WO2005016973A1 - Polysialic acid derivatives - Google Patents

Polysialic acid derivatives Download PDF

Info

Publication number
WO2005016973A1
WO2005016973A1 PCT/GB2004/003488 GB2004003488W WO2005016973A1 WO 2005016973 A1 WO2005016973 A1 WO 2005016973A1 GB 2004003488 W GB2004003488 W GB 2004003488W WO 2005016973 A1 WO2005016973 A1 WO 2005016973A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
polysaccharide
unit
groups
functional
Prior art date
Application number
PCT/GB2004/003488
Other languages
French (fr)
Inventor
Dale Howard Hreczuk-Hirst
Sanjay Jain
Peter Laing
Gregory Gregoriadis
Iaonnis Papaioannou
Original Assignee
Lipoxen Technologies Limited
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Lipoxen Technologies Limited filed Critical Lipoxen Technologies Limited
Priority to JP2006523054A priority Critical patent/JP2007501888A/en
Priority to KR1020067002875A priority patent/KR101270692B1/en
Priority to DE602004009314T priority patent/DE602004009314T2/en
Priority to EP04768054A priority patent/EP1654289B1/en
Priority to US10/568,111 priority patent/US7691826B2/en
Publication of WO2005016973A1 publication Critical patent/WO2005016973A1/en
Priority to CNA2005800345881A priority patent/CN101039965A/en
Priority to EP16176511.0A priority patent/EP3184551A1/en
Priority to US11/660,128 priority patent/US7875708B2/en
Priority to JP2007525356A priority patent/JP5197009B2/en
Priority to ES05794259.1T priority patent/ES2593318T3/en
Priority to EP05794259.1A priority patent/EP1776389B1/en
Priority to PCT/GB2005/003160 priority patent/WO2006016168A2/en
Priority to US12/717,073 priority patent/US20100221808A1/en
Priority to US12/987,878 priority patent/US8293888B2/en
Priority to US13/647,326 priority patent/US8765936B2/en
Priority to US13/650,048 priority patent/US9102714B2/en
Priority to US14/251,596 priority patent/US9102699B2/en
Priority to US14/791,468 priority patent/US9920137B2/en
Priority to US14/791,467 priority patent/US20150344591A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08BPOLYSACCHARIDES; DERIVATIVES THEREOF
    • C08B37/00Preparation of polysaccharides not provided for in groups C08B1/00 - C08B35/00; Derivatives thereof
    • C08B37/0006Homoglycans, i.e. polysaccharides having a main chain consisting of one single sugar, e.g. colominic acid
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/715Polysaccharides, i.e. having more than five saccharide radicals attached to each other by glycosidic linkages; Derivatives thereof, e.g. ethers, esters
    • 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
    • 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
    • A61K47/68Medicinal 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 antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/006General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length of peptides containing derivatised side chain amino acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/107General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length by chemical modification of precursor peptides
    • C07K1/1072General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length by chemical modification of precursor peptides by covalent attachment of residues or functional groups
    • C07K1/1077General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length by chemical modification of precursor peptides by covalent attachment of residues or functional groups by covalent attachment of residues other than amino acids or peptide residues, e.g. sugars, polyols, fatty acids
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/13Labelling of peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/001Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof by chemical synthesis
    • 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
    • C08B37/00Preparation of polysaccharides not provided for in groups C08B1/00 - C08B35/00; Derivatives thereof
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/96Stabilising an enzyme by forming an adduct or a composition; Forming enzyme conjugates

