WO2002057435A2 - Bifunctional fusion proteins with glucocerebrosidase activity - Google Patents

Bifunctional fusion proteins with glucocerebrosidase activity Download PDF

Info

Publication number
WO2002057435A2
WO2002057435A2 PCT/EP2001/015328 EP0115328W WO02057435A2 WO 2002057435 A2 WO2002057435 A2 WO 2002057435A2 EP 0115328 W EP0115328 W EP 0115328W WO 02057435 A2 WO02057435 A2 WO 02057435A2
Authority
WO
WIPO (PCT)
Prior art keywords
gcr
fusion protein
molecule
protein
dna
Prior art date
Application number
PCT/EP2001/015328
Other languages
French (fr)
Other versions
WO2002057435A3 (en
Inventor
Silke Schumacher
Stephen Gillies
Original Assignee
Merck Patent Gmbh
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to PL01362394A priority Critical patent/PL362394A1/en
Priority to BR0116803-7A priority patent/BR0116803A/en
Priority to CA002435037A priority patent/CA2435037A1/en
Priority to KR10-2003-7009521A priority patent/KR20030067755A/en
Priority to HU0401300A priority patent/HUP0401300A3/en
Priority to MXPA03006294A priority patent/MXPA03006294A/en
Application filed by Merck Patent Gmbh filed Critical Merck Patent Gmbh
Priority to JP2002558488A priority patent/JP2004525621A/en
Priority to US10/466,593 priority patent/US20040043457A1/en
Priority to EP01989057A priority patent/EP1392826A2/en
Publication of WO2002057435A2 publication Critical patent/WO2002057435A2/en
Priority to NO20033247A priority patent/NO20033247D0/en
Publication of WO2002057435A3 publication Critical patent/WO2002057435A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
    • 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
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • 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)
    • C12N9/24Hydrolases (3) acting on glycosyl compounds (3.2)
    • C12N9/2402Hydrolases (3) acting on glycosyl compounds (3.2) hydrolysing O- and S- glycosyl compounds (3.2.1)
    • 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)
    • C12Y302/01045Glucosylceramidase (3.2.1.45), i.e. beta-glucocerebrosidase
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/05Animals comprising random inserted nucleic acids (transgenic)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto

Definitions

  • GCR fusion proteins By selective altering of the amino acid sequences of the Ig moiety, GCR fusion proteins with improved properties, e.g. enhanced stability, can be obtained. Furthermore, fusion proteins can be provided, wherein shortened versions of GCR and the Ig chain are used.
  • the present invention relates also to pharmaceutical compositions and therapeutic methods and systems comprising such GCR fusion proteins and methods of treating Gaucher's disease or another disease caused by glycolipid storage disorders, such as Fabry's and Tay-Sachs disease, comprising administering to a subject afflicted with this disease, a pharmaceutical composition comprising a therapeutic amount of recombinantly produced GCR fusion protein in a pharmaceutically acceptable carrier.
  • the dose of the enzyme to treat these diseases is about 60 units per kilogram body weight every two weeks, that means that the average costs per year for the treatment of a 70 kg patient are about US$ 380.000,- for the enzyme alone. This is due to the short intracellular half-life of exogenous acid ⁇ -glucosidase.
  • Preferred truncated forms are for example those which consist of about one third to one half of the amino acid sequence of the natural GCR enzyme truncated from the carboxy terminal site of the enzyme.
  • Those truncated proteins may be derived from the full length protein by cleaving off the desired chain with a suitable reagent such as a restriction enzyme or the like.
  • GCR m is an GCR according to this invention which is mutated but not truncated in its amino acid sequence.
  • the number of mutations is not limited but is restricted to the loss of the biological activity of the molecule. In a preferred embodiment the degree of mutation is between 5 and 30 per cent, in a especially preferred embodiment between 5 and 20 per cent of the amino acid residues.
  • Variants with increased GCR biological activity can be generated by procedures described known in the art.
  • L has the meaning of a series of peptides such as. e.g., glycine and/or serine.
  • the peptide linker is a mixed series of glycine and serine peptides about 5 - 25, preferably 10 - 20 residues in length.
  • proteolytically cleavable linkers especially linkers which are cleavable by lysosomal proteases like cathepsins.
  • the Ig moiety is specific for a cell bearing an Fc receptor.
  • a preferred fragment of an Ig molecule to be linked to GCR is the Fc region.
  • the Fc region of an immunoglobulin is the amino acid sequence for the carboxyl-terminal portion of an immunoglobulin heavy chain constant region. The Fc regions are particularly important in determining the biological functions of the immunoglobulin and these biological functions are termed effector functions.
  • the heavy chains of the immunoglobulin subclasses comprise four or five domains: IgM and IgE have five heavy chain domains, and IgA, IgD and IgG have four heavy chain domains.
  • Fc portion means the carboxyl-terminal portion of an immunoglobulin heavy chain constant region, or an analog or portion thereof. That is, e.g., an immunoglobulin Fc region of Ig, preferably IgG, which may comprise at least a portion of a hinge region, a CH2 domain, and a CH3 domain.
