbad, Calif., USA) as described in the instructions given by Invitrogen. The gel is stained with Roti-Blue Coomassie staining reagent (Roth, Düsseldorf, D) overnight.
  • the experimental result is shown in FIG. 2 .
  • a successful conjugation is indicated by the migration of the protein band to higher molecular weights.
  • the increased bandwidth is due to the molecular weight distribution of the HES derivatives used and the number of HES derivatives linked to the protein.
  • Oxidized EPO (1.055 ⁇ g/ ⁇ l) in 20 mM PBS buffer was adjusted to pH 5.3 with 5 M sodium acetate buffer, pH 5.2.
  • 18 ⁇ l of a solution of the HES derivate as produced according to example 3.1 was added, and the mixture was incubated for 16 h at 25° C.
  • the crude product was analyzed by SDS-Page with NUPAGE 10% Bis-Tris Gels/MOPS buffer (Invitrogen, Carlsbad, Calif., USA) as described in the instructions given by Invitrogen. The gel is stained with Roti-Blue Coomassie staining reagent (Roth, Düsseldorf, D) overnight.
  • the experimental result is shown in FIG. 3 .
  • a successful conjugation is indicated by the migration of the protein band to higher molecular weights.
  • the increased bandwidth is due to the molecular weight distribution of the HES derivatives used and the number of HES derivatives linked to the protein.
  • Oxidized EPO (1.055 ⁇ g/ ⁇ l) in 20 mM PBS buffer was adjusted to pH 5.3 with 5 M sodium acetate buffer, pH 5.2.
  • 18 ⁇ l of a solution of the HES derivate as produced according to example 3.1 was added, and the mixture was incubated for 16 h at 25° C.
  • the crude product was analyzed by SDS-Page with NuPAGE 10% Bis-Tris Gels/MOPS buffer (Invitrogen, Carlsbad, Calif., USA) as described in the instructions given by Invitrogen. The gel is stained with Roti-Blue Coomassie staining reagent (Roth, Düsseldorf, D) overnight.
  • the experimental result is shown in FIG. 3 .
  • a successful conjugation is indicated by the migration of the protein band to higher molecular weights.
  • the increased bandwidth is due to the molecular weight distribution of the HES derivatives used and the number of HES derivatives linked to the protein.
  • EPO-GT-1 erythropoietin
  • a 0.1 M sodium borate buffer 5 mM EDTA, 10 mM DTT (Lancaster, Morcambe, UK), pH 8.3, were incubated for 1 h at 37° C.
  • the DTT was removed by centrifugal filtration with a VIVASPIN 0.5 ml concentrator, 10 KD MWCO (VIVASCIENCE, Hannover, D) at 13,000 rpm, subsequent washing 3 times with the borate buffer and twice with a phosphate buffer (0.1 M, 9.15 M NaCl, 50 mM EDTA, pH 7.2).
  • the gel is stained with Roti-Blue Coomassie staining reagent (Roth, Düsseldorf, D) overnight.
  • N-(alpha-maleimidoacetoxy) succinimide ester was used as crosslinking compound.
  • the experimental result is shown in FIG. 4 .
  • a successful conjugation is indicated by the migration of the protein band to higher molecular weights.
  • the increased bandwidth is due to the molecular weight distribution of the HES derivatives used and the number of HES derivatives linked to the protein.
  • the experimental result is shown in FIG. 5 .
  • a successful conjugation is indicated by the migration of the protein band to higher molecular weights.
  • the increased bandwidth is due to the molecular weight distribution of the HES derivatives used and the number of HES derivatives linked to the protein.
  • the experimental result is shown in FIG. 5 .
  • a successful conjugation is indicated by the migration of the protein band to higher molecular weights.
  • the increased bandwidth is due to the molecular weight distribution of the HES derivatives used and the number of HES derivatives linked to the protein.
  • the experimental result is shown in FIG. 4 .
  • a successful conjugation is indicated by the migration of the protein band to higher molecular weights.
  • the increased bandwidth is due to the molecular weight distribution of the HES derivatives used and the number of HES derivatives linked to the protein.
  • Remaining AMAS was removed by centrifugal filtration with a VIVASPIN 0.5 ml concentrator, 5 KD MWCO (VIVASCIENCE, Hannover, D) at 13,000 rpm, washing 4 times and 30 min with the phosphate buffer.
  • the experimental result is shown in FIG. 5 .
  • a successful conjugation is indicated by the migration of the protein band to higher molecular weights.
  • the increased bandwidth is due to the molecular weight distribution of the HES derivatives used and the number of HES derivatives linked to the protein.
  • Remaining AMAS was removed by centrifugal filtration with a VIVASPIN 0.5 ml concentrator, 5 KD MWCO (VIVASCIENCE, Hannover, D) at 13,000 rpm, washing 4 times and 30 min with the phosphate buffer.
  • the experimental result is shown in FIG. 5 .
  • a successful conjugation is indicated by the migration of the protein band to higher molecular weights.
  • the increased bandwidth is due to the molecular weight distribution of the HES derivatives used and the number of HES derivatives linked to the protein.
  • the experimental result is shown in FIG. 6 .
  • a successful conjugation is indicated by the migration of the protein band to higher molecular weights.
  • the increased bandwidth is due to the molecular weight distribution of the HES derivatives used and the number of HES derivatives linked to the protein.
  • the experimental result is shown in FIG. 6 .
  • a successful conjugation is indicated by the migration of the protein band to higher molecular weights.
  • the increased bandwidth is due to the molecular weight distribution of the HES derivatives used and the number of HES derivatives linked to the protein.
  • the experimental result is shown in FIG. 6 .
  • a successful conjugation is indicated by the migration of the protein band to higher molecular weights.
  • the increased bandwidth is due to the molecular weight distribution of the HES derivatives used and the number of HES derivatives linked to the protein.
  • the experimental result is shown in FIG. 6 .
  • a successful conjugation is indicated by the migration of the protein band to higher molecular weights.
  • the increased bandwidth is due to the molecular weight distribution of the HES derivatives used and the number of HES derivatives linked to the protein.
  • the experimental result is shown in FIG. 6 .
  • a successful conjugation is indicated by the migration of the protein band to higher molecular weights.
  • the increased bandwidth is due to the molecular weight distribution of the HES derivatives used and the number of HES derivatives linked to the protein.
  • the experimental result is shown in FIG. 6 .
  • a successful conjugation is indicated by the migration of the protein band to higher molecular weights.
  • the increased bandwidth is due to the molecular weight distribution of the HES derivatives used and the number of HES derivatives linked to the protein.
  • HES-EPO conjugates were synthesized by coupling of HES derivatives (average mw of 18,000 Dalton; hydroxyethyl substitution degree of 0.4) to the partially (mild periodate) oxidized sialic acid residues on the oligosaccharide chains of recombinant human EPO. Based on carbohydrate structural analysis the modifications introduced did not affect the structural integrity of the core oligosaccharide chains since MALDI/TOF-MS of the mild acid treated HES-modified glycans revealed intact neutral N-acetyllactosamine-type chains which were indistinguishable from those observed in unmodified EPO product.
  • the results obtained indicate that at least 3 modified HES-residues are attached per EPO molecule in the case of the EPO preparation which was subjected to modification without prior partial sialic acid removal.
  • An EPO variant lacking about 50% of the sialic acid residues of the former protein showed a similar apparent high molecular weight mobility in SDS-PAGE (60-110 KDa vs 40 KDa for the BRP EPO standard).
  • the HES modified EPO is stable under standard ion-exchange chromatography conditions at room temperature at pH 3-10.
  • the EPO-bioassay in the normocythaemic mouse system indicates that the HES-modified EPO has 2.5-3.0 fold higher specific activity (IU/mg) in this assay when compared to the International BRP EPO reference standard based on protein determination using the UV absorption value from the European Pharmacopeia and an RP-HPLC EPO protein determination method calibrated against the BRP EPO standard preparation.
  • the oligosaccharides in the pooled supernatants were dried in a vacuum centrifuge (Speed Vac concentrator, Savant Instruments Inc., USA).
  • the glycan samples were desalted using Hypercarb cartridges (25 mg or 100 mg of HyperCarb) as follows prior to use: the columns were washed with 3 ⁇ 500 ⁇ l of 80% acetonitrile (v/v) in 0.1% TFA followed by washes with 3 ⁇ 500 ⁇ l of water.
  • the samples were diluted with water to a final volume of 300 ⁇ l-600 ⁇ l before loading onto the cartridge which then was rigorously washed with water.
  • Oligosaccharides were eluted with 1.2 ml (25 mg cartridges; 1.8 ml in the case of 100 mg cartridges) 25% acetonitrile in water containing 0.1% trifluoroacetic acid (v/v).
  • the eluted oligosaccharides were neutralized with 2 M NH 4 OH and were dried in a Speed Vac concentrator.
  • desalting of N-glycosidase released oligosaccharides was performed by adsorption of the digestion mixture from samples ⁇ 100 ⁇ g of total (glyco)protein onto 100 mg Hypercarb cartridges.
  • TOF/TOF time-of-flight
  • EPO was expressed from recombinant CHO cells as described (Mueller P P et al., 1999, Dorner A J et al., 1984) and the preparations were characterized according to methods described in the Eur. Phar. ( Ph. Eur. 4, Monography January 2002:1316: Erythropoietin concentrated solution ). The final product had a sialic acid content of 12 nMol ( ⁇ 1.5 nMol) per nMol of protein.
  • the structures of N-linked oligosaccharides were determined by HPAEC-PAD and by MALDI/TOF-MS as described Nimtz et al., 1999, Grabenhorst, 1999).
  • the EPO preparations that were obtained contained di-, tri- and tetrasialylated oligosaccharides (2-12%, 15-28% and 60-80%, respectively, sulphated and pentasialylated chains were present in small amounts).
  • the overall glycosylation characteristics of EPO preparations were similar to that of the international BRP EPO standard preparation.
  • the isoelectric focusing pattern of the recombinant EPO was comparable to that of the international BRP Reference EPO standard preparation showing the corresponding isoforms. 25% of the EPO protein lacked O-glycosylation at Ser 126 of the polypeptide chain.
  • EPO GT-1 protein (2.84 mg/ml) was heated to 80° C. in 20 mM Na-phosphate buffer pH 7.0 and then 100 ⁇ l of 1 N H 2 SO 4 was added per 1 ml of the EPO solution; incubation was continued for 5 min, 10 min and 60 min, respectively, yielding EPO preparations of different degree of sialylation. Quantitation of oligosaccharides with 0-4 sialic acids was performed after liberation of oligosaccharides with polypeptide N-glycosidase and isolation of N-linked chains was performed by desalting using Hypercarb cartridges (25 mg HyperSep Hypercarb; ThermoHypersil-Keystone, UK). EPO preparations were neutralized by addition of 1 N NaOH and were frozen in liquid N 2 and were stored at ⁇ 20° C. until further use.
  • the partially oxidized EPO forms were separated from reagents by desalting using VIVASPIN concentrators (10,000 MWCO, PES Vivascience AG, Hannover, Germany) according to manufacturer's recommendation at 3000 rpm in a laboratory centrifuge equipped with a fixed angle rotor. After freezing in liquid nitrogen the EPO preparations were stored in a final volume of 4 ml at ⁇ 20° C.
  • VIVASPIN concentrators 10,000 MWCO, PES Vivascience AG, Hannover, Germany
  • oligosaccharides were isolated using Hypercarb cartridges as described. Oligosaccharides were desialylated by mild acid treatment and were analyzed by HPAEC-PAD and their retention times were compared to those of authentic standard oligosaccharides as described (Nimtz et al., 1990 and 1993).
  • EPO-GT-1 5 mg was incubated in 5 ml of 0.1 M Tris/HCl buffer pH 8.1 in the presence of 30 mM dithioerythreitol (DTT) at 37° C. for 60 minutes; removal of DTT was achieved by using a Vivaspin concentrator at 4° C., 4 cycles of buffer exchange.
  • DTT dithioerythreitol
  • EPO protein Quantitative determination of EPO protein was performed by measuring UV absorption at 280 nm according to the Eur. Phar. (European Pharmacopeia 4, Monography January 2002: 1316: erythropoietin concentrated solution) in a cuvette with 1 cm path length.
  • EPO was quantitated by applying a RP-HPLC method using a RP-C4 column (Vydac Protein C4, Cat.# 214TP5410, Grace Vydac, Calif., US); the HPLC method was calibrated using the erythropoietin BRP 1 reference standard (European Pharmacopeia, Riverside de l'Europe B.P. 907-F67029, France Cedex 1).
  • EPO incubation mixtures (approximately 5 mg of EPO protein) were diluted 1:10 with buffer A (20 mM N-morpholine propane sulfonic acid [MOPS/NaOH] in H 2 O bidest, pH 8.0) and were applied to a column containing 3 ml Q-Sepharose HP (Pharmacia Code no. 17-1014-03, Lot no. 220211) equilibrated with 10 column volumes (CV) of buffer A by using a flow rate of 0.5 ml/min.
  • buffer A 20 mM N-morpholine propane sulfonic acid [MOPS/NaOH] in H 2 O bidest, pH 8.0
  • buffer B 20 mM morpholine ethane sulfonic acid [MES/NaOH], 0.5 M NaCl in H 2 O bidest, pH 6.5
  • EPO was detected by UV absorption at 280 nm and eluted in about 6 ml.
  • Buffer exchange of EPO eluates obtained from the Q-Sepharose step was performed using Vivaspin concentrators and phosphate buffered saline (PBS) with each 3 centrifugation cycles per sample; samples were adjusted to 2 ml with PBS and were stored at ⁇ 20° C.
  • PBS phosphate buffered saline
  • HPAEC-PAD High-pH Anion-Exchange Chromatography with Pulsed Amperometric Detection
  • Purified native and desialylated oligosaccharides were analyzed by high-pH anionexchange (HPAE) chromatography using a Dionex BioLC system (Dionex, USA) equipped with a CarboPac PA1 column (0.4 ⁇ 25 cm) in combination with a pulsed amperometric detector (PAD) (Schröter et al., 1999; Nimtz et al., 1999).
  • HPAE high-pH anionexchange
  • Detector potentials (E) and pulse durations (T) were: E1: +50 mV, T1: 480 ms; E2: +500 mV, T2: 120 ms; E3: ⁇ 500 mV, T3: 60 ms, and the output range was 500-1500 nA.
  • the oligosaccharides were then injected onto the CarboPac PA1 column which was equilibrated with 100% solvent A.
  • For desialylated oligosaccharides elution (flow rate: 1 ml-min ⁇ 1 ) was performed by applying a linear gradient (0-20%) of solvent B over a period of 40 min followed by a linear increase from 20-100% solvent B over 5 min.
  • Solvent A was 0.2 M NaOH in bidistilled H 2 O
  • solvent B consisted of 0.6 M NaOAc in solvent A
  • elution flow rate: 1 ml-min ⁇ 1
  • Solvent D consisted of 0.6 M NaAc in solvent C.
  • Monosaccharides were identified by their retention time and characteristic fragmentation pattern. The uncorrected results of electronic peak integration were used for quantification. Monosaccharides yielding more than one peak due to anomericity and/or the presence of furanoid and pyranoid forms were quantified by adding all major peaks. 0.5 ⁇ g of myo-inositol was used as an internal standard compound.