Definitions

  • POLYSIALIC ACID DERIVATIVES The present invention relates to polysialic acid derivatives which are useful for conjugation to drugs, proteins and peptides, or to drug delivery systems such as liposomes, having sulfhydryl groups, as well as to conjugated products process for synthesising the derivatives and the conjugates and novel synthetic intermediates.
  • the extended presence of drugs either within the vascular system or in extravascular use is often a pre-requisite for their optimal use.
  • Many antibiotics and cytostatics for instance, as well as a variety of therapeutic peptides and proteins, and liposomes are removed from the circulation prematurely and before effective concentrations in target tissues can be achieved.
  • polysialic acid is rendered reactive, by generating an aldehyde group at the non-reducing end by oxidation of the 7, 8-vicinaldiol moiety with sodium periodate.
  • the aldehyde group was then reacted with primary amine groups on proteins, generally assumed to be epsilon-amino groups of lysine moieties of the protein or N-terminal amine groups.
  • the reaction forms a Schiff base which is reduced by cyanoborohydride to a secondary amine.
  • derivatisation with other molecules could be carried out in the presence of denaturant to achieve increased levels of derivatisation. Examples of other derivatising agents are polyethylene glycol compounds.
  • Activated PEG compounds such as tresyl- PEG and succinimidyl succinate ester of PEG were mentioned.
  • succinimidyl succinate activated PEG, which is believed to react with amine groups.
  • PEG derivatives having functional groups for coupling to thiol groups are commercially available.
  • the functional groups may be maleimide, vinyl sulfone, iodoacetamide or orthopyridyl disulphide groups. Since these reagents react specifically with cysteines, and since proteins have fewer cysteines on their surfaces than lysine groups, the derivatisation is more controllable. Furthermore, in the absence of a free cysteine in a native protein, one or more free cysteines may be added by genetic engineering.
  • a novel compound comprising a polysialic acid having a moiety linked at one or each terminal unit which includes a functional group selected from N-maleimido groups vinysulphone groups, N-iodoacetamide groups and orthopyridyl disulphide groups.
  • the terminal unit to which the moiety is linked may be at the non- reducing end of the polysialic acid or at the reducing end of the polysialic acid.
  • the terminal sialic acid unit has been subjected to a preliminary chemical reaction to generate useful functional groups to which a maleimide-group containing reagent may be linked.
  • a first reduction step converts the ketal unit at the reducing end into a reduced ring opened form, having vicinal diols.
  • the vicinal diols are subsequently oxidised using sodium periodate to form an aldehyde moiety at the carbon atom previously forming the 7-carbon of the reducing terminal unit.
  • the terminal unit will be simultaneously activated.
  • the moiety which includes the functional group may be linked directly to the polysialic acid unit or, more conveniently, may be linked via a difunctional organic group, such as an alkane diyl group, an arylene group or an oligo(alkyleneoxy) alkane group, or alternatively an oligo peptidyl group.
  • the linkage to the polysialic acid may be a secondary amine, a hydrazone, an alkyl hydrazine an ester or a peptidyl group.
  • the moiety may be generated by reaction of a polysialic acid substrate with a heterobifunctional reagent. The process form a further aspect of the invention.
  • Reagents useful for introducing the selected functional groups are commercially available.
  • a compound which will introduce a maleimide group onto an amine moiety without introducing any additional linker moiety is N- methoxy-carbonyl-maleimide.
  • the reagents include a second functional group for reaction with a group on the polysialic acid which may be the aldehyde group described above, or an amine group.
  • Suitable second functional groups include N-hydroxy succinimide esters and their sulfosuccimide analogues and hydrazides.
  • the compound is a N- maleimido-alkanoic acid hydrazide or an N-maleimidoarylalkanoic acid hydrazide i.e.
  • X is a N-maleimido, N-iodoacetamido, S-vinylsulphonyl or S- orthopyridyldisulphide group
  • R is alkane-diyl, arylene or aralkylene alkarylene, alkylene- oxaalkylene, or alkylene-oligooxa-alkylene or alkyl-oligopeptidyl-alkyl group
  • Y is a hydrazide, amine or N-hydroxysuccinimide group.
  • R is C,_ 6 alkanediyl, C 2.3 -alkyl-oxa-C 2 . 3 -alkylene, C 2 . 3 alkyl-oligo(oxa-C 2 . 3 alkylene), or C 2 . 6 alkylene phenyl.
  • X is N-maleimido or orthopyridyldisulphide.
  • Y is a hydrazide or a hydroxyl succinimide
  • Compounds which may be reacted with an aldehyde group, and which include a linker moiety and introduce a maleimide group include N-[ ⁇ - maleimidopropionic acid] hydrazide and 4-(4-N-maleimidophenyl)butyric acid hydrazide.
  • the hydrazide group reacts with the aldehyde to form a stable hydrazone group.
  • a suitable heterobifunctional compound which includes an oligo(ethyleneoxy) ethylene group is a compound comprising a polyethylene glycol (poly(ethyelenoxy)) group with, at one end, N-hydroxy succinimide (NHS) group and at the other end the functional group.
  • the NHS group reacts with amine groups to form stable amide linkages.
  • Heterobifunctional polyethyleneglycols with NHS at one end and either vinylsulphone or maleimide at the other end are available.
  • heterobifunctional reagents include, 3-(2-pyridyldithio)propionyl hydrazide, N-succinimidyl-3-[2-pyridyldithio]propionate, succinimidyl-H-[N- maleimidomethyl]cyclohexane-1-carboxylate), m-maleimidobenzoyl-N- hydroxysuccinimide ester, N-succinimidyl-[4-iodoacetyl] amino benzoate, N- [gamma-maleimidobutyryloxyjsuccinimide ester, N-[epsilon- maleimidocaproyloxy]succinimide ester and N-succinimidyl iodoacetate.
  • Sialic acids also known as nonulosonic acids
  • N-acetylneuraminic acid also known as 5-(acetylamino)-3,5-dideoxy-D-glycero-D-galacto-nonulosonic, lactaminic acid and O-sialic acid
  • Polysialic acids may be linked 2-8 and/or 2-9, usually in the ⁇ - configuration. In some polysialic acids the linkages are alternating 2 ⁇ 8 and 2 ⁇ 9.
  • the invention is also of utility for heteropolymeric polysaccharides comprising glycosyl units other than sialic acid units. Polysialic acids are generally found to be non-toxic and substantially non-immunogenic. Furthermore the biodegration units, sialic acid, is not known to be toxic and, indeed sialic acids are widely found in animal proteins and cells, including blood cells and circulating proteins.
  • Polysaccharide compounds containing many sialic acid units are polysaccharides produced by Esche chia coli, Moraxella nonliquifaciens, Pasteurella aeroginosis and Neisseria meningitidis or derivatives thereof.
  • colominic acid derived (by hydrolysis to shorten the chain lengths) from E. coli Kl comprises ⁇ 2-8 linked sialic acid units.
  • Polysaccharide from E. coli K92 strain comprises alternating 2-8 and 2-9 linked sialic acid units.
  • Polysaccharide C of N. meningitidis group C has 2-9 linked sialic acid units.
  • group B polysaccharides One group of polysaccharide compounds which has been found to be of particular utility in the invention is group B polysaccharides.
  • These compounds are produced by N. meningitidis, M. nonliquifaciens, P. aeroginosis A2 and E. coli K1. These compounds comprise a polysaccharide component comprising sialic acid residues and a phospholipid component.
  • the sialic acid residues are linked (2 - 8)- ⁇ , the naturally occurring polymer consisting of about 200 residues.
  • Some of the glycolipid molecules, especially the high molecular weight compounds appear to have a covalently attached phospholipid at the reducing end of the polysaccharide component. It is preferable for the bacteria from which the polysaccharide compound is derived to be non-pathogenic for convenience during production.
  • the polysaccharide is particularly suitable therefore for the polysaccharide to be derived from a non-pathogenic strain of E. coli such as E. coli K92 or, preferably, K1 which is non-immunogenic.
  • E. coli K92 and K1 isolates are well-known and any such type of any such strains can be used as sources of suitable polysaccharide.
  • the polysialic acid should have at least 2, preferably at least 5, more preferably at least 10, for instance more than 50 sialic acid units.
  • a conjugate of a protein and the novel activated polysaccharide is also provided.
  • the novel compound comprises a protein with at least one cysteine unit and, linked through a thioether bond to the side chain of a cysteine unit, a polysialic acid through a moiety joined at one or both terminal unit of the polysialic acid.
  • the polysialic acid derivative was a N-maleimido group
  • the moiety will include a N-succinimidyl group, with the thioester linked at the ⁇ - carbon atom.
  • the moiety also comprises a secondary amine, a hydrazone or an amide bond.
  • a polysialic acid is reacted with a heterobifunctional reagent having a first functional group for reaction with sulfhydryl groups and a second functional group different to the first group whereby the said second functional group reacts with a terminal unit of the polysialic acid to form a covalent bond therewith and form a capable of reaction with a sulfhydryl group functional polysialic acid.
  • the second functional group is a nucleophilic group, preferably hydrazine.
  • the polysialic acid comprises an aldehyde group in the terminal unit whose carbonyl group is attacked by the nucleophilic group.
  • the second functional group is electrophilic such as an N-alkoxyl carbonyl-imide such as N- hydroxysuccinimide ester or sulphosuccinimide ester, or carbodiimide.
  • the terminal group in such cases is preferably nucleophilic such as amine.
  • the reagent comprises a bifunctional organic group linking the first and second functional groups.
  • the bifunctional organic group is selected from a C 2 .
  • the process involves a subsequent step in which the functional polysialic acid is reacted with a polypeptide or a protein having at least one free and unprotected Cys unit whereby the functional group forms a thioether linkage with the thiol group of a Cys unit to form a polysialyated polypeptide or protein.
  • the process is particularly suitable where the protein contains a single Cys unit, whereby site-controlled derivatisation is achieved.
  • the invention is illustrated in the accompanying examples.
  • cysteine is reacted with the maleimide on the polymer preventing further reaction with Ellman's Reagent (5,5'-dithiobis(2- nitrobenzoic acid)) which contains a disulphide which forms an intense yellow colour when it is substituted for a thiol not adjacent to an aromatic ring.
  • Ellman's Reagent 5,5'-dithiobis(2- nitrobenzoic acid)
  • maleimide content can be calculated by measuring the inhition of reaction between cysteine and Ellman's reagent assay.
  • Ovine FAb (anti Desipramine /Norityrptaline, 4mg, 7.2 x10 nol) was dissolved in 0.25 ml PBS + 10 mM EDTA to this was added 0.498 mg 2- iminothiolane (2-IT, Traut's reagent 50 mol equiv 3.6 x10 "6 mol) in 0.25 ml of the same buffer. The tube was wrapped in foil and left to incubate stirring end over end for 1 h at 37°C.
  • Thiolated Fab was purified from free 2-IT Traut's reagent by gel filtration (PD-10) and 0.5 ml fractions assayed for presence of protein (BCA assay) or thiol ( Ellman's assay). The first eluting peak containing both was pooled and protein and thiols quantified.
  • 1.5 Conjugation of Fab-thiol to CAM To thiolated FAb ( 3.6 mg, 6.6x10 "8 mol) in 2 ml PBS/EDTA 22.5 mg CAM was added (9 x10 "7 mol, 15 molar equiv). The tube was sealed and allowed to incubate at 37°C for 1 h whilst gently mixing.
  • Figure 1 shows an SDS-PAGE gel of triplicate samples and relevant controls 1.6 Conclusion The results show that in all control wells (samples of thiolated FAb) the migration of the sample is similar to that for fresh FAb (below that of the 50 kDa marker) indicating no cross linking of FAb molecules during the process of conjugation. In the replicate lanes there is considerable band broadening with maximum intensity between the 98 and 250 KDa markers which typically indicates an increase in mass which is indicative of a of polysialylated product.
  • Step 1 amination of oxidised CA Oxidised colominic acid at (CAO) 10-100 mg/ml was dissolved in 2 ml of deionised water with a 300-fold molar excess of NH 4 CI, in a 50 ml tube 5 and then NaCNBH 3 (5 M stock in 1 N NaOH(aq) was added at a final concentration of 5 mg/ml. The mixture was incubated at room temperature for 5 days. A control reaction was also set up with colominic acid (A) instead of CAO. Product colominic acid amine derivative was precipitated by the addition of 5 ml ice-cold ethanol.
  • the precipitate was recovered by0 centrifugation at 4000 rpm, 30 minutes, room temperature in a benchtop centrifuge. The pellet was retained and resuspended in 2 ml of deionised water, then precipitated again with 5 ml of ice-cold ethanol in a 10 ml ultracentrifuge tube. The precipitate was collected by centrifugation at 30000 rpm for 30 minutes at room temperature. The pellet was again resuspended5 in 2 ml of deionised water and freeze-dried. 2.2.
  • TNBS picrylsulphonic acid or 2, 4, 6-tri-nitro-benzene sulphonic Acid
  • TNBS picrylsulphonic acid or 2, 4, 6-tri-nitro-benzene sulphonic Acid
  • Example 2.1 dissolved in 1 ml of PBS pH 7.4 was added 5 mg of N-succinimidyl iodoacetate (SIA). The mixture was left to react for 1 h at 37°C, after which excess SIA was removed by gel filtration over a 5ml HightrapTM Desalting column(AP Bioscience) eluted with PBS. 0.5 ml fractions were collected from the column and samples from each fraction tested for colominic acid content (resorcinol assay) and reactivity with cysteine indicating Iodide (Ellman's Assay). Fractions positive for both iodide and CA were pooled.
  • 3.2 Conjugation of CAI to ⁇ -galactosidase To E.coli ⁇ -galactosidase ( 5.0 mg, 4.3 x10 "8 mol) in 1 ml PBS 15 mg
  • CAI was added (6.59 x10-7 mol, 15 molar equiv). The tube was sealed wrapped in foil and allowed to incubate at room temperature for 1 h whilst gently mixing. The resulting conjugate was analysed by SDS page and then purified according to accepted protocols to remove free CAI. Samples were assayed for polymer and protein content as outlined elsewhere. Control reactions were carried out with CA as a negative control.
  • 25:1 linker to CA Colominic acid (CA; 22.7K; 73 mg for MBPH, tube A; 99.3 mg for PDPH; tube B and 76.6 mg for BMPH; tube C) was dissolved individually into 800 ml 0.1 M sodium acetate (pH 5.5) separately.
  • To tube A 25 mg of MBPH (dissolved in 200 ⁇ l of DMSO), to tube B 25 mg of PDPH (dissolved in 200 ⁇ l of DMSO) and to vial C 25 mg of BMPH (dissolved in 200 ⁇ l of sodium acetate buffer) were added. The pH of the reaction mixture was adjusted to 5.5. These mixture was then vortex mixed, wrapped in foil and allowed to incubate at 37°C for 72h on a orbital mixer.
  • the MBPH and PDPH respectively on the non-reducing (highly reactive ) end were 41.5, 32.5 and 48.3 mol % for MBPH, PDPH and BMPH respectively on the reducing end (weakly reactive).
  • the values on the reducing end are average of two values in each case.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • Genetics & Genomics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • Analytical Chemistry (AREA)
  • Polymers & Plastics (AREA)
  • Materials Engineering (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • General Chemical & Material Sciences (AREA)
  • General Engineering & Computer Science (AREA)
  • Microbiology (AREA)
  • Biotechnology (AREA)
  • Biomedical Technology (AREA)
  • Immunology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicinal Preparation (AREA)
  • Polysaccharides And Polysaccharide Derivatives (AREA)
  • Farming Of Fish And Shellfish (AREA)
  • Macromonomer-Based Addition Polymer (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

A polysialic acid compound is reacted with a hetero-­bifunctional reagent to introduce a pendant functional group for site­-specific conjugation to sulfhydryl groups, for instance side chains of cysteine units in drugs, drug delivery systems, proteins or peptides. The functional group is, for instance, an N-maleimide group.

