US20090246187A1 - Process for concentration of a polypeptide - Google Patents

Process for concentration of a polypeptide Download PDF

Info

Publication number
US20090246187A1
US20090246187A1 US12/295,848 US29584807A US2009246187A1 US 20090246187 A1 US20090246187 A1 US 20090246187A1 US 29584807 A US29584807 A US 29584807A US 2009246187 A1 US2009246187 A1 US 2009246187A1
Authority
US
United States
Prior art keywords
polypeptide
composition
interest
colour
clarity
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/295,848
Other languages
English (en)
Inventor
Stefan Nilsson
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.)
Zymenex AS
Shire Pharmaceuticals Ireland Ltd
Original Assignee
Shire Pharmaceuticals Ireland Ltd
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 Shire Pharmaceuticals Ireland Ltd filed Critical Shire Pharmaceuticals Ireland Ltd
Publication of US20090246187A1 publication Critical patent/US20090246187A1/en
Assigned to ZYMENEX A/S reassignment ZYMENEX A/S ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: NILSSON, STEFAN
Assigned to SHIRE PHARMACEUTICALS IRELAND LIMITED reassignment SHIRE PHARMACEUTICALS IRELAND LIMITED ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ZYMENEX A/S
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/45Transferases (2)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/465Hydrolases (3) acting on ester bonds (3.1), e.g. lipases, ribonucleases
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/47Hydrolases (3) acting on glycosyl compounds (3.2), e.g. cellulases, lactases
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • 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
    • A61P25/02Drugs for disorders of the nervous system for peripheral neuropathies
    • 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
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/14Extraction; Separation; Purification
    • C07K1/16Extraction; Separation; Purification by chromatography
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/14Extraction; Separation; Purification
    • C07K1/34Extraction; Separation; Purification by filtration, ultrafiltration or reverse osmosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K1/00General methods for the preparation of peptides, i.e. processes for the organic chemical preparation of peptides or proteins of any length
    • C07K1/14Extraction; Separation; Purification
    • C07K1/36Extraction; Separation; Purification by a combination of two or more processes of different types
    • 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
    • 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
    • C12N9/10Transferases (2.)
    • C12N9/1085Transferases (2.) transferring alkyl or aryl groups other than methyl groups (2.5)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/16Hydrolases (3) acting on ester bonds (3.1)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y205/00Transferases transferring alkyl or aryl groups, other than methyl groups (2.5)
    • C12Y205/01Transferases transferring alkyl or aryl groups, other than methyl groups (2.5) transferring alkyl or aryl groups, other than methyl groups (2.5.1)
    • C12Y205/01061Hydroxymethylbilane synthase (2.5.1.61)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y301/00Hydrolases acting on ester bonds (3.1)
    • C12Y301/06Sulfuric ester hydrolases (3.1.6)
    • C12Y301/06001Arylsulfatase (3.1.6.1)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y302/00Hydrolases acting on glycosyl compounds, i.e. glycosylases (3.2)
    • C12Y302/01Glycosidases, i.e. enzymes hydrolysing O- and S-glycosyl compounds (3.2.1)
    • C12Y302/01024Alpha-mannosidase (3.2.1.24)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y302/00Hydrolases acting on glycosyl compounds, i.e. glycosylases (3.2)
    • C12Y302/01Glycosidases, i.e. enzymes hydrolysing O- and S-glycosyl compounds (3.2.1)
    • C12Y302/01046Galactosylceramidase (3.2.1.46)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y301/00Hydrolases acting on ester bonds (3.1)
    • C12Y301/06Sulfuric ester hydrolases (3.1.6)
    • C12Y301/06008Cerebroside-sulfatase (3.1.6.8)