  • EPO-GT-1 preparations subjected to mild acid treatment for 5, 10 or 60 min. were analyzed by SDS-PAGE before and after liberation of N-linked oligosaccharides by incubation with N-glycosidase as shown in FIG. 7 .
  • N-linked oligosaccharides were subjected to HPAEC-PAD oligosaccharide mapping ( FIG. 8 ).
  • the untreated EPO-GT-1 contained >90% of N-linked oligosaccharides with 3 or 4 sialic acid residues whereas after 5 min. of incubation in the presence of mild acid ⁇ 40% of carbohydrate chains had 3 or 4 sialic acid residues.
  • HPAEC-PAD of the desialylated N-glycans revealed that the ratio of neutral oligosaccharides that were detected for the untreated EPO-GT-1 and remained stable in the preparations subjected to acid treatment for 5, 10 or 60 min.
  • MALDI/TOF-MS of the desialylated glycans revealed that ⁇ 90% of the proximal fucose was present after mild acid treatment of the protein.
  • hydroxylamine-modified HES derivative X 400 ⁇ g was added to 20 ⁇ g of EPO-GT-1-A (mild periodate oxidized EPO, not acid hydrolyzed prior to mild periodate oxidation) in 20 ⁇ L of 0.5 M NaOAc buffer pH 5.5 and the reaction was stopped after 30 min, 2, 4, and 17 hours, respectively, by freezing samples in liquid nitrogen. Subsequently samples were stored at ⁇ 20° C. until further analysis.
  • HES-EPO conjugates I originating from EPO-GT-1 after mild periodate oxidation, i.e. from EPO-GT-1-A
  • II resulting from EPO-GT-1 subjected to 5 min acid hydrolysis and mild periodate oxidation
  • III resulting from EPO-GT-1 subjected to 10 min acid hydrolysis and mild periodate oxidation
  • K was included containing unmodified EPO-GT-1 under the same buffer conditions to which an equivalent amount of unmodified HES was added. The incubation mixtures were subjected to further purification for subsequent biochemical analysis of the HES-EPO derivatives.
  • HES-modified EPO sample A and K were compared to periodate oxidized form EPO-GT-1-A.
  • the samples were subjected to N-glycosidase treatment and as is depicted in FIGS. 14 a and 14 b the release of N-glycans resulted in the two low molecular weight bands at the position of the O-glycosylated and nonglycosylated EPO forms of the standard EPO preparation.
  • sample A a further band migrating at the position of the 28 KDa mw standard was detected suggesting HES-modification at the O-glycan of this EPO variant (cf. Example 8.11(c)(aa)).
  • the oligosaccharide fractions from the RP-C18 step of N-glycosidase-treated sample A, EPO GT-1-A and sample K were neutralized and subjected to desalting using Hypercarb cartridges as described before.
  • the isolated oligosaccharides were subjected to HPAEC-PAD mapping before (see FIGS. 15 ) and after mild acid treatment under conditions which enabled quantitative removal of sialic acids from glycans (see FIGS. 16 ).
  • the HPAEC-PAD profile for the native material obtained from the HES-modified sample A showed only neglectable signals for oligosaccharides whereas EPO GT-1-A-derived oligosaccharides exhibited the same glycan profile as the one shown in FIG. 11 (sample named EPO-GT-1 after mild periodate treatment).
  • the elution profile of oligosaccharides obtained from the control EPO sample (K) yielded the expected pattern (compare profile in FIG. 8 ).
  • the native oligosaccharide profile of the international BRP-EPO standard is included for comparison and as reference standard.
  • EPO samples were digested with N-glycosidase and the EPO protein was adsorbed onto RP-C18 cartridges whereas oligosaccharide material was washed off as described above.
  • oligosaccharide material was washed off as described above.
  • glucose and hydroxyethylated glucose derivatives were detected only in the EPO protein which was subjected to HES-modification at cysteine residues and in oligosaccharide fractions of EPO sample A2.
  • the EPO-bioassay in the normocythaemic mouse system indicates was performed according to the procedures described in the European Pharmacopeia; the laboratory that carried out the EPO assay was using the International BRP EPO reference standard preparation.
  • the HES-modified EPO A2 preparation a mean value for the specific activity of 294,600 units per mg EPO of protein was determined indicating an approximately 3-fold higher specific activity when compared to the International BRP EPO reference standard preparation that was included in the samples sent for activity assays.
  • Recombinant EPO was produced in CHO cells as follows
  • a plasmid harbouring the human EPO cDNA was cloned into the eukaryotic expression vector (pCR3 and named afterwards pCREPO).
  • Site directed mutagenesis was performed using standard procedures as described (Grabenhorst, Nimtz, Costa et al., 1998, In vivo specificity of human alpha 1,3/4-fucosyltransferases III-VII in the biosynthesis of Lewis(x) and sialyl Lewis(x) motifs on complex-type N-glycans—Coexpression studies from BHK-21 cells together with human beta-trace protein, J. Biol. Chem., 273(47), 30985-30994).
  • CHO cells stably expressing human EPO or amino acid variants e.g. Cys-29 ⁇ Ser/Ala, or Cys-33 ⁇ Ser/Ala, Ser-126 ⁇ Ala etc.
  • amino acid variants e.g. Cys-29 ⁇ Ser/Ala, or Cys-33 ⁇ Ser/Ala, Ser-126 ⁇ Ala etc.
  • the cells were subcultivated 1:5 and selected in DMEM containing 10% FBS and 1.5 g/liter G418 sulfate.
  • EPO was produced from stable subclones in spinner flasks or in 21 perfusion reactors. Different glycoforms of EPO with different amounts of NeuAc (e.g. 2-8, 4-10, 8-12 NeuAc residues) were isolated according to published protocols using combinations various chromatographic procedures as described below.
  • NeuAc e.g. 2-8, 4-10, 8-12 NeuAc residues
  • Recombinant human EPO is produced from insect cell lines SF9 and SF 21 after infection of cells with recombinant baculovirus vector containing the human EPO cDNA under control of the polyhedrin promoter as described in the literature.
  • EPO is purified by Blue sepharose chromatography, ion-exchange chromatography on Q-Sepharose and finally RP-HPLC on C 4 -Phase.
  • Oxo-HES12KD (Fresenius German Patent DE 196 28 705 A1) are dissolved in 0.3 mL absolute dimethyl sulfoxide (DMSO) and are added dropwise under nitrogen to a mixture of 34 mg (0.15 mmol) EMCH (Perbio Science, GmbH, Bonn, Germany) in 1.5 mL DMSO. After stirring for 19 h at 60° C. the reaction mixture is added to 16 mL of a 1:1 mixture of ethanol and acetone. The precipitate is collected by centrifugation, redissolved in 3 mL DMSO and again precipitated as described. The SH-reactiv-HES12KD B is obtained by centrifugation and drying in vaccuo. The conjugation reaction with Thio-EPO is described in Example 11, 2.2. Alternatives:
  • cross-linkers which exhibit a hydrazide- and a maleimide function, separated by a spacer. Further examples for molecules of that group, available from Perbio Science, Kunststoff GmbH, Bonn, Germany, are shown in table 2; marked with an “A”. Furthermore, another group of cross-linkers exhibiting an activated disulfide function instead of a maleimide function could also be used.
  • HES12KD A 1 mg sample of HES12KD is dissolved in 3 mL of saturated ammonium bicarbonate. Additional solid ammonium bicarbonate is then added to maintain saturation of the solution during incubation for 120 h at 30° C.
  • the Amino-HES12KD C is desalted by direct lyophilization of the reaction mixture.
  • Bromoacetic anhydride is prepared as described by Thomas.3 A 1 mg sample of amino-HES12KD C is dissolved in 0.1 mL of dry DMF and cooled on ice and 5 mg bromoacetic anhydride is added. The reaction mixture is brought slowly to room temperature and the solution is stirred for 3 h. The reaction mixture is added to 1 mL of a 1:1 mixture of ethanol and acetone with ⁇ 20° C. The precipitate is collected by centrifugation, redissolved in 0.1 mL DMF and again precipitated as described. The Bromoacetamide-HES D2 (X ⁇ Br) is obtained by centrifugation and drying in vaccuo. The conjugation reaction with Thio-EPO is described in Example 11, 1.2.
  • acylation of amino groups other activated forms of halogen acidic acids can be used, e.g.
  • cross-linkers having an amino reactive group and a halogen acetyl function, separated by a spacer could be used.
  • An example thereof is SBAP. This molecule and others are available from Perbio Science GmbH, Bonn, Germany. They are marked in table 2 with an “D”.
  • cross-linkers for the ligation of amino-HES with thio-EPO without isolation of the halogenacetamide-HES derivatives see remarks in example 11, 1.2.
  • Oxo-HES12KD 1 . 44 g (0.12 mmol) of Oxo-HES12KD are dissolved in 3 mL dry dimethyl sulfoxide (DMSO) and are added dropwise under nitrogen to a mixture of 1.51 mL (15 mmol) 1,4-diaminobutane in 15 mL DMSO. After stirring for 19 h at 40° C. the reaction mixture is added to 160 mL of a 1:1 mixture of ethanol and acetone.
  • DMSO dry dimethyl sulfoxide
  • the precipitate Amino-HES12KD E is collected by centrifugation, redissolved in 40 mL of water an dialyzed for 4 days against water (SnakeSkin dialysis tubing, 3.5 KD cut off, Perbio Science GmbH, Bonn, Germany) and lyophilized.
  • Chloroacetamide-HES12KD F1 is Prepared as Described for Chloroacetamide-HES12KD D1 in 1.3 above.
  • Oxo-HES12KD 1.44 g (0.12 mmol) of Oxo-HES12KD are dissolved in 3 mL absolute dimethyl sulfoxide (DMSO) and are added dropwise under nitrogen to a mixture of 0.47 mL (15 mmol) hydrazine in 15 mL DMSO. After stirring for 19 h at 40° C. the reaction mixture is added to 160 mL of a 1:1 mixture of ethanol and acetone.
  • DMSO dimethyl sulfoxide
  • the precipitated product J is collected by centrifugation, redissolved in 40 mL of water and dialyzed for 2 days against a 0.5% (v/v) triethylamine in water solution and for 2 days against water (SnakeSkin dialysis tubing, 3.5 KD cut off, Perbio Science GmbH, Bonn, Germany) and lyophilized.
  • the conjugation reaction with oxidized Glyco-EPO is described in Example 12, 2.2.
  • derivatives can be used, wherein 2 hydrazide groups are separated by any spacer.
  • O-[2-(2-aminooxy-ethoxy)-ethyl]-hydroxylamine is synthesized as described by Boturyn et al in 2 steps from commercially available materials.5 1.44 g (0.12 mmol) of Oxo-HES12KD are dissolved in 3 mL absolute dimethyl sulfoxide (DMSO) and are added dropwise under nitrogen to a mixture of 2.04 g (15 mmol) O-[2-(2-aminooxy-ethoxy)-ethyl]-hydroxylamine in 15 mL DMSO. After stirring for 48 h at 65° C. the reaction mixture is added to 160 mL of a 1:1 mixture of ethanol and acetone.
  • DMSO dimethyl sulfoxide
  • the precipitated product K is collected by centrifugation, redissolved in 40 mL of water and dialyzed for 4 days against water (SnakeSkin dialysis tubing, 3.5 KD cut off, Perbio Science GmbH, Bonn, Germany) and lyophilized.
  • the conjugation reaction with oxidized Glyco-EPO is described in Example 12, 3.1. 5Boturyn, Boudali, Constant, Defrancq, L Subscribe, 1997 , Tetrahedron, 53, 5485
  • derivatives could be used, wherein the two hydroxylamine groups are separated by any spacer.
  • Oxo-HES12KD 1.44 g (0.12 mmol) of Oxo-HES12KD are dissolved in 3 mL absolute dimethyl sulfoxide (DMSO) and are added to a mixture of 1.16 g (15 mmol) cysteamine in 15 mL DMSO under nitrogen dropwise. After stirring for 24 h at 40° C. the reaction mixture is added to 160 mL of a 1:1 mixture of ethanol and acetone.
  • DMSO dimethyl sulfoxide
  • the precipitated product M is collected by centrifugation, redissolved in 40 mL of water and dialyzed for 2 days against a 0.5% (v/v) triethylamine in water solution and for 2 days against water (SnakeSkin dialysis tubing, 3.5 KD cut off, Perbio Science GmbH, Bonn, Germany) and lyophilized.
  • the conjugation reaction with oxidized Glyco-EPO is described in Example 12,2.1.
  • Derivatives could be used, wherein the amino group and the thio-function are separated by any spacer. Furthermore, the amino group in the derivatives could be replaced by a hydrazine, a hydrazide or a hydroxylamine.
  • the thio-function could be protected in the form of e.g. a disulfide or a trityl-derivative. However, in this case, a further deprotection step must be preformed before the conjugation, which would release a component being analogous to M.
  • the deprotection is performed in a 50 mM sodium phosphate buffer, containing 25 mM EDTA and 0.5M hydroxylamine, pH7.5 for 2 hours at room temperature and the product O is purified by dialysis against a 0.1 M sodium acetate buffer pH 5.5, containing 1 mM EDTA.
  • the deprotection reaction is performed immediately before the conjugation reaction which is described in Example 12, 2.1. b) Modification with SPDP
  • the deprotection is performed in a solution of 12 mg dithiothreitol (DTT) per 0.5 mL 100 mM sodium acetate buffer, containing 100 mM sodium chloride at pH 4.5 for 30 min at room temperature and the product Q was purified by dialysis against a 0.1 M sodium acetate buffer pH 5.5, containing 1 mM EDTA.
  • DTT dithiothreitol
  • thio-HES derivatives For the conversion of amino- to thiol-groups, either in free form or protected, several reagants are available. After the modification, the products could be isolated. Alternatively, as accepted for the use of cross-linkers, they could be directly used for the conjugation reaction, preferably after a purification step. For the isolation and storage of thio-HES derivatives, the synthesis of thio-HES derivatives in a protected form may be useful. For this, all derivatives being analogous to SATA could be used, which have an active ester-function and a thioester-function, separated by any spacer. SATP, being a further member of this group, is found in table 2, marked with an “H”.
  • the derivatives being analogous to SPDP could have an active ester-function and a disulfide-function, separated by any spacer. Further members of these groups are found in table 2, marked with an “F”. Further analogous derivatives could have an active ester-function and a thiol-function, protected as a trityl derivative, separated by any spacer.
  • Oxo-HES18KD 1.44 g (0.12 mmol) of Oxo-HES18KD were dissolved in 3 mL dry dimethyl sulfoxide (DMSO) and were added dropwise under nitrogen to a mixture of 1.51 mL (15 mmol) 1,4-diaminobutane in 15 mL DMSO. After stirring for 19 h at 40° C. the reaction mixture was added to 160 mL of a 1:1 mixture of ethanol and acetone.
  • DMSO dry dimethyl sulfoxide
  • the precipitated product J was collected by centrifugation, redissolved in 40 mL of water and dialyzed for 2 days against a 0.5% (v/v) triethylamine in water solution and for 2 days against water (SnakeSkin dialysis tubing, 3.5 KD cut off, Perbio Science GmbH, Bonn, Germany) and lyophilized.
  • the conjugation reaction with oxidized Glyco-EPO is described in Example 12, 2.2.
  • O-[2-(2-aminooxy-ethoxy)-ethyl]-hydroxylamine was synthesized as described by Boturyn et al in 2 steps from commercially available materials.7 1.44 g (0.12 mmol) of Oxo-HES18KD were dissolved in 3 mL absolute dimethyl sulfoxide (DMSO) and were added dropwise under nitrogen to a mixture of 2.04 g (15 mmol) O-[2-(2-aminooxy-ethoxy)-ethyl]-hydroxylamine in 15 mL DMSO. After stirring for 48 h at 65° C. the reaction mixture was added to 160 mL of a 1:1 mixture of ethanol and acetone.
  • DMSO dimethyl sulfoxide
  • the precipitated product K was collected by centrifugation, redissolved in 40 mL of water and dialyzed for 4 days against water (SnakeSkin dialysis tubing, 3.5 KD cut off, Perbio Science GmbH, Bonn, Germany) and lyophilized.