Description

POLYSIALIC ACID DERIVATIVES The present invention relates to polysialic acid derivatives which are useful for conjugation to drugs, proteins and peptides, or to drug delivery systems such as liposomes, having sulfhydryl groups, as well as to conjugated products process for synthesising the derivatives and the conjugates and novel synthetic intermediates. The extended presence of drugs either within the vascular system or in extravascular use is often a pre-requisite for their optimal use. Many antibiotics and cytostatics for instance, as well as a variety of therapeutic peptides and proteins, and liposomes are removed from the circulation prematurely and before effective concentrations in target tissues can be achieved. The half lives of a number of short-lived proteins (for instance enzymes, cytokines, etc) have been augmented by conjugating these to low poly(ethylene glycol). It appears that PEG molecules prolong the circulation time of proteins and particles by forming a cloud around their surface, thus sterically hindering interaction with factors responsible for their clearance. However PEG is non-biodegradable and accumulation of PEGylated proteins intracellulary may be undesirable especially on chronic use [Bendele, A., Seely, J., Richey, C, Sennello, G., Shopp, G., Renal tubular vacuolation in animals treated with polyethylene-glycol conjugated proteins, Toxicological sciences, 42 (1998) 152-157; Convers, C. D. , Lejeune, L. , Shum, K. , Gilbert, C. , Shorr, R.G.L, Physiological effect of polyethylene glycol conjugation on stroma-free bovine hemoglobin in the conscious dog after partial exchange transfusion, Artificial organ, 21 (1997) 369-378]. We have described the conjugation of a polysaccharide comprising 2→8 and or 2→9 (e.g. alternating 2→8 and 2→9) linked sialic acid units conjugated to proteins to increase their half life, reduce their immunogenicity/antigenicity or increase the stability of a variety of proteins. In W092/22331 , polysialic acids are reacted with a model drug and shown to extend the half life in the circulation of mice. In Cell. Mol. Life Sci. 57 (2000) 1964 to 1969 and in Biotechnol. Genet. Eng. Rev. 16 (1999) 203 to 215, Gregoriadis et al describe the conjugation of polysialic acids to asparaginase and catalase, and show that the clearance rates from circulation reduced whilst enzyme activity was retained. We have also polysialylated insulin (Biochim. Biophys. Acta 1622 (2003) 42-49 and shown it to be active. We have also polysialylated interferon (AAPS Annual meeting 2002, Toronto, Canada, M1056). We have also polysialylated antibody fragment Fab (Epenetos, A. et al., Proceedings of ASCO (Clinical Pharmacy) 21 (2002) 2186). In all of these publications, polysialic acid is rendered reactive, by generating an aldehyde group at the non-reducing end by oxidation of the 7, 8-vicinaldiol moiety with sodium periodate. The aldehyde group was then reacted with primary amine groups on proteins, generally assumed to be epsilon-amino groups of lysine moieties of the protein or N-terminal amine groups. The reaction forms a Schiff base which is reduced by cyanoborohydride to a secondary amine. In WO-A-01/87922 we also suggest that derivatisation with other molecules could be carried out in the presence of denaturant to achieve increased levels of derivatisation. Examples of other derivatising agents are polyethylene glycol compounds. Activated PEG compounds such as tresyl- PEG and succinimidyl succinate ester of PEG were mentioned. The examples used succinimidyl succinate activated PEG, which is believed to react with amine groups. PEG derivatives having functional groups for coupling to thiol groups are commercially available. The functional groups may be maleimide, vinyl sulfone, iodoacetamide or orthopyridyl disulphide groups. Since these reagents react specifically with cysteines, and since proteins have fewer cysteines on their surfaces than lysine groups, the derivatisation is more controllable. Furthermore, in the absence of a free cysteine in a native protein, one or more free cysteines may be added by genetic engineering. The advantage of this approach is that it makes possible site-specific derivatisation at areas on the protein which will minimise a loss in biological activity. PEGylated proteins have been found to generate anti PEG antibodies that could also influence the residence time of the conjugate in the blood circulation (Cheng T, Wu, M., Wu,P., Chern, J, Roffer, SR., Accelerated clearance of polyethylene glycol modified proteins by anti-polyethylene glycol IgM. Bioconjugate chemistry, 10 (1999) 520-528. Despite, the established history of PEG as a parenterally administered polymer conjugated to therapeutics, a better understanding of its immunotoxicology, pharmacology and metabolism will be required (Hunter, A. C, Moghimi, S. M., Therapeutic synthetic polymers: a game of Russian Roulette. Drug Discovery Today, 7 (2002) 998-1001 ; Brocchini, S., Polymers in medicine: a game of chess. Drug Discovery Today, 8, (2003) 111-112). It would be useful for modification by polysialic acid to be targeted towards thiol (sulfhydryl) groups. It would also be desirable for the efficiency of derivatisation by sialic acid to be increased, the processes described in our prior art mentioned above requiring high excesses of active polysialic acid. It would also be desirable to avoid the use of cyano borohydride. According to the present invention there is provided a novel compound comprising a polysialic acid having a moiety linked at one or each terminal unit which includes a functional group selected from N-maleimido groups vinysulphone groups, N-iodoacetamide groups and orthopyridyl disulphide groups. The terminal unit to which the moiety is linked may be at the non- reducing end of the polysialic acid or at the reducing end of the polysialic acid. Generally the terminal sialic acid unit has been subjected to a preliminary chemical reaction to generate useful functional groups to which a maleimide-group containing reagent may be linked. We have found it convenient to use the chemistry disclosed in our earlier publications in which an aldehyde group is generated, as a preliminary step to generate the functional group via which the maleimide moiety may be linked. In our earlier publications mentioned above, it is the non-reducing terminal unit which is converted into an aldehyde moiety by oxidation of the 7, 8-vicinal diol moiety with sodium periodate to form the carbon 7-aldehyde compound. This is an appropriate reaction for the present invention. As an alternative, it is possible to provide the aldehyde moiety at the reducing terminal unit. In this case, it is preferred (but is not essential) to carry out a preliminary step in which the non-reducing end is deactivated, by preliminary oxidation and reduction steps. A first reduction step converts the ketal unit at the reducing end into a reduced ring opened form, having vicinal diols. The vicinal diols are subsequently oxidised using sodium periodate to form an aldehyde moiety at the carbon atom previously forming the 7-carbon of the reducing terminal unit. Where the non-reducing terminal glycosyl (usually sialic acid) is not deactivated by a preliminary oxidation step, the terminal unit will be simultaneously activated. In the invention, the moiety which includes the functional group, may be linked directly to the polysialic acid unit or, more conveniently, may be linked via a difunctional organic group, such as an alkane diyl group, an arylene group or an oligo(alkyleneoxy) alkane group, or alternatively an oligo peptidyl group. The linkage to the polysialic acid (from the linker or the moiety including the functional group may be a secondary amine, a hydrazone, an alkyl hydrazine an ester or a peptidyl group. The moiety may be generated by reaction of a polysialic acid substrate with a heterobifunctional reagent. The process form a further aspect of the invention. Reagents useful for introducing the selected functional groups are commercially available. A compound which will introduce a maleimide group onto an amine moiety without introducing any additional linker moiety is N- methoxy-carbonyl-maleimide. Generally the reagents include a second functional group for reaction with a group on the polysialic acid which may be the aldehyde group described above, or an amine group. Suitable second functional groups include N-hydroxy succinimide esters and their sulfosuccimide analogues and hydrazides. Preferably the compound is a N- maleimido-alkanoic acid hydrazide or an N-maleimidoarylalkanoic acid hydrazide i.e. a compound having the general formula X-R-Y in which: X is a N-maleimido, N-iodoacetamido, S-vinylsulphonyl or S- orthopyridyldisulphide group, R is alkane-diyl, arylene or aralkylene alkarylene, alkylene- oxaalkylene, or alkylene-oligooxa-alkylene or alkyl-oligopeptidyl-alkyl group; and Y is a hydrazide, amine or N-hydroxysuccinimide group.
Preferably R is C,_6 alkanediyl, C2.3-alkyl-oxa-C2.3-alkylene, C2.3 alkyl-oligo(oxa-C2.3 alkylene), or C2.6 alkylene phenyl. Preferably X is N-maleimido or orthopyridyldisulphide. Preferably Y is a hydrazide or a hydroxyl succinimide Compounds which may be reacted with an aldehyde group, and which include a linker moiety and introduce a maleimide group include N-[β- maleimidopropionic acid] hydrazide and 4-(4-N-maleimidophenyl)butyric acid hydrazide. The hydrazide group reacts with the aldehyde to form a stable hydrazone group. A suitable heterobifunctional compound which includes an oligo(ethyleneoxy) ethylene group is a compound comprising a polyethylene glycol (poly(ethyelenoxy)) group with, at one end, N-hydroxy succinimide (NHS) group and at the other end the functional group. The NHS group reacts with amine groups to form stable amide linkages. Heterobifunctional polyethyleneglycols with NHS at one end and either vinylsulphone or maleimide at the other end are available. Other examples of heterobifunctional reagents include, 3-(2-pyridyldithio)propionyl hydrazide, N-succinimidyl-3-[2-pyridyldithio]propionate, succinimidyl-H-[N- maleimidomethyl]cyclohexane-1-carboxylate), m-maleimidobenzoyl-N- hydroxysuccinimide ester, N-succinimidyl-[4-iodoacetyl] amino benzoate, N- [gamma-maleimidobutyryloxyjsuccinimide ester, N-[epsilon- maleimidocaproyloxy]succinimide ester and N-succinimidyl iodoacetate. Other reagents are available from Pierce Biotechnology and Shearwater Corporation (polyethylene glycol-based). Sialic acids (also known as nonulosonic acids) are members of a family of amino containing sugars containing 9 or more carbon atoms. The most important of the sialic acids is N-acetylneuraminic acid (also known as 5-(acetylamino)-3,5-dideoxy-D-glycero-D-galacto-nonulosonic, lactaminic acid and O-sialic acid) which has the formula:
Figure imgf000007_0001
Polysialic acids may be linked 2-8 and/or 2-9, usually in the α- configuration. In some polysialic acids the linkages are alternating 2→8 and 2→9. The invention is also of utility for heteropolymeric polysaccharides comprising glycosyl units other than sialic acid units. Polysialic acids are generally found to be non-toxic and substantially non-immunogenic. Furthermore the biodegration units, sialic acid, is not known to be toxic and, indeed sialic acids are widely found in animal proteins and cells, including blood cells and circulating proteins. Polysaccharide compounds containing many sialic acid units are polysaccharides produced by Esche chia coli, Moraxella nonliquifaciens, Pasteurella aeroginosis and Neisseria meningitidis or derivatives thereof. For instance colominic acid derived (by hydrolysis to shorten the chain lengths) from E. coli Kl comprises α 2-8 linked sialic acid units. Polysaccharide from E. coli K92 strain comprises alternating 2-8 and 2-9 linked sialic acid units. Polysaccharide C of N. meningitidis group C has 2-9 linked sialic acid units. One group of polysaccharide compounds which has been found to be of particular utility in the invention is group B polysaccharides. These compounds are produced by N. meningitidis, M. nonliquifaciens, P. aeroginosis A2 and E. coli K1. These compounds comprise a polysaccharide component comprising sialic acid residues and a phospholipid component. The sialic acid residues are linked (2 - 8)-α, the naturally occurring polymer consisting of about 200 residues. Some of the glycolipid molecules, especially the high molecular weight compounds appear to have a covalently attached phospholipid at the reducing end of the polysaccharide component. It is preferable for the bacteria from which the polysaccharide compound is derived to be non-pathogenic for convenience during production. It is particularly suitable therefore for the polysaccharide to be derived from a non-pathogenic strain of E. coli such as E. coli K92 or, preferably, K1 which is non-immunogenic. E. coli K92 and K1 isolates are well-known and any such type of any such strains can be used as sources of suitable polysaccharide. Preferably the polysialic acid should have at least 2, preferably at least 5, more preferably at least 10, for instance more than 50 sialic acid units. According to the invention, there is also provided a conjugate of a protein and the novel activated polysaccharide. The novel compound comprises a protein with at least one cysteine unit and, linked through a thioether bond to the side chain of a cysteine unit, a polysialic acid through a moiety joined at one or both terminal unit of the polysialic acid. Where the polysialic acid derivative was a N-maleimido group the moiety will include a N-succinimidyl group, with the thioester linked at the α- carbon atom. Preferably the moiety also comprises a secondary amine, a hydrazone or an amide bond.