Definitions

  • the present invention relates to a method for concentrating a polypeptide of interest, to the use of a composition comprising a concentrated polypeptide of interest as a medicament for subcutaneous injection and to a composition comprising at least 10 mg/ml polypeptide of interest.
  • polypeptides are useful as a medicament for the prevention and/or treatment of certain diseases.
  • the ability to inject a medicament subcutaneously is an advantage as it makes it easy for the patients to administer the medication to themselves.
  • medicaments which are to be administered subcutaneously that they are available in a high concentration so as to ensure that the patient receives an adequate amount of the medicament and/or to avoid multiple subcutaneous injections.
  • WO 99/37325 discloses methods of treating and preventing disease caused by absence or deficiency of the activity of enzymes belonging to the heme biosynthetic pathway.
  • WO 03/002731 discloses a process for purification of recombinant porphobilinogen deaminase on an industrial scale and to the use of the purified product for the preparation of a medicament.
  • WO 02/099092 and WO 2005/094874 provides lysosomal alpha-mannosidase and therapeutic use hereof.
  • WO 2005/073367 provides a process for purification of aryl sulfatase A and use of the enzyme in the treatment of metachromatic leukodystrophy.
  • the present invention relates to a method for concentrating a polypeptide of interest and to the use of a composition comprising a concentrated polypeptide of interest for the manufacture of a medicament for subcutaneous injection into mammal.
  • the present invention relates in one aspect to a method of concentrating a composition comprising a polypeptide of interest comprising:
  • the present invention relates to a composition comprising at least 10 mg/ml polypeptide of interest.
  • the present invention relates to use of a composition comprising 75-250 mg/ml polypeptide of interest for the manufacture of a medicament for subcutaneous injection into a mammal.
  • the present invention relates to a method of treating a mammal for Acute Intermittent Porphyria comprising injecting subcutaneously a composition of 500-300 mg/ml PBGD.
  • the present invention relates a method of treating a mammal for metachromatic leukodystrophy comprising subcutaneous injection of a composition of 50-300 mg/ml aryl sulfatase A.
  • the present invention relates a method of treating a mammal for the lysosomal storage disorder alpha-mannosidosis comprising subcutaneous injection of a composition of 50-300 mg/ml lysosomal alpha-mannosidase.
  • the present invention relates a method of treating a mammal for Krabbe disease comprising subcutaneous injection of a composition of 50-300 mg/ml galactosylcerebrosidase.
  • alignments of sequences and calculation of homology scores may be done using a full Smith-Waterman alignment, useful for both protein and DNA alignments.
  • the default scoring matrices BLOSUM50 and the identity matrix are used for protein and DNA alignments respectively.
  • the penalty for the first residue in a gap is ⁇ 12 for proteins and ⁇ 16 for DNA, while the penalty for additional residues in a gap is ⁇ 2 for proteins and ⁇ 4 for DNA.
  • Alignment may be made with the FASTA package version v20u6 (W. R. Pearson and D. 3. Lipman (1988), “Improved Tools for Biological Sequence Analysis”, PNAS 85:2444-2448, and W. R. Pearson (1990) “Rapid and Sensitive Sequence Comparison with FASTP and FASTA”, Methods in Enzymology, 183:63-98).
  • E. C.” Enzyme Class
  • enzyme Class refers to the internationally recognized enzyme classification system, Recommendations of the Nomenclature Committee of the International Union of Biochemistry and Molecular Biology, Academic Press, Inc.
  • amino acid sequences e.g. proteins, or nucleic acid sequences
  • amino acid sequences e.g. proteins, or nucleic acid sequences
  • Said sequence may be expressed by another organism using gene technology methods well known to a person skilled in the art. This also encompasses sequences which have been chemically synthesized. Furthermore, said sequences may comprise minor changes such as codon optimization, i.e. changes in the nucleic acid sequences which do not affect the amino acid sequence.
  • the polypeptide of the present invention may in particular be a hormone or hormone variant, an enzyme, a receptor or portion thereof, an antibody or portion thereof, an allergen or a reporter.
  • the polypeptide of interest may in particular be an enzyme selected from one of six major enzyme groups, such as an oxidoreductase (E.C. 1), a transferase (E.C. 2), a hydrolase (E.C. 3), a lyase (E.C. 4), an isomerase (E.C. 5), or a ligase (E.C. 6).
  • the polypeptide of interest may be an aminopeptidase, amylase, carbohydrase, carboxypeptidase, catalase, cellulase, cellobiohydrolase, chitinase, cutinase, cyclodextrin glycosyltransferase, deoxyribonuclease, endoglucanase, esterase, alpha-galactosidase, beta-galactosidase, glucoamylase, alpha-glucosidase, beta-glucosidase, invertase, laccase, lipase, mannosidase, mutanase, oxidase, pectinolytic enzyme, peroxidase, phospholipase, phytase, polyphenoloxidase, proteolytic enzyme, ribonuclease, transglutaminase, xylanase, or
  • the polypeptide of interest may in particular be a polypeptide which is useful as a medicament.
  • a suitable polypeptide of interest examples include but is not limited to one selected from the group consisting of a phorphobilinogen deaminase, an aryl sulfatase, an alpha-mannosidase and a galactocerebrosidase.
  • polypeptide of interest derivable from any source may be treated according to the methods of the present invention.
  • the polypeptide of interest may be of human origin.
  • the polypeptide may be of human origin as this may minimize the risk of unwanted allergic reactions.
  • Natural variations of human polypeptide due to e.g. polymorphism are in the context of the present invention included in the term “human origin”.
  • the polypeptide of interest may in particular be produced as a recombinant protein, i.e. a nucleotide sequence encoding the polypeptide of interest may be introduced into a cell for expression of the polypeptide of interest.
  • the recombinant expression may be homologous or heterologous, i.e. the polypeptide of interest may be expressed in cell which it is naturally expressed by (homologous expression) or it may be expressed by a cell which it is not naturally expressed by (heterologous expression).
  • the recombinant polypeptide of interest may be expressed by any cell suitable for recombinant production of the particular polypeptide of interest.
  • suitable cells include but are not limited to prokaryotic cells, such as an E. coli cell or a Bacillus cell.
  • suitable eukaryotic cells include but are not limited to a yeast cell or a mammalian cell such as a Chinese Hamster Ovary (CHO). Alternatively, it may be a human cell.
  • Suitable host cells for the expression of glycosylated polypeptide are derived from multicellular organisms.
  • invertebrate cells include plant and insect cells.
  • the host cell may also be a vertebrate cell, and propagation of vertebrate cells in culture (tissue culture) has become a routine procedure
  • recombinant polypeptide or “recombinant polypeptide of interest” denotes herein a recombinant produced polypeptide.
  • references to a particular polypeptide of interest includes in the context of the present invention also functionally equivalent parts or analogues of the polypeptide of interest.
  • the polypeptide of interest is an enzyme
  • a functionally equivalent part of the enzyme could be a domain or subsequence of the enzyme which includes the necessary catalytic site to enable the domain or subsequence to exert substantially the same enzymatic activity as the full-length enzyme or alternatively a gene coding for the catalyst.
  • substantially the same enzymatic activity refers to an equivalent part or analogue having at least 50%, preferably at least 60%, more preferably at least 70%, more preferably at least 75%, more preferably at least 80%, more preferably at least 85%, more preferably at least 90%, more preferably at least 95% and most preferably at least 97%, at least 98% or at least 99% of the activity of the natural enzyme.
  • An example of an enzymatically equivalent analogue of the enzyme could be a fusion protein which includes the catalytic site of the enzyme in a functional form, but it can also be a homologous variant of the enzyme derived from another species. Also, completely synthetic molecules that mimic the specific enzymatic activity of the relevant enzyme would also constitute “enzymatic equivalent analogues”.
  • the polypeptide of interest of the invention may be porphobilinogen deaminase, (also known as porphobilinogen ammonia-lyase (polymerizing)), E.C. 4.3.1.8. (Waldenström 1937, 3. Acta. Med. Scand. Suppl. 8). Porphobilinogen deaminase is the third enzyme in the heme biosynthetic pathway. E.C. 4.3.1.8 has been transferred to E.C. 2.5.1.61, so porphobilinogen deaminase (PBGD) is now placed under this E.C. number.
  • Porphobilinogen deaminase catalyzes the reaction of 4 porphobilinogen+H 2 O hydroxymethylbilane+4 NH 3 .
  • PBDG is important in relation to Acute intermittent porphyria (AIP), which is an autosomal dominant disorder in man caused by a defect (50% reduction of activity) of PBDG (see WO01/07065 for further details in relation to this).
  • AIP Acute intermittent porphyria
  • Porphobilinogen deaminase is in short known as PBGD and in the context of the present invention these two terms may be used inter-changeably with one another.
  • a host cell may in particular be a yeast cell or an E. coli cell.
  • rPBGD porphobilinogen deaminase
  • substantially the same enzymatic activity refers to an equivalent part or analogues enzyme having at least 50%, preferably at least 60%, more preferably at least 70%, more preferably at least 75%, more preferably at least 80%, more preferably at least 85%, more preferably at least 90%, more preferably at least 95% and most preferably at least 97%, at least 98% or at least 99% of the activity of natural human rhPBGD measured in the rhPBGD activity assay described in example 2 of WO 03/002731.
  • an enzymatically equivalent analogue of the enzyme could be a fusion protein which includes the catalytic site of the enzyme in a functional form, but it can also be a homologous variant of the enzyme derived from another species. Also, completely synthetic molecules that mimic the specific enzymatic activity of the relevant enzyme would also constitute “enzymatic equivalent analogues”.
  • PBGD An example of PBGD which may be used in the present invention includes any of those shown in Sequence 1-10 of the present application, or in Genebank no. X04217, X04808 or M95623.
  • polypeptide of interest may be an arylsulfatase A.
  • ASA has been purified from a variety of sources including human liver, placenta, and urine. It is an acidic glucoprotein with a low isoelectric point. Above pH 6.5, the enzyme exists as a dimer with a molecular weight of approximately 110 kDa. ASA undergoes a pH-dependent polymerisation forming an octamer at pH 4.5. In human urine, the enzyme consists of two nonidentical subunits of 63 and 54 kDa. ASA purified from human liver, placenta, and fibroblasts also consist of two subunits of slightly different sizes varying between 55 and 64 kDa.
  • ASA is synthesised on membrane-bound ribosomes as a glycosylated precursor. It then passes through the endoplasmic reticulum and Golgi, where its N-linked oligosaccharides are processed with the formation of phosphorylated and sulfated oligosaccharide of the complex type (Waheed A et al. Biochim Biophys Acta. 1985, 847, 53-61, Braulke T et al. Biochem Biophys Res Commun. 1987, 143, 178-185).
  • a precursor polypeptide of 62 kDa is produced, which translocates via mannose-6-phosphate receptor binding (Braulke T et al. 3 Biol. Chem. 1990, 265, 6650-6655) to an acidic prelysosomal endosome (Kelly B M et al. Eur J Cell Biol. 1989, 48, 71-78).
  • the arylsulfatase A may in particular be of human origin.
  • the length (18 amino acids) of the human ASA signal peptide is based on the consensus sequence and a specific processing site for a signal sequence. Hence, from the deduced human ASA cDNA (EMBL GenBank accession numbers J04593 and X521151) the cleavage of the signal peptide should be done in all cells after residue number 18 (Ala), resulting in the mature form of the human ASA.
  • rASA recombinant arylsulfatase A
  • mASA mature form of arylsulfatase A including the mature form of human ASA
  • mrhASA mature recombinant human ASA
  • a protein modification has been identified in two eukaryotic sulfatases (ASA and arylsulfatase B (ASB)) and for one from the green alga Volvox carteri (Schmidt B et al. Cell. 1995, 82, 271-278, Selmer T et al. Eur J Biochem. 1996, 238, 341-345).
  • This modification leads to the conversion of a cysteine residue, which is conserved among the known sulfatases, into a 2-amino-3-oxopropionic acid residue (Schmidt B et al. Cell. 1995, 82, 271-278).
  • the novel amino acid derivative is also recognised as C*-formylglycin (FGly).
  • Cys-69 is referred to the precursor ASA which has an 18 residue signal peptide.
  • cysteine residue is Cys-51.
  • the arylsulfatase A may in particular be a form of arylsulfatase A, which is capable of crossing the blood brain barrier and/or a form of rASA, which possesses specific tags for entry into target cells within the brain.
  • rASA a form of rASA, which possesses specific tags for entry into target cells within the brain.
  • it may be a rASA, which is efficiently endocytosed in vivo via the mannose-6-phosphate pathway.
  • the ASA may in particular be covalently bound to a so-called tag, peptides or proteins as vehicles or toxins as vehicles which are capable of increasing and/or facilitating transport of ASA over the blood-brain barrier and/or across cellular membranes in general (Schwarze et al., Trends Cell Biol. 2000; 10(7): 290-295; Lindgren et al., Trends Pharmacol. Sci. 2000; 21(3): 99-103).
  • An ASA molecule containing such peptide sequences can be produced by expression techniques.
  • the protein transduction process is not cell type specific and the mechanism by which it occurs is not fully elucidated, however, it is believed that it takes place by some sort of membrane perturbation and penetration process that is receptor independent. A partially unfolded state of the molecule may facilitate the process but is not essential.
