US20140315854A1 - Biotechnological sulphated chondroitin sulphate at position 4 or 6 on the same polysaccharide chain, and process for the preparation thereof - Google Patents

Biotechnological sulphated chondroitin sulphate at position 4 or 6 on the same polysaccharide chain, and process for the preparation thereof Download PDF

Info

Publication number
US20140315854A1
US20140315854A1 US14/115,184 US201214115184A US2014315854A1 US 20140315854 A1 US20140315854 A1 US 20140315854A1 US 201214115184 A US201214115184 A US 201214115184A US 2014315854 A1 US2014315854 A1 US 2014315854A1
Authority
US
United States
Prior art keywords
chondroitin
acid
chondroitin sulphate
sulphate
sodium salt
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
US14/115,184
Other languages
English (en)
Inventor
Davide Bianchi
Marco Valetti
Paola Bazza
Niccolo Miraglia
Ermanno Valoti
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.)
Gnosis SpA
Original Assignee
Gnosis SpA
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from IT000829A external-priority patent/ITMI20110829A1/it
Priority claimed from IT000136A external-priority patent/ITMI20120136A1/it
Application filed by Gnosis SpA filed Critical Gnosis SpA
Assigned to GNOSIS S.P.A. reassignment GNOSIS S.P.A. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: VALOTI, ERMANNO, MIRAGLIA, NICCOLO, BAZZA, Paola, BIANCHI, DAVIDE, VALETTI, MARCO
Publication of US20140315854A1 publication Critical patent/US20140315854A1/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
    • C08B37/00Preparation of polysaccharides not provided for in groups C08B1/00 - C08B35/00; Derivatives thereof
    • C08B37/006Heteroglycans, i.e. polysaccharides having more than one sugar residue in the main chain in either alternating or less regular sequence; Gellans; Succinoglycans; Arabinogalactans; Tragacanth or gum tragacanth or traganth from Astragalus; Gum Karaya from Sterculia urens; Gum Ghatti from Anogeissus latifolia; Derivatives thereof
    • C08B37/0063Glycosaminoglycans or mucopolysaccharides, e.g. keratan sulfate; Derivatives thereof, e.g. fucoidan
    • C08B37/0069Chondroitin-4-sulfate, i.e. chondroitin sulfate A; Dermatan sulfate, i.e. chondroitin sulfate B or beta-heparin; Chondroitin-6-sulfate, i.e. chondroitin sulfate C; Derivatives thereof
    • 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
    • C08B37/0024Homoglycans, i.e. polysaccharides having a main chain consisting of one single sugar, e.g. colominic acid beta-D-Glucans; (beta-1,3)-D-Glucans, e.g. paramylon, coriolan, sclerotan, pachyman, callose, scleroglucan, schizophyllan, laminaran, lentinan or curdlan; (beta-1,6)-D-Glucans, e.g. pustulan; (beta-1,4)-D-Glucans; (beta-1,3)(beta-1,4)-D-Glucans, e.g. lichenan; Derivatives thereof
    • C08B37/00272-Acetamido-2-deoxy-beta-glucans; Derivatives thereof
    • C08B37/003Chitin, i.e. 2-acetamido-2-deoxy-(beta-1,4)-D-glucan or N-acetyl-beta-1,4-D-glucosamine; Chitosan, i.e. deacetylated product of chitin or (beta-1,4)-D-glucosamine; Derivatives thereof
    • 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
    • A61K31/737Sulfated polysaccharides, e.g. chondroitin sulfate, dermatan sulfate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08LCOMPOSITIONS OF MACROMOLECULAR COMPOUNDS
    • C08L5/00Compositions of polysaccharides or of their derivatives not provided for in groups C08L1/00 or C08L3/00
    • C08L5/08Chitin; Chondroitin sulfate; Hyaluronic acid; Derivatives thereof
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to a method for the production of chondroitin sulphate by chemical sulphation starting from an unsulphated chondroitin backbone.
  • the process according to the invention allows simultaneous sulphation, within the same polysaccharide chain, of position 4 or position 6 of the N-acetyl-D-galactosamine residue.
  • the chondroitin sulphate thus obtained presents the same sulphation pattern as observed in natural chondroitin sulphate, unlike that obtained with the synthesis methods described so far.
  • the invention also relates to a chondroitin sulphate which has an average molecular weight determined by SEC (Mw) of 4-9 kDa, and a distribution of mono-sulphated groups ranging from 90% 4-sulphate and 10% 6-sulphate to 10% 4-sulphate and 90% 6-sulphate.
  • Mw average molecular weight determined by SEC
  • Chondroitin sulphate is a complex natural polysaccharide belonging to the glycosaminoglycan (GAG) class, consisting of disaccharide sequences formed by residues of glucuronic acid (GlcA) and N-acetyl-D-galactosamine (GalNAc) sulphated in different positions and bonded by beta 1-3 bonds.
  • GAG glycosaminoglycan
  • CS is present in animal tissues, with structural and physiological functions. Depending on its origin, CS mainly consists of variable percentages of two types of disaccharide unit monosulphated at position 4 or position 6 of GalNAc (disaccharides A and C respectively). However, disaccharides in which the sulphate groups are present in different numbers and different positions may be present in various percentages in the polysaccharide chains.
  • the CS backbone also contains unsulphated disaccharide, generally in small quantities.
  • Disulphated disaccharides having two sulphate groups bonded through the oxygen atom in various positions can be present in the CS backbone in variable percentages, depending on the specific animal sources (Volpi N. J Pharm Pharmacol 61, 1271, 2009. Volpi N. J Pharm Sci 96, 3168, 2007. Volpi N. Curr Pharm Des 12, 639, 2006).
  • the repeating disaccharide unit found in CS has the following chemical formula:
  • R 2 , R 4 and R 6 are independently H or SO 3 ⁇ .
  • the negative charges of the carboxylate and sulphate groups in the repeating disaccharide unit are neutralised by sodium ions.
  • Samples of CS originating from different animal sources are also characterised by different molecular weights and charge densities, this latter parameter being directly correlated with the specific sulphated groups.
  • Table 1 shows the main disaccharides found in natural CS extracted from cartilage and other tissues of various animal species:
  • CS derived from land animals has similar molecular mass parameters (Mn and Mw), whereas it is different from that originating from fish species, which have higher molecular mass values.
  • the terrestrial CS samples are also characterised by charge density (CD) values below 1.0, whereas the marine CS samples always have CD values exceeding 1.0. This characteristic is due to the different distribution of the sulphated disaccharides. Generally, disulphated disaccharides are found in trace amounts in terrestrial CS, and no polysulphated disaccharides (tri- and tetra-sulphates) are observed in natural CS.
  • FACE Fluorophore-Assisted Carbohydrate Electrophoresis
  • CS has anti-inflammatory activity, and is currently recommended in the treatment of osteoarthritis (OA) as a Symptomatic Slow-Acting Drug for OsteoArthritis (SYSADOA) in Europe, in particular for the treatment of osteoarthritis of the knee (Jordan K M et al., Ann Rheum Dis 62, 1145, 2003), hip (Jordan K M et al. Ann Rheum Dis 62, 1145, 2003) and hand (Zhang W et al., Ann Rheum Dis 66, 377, 2007) on the basis of clinical evidence and corresponding meta-analyses of numerous clinical trials.
  • OA osteoarthritis
  • SYSADOA Symptomatic Slow-Acting Drug for OsteoArthritis