©II HC- C^T-fTS FTβ— M I I— r~s- m i II There is also provided in the invention a new process in which a polysialic acid is reacted with a heterobifunctional reagent having a first functional group for reaction with sulfhydryl groups and a second functional group different to the first group whereby the said second functional group reacts with a terminal unit of the polysialic acid to form a covalent bond therewith and form a capable of reaction with a sulfhydryl group functional polysialic acid. In one embodiment the second functional group is a nucleophilic group, preferably hydrazine. This is of particular utility where the polysialic acid comprises an aldehyde group in the terminal unit whose carbonyl group is attacked by the nucleophilic group. In another embodiment of the process the second functional group is electrophilic such as an N-alkoxyl carbonyl-imide such as N- hydroxysuccinimide ester or sulphosuccinimide ester, or carbodiimide. The terminal group in such cases is preferably nucleophilic such as amine. In the process it is preferred that the reagent comprises a bifunctional organic group linking the first and second functional groups. Preferably the bifunctional organic group is selected from a C2.18-alkanediyl group, an arylene group, an oligo peptide and an oligo(alkoxy)alkyl group. Examples of suitable reagents are given above. Most usefully the process involves a subsequent step in which the functional polysialic acid is reacted with a polypeptide or a protein having at least one free and unprotected Cys unit whereby the functional group forms a thioether linkage with the thiol group of a Cys unit to form a polysialyated polypeptide or protein. The process is particularly suitable where the protein contains a single Cys unit, whereby site-controlled derivatisation is achieved. The invention is illustrated in the accompanying examples. Example 1
Figure imgf000010_0001
1.1 Synthesis Three separate preparations were carried out as follows: Colominic acid aldehyde (CAO) produced according to WO-A-9222331
(100 mg, 4.4 x10"6 mol) was dissolved into 500 μl 0.1 M sodium acetate, to this 5 molar equivalents of Λ/-[β-Maleimidopropionic acid] hydrazide ( 6.5 mg, 2.2 x10"5 mol) was added. This mixture was then vortex mixed and wrapped in foil and allowed to incubate at 37°C for 2h on a rotary mixer. The polymer was then precipitated by the addition of 2 volumes ( 1.0 ml) of ethanol. The precipitate was collected by centrifugation ( 13,000 rpm 2 min) in a bench top microcentrifuge. The supernatant was discarded and the pellet dissolved in 500 μml 0.1 M acetate. This process was repeated a further 2 times and the final pellet dissolved in deionised water and freeze dried overnight. 1.2 Assay for Maleimide Content In this assay cysteine is reacted with the maleimide on the polymer preventing further reaction with Ellman's Reagent (5,5'-dithiobis(2- nitrobenzoic acid)) which contains a disulphide which forms an intense yellow colour when it is substituted for a thiol not adjacent to an aromatic ring. Thus maleimide content can be calculated by measuring the inhition of reaction between cysteine and Ellman's reagent assay. First an initial stock of cysteine at 12 x10"3 M (0.145 mg/ml) was prepared in PBS. In a clean microtitre plate 100 μl volume doubling dilutions from 12 x10"3M to 0.375 x10"3 M were made from row B to row H. In row A 100 μl PBS was used as a zero standard. Samples of CA and CA-maleimide (CAM) were prepared at 5 or 10 mg/ml and 100 ul of each sample added to duplicate columns of the cysteine dilutions. In one set 100ul PBS without any CA was added as a control. The plate was covered and allowed to incubate at 37°C for 1 h. After this time 20 μl of Ellman's reagent (4 mg/ml in PBS) was added to each well and the plate incubated in the dark at room temperature for 15 min. Absorbance was the measured in wells at 405 nm. Standard curves were then plotted for the samples and the amount of maleimide present calculated from inhibition of the signal. 1.4 Thiolation of FAb and conjugation to CAM In the first step a thiol group is introduced into a model protein by thiolation of an amine of lysine. Ovine FAb (anti Desipramine /Norityrptaline, 4mg, 7.2 x10 nol) was dissolved in 0.25 ml PBS + 10 mM EDTA to this was added 0.498 mg 2- iminothiolane (2-IT, Traut's reagent 50 mol equiv 3.6 x10"6 mol) in 0.25 ml of the same buffer. The tube was wrapped in foil and left to incubate stirring end over end for 1 h at 37°C.
Figure imgf000012_0001
Thiolated Fab was purified from free 2-IT Traut's reagent by gel filtration ( PD-10) and 0.5 ml fractions assayed for presence of protein (BCA assay) or thiol ( Ellman's assay). The first eluting peak containing both was pooled and protein and thiols quantified. 1.5 Conjugation of Fab-thiol to CAM To thiolated FAb ( 3.6 mg, 6.6x10"8 mol) in 2 ml PBS/EDTA 22.5 mg CAM was added (9 x10"7 mol, 15 molar equiv). The tube was sealed and allowed to incubate at 37°C for 1 h whilst gently mixing. The resulting conjugate was then purified according to accepted protocols to remove free CAM. Both CA and protein content were assayed on the conjugate to calculate conjugation ratio. Control reactions were carried out with CA as a negative control. Several batches of CAM-Fab were prepared with various degrees of thiolation but maintaining the 15:1 ratio of CAM: Fab. Results are shown in table 1 below: Table 1
Figure imgf000013_0001
Figure 1 shows an SDS-PAGE gel of triplicate samples and relevant controls 1.6 Conclusion The results show that in all control wells (samples of thiolated FAb) the migration of the sample is similar to that for fresh FAb (below that of the 50 kDa marker) indicating no cross linking of FAb molecules during the process of conjugation. In the replicate lanes there is considerable band broadening with maximum intensity between the 98 and 250 KDa markers which typically indicates an increase in mass which is indicative of a of polysialylated product. 1.7 Conjugation of CAM to beta galactosidase To E.coli β-galactosidase (β-gal) 5.0 mg, 4.3 x10"8 mol) in 1 ml PBS 15 mg CAM was added (6.59 x10-7 mol, 15 molar equiv). The tube was sealed wrapped in foil and allowed to incubate at room temperature for 1 h whilst gently mixing. The resulting conjugate was analysed by SDS page and then purified according to accepted protocols to remove free CAM. Samples were assayed for polymer and protein content as outlined elsewhere. Control reactions were carried out with CA as a negative control. 1.8 Assay for Enzyme Activity Standards from 60 μg/ml to 3.75 μg/ml of fresh β-galactosidase were prepared in PBS. Sample of CAM-β-gal were diluted to 60 μg/ml in the same buffer. Enzyme activity of the conjugates was measured as follows: In a microtitre plate, to 100 μl of sample or standard was added 100 μl of All-in-One β-gal substrate (Pierce). The plate was incubated at 37°C for 30 min and absorbance read at 405nm. A calibration curve was prepared from the standards and the activity of the samples calculated from the equation for the linear regression of the curve. 1.9 Results From the protein and polymer assays the conjugation ratio was determined to be 1.23 CAM:1 β-Gal. There was also a corresponding increase in apparent molecular mass from the SDS page of the samples ( Fig 2). Enzyme activity in the purified sample was calculated to be 100.4 % compared to the free enzyme. Example 2 Synthesis Route 2 Step 1 amination of CA-aldehyde (CHO)
Figure imgf000014_0001
Step 2 introduction of maleimide ring
Figure imgf000014_0002
Synthesis 2.1. Step 1 amination of oxidised CA Oxidised colominic acid at (CAO) 10-100 mg/ml was dissolved in 2 ml of deionised water with a 300-fold molar excess of NH4CI, in a 50 ml tube 5 and then NaCNBH3 (5 M stock in 1 N NaOH(aq) was added at a final concentration of 5 mg/ml. The mixture was incubated at room temperature for 5 days. A control reaction was also set up with colominic acid (A) instead of CAO. Product colominic acid amine derivative was precipitated by the addition of 5 ml ice-cold ethanol. The precipitate was recovered by0 centrifugation at 4000 rpm, 30 minutes, room temperature in a benchtop centrifuge. The pellet was retained and resuspended in 2 ml of deionised water, then precipitated again with 5 ml of ice-cold ethanol in a 10 ml ultracentrifuge tube. The precipitate was collected by centrifugation at 30000 rpm for 30 minutes at room temperature. The pellet was again resuspended5 in 2 ml of deionised water and freeze-dried. 2.2. Assay for amine content The TNBS (picrylsulphonic acid or 2, 4, 6-tri-nitro-benzene sulphonic Acid) assay was used to determine the amount of amino groups present in the product.0 In the well of a microtitre plate TNBS (0.5 μl of 15 mM TNBS) was added to 90 μl of 0.1 M borate buffer pH 9.5. To this was added 10 μl of a 50 mg/ml solution of CA-amide the plate was allowed to stand for 20 minutes at room temperature, before reading the absorbance at 405nm. Glycine was used as a standard, at a concentration range of 0.1 to 1 mM. TNBS5 trinitrophenylates primary amine groups. The TNP adduct of the amine is detected. Testing the product purified with a double cold-ethanol precipitation using the TNBS assay showed close to 90 % conversion. 2.3. Maleimidation of CA-amine o CA-Amine (17 mg) was dissolved in 1 ml deionised water, to this was added 6 mg methoxy-carbonyl-maleimide (MCM). The mixture was left to react at room temperature for 30 min. To the sample 1 100 μl water and 200 μl acetonitrile was added and then incubated at room temperature for 4 h, after which 300 μl CHCI3 was added the tube shaken and the aqueous fraction collected. Then the fraction was purified over a PD-10 column to remove small molecules. The eluate was freeze dried and assayed for maleimide content. The molar concentration of maleimido was 44 mol %. Example 3: Preparation of lodoacetate derivative of colominic acid (CAI).
Figure imgf000016_0001
3.1 Synthesis To 40 mg colominic acid amine (85 mol % amine) as (described in
Example 2.1 ) dissolved in 1 ml of PBS pH 7.4 was added 5 mg of N-succinimidyl iodoacetate (SIA). The mixture was left to react for 1 h at 37°C, after which excess SIA was removed by gel filtration over a 5ml Hightrap™ Desalting column(AP Bioscience) eluted with PBS. 0.5 ml fractions were collected from the column and samples from each fraction tested for colominic acid content (resorcinol assay) and reactivity with cysteine indicating Iodide (Ellman's Assay). Fractions positive for both iodide and CA were pooled. 3.2 Conjugation of CAI to β-galactosidase To E.coli β-galactosidase ( 5.0 mg, 4.3 x10"8 mol) in 1 ml PBS 15 mg
CAI was added (6.59 x10-7 mol, 15 molar equiv). The tube was sealed wrapped in foil and allowed to incubate at room temperature for 1 h whilst gently mixing. The resulting conjugate was analysed by SDS page and then purified according to accepted protocols to remove free CAI. Samples were assayed for polymer and protein content as outlined elsewhere. Control reactions were carried out with CA as a negative control.
All samples were analysed for β-gal activity as example 1.8 above. 3.3 Conclusions Fractions 3-6 were positive for both polymer and iodoacetate and were pooled. The SDS page ( 4-12 % Bis/Tris gel; Figure 2) showed an increase in apparent molecular mass for samples incubated with the iodoacetamide derivative but not with control polymer. From the protein and polymer assays the conjugation ratio was determined to be 1.63 CAM β-gal. β-gal activity was calculated to be 100. 9 % for the conjugated sample, compared to the free enzyme. Example 4
4.1 Synthesis: Non-reducing end activation with 4(4-N- maleimidophenyl) butyric acid hydrazide (CA-MBPH) and 3-(2- pyridyldithio) propionyl hydrazide (CA-PDPH) The preparations were made as follows: Colominic acid aldehyde (CAO; 22.7kDa, Camida, Ireland) produced according to WO-A-9222331 (73 mg for MBPH, tube A; 99.3 mg for PDPH; tube B) was dissolved individually into 800 ml 0.1 M sodium acetate (pH 5.5). To tube A 15 mg of MBPH (15: 1 linker : CA ratio) dissolved in 200μl of DMSO was added. To tube B 15 mg (15: 1 linker : CA ratio) of PDPH dissolved in 200μl of DMSO was added. The pH was adjusted to 5.5 in each case for each reaction vessel. These mixtures were then vortex mixed and wrapped in foil and allowed to incubate at 37°C for 2h on a orbital mixer. Each polymer solution was purified by gel filtration (PD 10 column) eluting with PBS pH 7.4 and the 1 ml fraction containing CA ( by resorcinol assay) collected. These sample were freeze dried overnight and assayed for maleimide content as described in example 1.2. 4.2 Synthesis: Reducing end activation with 4(4-N- maleimidophenyl)butyric acid hydrazide (MBPH-CA), (2- pyridyldithio)propionyl hydrazide (PDPH-CA) and N-β- Maleimidopropionic acid hydrazide) hydrazide (BMPH-CA) The preparations were carried out as follows all at molar ratios of
25:1 linker to CA: Colominic acid (CA; 22.7K; 73 mg for MBPH, tube A; 99.3 mg for PDPH; tube B and 76.6 mg for BMPH; tube C) was dissolved individually into 800 ml 0.1 M sodium acetate (pH 5.5) separately. To tube A, 25 mg of MBPH (dissolved in 200μl of DMSO), to tube B 25 mg of PDPH (dissolved in 200μl of DMSO) and to vial C 25 mg of BMPH (dissolved in 200μl of sodium acetate buffer) were added. The pH of the reaction mixture was adjusted to 5.5. These mixture was then vortex mixed, wrapped in foil and allowed to incubate at 37°C for 72h on a orbital mixer. Each polymer solution was purified by gel filtration (PD 10 column) eluting with PBS pH 7.4 and the 1 ml fraction containing CA ( by resorcinol assay) collected. These sample were freeze dried overnight and assayed for maleimide content as described in example 1.2. 4.3 Results The molar concentration of maleimido was 49.0 and 35.0 mol % for
MBPH and PDPH respectively on the non-reducing (highly reactive ) end. The molar concentration of maleimido was 41.5, 32.5 and 48.3 mol % for MBPH, PDPH and BMPH respectively on the reducing end (weakly reactive). The values on the reducing end are average of two values in each case. 4.4 Conjugation of β-galactosidase (β-gal) to Maleimide activated colominic acids (reducing end and non-reducing end) To β-gal (1.0 mg; 1 ml in PBS/EDTA) in separate tubes, a 15 molar excess of each maleimide activated CA (MBPH, PDPH and BMPH on non-reducing or reducing end, from above examples) was added separately. Each tube was sealed and allowed to incubate at 37°C for 1 h whilst gently mixing. The resulting conjugate was then purified according to accepted protocols to remove free activated polymer. All samples were analysed by SDS PAGE and for β-gal activity as in example 1.8 4.5 Results The results (figure 3) show that in all control well (with β-gal) the migration of the sample is similar to that for fresh β-gal. In the conjugate lanes there is considerable band broadening with maximum intensity between the 98 and 250 kDa markers which typically indicates an increase in mass which is indicative of a of polysialylated-β-gal. β-gal activity was calculated to be 91.0 - 106 % for the conjugated samples.