  • An example of a suitable tag includes but is not limited to the mannose-6-phosphate tag.
  • peptides or proteins as vehicle include but are not limited to so-called protein-transducing domains.
  • suitable protein-transducing domains include but are not limited to those mentioned in WO 2005/073367, which is incorporated herein by reference.
  • the protein-transducing domain may be the 11 residue basic peptide from the HIV TAT protein -YGRKKRRQRRR (Schwarze et al., Trends Cell Biol. 2000; 10(7): 290-295), a synthetic version of TAT-YARAAARQARA that confers more alpha-helicity and amphipathic nature to the sequence (Ho et al., Cancer Res.
  • Suitable toxins as vehicles include but are not limited to those described in WO 2005/073367, which is incorporated herein by reference.
  • the ASA may in particular comprise a nucleic acid sequence, which encodes:
  • an amino acid sequence or a portion of an amino acid sequence which is a polypeptide capable of hydrolysing an amount of the arylsulfatase A substrate pNCS at 37° C. a rate corresponding to a specific activity of at least 20 U/mg polypeptide (preferably 50 U/mg polypeptide) when determined in an assay for measuring arylsulfatase A activity as described in example 1 of WO 2005/073367, and/or a polypeptide, which is capable of hydrolysing at least 40% of labelled arylsulfatase A substrate, fx. 14C palmitoyl sulfatide, loaded into MLD fibroblasts, when assayed by incubation at a dose level of 25 mU/ml in an assay as described in example 2 of WO 2005/073367.
  • the degree of sequence identity between the above mentioned nucleic acid sequence and SEQ ID NO: 1 of WO 2005/073367 is at least 80%, such as at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, or at least 99%. It may be equally preferred that the degree of sequence identity between the amino acid sequence encoded by the above mentioned nucleic acid sequence and SEQ ID NO: 2 WO 2005/073367 is at least 80%, such as at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, or at least 99%.
  • arylsulfatase A is a recombinant enzyme, particularly preferred is recombinant human arylsulfatase A (rhASA).
  • rASA is produced in a mammalian cell or cell line and that said mammalian cell or cell line produces a glycoform of rASA, which is efficiently endocytosed in vivo via the mannose-6-phosphate receptor pathway.
  • the preferred glycoform of rASA comprises an amount of exposed mannose-6-phosphate, which allows efficient endocytosis of rASA in vivo via the mannose-6-phosphate pathway.
  • At least one of the produced glycoforms of rASA is similar to a glycoform produced in CHO cells.
  • the post translational modification of the cysteine residue in position 51 in the mature human arylsulfatase A is relevant for the activity of the enzyme. Accordingly, in a preferred embodiment of the present invention production of the arylsulfatase A or its equivalent occurs at a rate and under conditions, which result in a product comprising an isoform of the enzyme in which the amino acid corresponding to Cys-69 in SEQ ID NO: 2 of WO 2005/073367 is converted to Formylglycine, corresponding to Fgly-51 in SEQ ID NO: 3 of WO 2005/073367.
  • SEQ ID NO: 4 of WO 2005/073367 represents mature human arylsulfatase A after cleavage of the 18 amino acid signal peptide but prior to modification of C-51.
  • the degree of sequence identity between the enzyme produced according to the invention and SEQ ID NO: 3 of WO 2005/073367 or SEQ ID NO: 4 of WO 2005/073367 is at least 80%, such as at least 85%, at least 90%, at least 95%, at least 97%, at least 98%, or at least 99%.
  • the ASA of the present invention may in terms of its structure be different from the rASA according to SEQ ID NO: 3 of 2005/073367. It may be an advantage that the sequence of amino acid residues surrounding the Cys-51 is identical or has a high degree of sequence identity to the corresponding sequence in SEQ ID NO: 3. Thus, it may be preferred that a linear sequence of 20 amino acids, such as 19, 18, 17, 16, 15, 14, 13, 12, 11, 10, 9, 8, 7, 6, 5 or 4 amino acid residues surrounding the Cys-51 in the arylsulfatase A is identical or at least 90% identical, such as 95%, 96%, 97%, 98%, or 99% identical to the corresponding sequence in SEQ ID NO: 3 of 2005/073367.
  • rASA As the active form of rASA within the lysosymes is an octamer the ASA of the present invention may in particular be a rASA which is an octamer or assembles into an octamer under physiological conditions.
  • the enzyme activity of ASA which is to be understood as the catalytic activity of the rASA, may be measured in an enzyme assay based on the rASA mediated hydrolysis of either a detectable substrate or a substrate, which leads to a detectable end product.
  • the assay is based on hydrolysis of the synthetic, chromogenic substrate, para-Nitrocatechol sulphate (pNCS) which has an end product, para-Nitrocatechol (pNC) that absorbs light at 515 nm.
  • the polypeptide of interest may be a lysosomal alpha-mannosidase (LAMAN).
  • Lysomal alpha-mannosidase belongs to EC 3.2.1.24 and is an exoglycosidase which hydrolyses the terminal, non-reducing alpha-D-mannose residues in alpha-D-mannosides from the non-reducing end during the ordered degradation of N-linked glycoproteins (Aronson and Kuranda FASEB J 3:2615-2622. 1989).
  • the term lysosomal alpha-mannosidase may be used interchangeably with the short term LAMAN.
  • the LAMAN of the present invention may in particular be of human origin.
  • the human enzyme is synthesised as a single polypeptide of 1011 amino acids with a putative signal peptide of 49 residues that is processed into three main glycopeptides of 15, 42, and 70 kD (Nilssen et al. Hum. Mol. Genet. 6, 717-726. 1997).
  • LAMAN The gene coding for LAMAN (MANB) is located at chromosome 19 (19cen-ql2), (Kaneda et al. Chromosoma 95:8-12. 1987). MANB consists of 24 exons, spanning 21.5 kb (GenBank accession numbers U60885-U60899; Riise et al. Genomics 42:200-207. 1997). The LAMAN transcript is >>3,500 nucleotides (nts) and contains an open reading frame encoding 1,011 amino acids (GenBank U60266.1).
  • the lysosomal alpha mannosidase comprises the amino acid sequence of SEQ ID NO.: 1 of WO 2005/094874.
  • the LAMAN of the present invention is produced recombinant.
  • recombinant production it may also be possible to obtain a preparation of the enzyme wherein a large fraction contains mannose-6-phosphate.
  • Recombinant production may be achieved after transfection of a cell using a nucleic acid sequence comprising the sequence of SEQ ID NO: 2 of WO 2005/094874.
  • the alpha-mannosidase is preferably made in a mammalian cell system as this will result in a glycosylation profile, which ensures efficient receptor mediated uptake in cells of for instance visceral organs of the body.
  • a glycosylation profile which ensures efficient receptor mediated uptake in cells of for instance visceral organs of the body.
  • production of the enzyme in CHO, COS or BHK cells ensures adequate post-translational modification of the enzyme by addition of mannose-6-phosphate residues.
  • a correct sialylation profile is obtained. Correct sialylation is known to be important in order to prevent uptake by the liver, because of exposed galactose residues.
  • the mammalian cell system is therefore selected from the group comprising CHO, COS cells or BHK cells (Stein et al. 3 Biol. Chem. 1989, 264, 1252-1259). It may further be preferred that the mammalian cell system is a human fibroblast cell line.
  • the mammalian cell system is a CHO cell line.
  • the lysosomal alpha-mannosidase may be a preparation of lysosomal alpha-mannosidase wherein a fraction of said preparation consists of lysosomal alpha mannosidase having one or more N-linked oligosaccharides carrying mannose 6-phosphate groups.
  • a fraction of a preparation of said lysosomal alpha-mannosidase is capable of binding to mannose 6-phosphate receptors.
  • the ability of the enzyme to bind to mannose-6-phosphate receptors may be determined in an in vitro assay as described in example 1 of WO 2005/094874.
  • binding of the enzyme to a MPR affinity 300 Matrix provides a measure of its ability to bind to mannose-6-phosphate receptors.
  • binding of the enzyme to mannose-6-phosphate receptors occurs in vitro.
  • this fraction corresponds to from 1 to 75% of the activity of a preparation of lysosomal alpha-mannosidase, such as from 2 to 70%, such as from 5 to 60%, such as from 10 to 50% such as from 15 to 45%, such as from 20 to 40%, such as from 30 to 35%.
  • the lysosomal alpha-mannosidase has a content of mannose 6-phosphate residues allowing mannose 6-phosphate dependent binding of from 2 to 100%, 5 to 95%, 10 to 90%, 20 to 80%, 30 to 70% or 40 to 60% of the amount of enzyme to a Man-6-P-receptor matrix.
  • the degree of phosphorylation has been analysed in several batches of enzyme and, typically, from 30 to 45% of the enzyme is phosphorylated and binds the affinity matrix.
  • a fraction constituting from 2-100%, 5-90%, 10-80%, 20-75%, 30-70%, 35-65% or 40-60% of the amount of said lysosomal alpha-mannosidase binds to the Man-6-P-receptor with high affinity.
  • two mannose 6-phosphate groups must be positioned close to each other in order for the enzyme to bind a Man-6-P-receptor with high affinity.
  • the distance between the phosphorylated mannose residues must be 40 ⁇ or less in order to obtain high affinity binding.
  • the two mannose 6-phosphate residues may be situated at the asparagines residues in positions 367 and 766. Accordingly, it is preferred that the medicament according to the present invention comprises lysosomal alpha-mannosidase, a fraction of which carries mannose 6-phosphate groups at both of these asparagine residues.
  • the alpha-mannosidase is made by recombinant techniques.
  • the alpha-mannosidase is of human origin (hLAMAN) and still more preferred a mature human alpha-mannosidase (mhLAMAN) or a fragment thereof. The fragment may be modified, however the active sites of the enzyme should be preserved.
  • one fraction of the enzyme is represented by its precursor form, while other fractions represent the proteolytically processed forms of approximately 55 and 70 kDa.
  • the polypeptide of interest may be a galactocerebrosidase, which may be shortended to GALC.
  • the GALC enzyme derived from humans is a glycosylated lysosomal enzyme comprising 643 amino acids and with a molecular weight of 72.8 kDa.
  • the GALC of the present invention may in particular be of human origin.
  • the GALC may be expressed recombinant in one of the previously mentioned host cells.
  • the host cell for recombinant expression of GALC may in particular be a CHO cell.
  • PBGD coding sequence 1 SEQ ID NO.: 1 PBGD coding sequence 2 SEQ ID NO.: 2 PBGD coding sequence 3 SEQ ID NO.: 3 PBGD coding sequence 4 SEQ ID NO.: 4 PBGD coding sequence 5 SEQ ID NO.: 5 PBGD coding sequence 6 SEQ ID NO.: 6 PBGD coding sequence 7 SEQ ID NO.: 7 PBGD coding sequence 8 SEQ ID NO.: 8 PBGD coding sequence 9 SEQ ID NO.: 9 PBGD coding sequence 10 SEQ ID NO.: 10 PBGD coding sequence, GenBank Acc. No. X04217 SEQ ID NO.: 11 PBGD coding sequence, GenBank Acc. No.
  • X04808 SEQ ID NO.: 12 PBGD coding sequence, GenBank Acc. No. M95623 SEQ ID NO.: 13 PBGD aa sequence from coding sequence, GenBank SEQ ID NO.: 14 Acc. No. M95623, Constitutive form PBGD aa sequence from coding sequence, GenBank SEQ ID NO.: 15 Acc. No. M95623, Erythropoietic form ASA coding sequence Genbank Acc. No.
  • polypeptide of interest comprises an amino acid selected from the group consisting of:
  • the analogue in iii) is at least 80% identical to a sequence as defined in i) or ii), such as at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or such as at least 99.5% identical to a sequence as defined in i) or ii).
  • polypeptide of interest may be obtained by recombinant expression using a nucleic acid sequence comprising a sequence selected from the group consisting of:
  • the acid sequence in ii) is at least 80% identical to a sequence as defined in i), such as at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or such as at least 99.5% identical to a sequence as defined in i).
  • composition Comprising a Polypeptide of Interest
  • the present invention also relates to a composition
  • a composition comprising at least 10 mg/ml polypeptide of interest, wherein the polypeptide of interest may be any polypeptide according to the present invention, such as in particular rhPBGD, aryl sulfatase, alpha-mannosidase or galactocerebrosidase.
  • Said composition may in particular comprise at least 25 mg/ml polypeptide of interest, such as at least 50 mg/ml or at least 75 mg/ml or at least 100 mg/ml polypeptide of interest.
  • composition may in particular comprise between 10-1000 mg/ml polypeptide of interest, such as between 10-500 mg/ml or between 10-300 mg/ml or between 10-200 mg/ml or between 25-500 mg/ml or between 25-400 mg/ml or between 40-400 mg/ml or between 40-300 mg/ml or between 50-400 mg/ml or between 50-300 mg/ml or between 75-400 mg/ml or between 75-300 mg/ml or between 100-200 mg/ml or between 100-150 mg/ml polypeptide of interest.
  • composition comprising a polypeptide of interest may in particular be an aqueous solution.
  • composition may in particular further comprise no aggregates of the polypeptide of interest or at least only very few aggregates.
  • amount of polypeptide of interest present as aggregates may in particular constitute less than 5 w/w % of the total amount of polypeptide of interest in the composition.
  • said aggregates may constitute less than 4 w/w %, such as less than 3 w/w %, or less than 2 w/w %, or less than 1 w/w %, or less than 0.5 w/w %, or less than 0.1 w/w % of the total amount of polypeptide of interest.
  • aggregates means any form of the polypeptide of interest which is not monomeric. Thus the term encompasses any dimer or multimer of the polypeptide of interest.
  • said composition comprises only the polypeptide of interest or at least only minor traces of other proteins, i.e. proteins different from polypeptide of interest.
  • said composition comprises less than 1 w/w %, such as less than 0.5 w/w %, or less than 0.1 w/w %, or less than 0.05 w/w %, or less than 0.01 w/w % other proteins than the polypeptide of interest.
  • a range of factors affect the stability and activity of polypeptides and the composition comprising a polypeptide of interest may therefore in particular be optimized to keep the polypeptide of interest as stable as possible.