  • IgG histidins located at the junction between the CH2 and CH3 domains (residues 310 and 433) of the IgG heavy chain contribute to the pH-dependent binding to the FcRn receptor (Raghavan, et al., Biochemistry 34(45): 14649-57 (1995)). Also lie 253 and His 435 and 436 (Kim et al., Eur. J. Immunol.
  • a typical signal peptide consists of three regions: a basic N-terminal region, a central hydrophobic region, and a more polar
  • compositions comprising effective amounts of protein or derivative products of the invention together with pharmaceutically acceptable diluents, preservatives, solubilizers, emulsifiers, adjuvants and/or carriers.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • General Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Biochemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Diabetes (AREA)
  • General Engineering & Computer Science (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Gastroenterology & Hepatology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Obesity (AREA)
  • Hematology (AREA)
  • Epidemiology (AREA)
  • Immunology (AREA)
  • Microbiology (AREA)
  • Biotechnology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Biomedical Technology (AREA)
  • Endocrinology (AREA)
  • Emergency Medicine (AREA)
  • Biophysics (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)
  • Enzymes And Modification Thereof (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

The present invention relates to novel Glucocerebrosidase bifunctional fusion proteins consisting essentially of an Immunoglobulin (Ig) molecule and a protein having the biological activity of Glucocerebrosidase, for enzyme replacement therapy and/or augmentation of glycolipid metabolism by the administration of bifunctional fusion proteins using a therapy based on the treatment of glycolipid storage disorders such as Gaucher's, Fabry's and Tay-Sachs diseases.

Description

Bifunctional Fusion Proteins with Glucocerebrosidase activity
Field of the invention
The present invention relates to Glucocerebrosidase (GCR) bifunctional fusion proteins (GCR fusion proteins) consisting essentially of an Immunoglobulin (Ig) molecule (whole antibody, an Ig heavy or light chain or a fragment thereof) and a protein (the term includes also oligopeptides) having the biological activity of GCR (GCR-like protein), for enzyme replacement therapy and/or augmentation of glycolipid metabolism by the administration of bifunctional fusion proteins using a therapy based on the treatment of glycolipid storage disorders such as Gaucher's, Fabry's and Tay-Sachs diseases.
By selective altering of the amino acid sequences of the Ig moiety, GCR fusion proteins with improved properties, e.g. enhanced stability, can be obtained. Furthermore, fusion proteins can be provided, wherein shortened versions of GCR and the Ig chain are used. The present invention relates also to pharmaceutical compositions and therapeutic methods and systems comprising such GCR fusion proteins and methods of treating Gaucher's disease or another disease caused by glycolipid storage disorders, such as Fabry's and Tay-Sachs disease, comprising administering to a subject afflicted with this disease, a pharmaceutical composition comprising a therapeutic amount of recombinantly produced GCR fusion protein in a pharmaceutically acceptable carrier.
Background
The administration of exogenous β-glucosidase to treat diseases caused by glycolipid storage disorders like Gaucher's, Tay-Sachs' or Fabry's disease as attempts of enzyme augmentation in an organism suffering from such a disease rather than splenectomy or bone marrow transplantation are already described in literature to treat lysosomal storage defects. See, for example, De Duve, C. in Fed. Proc. 23, 1045 (1964) and Barton, N. W. et al. in Proc. Natl. Acad. Sci. 87, 1913 (1990), which describe the use of β-glucosidases and especially GCR to treat Gaucher's disease and the difficulties combined therewith to get a therapeutic response. However, the dose of the enzyme to treat these diseases is about 60 units per kilogram body weight every two weeks, that means that the average costs per year for the treatment of a 70 kg patient are about US$ 380.000,- for the enzyme alone. This is due to the short intracellular half-life of exogenous acid β-glucosidase.
Antibody-enzyme fusion proteins have been described which have a considerable improved in-vivo half-life and promote targeting to specific cell types such as tumor cells. For example the cytokine interleukin 2 (IL-2) has been fused to a monoclonal antibody heavy chain immunoreactive with, in two separate fusion proteins, the tumor antigens epithelial cell adhesion molecule (Ep-CAM) or the disialoganglios.de GD2 by use of the antibodies KS1/4 and ch14.18, respectively, to form the fusion proteins ch14.18-IL-2 and KS1/4-IL-2, respectively. See, for example, U.S. Patent No. 5,650,150.
Therefore, the object of the invention was to find suitable compounds for the effective treatment of glycolipid storage disorders, such as Gaucher's, Fabry's and Tay-Sachs disease which allow an efficient way and mode of administration, which is cheaper than the known costly procedures and within the price range of most patients, especially those in developing countries. The goal of the invention was to provide molecules for the treatment of Gaucher's, Fabry's and Tay-Sachs disease which can be administered in low dosages and have a longer half life in an organism without a significantly reduced activity and better targeting to specific cells where the glycolipid metabolism takes place and therefore enable a cheaper and more effective treatment of these diseases. Summary of the invention
It has now been discovered that there is an unexpected synergy in effectiveness and a prolonged half-life, if proteins having the biological activity of GCR are linked to an Ig molecule like an whole antibody, an Ig heavy or light chain, a fragment of an Ig heavy chain, for example the constant region of the heavy chain (CH), or the Fc or Fab fragment.