Claims

CLAIMS 1. A compound comprising a polysaccharide acid a pendant moiety linked at least one terminal unit derived from a sialic acid unit which includes a functional group selected from N-maleimide groups,
5 vinylsulphone groups, N-iodoacetamide groups orthopyridyl disulphide groups. 2. A compound according to claim 1 in which the pendant moiety is linked at the reducing terminal unit of the polysaccharide. 3. A compound according to claim 1 or claim 2 in which the0 moiety is linked at the non-reducing terminal unit of the polysaccharide. 4. A compound according to any preceding claim in which the moiety comprises an alkanediyl group and/or an arylene group and a linkage optionally in combination with a oxalkylene or oligoaxa-alkylene group which is a secondary amine linkage, a hydrazone, an alkyl hydrazide linkage or a5 peptide linkage. 5. A compound according to any preceding claim in which the functional group is N-maleimido. 6. A compound according to any preceding claim in which the polysaccharide is a polysialic acid, preferably consisting substantially only of 0 sialic acid units 7. A compound which the compound has the formula
Figure imgf000020_0001
in which one of the following groups of definitions apply: i) R1 is H or -CHOHCH2OH, R2 is OH and R3 is either o -CH2CHR4R5 or -CH(CH2OH)CHR4R5 in which R4 and R5 together represent =N-NR6 or R4 is H and R5 is -NR6R7 in which R6 is an organic group comprising the said functional group or is H, and R7 is H or R6 and R7 together are a 1 ,3-but-2-enedioyl group; ii) R1 and R2 together represent =N-NR6or R1 is H and R2 is -NR6R7 in which R6 is an organic group comprising the said functional group or is H, and R7 is H or R6 and R7 together are a 1 ,3-but-2-enedioyl group; Gly-0 is a glycosyl (saccharide) group; n is 0 or more; and Ac is acetyl. 8. A compound according to claim 7 in which each Gly is a sialic acid unit. 9. A compound comprising a protein with at least one free cysteine unit and, linked through a thioester bond to the side chain of the cysteine unit, with a polysialic acid, through a moiety joined at one or each terminal units of the polysialic acid. 10. A compound according to any preceding claim in which the polysaccharide has at least 2, preferably at least 10, more preferably at least 50 saccharide units, preferably sialic acid units 2,8 and/or 2,9 linked to one another. 11. A process in which a polysaccharide comprising at least one terminal unit which is derived from a sialic acid unit is reacted with a heterobifunctional reagent having a first functional group selected from N- maleimido groups, vinylsulphone groups, N-iodoacetamide groups orthopyridyl disulphide groups and a second functional group different from the first group whereby the said second functional group reacts with a terminal sialic acid derivative unit to form a covalent bond therewith and form a functional polysaccharide suitable for selective conjugation to a thiol group. 12. A process according to claim 11 in which the said second functional group is a nucleophilic group, preferably hydrazine.
13. A process according to claim 11 in which the terminal unit of the polysaccharide has a carbonyl group which reacts with said nucleophilic group. 14. A process according to claim 11 in which the said second functional group is an electrophilic group, preferably an N-alkoxycarbonyl- imide or carbodiimide moiety. 15. A process according to claim 14 in which the terminal unit of the polysaccharide has an amine group which reacts with said electrophilic group, preferably to form a peptide or a urethane linkage. 16. A process according to any of claims 11 to 15 in which the reagent comprises a bifunctional organic group linking the first and second functional groups. 17. A process according to claim 16 in which the bifunctional organic group is selected from a C28-alkanediyl group, an arylene group, an oligo peptide and an oligo(alkoxy)alkyl group. 18. A process according to any of claims 11 to 17 in which the first functional group is a N-maleimide group. 19. A process according to claim 11 in which the reagent has the general formula X-R-Y in which: X is a N-maleimido, N-iodoacetamido, S-vinylsulphonyl or S- orthopyridyldisulphide group, R is alkane-diyl, arylene or aralkylene alkarylene, alkylene- oxaalkylene, or alkylene-oligooxa-alkylene or alkyl-oligopeptidyl-alkyl group; and Y is a hydrazide, amine or N-hydroxysuccinimide group. 20. A process according to any of claims 11 to 19 in which the polysaccharide acid has at least 2, preferably at least 10, more preferably at least 50, sialic acid units, preferably 2→8 and 2→9 linked to one another.
21. A process according to any one of claims 11 to 20 in which the maleimido-functional polysialic acid is reacted with a polypeptide or a protein having at least one free unprotected Cys unit whereby the maleimide group forms a thioether linkage with the thiol group of a Cys unit to form a polysialyated polypeptide or protein. 22. A process in which a compound according to any of claims 1 to 6 is reacted with a polypeptide or a protein having at least one free and unprotected Cys unit whereby the said functional group forms a thioether linage with the thiol group of a Cys unit to form a conjugate of the polysaccharide with the polypeptide or protein.
PCT/GB2004/003488 2003-08-12 2004-08-12 Polysialic acid derivatives WO2005016973A1 (en)