  • the pH generally affects the stability of a polypeptide of interest, thus the pH of a composition comprising a polypeptide of interest may in particular be in the range of 7.5-8.5, such as in particular between pH 7.7-8.2, more particularly between pH 7.8-8.0 or between pH 7.85-7.95, such as pH 7.8 or pH 7.9. This may in particular be the case if the polypeptide of interest is PBGD.
  • composition comprising a polypeptide of interest may in particular comprise a buffer capable of keeping the composition within the described pH range.
  • buffers include but are not limited to TRIS-HCL, Na-Citrate and Na 2 HPO 4 .
  • concentration of such a buffer may depend on the choice of the particular buffer and the presence of other components in the composition.
  • the concentration of Na 2 HPO 4 may be in the range of 0.5-15 mM, such as in the range of 1-10 mM, or in the range of 1.5-7.5 mM, such as in the range of 1.83-7.4 mM, or in the range of 1.5-3 mM, such as in the range of 1.83-3.7 mM, or in the range of 1.83-2.45 mM, or in the range of 3.5-7.5 mM, such as in the range of 3.6-7.4 mM, or in the range of 5.4-7.4 mM, such as 1.84 mM, or 2.45 mM, or 3.67 mM or 5.51 mM or 7.34 mM.
  • the concentration of TRIS-HCL may in particular be in the range of 2-50 mM, such as 2-40 mM, or 2-30 mM, or 2-20 mM, or 2-10 mM, or 5-25 mM, or 5-20 mM, or 8-12 mM, or 9-11 mM, e.g. 10 mM.
  • suitable compounds include but are not limited to glycine, mannitol, sucrose, L-serine, Tween 80 or a combination of one or more of said compounds.
  • concentration of these compounds depend on the particular compound, but for glycine the concentration may in particular be in the range of 1-200 mM, such as in the range of 5-190 mM, or in the range of 10-180 mM, or in the range of 10-170 mM, or in the range of 20-160 mM, or in the range of 20-150 mM, or in the range of 25-125 mM, or in the range of 5-100 mM, or in the range of 5-90 mM, or in the range of 5-80 mM, or in the range of 5-70 mM, or in the range of 5-60 mM, or in the range of 10-100 mM, or in the range of 10-90 mM, or in the range of 10-80 mM, or in the range of 10-70 mM, or in the range of 10-60 mM, or in the range of 12-60 mM, or in the range of 12-55 mM, or in the range of 13.5-54 mM, or in the range of 10
  • the concentration of mannitol may in particular be in the range of 50-1000 mM, such as in the range of 50-900 mM, or in the range of 50-800 mM, or in the range of 50-700 mM, or in the range of 50-600 mM, or in the range of 100-900 mM, or in the range of 100-800 mM, or in the range of 100-700 mM, or in the range of 100-600 mM, or in the range of 100-500 mM, or in the range of 120-525 mM, or in the range of 125-500 mM, or in the range of 100-300 mM, such as in the range of 120-275 mM, or in the range of 120-170 mM, or in the range of 200-600 mM, such as in the range of 225-550 mM, or in the range of 240-510 mM, or in the range of 370-525 mM, such as 120, 125, 130
  • the concentration of sucrose may in particular be in the range of 1-200 mM, such as in the range of 5-190 mM, or in the range of 10-180 mM, or in the range of 10-170 mM, or in the range of 20-160 mM, or in the range of 20-150 mM, or in the range of 25-125 mM, or in the range of 5-100 mM, or in the range of 5-90 mM, or in the range of 5-80 mM, or in the range of 5-70 mM, or in the range of 5-60 mM, or in the range of 10-100 mM, or in the range of 10-90 mM, or in the range of 10-80 mM, or in the range of 10-70 mM, or in the range of 10-60 mM, or in the range 35 of 12-60 mM, or in the range of 12-55 mM, or in the range of 13.5-54 mM, or in the range of 10-30 mM, such as in the range of 13.
  • sucrose is included in a composition which also comprises mannitol
  • concentration of mannitol may in particular be lowered corresponding to the concentration of sucrose; i.e. the concentration of mannitol and sucrose together may in particular be the same as the concentration of mannitol if this was to be used alone.
  • the concentration of Tween 80 may in particular be in the range of 0.001-1 w/v %, such as in the range of 0.005-1 w/v %, or in the range of 0.01-1 w/v %, or in the range of 0.001-0.5 w/v %, or in the range of 0.005-0.5 w/v %, or in the range of 0.01-0.5 w/v %, or in the range of 0.05-0.4 w/v %, or in the range of 0.05-0.3 w/v %, or in the range of 0.05-0.2 w/v %, or in the range of 0.075-0.4 w/v %, or in the range of 0.075-0.3 w/v %, or in the range of 0.075-0.2 w/v %, or in the range of 0.09-0.2 w/v %, such as 0.075, 0.08, 0.09, 0.1, 0.125, 0.15, 0.175 or 0.2 w/v
  • composition comprising a polypeptide of interest, wherein the polypeptide in particular may be a PBGD, an aryl sulfatase, a lysosomal alpha-mannosidase or a galactocerebrosidase, may in particular comprise a combination of one or more of the above-mentioned compounds.
  • a suitable example of such a composition may be one which besides the polypeptide of interest comprises Na 2 HPO 4 , glycine and mannitol.
  • the pH of the composition and the concentration of the different compounds may be as described above.
  • composition may in one embodiment comprise 0.5-15 mM Na 2 HPO 4 , 1-200 mM glycine, 50-1000 mM mannitol and a pH in the range of 7.5-8.5.
  • concentrations of compounds and pH are encompassed by the present invention.
  • a specific example of a suitable combination of other compounds and pH in the composition comprising a polypeptide of interest is one which comprises 3.67 mM Na 2 HPO 4 , 27 mM glycine, 250 mM mannitol and has a pH in the range of 7.7 to 7.9.
  • compositions include, but are not limited to any of the following:
  • composition comprising a polypeptide of interest may in particular be used for therapeutic applications in mammals.
  • the composition comprising a polypeptide of interest may in particular be isotonic with regard to the tissue of mammals, e.g. it may in particular have an osmolality in the range of 200-400 mOsm/kg, such as in the range of 250-350 mOsm/kg or in the range of 275-325 mOsm/kg or in the range of 295-305 mOsm/kg, such as 295 mOsm/kg or 300 mOsm/kg or 305 mOsm/kg.
  • the method of the present invention comprises the steps of a) centrifugation and/or filtration of a composition comprising a polypeptide of interest and b) concentrating the composition from step a).
  • the inventors of the present invention have found that by centrifugation and/or filtrating a composition comprising a polypeptide of interest prior to concentrating said composition it is possible to obtain a composition comprising a highly concentrated polypeptide of interest without any or with at least only few aggregates of the polypeptide of interest. Furthermore, it is generally an advantage for therapeutic applications of a polypeptide that the amount of polypeptide aggregates is reduced, e.g. as they may increase the risk of eliciting an immune response towards the polypeptide.
  • polypeptide composition For administration of a polypeptide subcutaneously it is an advantage that the polypeptide composition has a high activity in a small volume as only small volumes can be injected subcutaneously.
  • Proteins or polypeptides may in general form aggregates when they are concentrated. Thus it is an advantage that when the method of the present invention is used to concentrate a polypeptide of interest it does not cause a high rate of polypeptide aggregate formation. As shown in the examples the amount of PBGD aggregates in the composition obtained by the concentration method of the present invention is similar to that of a non-concentrated PBGD composition.
  • step a) of the method is performed prior to step b).
  • the inventors of the present invention have found that prior to concentrating a composition comprising a polypeptide of interest it is an advantage to pre-treat the composition by centrifugation and/or filtration of the composition as by this pre-treatment many or most of the polypeptide aggregates are removed.
  • the concentration of the composition in step b) is performed by a method which relies on the use of a filter or membrane, such as ultrafiltration, the presence of aggregates may block the filter or membrane so that small molecules and liquid are not able to cross the filter or membrane. This may decrease the speed by which the composition is concentrated and/or completely block any further concentration.
  • the pre-treatment according to step a) is an advantage as removal of the aggregates makes it possible to obtain compositions of a polypeptide of interest which are more concentrated than if said composition were not been pre-treated.
  • the pre-treatment in step a) has the advantage that it reduces the amount of aggregates present in the concentrated composition.
  • Step a) may be performed by one of the following three alternatives:
  • step a) comprises both centrifugation and filtration it is an advantage to perform the centrifugation prior to the filtration as the inventors of the present invention have found that the centrifugation removes most of large aggregates and the filtration subsequently removes the remaining smaller aggregates.
  • composition comprising a polypeptide of interest may be centrifuged at a force in the range of 1500-3000 g, such as in the range of 1800-2500 g, or in the range of 2000-2300 g.
  • composition may be centrifuged for 10-60 minutes, such as for 15-50 minutes or for 20-40 minutes.
  • the centrifugation may be performed at a temperature in the range of 2-20° C., such as from 3-15° C. or in the range of 3-10° C., or in the range of 3-8° C., such as at 4° C. or 5° C. or 6° C.
  • the centrifugation results in that the polypeptide of interest aggregates sediment, i.e. they form a pellet, while the individual polypeptide of interest molecules stays in the solution. So it is the supernatant of the centrifuged composition which is subsequently used in the method of the present invention.
  • composition comprising a polypeptide of interest may be filtered through a filter having a pore-size in the range of 0.20-5 ⁇ m, such as in the range of 0.2-2.5 ⁇ m.
  • Suitable membrane filters include but are not limited to polyethersulfone (PES), cellulose acetate, regenerated cellulose and polyvinylidene flouride (PVDF).
  • PES polyethersulfone
  • PVDF polyvinylidene flouride
  • the polypeptide of interest may generally be present in the retentate. Hence it is generally the retentate from the filtration which is used in the subsequent steps of the present invention.
  • any method of concentrating the polypeptide of interest composition may be used in step b) of the present invention.
  • Examples of such suitable methods include but are not limited to ultrafiltration and concentration by removal of water.
  • Ultrafiltration is a separation method in which hydraulic pressure is used to force molecules and solvent across a membrane comprising pores of a particular size, also known as the cut-off size of value. Only molecules which have a molecular weight smaller than the cut-off value of the membrane are able to cross the membrane while those with a larger molecular weight do not cross the membrane and form the so called retentate. The molecules present in the retentate are thereby concentrated as the solvent flows across the membrane.
  • the concentration of the solution or composition comprising a polypeptide of interest may be performed by Tangential flow filtration (TFF).
  • TMF Tangential flow filtration
  • the TFF technique is based on the use of a particular apparatus which causes the solution which is to be filtrated to flow across a semi-permeable membrane; only molecules which are smaller than the membrane pores will pass through the membrane, forming the filtrate, leaving larger matter to be collected (retentate).
  • two different pressures are applied; one to pump the solution into the system and to circulate it in the system (inlet pressure), and another pressure is applied over the membrane (membrane pressure) to force the small molecules and the solvent across the membrane.
  • the inlet pressure may typically be in the range of 1-3 bar, such as between 1.5-2 bar.
  • the membrane pressure may typically be larger than 1 bar.
  • the concentrated composition of a polypeptide of interest may be collected as the retentate when TFF is used to concentrate the composition.
  • Membranes useful for TFF may typically be made of regenerated cellulose or polyethersolufone (PES).
  • the pore-size of the membrane may typically have a molecular weight cut-off which is smaller than 10.000 Mw, such as in the range of 10-10.000 Mw.
  • the concentration of the composition comprising a polypeptide of interest may be performed by the use of a centrifugal device.
  • the principle of this method is that the solution is filtrated over a membrane by the application of a centrifugal force over the membrane.
  • Such membranes are often characterized by a molecular weight (Mw) cut-off, i.e. this is the maximum molecular size of compounds which are able to cross the membrane and compound with a molecular size larger than this will not cross the membrane.
  • Mw cut-off of the membranes used in the present invention may in particular be smaller than 30.000 Mw, such as between 10-30.000 Mw.
  • the membrane may in particular be made of polyethersulfone (PES) or regenerated cellulose.
  • Examples of such suitable commercial filter devices may be Centricon Plus-80 or Centricon Plus-15.
  • the concentration may typically be performed by centrifugation at 2000-4500 g, such as between 2500-4000 g, or between 2750-3500 g, or between 3000-3500 g, such as at 3000 g or 3100 g or 3200 g or 3300 g or 3400 g or 3500 g.
  • centrifugation may be run for several hours, e.g. for more than one hour, such as for 1-10 hours.
  • the centrifugation may in particular be performed at a temperature in the range of 2-20° C., such as in the range of 3-15° C. or in the range of 3-10° C. or in the range of 3-6° C.
  • the principle of concentration by removal of water is usually that all, or most, of the water is removed to obtain a solid, and then subsequently diluting or dissolving this solid in a volume of water which is less than what it was previously diluted or dissolved in. However, it may in principle be performed by just removing the necessary amount of water to obtain the desired concentration without subsequently re-diluting or re-dissolving the compound.
  • Examples of suitable methods of concentrating by removal of water include freeze-drying and evaporation.
  • the method of freeze-drying may be comprise the following three or four steps; a freezing-phase, a primary drying phase and a secondary drying phase and optionally a step of annealing after the freezing phase. Freeze-drying may in particular be performed as described with regard to freeze-drying included as a further step of the method of the present invention.