Fusion proteins and modification of specified fusion proteins are known in the art. For example, fusion proteins may effectively block a proteolytic enzyme from physical contact with the protein backbone itself, and thus prevent degradation. Additional advantages include, under certain circumstances, improved yield in a specific expression system, correct folding of a target protein, and increasing the stability, circulation time, and the biological activity of the therapeutic protein. One such modification is the use of the Fc region of immunoglobulins. Antibodies comprise two functionally independent parts, a variable domain known as "Fab", which binds antigen, and a constant domain, known as "Fc" which provides the link to effector functions such as complement or phagocytic cells. The Fc portion of an immunoglobulin mediates a long plasma half life when fused to certain proteins that have particularly short half lives (Capon, et al., Nature 337: 525-531 (1989)).
Therapeutic fusion proteins have also been constructed using the Fc domain to incorporate functions such as Fc receptor binding, protein A binding, complement fixation and placental transfer which all reside in the Fc proteins of immunoglobulins. For example, the Fc region of an lgG1 antibody has been fused to the N-terminal end of CD30-L, a molecule which binds CD30 receptors expressed on Hodgkin's Disease tumor cells, anaplastic lymphoma cells, T-cell leukemia cells and other malignant cell types (U.S. Patent No. 5,480,981). Furthermore, it has been reported in 1996 that efficient expression and secretion of certain non- mutant target proteins can be achieved by expression of fusion proteins comprising an Fc portion of an immunoglobulin and said target proteins followed by proteolytic cleavage of the target protein (WO 96/08570, US 5,541 ,087). A suitable GCR-like protein to be fused with an Ig polypeptide chain can have an amino acid sequence and a relating DNA sequence as given in the U.S. Patent No. 5,879,680 or can be a truncated or mutated form derived therefrom. Exemplary these proteins and the method of synthesis and conditions thereof, excluding the truncated and mutated forms, are described in the teachings of U.S. Patent No. 5,879,680, the disclosures of which relating to the preparation and use are specifically incorporated herein by reference.
Preferred truncated forms are for example those which consist of about one third to one half of the amino acid sequence of the natural GCR enzyme truncated from the carboxy terminal site of the enzyme. Those truncated proteins may be derived from the full length protein by cleaving off the desired chain with a suitable reagent such as a restriction enzyme or the like.
Assays for the identification of an effective GCR-like protein which is a suitable candidate for the fusion with an Ig polypeptide chain, as well as for the proof of activity for a fused compound according to this invention are described in the referenced U.S. Patent, and therefore it is considered that alternate fusion proteins for the treatment of glycolipid storage disorders such as Gaucher's, Fabry's and Tay-Sachs disease can be readily identified for practicing the present invention.
The invention presents novel proteins that have GCR-like activity in their ability to hydrolyze glucocerebrosides in an animal, but with additional advantageous properties such as higher expression level, higher solubility, better tissue distribution and better targeting to macrophages. These novel proteins include fusion proteins of GCR-like proteins and Ig molecules like a whole antibody or fragments thereof (an Ig heavy or light chain or a fragment of the heavy chain, like the CH, Fc or Fab fragment), forms of these fusion proteins that have altered glycosylation either in the GCR-like protein or in the Ig portion, forms of GCR fusion proteins that have a truncated or mutated amino acid sequence, having, for example, a reduced affinity e.g. to neonatal Fc receptors (FcRn) and GCR fusion proteins having specific linkers. Detailed Description
It is an object of the present invention to provide a protein with GCR-like activity having improved properties, wherein said protein is a fusion protein comprising an Ig molecule like a whole antibody, an Ig heavy or light chain or a fragment of the heavy chain (e.g. the CH, Fc or Fab fragment) and an GCR-like protein, wherein said Ig moiety is fused covalently directly or indirectly (via a linker molecule) to said GCR-like protein. In a preferred embodiment, the Ig moiety is fused covalently via its C-terminus directly or indirectly (via a linker molecule) to said GCR-like protein by its N-terminus, and the Ig portion as well as the GCR portion may be modified or mutated, selected from the group:
(I) H2N-lg - GCR-COOH
(II) H2N-lg - L - GCR-COOH (III) H2N-lg - GCRm-COOH
(IV) H2N-lgm - GCR-COOH
(V) H2N-lgm - GCRm-COOH
(VI) H2N-lgm - L - GCR-COOH
(VII) H2N-lg - L - GCRm-COOH (VIII) H2N-lg - GCRtrunc-COOH
(IX) H2N-lg - L - GCRtrunc-COOH
(X) H2N-GCR - Ig-COOH
(XI) H2N-GCR - L - Ig-COOH
(XII) H2N-GCRm - Ig-COOH (XIII) H2N-GCR - lgm-COOH
(XIV) H2N-GCRm - lgm-COOH
(XV) H2N-GCR - L - lgm-COOH
(XVI) H2N-GCRm - L - Ig-COOH
(XVII) H2N-GCRtrunc " Ig-COOH (XVIII) H2N-GCRtrUnc - - Ig-COOH
Herein, Ig has the meaning of a Ig heavy or light chain or a fragment of an Ig heavy chain (e.g. the CH, Fc or FAB fragment). GCR has the meaning of naturally occurring GCR from mammalian, preferably human origin, especially preferred from human lysosomal origin, and includes also recombinant GCR engineered from natural sources. GCRtrunc is an GCR according to this invention which is truncated but not mutated in its amino acid sequence. Truncated forms are protein fragments having essentially the full or only a slightly reduced biological activity of glucocerebrosidase. Preferred truncated forms of GCR according to this invention are those which consist of about one third to one half of the amino acid sequence of the natural glucocerebrosidase enzyme shortened at the C-terminus. GCRm is an GCR according to this invention which is mutated but not truncated in its amino acid sequence. The number of mutations is not limited but is restricted to the loss of the biological activity of the molecule. In a preferred embodiment the degree of mutation is between 5 and 30 per cent, in a especially preferred embodiment between 5 and 20 per cent of the amino acid residues. Variants with increased GCR biological activity can be generated by procedures described known in the art.