  • the conjugation reaction with oxidized Glyco-EPO is described in Example 12, 3.1. 7Boturyn, Boudali, Constant, Defrancq, L Subscribe, 1997, Tetrahedron, 53, 5485
  • Oxo-HES18KD 1.44 g (0.12 mmol) of Oxo-HES18KD were dissolved in 3 mL absolute dimethyl sulfoxide (DMSO) and were added to a mixture of 1.16 g (15 mmol) cysteamine in 15 mL DMSO under nitrogen dropwise. After stirring for 24 h at 40° C. the reaction mixture was added to 160 mL of a 1:1 mixture of ethanol and acetone.
  • DMSO dimethyl sulfoxide
  • the precipitated product M was collected by centrifugation, redissolved in 40 mL of water and dialyzed for 2 days against a 0.5% (v/v) triethylamine in water solution and for 2 days against water (SnakeSkin dialysis tubing, 3.5 KD cut off, Perbio Science GmbH, Bonn, Germany) and lyophilized.
  • the conjugation reaction with oxidized Glyco-EPO is described in Example 12, 2.1.
  • A. Borate buffer. Composition is 50 mM sodium borate, pH 8.3, 5 mM EDTA.
  • PBS phosphate buffered saline:10 mM sodium phosphate, 150 mM NaCl, pH 7.4.
  • ThioEPO solution 5 mg/mL of ThioEPO 1 in borate buffer.
  • Microconcentrator Microcon YM-3 (amicon, Milipore GmbH, Eschborn, Germany) 8Cumber, Forrester, Foxwell, Ross, Thorpe, 1985 , Methods Enzymol., 112, 207
  • SIA solution 100 ⁇ L SIA solution is added to 400 ⁇ L of the aminoHES12KD E solution and is allowed to react with agitation for 0.5 hours at room temperature.
  • the excess crosslinker is removed by centrifuging the sample at 14000 ⁇ g for 60 minutes using a microconcentrator. After centrifuging the sample is brought up to its original volume in borate buffer and this process is repeated two more times.
  • the residual solution is added to 1 mL of ThioEPO solution and the reaction mixture is incubated for 16 hour at room temperature. Reactivity of the excess iodoacetamide is quenched at the end of the incubation period by the addition of cysteine to a final concentration of 10 mM.
  • the reaction mixture is applied to a desalting column equilibrated with PBS buffer and the protein content of the fractions are monitored with a Coomassie protein assay reagent. All fractions containing the protein conjugate are pooled and the conjugate was obtained by lyophylization after dialysis against water over night.
  • cross-linkers could be used, which have a succinimide- or a sulfosuccinimide function and a iodoacetamide function separated by a spacer. Further examples are found in table 2. They are marked with a “C” and are available from Perbio Science GmbH, Bonn, Germany.
  • A. Phosphate buffer. Composition is 100 mM sodium phosphate, pH 6.1, 5 mM EDTA.
  • PBS phosphate buffered saline:10 mM sodium phosphate, 150 mM NaCl, pH 7.4.
  • amino HES12KD (E, H, I) could be linked with a cross-linker via a succinimide- and a bromoacetamide function (see 1.1 above).
  • SBAP is a member of this group of cross-linkers and is found in table 2, marked with a “D”.
  • HES12KD 10 mg/mL in 0.1 M sodium acetate buffer, pH 5.5
  • Microconcentrator Microcon YM-3 (amicon, Milipore GmbH, Eschborn, Germany)
  • PBS phosphate buffered saline: 10 mM sodium phosphate, 150 mM NaCl, pH 7.4.
  • M 2 C 2 H solution is added to 400 ⁇ L of the HES12KD solution to a final concentration of 1 mM and is allowed to react with agitation for 2 hours at room temperature.
  • the excess cross-linker is removed by centrifuging the sample at 14000 ⁇ g for 60 minutes using a microconcentrator. After centrifuging the sample is brought up to its original volume in phosphate/NaCl buffer and this process is repeated two more times.
  • To the M 2 C 2 H-modified HES12KD 0.5 mL of ThioEPO solution is added and the reaction mixture is incubated for 2 hours at room temperature. Reactivity of the excess maleimides is quenched at the end of the incubation period by the addition of cysteine to a final concentration of 10 mM.
  • the reaction mixture is applied to Sephadex® G-200 (1.5 ⁇ 45 cm) equilibrated with PBS buffer and 1 mL fractions are collected. The protein content of the fractions is monitored with a Coomassie protein assay reagent. All fractions containing the protein conjugate are pooled and the conjugate was obtained by lyophylization after dialysis against water over night.
  • the hydrazone adduct is slightly less stable at extremes of pH. For applications that may involve treatment at low pH, we reduced the hydrazone by treatment with 30 mM sodium cyanoborohydride in PBS buffer to a hydrazine. For most applications, this extra step is unnecessary.
  • Phosphate/NaCl 0.1 M sodium phosphate, 50 mM NaCl, pH 7.0
  • PBS phosphate buffered saline: 10 mM sodium phosphate, 150 mM NaCl, pH 7.4.
  • Microconcentrator Microcon YM-10 (amicon, Milipore GmbH, Eschborn, Germany).
  • PBS phosphate buffered saline: 10 mM sodium phosphate, 150 mM NaCl, pH 7.4.
  • ThioEPO 1 solution 5 mg/mL of ThioEPO 1 in PBS buffer.
  • Reactivity of the excess maleimide is quenched at the end of the incubation period by the addition of cysteine to a final concentration of 10 mM.
  • the reaction mixture is applied to a desalting column equilibrated with PBS buffer.
  • the protein content of the fractions are monitored with a Coomassie protein assay reagent, all fractions containing the protein conjugate are pooled and the conjugate is obtained by lyophylization after dialysis against water over night.
  • cross-linkers could be used which have a succinimide- or a sulfosuccinimide function and a maleimide-function, separated by a spacer. Further examples for this group of molecules, available from Perbio Science GmbH, Bonn, Germany, are found in table, 2, marked with an “E”. There is a further group of cross-linkers, which have instead of a maleimide function an activated disulfide function. These cross-linkers could also be used for the conjugation. However, the disulfide bond of the conjugate is cleavable under reductive conditions. Members of this group are marked in table 2 with a “F”. A third group of cross-linkers uses instead of a maleimide function a vinylsulfone function as a SH-reactive group. A member of this group “SVSB” is marked in table 2 with a “G”.
  • Glyco-EPO solution 10 mg/mL of Glyco-EPO in acetate buffer
  • Sodium meta-periodate solution 10 mM or 100 mM sodium periodate in acetate buffer, prepared fresh. Keep in dark. Using these solutions, the final concentration of sodium periodate in the oxidation mixture is 1 mM or 10 mM, respectively.
  • Microconcentrator Microcon YM-3 (Amicon, Milipore GmbH, Eschborn, Germany)
  • M 2 C 2 H stock 10 mg/mL M 2 C 2 H in DMSO, prepared fresh
  • Acetate buffer 0.1 M sodium acetate buffer, pH 5.5
  • Microconcentrator Microcon YM-3 (amicon, Milipore GmbH, Eschborn, Germany)
  • M 2 C 2 H stock solution is added to 1 mL of oxidized Glyco-EPO to a final concentration of 1 mM and is allowed to react with agitation for 2 hours at room temperature.
  • the excess crosslinker is removed by centrifuging the sample at 14000 ⁇ g for 60 minutes using a microconcentrator. After centrifuging the sample is brought up to its original volume in phosphate/NaCl buffer and this process was repeated two more times.
  • O or Q solution is added and the reaction mixture is incubated for 16 hours at room temperature.
  • Reactivity of the excess maleimides is quenched at the end of the incubation period by the addition of cysteine.
  • the reaction mixture is applied to Sephadex® G-200 (1.5 ⁇ 45 cm) equilibrated with PBS and 1 mL fractions are collected.
  • the protein content of the fractions is monitored with a Coomassie protein assay reagent, all fractions containing the protein conjugate are pooled and the conjugate is obtained by lyophylization after dialysis against water over night.
  • the hydrazone adduct is slightly less stable at extremes of pH. For applications that may involve treatment at low pH, we reduced the hydrazone by treatment with 30 mM sodium cyanoborohydride in PBS buffer to a hydrazine. For most applications, this extra step was unnecessary.
  • Acetate buffer 0.1 M sodium acetate buffer, pH 5.5
  • PBS phosphate buffered saline: 10 mM sodium phosphate, 150 mM NaCl, pH 7.4
  • the hydrazone adduct is slightly less stable at extremes of pH.
  • the hydrazone may be reduced by treatment with 30 mM sodium cyanoborohydride in PBS buffer to a hydrazine. For most applications, this extra step is unnecessary.
  • Acetate buffer 0.1 M sodium acetate buffer, pH 5.5
  • EPO or partially desialylated EPO forms were incubated with galactose oxidase in the presence of catalase at 37° C. from 30 min-4 hours at 37° C. in 0.05 M Na-phosphate buffer pH 7.0. Progress of the reaction was monitored by removal of 50 ⁇ g aliquots of the EPO and subsequent treatment of the protein with polypeptide N-glycanase.
  • HAS modified EPO forms from nonreacted EPO and HAS-precursor molecules was achieved by gel filtration using e.g. Ultrogel AcA 44/54 or similar gel filtration media.
  • nonreacted HAS was removed by immuno affinity isolation of EPO on a 4 mL column containing a monoclonal antibody coupled to Affigel (BioRad) and subsequent separation of unmodified EPO by gel filtration (e.g. using a matrix enabling the separation of globular proteins of a relative molecular mass between 20 kDa and 200 kDa).
  • HAS modified EPOs were identified by SDS-PAGE analysis (using 12.5 or 10% acrylamide gels) through detection of their higher molecular weight compared to unmodified EPO upon staining of gels with Coomassie Brilliant Blue. The higher molecular weight of HAS modified EPO polypeptides was also identified by Western Blot analysis of samples using a polyclonal antibody raised against recombinant human EPO.
  • N-glycan modification of EPO forms was demonstrated by their successful removal from the EPO protein with polypeptide N-glycanase (recombinant N-glycosidase from Roche, Germany employing 25 units/mg EPO protein at 37° C. for 16 hours); analysis by SDS-PAGE resulted in a typical shift of the EPO protein to a migration position of the N-glycosidase treated unmodified EPO of approximately 20 KDa.
  • Modification of the single desialylated and galactose oxidase treated EPO O-glycan at Ser 126 was demonstrated by SDS-PAGE migration of the de-N-glycosylated product by detection of its migration position compared to nonreacted de-N-glycosylated EPO. If required, modified EPO was fractionated by RP-HULK on a C8-phase before SDS-PAGE analysis. HAS O-glycan modification of EPO was also analyzed by ⁇ -elimination of the O-glycan and detection of the de-O-glycosylated form of EPO in Western blots using a polyclonal antibody raised against recombinant human EPO.
  • EPO forms where quantitated by UV measurements as described in Ph.Eur (2000, Erythropoietini solutio concentrata, 1316, 780-785) and compared to the international BRP reference EPO standard.
  • EPO concentrations were determined by a RP-HPLC assay using a RP-C4-column and absorption at 254 nm employing 20, 40, 80 and 120 ⁇ g of the BRP standard EPO reference preparation for calibration.
  • Purified HES-modified EPO is tested for activity using the erythropoietin bioactivity assay as described by Krystal [Krystal, 1984, Exp. Heamatol., 11, 649-660].
  • Anemia is induced in NMRI mice by treatment with phenylhydrazine hydrochloride and spleen cells are collected and used as described in [Fibi et al., 1991, Blood, 77, 1203 ff.]. Dilutions of EPO are incubated with 3 ⁇ 10 5 cells/well in 96-well microtiter plates. After 24 hours at 37° C. in a humidified atmosphere (5% CO 2 ) cells are labeled for 4 hours with 1 ⁇ Ci of 3 H-thymidine per well. Incorporated radioactivity is determined by liquid scintillation counting. The International reference EPO standard (BRP-standard) is used for comparison.
  • EPO bioactivity may be measured by an in vitro assay using the EPO-sensitive cell line TF-1 (Kitamura et. al., [J. cell Phys., 140. 323-334]. Exponentially growing cells are washed free of growth factors and are incubated in the presence of serial dilutions of the EPO for further 48 hours. Proliferation of the cells is assessed by using the MTT reduction assay as described by Mosmann [Mosmann, 1983, J. Immunol. Methods, 65, 55-63].
  • In vivo activity determinations are performed in normocythemic mice by measuring the increase of reticulocytes after 4 days after animals received the foreseen dose of EPO or modified EPO forms. Assays are performed using the BRP EPO standard which is calibrated against the WHO EPO standard in the polycythemic mouse assay. EPO samples are diluted in phosphate buffered saline containing 1 mg/ml of bovine serum albumin (Sigma).
  • EPO test solution 0.5 ml of the EPO test solution in Dulbecco's buffered saline (corresponding to an EPO protein equivalent of a 100, 80, 40 or 20 IU/ml of the BRP standard EPO) are infected subcutaneously per animal.
  • Blood samples are taken after 4 days after injection and reticulocytes are stained with acridine orange; quantitation of reticulocytes is performed by flow-cytometry by counting a total of 30,000 blood cells within 5 hours after the blood sample was taken (see Ph. Eur, 2000, Erythropoietini solutio concentrata, 1316, pages 780-785) and European Pharmacopoeia (1996/2000, attachment 2002).
  • Rabbits are injected intravenously with specified amounts of unmodified or HAS-modified EPO forms. Blood samples are obtained at specified times, and serum is prepared. Serum erythropoietin levels are determined by in vitro bioassay or by an EPO-specific commercial ELISA.
  • mice Each animal receive 300 IU EPO/kg subcutaneously. Seven days after the post-treatment hematocrit of each animal is determined. A substantial increase in hematocrit is observed in all animals treated with modified EPO.
  • Rabbits are treated with a single dose of unmodified or HAS-modified EPO corresponding to 200 or up to 800 ng/kg body weight. After 2, 6, 16, 24 and 48 hours blood samples are analyzed by using a commercial EPO-specific ELISA for determination of plasma concentrations. Mean plasma EPO concentrations are determined and the average initial half-lives ( ⁇ -phase) and the terminal half-lives ( ⁇ -phase) are calculated from the ELISA values as described: (Zettlmissl et al., 1989, J. Biol. Chem., 264, 21153-21159).
  • Modified IL2 is recovered by gel filtration on Ultrogel AcA 54. Aliquots of corresponding fraction are sterile filtrated and IL2 bioactivity is determined by using the IL2 dependent murine CTLL-2 cell line [Gillis, Ferm, On, and Smith, 1978, J. Immunol., 120, 2027-2032]. Activity is related to the international reference IL2 standard preparation.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Polymers & Plastics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Materials Engineering (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Toxicology (AREA)
  • Zoology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Dermatology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Polysaccharides And Polysaccharide Derivatives (AREA)
  • Peptides Or Proteins (AREA)
  • Medicinal Preparation (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
US11/078,098 2002-09-11 2005-03-11 Method of producing hydroxyalkyl starch derivatives Abandoned US20050238723A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US11/078,098 US20050238723A1 (en) 2002-09-11 2005-03-11 Method of producing hydroxyalkyl starch derivatives
US12/824,618 US20120046240A9 (en) 2002-09-11 2010-06-28 Method of Producing Hydroxyalkyl Starch Derivatives