  • the polypeptide of interest may derive from a natural source, i.e. from cells naturally expressing the polypeptide of interest, or it may in particular be expressed recombinant.
  • polypeptide of interest may have been purified before being subjected to a method of the present invention.
  • composition comprising a polypeptide of interest comprises less than 5 w/w %, or less than 1 w/w % or less 0.5 w/w % or less than 0.1 w/w % or less than 0.05 w/w % or less than 0.01 w/w % other proteins than the polypeptide of interest.
  • proteins which are expressed by e.g. a host cell may be removed from the composition comprising a polypeptide of interest before it is used in a method of the present invention.
  • the method of the present invention may comprise one or more of following steps prior to step a):
  • Recombinant expression of a polypeptide of interest may in particular be performed as described previously with regard to the polypeptide of interest.
  • PBGD polypeptide of interest
  • suitable types of chromatography include but are not limited to affinity chromatography, Ion Exchange Chromatography (IEC) and chromatography on a hydroxyapatite column. In principle any combination of these chromatography methods may be used.
  • the inventors of the present invention have previously found for PBGD that it is an advantage to perform at least the step of affinity chromatography and if this is combined with any of the other methods of chromatography it is an advantage to perform the step of affinity chromatography prior to the other chromatography steps (see e.g. WO 03/002731).
  • affinity chromatography columns include affinity coupling, group specific affinity, and metal chelate affinity columns.
  • Metal chelate affinity columns are specially preferred for purifying proteins via metal ion complex formation with exposed histidine groups.
  • Example 3 of WO01/07065 describes construction of a recombinant human Porphobilinogen deaminase with a “His-Tag” (rhPBGD-His).
  • rhPBGD-His a recombinant human Porphobilinogen deaminase with a “His-Tag”
  • a metal chelate affinity column such as a column having a cobalt metal affinity resin.
  • Examples of other suitable methods of affinity chromatography include but are not limited to columns having porcine heparin as ligand or columns having 1-Amino-4-[[4-[[4-chloro-6-[[3 (or 4)-sulfophenyl]amino]-1,3,5-triazin-2-yl]amino]-3-sulfophenyl]amino]-9,10-dihydro-9,10-dioxo-2-anthracenesulfonic acid, also known as Cibracon Blue 3G, as ligand and using Triazine coupling as the ligand coupling method.
  • a commercially available example of the latter is Blue Sepharose 6 Fast Flow (FF) from Amersham Pharmacia Biotech.
  • a preferred embodiment of the invention relates to the process, as described herein, wherein the affinity chromatography column of step (i) is a column using a triazine coupling as ligand coupling method, and more preferably wherein the ligand is Cibacron Blue 3G.
  • IEC Ion Exchange Chromatography
  • IEC columns are columns such as a Q Sepharose column, a Q SP Sepharose column, or a CM Sepharose column, it may in particular be a DEAE Sepharose column.
  • hydroxyapatite column is a ceramic hydroxyapatite column.
  • Hydroxyapatite (Ca 5 (PO 4 ) 3 OH) 2 is a form of calcium phosphate that can be used for the separation and purification of proteins, enzymes, nucleic acids, viruses, and other macromolecules.
  • Ceramic hydroxyapatite is a spherical, macroporous form of hydroxyapatite.
  • CHT Type I Bio-Rad
  • the method of the present invention may comprise the following steps prior to step a):
  • the method of the present invention may comprise the following steps prior to step a):
  • Both of these methods may optionally include a further step of dilution of diafiltration of the PBGD composition obtained from step ii).
  • step should be after step ii) and before iii), i.e. a step iia).
  • Step iia) has the purpose of reducing the concentration of salts to suitable conductivity, e.g. ⁇ 10 mS/cm. This may in particular be relevant if DEAE Sepharose is used as resin in the ion exchange chromatography step, i.e. step iii), as this may facilitate binding of the captured PBGD to the DEAE Sepharose resin.
  • Dilution may be obtained by addition of purified water directly or by ultrafiltration against purified water.
  • PBGD PBGD
  • step i) The recombinant expression of PBGD, step i) may be performed by any of the methods described above.
  • Suitable affinity chromatography columns in step ii) may be any of the above mentioned.
  • Examples of suitable methods of performing ion exchange chromatography in step iii) may be any of the above mentioned.
  • Suitable hydroxyapatite chromatography columns in step iv) may be any of the above mentioned.
  • the affinity chromatography column may be a column using a triazine coupling as ligand coupling method, and in particular such a method wherein the ligand is Cibracon Blue 3G.
  • This may in particular be a Blue Sepharose 6 Fast Flow column, and the ion exchange chromatography column may be DEAE Sepharose column, and in the embodiment wherein the method also comprises a step iv) this column may in particular be a ceramic hydroxyapatite column.
  • the method of the present invention may also comprise further steps after step b) of the method. Such steps include but are not limited to one or more of the following:
  • a suitable freeze-dryer includes but is not limited to a Lyostar (FTM-systems) freeze-drier as used the examples of the present invention, where the solutions comprising a concentrated polypeptide of interest, i.e. in this case PBGD, were filled in 2 and 6 ml injection glass vials (type 1) and stoppered with rubber stoppers (chlorobutyl).
  • the freeze-drying may be performed by the following three steps;
  • Step i) freezing may in particular be performed by first loading a sample in ambient temperature and cooling it to 0° C. and keeping it at 0° C. for 30 minutes, before lowering the temperature by 1° C. per minute to ⁇ 40° C. and keeping it at ⁇ 40° C. for 30 minutes.
  • Step ii) primary drying may in particular be performed by drawing the vacuum pressure 126 mTorr, raising the temperature by 1° C. per minute to 0° C. and keeping the sample at 0° C. for 360 minutes
  • Step iii) secondary drying may in particular be performed by drawing the full vacuum simultaneously with raising the temperature by 0.5° C. per minute to +30° C. and keeping the sample at +30° C. for 360 minutes. After the secondary drying the sample may further be closed under vacuum or closed after filling with nitrogen.
  • An example of a suitable freeze-drying method includes the one described in the examples of the present invention.
  • the freeze-drying may in further embodiment comprise an annealing step prior to the primary drying phase.
  • the inventors of the present invention have found that inclusion of an annealing step in the freeze-drying method improves the visual appearance, as visualised by fewer cracks, and/or results in a shorter reconstitution time of the freeze-dried product compared to when the same method of freeze-drying is used but without the annealing step. It is an advantage that the time for reconstitution of a freeze-dried product is reduced, especially if it is to be used as a pharmaceutical which is administered as a solution. An improved visual appearance is usually also regarded as an advantage for most products.
  • freeze-drying may comprise the following steps:
  • the freezing, primary drying and secondary drying steps may in particular be performed as described above.
  • the annealing step, i.e. step ii) may in particular be performed by after 30 minutes of freezing, raising the temperature at e.g. a rate of 2° C. per minute to ⁇ 10° C. or ⁇ 20° C. and keeping this temperature for 120 or 420 minutes and then lowering the temperature e.g. a rate of 2° C. per minute to ⁇ 40° C. at which temperature the sample may be kept at 60-90 minutes before start of the step of primary drying.
  • Changing the buffer of the composition comprising a concentrated polypeptide of interest may in particular be performed by a) diluting, e.g. 5-15 times, the composition comprising a concentrated polypeptide of interest in a buffer or formulation, b) concentrating the diluted composition again and performing the steps a) and b) a sufficient number of times so that amount of the excipients in the buffer or formulation present in the composition before these steps constitute less than e.g. 5 v/v % or less than 1 v/v % of excipients in the buffer or formulation present in said composition after said steps were performed.
  • composition comprising a polypeptide of interest obtained from step b) of the present invention may in particular further comprise a step of sterile filtration of said composition and/or a step of freeze-drying the composition.
  • Sterile filtration is generally performed by filtration of the composition through a filter with a pore-size of 0.22 ⁇ m or 0.20 ⁇ m. Freeze-drying may in particular be performed as described above.
  • the present invention also relates to a freeze-dried composition obtained by a method of the present invention.
  • the present invention also relates to the use of a composition comprising in the range of 50-300 mg/ml polypeptide of interest for the manufacture of a medicament for subcutaneous injection into a mammal.
  • the polypeptide of interest may be any polypeptide of interest according to the present invention, including but not limited to PBGD, aryl sulfatase A, lysosomal alpha-mannosidase and galactocerebrosidase.
  • subcutaneous is often shortened to s.c. and the two terms may be used interchangeably in the context of the present invention.
  • the volume of the composition comprising a polypeptide of interest which needs to be administered to the patient and of the concentration of polypeptide of interest in said composition.
  • the composition comprising a polypeptide of interest comprises a high concentration of the polypeptide of interest and that this high concentration of the polypeptide of interest can be obtained without the formation of large amounts of polypeptide aggregates.
  • concentrated polypeptide of interest compositions makes it possible to inject a smaller volume of said composition and at the same time ensure that the patient receives an adequate amount of the polypeptide of interest; thus making it easier to administer the polypeptide of interest subcutaneously.
  • composition comprising a polypeptide of interest may in particular comprise between 75-250 mg/ml, such as between 75-200 mg/ml or between 75-150 mg/ml or between 100-150 mg/ml or between 100-125 mg/ml or between 125-150 mg/ml of polypeptide of interest.
  • the volume of composition comprising a polypeptide of interest which it is necessary to inject into the patient to ensure that the patient receives an adequate amount of the polypeptide of interest correlates with the concentration of the polypeptide of interest in said composition.
  • the volume of such a composition will generally be adjusted according to the concentration of the polypeptide of interest in the composition.
  • the volume may generally be in the range of 0.1-1.5 ml, such as in the range of 0.1-1.5 ml or in the range of 0.5-1.5 ml or in the range of 0.5-1.5 ml or in the range of 0.75-1.5 ml or in the range of 0.75-1.5 ml or in the range of 1-1.5 ml or in the range of 1-1.5 ml.
  • the amount of polypeptide of interest which it is relevant to administer to a patient generally depends on the weight of the individual and the particular polypeptide of interest.
  • the present invention relates to a method of treating a mammal for Acute Intermittent Porphyria comprising subcutaneous injection of a composition of 50-300 mg/ml PBGD.
  • PBGD may in particular be useful for the treatment of Acute Intermittent Porphyria.
  • administration of PBGD also may be useful for the treatment of other porphyrias, such as Hereditary coproporphyria or Variegata porphyria.
  • Porphyria is a term used to collectively describe a number of diseases caused by different deficiencies in the heme biosynthetic pathway.
  • administration of PBGD e.g. in combination with other therapeutics, to a patient suffering from any type of porphyria may help to increase the overall turnover of the different intermediates in the pathway.
  • the present invention relates to a method of treating a mammal for metachromatic leukodystrophy comprising subcutaneous injection of a composition of 50-300 mg/ml aryl sulfatase A.
  • Metachromatic leukodystrophy is caused by an autosomal recessive genetic defect in the lysosomal enzyme Arylsulfatase A (ASA), resulting in a progressive breakdown of membranes of the myelin sheath (demyelination) and accumulation of galactosyl sulphatide (cerebroside sulphate) in the white matter of both the central nervous system (CNS) and the peripheral nervous system.
  • ASA autosomal recessive genetic defect in the lysosomal enzyme Arylsulfatase A
  • galactosyl sulphatide cerebroside sulphate
  • CNS central nervous system
  • galactosyl sulphatide forms spherical granular masses that stain metachromatically.
  • Galactosyl sulphatide also accumulates within the kidney, gallbladder, and certain other visceral organs and is excreted in excessive amounts in the urine.
  • Galactosyl sulfatide is normally metabolised by the hydrolysis of 3-O-sulphate linkage to form galactocerebroside through the combined action of the lysosomal enzyme arylsulfatase A (EC 3.1.6.8) (Austin et al. Biochem J. 1964, 93, 15C-17C) and a sphingolipid activator protein called saposin B.
  • arylsulfatase A EC 3.1.6.8
  • sphingolipid activator protein called saposin B.
  • ASA sphingolipid activator protein
  • the present invention relates to a method of treating a mammal for the lysosomal storage disorder alpha-mannosidosis comprising subcutaneous injection of a composition of 50-300 mg/ml lysosomal alpha-mannosidase.
  • Alpha-mannosidosis is a recessive, autosomal disease that occurs world wide with a frequency of between 1/1.000.000 and 1/500.000. Mannosidosis is found in all ethnic groups in Europe, America, Africa and also Asia. It is detected in all countries with a good diagnostic service for lysosomal storage disorders, at a similar frequency. They are born apparently healthy; however the symptoms of the diseases are progressive. Alpha-mannosidosis displays clinical heterogeneity, ranging from very serious to very mild forms.