GCR, GCRm, GCRtrunc according to the invention is glycosylated, non- glycosylated, partially glycosylated or otherwise modified in its glycosylation pattern. The GCR fusion protein can be purified by standard techniques, for example, on a protein A column.
L has the meaning of a series of peptides such as. e.g., glycine and/or serine. Preferably, the peptide linker is a mixed series of glycine and serine peptides about 5 - 25, preferably 10 - 20 residues in length. Especially preferred are proteolytically cleavable linkers, especially linkers which are cleavable by lysosomal proteases like cathepsins.
In a preferred embodiment the Ig moiety is specific for a cell bearing an Fc receptor. Therefor, a preferred fragment of an Ig molecule to be linked to GCR is the Fc region. The Fc region of an immunoglobulin is the amino acid sequence for the carboxyl-terminal portion of an immunoglobulin heavy chain constant region. The Fc regions are particularly important in determining the biological functions of the immunoglobulin and these biological functions are termed effector functions. As known, the heavy chains of the immunoglobulin subclasses comprise four or five domains: IgM and IgE have five heavy chain domains, and IgA, IgD and IgG have four heavy chain domains. The Fc region of IgA, IgD and IgG is a dimer of the hinge-CH2-CH3 domains, and in IgM and IgE it is a dimer of the hinge-CH2-CH3-CH4 domains (see, W.E.Paul, ed.,1993, Fundamental Immunology, Raven Press, New York, New York).
As used herein, the term "Fc portion" means the carboxyl-terminal portion of an immunoglobulin heavy chain constant region, or an analog or portion thereof. That is, e.g., an immunoglobulin Fc region of Ig, preferably IgG, which may comprise at least a portion of a hinge region, a CH2 domain, and a CH3 domain.
The Fc region can be joined at its amino-terminus by a peptide bond to the carboxy-terminal amino acid of the GCR, or, in a preferred embodiment, the Fc region is linked at its carboxy-terminus by a peptide bond to the amino-terminal amino acid of the GCR.
In some circumstances, it is useful to mutate certain amino acids within the Ig molecule, especially in the Fc region of the fusion protein. For example, the neonatal Fc receptor (FcRn) binds IgG, and might reduce the clinical efficacy of the fusion protein.
Thus, Fcm is a Fc portion as defined above which is mutated in its amino acid sequence and / or modified in its glycosylation pattern. Such modified Fc portions lead to fusion proteins with improved properties. In this context Fcm includes additionally modified or mutated Fc portions which have a reduced affinity to
FcRn receptors. For example, it is known that IgG histidins located at the junction between the CH2 and CH3 domains (residues 310 and 433) of the IgG heavy chain contribute to the pH-dependent binding to the FcRn receptor (Raghavan, et al., Biochemistry 34(45): 14649-57 (1995)). Also lie 253 and His 435 and 436 (Kim et al., Eur. J. Immunol. 34: 2429-34 (1994)) as well as residues 309 (Leu, Val, Gin or Met in rat, murine and human IgGs) and 311 (Gin or Arg in rat, murine and human IgGs) (Kabat et al., in: Sequences of proteins of immunological interest. US Department of Health and Human Services, Bethesda, MD, USA (1991)) seems to form an interaction with the FcRn receptor. Thus, it is an object of the invention to provide a fusion protein with enhanced in vivo circulating half-life having a mutation, deletion or insertion at one or more amino acids in the domains responsible for FcRn receptor binding. In a preferred embodiment of the invention the GCR fusion protein comprises a Fc portion of an lgG1 , wherein said mutations are: position 253 is not He, position 309 is not Leu, Val, Gin or Met, position 310 is not His, position 311 is not Gin or Arg, position 433 is not His, position 435 is not His, and position 436 is not His. These and other variant proteins according to the invention may establish enhanced binding to the Fc receptor, enhanced stability, enhanced adoption of a correct active conformation, enhanced pharmacokinetic properties, enhanced synthesis, or other advantageous features. A specific method for improvement of GCR fusion proteins uses site-directed mutagenesis techniques. It is important to note that a wide variety of site-directed mutagenesis techniques are available, and can be used as alternatives to achieve similar results. The strategies for choosing among these techniques is well-known to those skilled in the art of molecular biology. Similarly, there is a wide variety of techniques for achieving random and semi-random mutagenesis of a target DNA. These techniques are also well-known to those skilled in the art of molecular biology.