  • CS is also widely used as a nutraceutical in Europe and the USA, either alone or in combination with other ingredients (McAlindon T E et al., JAMA 283, 1469, 2000. Volpi N et al., Food Anal Meth 1, 195, 2008. Volpi N et al., Separation Sc 1, 22, 2009).
  • CS Commercial CS is obtained by extraction from animal tissue, such as bovine and porcine tissue (Fuentes E P et al., Acta Farm Bonaerense 17, 135, 1998), bird tissue (Luo X M et al., Poult Sci 81, 1086-1089, 2002) and fish cartilage (Sugahara K et al., Eur J Biochem 239, 871, 1996. Lignot B et al., J Biotechnol 103, 281, 2003).
  • animal tissue such as bovine and porcine tissue (Fuentes E P et al., Acta Farm Bonaerense 17, 135, 1998), bird tissue (Luo X M et al., Poult Sci 81, 1086-1089, 2002) and fish cartilage (Sugahara K et al., Eur J Biochem 239, 871, 1996. Lignot B et al., J Biotechnol 103, 281, 2003).
  • BSE bovine spongiform encephalopathy
  • Another example of this strategy is the production of biotechnological CS from capsular polysaccharide K4 of E. coli O5:K4:H4, as described in EP 1304338 B1.
  • Said patent discloses a process wherein polysaccharide K4 produced in liquid cultures is extracted and purified, and then redissolved and subjected to acid hydrolysis to eliminate the fructose residues bonded to the GlcA residues of the polymer.
  • the defructosylated polymer identical to the unsulphated backbone of CS (CH), is then sulphated at position 4 or position 6 of the GalNAc residue according to two different chemical synthesis methods.
  • Said patent also discloses a third method whereby a disulphated CS in both positions 4 and 6 is obtained.
  • the CS described therein has a content of at least 70% of sulphated polysaccharides consisting of mono- and/or disulphated at position 4 and 6 of the GalNAc residue, position 2′ of the GlcA residue being unsulphated, and has a molecular weight (Mw) of 6-25 kDa and a charge density (CD) of 0.7-2.0.
  • FIG. 1 relates to natural chondroitin sulphate of bovine origin treated with chondroitinase C.
  • Various oligosaccharides of different length demonstrating the presence of sulphate groups at position 4 or 6 of the GalNAc residue on the same polysaccharide chain are formed.
  • the chromatogram was obtained by gradient separation on a strong anion-exchange column (SAX-HPLC) and UV detection at 232 nm.
  • the gradient was obtained by 50 mM NaCl up to 1.2 M NaCl from 0 to 60 minutes.
  • FIG. 2 relates to natural chondroitin sulphate of porcine origin treated with chondroitinase C.
  • Various oligosaccharides of different length demonstrating the presence of sulphate groups at position 4 or 6 of the GalNAc residue on the same polysaccharide chain are formed.
  • the chromatogram was obtained by gradient separation on a strong anion-exchange column (SAX-HPLC) and UV detection at 232 nm.
  • FIG. 3 relates to biotechnological chondroitin sulphate according to the present invention treated with chondroitinase C. Also for this polysaccharide various oligosaccharides of different length demonstrating the presence of sulphate groups at position 4 or 6 of the GalNAc residue on the same polysaccharide chain are formed.
  • the chromatogram was obtained by gradient separation on a strong anion-exchange column (SAX-HPLC) and UV detection at 232 nm.
  • the present invention describes a method for the production of CS following chemical sulphation starting from an unsulphated chondroitin backbone (CH), this CH being obtained by acid hydrolysis of a natural microbial polysaccharide i.l. (K4), or produced directly from a genetically modified E. coli , such as E. coli strain DSM23644, described in patent applications MI2010A001300 and MI2010A001264.
  • the bacterial strain described therein carries a mutation that causes the inactivation of the KfoE gene for fructosylation of K4.
  • the CS obtained by the process according to the invention presents the characteristics of a natural CS with a titre exceeding 95% on the basis of the analytic methods described in the European Pharmacopoeia.
  • the CS obtained with the process according to the invention has an average molecular weight (Mw), measured by SEC, of 10-30 kDa, preferably 20-30 kDa, and presents a distribution of mono-sulphated groups ranging from 90% of 4-sulphate and 10% of 6-sulphate to 10% of 4-sulphate and 90% of 6-sulphate (Table 2).
  • Mw average molecular weight
  • the CS obtained with the process according to the invention contains a small amount ( ⁇ 10%) of unsulphated disaccharide and very low percentages ( ⁇ 5%) of disulphated disaccharides; trisulphated disaccharides cannot be identified.
  • the CS obtained with the process according to the invention is characterised by charge density values of 0.8-1.0.
  • the CS obtained shows a ratio between the sulphated disaccharide at position 4 (Di-4S) and the sulphated disaccharide at position 6 (Di-6S) of less than 1, whereas in other forms it shows a ratio between (4S) disaccharide and (6S) disaccharide greater than 1.
  • the process according to the present invention allows site-specific sulphation to be modulated to produce a CS with a specific 4S/6S ratio within the range specified above.
  • the present invention also relates to the production of chondroitin sulphate (CS) with low molecular weight (LMW-CS BIOTEC, 4,000-9,000 daltons) by chemical sulphation from a non-sulphated chondroitin backbone, which in turn is obtained by acid hydrolysis of the capsular polysaccharide K4 produced by E. coli strain O5:K4:H4, or directly produced from a genetically modified E. coli .
  • CS chondroitin sulphate
  • LMW-CS BIOTEC low molecular weight
  • the chondroitin sulphate with low molecular weight that is object of the invention is characterised by a molecular weight interval of 4,000-9,000 daltons, which is much less than that of chondroitin sulphates of natural origin, whether terrestrial, in particular of bovine, porcine or avian origin (14,000-26,000 daltons) or of marine origin, for example obtained from sharks, squid, rays or bony fish (generally >40,000 daltons).
  • the chondroitin sulphate according to the invention presents higher absorption after oral administration and therefore better bioavailability in humans than highly pure natural chondroitin sulphate or chondroitin sulphate produced by biotechnological/chemical processes.
  • the chondroitin sulphate according to the invention possesses anti-inflammatory and antiarthritic activity comparable with those of highly pure natural chondroitin sulphate.
  • the chondroitin sulphate according to the invention is suitable for use in the treatment of inflammatory and osteoarthritic/arthritic processes.
  • the LMW-CS BIOTEC according to the invention has an average molecular weight, measured by SEC (Mw), of 4-9 kDa, and a distribution of mono-sulphated groups ranging from 90% 4-sulphate and 10% 6-sulphate to 10% 4-sulphate and 90% 6-sulphate.
  • Mw average molecular weight
  • mono-sulphated groups ranging from 90% 4-sulphate and 10% 6-sulphate to 10% 4-sulphate and 90% 6-sulphate.
  • the characteristics of the low molecular weight CS according to the invention are substantially identical to those of the higher molecular weight derivatives reported in Table 2 above.
  • the LMW-CS BIOTEC according to the invention has a small quantity ( ⁇ 10%) of non-sulphated disaccharide and very low percentages ( ⁇ 5%) of disulphated disaccharides, while no trisulphated disaccharides are identifiable.