Priority Applications (19)

Application Number Priority Date Filing Date Title
JP2006523054A JP2007501888A (en) 2003-08-12 2004-08-12 Polysialic acid derivatives
KR1020067002875A KR101270692B1 (en) 2003-08-12 2004-08-12 Polysialic acid derivatives
DE602004009314T DE602004009314T2 (en) 2003-08-12 2004-08-12 POLYSIALINSÄUREDERIVATE
EP04768054A EP1654289B1 (en) 2003-08-12 2004-08-12 Polysialic acid derivatives
US10/568,111 US7691826B2 (en) 2003-08-12 2004-08-12 Polysialic acid derivatives
EP16176511.0A EP3184551A1 (en) 2004-08-12 2005-08-12 Sialic acid derivatives
JP2007525356A JP5197009B2 (en) 2004-08-12 2005-08-12 Sialic acid derivatives
PCT/GB2005/003160 WO2006016168A2 (en) 2004-08-12 2005-08-12 Sialic acid derivatives
US11/660,128 US7875708B2 (en) 2004-08-12 2005-08-12 Sialic acid derivatives
CNA2005800345881A CN101039965A (en) 2004-08-12 2005-08-12 Sialic acid derivatives
ES05794259.1T ES2593318T3 (en) 2004-08-12 2005-08-12 Sialic acid derivatives
EP05794259.1A EP1776389B1 (en) 2004-08-12 2005-08-12 Sialic acid derivatives
US12/717,073 US20100221808A1 (en) 2003-08-12 2010-03-03 Polysialic acid derivatives
US12/987,878 US8293888B2 (en) 2004-08-12 2011-01-10 Sialic acid derivatives
US13/647,326 US8765936B2 (en) 2004-08-12 2012-10-08 Sialic acid derivatives
US13/650,048 US9102714B2 (en) 2003-08-12 2012-10-11 Polysialic acid derivatives
US14/251,596 US9102699B2 (en) 2004-08-12 2014-04-12 Sialic acid derivatives
US14/791,468 US9920137B2 (en) 2003-08-12 2015-07-05 Polysialic acid derivatives
US14/791,467 US20150344591A1 (en) 2004-08-12 2015-07-05 Sialic Acid Derivatives

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
EP03254988.3 2003-08-12
EP03254988 2003-08-12
EP03255200 2003-08-21
EP03255200.2 2003-08-21

Related Child Applications (4)

Application Number Title Priority Date Filing Date
US10568111 A-371-Of-International 2004-08-12
US10/568,111 A-371-Of-International US7691826B2 (en) 2003-08-12 2004-08-12 Polysialic acid derivatives
US12/707,073 Division US8262902B2 (en) 2009-02-19 2010-02-17 Magnetic removal of material from a mixture based on sulfided diluent in the mixture
US12/717,073 Division US20100221808A1 (en) 2003-08-12 2010-03-03 Polysialic acid derivatives

Publications (1)

Publication Number Publication Date
WO2005016973A1 true WO2005016973A1 (en) 2005-02-24

Family

ID=34196160

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/GB2004/003488 WO2005016973A1 (en) 2003-08-12 2004-08-12 Polysialic acid derivatives

Country Status (9)

Country Link
US (4) US7691826B2 (en)
EP (1) EP1654289B1 (en)
JP (1) JP2007501888A (en)
KR (1) KR101270692B1 (en)
AT (1) ATE374788T1 (en)
DE (1) DE602004009314T2 (en)
ES (1) ES2294535T3 (en)
RU (1) RU2327703C2 (en)
WO (1) WO2005016973A1 (en)

Cited By (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006090119A1 (en) * 2005-02-23 2006-08-31 Lipoxen Technologies Limited Activated sialic acid derivatives for protein derivatisation and conjugation
EP1877099A2 (en) * 2005-04-06 2008-01-16 Genzyme Corporation Peg and polysialic lysosomal enzyme conjugates via acid labile linkers for therapeutic targeting
WO2008012540A1 (en) * 2006-07-25 2008-01-31 Lipoxen Technologies Limited N-terminal polysialylation
JP2009531389A (en) * 2006-03-30 2009-09-03 グラクソスミスクライン バイオロジカルズ ソシエテ アノニム Joining method
WO2010011944A2 (en) 2008-07-25 2010-01-28 Wagner Richard W Protein screeing methods
WO2010033240A2 (en) * 2008-09-19 2010-03-25 Nektar Therapeutics Carbohydrate-based drug delivery polymers and conjugates thereof
US7875708B2 (en) 2004-08-12 2011-01-25 Lipoxen Technologies Limited Sialic acid derivatives
WO2011012850A2 (en) 2009-07-27 2011-02-03 Lipoxen Technologies Limited Glycopolysialylation of non-blood coagulation proteins
WO2011017055A2 (en) 2009-07-27 2011-02-10 Baxter International Inc. Blood coagulation protein conjugates
EP2532369A2 (en) 2006-12-15 2012-12-12 Baxter International Inc Factor VIIa-(poly)sialic acid conjugate having prolonged in vivo half-life
US8399657B2 (en) 2001-01-18 2013-03-19 Genzyme Corporation Methods for introducing mannose 6-phosphate and other oligosaccharides onto glycoproteins and its application thereof
US8652334B2 (en) 2004-08-12 2014-02-18 Lipoxen Technologies Limited Fractionation of charged polysaccharide
US8759501B2 (en) 2007-01-18 2014-06-24 Genzyme Corporation Oligosaccharides comprising an aminooxy group and conjugates thereof
US8835614B2 (en) 2008-12-16 2014-09-16 Genzyme Corporation Oligosaccharide-protein conjugates
EP2799088A2 (en) 2008-08-01 2014-11-05 Baxter International Inc Factor VIII polymer conjugates
US9474806B2 (en) 2008-07-21 2016-10-25 The Brigham And Women's Hospital, Inc. Methods and compositions relating to synthetic beta-1,6 glucosamine oligosaccharides
EP3093029A1 (en) 2009-07-27 2016-11-16 Baxalta GmbH Blood coagulation protein conjugates
WO2018197545A1 (en) 2017-04-25 2018-11-01 Lipoxen Technologies Limited Methods of treating multiple myeloma cancers expressing high levels of epo-receptor using psa-epo
WO2018197547A1 (en) 2017-04-25 2018-11-01 Lipoxen Technologies Limited Methods of treating diseases related to net formation with parenteral administration of polysialylated dnase i
US10188739B2 (en) 2014-02-27 2019-01-29 Xenetic Biosciences, Inc. Compositions and methods for administering insulin or insulin-like protein to the brain
ES2711669A1 (en) * 2017-11-02 2019-05-06 Univ Santiago Compostela SYSTEMS FOR THE RELEASE OF POLYSIOLOGIC ACID DRUGS AND METHODS (Machine-translation by Google Translate, not legally binding)
WO2020099513A1 (en) 2018-11-13 2020-05-22 Lipoxen Technologies Limited Glycopolysialylation of blinatumomab
US10919956B2 (en) 2002-11-12 2021-02-16 The Brigham And Women's Hospital, Inc. Polysaccharide vaccine for staphylococcal infections

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7691826B2 (en) * 2003-08-12 2010-04-06 Lipoxen Technologies Limited Polysialic acid derivatives
WO2005016974A1 (en) 2003-08-12 2005-02-24 Lipoxen Technologies Limited Sialic acid derivatives for protein derivatisation and conjugation
DK2173383T3 (en) * 2007-06-26 2017-09-25 Baxalta GmbH HYDROLYSIBLE POLYMERIC FMOC LINKER
TW201042257A (en) * 2009-05-26 2010-12-01 Baxter Int Detection of antibody that binds to water soluble polymer-modified polypeptides
US8809496B2 (en) 2009-09-03 2014-08-19 The Noguchi Institute Production method of 11-sugar sialylglycopeptide
RU2522846C2 (en) * 2009-11-30 2014-07-20 Российская Федерация, От Имени Которой Выступает Министерство Промышленности И Торговли Российской Федерации Drug preparation and method for improving rheological properties of sputum and pulmonary administration of this preparation
JP5285022B2 (en) * 2010-04-30 2013-09-11 旭化成株式会社 Glycopeptide derivative and method for producing the same
RU2559522C2 (en) * 2010-11-29 2015-08-10 Российская Федерация в лице Министерства промышленности и торговли Российской Федерации Inhalation drug form of polysialylated human doexyribonuclease i and method of obtaining thereof
JP5680576B2 (en) * 2011-03-03 2015-03-04 旭化成株式会社 Method for producing 11-saccharide sialyl-oligosaccharide asparagine

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SU675053A1 (en) * 1977-08-10 1979-07-25 Институт Белка Ан Ссср 9-iodacetamino-methyl-antthracene as luminescent label for proteins and protein-like polymers
US5329028A (en) * 1992-08-05 1994-07-12 Genentech, Inc. Carbohydrate-directed cross-linking reagents
US5965532A (en) * 1996-06-28 1999-10-12 Trustees Of Tufts College Multivalent compounds for crosslinking receptors and uses thereof
WO2001087922A2 (en) * 2000-05-16 2001-11-22 Lipoxen Technologies Limited Derivatisation of proteins in aqueous solution