The Ig molecule and the GCR-like protein according to this invention may also be linked by linker molecules, wherein the amino acid linkers are of varying length. The linker of the invention (L) is a linker molecule as defined below which may have also a protease cleavage site. The peptide linker often is a series of peptides such as. e.g., glycine and/or serine. Preferably, the peptide linker is a mixed series of glycine and serine peptides about 5 - 25, preferably 10 - 20 residues in length. Especially preferred are proteolytically cleavable linkers, especially linkers which are cleavable by lysosomal proteases like cathepsins. Preferred amino acid linkers L are used and include the following sequences:
1. Ala Ala Ala
2. Ala Ala Ala Ala,
3. Ala Ala Ala Ala Ala, 4. Ser,
5. Ser Ser,
6. Gly Gly Gly,
7. Gly Gly Gly Gly,
8. Gly Gly Gly Gly Gly, 9. Gly Gly Gly Gly Gly Gly Gly, 10. Gly Pro Gly, H . GIy Gly Pro Gly Gly,
12. Gly Gly Gly Gly Ser, and, if the linker shall have a protease cleavage site
13. Gly Gly Tyr Leu 14. Gly Gly Tyr
15. Gly Phe Ala Leu
16. Gly Pro Arg Leu and
17. any combinations of subparts 1-16
Additional suitable linkers are disclosed in Robinson et al., 1998, Proc. Natl. Acad. Sci. USA; 95, 5929.
As used herein, "proteolytic cleavage site" means amino acid sequences which are preferentially cleaved by a proteolytic enzyme or other proteolytic cleavage agents. Proteolytic cleavage sites include amino acids sequences which are recognized by proteolytic enzymes especially cathepsins or other lysosomal proteases.
It is another object of the present invention to construct GCR fusion proteins, wherein a whole antibody is used. Such fusion molecules comprise the variable regions of heavy and light chains of an antibody and the epitopes binding to a specific antigen. For example, GCR is fused to the C-terminus of an antibody heavy chain. DNA constructs encoding whole antibody fusion proteins may be constructed as described previously (Gillies et al. [1991] Hybridoma 10:347-356).
The invention also relates to a DNA molecule that encodes any of the fusion proteins disclosed above and depicted in the claims.
As a preferred embodiment a DNA molecule is disclosed that encodes a fusion protein as defined above and in the claims comprising: (a) a signal / leader sequence (b) a sequence of an Ig molecule
(c) a target protein sequence having the biological activity of GCR.
The signal sequence of the invention as indicated above is a polynucleotide which encodes an amino acid sequence that initiates transport of a protein across the membrane of the endoplasmic reticulum. Signal sequences which will be useful in the invention include antibody light chain signal sequences, e.g., antibody 14.18 (Gillies et. al., Jour, of Immunol. Meth., 125:191 , (1989)), antibody heavy chain signal sequences, e.g., the MOPC141 antibody heavy chain signal sequence (Sakano et al., Nature 286:5774(1980)), and any other signal sequences which are known in the art (see for example, Watson, Nucleic Acids Research 12:5145, (1984)). Each of these references is incorporated herein by reference. Signal sequences have been well characterised in the art and are known typically to contain 16 to 30 amino acid residues, and may contain greater or fewer amino acid residues. A typical signal peptide consists of three regions: a basic N-terminal region, a central hydrophobic region, and a more polar
C-terminal region. The central hydrophobic region contains 4 to 12 hydrophobic residues that anchor the signal peptide across the membrane lipid bilayer during transport of the nascent polypeptide. Following initiation, the signal peptide is usually cleaved within the lumen of the endoplasmic reticulum by cellular enzymes known as signal peptidases.
Potential cleavage sites of the signal peptide generally follow the "(-3, -1) rule". Thus a typical signal peptide has small, neutral amino acid residues in positions -1 and -3 and lacks proline residues in this region. The signal peptidase will cleave such a signal peptide between the -1 and +1 amino acids. Thus, the portion of the DNA encoding the signal sequence may be cleaved from the amino-terminus of the fusion protein during secretion. This results in the secretion of a fusion protein consisting of the Ig region and the target protein. A detailed discussion of signal peptide sequences is provided by von Heijne (Nucleic Acids Res., 14:4683,(1986)). As would be apparent to one of skilled in the art, the suitability of a particular signal sequence for use in a secretion cassette may require some routine experimentation. A signal sequence is also referred to as a "signal peptide", "leader sequence" or "leader peptides" and each of these terms having meanings synonymous to signal sequence may be used herein.