  • LMW-CS BIOTEC is characterised by charge density values of 0.8-1.0, which are comparable with those of natural CS of terrestrial origin (see Table 1).
  • the process according to the invention also allows site-specific sulphation to be modulated in order to supply a CS with a specific 4S/6S ratio within the limits specified above, which are similar to those present in CS of natural origin.
  • the LMW-CS BIOTEC according to the invention is recognised and digested by chondroitinase ABC, a lytic enzyme which has the task of catabolising the natural CS in specific organisms, thus demonstrating that the polysaccharide chains of biotechnological LMW-CS have not undergone structural modifications liable to prejudice the specific, highly sensitive recognition of natural enzymes.
  • FIG. 1 in particular describes natural chondroitin sulphate of bovine origin treated with chondroitinase C. Oligosaccharides of different lengths can be seen which indicate the presence of sulphate groups in position 4 or 6 of the GalNAc residue on the same polysaccharide chain.
  • SAX-HPLC strong anion-exchange column
  • UV detection at 232 nm. The gradient was obtained with 50 mM NaCl to 1.2 M NaCl from 0 to 60 minutes;
  • FIG. 2 describes natural chondroitin sulphate of porcine origin treated with chondroitinase C. Oligosaccharides of different lengths can be seen which indicate the presence of sulphate groups in position 4 or 6 of the GalNAc residue on the same polysaccharide chain.
  • the chromatogram was obtained by gradient separation on strong anion-exchange column (SAX-HPLC) and UV detection at 232 nm;
  • FIG. 3 describes the LMW-CS BIOTEC of the present invention, treated with chondroitinase C.
  • the chromatogram was obtained by gradient separation on strong anion-exchange column (SAX-HPLC) and UV detection at 232 nm.
  • the LMW-CS BIOTEC according to the invention has been evaluated for oral absorption and bioavailability in humans by comparison with highly pure natural CS of bovine origin, the first standard of the European Pharmacopoeia. This is particularly important because the presence of a bacterium able to biosynthesise a lytic enzyme specific for the breakdown of CS (and derivatives with low molecular weight) has been described in human but not animal bacterial flora (Ahn M Y, et al., Can J Microbiol 1998; 44: 423-9).
  • the LMW-CS BIOTEC according to the invention was evaluated for possible anti-inflammatory activity using specific tests such as:
  • the LMW-CS BIOTEC according to the invention was also evaluated for antiarthritic properties in an animal model, the “Adjuvant Arthritis (AA) model”, which is widely recognised by the scientific community and has been published in numerous scientific papers. Once again, the results were compared with those previously obtained with the reference molecule: the European Pharmacopoeia standard, a highly pure natural CS of bovine origin (Volpi N. J Pharm Sci 96, 3168, 2007). In fact, animal models of OA and rheumatoid arthritis (AR) are useful tools for the study of these pathogenic processes. “Adjuvant Arthritis” (AA) is one of the most commonly used models.
  • AA in the rat is an experimental model of polyarthritis which has been widely used to test numerous antiarthritic agents and medicaments before and after thorough clinical trials (Bendele A et al., Toxicol Pathol 27, 134, 1999; Rovensky J et al., Rheumatol Int. 31, 507, 2011; Bauerova K et al., Interdisc Toxicol 4, 101, 2011). Numerous studies have also been conducted wherein the data on animals obtained with the AA test were compared with the results in humans (Kannan K et al., Pathophysiology 12, 167, 2005).
  • compositions of the CS according to the invention relates to the composition of the CS according to the invention and a carrier acceptable in the pharmaceutical or nutraceutical field.
  • Said composition can be formulated in various solid forms, such as tablets, rigid capsules, soft gelatin capsules or powdered mixtures for drinks, or in liquid forms (solutions), preferably in the form of pharmaceutical or nutraceutical preparations for parenteral or oral administration.
  • the composition can contain other active or inactive ingredients.
  • the composition can also, preferably, contain at least one of the following substances: glucosamine hydrochloride, glucosamine sulphate, N-acetyl glucosamine, hyaluronic acid, heparin, keratin, dermatin, methyl sulphonyl methane, folates and reduced folates, Group B vitamins, S-adenosylmethionine (SAMe), ascorbic acid or manganese ascorbate.
  • SAMe S-adenosylmethionine
  • the composition can be administered to patients in effective quantities based on their needs.
  • the CS or the composition described in the present invention can be administered in a quantity of between 100 and 3000 mg a daily, preferably between 1000 and 2000 mg a daily, and more preferably between 1250 and 1750 mg a daily, divided into two doses of approx. 600 mg or three doses of 400 mg a daily.
  • the present invention also relates to the use of the CS described, or a composition thereof, for the treatment or prevention of osteoarthritis or for the maintenance of musculoskeletal well-being as an ingredient of a medicament or nutritional supplement.
  • the CS described or a composition thereof can be used to make a pharmaceutical preparation, dietary additive or nutritional supplement for the prevention and/or treatment of osteoarthritis of the hip, hand or knee and the main symptoms thereof (pain, joint swelling, inflammation), Alzheimer's disease, microbial infections, arteriosclerosis and osteoporosis, and as adjuvant in antitumoral treatment and tissue regeneration, including nerve tissue.
  • An advantageous characteristic of the process according to the invention is that the sulphation at position 4 or 6 of the GalNAc residue takes place simultaneously in the same polysaccharide chain, simulating the sulphation pattern observed in natural CS, unlike that obtained with the synthesis methods described to date.
  • the CS according to the present invention can be obtained using as starting substrate the capsular polysaccharide K4 naturally produced by E. coli strain O5:K4:H4 (EP 1304338 B1) or another polysaccharide having the structure of unsulphated chondroitin (CH).
  • capsular polysaccharide K4 naturally produced by E. coli strain O5:K4:H4 (EP 1304338 B1) or another polysaccharide having the structure of unsulphated chondroitin (CH).
  • polysaccharide K4 obtained from a culture broth of E. coli strain O5:K4:H4, is defructosylated at the end of fermentation by thermoacid hydrolysis, and the chondroitin is purified in accordance with an adaptation of the methods described by Rodriguez and Jann (Eur. J. Biochem. 117, 117-124, FEBS 1988).
  • the starting polysaccharide is obtained, for example, from the culture of E. coli strain DSM23644 described in MI2010A001300 which, due to a mutation induced in the KfoE gene responsible for the fructosylation of K4, produces a polysaccharide identical to natural unsulphated CH. Defructosylation is not necessary in this case; however, the thermoacid hydrolysis step is maintained to eliminate some impurities, including the bacterial endotoxins that precipitate as a result of the treatment. The chondroitin (CH) is then purified by centrifugation, dialysis and spray drying.