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US40978102P 2002-09-11 2002-09-11
EP02020425A EP1400533A1 (fr) 2002-09-11 2002-09-11 Polypeptides hasylés, érythropoiétine hasylée en particulier
EP02020425.1 2002-09-11
PCT/EP2003/008829 WO2004024776A1 (fr) 2002-09-11 2003-08-08 Procede de production de derives d'amidon hydroxyalkyle
US11/078,098 US20050238723A1 (en) 2002-09-11 2005-03-11 Method of producing hydroxyalkyl starch derivatives

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2003/008829 Continuation-In-Part WO2004024776A1 (fr) 2002-09-11 2003-08-08 Procede de production de derives d'amidon hydroxyalkyle

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/824,618 Continuation US20120046240A9 (en) 2002-09-11 2010-06-28 Method of Producing Hydroxyalkyl Starch Derivatives

Publications (1)

Publication Number Publication Date
US20050238723A1 true US20050238723A1 (en) 2005-10-27

Family

ID=31995520

Family Applications (5)

Application Number Title Priority Date Filing Date
US11/077,906 Expired - Fee Related US7815893B2 (en) 2002-09-11 2005-03-11 Hydroxyalkyl starch derivatives
US11/078,582 Expired - Fee Related US8618266B2 (en) 2002-09-11 2005-03-11 Hasylated polypeptides
US11/078,098 Abandoned US20050238723A1 (en) 2002-09-11 2005-03-11 Method of producing hydroxyalkyl starch derivatives
US12/824,618 Abandoned US20120046240A9 (en) 2002-09-11 2010-06-28 Method of Producing Hydroxyalkyl Starch Derivatives
US12/855,381 Expired - Fee Related US8475765B2 (en) 2002-09-11 2010-08-12 Hydroxyalkyl starch derivatives

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US11/077,906 Expired - Fee Related US7815893B2 (en) 2002-09-11 2005-03-11 Hydroxyalkyl starch derivatives
US11/078,582 Expired - Fee Related US8618266B2 (en) 2002-09-11 2005-03-11 Hasylated polypeptides

Family Applications After (2)

Application Number Title Priority Date Filing Date
US12/824,618 Abandoned US20120046240A9 (en) 2002-09-11 2010-06-28 Method of Producing Hydroxyalkyl Starch Derivatives
US12/855,381 Expired - Fee Related US8475765B2 (en) 2002-09-11 2010-08-12 Hydroxyalkyl starch derivatives

Country Status (25)

Country Link
US (5) US7815893B2 (fr)
EP (8) EP2272865A3 (fr)
JP (4) JP2006516534A (fr)
KR (3) KR101174510B1 (fr)
CN (2) CN100348618C (fr)
AR (3) AR041097A1 (fr)
AT (3) ATE406387T1 (fr)
AU (6) AU2003260393B2 (fr)
BR (3) BR0314107A (fr)
CA (2) CA2496317C (fr)
CY (1) CY1109813T1 (fr)
DE (7) DE60323192D1 (fr)
DK (3) DK1398322T3 (fr)
ES (4) ES2213506T3 (fr)
HK (4) HK1063477A1 (fr)
IL (5) IL166506A0 (fr)
MX (4) MXPA05002593A (fr)
NO (2) NO20051427L (fr)
PL (3) PL375037A1 (fr)
PT (3) PT1398322E (fr)
RU (3) RU2328505C2 (fr)
SI (2) SI1398327T1 (fr)
TW (1) TW200418875A (fr)
WO (3) WO2004024761A1 (fr)
ZA (1) ZA200600651B (fr)

Cited By (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20050063943A1 (en) * 2001-03-16 2005-03-24 Klaus Sommermeyer Conjugated of hydroxyalkyl starch and an active agent
US20050181985A1 (en) * 2002-03-06 2005-08-18 Jurgen Hemberger Coupling proteins to a modified polysaccharide
US20050234230A1 (en) * 2002-09-11 2005-10-20 Norbert Zander Hydroxyalkyl starch derivatives
US20060052342A1 (en) * 2002-12-04 2006-03-09 Klaus Sommermeyer Aldonic acid esters, methods for producing the same, and methods for producing pharmaceutical active ingredients coupled to polysaccharides or polysaccharide derivatives on free amino groups
US20060100163A1 (en) * 2002-10-08 2006-05-11 Michele Orlando Pharmaceutically active oligosaccharide conjugates
US20060121062A1 (en) * 2002-09-11 2006-06-08 Wolfram Eichner Conjugated hydroxyalkyl starch allergen compounds
US20060217293A1 (en) * 2002-03-06 2006-09-28 Michele Orlando Coupling low-molecular substances to a modified polysaccharide
US20070087961A1 (en) * 2004-03-11 2007-04-19 Wolfram Eichner Conjugates of hydroxyalkyl starch and erythropoietin
US20080206182A1 (en) * 2003-08-08 2008-08-28 Fresenius Kabi Deutschland Gmbh Conjugates of a Polymer and a Protein Linked by an Oxime Group
US20080207562A1 (en) * 2005-09-12 2008-08-28 Fresenius Kabi Deutschland Gmbh Conjugates of Hydroxyalkyl Starch and Active Substance, Prepared by Chemical Ligation Via Thiazolidine
US20080274948A1 (en) * 2003-08-08 2008-11-06 Fresenius Kabi Deutschland Gmbh Conjugates of Hydroxyalkyl Starch and G-Csf
US20090047251A1 (en) * 2004-03-11 2009-02-19 Wolfram Eichner Conjugates of hydroxyalkyl starch and a protein
EP2070950A1 (fr) * 2007-12-14 2009-06-17 Fresenius Kabi Deutschland GmbH Dérivés hydroxyalkylés de l'amidon et leur procédé de préparation
EP2070951A1 (fr) * 2007-12-14 2009-06-17 Fresenius Kabi Deutschland GmbH Procédé de production d'un dérivé hydroxyalkyle de l'amidon avec deux liens
WO2010011735A2 (fr) 2008-07-23 2010-01-28 Ambrx, Inc. Polypeptides g-csf bovins modifiés et leurs utilisations
US20100062973A1 (en) * 2005-03-11 2010-03-11 Fresenius Kabi Deutschland Gmbh Production of bioactive glycoproteins from inactive starting material
US20100311670A1 (en) * 2004-03-11 2010-12-09 Nobert Zander Conjugates of hydroxyalkyl starch and a protein, prepared by native chemical ligation
WO2012004007A1 (fr) * 2010-07-09 2012-01-12 Fresenius Kabi Deutschland Gmbh Conjugués comprenant un amidon hydroxyalkylé et un agent cytotoxique et leur procédé de préparation
WO2012004005A1 (fr) * 2010-07-09 2012-01-12 Fresenius Kabi Deutschland Gmbh Conjugués comprenant de l'hydroxyalkylamidon et un agent cytotoxique et leur procédé de synthèse
WO2012004009A1 (fr) * 2010-07-09 2012-01-12 Fresenius Kabi Deutschland Gmbh Conjugués comprenant de l'hydroxyalkylamidon et un agent cytotoxique et leur procédé de synthèse
US8287850B2 (en) 2004-03-11 2012-10-16 Fresenius Kabi Deutschland Gmbh Conjugates of hydroxyalkyl starch and a protein, prepared by reductive amination
EP2805964A1 (fr) 2009-12-21 2014-11-26 Ambrx, Inc. Polypeptides modifiés de somatotrophine bovine et leurs utilisations
EP2805965A1 (fr) 2009-12-21 2014-11-26 Ambrx, Inc. Polypeptides modifiés de somatotrophine bovine et leurs utilisations
US10787476B2 (en) 2013-09-24 2020-09-29 Ajinomoto Co., Inc. Glycoamino acid and use thereof
US11273202B2 (en) 2010-09-23 2022-03-15 Elanco Us Inc. Formulations for bovine granulocyte colony stimulating factor and variants thereof