The invention also relates to expression vectors comprising said DNA molecules which promote expression of the target protein, that is a GCR fusion protein. As used herein, "vector" means any nucleic acid comprising a nucleotide sequence competent to be incorporated into a host cell and to be recombined with and integrated into the host cell genome, or to replicate autonomously as an episome. Such vectors include linear nucleic acids, plasmids, phagemids, cosmids, RNA vectors, viral vectors and the like. Non-limiting examples of a viral vector include a retrovirus, an adenovirus and an adeno-associated virus. As used herein, "expression of a target protein" is understood to mean the transcription of the DNA sequence, translation of the mRNA transcript, and secretion of a protein product that is folded into a correct, active conformation.
According to the invention eukaryotic, preferably mammalian, host cells are used that are suitable for expressing a fusion protein as defined in this application. Methods of transfecting such host cells with said vector, expressing, purifying and isolating the fusion proteins of this invention are well known in the art. Therefore, the method according to this invention comprises: (i) constructing a DNA encoding a precursor protein that comprises a leader sequence for secretion, the Ig portion, the GCR, GCRm or GCR t nc moiety and optionally a linker sequence between the Ig and GCR portion, (ii) placing said fused DNA in an approbiate expression vector, (iii) expressing said fusion protein in a eukaryotic cell, and (iv) purifying said secreted fusion protein.
The invention also relates to pharmaceutical compositions comprising at least one of the GCR fusion protein as defined above and below, preferably a fusion protein wherein a Fc portion of a IgG is linked at its C-terminal amino acid by a peptide bond to the N-terminal amino acid of the GCR-like protein, together with pharmaceutically acceptable carriers, diluents, and excipients. These pharmaceutical compositions may optionally contain other drugs or medicaments that are helpful in co-treating GCR deficient diseases. Such pharmaceutical compositions may be for intravenous, subcutaneous, intramuscular, orthotopic injection, orthotopic infusion, or for oral, pulmonary, nasal, transdermal or other forms of administration. Administration can be accomplished by periodic unit dosages, by continuous infusion, peristaltic delivery, by bolus injection, and the like. Routes can include
In general, comprehended by the invention are pharmaceutical compositions comprising effective amounts of protein or derivative products of the invention together with pharmaceutically acceptable diluents, preservatives, solubilizers, emulsifiers, adjuvants and/or carriers. Such compositions include diluents of various buffer content (e.g., Tris-HC1 , acetate, phosphate), pH and ionic strength; additives such as detergents and solubilizing agents (e.g., Tween 80, Polysorbate 80), anti-oxidants (e.g., ascorbic acid, sodium metabisulfite), preservatives (e.g., Thimersol, benzyl alcohol) and bulking substances (e.g., lactose, mannitol). The term "parenteral" as mentioned above and below includes subcutaneous, intravenous, intra-articular and intratracheal injection and infusion techniques. The parenteral administration is preferred.
As used herein, the term "pharmaceutically acceptable carrier or excipient" means an inert, non toxic liquid filler, diluent, solvent or solution, not reacting adversely with the active compounds or with the patient. Suitable liquid carriers are well known in the art such as steril water, saline, aqueous dextrose, sugar solutions, ethanol, glycols and oils, including those of petroleum, animal, vegetable, or synthetic origin. The formulations may also contain adjuvants or vehicles which are typical for parenteral administration. With respect to said suitable formulations it should be pointed out that the Fusion proteins of the present invention may eventually form pharmaceutically acceptable salts with any non-toxic, organic or inorganic acid showing changed solubility. Inorganic acids are, for example, hydrochloric, sulphuric or phosphoric acid and acid metal salts such as sodium monohydrogen orthophosphate and potassium hydrogen sulfate. Examples for organic acids are the mono, di and tri carboxylic acids such as acetic, glycolic, lactic, pyruvic, malonic, succinic, glutaric, fumaric, malic, tartaric, citric, ascorbic, maleic, benzoic, phenylacetic, cinnamic, salicylic and sulfonic acids. Salts of the carboxy terminal amino acid moiety include the non-toxic carboxylic acid salts formed with any suitable inorganic or organic bases. These salts include, for example, alkali metals such as sodium and potassium, alkaline earth metals such as calcium and magnesium, and organic primary, secondary and tertiary amines such as trialkylamines.