  • Hydrolysis is conducted on the culture supernatant, separated from the biomass by continuous centrifugation. Partial hydrolysis and defructosylation of K4 is performed by incubation at 90-95° C. for 30-50 min at pH 2.8-3.0.
  • the resulting suspension is cooled at a temperature below 40° C., preferably 20-30° C., to quench the hydrolysis reaction, and the pH is simultaneously adjusted to 4-4.5.
  • the resulting suspension undergoes, in sequence, clarification by continuous centrifugation, ultrafiltration and finally, dialysis with water through a 30 kDa membrane.
  • the dialysed retentate (approx. 1/10th of the volume of the initial culture broth) is filtered and finally dried with a spray dryer to obtain a polysaccharide having the structure of CH, to be subjected to the sulphation process.
  • the CH obtained has a titre of 80-90% on a dry basis (w/w), as determined by capillary electrophoresis (CE) or HPLC.
  • the CH thus obtained takes the form of the sodium salt, and in order to be sulphated needs to be converted to free acid or a salt thereof.
  • the sulphation process according to the present invention which allows positions 4 or 6 of the GalNAc residue of the same polysaccharide chain to be monosulphated randomly, comprises the formation of an orthoester which simultaneously involves GalNAc positions 4 and 6 and its subsequent rearrangement to an ester which, surprisingly, can be modulated to release mainly the hydroxyl in 4 or in 6, thus allowing selective sulphation of those hydroxyls.
  • the process according to the invention comprises the following steps:
  • chondroitin sodium salt Conversion of the chondroitin sodium salt to free acid or, alternatively, to a salt thereof with a quaternary ammonium ion, such as tetramethyl-, tetraethyl- or tetrabutyl-ammonium, or with pyridine.
  • Tetrabutylammonium (TBA) salt is preferably used.
  • chondroitin (CH) in acid form is converted to its methyl ester after reaction in methanol and acetyl chloride.
  • R, R 1 are as defined above.
  • orthoesters which can be used are trimethyl orthoacetate, triethyl orthoacetate, trimethyl orthoformate, triethyl orthoformate, trimethyl orthopropionate, triethyl orthopropionate or trimethyl orthobenzoate. Trimethyl orthoacetate or triethyl orthoacetate is preferably used. The use of trimethyl orthoacetate is particularly preferred.
  • R, R 1 and R 2 are as defined above.
  • Acetic anhydride is preferably used.
  • R and R 2 are as defined above;
  • R and R 2 are as defined above.
  • the water-soluble organic acid is selected from acetic, formic, propionic, tartaric citric acid or a cationic resin such as for example Sepra SCX 50 ⁇ m 65A, preferably acetic acid or propionic acid, and more preferably acetic acid.
  • LMW low molecular weight
  • Chondroitin can also be depolymerised at the orthoester rearrangement stage, using the acid as solvent or co-solvent of the reaction.
  • the high concentration of acid at this stage leads to rupture of the polysaccharide chain, with consequent production of low molecular weight chains, in the 4-9 kD range.
  • the LMW-CS BIOTEC 4,000-9,000 daltons, obtained by the process described, was evaluated for efficacy in an experimental animal arthritis model (Adjuvant Arthritis AA) in the rat, and the results were compared with those for pharmaceutical grade natural CS of extracted origin used in the same experimental model (Bauerova K. et al., Osteoarthritis Cartilage 2011, Epub ahead of print) after daily oral treatment with 900 mg/kg.
  • AA was induced by a single intradermal injection of Mycobacterium butyricum in incomplete Freund's adjuvant.
  • the experiments comprised healthy animals, untreated arthritic animals and treated arthritic animals.
  • one group of animals was subjected to pre-treatment consisting of administration of 900 mg/kg of LMW-CS BIOTEC a day for 14 days before arthritis was induced, continuing for 28 days after the induction of AA.
  • Another group of animals was treated with 900 mg/kg of LMW-CS BIOTEC a day solely during the 28 days after the induction of AA.
  • Pre-treatment with the LMW-CS BIOTEC according to the invention significantly reduced oedema throughout the experiment compared with the untreated controls.
  • Pre-treatment with LMW-CS BIOTEC also restores the body weight by approx. 8-15% compared with the untreated arthritic control.
  • the severity of the arthritis was quantified on the basis of increasing levels of swelling and periarticular erythema. 900 mg/kg/day of LMW-CS BIOTEC, administered as both pre-treatment and treatment, is significantly effective in reducing the arthritis score. Moreover, pre-treatment is effective throughout the subacute stage (from day 14 to day 28 after induction of AA), whereas the treatment is only effective in the medium-long term, in days 21-28 after induction of AA, not at the acute stage (the first 14 days after induction of AA).
  • Oxidative stress a consequence of the chronic inflammatory processes that take place in arthritic/osteoarthritic processes, significantly increases in the animal model at both the acute and the subchronic stage. Increased oxidative stress induces high consumption of endogenous antioxidants in the plasma, and consequently causes a reduction in the plasma antioxidant capacity, measured as the total antioxidant status. Pre-treatment with LMW-CS BIOTEC is effective in correcting the total antioxidant status in the animal model, significantly reducing the consumption of endogenous antioxidants.
  • ⁇ -glutamyl transferase which increases in correspondence with oxidative stress and is therefore considered to be a good marker for oxidative stress, measured in joint tissue homogenates, proved considerably greater in animals with experimentally induced polyarthritis, and considerably lower in the animals treated with LMW-CS BIOTEC, by comparison with the untreated animals.
  • Interleukin-1 ⁇ IL- ⁇ and interleukin-6 (IL-6), pro-inflammatory cytokines, significantly increased in the animal model of experimentally induced arthritis, with a dramatic increase in IL-6 at the acute stage, presenting a level 10 times higher than the healthy controls.
  • the therapeutic effect of LMW-CS BIOTEC was already evident from day 14, at the acute stage, reducing the IL-6 concentration by approx. 30-40% compared with the animals suffering from AA.
  • CRP C-reactive protein
  • the differences observed between the healthy control and the control suffering from induced experimental AA were significant in the case of increased phagocytic activity.
  • the administration of LMW-CS BIOTEC on a pre-treatment basis induced a significant reduction in phagocytosis and the oxidative burst.
  • the LMW-CS BIOTEC significantly reduces the severity of the arthritic processes and the oxidative stress generated as a result of chronic inflammatory processes.
  • Pre-treatment with LMW-CS BIOTEC is effective throughout the subacute stage, whereas treatment from day 1 of onset of AA is only effective during the chronic period. The effects are confirmed by an improvement in the total antioxidant status and activity of ⁇ -glutamyl transferase.
  • LMW-CS BIOTEC administered as a pre-treatment, also reduces the production of pro-inflammatory cytokines, C-reactive protein in the plasma, phagocytic activity and the intracellular oxidative burst of the neutrophils.