Families Citing this family (132)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1400533A1 (fr) * 2002-09-11 2004-03-24 Fresenius Kabi Deutschland GmbH Polypeptides hasylés, érythropoiétine hasylée en particulier
CA2534418A1 (fr) * 2003-08-08 2005-02-17 Fresenius Kabi Deutschland Gmbh Conjugues d'amidon hydroxyalkyle et de g-csf
US8754031B2 (en) 2004-03-08 2014-06-17 Oncolix, Inc. Use of prolactin receptor antagonists in combination with an agent that inactivates the HER2/neu signaling pathway
US7589063B2 (en) 2004-12-14 2009-09-15 Aplagen Gmbh Molecules which promote hematopoiesis
WO2006094826A2 (fr) * 2005-03-11 2006-09-14 Fresenius Kabi Deutschland Gmbh Procede permettant de coupler des glycoconjugues actives sur le plan enzymatique a un compose de modification
CN101528263B (zh) 2006-06-30 2013-01-23 Sku资产管理有限公司 用于癌症的先天性免疫方法的缀合物
JP2009544681A (ja) 2006-07-25 2009-12-17 リポクセン テクノロジーズ リミテッド エリスロポエチンの多糖誘導体
EP2120998B1 (fr) 2006-11-28 2013-08-07 HanAll Biopharma Co., Ltd. Polypeptides d'érythropoïétine modifiés et utilisation de ceux-ci dans des traitements
DK2101821T3 (da) * 2006-12-15 2014-10-06 Baxter Healthcare Sa Faktor VIIA-(poly)sialinsyre-konjugat med forlænget halveringstid in vivo
CN102978185B (zh) 2007-01-18 2015-02-11 建新公司 包含氨基氧基基团的寡糖及其轭合物
CN101485897B (zh) 2008-01-14 2015-05-20 纪欣 生物相容性止血、防粘连、促愈合、外科封闭的变性淀粉材料
ES2750254T3 (es) 2007-09-27 2020-03-25 Amgen Inc Formulaciones farmacéuticas
EP2219602A1 (fr) 2007-11-15 2010-08-25 Amgen, Inc Préparation aqueuse de protéine de stimulation de l'érythropoïèse stabilisée par des antioxydants pour une administration par voie parentérale
EP2803675A3 (fr) 2008-01-25 2014-12-24 Amgen, Inc Anticorps anti-ferroportine et procédés d'utilisation
EP2310423A1 (fr) * 2008-04-28 2011-04-20 SurModics, Inc. Matrices à base de poly- (1 4)glucopyranose réticulées avec un hydrazide
AU2009246946B2 (en) 2008-05-01 2013-09-26 Amgen Inc. Anti-hepcidin antibodies and methods of use
EP2166085A1 (fr) 2008-07-16 2010-03-24 Suomen Punainen Risti Veripalvelu Cellules modifiées bivalentes
WO2010033207A1 (fr) 2008-09-19 2010-03-25 Nektar Therapeutics Conjugués polymères de peptides thérapeutiques
US8318167B2 (en) 2008-11-13 2012-11-27 The General Hospital Corporation Methods and compositions for regulating iron homeostasis by modulation of BMP-6
EP2370105B1 (fr) 2008-12-06 2016-03-16 B. Braun Melsungen AG Composés consistant en un glycosaminoglycane et un amidon pour le transport d'un médicament ou d'un marqueur fluorescent lié de manière covalente
US8648046B2 (en) 2009-02-26 2014-02-11 Oncolix, Inc. Compositions and methods for visualizing and eliminating cancer stem cells
JP5963443B2 (ja) 2009-02-26 2016-08-03 オンコリックス, インコーポレイテッド がん幹細胞を可視化・排除するための組成物および方法
ES2859625T3 (es) * 2009-03-31 2021-10-04 Braun Melsungen Ag Productos de enlace de polisacáridos aminados
US8809501B2 (en) 2009-07-27 2014-08-19 Baxter International Inc. Nucleophilic catalysts for oxime linkage
US8642737B2 (en) 2010-07-26 2014-02-04 Baxter International Inc. Nucleophilic catalysts for oxime linkage
HUE028056T2 (en) 2009-07-27 2016-11-28 Baxalta GmbH Blood coagulation protein conjugates
JP5908401B2 (ja) 2009-07-27 2016-04-26 バクスター・インターナショナル・インコーポレイテッドBaxter International Incorp0Rated 血液凝固タンパク質複合体
WO2011012850A2 (fr) * 2009-07-27 2011-02-03 Lipoxen Technologies Limited Glyco-polysialylation de protéines de coagulation non sanguine
WO2011050333A1 (fr) 2009-10-23 2011-04-28 Amgen Inc. Adaptateur de fiole et système
WO2011098095A1 (fr) 2010-02-09 2011-08-18 Aplagen Gmbh Peptides se liant au récepteur de tpo
FI2575935T4 (fi) 2010-06-07 2023-11-23 Amgen Inc Lääkeaineen vapautuslaite
US20140073602A9 (en) 2010-07-09 2014-03-13 Fresenius Kabi Deutschland Gmbh Nitric oxide delivering hydroxyalkyl starch derivatives
BR112013001611B1 (pt) 2010-07-30 2022-05-10 Takeda Pharmaceutical Company Limited Método para conjugar um polímero solúvel em água a uma fração de carboidrato oxidado de uma proteína terapêutica, proteína terapêutica modificada, método para formar uma ligação oxima entre uma fração de carboidrato oxidado em uma proteína terapêutica e um polímero solúvel em água ativado contendo um grupo amino-óxi ativo emétodo para formar uma ligação hidrazona entre uma fração de carboidrato oxidado em uma proteína terapêutica e um polímero solúvel em água ativado contendo um grupo hidrazida ativo
CA2822591C (fr) 2010-12-22 2020-12-29 Baxter International Inc. Materiels et methodes pour la conjugaison d'un derive d'acide gras soluble dans l'eau a une proteine
EP2691065B1 (fr) 2011-03-31 2017-03-01 Amgen Inc. Adaptateur de flacon et système
CA2833748C (fr) 2011-04-20 2019-07-16 Amgen Inc. Appareil auto-injecteur
AU2012262428C1 (en) 2011-05-27 2018-10-11 Takeda Pharmaceutical Company Limited Therapeutic proteins with increased half-life and methods of preparing same
US20150044714A1 (en) * 2011-06-07 2015-02-12 Philadelphia Health & Education Corporation D/B/A Drexel University College Of Medicine sPLA2 MONITORING STRIP
WO2013021056A1 (fr) 2011-08-10 2013-02-14 Ludwig-Maximilians-Universität München Procédé d'administration intracellulaire contrôlée d'acides nucléiques
BR112014008993B1 (pt) 2011-10-14 2021-06-08 Amgen Inc injetor e método de montagem
WO2013113503A1 (fr) 2012-01-31 2013-08-08 Fresenius Kabi Deutschland Gmbh Conjugués d'amidon hydroxyalkylé et d'un oligonucléotide
AU2013204754C1 (en) 2012-05-16 2018-10-11 Takeda Pharmaceutical Company Limited Nucleophilic Catalysts for Oxime Linkage
CA2884887C (fr) 2012-11-21 2023-09-12 Amgen Inc. Dispositif de livraison de medicament comportant un element d'insertion et un reservoir
WO2014084110A1 (fr) * 2012-11-30 2014-06-05 株式会社糖鎖工学研究所 Lieur à chaîne glucidique liée, composé contenant un lieur à chaîne glucidique liée et substance physiologiquement active ou son sel, et son procédé de production
CN105263514B (zh) 2013-03-15 2019-04-26 本质生命科学有限公司 抗铁调素抗体及其用途
TWI580452B (zh) 2013-03-15 2017-05-01 安美基公司 用於注射器之匣盒、注射器及使用包括自動注射器及匣盒之設備之方法
JP6336564B2 (ja) 2013-03-15 2018-06-06 アムゲン・インコーポレーテッド 薬物カセット、自動注入器、および自動注入器システム
AU2014234270A1 (en) * 2013-03-20 2015-10-08 Fresenius Kabi Deutschland Gmbh Hydroxyalkyl starch derivatives as reactants for coupling to thiol groups
AU2014234272A1 (en) * 2013-03-20 2015-10-08 Fresenius Kabi Deutschland Gmbh Hydroxyalkyl starch derivatives as reactants for coupling to thiol groups
EP2976365A1 (fr) * 2013-03-20 2016-01-27 Fresenius Kabi Deutschland GmbH Procédé pour la préparation de dérivés d'hydroxyalkylamidon fonctionnalisés par thiol
SG11201507878SA (en) 2013-03-22 2015-10-29 Amgen Inc Injector and method of assembly
EP3060281B1 (fr) 2013-10-24 2019-01-30 Amgen Inc. Système de distribution de médicaments équipé d'un dispositif de commande sensible à la température
WO2015061386A1 (fr) 2013-10-24 2015-04-30 Amgen Inc. Injecteur et procédé d'assemblage
US10994112B2 (en) 2014-02-05 2021-05-04 Amgen Inc. Drug delivery system with electromagnetic field generator
EP3104964A4 (fr) * 2014-02-10 2018-02-14 Honeywell International Inc. Conception de réacteur de fluoration en phase liquide
EP3114138B1 (fr) 2014-03-05 2021-11-17 Pfizer Inc. Mutéines améliorées du facteur viii de coagulation
EP3785749A1 (fr) 2014-05-07 2021-03-03 Amgen Inc. Auto-injecteur doté d'éléments de réduction de choc
WO2015187797A1 (fr) 2014-06-03 2015-12-10 Amgen Inc. Système d'administration de médicament pouvant être commandé et son procédé d'utilisation
CN104072608A (zh) * 2014-06-23 2014-10-01 山东齐都药业有限公司 含羧基羟乙基淀粉衍生物修饰的牛血清白蛋白偶联物及其制备方法
CA2961917A1 (fr) 2014-09-22 2016-03-31 Intrinsic Lifesciences Llc Anticorps anti-hepcidine humanises et utilisations de ceux-ci
US10695506B2 (en) 2014-10-14 2020-06-30 Amgen Inc. Drug injection device with visual and audio indicators
ES2898469T3 (es) 2014-12-19 2022-03-07 Amgen Inc Dispositivo de administración de medicamentos con sensor de proximidad
EP3689394A1 (fr) 2014-12-19 2020-08-05 Amgen Inc. Dispositif d'administration de médicaments avec bouton mobile ou panneau d'interface utilisateur
WO2016133947A1 (fr) 2015-02-17 2016-08-25 Amgen Inc. Dispositif d'administration de médicament à sécurisation assistée par dépression et/ou retour d'informations
WO2016138434A1 (fr) 2015-02-27 2016-09-01 Amgen Inc. Dispositif d'administration de médicament ayant un mécanisme de protection d'aiguille présentant un seuil réglable de résistance au mouvement de l'élément de protection d'aiguille
WO2017039786A1 (fr) 2015-09-02 2017-03-09 Amgen Inc. Adaptateur d'ensemble de seringue pour une seringue
JP7082568B2 (ja) 2015-12-09 2022-06-08 アムジエン・インコーポレーテツド 信号伝達キャップ付き自動注射器
US11154661B2 (en) 2016-01-06 2021-10-26 Amgen Inc. Auto-injector with signaling electronics
ES2814287T3 (es) 2016-03-15 2021-03-26 Amgen Inc Reducir la probabilidad de rotura de cristal en dispositivos de administración de fármaco
US11541168B2 (en) 2016-04-29 2023-01-03 Amgen Inc. Drug delivery device with messaging label
US11389588B2 (en) 2016-05-02 2022-07-19 Amgen Inc. Syringe adapter and guide for filling an on-body injector
EP3455142B1 (fr) 2016-05-13 2023-08-09 Amgen Inc. Ensemble manchon de flacon
EP3458988B1 (fr) 2016-05-16 2023-10-18 Amgen Inc. Chiffrement de données dans des dispositifs médicaux à capacité de calcul limitée
CA3024349A1 (fr) 2016-05-20 2017-11-23 Octapharma Ag Proteines de fusion vwf glycosylees a pharmacocinetique amelioree
GB201609083D0 (en) 2016-05-24 2016-07-06 Syntab Therapeutics Gmbh Synthetic compound
US11541176B2 (en) 2016-06-03 2023-01-03 Amgen Inc. Impact testing apparatuses and methods for drug delivery devices
EP3478342A1 (fr) 2016-07-01 2019-05-08 Amgen Inc. Dispositif d'administration de médicament présentant un risque réduit au minimum de fracture de composant lors d'événements d'impact
US20190328965A1 (en) 2016-08-17 2019-10-31 Amgen Inc. Drug delivery device with placement detection
WO2018081234A1 (fr) 2016-10-25 2018-05-03 Amgen Inc. Injecteur porté sur le corps
AU2018210301A1 (en) 2017-01-17 2019-08-01 Amgen Inc. Injection devices and related methods of use and assembly
MX2019009625A (es) 2017-02-17 2019-10-09 Amgen Inc Dispositivo de administracion de farmacos con trayectoria de flujo de fluido esteril y metodo relacionado de ensamblaje.
JP7280189B2 (ja) 2017-02-17 2023-05-23 アムジエン・インコーポレーテツド 薬物送達装置用の挿入機構
MX2019010544A (es) 2017-03-06 2019-10-21 Amgen Inc Dispositivo de administracion de farmacos con caracteristica de prevencion de la activacion.
SG11201908058UA (en) 2017-03-07 2019-09-27 Amgen Inc Needle insertion by overpressure
MX2019010671A (es) 2017-03-09 2019-10-21 Amgen Inc Mecanismo de insercion para dispositivo de administracion de farmacos.
WO2018172219A1 (fr) 2017-03-20 2018-09-27 F. Hoffmann-La Roche Ag Procédé de glyco-ingénierie in vitro d'une protéine stimulant l'érythropoïèse
EP3600491B1 (fr) 2017-03-28 2023-08-09 Amgen Inc. Tige de piston ainsi que système et procédé d'assemblage de seringue
AU2018280054B2 (en) 2017-06-08 2023-07-13 Amgen Inc. Syringe assembly for a drug delivery device and method of assembly
MX2019014615A (es) 2017-06-08 2020-02-07 Amgen Inc Dispositivo de administracion de farmacos accionado por par de torsion.
JP7195276B2 (ja) 2017-06-22 2022-12-23 アムジエン・インコーポレーテツド デバイス起動による衝突/衝撃の低減
WO2018237225A1 (fr) 2017-06-23 2018-12-27 Amgen Inc. Dispositif électronique d'administration de médicament comprenant un bouchon activé par un ensemble commutateur
ES2972207T3 (es) 2017-07-14 2024-06-11 Amgen Inc Sistema de inserción-retracción de agujas con sistema de resorte de torsión doble
IL271173B2 (en) 2017-07-21 2024-04-01 Amgen Inc Gas permeable sealing element for drug container and methods of assembly
JP7242562B2 (ja) 2017-07-25 2023-03-20 アムジエン・インコーポレーテツド 容器アクセスシステムを有する薬物送達デバイス及び関連する組立方法
MA49677A (fr) 2017-07-25 2021-04-21 Amgen Inc Dispositif d'administration de médicament avec module d'engrenage et procédé d'assemblage associé
EP3664863A2 (fr) 2017-08-09 2020-06-17 Amgen Inc. Systèm de administration de médicaments avec pression hydraulique-pneumatique de chambre
EP3668567A1 (fr) 2017-08-18 2020-06-24 Amgen Inc. Injecteur sur-corps avec patch adhésif stérile
US11103636B2 (en) 2017-08-22 2021-08-31 Amgen Inc. Needle insertion mechanism for drug delivery device
US11759565B2 (en) 2017-10-04 2023-09-19 Amgen Inc. Flow adapter for drug delivery device
EP3691716B1 (fr) 2017-10-06 2023-11-29 Amgen Inc. Dispositif d'administration de médicament comprenant un ensemble de verrouillage et procédé d'assemblage associé
WO2019074579A1 (fr) 2017-10-09 2019-04-18 Amgen Inc. Dispositif d'administration de médicament comprenant un ensemble d'entraînement et procédé d'assemblage associé
EP3703779A1 (fr) 2017-11-03 2020-09-09 Amgen Inc. Systèmes et approches pour stériliser un dispositif d'administration de médicament
WO2019090303A1 (fr) 2017-11-06 2019-05-09 Amgen Inc. Ensembles de remplissage-finition et procédés associés
JP2021501616A (ja) 2017-11-06 2021-01-21 アムジエン・インコーポレーテツド 配置及び流量検出を備える薬物送達デバイス
CN116832271A (zh) 2017-11-10 2023-10-03 安进公司 用于药物递送装置的柱塞
AU2018368340B2 (en) 2017-11-16 2024-03-07 Amgen Inc. Door latch mechanism for drug delivery device
EP3710089A1 (fr) 2017-11-16 2020-09-23 Amgen Inc. Auto-injecteur avec détection de décrochage et de point d'extrémité
CN108219019B (zh) * 2018-02-08 2020-03-20 华中科技大学 一种巯基化羟乙基淀粉及其修饰的纳米材料和制备方法
CN108503718B (zh) * 2018-02-08 2020-03-20 华中科技大学 一种羟烷基淀粉偶联物及其制备方法和应用
CN108403641B (zh) * 2018-02-08 2020-03-20 华中科技大学 一种载药纳米材料及其制备方法
EA202090767A1 (ru) * 2018-05-04 2020-08-03 Борис Славинович ФАРБЕР Фармацевтическая композиция на основе полимиксина для лечения инфекционных заболеваний
US10835685B2 (en) 2018-05-30 2020-11-17 Amgen Inc. Thermal spring release mechanism for a drug delivery device
US11083840B2 (en) 2018-06-01 2021-08-10 Amgen Inc. Modular fluid path assemblies for drug delivery devices
EP3826699A1 (fr) 2018-07-24 2021-06-02 Amgen Inc. Dispositifs d'administration pour l'administration de médicaments
MX2021000748A (es) 2018-07-24 2021-03-26 Amgen Inc Dispositivos de suministro para administrar farmacos.
WO2020023336A1 (fr) 2018-07-24 2020-01-30 Amgen Inc. Dispositifs hybrides d'administration de médicament dotés d'une partie de préhension
US20210260279A1 (en) 2018-07-24 2021-08-26 Amgen Inc. Hybrid drug delivery devices with optional grip portion and related method of preparation
CA3103105A1 (fr) 2018-07-31 2020-02-06 Amgen Inc. Ensemble de trajet de fluide pour dispositif d'administration de medicament
CA3106452A1 (fr) 2018-09-24 2020-04-02 Amgen Inc. Systemes et procedes de dosage interventionnel
IL281469B1 (en) 2018-09-28 2024-04-01 Amgen Inc Assembling a memory alloy ejector activation assembly for a drug delivery device
TW202027806A (zh) 2018-10-02 2020-08-01 美商安進公司 具有內部力傳遞的用於藥物遞送之注射系統
AR116607A1 (es) 2018-10-05 2021-05-26 Amgen Inc Dispositivo de administración de fármacos con indicador de dosis
MA53913A (fr) 2018-10-15 2022-01-19 Amgen Inc Procédé d'assemblage de plate-forme pour dispositif d'administration de médicament
CN112789073B (zh) 2018-10-15 2023-09-15 安进公司 具有阻尼机构的药物递送装置
WO2020091956A1 (fr) 2018-11-01 2020-05-07 Amgen Inc. Dispositifs d'administration de médicament avec rétraction partielle de l'organe d'administration de médicament
TWI831847B (zh) 2018-11-01 2024-02-11 美商安進公司 部分針頭縮回之藥物遞送裝置及其操作方法
US20220031939A1 (en) 2018-11-01 2022-02-03 Amgen Inc. Drug delivery devices with partial drug delivery member retraction
CN111157736A (zh) * 2018-11-08 2020-05-15 中国科学院大连化学物理研究所 一种基于化学酶促的人血清o糖基化鉴定方法
JP7510952B2 (ja) 2019-04-24 2024-07-04 アムジエン・インコーポレーテツド シリンジ滅菌確認アセンブリ及び方法
JP2022545227A (ja) 2019-08-23 2022-10-26 アムジエン・インコーポレーテツド 構成可能な針シールド係合構成要素を備えた薬物送達デバイス及び関連方法
KR20240011135A (ko) 2021-05-21 2024-01-25 암젠 인크 약물 용기를 위한 충전 레시피를 최적화하는 방법
WO2024094457A1 (fr) 2022-11-02 2024-05-10 F. Hoffmann-La Roche Ag Procédé de production de compositions de glycoprotéines