  • Typical clinical symptoms are: mental retardation, skeletal changes, impaired immune system resulting in recurrent infections, hearing impairment and often the disease is associated with a typical facial characteristics such as a coarse face, a prominent forehead, a flattened nasal bridge, a small nose, and a broad mouth.
  • a typical facial characteristics such as a coarse face, a prominent forehead, a flattened nasal bridge, a small nose, and a broad mouth.
  • mannosidosis type I the children suffer from hepatosplenomegaly, and they die during the first years of life. Possibly this early death is caused by severe infections due to the immunodeficiency caused by the disease.
  • severe cases mannosidosis type 2
  • the patients In milder cases (mannosidosis type 2) the patients usually reach adult age.
  • the skeletal weaknesses of the patients result in the needs of wheeling chairs at age 20 to 40.
  • the disease causes a diffuse dysfunction of the brain often resulting in weak mental performances that excludes anything but the most basic skills of simple reading and writing. These problems associated with hearing inabilities and other clinical manifestations preclude the patient from an independent life, the consequence being that life long caretaking is needed.
  • the present invention relates to a method of treating a mammal for Krabbe disease comprising subcutaneous injection of a composition of 50-300 mg/ml galactosylcerebrosidase.
  • a deficiency in the GALC enzyme results in an autosomal inherited genetic Lysosomal Storage disease known as Krabbe disease or Globoid Cell Leukodystrophy.
  • the enzyme is generally expressed in the testis, kidneys, placenta, liver and brain of human beings and a deficiency in the GALC enzyme generally results in a disorder in the myelin metabolism and in the central and peripheral nervous systems (the CNS and PNS, respectively).
  • compositions comprising a polypeptide of interest also apply to the other aspects of the invention.
  • all of the embodiments described for the composition comprising a polypeptide of interest such as the presence of further compounds, buffers and pH also apply to the composition comprising a polypeptide of interest used in the present applications.
  • rhPBGD used in the following experiments were obtained according to process 2 in example 1 of WO 03/002731, where process 2 is the process which includes step 1V, i.e. the ceramic hydroxyapatite chromatography step.
  • the recombinant and purified rhPBGD was present in the following aqueous formulation buffer:
  • the formulation buffer was then sterile-filtered trough a 0.22 ⁇ m filter.
  • the freeze-drying of the purified rhPBGD solutions were performed in a Lyostar (FTM-systems) freeze-drier according to the following schedule:
  • Freezing phase 0° C. 30 min 760 Torr 0° C. to ⁇ 40° C. 1° C./min 760 Torr ⁇ 40° C. 30 min 760 Torr Primary drying ⁇ 40° C. to 0° C. 1° C./min 169 mTorr 0° C. 240 min 169 mTorr Secondary drying 0° C. to 30° C. 10° C./60 min, 20 mTorr 180 min 30° C. 720 min 20 mTorr
  • the pH-meter (Metrohm 691 pH Meter) and electrode (combined LL pH electrode) were calibrated with 3 standard reference solutions (Merck) in the range 4.00 to 9.00. The liquid was finally analysed.
  • Protein concentration in extract, in-process samples, bulk drug substance and final product was determined by a method that utilizes principles of the reduction of Cu2+ to Cu+ by protein in an alkaline medium (the Biuret reaction).
  • the Cu+ ions were then reacted with a reagent containing bicinchoninic acid resulting in a highly sensitive and selective colorimetric detection.
  • rhPBGD Porphobilinogen Deaminase
  • rhPBGD variants were separated according to their ability to adsorb and desorb to silica based stationary media depending on the percentage of organic modifier (acetonitrile) in the mobile phase.
  • Porphobilinogen deaminase catalyzes the addition of 4 molecules of porphobilinogen (PBG) to form a linear tetramer, preuroporphyrinogen, which is released from the enzyme and in vivo circularized to uroporphyrinogen III by the action of Uroporphyrinogen III synthase.
  • PBG porphobilinogen
  • Preuroporphyrinogen can be chemically oxidized with benzoquinone to form uroporphyrin, which absorbs light at 405 nm.
  • rhPBGD freeze-dried rhPBGD was resuspended in 1.00 ml MilliQ-water. The vial of frozen aqueous solution of rhPBGD was thawed. The osmometer (Vapro osmometer) was calibrated with 3 standard solutions in the range 100-1000 mOsm/kg (100, 290, 1000 mOsm/kg). The liquid was then analyzed.
  • PBGD-bulk solution (7 mg/mL rhPBGD, 3.67 mM Na 2 HPO 4 , 27 mM glycine, 250 mM Mannitol, pH 7.9) was thawed in a water-bath at 20° C., centrifuged at 3200 g for 10 min and thereafter sterile-filtrated by 0.20 ⁇ m-PES filters (Nalgene Polyethersulfone filters).
  • the PBGD-bulk solution was concentrated to 100 mg/ml by running the Centrifugal Filter Devices Centricon Plus-80 (Mw cut-off 30000) and Centricon Plus-15 (Mw cut-off 30000) at 3200 g for several hours.
  • the concentrated solution i.e.
  • the retentate was sterile-filtrated by 0.22 ⁇ m-filters (Millex GV) and finally a part of this solution was diluted with sterile formulation buffer to get 50 mg/ml.
  • the 5 mg/ml-solution was prepared by directly diluting the recombinant and purified hPBGD with sterile formulation buffer.
  • the 5 mg/mL, 50 mg/mL and 100 mg/mL rhPBGD were then freeze-dried as described above.
  • Several vials of each the above-mentioned freeze-dried rhPBGD solutions with 5, 50 and 100 mg/mL rhPBGD and of the aqueous 5 mg/mL rhPBGD solution were stored at 40° C. ⁇ 2° C., 75% ⁇ 5% relative humidity (RH).
  • the vials were stored protected from light in a well sealed secondary package (paper box).
  • PBGD-bulk solution (7 mg/mL rhPBGD, 3.67 mM Na 2 HPO 4 , 27 mM glycine, 250 mM Mannitol, pH 7.9) was thawed in a water-bath at 20° C., centrifuged at 3200 g for 10 min and thereafter sterile-filtrated by 0.20 ⁇ m-PES filters (Nalgene Polyethersulfone filters).
  • the PBGD-bulk solution was concentrated to 100 mg/ml by running the Centrifugal Filter Devices Centricon Plus-80 (Mw cut-off 30000) and Centricon Plus-15 (Mw cut-off 30000) at 3200 g for several hours.
  • the concentrated solution i.e.
  • the retentate was sterile-filtered by 0.22 ⁇ m-filters (Millex GV) and diluted with sterile filtered formulation buffer (see above) to get solutions of lower concentrations. A fraction in volume of each concentration was freeze-dried as described above.
  • the different concentrations of freeze-dried rhPBGD and aqueous solution of rhPBGD were stored at 5° C. ⁇ 3° C. or at ⁇ 20° C. ⁇ 5° C. (ambient relative humidity (RH)). All vials were stored protected from light in a well-sealed secondary package (paper box).
  • a vial of each freeze-dried samples were resuspended in 1.00 mL Millipore water and then tested together with the aqueous solution of rhPBGD by visually observing the colour, clarity and precipitates, and by measuring pH, protein concentration, purity, osmolality and rhPBGD activity.
  • the thawed solution was then centrifuged with 200 mL conical centrifuge tubes for approximately 10 minutes at 2200 g.
  • the concentration by TFF was performed with a Millipore Labscale TFF System and Millipore Pellicon® XL Filter with a pump inlet pressure of approximately 20-25 psi and a pressure over the Pellicon® XL Filter of approximately 4-6 psi.
  • the rhPBGD was protected from light during the procedure by covering the sample container of the TFF System by sheets of aluminium foil.
  • the concentrated rhPBGD solution obtained from the TFF procedure was then buffer-changed against a formulation buffer containing 3.67 mM Na 2 HPO 4 ⁇ 2H 2 O, 27 mM glycin and 220 mM Mannitol prepared in sterile water. This was performed by continuously adding said buffer to the TFF-system and pressing it across the membrane until said buffer has replaced the previous buffer.
  • the concentrated and buffer-changed rhPBGD solution was then sterile filtered by passing it through a filter with a pore-size of 0.22 ⁇ m. This sterile filtration was performed twice with a new filter each time.
  • the sterile concentrated rhPBGD solution was then placed in vials before it was freeze-dried as described in the method section.
  • PBGD was concentrated as described in example 3 and after the exchange of the buffer was the concentration of PBGD determined.
  • the concentrated PBGD solution was then freeze-dried in a Lyostar (FTM-systems) freeze-dryer.
  • the solutions were filled in 2 and 6 ml injection glass vials (type 1) and stoppered with rubber stoppers (chlorobutyl).
  • the samples were loaded in ambient temperature and the shelves were cooled down to 0° C. for 30 minutes. The temperature were lowered to ⁇ 40° C. (1° C. per minute) and held there for 30 minutes and then the vacuum pressure was drawn to 126 mTorr and the primary drying began by raising the temperature to 0° C. (1° C. per minute). After 360 minutes of primary drying the temperature was raised to +30° C. (0.5° C. per minute) and full vacuum was drawn simultaneously (start of secondary drying). The temperature was held at +30° C. for 360 minutes and the vials were then stoppered under vacuum.
  • the temperature was raised with a rate of 2° C. per minute to ⁇ 10° C. or ⁇ 20° C. at which temperature they were kept for 120 or 420 minutes before the temperature was lowered again with 2° C. per minute to ⁇ 40° C. were the samples were kept for 60-90 minutes before start of primary drying.
  • 1 ⁇ amount of excipients refers to that the PBGD solution comprises 3.67 mM Na 2 HPO 4 ⁇ 2H 2 O, 27 mM glycin and 220 mM Mannitol prepared in sterile water.
  • 1.5 ⁇ amount excipients refers to that the PBGD solution comprises 5.51 mM Na 2 HPO 4 ⁇ 2H 2 O, 40.5 mM glycin and 375 mM Mannitol prepared in sterile water, i.e. 1.5 ⁇ of each of the components present in the 1 ⁇ buffer.
  • 2 ⁇ excipients refers to that the PBGD solution comprises 7.34 mM Na 2 HPO 4 ⁇ 2H 2 O, 54 mM glycin and 500 mM Mannitol prepared in sterile water, i.e. 2 ⁇ of each of the components present in the 1 ⁇ buffer.
  • the original freeze-drying cycle is as described above.
  • the annealing freeze-drying cycle is as described above where the annealing step comprises raising the temperature to ⁇ 10° C. at keeping the sample at this temperature for 120 minutes before lowering it to ⁇ 40° C. again.
  • the extended annealing freeze-drying cycle is as described above where the annealing step comprises raising the temperature to ⁇ 20° C. at keeping the sample at this temperature for 420 minutes before lowering it to ⁇ 40° C. again.
  • freeze-dried products were prepared from a 150, 175 and 200 mg/ml, respectively, rhPBGD solution.
  • the freeze-dried products each comprised 1.5 ⁇ amount of excipients and they were freeze-dried with the annealing cycle. None of the freeze-dried products comprised any cracks.
  • Two freeze-dried rhPBGD products were prepared from a 150 mg/ml rhPBGD solution. One of them comprised 1 ⁇ amount of excipients and was prepared according to the original freeze-drying cycle, while the other comprised 1.5 ⁇ amount of excipients and was prepared according to the extended annealing free-drying cycle.
  • the product comprising 1.5 ⁇ amount of excipients and prepared according to the extended annealing freeze-drying cycle comprised fewer cracks than the product comprising 1 ⁇ amount of excipients and prepared according to the original freeze-drying cycle.
  • rhPBGD products Two freeze-dried rhPBGD products were prepared from a 150 mg/ml rhPBGD solution. One of them comprised 1 ⁇ amount of excipients and was prepared according to the original freeze-drying cycle, while the other comprised 0.1% Tween 80 in combination with the 1 ⁇ amount of excipients and was prepared according to the extended annealing freeze-drying cycle.
  • the product comprising the 0.1% Tween 80 in combination with the 1 ⁇ amount of excipients and which was prepared according to the extended annealing freeze-drying cycle comprised fewer cracks than the product which comprised 1 ⁇ amount of excipients and which was prepared according to the original freeze-drying cycle.
  • the “bulk solution” is a concentrated solution of PBGD before freeze-drying.
  • Table 21 shows the results of rhPBGD solutions having the following characteristics with regard to the concentration of rhPBGD, amount of excipients (were the same definitions as in example 4 are used), the mode of freeze-drying (were the same definitions as in example 4 are used) and the ratio of the filling volume (fill. Vol which is the volume of the composition before it is freeze-dried) versus the recovery volume (Rec. vol which is the volume in which the freeze-dried product is resuspended):
  • rhPBGD was concentrated as described in example 4 and then the buffer was changed as to one of the four buffers described below. The products were then freeze-dried as described in example 4 with an original annealing step included and the stability of the samples were tested as described in example 6.
  • Formulation A (corresponds to solution 9 in example 6): 250 mM mannitol, 27 mM glycine and 3.67 mM Na 2 HPO 4 .
  • Formulation B 250 mM mannitol, 27 mM glycine and 10 mM TRIS-HCL.
  • Formulation C 250 mM mannitol, 27 mM glycine, 3.67 mM Na 2 HPO 4 and 0.1% Tween 80.
  • Formulation D 221 mM mannitol, 29 mM sucrose, 27 mM glycine, 3.67 mM Na 2 HPO 4 and 0.1% Tween 80.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Medicinal Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Biochemistry (AREA)
  • Zoology (AREA)
  • Wood Science & Technology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • General Engineering & Computer Science (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Molecular Biology (AREA)
  • Biomedical Technology (AREA)
  • Epidemiology (AREA)
  • Immunology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Biophysics (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Analytical Chemistry (AREA)
  • Biotechnology (AREA)
  • Microbiology (AREA)
  • Water Supply & Treatment (AREA)
  • Psychiatry (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Hospice & Palliative Care (AREA)
  • Enzymes And Modification Thereof (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)
US12/295,848 2006-04-04 2007-04-04 Process for concentration of a polypeptide Abandoned US20090246187A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
DKPA200600488 2006-04-04
DKPA200600488 2006-04-04
DKPA200600922 2006-07-05
DKPA200600922 2006-07-05
PCT/DK2007/000177 WO2007112757A2 (en) 2006-04-04 2007-04-04 A process for concentration of a polypeptide