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB9112212D0 (en) * 1991-06-06 1991-07-24 Gregoriadis Gregory Pharmaceutical compositions
KR100307289B1 (en) * 1993-01-22 2001-12-28 제임스 에스. 쿼크 Ganglioside-KLH Conjugate Vaccine Containing QS-21
US7691826B2 (en) * 2003-08-12 2010-04-06 Lipoxen Technologies Limited Polysialic acid derivatives
WO2006016168A2 (en) * 2004-08-12 2006-02-16 Lipoxen Technologies Limited Sialic acid derivatives
JP5096466B2 (en) * 2006-07-25 2012-12-12 リポクセン テクノロジーズ リミテッド N-terminal polysialylation

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SU675053A1 (en) * 1977-08-10 1979-07-25 Институт Белка Ан Ссср 9-iodacetamino-methyl-antthracene as luminescent label for proteins and protein-like polymers
US5329028A (en) * 1992-08-05 1994-07-12 Genentech, Inc. Carbohydrate-directed cross-linking reagents
US5965532A (en) * 1996-06-28 1999-10-12 Trustees Of Tufts College Multivalent compounds for crosslinking receptors and uses thereof
WO2001087922A2 (en) * 2000-05-16 2001-11-22 Lipoxen Technologies Limited Derivatisation of proteins in aqueous solution

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
DATABASE WPI Section Ch Week 198013, Derwent World Patents Index; Class A60, AN 1980-23224C, XP002267699 *
JAIN S ET AL: "Polysialylated insulin: synthesis, characterization and biological activity in vivo", BBA - GENERAL SUBJECTS, ELSEVIER SCIENCE PUBLISHERS, NL, vol. 1622, no. 1, 20 June 2003 (2003-06-20), pages 42 - 49, XP004433607, ISSN: 0304-4165 *
MACMILLAN D ET AL: "SELECTIVE IN VITRO GLYCOSYLATION OF RECOMBINANT PROTEINS: SEMI-SYNTHESIS OF NOVEL HOMOGENEOUS GLYCOFORMS OF HUMAN ERYTHROPOIETIN", CHEMISTRY AND BIOLOGY, CURRENT BIOLOGY, LONDON, GB, vol. 8, no. 2, 2001, pages 133 - 145, XP000998149, ISSN: 1074-5521 *
S. M. CHAMOW ET AL.: "Conjugation of Soluble CD4 without Loss of Biological Activity via a Novel Carbohydrate-directed Cross-linking Reagent", THE JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 267, no. 22, 5 August 1992 (1992-08-05), USA, pages 15916 - 15922, XP002267698 *

Cited By (94)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10363291B2 (en) 2001-01-18 2019-07-30 Genzyme Corporation Methods for introducing mannose 6-phosphate and other oligosaccharides onto glycoproteins and applications thereof
US8841427B2 (en) 2001-01-18 2014-09-23 Genzyme Corporation Methods for introducing mannose 6-phosphate and other oligosaccharides onto glycoproteins and applications thereof
US10973887B2 (en) 2001-01-18 2021-04-13 Genzyme Corporation Methods for introducing mannose 6-phosphate and other oligosaccharides onto glycoproteins and its application thereof
US8399657B2 (en) 2001-01-18 2013-03-19 Genzyme Corporation Methods for introducing mannose 6-phosphate and other oligosaccharides onto glycoproteins and its application thereof
US9687531B2 (en) 2001-01-18 2017-06-27 Genzyme Corporation Methods for introducing mannose 6 phosphate and other oligosaccharides onto glycoproteins and applications thereof
US10919956B2 (en) 2002-11-12 2021-02-16 The Brigham And Women's Hospital, Inc. Polysaccharide vaccine for staphylococcal infections
US20150344591A1 (en) * 2004-08-12 2015-12-03 Lipoxen Technologies Limited Sialic Acid Derivatives
US8293888B2 (en) 2004-08-12 2012-10-23 Lipoxen Technologies Limited Sialic acid derivatives
US9200052B2 (en) 2004-08-12 2015-12-01 Lipoxen Technologies Limited Fractionation of charged polysaccharide
US9828443B2 (en) 2004-08-12 2017-11-28 Lipoxen Technologies Limited Fractionation of charged polysaccharide
US7875708B2 (en) 2004-08-12 2011-01-25 Lipoxen Technologies Limited Sialic acid derivatives
US9790288B2 (en) 2004-08-12 2017-10-17 Lipoxen Technologies Limited Fractionation of charged polysaccharide
EP3184551A1 (en) 2004-08-12 2017-06-28 Lipoxen Technologies Limited Sialic acid derivatives
US20140309409A1 (en) * 2004-08-12 2014-10-16 Lipoxen Technologies Limited Sialic Acid Derivatives
US8652334B2 (en) 2004-08-12 2014-02-18 Lipoxen Technologies Limited Fractionation of charged polysaccharide
US9102699B2 (en) 2004-08-12 2015-08-11 Lipoxen Technologies Limited Sialic acid derivatives
US8952141B2 (en) 2005-02-23 2015-02-10 Lipoxen Technologies Incorporated Activated sialic acid derivatives for protein derivatisation and conjugation
US8735557B2 (en) * 2005-02-23 2014-05-27 Lipoxen Technologies Limited Activated sialic acid derivatives for protein derivatisation and conjugation
EP3138855A1 (en) 2005-02-23 2017-03-08 Lipoxen Technologies Limited Activated sialic acid derivatives for protein derivatisation and conjugation
US20150150997A1 (en) * 2005-02-23 2015-06-04 Lipoxen Technologies Limited Activated Sialic Acid Derivatives For Protein Derivatisation And Conjugation
US9216227B2 (en) 2005-02-23 2015-12-22 Lipoxen Technologies Limited Activated sialic acid derivatives for protein derivatisation and conjugation
US8217154B2 (en) 2005-02-23 2012-07-10 Lipoxen Technologies Limited Activated sialic acid derivatives for protein derivatisation and conjugation
US20130095548A1 (en) * 2005-02-23 2013-04-18 Sanjay Jain Activated sialic acid derivatives for protein derivatisation and conjugation
WO2006090119A1 (en) * 2005-02-23 2006-08-31 Lipoxen Technologies Limited Activated sialic acid derivatives for protein derivatisation and conjugation
US10155046B2 (en) 2005-02-23 2018-12-18 Lipoxen Technologies Limited Activated sialic acid derivatives for protein derivatisation and conjugation
US10155045B2 (en) 2005-02-23 2018-12-18 Lipoxen Technologies Limited Activated sialic acid derivatives for protein derivatisation and conjugation
US8124073B2 (en) 2005-04-06 2012-02-28 Genzyme Corporation Targeting of glycoprotein therapeutics
US9498518B2 (en) 2005-04-06 2016-11-22 Genzyme Corporation Targeting of glycoprotein therapeutics
US10792342B2 (en) 2005-04-06 2020-10-06 Genzyme Corporation Targeting of glycoprotein therapeutics
EP1877099B1 (en) * 2005-04-06 2012-09-19 Genzyme Corporation Therapeutic conjugates comprising a lysosomal enzyme, polysialic acid and a targeting moiety
US8906379B2 (en) 2005-04-06 2014-12-09 Genzyme Corporation Targeting of glycoprotein therapeutics
EP1877099A2 (en) * 2005-04-06 2008-01-16 Genzyme Corporation Peg and polysialic lysosomal enzyme conjugates via acid labile linkers for therapeutic targeting
JP2009531389A (en) * 2006-03-30 2009-09-03 グラクソスミスクライン バイオロジカルズ ソシエテ アノニム Joining method
US8933218B2 (en) 2006-03-30 2015-01-13 Glaxosmithkline Biologicals S.A. Conjugation process for PNAG and a carrier protein
JP2013121982A (en) * 2006-07-25 2013-06-20 Lipoxen Technologies Ltd N-terminal derivatisation of proteins with polysaccharides
US9579393B2 (en) 2006-07-25 2017-02-28 Lipoxen Technologies Limited Derivatisation of erythropoietin (EPO)
WO2008012540A1 (en) * 2006-07-25 2008-01-31 Lipoxen Technologies Limited N-terminal polysialylation
JP2009544681A (en) * 2006-07-25 2009-12-17 リポクセン テクノロジーズ リミテッド Polysaccharide derivatives of erythropoietin
US8933026B2 (en) 2006-07-25 2015-01-13 Lipoxen Technologies Limited Derivatisation of erythropoietin (EPO)
US8796207B2 (en) 2006-07-25 2014-08-05 Lipoxen Technologies Limited Derivatisation of erythropoietin (EPO)
US8946406B2 (en) 2006-07-25 2015-02-03 Lipoxen Technologies Limited Derivatisation of erythropoietin (EPO)
JP2009544680A (en) * 2006-07-25 2009-12-17 リポクセン テクノロジーズ リミテッド N-terminal derivatization of proteins with polysaccharides
US8981050B2 (en) 2006-07-25 2015-03-17 Lipoxen Technologies Limited N-terminal derivatisation of proteins with polysaccharides
US9040478B2 (en) 2006-07-25 2015-05-26 Lipoxen Technologies Limited Derivatisation of granulocyte colony-stimulating factor
JP2009544677A (en) * 2006-07-25 2009-12-17 リポクセン テクノロジーズ リミテッド Derivatization of granulocyte colony-stimulating factor
US10300144B2 (en) 2006-07-25 2019-05-28 Lipoxen Technologies Limited N-terminal polysialylation
JP2015145400A (en) * 2006-07-25 2015-08-13 リポクセン テクノロジーズ リミテッド N-terminal derivatization of proteins with polysaccharides
EP3299033A1 (en) 2006-07-25 2018-03-28 Lipoxen Technologies Limited N-terminal polysialylation
JP2013241446A (en) * 2006-07-25 2013-12-05 Lipoxen Technologies Ltd N-terminal derivatization of protein with polysaccharide
EP2630972A2 (en) 2006-07-25 2013-08-28 Lipoxen Technologies Limited N-terminal polysialylation
US8394921B2 (en) 2006-07-25 2013-03-12 Lipoxen Technologies Limited N-terminal derivatisation of proteins with polysaccharides
US9234020B2 (en) 2006-07-25 2016-01-12 Lipoxen Technologies Limited Derivatisation of erythropoietin (EPO)
US9266936B2 (en) 2006-07-25 2016-02-23 Lipoxen Technologies Limited Derivatisation of erythropoietin (EPO)
JP2016128493A (en) * 2006-07-25 2016-07-14 リポクセン テクノロジーズ リミテッド N-terminal derivatization of protein by polysaccharide
US9789163B2 (en) 2006-07-25 2017-10-17 Lipoxen Technologies Limited N-terminal derivatisation of proteins with polysaccharides
US8299026B2 (en) 2006-07-25 2012-10-30 Lipoxen Technologies Limited Derivatisation of erythropoietin (EPO)
US9474805B2 (en) 2006-07-25 2016-10-25 Lipoxen Technologies Limited Derivatisation of granulocyte colony-stimulating factor
US8299015B2 (en) 2006-07-25 2012-10-30 Lipoxen Technologies Limited Derivatisation of granulocyte colony-stimulating factor
US9549990B2 (en) 2006-07-25 2017-01-24 Lipoxen Technologies Limited Derivatisation of erythropoietin (EPO)
US9492556B2 (en) 2006-07-25 2016-11-15 Lipoxen Technologies Limited N-terminal derivatisation of proteins with polysaccharides
US9492557B2 (en) 2006-07-25 2016-11-15 Lipoxen Technologies Limited Derivatisation of granulocyte colony-stimulating factor
EP3323430A1 (en) 2006-12-15 2018-05-23 Baxalta GmbH Factor viia-(poly)sialic acid conjugate having prolonged in vivo half-life
EP2532369A2 (en) 2006-12-15 2012-12-12 Baxter International Inc Factor VIIa-(poly)sialic acid conjugate having prolonged in vivo half-life
US8637007B2 (en) 2006-12-15 2014-01-28 Baxter International Inc. Factor VIIa-polysialic acid conjugate having prolonged in vivo half-life
US8759501B2 (en) 2007-01-18 2014-06-24 Genzyme Corporation Oligosaccharides comprising an aminooxy group and conjugates thereof
US10907142B2 (en) 2007-01-18 2021-02-02 Genzyme Corporation Oligosaccharides comprising an aminooxy group and conjugates thereof
US9469850B2 (en) 2007-01-18 2016-10-18 Genzyme Corporation Oligosaccharides comprising an aminooxy group and conjugates thereof
US9474806B2 (en) 2008-07-21 2016-10-25 The Brigham And Women's Hospital, Inc. Methods and compositions relating to synthetic beta-1,6 glucosamine oligosaccharides
US11123416B2 (en) 2008-07-21 2021-09-21 The Brigham And Women's Hospital, Inc. Methods and compositions relating to synthetic beta-1,6 glucosamine oligosaccharides
US10034927B2 (en) 2008-07-21 2018-07-31 The Brigham And Women's Hospital, Inc. Methods and compositions relating to synthetic beta-1,6 glucosamine oligosaccharides
EP3629022A1 (en) 2008-07-25 2020-04-01 Richard W. Wagner Protein screening methods
WO2010011944A2 (en) 2008-07-25 2010-01-28 Wagner Richard W Protein screeing methods
EP2810662A1 (en) 2008-08-01 2014-12-10 Baxter International Inc Factor VIII polymer conjugates
EP2799088A2 (en) 2008-08-01 2014-11-05 Baxter International Inc Factor VIII polymer conjugates
US9173951B2 (en) 2008-09-19 2015-11-03 Nektar Therapeutics Carbohydrate-based drug delivery polymers and conjugates thereof
WO2010033240A3 (en) * 2008-09-19 2010-06-17 Nektar Therapeutics Carbohydrate-based drug delivery polymers and conjugates thereof
US8680263B2 (en) 2008-09-19 2014-03-25 Nektar Therapeutics Carbohydrate-based drug delivery polymers and conjugates thereof
WO2010033240A2 (en) * 2008-09-19 2010-03-25 Nektar Therapeutics Carbohydrate-based drug delivery polymers and conjugates thereof
US9682153B2 (en) 2008-09-19 2017-06-20 Nektar Therapeutics Polymer conjugates of therapeutic peptides
US8835614B2 (en) 2008-12-16 2014-09-16 Genzyme Corporation Oligosaccharide-protein conjugates
US11279725B2 (en) 2008-12-16 2022-03-22 Genzyme Corporation Oligosaccharide-protein conjugates
US10464962B2 (en) 2008-12-16 2019-11-05 Genzyme Corporation Oligosaccharide-protein conjugates
US9493498B2 (en) 2008-12-16 2016-11-15 Genzyme Corporation Oligosaccharide-protein conjugates
WO2011012850A2 (en) 2009-07-27 2011-02-03 Lipoxen Technologies Limited Glycopolysialylation of non-blood coagulation proteins
US10772968B2 (en) 2009-07-27 2020-09-15 Lipoxen Technologies Limited Glycopolysialylation of non-blood coagulation proteins
US9795683B2 (en) 2009-07-27 2017-10-24 Lipoxen Technologies Limited Glycopolysialylation of non-blood coagulation proteins
EP3093029A1 (en) 2009-07-27 2016-11-16 Baxalta GmbH Blood coagulation protein conjugates
WO2011017055A2 (en) 2009-07-27 2011-02-10 Baxter International Inc. Blood coagulation protein conjugates
EP3081233A1 (en) 2009-07-27 2016-10-19 Baxalta GmbH Glycopolysialylation of proteins other than blood coagulation proteins
US10188739B2 (en) 2014-02-27 2019-01-29 Xenetic Biosciences, Inc. Compositions and methods for administering insulin or insulin-like protein to the brain
WO2018197547A1 (en) 2017-04-25 2018-11-01 Lipoxen Technologies Limited Methods of treating diseases related to net formation with parenteral administration of polysialylated dnase i
WO2018197545A1 (en) 2017-04-25 2018-11-01 Lipoxen Technologies Limited Methods of treating multiple myeloma cancers expressing high levels of epo-receptor using psa-epo
ES2711669A1 (en) * 2017-11-02 2019-05-06 Univ Santiago Compostela SYSTEMS FOR THE RELEASE OF POLYSIOLOGIC ACID DRUGS AND METHODS (Machine-translation by Google Translate, not legally binding)
WO2020099513A1 (en) 2018-11-13 2020-05-22 Lipoxen Technologies Limited Glycopolysialylation of blinatumomab