Citations (52)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3191291A (en) * 1959-01-21 1965-06-29 Continental Can Co Art of producing very thin steel and like sheets in wide strips
US4001401A (en) * 1975-02-02 1977-01-04 Alza Corporation Blood substitute and blood plasma expander comprising polyhemoglobin
US4001200A (en) * 1975-02-27 1977-01-04 Alza Corporation Novel polymerized, cross-linked, stromal-free hemoglobin
US4053590A (en) * 1975-02-27 1977-10-11 Alza Corporation Compositions of matter comprising macromolecular hemoglobin
US4061736A (en) * 1975-02-02 1977-12-06 Alza Corporation Pharmaceutically acceptable intramolecularly cross-linked, stromal-free hemoglobin
US4064118A (en) * 1975-10-22 1977-12-20 Hematech Inc. Blood substitute based on hemoglobin
US4125492A (en) * 1974-05-31 1978-11-14 Pedro Cuatrecasas Affinity chromatography of vibrio cholerae enterotoxin-ganglioside polysaccharide and the biological effects of ganglioside-containing soluble polymers
US4179337A (en) * 1973-07-20 1979-12-18 Davis Frank F Non-immunogenic polypeptides
US4261973A (en) * 1976-08-17 1981-04-14 Pharmacia Ab Allergen-containing substances
US4412989A (en) * 1981-06-10 1983-11-01 Ajinomoto Company Incorporated Oxygen carrier
US4667016A (en) * 1985-06-20 1987-05-19 Kirin-Amgen, Inc. Erythropoietin purification
US4703008A (en) * 1983-12-13 1987-10-27 Kiren-Amgen, Inc. DNA sequences encoding erythropoietin
US4766106A (en) * 1985-06-26 1988-08-23 Cetus Corporation Solubilization of proteins for pharmaceutical compositions using polymer conjugation
US4847325A (en) * 1988-01-20 1989-07-11 Cetus Corporation Conjugation of polymer to colony stimulating factor-1
US4863964A (en) * 1985-07-02 1989-09-05 Biomedical Frontiers, Inc. Method for the stabilization of deferoxamine to chelate free ions in physiological fluid
US4900780A (en) * 1988-05-25 1990-02-13 Masonic Medical Research Laboratory Acellular resuscitative fluid
US4904584A (en) * 1987-12-23 1990-02-27 Genetics Institute, Inc. Site-specific homogeneous modification of polypeptides
US4925677A (en) * 1988-08-31 1990-05-15 Theratech, Inc. Biodegradable hydrogel matrices for the controlled release of pharmacologically active agents
US4952496A (en) * 1984-03-30 1990-08-28 Associated Universities, Inc. Cloning and expression of the gene for bacteriophage T7 RNA polymerase
US5068321A (en) * 1988-10-27 1991-11-26 Wolff Walsrode Aktiengesellschaft Carbonic acid esters of polysaccharides and a process for their production
US5079337A (en) * 1986-07-02 1992-01-07 Pasteur Merieux Serums Et Vaccins S.A. Macromolecular conjugates of hemoglobin, a procedure for their preparation and their uses
US5214132A (en) * 1986-12-23 1993-05-25 Kyowa Hakko Kogyo Co., Ltd. Polypeptide derivatives of human granulocyte colony stimulating factor
US5217998A (en) * 1985-07-02 1993-06-08 Biomedical Frontiers, Inc. Composition for the stabilization of deferoxamine to chelate free ions in physiological fluid
US5218108A (en) * 1989-06-16 1993-06-08 Fresenius Ag Hydroxylethylstarch (hes) as plasma expander and process for preparing hes
US5218092A (en) * 1988-09-29 1993-06-08 Kyowa Hakko Kogyo Co., Ltd. Modified granulocyte-colony stimulating factor polypeptide with added carbohydrate chains
US5281698A (en) * 1991-07-23 1994-01-25 Cetus Oncology Corporation Preparation of an activated polymer ester for protein conjugation
US5362853A (en) * 1986-12-23 1994-11-08 Kyowa Hakko Kogyo Co., Ltd. Polypeptide derivatives of human granulocyte colony stimulating factor
US5470843A (en) * 1992-12-11 1995-11-28 Hoechst Aktiengesellschaft Carbohydrate-containing polymers, their preparation and use
US5484903A (en) * 1991-09-17 1996-01-16 Wolff Walsrode Aktiengesellschaft Process for the production of polysaccharide carbonates
US5543332A (en) * 1991-07-04 1996-08-06 Immunodex K/S Water-soluble, polymer-based reagents and conjugates comprising moieties derived from divinyl sulfone
US5581476A (en) * 1993-01-28 1996-12-03 Amgen Inc. Computer-based methods and articles of manufacture for preparing G-CSF analogs
US5622718A (en) * 1992-09-25 1997-04-22 Keele University Alginate-bioactive agent conjugates
US5876980A (en) * 1995-04-11 1999-03-02 Cytel Corporation Enzymatic synthesis of oligosaccharides
US5880270A (en) * 1995-06-07 1999-03-09 Cellpro, Incorporated Aminooxy-containing linker compounds for formation of stably-linked conjugates and methods related thereto
US5952347A (en) * 1997-03-13 1999-09-14 Merck & Co., Inc. Quinoline leukotriene antagonists
US6011008A (en) * 1997-01-08 2000-01-04 Yissum Research Developement Company Of The Hebrew University Of Jerusalem Conjugates of biologically active substances
US6083909A (en) * 1996-07-08 2000-07-04 Fresenius Ag Haemoglobin-hydroxyethyl starch conjugates as oxygen carriers
US6261800B1 (en) * 1989-05-05 2001-07-17 Genentech, Inc. Luteinizing hormone/choriogonadotropin (LH/CG) receptor
US6299881B1 (en) * 1997-03-24 2001-10-09 Henry M. Jackson Foundation For The Advancement Of Military Medicine Uronium salts for activating hydroxyls, carboxyls, and polysaccharides, and conjugate vaccines, immunogens, and other useful immunological reagents produced using uronium salts
US6340746B1 (en) * 1997-08-07 2002-01-22 University Of Utah Prodrugs and conjugates of thiol- and selenol- containing compounds and methods of use thereof
US6500930B2 (en) * 1998-03-31 2002-12-31 Hemosol Inc. Hemoglobin-polysaccharide conjugates
US6555660B2 (en) * 2000-01-10 2003-04-29 Maxygen Holdings Ltd. G-CSF conjugates
US6586398B1 (en) * 2000-04-07 2003-07-01 Amgen, Inc. Chemically modified novel erythropoietin stimulating protein compositions and methods
US6596861B1 (en) * 1999-03-12 2003-07-22 Aventis Pasteur S.A. Method for the reductive amination of polysaccharides
US20040043446A1 (en) * 2001-10-19 2004-03-04 Neose Technologies, Inc. Alpha galalctosidase a: remodeling and glycoconjugation of alpha galactosidase A
US20040180858A1 (en) * 2001-06-21 2004-09-16 Klaus Sommermeyer Water-soluble antibiotic comprising an amino sugar, in the form of a polysaccharide conjugate
US20050063943A1 (en) * 2001-03-16 2005-03-24 Klaus Sommermeyer Conjugated of hydroxyalkyl starch and an active agent
US6875594B2 (en) * 1997-11-13 2005-04-05 The Rockefeller University Methods of ligating expressed proteins
US20050181985A1 (en) * 2002-03-06 2005-08-18 Jurgen Hemberger Coupling proteins to a modified polysaccharide
US20060019877A1 (en) * 2002-09-11 2006-01-26 Conradt Harald S Hasylated polypeptides
US20060217293A1 (en) * 2002-03-06 2006-09-28 Michele Orlando Coupling low-molecular substances to a modified polysaccharide
US7285661B2 (en) * 2002-02-20 2007-10-23 Fresenius Kabi Deutschland Gmbh Starch derivatives, starch active substance conjugates, method for the production thereof and their use as medicaments

Family Cites Families (105)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE279486C (fr)
CH385787A (de) 1960-10-04 1964-09-15 Hoechst Ag Verfahren zur Herstellung von nassechten Färbungen auf Cellulosetextilmaterialien
DE2233977A1 (de) 1971-07-14 1973-02-01 Unilever Nv Verfahren zur herstellung oxydierter kohlenhydrate
GB1419080A (en) 1972-12-29 1975-12-24 Cheminova As Chemical compounds having juvenile hormone activity
DE2616086C2 (de) * 1976-04-13 1986-04-03 Dr. Eduard Fresenius, Chemisch-pharmazeutische Industrie KG, 6380 Bad Homburg Substanz zur Verwendung in einem kolloidalen Blutvolumenersatzmittel aus Hydroxyethylstärke und Hämoglobin
FR2378094A2 (fr) 1977-01-24 1978-08-18 Inst Nat Sante Rech Med Nouveaux reactifs biologiques constitues par des supports solides sur lesquels sont couples des composes organiques comportant un residu glucidique
EP0019403B1 (fr) * 1979-05-10 1985-07-31 American Hospital Supply Corporation Support de médicament en hydroxyalkylamidon
DE3029307A1 (de) * 1980-08-01 1982-03-04 Dr. Eduard Fresenius, Chemisch-pharmazeutische Industrie KG, 6380 Bad Homburg Haemoglobin enthaltendes blutersatzmittel
US4454161A (en) 1981-02-07 1984-06-12 Kabushiki Kaisha Hayashibara Seibutsu Kagaku Kenkyujo Process for the production of branching enzyme, and a method for improving the qualities of food products therewith
FI82266C (fi) 1982-10-19 1991-02-11 Cetus Corp Foerfarande foer framstaellning av il-2 -mutein.
DE3485810T2 (de) 1983-05-27 1992-12-10 Texas A & M University Syst Verfahren zur herstellung eines rekombinanten baculovirus-expressionsvektors.
EP0138572B1 (fr) 1983-10-11 1990-07-25 FIDIA S.p.A. Fractions d'acide hyaluronique ayant une activité pharmaceutique, méthodes pour leur préparation et compositions pharmaceutiques les contenant
NZ210501A (en) 1983-12-13 1991-08-27 Kirin Amgen Inc Erythropoietin produced by procaryotic or eucaryotic expression of an exogenous dna sequence
US4496689A (en) 1983-12-27 1985-01-29 Miles Laboratories, Inc. Covalently attached complex of alpha-1-proteinase inhibitor with a water soluble polymer
IL77081A (en) 1984-12-04 1999-10-28 Genetics Inst AND sequence encoding human erythropoietin, a process for its preparation and a pharmacological preparation of human erythropoietin
DE3501616A1 (de) 1985-01-17 1986-07-17 Schering AG, 1000 Berlin und 4709 Bergkamen Verfahren zur herstellung von hydroxylamin-derivaten
GB8610551D0 (en) 1986-04-30 1986-06-04 Hoffmann La Roche Polypeptide & protein derivatives
IT1203814B (it) 1986-06-30 1989-02-23 Fidia Farmaceutici Esteri dell'acido alginico
JP2594123B2 (ja) 1987-09-12 1997-03-26 株式会社林原生物化学研究所 減感作剤
EP0307827A3 (fr) 1987-09-15 1989-12-27 Kuraray Co., Ltd. Complexes moléculaires, procédé de préparation et utilisation en médecine
IL84252A (en) 1987-10-23 1994-02-27 Yissum Res Dev Co Phospholipase inhibiting compositions
DK110188D0 (da) 1988-03-02 1988-03-02 Claus Selch Larsen High molecular weight prodrug derivatives of antiinflammatory drugs
FR2630329B1 (fr) 1988-04-20 1991-07-05 Merieux Inst Conjugues macromoleculaires d'hemoglobine, leur procede de preparation et leurs applications
IT1219942B (it) 1988-05-13 1990-05-24 Fidia Farmaceutici Esteri polisaccaridici
US5420105A (en) 1988-09-23 1995-05-30 Gustavson; Linda M. Polymeric carriers for non-covalent drug conjugation
CA2006596C (fr) 1988-12-22 2000-09-05 Rika Ishikawa G-csf modifie chimiquement
JP2896580B2 (ja) 1989-08-25 1999-05-31 チッソ株式会社 アミロース―リゾチームハイブリッドと活性化糖およびその製造法
JP2838800B2 (ja) 1989-09-02 1998-12-16 株式会社林原生物化学研究所 減感作剤
WO1991005867A1 (fr) 1989-10-13 1991-05-02 Amgen Inc. Isoformes d'erythropoietine
JP2975632B2 (ja) 1990-03-30 1999-11-10 生化学工業株式会社 グリコサミノグリカン修飾プロテイン
US5169784A (en) 1990-09-17 1992-12-08 The Texas A & M University System Baculovirus dual promoter expression vector
US6172208B1 (en) 1992-07-06 2001-01-09 Genzyme Corporation Oligonucleotides modified with conjugate groups
AU5098193A (en) * 1992-09-01 1994-03-29 Berlex Laboratories, Inc. Glycolation of glycosylated macromolecules
NO934477L (no) 1992-12-09 1994-06-10 Ortho Pharma Corp PEG hydrazon- og PEG oksim-bindingdannende reagenser og proteinderivater derav
CA2110543A1 (fr) 1992-12-09 1994-06-10 David E. Wright Reactifs formant des chainons peg-hydrazone et peg-oxyme et derives proteiniques ainsi obtenus
NZ250375A (en) 1992-12-09 1995-07-26 Ortho Pharma Corp Peg hydrazone and peg oxime linkage forming reagents and protein derivatives
DK0646130T3 (da) 1993-03-16 1997-06-02 Hemosol Inc Selektiv tværbinding af hæmoglobiner ved hjælp af oxiderede, ringåbnede saccharider
WO1994028024A1 (fr) * 1993-06-01 1994-12-08 Enzon, Inc. Polymere modifie aux glucides presentant une activite erythropoïetique
IL110669A (en) 1993-08-17 2008-11-26 Kirin Amgen Inc Erythropoietin analogs
US5840900A (en) 1993-10-20 1998-11-24 Enzon, Inc. High molecular weight polymer-based prodrugs
JPH07188291A (ja) 1993-12-27 1995-07-25 Hayashibara Biochem Lab Inc 蛋白質とその製造方法並びに用途
US6214331B1 (en) 1995-06-06 2001-04-10 C. R. Bard, Inc. Process for the preparation of aqueous dispersions of particles of water-soluble polymers and the particles obtained
US5736533A (en) 1995-06-07 1998-04-07 Neose Technologies, Inc. Bacterial inhibition with an oligosaccharide compound
WO1997021452A2 (fr) 1995-12-14 1997-06-19 Advanced Magnetics, Inc. Promedicaments macromoleculaires d'analogues de nucleotides
US5723589A (en) * 1995-12-21 1998-03-03 Icn Pharmaceuticals Carbohydrate conjugated bio-active compounds
PL189698B1 (pl) 1996-03-12 2005-09-30 Pg Txl Co Kompozycja farmaceutyczna zawierająca lek przeciwnowotworowy i jej zastosowania
US5696152A (en) 1996-05-07 1997-12-09 Wisconsin Alumni Research Foundation Taxol composition for use as organ preservation and cardioplegic agents
TW517067B (en) * 1996-05-31 2003-01-11 Hoffmann La Roche Interferon conjugates
US5770645A (en) 1996-08-02 1998-06-23 Duke University Medical Center Polymers for delivering nitric oxide in vivo
US5851984A (en) 1996-08-16 1998-12-22 Genentech, Inc. Method of enhancing proliferation or differentiation of hematopoietic stem cells using Wnt polypeptides
US5990237A (en) 1997-05-21 1999-11-23 Shearwater Polymers, Inc. Poly(ethylene glycol) aldehyde hydrates and related polymers and applications in modifying amines
US5847110A (en) 1997-08-15 1998-12-08 Biomedical Frontiers, Inc. Method of reducing a schiff base
US6624142B2 (en) 1997-12-30 2003-09-23 Enzon, Inc. Trimethyl lock based tetrapartate prodrugs
DE19808079A1 (de) 1998-02-20 1999-08-26 Schering Ag Hydroxyethylstärke-Konjugate, Verfahren zu ihrer Herstellung und diese enthaltende pharmazeutische Mittel
EP1063317B1 (fr) 1998-03-05 2003-07-30 Asahi Glass Company Ltd. Cible de pulverisation cathodique, film conducteur transparent et son procede de production
US6153655A (en) 1998-04-17 2000-11-28 Enzon, Inc. Terminally-branched polymeric linkers and polymeric conjugates containing the same
US6660843B1 (en) 1998-10-23 2003-12-09 Amgen Inc. Modified peptides as therapeutic agents
US6261594B1 (en) 1998-11-25 2001-07-17 The University Of Akron Chitosan-based nitric oxide donor compositions
CZ299516B6 (cs) 1999-07-02 2008-08-20 F. Hoffmann-La Roche Ag Konjugát erythropoetinového glykoproteinu, zpusobjeho výroby a použití a farmaceutická kompozice sjeho obsahem
US7279176B1 (en) 1999-09-02 2007-10-09 Rice University Nitric oxide-producing hydrogel materials
US20020065410A1 (en) 1999-12-02 2002-05-30 Antrim Richard L. Branched starches and branched starch hydrolyzates
US6749865B2 (en) 2000-02-15 2004-06-15 Genzyme Corporation Modification of biopolymers for improved drug delivery
JP2001294601A (ja) 2000-04-11 2001-10-23 Akita Prefecture 高度分岐澱粉と該高度分岐澱粉の製造方法
JP2002003398A (ja) 2000-04-17 2002-01-09 Ltt Institute Co Ltd 徐放製剤、その製造法及びワクチン
DE10023051B4 (de) * 2000-05-11 2004-02-19 Roche Diagnostics Gmbh Verfahren zur Herstellung von Fluoresceinisothiocyanat-Sinistrin, dessen Verwendung und Fluoresceinisothiocyanat-Sinistrin enthaltende diagnostische Zubereitung
FR2811967B1 (fr) 2000-07-24 2002-12-13 Cebal Tube muni d'une tete de fixation pour divers bouchages et les divers bouchages munis de moyens de fixation sur ledit tube
DE10041541A1 (de) 2000-08-24 2002-03-14 Michael Duchene Rekombinante Allergene aus der Motte Plodia interpunctella
US6417347B1 (en) 2000-08-24 2002-07-09 Scimed Life Systems, Inc. High yield S-nitrosylation process
HUP0303854A2 (hu) 2000-09-08 2004-03-01 Gryhon Therapeutics, Inc. Szintetikus erythropoiesis-stimuláló proteinek
DE10105921A1 (de) 2001-02-09 2002-08-14 Braun Melsungen Ag An Kolloide gebundene Arzneiwirkstoffe
DE10126158A1 (de) 2001-05-30 2002-12-12 Novira Chem Gmbh Eine Methode zur Synthese von Gemischen einfach aktivierter und nicht aktivierter Polyoxyalkylene zur Modifizierung von Proteinen
DE10135694A1 (de) 2001-07-21 2003-02-06 Supramol Parenteral Colloids Amphiphile Stärke-und Hydroxyethylstärke-Konjugate
US7179617B2 (en) * 2001-10-10 2007-02-20 Neose Technologies, Inc. Factor IX: remolding and glycoconjugation of Factor IX
DK2431377T3 (en) 2001-10-26 2017-08-14 Noxxon Pharma Ag Modified L-nucleic acids
US6375846B1 (en) * 2001-11-01 2002-04-23 Harry Wellington Jarrett Cyanogen bromide-activation of hydroxyls on silica for high pressure affinity chromatography
DE10155098A1 (de) 2001-11-09 2003-05-22 Supramol Parenteral Colloids Mittel zur Prävention von mykotischen Kontaminationen bei der Zell- und Gewebekultur bakteriellen, pflanzlichen, animalischen und humanen Ursprungs, bestehend aus Polyen-Makrolid-Konjugaten mit Polysacchariden
US6916962B2 (en) 2001-12-11 2005-07-12 Sun Bio, Inc. Monofunctional polyethylene glycol aldehydes
JP4272537B2 (ja) 2002-03-13 2009-06-03 北京鍵▲凱▼科技有限公司 Y型分鎖親水性ポリマー誘導体、それらの調製方法、前記誘導体および薬剤分子の結合生成物、ならびに前記結合生成物を含む医薬組成物
DE10217994A1 (de) 2002-04-23 2003-11-06 Supramol Parenteral Colloids Konjugate von hyperverzweigten Polysacchariden
JP4583168B2 (ja) 2002-06-03 2010-11-17 ザ インスティテュート フォー システムズ バイオロジー 糖タンパク質を定量的プロテオ−ム分析する方法
US20040101546A1 (en) 2002-11-26 2004-05-27 Gorman Anne Jessica Hemostatic wound dressing containing aldehyde-modified polysaccharide and hemostatic agents
WO2004022630A2 (fr) 2002-09-09 2004-03-18 Nektar Therapeutics Al, Corporation Alcanales polymeres solubles dans l'eau
EP1591467A1 (fr) 2002-09-09 2005-11-02 Nektar Therapeutics Al, Corporation Conjugue d un polyethylene glycol comprenant un group terminal alcanal et un hormone de croissance humaine
DE10242076A1 (de) 2002-09-11 2004-03-25 Fresenius Kabi Deutschland Gmbh HAS-Allergen-Konjugate
EP1400533A1 (fr) 2002-09-11 2004-03-24 Fresenius Kabi Deutschland GmbH Polypeptides hasylés, érythropoiétine hasylée en particulier
ES2314238T3 (es) 2002-10-08 2009-03-16 Fresenius Kabi Deutschland Gmbh Conjugados de oligosacaridos farmaceuticamente activos.
DE10254745A1 (de) 2002-11-23 2004-06-03 Supramol Parenteral Colloids Gmbh Imidazolide von Polysaccharid Aldonsäuren, Verfahren zu ihrer Herstellung und Verwendung zur Kopplung an pharmazeutische Wirkstoffe
DE10256558A1 (de) 2002-12-04 2004-09-16 Supramol Parenteral Colloids Gmbh Ester von Polysaccharid Aldonsäuren, Verfahren zu ihrer Herstellung und Verwendung zur Kopplung an pharmazeutische Wirkstoffe
US20080274948A1 (en) 2003-08-08 2008-11-06 Fresenius Kabi Deutschland Gmbh Conjugates of Hydroxyalkyl Starch and G-Csf
US20080206182A1 (en) 2003-08-08 2008-08-28 Fresenius Kabi Deutschland Gmbh Conjugates of a Polymer and a Protein Linked by an Oxime Group
WO2005014655A2 (fr) 2003-08-08 2005-02-17 Fresenius Kabi Deutschland Gmbh Conjugues d'amidon d'hydroxyalkyle et de proteine
CA2534418A1 (fr) 2003-08-08 2005-02-17 Fresenius Kabi Deutschland Gmbh Conjugues d'amidon hydroxyalkyle et de g-csf
US20050169941A1 (en) 2004-01-29 2005-08-04 Andrew Lees Use of amino-oxy functional groups in the preparation of protein-polysaccharide conjugate vaccines
DE102004009783A1 (de) 2004-02-28 2005-09-15 Supramol Parenteral Colloids Gmbh Hyperverzweigte Stärkefraktion, Verfahren zu ihrer Herstellung und ihre Konjugate mit pharmazeutischen Wirkstoffen
WO2005092369A2 (fr) 2004-03-11 2005-10-06 Fresenius Kabi Deutschland Gmbh Conjugues d'hydroxy-ethyl-amidon et d'erythropoietine
JP5396019B2 (ja) 2004-03-11 2014-01-22 フレゼニウス・カビ・ドイチュラント・ゲゼルシャフト・ミット・ベシュレンクテル・ハフツング ヒドロキシアルキルデンプンとタンパク質とのコンジュゲート
CN101659704A (zh) 2004-03-11 2010-03-03 弗雷泽纽斯卡比德国有限公司 通过还原氨基化制备的羟烷基淀粉和蛋白质的偶联物
WO2005112954A1 (fr) 2004-05-14 2005-12-01 William Marsh Rice University Compositions de déclenchement d’oxyde nitrique et méthodes associées
KR20070110902A (ko) 2005-03-11 2007-11-20 프레제니우스 카비 도이치란트 게엠베하 비활성 출발 물질로부터 생물활성 당단백질의 생산
EP1762250A1 (fr) 2005-09-12 2007-03-14 Fresenius Kabi Deutschland GmbH Conjugués d' amidon hydroxyalkyle et une substance active obtenue par ligation chimique via thiazolidine
EP1942942A2 (fr) 2005-10-31 2008-07-16 Government of the United States of America, Represented by the Secretary, Department of Health and Human Services Diazeniumdiolates lies par un carbone, liberant de l'oxyde nitrique et derives de polysaccharides
JP2009093397A (ja) 2007-10-09 2009-04-30 Panasonic Corp タッチパネル及びこれを用いた入力装置
EP2070951A1 (fr) 2007-12-14 2009-06-17 Fresenius Kabi Deutschland GmbH Procédé de production d'un dérivé hydroxyalkyle de l'amidon avec deux liens
EP2070950A1 (fr) 2007-12-14 2009-06-17 Fresenius Kabi Deutschland GmbH Dérivés hydroxyalkylés de l'amidon et leur procédé de préparation
US9434610B2 (en) 2008-10-07 2016-09-06 Rexahn Pharmaceuticals, Inc. HPMA—docetaxel conjugates and uses therefore