  • LMW-CS BIOTEC has proved effective in slowing the development of experimental arthritis/osteoarthritis at both the acute and the subchronic stage, and in reducing the markers of the disease, thus supporting its beneficial activity, on a par with that of the reference compound.
  • a cation-exchange resin such as Amberjet 1200 H, Rohm and Haas, or equivalent.
  • the fractions eluted at pH 1.5-4.0, or preferably at pH 1.5-2.0, are collected, and an aqueous solution of an ion selected from tetramethyl-, tetraethyl- and tetrabutyl-ammonium or pyridinium is added until a pH of 6.0-8.0, or preferably 6.5-7.0, is obtained.
  • the solution is then evaporated to complete dryness by freeze-drying or spray drying to obtain the corresponding salt.
  • the salt obtained from chondroitin such as tetrabutyl ammonium (TBA) salt, is mixed with dimethylformamide (DMF) in a flask in the quantities of 5.2 g and 130 ml respectively.
  • DMF dimethylformamide
  • 8.49 g of trimethyl orthoacetate is dripped into the flask, followed by the addition of 300 mg of camphorsulphonic acid, and the reaction mixture is maintained at 70° C. for 72 h.
  • the reaction is then evaporated under vacuum to dryness, and further stove-dried at 40° C. for 20 h to obtain 6.1 g of chondroitin-MOE TBA in the form of a solid.
  • chondroitin originating from the preceding step protected as cyclic methyl orthoester (CH-cMOE) (4.79 g), is introduced into a reaction flask with 23.95 ml of acetonitrile, 15.69 ml of triethylamine (TEA), 6.21 ml of acetic anhydride and 78.96 mg of 4-dimethylaminopyridine (DMAP). After 2 hours' stirring at 25-26° C., 94 ml of di-isopropyl ether is added to obtain a viscous solid, which is then filtered through filter paper and stove-dried under vacuum at 45° C. for 24 h. The intermediate cyclic orthoester thus obtained has the appearance of a pink solid.
  • CH-cMOE cyclic methyl orthoester
  • the intermediate obtained from the preceding step (2.42 g) is introduced into a reaction flask, to which 18.8 ml of 96% acetic acid and 2.35 ml of demineralised water are added.
  • the mixture is stirred for 38 h at room temperature, after which 100 ml of an 0.6 M solution of NaCl are added and the mixture is ultrafiltered through a 5 kDa membrane and dialysed, to recover a retentate with a pH of 3.32.
  • the intermediate obtained as described in example 4 (0.76 g) is introduced into a flask with 46.0 ml of DMF starring the mixture at 30° C. for 10 min. 0.72 g of sulphur trioxide pyridinium are added and when the starting material has dissolved (approx. 10 min), the solution is left under stirring at 30° C. for 1 h. A further 0.72 g of sulphur trioxide pyridinium are then added, followed by a further 0.72 g of sulphur trioxide pyridinium. The solution is stirred for a further hour at 30° C.
  • the reaction is quenched by pouring the mixture into 50 ml of 10% NaHCO 3 in water at room temperature (pH 7.81). After filtration the solution is evaporated under vacuum (10 mBar) to dryness, the residue redissolved with 150 ml of 0.6 M NaCl and, finally, the solution is ultrafiltered.
  • the retentate After 6 changes of volume the retentate has a pH of 9.22; the pH is adjusted to 6.7 with 1N HCl and ultrafiltration continues, replacing the 0.6N NaCl solution with demineralised water.
  • the resulting solution is ultrafiltered again for 2 volumes, and then dialysed to a volume of 20 ml.
  • the dialysed solution is concentrated to dryness under vacuum (10 mBar, 45° C.).
  • the product thus obtained (0.88 g) is dissolved with 34.0 ml of 0.2N soda (NaOH) and heated to 40° C. under stirring for 2 h. Finally, the solution is diluted with an 0.6M aqueous solution of sodium chloride, ultrafiltered through a 5 kDa membrane, and dialysed with demineralised water. The retentate is concentrated to dryness under vacuum (45° C., 10 mBar), to obtain 0.67 g of chondroitin sulphate.
  • the end product which has a molecular weight of 29 kDa, determined by HPLC-SEC, shows:
  • the intermediate obtained as described in example 5 (1.12 g) is introduced into a flask with 67.2 ml of DMF, stirring the mixture at 50° C. for 10 min. 1.05 g of sulphur trioxide pyridinium are added, and when the starting material has dissolved (approx. 10 min), the solution is left under stirring at 50° C. for 1 h. A further 1.05 g of sulphur trioxide pyridinium are then added. The solution is stirred for a further hour at 50° C.
  • reaction is quenched by pouring the mixture into 60 ml of 10% NaHCO 3 in water at room temperature (RT) (pH 7.81). After filtration the solution is evaporated under vacuum (10 mBar) to dryness, and the residue is redissolved with 30 ml of 0.6 M NaCl. Finally, the solution is ultrafiltered.
  • the retentate After 6 changes of volume the retentate has a pH of 9.22; the pH is adjusted to neutrality (7.5) with 1 N HCl and microfiltration continues, replacing the 0.6 N NaCl solution with demineralised water.
  • the resulting solution is ultrafiltered again for 2 volumes, and then dialysed to a volume of 20 ml.
  • the dialysed solution is concentrated to dryness under vacuum (10 mBar, 45° C.), to obtain 1.53 g of product.
  • the product thus obtained has a molecular weight of 15.4 kDa, determined by HPLC-SEC; digestibility with chondroitinase ABC exceeding 95%; a 4S/6S ratio of 82/18; and a total charge density value of approx. 1.09.
  • the suspension is filtered and the solid is washed with 100 ml of methanol (2 ⁇ 50 ml) and dried at 50° C. under vacuum to recover 9.4 g of dry solid.
  • the reaction is repeated a second time with the same procedure, and when the second period has elapsed, the suspension is cooled at between 0 and 5° C. for 60 minutes before filtration.
  • the solid obtained is washed with cold methanol (0-5° C.) and stove-dried under vacuum for 3 hours at 50° C. to recover 6.3 g of solid.
  • DMF dimethylformamide
  • 6.0 g of the product obtained in the preceding step 150 ml of dimethylformamide (DMF) and 6.0 g of the product obtained in the preceding step are introduced into a 500 ml flask with a calcium chloride valve and nitrogen flow. 20.06 g of trimethyl orthoacetate and 0.71 g of camphorsulphonic acid are then added. The solution obtained is heated at 50° C. (internal temperature) for 18 hours.
  • the solution obtained is left under stirring for 3 hours; when that time has elapsed, 150 ml of isopropyl ether are added to the flask and an amorphous solid precipitates.
  • the waters are eliminated by decanting and 100 ml of isopropyl ether are added to the solid and left under stirring for 1 hour.
  • the solid is then filtered and washed with 50 ml of isopropyl ether and dried under vacuum at 40° C. to recover 8.52 g of product.
  • 630.44 g of sulphur trioxide pyridinium complex are added to the solution obtained and the resulting solution is heated at 50° C. (internal temperature) for 1 hour. 630.44 g of sulphur trioxide pyridinium complex are then added to the flask at the same temperature and again left under stirring for 1 hour.
  • the solution is cooled to RT and 40 ml of 3% NaHCO 3 are added to the flask at the same temperature to produce a solution which is concentrated under vacuum to obtain 2.3 g of solid mixed with inorganic salts.
  • the product obtained is diluted to 150 ml of 0.6 M sodium chloride and ultrafiltered through a 5 KDa membrane.
  • the product obtained in the preceding step is introduced into a 100 ml flask with 33 ml of 0.2 M soda.