Typically, the dosage of the GCR fusion protein for the treatment of glycolipid storage disorders like Gaucher's, Tay-Sachs' or Fabry's disease is 0.01 mg to 25 mg, preferably about 0.1 to 2 mg, and more preferably about 0.1 to 1 mg per kilogram body weight per day. The effective dosages may be determined using diagnostic tools which are known in the prior art. In general, the optimum therapeutically acceptable dosage and dose rate for a given patient within the above-said ranges depends on a variety of factors, such as the activity of the specific active material employed, the age, body weight, general health, sex, diet, time and route of administration, rate of clearance or the object of treatment. One skilled in the art will be able to ascertain effective dosages by administration and observing the desired therapeutic effect. The dosages may also vary over the course of therapy, with a relatively high dosage being used initially, until therapeutic benefit is seen, and lower dosages used to maintain the therapeutic benefits.
The invention relates also to therapeutic methods and therapeutic systems for treating a variety of glycolipid storage disorders such as Gaucher's, Fabry's and Tay-Sachs disease and the enzymes related to these diseases, by GCR fusion protein therapy. The therapeutic method comprises a variety of modalities for practicing the invention in terms of the steps. For example, the GCR fusion protein can be administered following admixture, i.e., simultaneously, or can be administered sequentially with an other drug and/or additive, such as a vitamine or as a single medication. Furthermore, the additives and/or additional drugs and the fusion protein can be separately administered with a time interval between administrations of from zero to 3 weeks, i.e., from substantially immediately after the first active agent is administered to up to 3 weeks after the first agent is administered. Additionally, it is contemplated that the order can be varied, i.e., that the additives and/or additional drugs could be administered prior to administration of the fusion protein, or that administration can be conducted in the reverse order.
In another embodiment, it is considered that the invention can be practiced in conjunction with surgical procedures where for example portions or all of the spleen has been removed. In this regard, the method can be practiced following a surgical procedure. Alternatively, the surgical procedure can be practiced during the interval between administration of the active agent. Exemplary of this method is the combination of the present method with surgical spleen removal.
Treatment according to the method will typically comprise administration of the active agent in one or more cycles of administration. For example, where a single or a simultaneous administration of GCR fusion protein is practiced, a therapeutic composition comprising the single lipid storage disease drug or afore- said drug and a additive and/or another drug is administered over a time period of from about 2 days to about 3 weeks in a single cycle. Thereafter, the treatment cycle can be repeated as needed according to the judgment of the practicing physician. Similarly, where a sequential application of two different agents is contemplated, the administration time will typically cover the same time period. The interval between cycles can vary from about zero to 2 months.
In another embodiment, the invention describes a method for the treatment of Gaucher's disease comprising administering to a patient a therapeutic composition comprising an amount of a GCR fusion protein as defined above as a supportive treatment in combination with a bone marrow transplantation, or a surgery, by removing an organ that serves as an important storage site of glycolipid, for example the spleen or to prepare a successful gene therapy by a previous enzyme augmentation treatment of a human being suffering from a glycolipid storage disorder.
In those cases of a supportive treatment of one of the diseases according to this invention by enzyme replacement or augmentation therapy, the administration of the fusion protein can be separately to the other operation, i.e. the surgery administered with a time interval between the operation and the administrations of the fusion protein of from zero to 3 weeks, i.e., from substantially immediately after the operation, such as bone marrow transplantation, or a surgery of the active agent up to 3 weeks after the agent is administered. Additionally, it is contemplated that the order can be varied, i.e., that the fusion protein could be administered prior to bone marrow transplantation, or a surgery, or that administration can be conducted in the reverse order.
Further, the invention contemplates systems comprising packaging and/or kits which provide the reagents necessary for practicing the methods of the present invention. A kit is therefore described for treating glycolipid storage disorders comprising a package comprising:
a) a therapeutic composition comprising an amount of the GCR fusion protein as defined above b) optionally a additive or a supportive drug for the treatment of afore-said diseases; and c) instructions for using the reagents in methods to treat Gaucher's, Tay-Sachs' or Fabry's diseases.
A reagent in a kit of this invention is typically formulated as a therapeutic composition as described herein, and therefore can be in any of a variety of forms suitable for distribution in a kit. Such forms can include a liquid, powder, tablet, suspension and the like formulation for providing the fusion protein of the present invention and optionally the supportive drug and/or additive. The reagents may be provided in separate containers suitable for administration separately according to the present methods, or alternatively may be provided combined in a composition in a single container in the package.
The package may contain an amount sufficient for one or more dosages of reagents according to the treatment methods described herein. Typically, a package will contain an amount sufficient for one cycle of treatment as described herein.
A kit of this invention also contains "instruction for use" of the materials contained in the package. The instructions relate to the use of the fusion protein and/or optionally for the supportive drug and/or the additive for treating the glycolipid storage disorders according to the methods. Insofar as the methods can vary widely depending upon the phase and the type of disease, the patient and the condition of the disease, the instructions can vary to specify procedures for administration accordingly. The invention is not to be considered as limiting as to the nature of the instructions other than the particularity regarding the use of the fusion protein according to the methods of the present invention.