Patent Citations (53)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3191291A (en) * 1959-01-21 1965-06-29 Continental Can Co Art of producing very thin steel and like sheets in wide strips
US4179337A (en) * 1973-07-20 1979-12-18 Davis Frank F Non-immunogenic polypeptides
US4125492A (en) * 1974-05-31 1978-11-14 Pedro Cuatrecasas Affinity chromatography of vibrio cholerae enterotoxin-ganglioside polysaccharide and the biological effects of ganglioside-containing soluble polymers
US4001401A (en) * 1975-02-02 1977-01-04 Alza Corporation Blood substitute and blood plasma expander comprising polyhemoglobin
US4061736A (en) * 1975-02-02 1977-12-06 Alza Corporation Pharmaceutically acceptable intramolecularly cross-linked, stromal-free hemoglobin
US4053590A (en) * 1975-02-27 1977-10-11 Alza Corporation Compositions of matter comprising macromolecular hemoglobin
US4001200A (en) * 1975-02-27 1977-01-04 Alza Corporation Novel polymerized, cross-linked, stromal-free hemoglobin
US4064118A (en) * 1975-10-22 1977-12-20 Hematech Inc. Blood substitute based on hemoglobin
US4261973A (en) * 1976-08-17 1981-04-14 Pharmacia Ab Allergen-containing substances
US4412989A (en) * 1981-06-10 1983-11-01 Ajinomoto Company Incorporated Oxygen carrier
US4703008A (en) * 1983-12-13 1987-10-27 Kiren-Amgen, Inc. DNA sequences encoding erythropoietin
US4952496A (en) * 1984-03-30 1990-08-28 Associated Universities, Inc. Cloning and expression of the gene for bacteriophage T7 RNA polymerase
US4667016A (en) * 1985-06-20 1987-05-19 Kirin-Amgen, Inc. Erythropoietin purification
US4766106A (en) * 1985-06-26 1988-08-23 Cetus Corporation Solubilization of proteins for pharmaceutical compositions using polymer conjugation
US4863964A (en) * 1985-07-02 1989-09-05 Biomedical Frontiers, Inc. Method for the stabilization of deferoxamine to chelate free ions in physiological fluid
US5217998A (en) * 1985-07-02 1993-06-08 Biomedical Frontiers, Inc. Composition for the stabilization of deferoxamine to chelate free ions in physiological fluid
US5079337A (en) * 1986-07-02 1992-01-07 Pasteur Merieux Serums Et Vaccins S.A. Macromolecular conjugates of hemoglobin, a procedure for their preparation and their uses
US5362853A (en) * 1986-12-23 1994-11-08 Kyowa Hakko Kogyo Co., Ltd. Polypeptide derivatives of human granulocyte colony stimulating factor
US5214132A (en) * 1986-12-23 1993-05-25 Kyowa Hakko Kogyo Co., Ltd. Polypeptide derivatives of human granulocyte colony stimulating factor
US4904584A (en) * 1987-12-23 1990-02-27 Genetics Institute, Inc. Site-specific homogeneous modification of polypeptides
US4847325A (en) * 1988-01-20 1989-07-11 Cetus Corporation Conjugation of polymer to colony stimulating factor-1
US4900780A (en) * 1988-05-25 1990-02-13 Masonic Medical Research Laboratory Acellular resuscitative fluid
US4925677A (en) * 1988-08-31 1990-05-15 Theratech, Inc. Biodegradable hydrogel matrices for the controlled release of pharmacologically active agents
US5218092A (en) * 1988-09-29 1993-06-08 Kyowa Hakko Kogyo Co., Ltd. Modified granulocyte-colony stimulating factor polypeptide with added carbohydrate chains
US5068321A (en) * 1988-10-27 1991-11-26 Wolff Walsrode Aktiengesellschaft Carbonic acid esters of polysaccharides and a process for their production
US6261800B1 (en) * 1989-05-05 2001-07-17 Genentech, Inc. Luteinizing hormone/choriogonadotropin (LH/CG) receptor
US5218108A (en) * 1989-06-16 1993-06-08 Fresenius Ag Hydroxylethylstarch (hes) as plasma expander and process for preparing hes
US5543332A (en) * 1991-07-04 1996-08-06 Immunodex K/S Water-soluble, polymer-based reagents and conjugates comprising moieties derived from divinyl sulfone
US5281698A (en) * 1991-07-23 1994-01-25 Cetus Oncology Corporation Preparation of an activated polymer ester for protein conjugation
US5484903A (en) * 1991-09-17 1996-01-16 Wolff Walsrode Aktiengesellschaft Process for the production of polysaccharide carbonates
US5622718A (en) * 1992-09-25 1997-04-22 Keele University Alginate-bioactive agent conjugates
US5470843A (en) * 1992-12-11 1995-11-28 Hoechst Aktiengesellschaft Carbohydrate-containing polymers, their preparation and use
US5581476A (en) * 1993-01-28 1996-12-03 Amgen Inc. Computer-based methods and articles of manufacture for preparing G-CSF analogs
US5876980A (en) * 1995-04-11 1999-03-02 Cytel Corporation Enzymatic synthesis of oligosaccharides
US5880270A (en) * 1995-06-07 1999-03-09 Cellpro, Incorporated Aminooxy-containing linker compounds for formation of stably-linked conjugates and methods related thereto
US6083909A (en) * 1996-07-08 2000-07-04 Fresenius Ag Haemoglobin-hydroxyethyl starch conjugates as oxygen carriers
US6011008A (en) * 1997-01-08 2000-01-04 Yissum Research Developement Company Of The Hebrew University Of Jerusalem Conjugates of biologically active substances
US5952347A (en) * 1997-03-13 1999-09-14 Merck & Co., Inc. Quinoline leukotriene antagonists
US6299881B1 (en) * 1997-03-24 2001-10-09 Henry M. Jackson Foundation For The Advancement Of Military Medicine Uronium salts for activating hydroxyls, carboxyls, and polysaccharides, and conjugate vaccines, immunogens, and other useful immunological reagents produced using uronium salts
US6340746B1 (en) * 1997-08-07 2002-01-22 University Of Utah Prodrugs and conjugates of thiol- and selenol- containing compounds and methods of use thereof
US6875594B2 (en) * 1997-11-13 2005-04-05 The Rockefeller University Methods of ligating expressed proteins
US6500930B2 (en) * 1998-03-31 2002-12-31 Hemosol Inc. Hemoglobin-polysaccharide conjugates
US6596861B1 (en) * 1999-03-12 2003-07-22 Aventis Pasteur S.A. Method for the reductive amination of polysaccharides
US6555660B2 (en) * 2000-01-10 2003-04-29 Maxygen Holdings Ltd. G-CSF conjugates
US6586398B1 (en) * 2000-04-07 2003-07-01 Amgen, Inc. Chemically modified novel erythropoietin stimulating protein compositions and methods
US20050063943A1 (en) * 2001-03-16 2005-03-24 Klaus Sommermeyer Conjugated of hydroxyalkyl starch and an active agent
US20040180858A1 (en) * 2001-06-21 2004-09-16 Klaus Sommermeyer Water-soluble antibiotic comprising an amino sugar, in the form of a polysaccharide conjugate
US7115576B2 (en) * 2001-06-21 2006-10-03 Fresenius Kabi Deutschland Gmbh Water-soluble antibiotic comprising an amino sugar, in the form of a polysaccharide conjugate
US20040043446A1 (en) * 2001-10-19 2004-03-04 Neose Technologies, Inc. Alpha galalctosidase a: remodeling and glycoconjugation of alpha galactosidase A
US7285661B2 (en) * 2002-02-20 2007-10-23 Fresenius Kabi Deutschland Gmbh Starch derivatives, starch active substance conjugates, method for the production thereof and their use as medicaments
US20050181985A1 (en) * 2002-03-06 2005-08-18 Jurgen Hemberger Coupling proteins to a modified polysaccharide
US20060217293A1 (en) * 2002-03-06 2006-09-28 Michele Orlando Coupling low-molecular substances to a modified polysaccharide
US20060019877A1 (en) * 2002-09-11 2006-01-26 Conradt Harald S Hasylated polypeptides