  • the solution is heated at 40° C. (internal temperature) for 2 hours, after which it is cooled to RT and neutralised with 1M HCl.
  • the solution is diluted to 150 ml of 0.6 M sodium chloride and ultrafiltered through a 5 KDa membrane. After dialysis and concentration of the solution under vacuum, 350 mg of solid are obtained.
  • the product obtained in this example has a molecular weight of 11 KDa, a 4S/6S ratio of 47/53, and a charge density value of 0.9.
  • the residual content of unprotected chondroitin after digestion is 4.6%.
  • the presence of the orthoester is demonstrated by the corresponding signal in FTIR.
  • Chondroitin orthoester (3.00 g), water (3.14 ml) and acetic acid (26.25 g; 437 mmols) were introduced into a 250 ml three-necked flask.
  • the suspension obtained was heated for 36 h at ambient temperature (20-25° C.). Water was then added to make up the solution to a total volume of 100 ml.
  • the solution thus obtained was ultrafiltered (5 KD membrane).
  • the retentate collected was dialysed to a small volume (20 ml), and then concentrated until dry by evaporation under vacuum, providing 1.55 g of solid residue corresponding to the desired product (triacetyl chondroitin).
  • 40 male Lewis rats weighing between 150 and 190 g were randomised to four groups of 10 animals each, housed in polypropylene cages in a environment maintained at the temperature of 22 ⁇ 2° C., and fed on a standard laboratory diet with unlimited access to water.
  • the LMW-CS BIOTEC was dissolved in distilled water at the concentration of 20 mg/ml and administered orally as a single daily dose by gavage.
  • the oedema that developed as a consequence of arthritis was measured by observing the increase in volume of the hind paw with a caliper suitable for the measurement. The measurements were performed before the induction of AA and on day 28 of the study.
  • the body weight of the rats was measured before induction of AA and at the end of the treatment (day 28). The effect of the treatment on this parameter was evaluated by comparing the various weight increases of the different groups during the treatment period.
  • the arthritis score was evaluated by attributing a score to the paw joint swelling and the extent of the periarticular erythema.
  • the arthritis score or arthrogram was measured as the sum total of oedema (in ml, max. 8 points), plus the diameter of the forepaw (in mm, max 5 points), plus the diameter of the scab at the site of application of Mycobacterium butyricum measured parallel to the spinal column (in mm, max 5 points), for each animal.
  • Oxidative stress was evaluated by measuring the activity of ⁇ -glutamyl transferase in homogenates of joint tissue taken from the rats at the end of the treatments with LMW-CS BIOTEC. ⁇ -glutamyl transferase is considered to be a marker for oxidative stress.
  • the activity of the cell ⁇ -glutamyl transferase was determined in homogenates of tissue taken from the hind paw, and evaluated by the Orlowski and Meister method (Orlowski M, Meister A. The gamma-glutamyl cycle: a possible transport system for amino acids. Proc Natl Acad Sci USA 1970; 67: 1248-1255) as modified by Ondrejickova et al. (Cardioscience 1993; 4: 225-230).
  • the samples were homogenised in a buffer (2.6 mM NaH 2 PO 4 , 50 mM Na 2 HPO 4 , 15 mM EDTA, 68 mM NaCl, pH 8.1) in a 1:9 (w/v) solution with UltraTurax TP 18/10 (Janke & Kunkel, Germany) for 1 min at 0° C.
  • the substrates 8.7 mM of ⁇ -glutamyl p-nitroanilide and 44 mM of methionine, were added to 65% of isopropyl alcohol at final concentrations of 2.5 mM and 12.6 mM respectively.
  • reaction mixtures in the absence of substrate or acceptor were used as reference samples.
  • C-reactive protein was assayed in the rat plasma with an ELISA kit (Immunology Consultant Laboratories, Inc., ICL).
  • the reaction of the biotin-conjugated secondary antibody with anti-rat C-reactive protein antibodies was evaluated by means of the activity of streptavidin-horseradish peroxidase (HRP).
  • HRP streptavidin-horseradish peroxidase
  • the reaction of methyl-benzidine with HRP bonded to immune complexes was then measured at 450 nm using a Labsystems Multiskan RC microplate reader. The results were calculated using the standard calibration curve in accordance with the ELISA kit instructions.
  • the neutrophil population was extracted from the blood of the rats at the end of the evaluation of their phagocytic activity and oxidative burst.
  • the measurement of phagocytosis namely ingestion of bacteria, was performed under controlled conditions using opsonised Staphylococcus aureus labelled with fluorescein (SPA-FITC) (Invitrogen Molecular Probes, USA). Aliquots of peripheral blood in lithium-heparin were then incubated with hydroethidine (Invitrogen molecular probes, USA) (15.75 mg in 5 ml of dimethylformamide, Merck, Germany) for 15 minutes at 37° C. After treatment with SPA-FITC for 15 minutes at 37° C., the reaction was interrupted by placing the test tubes in ice.
  • SPA-FITC opsonised Staphylococcus aureus labelled with fluorescein
  • the subsequent lysis of the erythrocytes was performed for 15 min with a lysis solution consisting of cold ammonium chloride/potassium chloride (200 ml deionised water, 1.658 g NH 4 Cl, 0.2 g KHCO 3 and 7.4 mg Na 2 EDTA, pH 7.2-7.4).
  • the average percentage of phagocyte cells represents the percentage of granulocytes which ingested at least one particle of SPA-FITC, and the average percentage of the respiratory burst represents the percentage of granulocytes labelled with ethidium.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Engineering & Computer Science (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Molecular Biology (AREA)
  • Polymers & Plastics (AREA)
  • Dermatology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Neurology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Rheumatology (AREA)
  • Materials Engineering (AREA)
  • Biochemistry (AREA)
  • Epidemiology (AREA)
  • Biomedical Technology (AREA)
  • Neurosurgery (AREA)
  • Urology & Nephrology (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Vascular Medicine (AREA)
  • Hospice & Palliative Care (AREA)
  • Oncology (AREA)
  • Pain & Pain Management (AREA)
  • Immunology (AREA)
  • Communicable Diseases (AREA)
  • Psychiatry (AREA)
  • Polysaccharides And Polysaccharide Derivatives (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
US14/115,184 2011-05-12 2012-05-10 Biotechnological sulphated chondroitin sulphate at position 4 or 6 on the same polysaccharide chain, and process for the preparation thereof Abandoned US20140315854A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
ITMI2011A000829 2011-05-12
IT000829A ITMI20110829A1 (it) 2011-05-12 2011-05-12 Condroitin solfato biotecnologico solfatato in posizione 4 o 6 sulla stessa catena polisaccaridica e procedimento per la sua preparazione
IT000136A ITMI20120136A1 (it) 2012-02-02 2012-02-02 Condroitin solfato biotecnologico a basso peso molecolare solfatato in posizione 4 o 6 sulla stessa catena polisaccaridica dotato di attivita' antiinfiammatoria e antiartritica e procedimento per la sua preparazione
ITMI2012A000136 2012-02-02
PCT/EP2012/058654 WO2012152872A1 (en) 2011-05-12 2012-05-10 "biotechnological sulphated chondroitin sulphate at position 4 or 6 on the same polysaccharide chain, and process for the preparation thereof"