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/DK2007/000177 A-371-Of-International WO2007112757A2 (en) 2006-04-04 2007-04-04 A process for concentration of a polypeptide

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US13/094,321 Division US8809055B2 (en) 2006-04-04 2011-04-26 Process for concentration of a polypeptide
US15/041,841 Division US9713634B2 (en) 2006-04-04 2016-02-11 Process for concentration of a polypeptide

Publications (1)

Publication Number Publication Date
US20090246187A1 true US20090246187A1 (en) 2009-10-01

Family

ID=38472949

Family Applications (6)

Application Number Title Priority Date Filing Date
US12/295,848 Abandoned US20090246187A1 (en) 2006-04-04 2007-04-04 Process for concentration of a polypeptide
US13/094,321 Active US8809055B2 (en) 2006-04-04 2011-04-26 Process for concentration of a polypeptide
US14/321,642 Abandoned US20140314735A1 (en) 2006-04-04 2014-07-01 Process for concentration of a polypeptide
US15/041,841 Active US9713634B2 (en) 2006-04-04 2016-02-11 Process for concentration of a polypeptide
US15/473,460 Abandoned US20170202927A1 (en) 2006-04-04 2017-03-29 Process for concentration of a polypeptide
US15/585,916 Abandoned US20180289777A1 (en) 2006-04-04 2017-05-03 Process for concentration of a polypeptide

Family Applications After (5)

Application Number Title Priority Date Filing Date
US13/094,321 Active US8809055B2 (en) 2006-04-04 2011-04-26 Process for concentration of a polypeptide
US14/321,642 Abandoned US20140314735A1 (en) 2006-04-04 2014-07-01 Process for concentration of a polypeptide
US15/041,841 Active US9713634B2 (en) 2006-04-04 2016-02-11 Process for concentration of a polypeptide
US15/473,460 Abandoned US20170202927A1 (en) 2006-04-04 2017-03-29 Process for concentration of a polypeptide
US15/585,916 Abandoned US20180289777A1 (en) 2006-04-04 2017-05-03 Process for concentration of a polypeptide

Country Status (20)

Country Link
US (6) US20090246187A1 (pl)
EP (4) EP2628746B1 (pl)
JP (6) JP2009532394A (pl)
KR (3) KR20140057678A (pl)
CN (1) CN105233276A (pl)
AU (2) AU2007234195B2 (pl)
BR (1) BRPI0709737A2 (pl)
CA (3) CA2632528C (pl)
DK (1) DK2628746T3 (pl)
ES (3) ES2928096T3 (pl)
HU (1) HUE042402T2 (pl)
IL (4) IL194267A (pl)
MX (1) MX2008012748A (pl)
NO (3) NO347673B1 (pl)
NZ (7) NZ588903A (pl)
PL (3) PL2100898T3 (pl)
PT (1) PT2628746T (pl)
SI (1) SI2628746T1 (pl)
WO (1) WO2007112757A2 (pl)
ZA (2) ZA200805346B (pl)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011163650A3 (en) * 2010-06-25 2013-06-13 Shire Human Genetic Therapies, Inc. Methods and compositions for cns delivery of arylsulfatase a
US9283181B2 (en) 2010-06-25 2016-03-15 Shire Human Genetic Therapies, Inc. CNS delivery of therapeutic agents
US9320711B2 (en) 2010-06-25 2016-04-26 Shire Human Genetic Therapies, Inc. Methods and compositions for CNS delivery of heparan N-sulfatase
US9713634B2 (en) 2006-04-04 2017-07-25 Shire Pharmaceuticals Ireland Limited Process for concentration of a polypeptide
US9814764B2 (en) 2010-06-25 2017-11-14 Shire Human Genetic Therapies, Inc. Treatment of sanfilippo syndrome type b by intrathecal administration of alpha-n-acetylglucosaminidase
US9957489B2 (en) 2004-01-30 2018-05-01 Shire Pharmaceuticals Ireland Limited Production and purification of recombinant arylsulfatase A
US10336992B2 (en) 2011-07-08 2019-07-02 Shire Human Genetic Therapies, Inc. Methods for purification of arylsulfatase A
US10660944B2 (en) 2011-12-23 2020-05-26 Shire Human Genetic Therapies, Inc. Stable formulations for CNS delivery of arylsulfatase A
US11407984B2 (en) 2013-01-09 2022-08-09 Takeda Pharmaceutical Company Limited Methods for purification of arysulfatase A

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8974780B2 (en) * 2010-02-24 2015-03-10 Zymenex A/S Process for production and purification of recombinant lysosomal alpha-mannosidase
EP2920198B1 (en) * 2012-11-13 2019-09-25 Chiesi Farmaceutici S.p.A. Purification of recombinant human galactocerebroside beta-galactosidase (rhgalc)
CA2893609A1 (en) * 2012-12-11 2014-06-19 Centogene Ag Method for the diagnosis of metachromatic leukodystrophy
TWI788044B (zh) 2013-03-15 2022-12-21 美商百歐維拉提夫治療公司 因子ix多肽調配物
EA038573B1 (ru) * 2014-03-24 2021-09-16 Биовератив Терапьютикс Инк. Лиофилизированный состав, содержащий фактор ix, для предупреждения приступов кровотечения
EP3676375A4 (en) 2017-08-31 2021-04-07 Green Cross Corporation METHOD OF PURIFYING A SULFATASE PROTEIN
KR102290596B1 (ko) * 2020-09-10 2021-08-19 주식회사 파이안바이오테크놀로지 분리된 미토콘드리아를 포함하는 주사용 조성물 및 이의 용도

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6197507B1 (en) * 1996-01-15 2001-03-06 Thomas Berg Genetic test for α-mannosidosis
US20030199073A1 (en) * 2001-06-07 2003-10-23 Hemebiotech A/S Production of recombinant human lysosomal alpha-mannosidase
US20040126370A1 (en) * 2001-09-28 2004-07-01 St. Jude Children's Research Hospital, Inc. Targeting proteins to cells expressing mannose receptors via expression in insect cells
US6812339B1 (en) * 2000-09-08 2004-11-02 Applera Corporation Polymorphisms in known genes associated with human disease, methods of detection and uses thereof
US20080003211A1 (en) * 2004-01-30 2008-01-03 Zymenex A/S Production and Purification of Recombinant Arylsulftase

Family Cites Families (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2041280A1 (en) 1990-05-10 1991-11-11 Yosuke Sawada Arylsulfatase
DK1049487T3 (da) 1998-01-27 2002-09-16 Hemebiotech As Behandling af akut intermitterende porfyri og andre porfyriske lidelser
IL140155A (en) * 1998-06-09 2005-12-18 Statens Seruminstitut Process for producing immunoglobulins for intravenous administration and other immunoglobulin products
DK2270044T3 (en) * 1998-06-09 2015-01-26 Csl Behring Ag Liquid immunoglobulin G (IgG) product
US6537777B1 (en) 1998-07-27 2003-03-25 Hemebiotech A/S Human porphobilinogen deaminase sequences
AUPQ026799A0 (en) * 1999-05-10 1999-06-03 Csl Limited Method of increasing protein stability by removing immunoglobulin aggregates
JP2003505057A (ja) 1999-07-27 2003-02-12 ヘム−バイオテック エイ/エス rhPBGDの製造及び急性間欠性ポルフィリン症(AIP)及び他のポルフィリン症の患者を処置するための新規な治療方法
CA2429210A1 (en) 2000-11-15 2002-05-23 Jason C. Goodrick Expression system for recombinant proteins
WO2002098455A2 (en) * 2001-06-07 2002-12-12 Hemebiotech A/S Production of recombinant human arylsulfatase a
EP1402014A1 (en) * 2001-06-29 2004-03-31 Hemebiotech A/S A process for purification of recombinant porphobilinogen deaminase
EP1463512B1 (en) 2002-01-11 2014-05-28 biOasis Technologies Inc. Use of p97 as an enzyme delivery system for the delivery of therapeutic lysosomal enzymes
WO2003066669A2 (en) * 2002-02-04 2003-08-14 Millipore Corporation Process for removing protein aggregates and virus from a protein solution
CH694697A5 (de) 2004-01-29 2005-06-15 Koelbl Engineering Und Consult Gummimehl-enthaltende Elastomerlegierungen.
DK1740204T3 (en) 2004-04-01 2018-05-22 Chiesi Farm Spa MEDICAL USE OF ALFA MANNOSIDASE
US20060051347A1 (en) * 2004-09-09 2006-03-09 Winter Charles M Process for concentration of antibodies and therapeutic products thereof
NZ588903A (en) * 2006-04-04 2012-08-31 Zymenex As A process for concentration of galactosycerebrosidase or porphobuilinogen deaminase