Cited By (52)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060188472A1 (en) * 2001-03-16 2006-08-24 Fresenius Kabi Deutschland Gmbh, A Germany Corporation HAS-active ingredient conjugates
US7816516B2 (en) 2001-03-16 2010-10-19 Fresenius Kabi Deutschland Gmbh Conjugates of hydroxyalkyl starch and an active agent
US20050063943A1 (en) * 2001-03-16 2005-03-24 Klaus Sommermeyer Conjugated of hydroxyalkyl starch and an active agent
US20060217293A1 (en) * 2002-03-06 2006-09-28 Michele Orlando Coupling low-molecular substances to a modified polysaccharide
US8916518B2 (en) 2002-03-06 2014-12-23 Fresenius Kabi Deutschland Gmbh Coupling proteins to a modified polysaccharide
US20050181985A1 (en) * 2002-03-06 2005-08-18 Jurgen Hemberger Coupling proteins to a modified polysaccharide
US7541328B2 (en) * 2002-03-06 2009-06-02 Fresenius Kabi Deutschland Gmbh Coupling proteins to a modified polysaccharide
US8466277B2 (en) 2002-03-06 2013-06-18 Fresenius Kabi Deutschland Gmbh Coupling low-molecular substances to a modified polysaccharide
US20060019877A1 (en) * 2002-09-11 2006-01-26 Conradt Harald S Hasylated polypeptides
US7815893B2 (en) 2002-09-11 2010-10-19 Fresenius Kabi Deutschland Gmbh Hydroxyalkyl starch derivatives
US8618266B2 (en) 2002-09-11 2013-12-31 Fresenius Kabi Deutschland Gmbh Hasylated polypeptides
US8475765B2 (en) 2002-09-11 2013-07-02 Fresenius Kabi Deutschland Gmbh Hydroxyalkyl starch derivatives
US20050234230A1 (en) * 2002-09-11 2005-10-20 Norbert Zander Hydroxyalkyl starch derivatives
US20060121062A1 (en) * 2002-09-11 2006-06-08 Wolfram Eichner Conjugated hydroxyalkyl starch allergen compounds
US20110054152A1 (en) * 2002-09-11 2011-03-03 Fresenius Kabi Deutschland Gmbh Hydroxyalkyl Starch Derivatives
US7538092B2 (en) 2002-10-08 2009-05-26 Fresenius Kabi Deutschland Gmbh Pharmaceutically active oligosaccharide conjugates
US20060100163A1 (en) * 2002-10-08 2006-05-11 Michele Orlando Pharmaceutically active oligosaccharide conjugates
US20060052342A1 (en) * 2002-12-04 2006-03-09 Klaus Sommermeyer Aldonic acid esters, methods for producing the same, and methods for producing pharmaceutical active ingredients coupled to polysaccharides or polysaccharide derivatives on free amino groups
US20080274948A1 (en) * 2003-08-08 2008-11-06 Fresenius Kabi Deutschland Gmbh Conjugates of Hydroxyalkyl Starch and G-Csf
US20080206182A1 (en) * 2003-08-08 2008-08-28 Fresenius Kabi Deutschland Gmbh Conjugates of a Polymer and a Protein Linked by an Oxime Group
US20120046235A9 (en) * 2004-03-11 2012-02-23 Nobert Zander Conjugates of hydroxyalkyl starch and a protein, prepared by native chemical ligation
US8017739B2 (en) 2004-03-11 2011-09-13 Fresenius Kabi Deutschland Gmbh Conjugates of hydroxyalkyl starch and a protein
US20090047251A1 (en) * 2004-03-11 2009-02-19 Wolfram Eichner Conjugates of hydroxyalkyl starch and a protein
US8840879B2 (en) 2004-03-11 2014-09-23 Fresenius Kabi Deutschland Gmbh Conjugates of hydroxyalkyl starch and a protein
US8287850B2 (en) 2004-03-11 2012-10-16 Fresenius Kabi Deutschland Gmbh Conjugates of hydroxyalkyl starch and a protein, prepared by reductive amination
US20100311670A1 (en) * 2004-03-11 2010-12-09 Nobert Zander Conjugates of hydroxyalkyl starch and a protein, prepared by native chemical ligation
US20070087961A1 (en) * 2004-03-11 2007-04-19 Wolfram Eichner Conjugates of hydroxyalkyl starch and erythropoietin
US20110200555A1 (en) * 2004-03-11 2011-08-18 Fresenius Kabi Deutschland Gmbh Conjugates of hydroxyalkyl starch and a protein
US20100062973A1 (en) * 2005-03-11 2010-03-11 Fresenius Kabi Deutschland Gmbh Production of bioactive glycoproteins from inactive starting material
US20080207562A1 (en) * 2005-09-12 2008-08-28 Fresenius Kabi Deutschland Gmbh Conjugates of Hydroxyalkyl Starch and Active Substance, Prepared by Chemical Ligation Via Thiazolidine
WO2009077153A1 (fr) * 2007-12-14 2009-06-25 Fresenius Kabi Deutschland Gmbh Procédé de production d'un dérivé d'amidon hydroxyalkylé comportant deux séquences de liaison
AU2008337930B2 (en) * 2007-12-14 2013-12-05 Fresenius Kabi Deutschland Gmbh Method for producing a hydroxyalkyl starch derivative with two linkers
KR101578955B1 (ko) 2007-12-14 2015-12-21 프레제니우스 카비 도이치란트 게엠베하 히드록시알킬 전분 유도체 및 이의 제조 방법
EP2070950A1 (fr) * 2007-12-14 2009-06-17 Fresenius Kabi Deutschland GmbH Dérivés hydroxyalkylés de l'amidon et leur procédé de préparation
US20100297078A1 (en) * 2007-12-14 2010-11-25 Fresenius Kabi Deutschland Gmbh Method for producing a hydroxyalkyl starch derivative with two linkers
EP2070951A1 (fr) * 2007-12-14 2009-06-17 Fresenius Kabi Deutschland GmbH Procédé de production d'un dérivé hydroxyalkyle de l'amidon avec deux liens
US20100305033A1 (en) * 2007-12-14 2010-12-02 Fresenius Kabi Deutschland Gmbh Hydroxyalkyl starch derivatives and process for their preparation
US8404834B2 (en) 2007-12-14 2013-03-26 Fresenius Kabi Deutschland Gmbh Hydroxyalkyl starch derivatives and process for their preparation
WO2009077154A1 (fr) * 2007-12-14 2009-06-25 Fresenius Kabi Deutschland Gmbh Dérivés d'amidon d'hydroxyalkyle et leur procédé de préparation
EA018222B1 (ru) * 2007-12-14 2013-06-28 Фрезениус Каби Дойчланд Гмбх Производные гидроксиалкилкрахмала и способ их получения
KR20100099282A (ko) * 2007-12-14 2010-09-10 프레제니우스 카비 도이치란트 게엠베하 히드록시알킬 전분 유도체 및 이의 제조 방법
US10138283B2 (en) 2008-07-23 2018-11-27 Ambrx, Inc. Modified bovine G-CSF polypeptides and their uses
WO2010011735A2 (fr) 2008-07-23 2010-01-28 Ambrx, Inc. Polypeptides g-csf bovins modifiés et leurs utilisations
EP3225248A1 (fr) 2008-07-23 2017-10-04 Ambrx, Inc. Polypeptides g-csf bovins modifiés et leurs utilisations
EP2805964A1 (fr) 2009-12-21 2014-11-26 Ambrx, Inc. Polypeptides modifiés de somatotrophine bovine et leurs utilisations
EP2805965A1 (fr) 2009-12-21 2014-11-26 Ambrx, Inc. Polypeptides modifiés de somatotrophine bovine et leurs utilisations
WO2012004009A1 (fr) * 2010-07-09 2012-01-12 Fresenius Kabi Deutschland Gmbh Conjugués comprenant de l'hydroxyalkylamidon et un agent cytotoxique et leur procédé de synthèse
WO2012004005A1 (fr) * 2010-07-09 2012-01-12 Fresenius Kabi Deutschland Gmbh Conjugués comprenant de l'hydroxyalkylamidon et un agent cytotoxique et leur procédé de synthèse
WO2012004007A1 (fr) * 2010-07-09 2012-01-12 Fresenius Kabi Deutschland Gmbh Conjugués comprenant un amidon hydroxyalkylé et un agent cytotoxique et leur procédé de préparation
WO2012004006A1 (fr) * 2010-07-09 2012-01-12 Fresenius Kabi Deutschland Gmbh Conjugués comprenant un amidon hydroxyalkylé et un agent cytotoxique et leur procédé de préparation
US11273202B2 (en) 2010-09-23 2022-03-15 Elanco Us Inc. Formulations for bovine granulocyte colony stimulating factor and variants thereof
US10787476B2 (en) 2013-09-24 2020-09-29 Ajinomoto Co., Inc. Glycoamino acid and use thereof

Also Published As

Publication number Publication date
PL374490A1 (en) 2005-10-31
RU2329274C2 (ru) 2008-07-20
AU2003260393A1 (en) 2004-04-30
US20100317609A1 (en) 2010-12-16
ES2399006T3 (es) 2013-03-25
WO2004024777A1 (fr) 2004-03-25
US20050234230A1 (en) 2005-10-20
MXPA05002594A (es) 2005-05-27
BR0314227A (pt) 2005-10-25
PT1398322E (pt) 2006-08-31
NO20051427L (no) 2005-05-31
MXPA05002593A (es) 2005-05-27
CN100348618C (zh) 2007-11-14
ES2213507T3 (es) 2010-04-15
EP1398322A1 (fr) 2004-03-17
EP2316850A3 (fr) 2011-11-09
RU2005110415A (ru) 2006-08-27
AU2003255406A1 (en) 2004-04-30
JP2010248528A (ja) 2010-11-04
HK1136226A1 (en) 2010-06-25
IL166506A (en) 2010-02-17
NO20051419L (no) 2005-05-30
JP5275293B2 (ja) 2013-08-28
ES2213507T1 (es) 2004-09-01
EP2316850A2 (fr) 2011-05-04
KR20050072417A (ko) 2005-07-11
DE03020423T1 (de) 2004-07-15
KR101174510B1 (ko) 2012-08-16
HK1063478A1 (en) 2004-12-31
EP2017287A2 (fr) 2009-01-21
ES2213505T3 (es) 2006-11-16
IL200150A0 (en) 2011-07-31
IL166930A (en) 2012-08-30
KR101045401B1 (ko) 2011-06-30
EP2143736A1 (fr) 2010-01-13
CA2496317A1 (fr) 2004-03-25
RU2326891C2 (ru) 2008-06-20
KR101045422B1 (ko) 2011-06-30
US8475765B2 (en) 2013-07-02
AR045234A1 (es) 2005-10-19
PL216545B1 (pl) 2014-04-30
CN1681846A (zh) 2005-10-12
SI1398328T1 (sl) 2010-02-26
PL375037A1 (en) 2005-11-14
WO2004024777A8 (fr) 2005-03-24
DK1398328T3 (da) 2010-03-08
ES2213505T1 (es) 2004-09-01
JP2010265464A (ja) 2010-11-25
US20060019877A1 (en) 2006-01-26
IL166506A0 (en) 2006-01-15
AU2011200219A1 (en) 2011-02-10
CN1681844A (zh) 2005-10-12
EP2154160A1 (fr) 2010-02-17
AU2009238372B2 (en) 2012-08-23
AU2011200158B2 (en) 2012-08-02
ZA200600651B (en) 2007-04-25
HK1063475A1 (en) 2004-12-31
BR0314107A (pt) 2005-07-19
DE20321836U1 (de) 2011-04-07
CY1109813T1 (el) 2014-09-10
DE60304640T2 (de) 2007-05-16
ES2213506T3 (es) 2009-02-16
KR20050075336A (ko) 2005-07-20
MX2010006175A (es) 2010-07-05
DE60304640D1 (de) 2006-05-24
EP2143736B1 (fr) 2012-11-07
PT1398327E (pt) 2008-10-31
US7815893B2 (en) 2010-10-19
ATE406387T1 (de) 2008-09-15
EP1398322B1 (fr) 2006-04-19
JP2011089125A (ja) 2011-05-06
AU2003255406B2 (en) 2009-09-10
CA2495242A1 (fr) 2004-03-25
HK1063477A1 (en) 2004-12-31
US20120046240A9 (en) 2012-02-23
PL374969A1 (en) 2005-11-14
AR041097A1 (es) 2005-05-04
DE60323192D1 (de) 2008-10-09
ATE323723T1 (de) 2006-05-15
KR20050061466A (ko) 2005-06-22
AU2003253399A1 (en) 2004-04-30
JP2006516534A (ja) 2006-07-06
AU2009238372A1 (en) 2009-12-17
AU2003260393B2 (en) 2009-10-01
EP1398328A1 (fr) 2004-03-17
EP2272865A3 (fr) 2014-06-18
JP5455821B2 (ja) 2014-03-26
MXPA05002591A (es) 2005-09-20
SI1398327T1 (sl) 2009-02-28
DK1398322T3 (da) 2006-08-21
EP1398327A1 (fr) 2004-03-17
ES2213506T1 (es) 2004-09-01
US8618266B2 (en) 2013-12-31
PL217085B1 (pl) 2014-06-30
WO2004024776A1 (fr) 2004-03-25
ATE449112T1 (de) 2009-12-15
CA2496317C (fr) 2014-02-18
EP1398328B1 (fr) 2009-11-18
RU2005110412A (ru) 2006-01-20
DE03020424T1 (de) 2004-07-15
DK1398327T3 (da) 2008-11-10
AU2003253399B2 (en) 2010-10-14
BR0314106A (pt) 2005-07-19
EP2272865A2 (fr) 2011-01-12
TW200418875A (en) 2004-10-01
DE60330098D1 (de) 2009-12-31
RU2005110423A (ru) 2006-01-20
US20110054152A1 (en) 2011-03-03
EP1398327B1 (fr) 2008-08-27
AU2011200158A1 (en) 2011-02-03
WO2004024761A1 (fr) 2004-03-25
AR045235A1 (es) 2005-10-19
DE03020425T1 (de) 2004-07-15
PT1398328E (pt) 2010-02-19
IL166931A (en) 2012-06-28
EP2017287A3 (fr) 2009-11-04
AU2011200219B2 (en) 2012-05-17
RU2328505C2 (ru) 2008-07-10

Similar Documents

Publication Publication Date Title
US8618266B2 (en) Hasylated polypeptides
CA2799437C (fr) Procede de production de derives d'amidon hydroxyalkyle
EP1681303B1 (fr) Polypeptides HASylés, en particulier érythropoiétine HASylée

Legal Events

Date Code Title Description
AS Assignment

Owner name: FRESENIUS KABI DEUTSCHLAND GMBH, GERMANY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:ZANDER, NORBERT;CONRADT, HARALD;EICHNER, WOLFRAM;REEL/FRAME:016491/0230;SIGNING DATES FROM 20050520 TO 20050607

STCB Information on status: application discontinuation

Free format text: EXPRESSLY ABANDONED -- DURING EXAMINATION