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2012/058654 A-371-Of-International WO2012152872A1 (en) 2011-05-12 2012-05-10 "biotechnological sulphated chondroitin sulphate at position 4 or 6 on the same polysaccharide chain, and process for the preparation thereof"

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US14/977,119 Division US9908947B2 (en) 2011-05-12 2015-12-21 Biotechnological sulphated chondroitin sulphate at position 4 or 6 on the same polysaccharide chain, and process for the preparation thereof

Publications (1)

Publication Number Publication Date
US20140315854A1 true US20140315854A1 (en) 2014-10-23

Family

ID=46208441

Family Applications (2)

Application Number Title Priority Date Filing Date
US14/115,184 Abandoned US20140315854A1 (en) 2011-05-12 2012-05-10 Biotechnological sulphated chondroitin sulphate at position 4 or 6 on the same polysaccharide chain, and process for the preparation thereof
US14/977,119 Active 2032-06-21 US9908947B2 (en) 2011-05-12 2015-12-21 Biotechnological sulphated chondroitin sulphate at position 4 or 6 on the same polysaccharide chain, and process for the preparation thereof

Family Applications After (1)

Application Number Title Priority Date Filing Date
US14/977,119 Active 2032-06-21 US9908947B2 (en) 2011-05-12 2015-12-21 Biotechnological sulphated chondroitin sulphate at position 4 or 6 on the same polysaccharide chain, and process for the preparation thereof

Country Status (25)

Country Link
US (2) US20140315854A1 (de)
EP (1) EP2707396B1 (de)
JP (1) JP6205350B2 (de)
KR (1) KR101883910B1 (de)
CN (1) CN103582653B (de)
AU (1) AU2012252415B2 (de)
BR (1) BR112013027389B1 (de)
CA (1) CA2835498C (de)
CY (1) CY1121099T1 (de)
DK (1) DK2707396T3 (de)
EA (1) EA023849B1 (de)
ES (1) ES2705734T3 (de)
GE (1) GEP201606591B (de)
HR (1) HRP20190183T1 (de)
HU (1) HUE042378T2 (de)
IL (1) IL229367A (de)
LT (1) LT2707396T (de)
MX (1) MX351660B (de)
PL (1) PL2707396T3 (de)
PT (1) PT2707396T (de)
RS (1) RS58293B1 (de)
SG (1) SG194898A1 (de)
SI (1) SI2707396T1 (de)
WO (1) WO2012152872A1 (de)
ZA (1) ZA201308458B (de)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ITMI20130117A1 (it) 2013-01-25 2014-07-26 Gnosis Spa Composizioni comprendenti condroitin solfato, enzimi proteolitici e composti sulfidrilati capaci di migliorare la biodisponibilita' del condroitin solfato
CZ2014451A3 (cs) * 2014-06-30 2016-01-13 Contipro Pharma A.S. Protinádorová kompozice na bázi kyseliny hyaluronové a anorganických nanočástic, způsob její přípravy a použití
CN111662394B (zh) * 2019-03-05 2022-11-04 中国医学科学院药物研究所 硫酸软骨素多糖半合成制备方法和应用
IT202000013633A1 (it) 2020-06-08 2021-12-08 Vivatis Pharma Gmbh Processo di estrazione di una condroitina solfato da un fungo, una condroitina solfato di origine vegetale e suo uso
IT202000013618A1 (it) 2020-06-08 2021-12-08 Vivatis Pharma Gmbh Processo di estrazione di un acido ialuronico da un fungo, un acido ialuronico di origine vegetale e suo uso