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6197507B1 (en) * 1996-01-15 2001-03-06 Thomas Berg Genetic test for α-mannosidosis
US6812339B1 (en) * 2000-09-08 2004-11-02 Applera Corporation Polymorphisms in known genes associated with human disease, methods of detection and uses thereof
US20030199073A1 (en) * 2001-06-07 2003-10-23 Hemebiotech A/S Production of recombinant human lysosomal alpha-mannosidase
US20040126370A1 (en) * 2001-09-28 2004-07-01 St. Jude Children's Research Hospital, Inc. Targeting proteins to cells expressing mannose receptors via expression in insect cells
US7232670B2 (en) * 2001-09-28 2007-06-19 St. Jude Children's Research Hospital Targeting proteins to cells expressing mannose receptors via expression in insect cells
US20080003211A1 (en) * 2004-01-30 2008-01-03 Zymenex A/S Production and Purification of Recombinant Arylsulftase

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Michael et al., Pharmaceutical Research, Vol. 19, No. 4, April 2002 *

Cited By (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9957489B2 (en) 2004-01-30 2018-05-01 Shire Pharmaceuticals Ireland Limited Production and purification of recombinant arylsulfatase A
US9713634B2 (en) 2006-04-04 2017-07-25 Shire Pharmaceuticals Ireland Limited Process for concentration of a polypeptide
EP2631242B1 (en) * 2006-04-04 2022-08-03 Takeda Pharmaceutical Company Limited A process for concentration of a polypeptide
US10456454B2 (en) 2010-06-25 2019-10-29 Shire Human Genetic Therapies, Inc. CNS delivery of therapeutic agents
US9320711B2 (en) 2010-06-25 2016-04-26 Shire Human Genetic Therapies, Inc. Methods and compositions for CNS delivery of heparan N-sulfatase
KR102094094B1 (ko) 2010-06-25 2020-03-27 샤이어 휴먼 지네틱 테라피즈 인크. 아릴설파타제 a의 cns 전달을 위한 방법들 및 조성물들
AU2011270668A8 (en) * 2010-06-25 2017-02-09 Takeda Pharmaceutical Company Limited CNS delivery of therapeutic agents
KR20200035167A (ko) * 2010-06-25 2020-04-01 샤이어 휴먼 지네틱 테라피즈 인크. 아릴설파타제 a의 cns 전달을 위한 방법들 및 조성물들
US9770410B2 (en) 2010-06-25 2017-09-26 Shire Human Genetic Therapies, Inc. Methods and compositions for CNS delivery of arylsulfatase A
US9814764B2 (en) 2010-06-25 2017-11-14 Shire Human Genetic Therapies, Inc. Treatment of sanfilippo syndrome type b by intrathecal administration of alpha-n-acetylglucosaminidase
US9283181B2 (en) 2010-06-25 2016-03-15 Shire Human Genetic Therapies, Inc. CNS delivery of therapeutic agents
KR20180059580A (ko) * 2010-06-25 2018-06-04 샤이어 휴먼 지네틱 테라피즈 인크. 아릴설파타제 a의 cns 전달을 위한 방법들 및 조성물들
RU2671503C2 (ru) * 2010-06-25 2018-11-01 Шир Хьюман Дженетик Терапис, Инк. Способы и композиции для доставки в цнс арилсульфатазы а
AU2017202289B2 (en) * 2010-06-25 2019-03-14 Takeda Pharmaceutical Company Limited Methods and compositions for cns delivery of arylsulfatase a
US10646554B2 (en) 2010-06-25 2020-05-12 Shire Human Genetic Therapies, Inc. Methods and compositions for CNS delivery of arylsulfatase A
KR101997935B1 (ko) 2010-06-25 2019-10-04 샤이어 휴먼 지네틱 테라피즈 인크. 아릴설파타제 a의 cns 전달을 위한 방법들 및 조성물들
WO2011163650A3 (en) * 2010-06-25 2013-06-13 Shire Human Genetic Therapies, Inc. Methods and compositions for cns delivery of arylsulfatase a
AU2011270668B2 (en) * 2010-06-25 2017-01-12 Takeda Pharmaceutical Company Limited CNS delivery of therapeutic agents
AU2011270670B2 (en) * 2010-06-25 2017-01-12 Takeda Pharmaceutical Company Limited Methods and compositions for CNS delivery of arylsulfatase A
KR102537084B1 (ko) 2010-06-25 2023-05-26 샤이어 휴먼 지네틱 테라피즈 인크. 아릴설파타제 a의 cns 전달을 위한 방법들 및 조성물들
US11471516B2 (en) 2010-06-25 2022-10-18 Shire Human Genetic Therapies, Inc. CNS delivery of therapeutic agents
US11065307B2 (en) 2010-06-25 2021-07-20 Shire Human Genetic Therapies, Inc. Therapeutic fusion protein comprising an alpha-n-acetylglucosaminidase and a lysosomal targeting moiety
US11065308B2 (en) 2010-06-25 2021-07-20 Shire Human Genetic Therapies, Inc. Methods and compositions for CNS delivery of heparan n-sulfatase
AU2019204051B2 (en) * 2010-06-25 2021-07-22 Takeda Pharmaceutical Company Limited Methods and compositions for cns delivery of arylsulfatase a
KR102318997B1 (ko) * 2010-06-25 2021-10-29 샤이어 휴먼 지네틱 테라피즈 인크. 아릴설파타제 a의 cns 전달을 위한 방법들 및 조성물들
KR20210131449A (ko) * 2010-06-25 2021-11-02 샤이어 휴먼 지네틱 테라피즈 인크. 아릴설파타제 a의 cns 전달을 위한 방법들 및 조성물들
US11260112B2 (en) 2010-06-25 2022-03-01 Shire Human Genetic Therapies, Inc. Methods and compositions for CNS delivery of iduronate-2-sulfatase
KR20130135821A (ko) * 2010-06-25 2013-12-11 샤이어 휴먼 지네틱 테라피즈 인크. 아릴설파타제 a의 cns 전달을 위한 방법들 및 조성물들
US10336992B2 (en) 2011-07-08 2019-07-02 Shire Human Genetic Therapies, Inc. Methods for purification of arylsulfatase A
US10660944B2 (en) 2011-12-23 2020-05-26 Shire Human Genetic Therapies, Inc. Stable formulations for CNS delivery of arylsulfatase A
US11407984B2 (en) 2013-01-09 2022-08-09 Takeda Pharmaceutical Company Limited Methods for purification of arysulfatase A
US11884945B2 (en) 2013-01-09 2024-01-30 Takeda Pharmaceutical Company Limited Methods for purification of arylsulfatase A

Also Published As

Publication number Publication date
EP2004672B1 (en) 2015-08-19
CA2882501A1 (en) 2007-10-11
JP2009532394A (ja) 2009-09-10
JP2014051503A (ja) 2014-03-20
IL241654A0 (en) 2015-11-30
BRPI0709737A2 (pt) 2011-07-26
NO20211305A1 (no) 2008-10-31
IL194267A (en) 2015-10-29
EP2628746A3 (en) 2014-03-19
CA2644642C (en) 2016-01-26
ES2744499T3 (es) 2020-02-25
JP2009273469A (ja) 2009-11-26
EP2628746A2 (en) 2013-08-21
WO2007112757A2 (en) 2007-10-11
NO346368B1 (no) 2022-06-27
KR20090018894A (ko) 2009-02-24
ES2928096T3 (es) 2022-11-15
US20170202927A1 (en) 2017-07-20
EP2631242B1 (en) 2022-08-03
EP2631242A3 (en) 2013-10-02
IL194267A0 (en) 2011-08-01
KR101504969B1 (ko) 2015-03-24
IL211905A0 (en) 2011-05-31
US20180289777A1 (en) 2018-10-11
SI2628746T1 (sl) 2019-04-30
NO20084776L (no) 2008-10-31
KR20140057678A (ko) 2014-05-13
PL2631242T3 (pl) 2023-03-20
ZA200805346B (en) 2018-11-28
HUE042402T2 (hu) 2019-06-28
NZ597548A (en) 2013-08-30
IL241654B (en) 2019-02-28
US9713634B2 (en) 2017-07-25
EP2004672A2 (en) 2008-12-24
NO347673B1 (no) 2024-02-19
NO20082510L (no) 2008-10-31
EP2100898A1 (en) 2009-09-16
JP2014058521A (ja) 2014-04-03
JP2016104000A (ja) 2016-06-09
KR20150103339A (ko) 2015-09-09
AU2008229659A1 (en) 2008-10-23
CN105233276A (zh) 2016-01-13
NZ582045A (en) 2011-07-29
JP5878152B2 (ja) 2016-03-08
PL2100898T3 (pl) 2019-11-29
NZ588903A (en) 2012-08-31
US20120076767A1 (en) 2012-03-29
CA2632528A1 (en) 2007-10-11
IL235982A0 (en) 2015-01-29
PT2628746T (pt) 2019-04-23
AU2007234195A1 (en) 2007-10-11
NZ623123A (en) 2015-04-24
JP5384232B2 (ja) 2014-01-08
EP2631242A2 (en) 2013-08-28
PL2628746T3 (pl) 2019-07-31
CA2632528C (en) 2022-07-12
NZ607595A (en) 2014-10-31
EP2100898B1 (en) 2019-06-19
NZ568728A (en) 2011-09-30
AU2008229659B2 (en) 2013-01-31
ZA200809540B (en) 2019-09-25
MX2008012748A (es) 2009-01-07
US8809055B2 (en) 2014-08-19
CA2644642A1 (en) 2007-10-11
US20160193304A1 (en) 2016-07-07
CA2882501C (en) 2021-09-07
EP2628746B1 (en) 2019-01-09
NZ571610A (en) 2011-07-29
US20140314735A1 (en) 2014-10-23
JP2013236633A (ja) 2013-11-28
ES2713488T3 (es) 2019-05-22
WO2007112757A3 (en) 2008-02-28
DK2628746T3 (en) 2019-04-01
AU2007234195B2 (en) 2013-02-07

Similar Documents

Publication Publication Date Title
US9713634B2 (en) Process for concentration of a polypeptide
EP1713904B1 (en) Production and purification of recombinant arylsulfatase a
US8343487B2 (en) Large-scale production of soluble hyaluronidase
DK1740204T3 (en) MEDICAL USE OF ALFA MANNOSIDASE
CA2956469A1 (en) Mannose-6-phosphate bearing peptides fused to lysosomal enzymes

Legal Events

Date Code Title Description
AS Assignment

Owner name: SHIRE PHARMACEUTICALS IRELAND LIMITED, IRELAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:ZYMENEX A/S;REEL/FRAME:025669/0795

Effective date: 20110118

Owner name: ZYMENEX A/S, SWEDEN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:NILSSON, STEFAN;REEL/FRAME:025669/0776

Effective date: 20110118

STCB Information on status: application discontinuation

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