Also Published As

Publication number Publication date
US9920137B2 (en) 2018-03-20
JP2007501888A (en) 2007-02-01
EP1654289A1 (en) 2006-05-10
US20060270830A1 (en) 2006-11-30
US20100221808A1 (en) 2010-09-02
RU2006107545A (en) 2007-09-27
RU2327703C2 (en) 2008-06-27
US20150307633A1 (en) 2015-10-29
ES2294535T3 (en) 2008-04-01
US7691826B2 (en) 2010-04-06
DE602004009314T2 (en) 2008-02-07
DE602004009314D1 (en) 2007-11-15
KR101270692B1 (en) 2013-06-03
EP1654289B1 (en) 2007-10-03
KR20060085329A (en) 2006-07-26
ATE374788T1 (en) 2007-10-15
US20130144043A1 (en) 2013-06-06
US9102714B2 (en) 2015-08-11

Similar Documents

Publication Publication Date Title
US9920137B2 (en) Polysialic acid derivatives
AU698944B2 (en) Conjugation-stabilized therapeutic agent compositions, delivery and diagnostic formulations
KR101113726B1 (en) Sialic acid derivatives for protein derivatisation and conjugation
JP5419689B2 (en) Derivatization of granulocyte colony-stimulating factor
US5283317A (en) Intermediates for conjugation of polypeptides with high molecular weight polyalkylene glycols
EP2889624B1 (en) Reversible covalent linkage of functional molecules
CA2478478C (en) Coupling hydroxyalkyl starch via a terminal aldehyde group or a functional group derived therefrom, to a protein
AU694919B2 (en) Conjugation-stabilized polypeptide compositions
US5006333A (en) Conjugates of superoxide dismutase coupled to high molecular weight polyalkylene glycols
EP0200467A2 (en) Superoxide dismutase combined with a poly(alkylene oxide)
MX2011000859A (en) Modified bovine g-csf polypeptides and their uses.
CA2394928A1 (en) Amphiphilic polymers and polypeptide conjugates comprising same
JPH07196925A (en) Reagent that forms peg hydrazone and peg oxime bonds and protein derivative thereof
JP2020037701A (en) Glycopolysialylation of non-blood coagulation protein
RU1776275C (en) Process for producing water-soluble conjugation of c@@@,-z@@@-dependent peroxide-dismutase
Ramírez-Andersen et al. Long-acting human growth hormone analogue by noncovalent albumin binding
EP0476408A1 (en) Chemical conjugation of morpholino anthracyclines to antibodies
EP1008355A1 (en) Method for identifying or analyzing polymer linkage sites on macromolecules using amino acid report binding
AU778790B2 (en) Polypeptides having a single covalently bound n-terminal water-soluble polymer
CN101385858A (en) Purified PEG human growth hormone conjugates and preparation thereof
AU2006200906A1 (en) Amphiphilic polymers and polypeptide conjugates comprising same

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GM KE LS MW MZ NA SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DPEN Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed from 20040101)
WWE Wipo information: entry into national phase

Ref document number: 2006523054

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 1020067002875

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 2004768054

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 903/DELNP/2006

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2006107545

Country of ref document: RU

WWP Wipo information: published in national office

Ref document number: 2004768054

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2006270830

Country of ref document: US

Ref document number: 10568111

Country of ref document: US

WWP Wipo information: published in national office

Ref document number: 1020067002875

Country of ref document: KR

WWP Wipo information: published in national office

Ref document number: 10568111

Country of ref document: US

WWG Wipo information: grant in national office

Ref document number: 2004768054

Country of ref document: EP