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3405120A (en) * 1966-01-27 1968-10-08 Green Cross Corp Low molecular chondroitin sulfate and method for manufacturing the same
WO2012062917A2 (en) * 2010-11-11 2012-05-18 Altergon S.A. Process for obtaining chondroitin sulphated at the 4- or 6- positions of n-acetyl-galactosamine residues

Family Cites Families (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4704356A (en) * 1984-03-27 1987-11-03 Rush-Presbyterian-St. Luke's Medical Center Method of diagnosing cartilage tissue abnormalities
JPS6144822A (ja) * 1984-08-09 1986-03-04 Seikagaku Kogyo Co Ltd コンドロイチン硫酸含有抗疲労剤
JPH0699485B2 (ja) * 1984-08-14 1994-12-07 生化学工業株式会社 合成コンドロイチン多硫酸の製造法
US5856174A (en) 1995-06-29 1999-01-05 Affymetrix, Inc. Integrated nucleic acid diagnostic device
JP4258856B2 (ja) * 1998-03-06 2009-04-30 ニプロ株式会社 抗癌性複合体
ITMI20012200A1 (it) * 2001-10-22 2003-04-22 Ibsa Inst Biochimique Sa Processo per la preparazione di condroitin solfati dal polisaccaride k4 e prodotti ottenuti
JP2003128705A (ja) * 2001-10-29 2003-05-08 Kyowa Technos:Kk 低分子キトサンおよびその製造方法
JP4317350B2 (ja) * 2002-06-12 2009-08-19 生化学工業株式会社 顆粒球マクロファージコロニー刺激因子分泌促進剤
US8455458B2 (en) * 2002-10-16 2013-06-04 Arthrodynamic Technologies, Animal Health Division, Inc. Composition and method for treating connective tissue damage
JP4567942B2 (ja) * 2002-12-27 2010-10-27 生化学工業株式会社 破骨細胞形成抑制剤
JP4914594B2 (ja) * 2005-09-29 2012-04-11 ユニテックフーズ株式会社 関節痛改善用食品組成物
CN1789287A (zh) * 2005-12-20 2006-06-21 山东大学 一种多硫酸化硫酸软骨素及其制备方法
EP2204387B1 (de) * 2007-10-01 2016-07-20 Seikagaku Corporation Neues chondroitinsulfat mit verringertem molekulargewicht und verwendung davon
US8394782B2 (en) * 2007-11-30 2013-03-12 Allergan, Inc. Polysaccharide gel formulation having increased longevity
WO2009149155A1 (en) * 2008-06-04 2009-12-10 Amano Enzyme Usa., Ltd. Microbial-derived chondroitin sulfate
JP5435538B2 (ja) * 2008-09-25 2014-03-05 国立大学法人鳥取大学 コンドロイチン硫酸の低分子化物の製造方法

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3405120A (en) * 1966-01-27 1968-10-08 Green Cross Corp Low molecular chondroitin sulfate and method for manufacturing the same
WO2012062917A2 (en) * 2010-11-11 2012-05-18 Altergon S.A. Process for obtaining chondroitin sulphated at the 4- or 6- positions of n-acetyl-galactosamine residues

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Fujikawa, Biomolecules 2005, 6, 2935-2942. *

Also Published As

Publication number Publication date
EA201391673A1 (ru) 2014-03-31
NZ617564A (en) 2015-06-26
CA2835498A1 (en) 2012-11-15
DK2707396T3 (en) 2019-01-28
EA023849B1 (ru) 2016-07-29
JP2014514424A (ja) 2014-06-19
CY1121099T1 (el) 2019-12-11
IL229367A (en) 2016-11-30
EP2707396A1 (de) 2014-03-19
GEP201606591B (en) 2017-01-10
HRP20190183T1 (hr) 2019-03-22
PT2707396T (pt) 2019-01-23
CA2835498C (en) 2019-06-25
ZA201308458B (en) 2015-02-25
IL229367A0 (en) 2014-01-30
RS58293B1 (sr) 2019-03-29
WO2012152872A1 (en) 2012-11-15
PL2707396T3 (pl) 2019-04-30
BR112013027389B1 (pt) 2021-06-01
MX2013013181A (es) 2013-12-12
US9908947B2 (en) 2018-03-06
SI2707396T1 (sl) 2019-02-28
BR112013027389A2 (pt) 2017-01-17
EP2707396B1 (de) 2018-11-14
AU2012252415B2 (en) 2016-11-24
CN103582653B (zh) 2016-08-17
KR20140034790A (ko) 2014-03-20
SG194898A1 (en) 2013-12-30
MX351660B (es) 2017-10-23
KR101883910B1 (ko) 2018-08-01
HUE042378T2 (hu) 2019-06-28
CN103582653A (zh) 2014-02-12
US20160108139A1 (en) 2016-04-21
LT2707396T (lt) 2019-01-10
JP6205350B2 (ja) 2017-09-27
ES2705734T3 (es) 2019-03-26
AU2012252415A1 (en) 2013-11-28

Similar Documents

Publication Publication Date Title
US9908947B2 (en) Biotechnological sulphated chondroitin sulphate at position 4 or 6 on the same polysaccharide chain, and process for the preparation thereof
US8664196B2 (en) Shark-like chondroitin sulphate and process for the preparation thereof
AU2011369262B2 (en) Shark-like chondroitin sulphate and process for the preparation thereof
US10632144B2 (en) Low polydispersity, low molecular weight biotechnological chondroitin sulfate with anti-inflammatory and antiarthritis activity and use thereof in the prevention of osteoarthritis
CA2489870A1 (en) Low molecular weight oversulfated polysaccharide
US9718947B2 (en) Biotechnological sulphated chondroitin sulphate at position 4 or 6 on the same polysaccharide chain, and process for the preparation thereof
JP4462826B2 (ja) 骨疾患治療剤
NZ617564B2 (en) "biotechnological sulphated chondroitin sulphate at position 4 or 6 on the same polysaccharide chain, and process for the preparation thereof"
ITMI20120136A1 (it) Condroitin solfato biotecnologico a basso peso molecolare solfatato in posizione 4 o 6 sulla stessa catena polisaccaridica dotato di attivita' antiinfiammatoria e antiartritica e procedimento per la sua preparazione
ZA200410358B (en) Low molecular weight oversulfated polysaccharide.

Legal Events

Date Code Title Description
AS Assignment

Owner name: GNOSIS S.P.A., ITALY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BIANCHI, DAVIDE;VALETTI, MARCO;BAZZA, PAOLA;AND OTHERS;SIGNING DATES FROM 20131127 TO 20131203;REEL/FRAME:031715/0851

STCB Information on status: application discontinuation

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