Sequence Information
The following amino acid sequences were used in this invention
SEQ ID NO:1 The human lysosomal GCR amino acid sequence (one-letter code)
MAGSLTGLLL LQAVSWASGA RPCIPKSFGY SSWCVCNAT YCDSFDPPTF PALGTFSRYE STRSGRRMEL SMGPIQANHT GTGLLLTLQP EQKFQKVKGF GGAMTDAAAL NILALSPPAQ NLLLKSYFSE EGIGYNIIRV PMASCDFSIR TYTYADTPDD FQLHNFSLPE EDTKLKIPLI HRALQLAQRP VSLLASPWTS PTWLKTNGAV NGKGSLKGQP GDIYHQTWAR YFVKFLDAYA EHKLQFWAVT AENEPSAGLL SGYPFQCLGF TPEHQRDFIA RDLGPTLANS THHNVRLLMLD DQRLLLPHWA KWLTDPEAA KYVHGIAVHW YLDFLAPAKA TLGETHRLFP NTMLFASEAC VGSKFWEQSV RLGSWDRGMQ YSHSIITNLL YHWGWTDWN
LALNPEGGPN WVRNFVDSPI IVDITKDTFY KQPMFYHLGH FSKFIPEGSQ
RVGLVASQKN DLDAVALMHP DGSAWWLN RSSKDVPLTI KDPAVGFLET ISPGYSIHTY LWRRQ
SEQ ID NO:2 Human lgG1 Fc region - mature protein coding sequence (one-letter code)
EPKSCDKTHT CPPCPAPELL GGPSVFLFPP KPKDTLMISR TPEVTCWVD VSHEDPEVKF NWYVDGVEVH NAKTKPREEQ YNSTYRWSV LTVLHQDWLN GKEYKCKVSN KALPAPIEKT ISKAKGQPRE PQVYTLPPSR EEMTKNQVSL TCLVKGFYPS DIAVEWESNG QPENNYKTTP PVLDSDGSFF LYSKLTVDKS RWQQGNVFSC SVMHEALHNH YTQKSLSLSP GK
SEQ ID NO:3 Human lgG2 Fc region - mature protein coding sequence
(one-letter code) ERKCCVECPP CPAPPVAGPS VFLFPPKPKD TLMISRTPEV TCVWDVSHE DPEVQFNWYV DGVEVHNAKT KPREEQFNST FRWSVLTW HQDWLNGKEY KCKVSNKGLP APIEKTISKT KGQPREPQVY TLPPSREEMT KNQVSLTCLV KGFYPSDIAV EWESNGQPEN NYKTTPPMLD SDGSFFLYSK L DKSRWQQ GNVFSCSVMH EALHNHYTQK SLSLSPGK
The following examples describe the invention in more detail without limiting it.
Example 1.
Expression of human Fc-GCR
A sequence encoding the mature form of GCR was completely synthesized from oligonucleoties by standard techniques. The synthesized DNA was engineered to have a Xmal-compatible overhang at the 5'end and an Xhol-compatible overhang at the 3'end.
The DNA was cloned and sequence analysis confirmed that encodes the mature
GCR protein without mutations.
The expression vector pdCs-Fc-GCR was constructed as follows. The Xmal-Xhol- restriction fragment containing the GCR cDNA was ligated to the Xmal-Xhol fragment of the pdCs-Fc vector according to Lo et al. [Protein Engineering (1998) 11 :495]. The resultant vector, pdCs-Fc-GCR, was used to transfect mammalian cells for the expression of Fc-GCR. This vector expresses the human imunoglobulin gamma 1 chain Fc-region. The Fc protein moiety also usually contains a glycosylation site. This site may be optinally changed to a non-glycosylated sequence by standard approaches.
Example 2.
Transfection and expression of Fc-GCR fusion proteins For transient transfection, the plasmids were introduced into BHK cells. Cells were transfected by coprecipitation of plasmid DNA with calcium phosphate [Sambrook et al. (1989) Molecular Cloning-A Laboratory Manual, Cold Spring, Harbor, NY] or by lipofection using Lipofectamine Plus (Life technologies, Gaithersburg, MD) according to suppliers protocol. To generate stable cell lines, NS/0 cells were used for both transient transfection and the generation of stable eel lines.
In order to obtain stably transfected clones, plasmid DNA was introduced into cells by electroporation. About 5x106 eels were washed once with PBS and resuspended in 0.5 ml PBS. 10 μg of linearized plasmid DNA were then incubated with the cells in a Gene Pulser Cuvette (0.4 cm electrode gap, Bio Rad) on ice for 10 min. Electroporation was performed using a Gene Pulser (Bio Rad, Hercules, CA) with sttings at 0.25 V and 500 microF. Cells were allowed to recover for 10 min on ice, after which they were resuspended in growth medium and then plated onto 96 well plates. Stably transfected clones were selected by growth in the presence of 100 nM methotrexate (MTX), which was introduced two days post transfection. The cells were fed every 3 days for 2 to 3 more times, and MTX-resistant clones appeared in 2 to 3 weeks. Supematants from clones were assayed by anti-Fc ELISA to identify high producers. High producing clones were isolated and propadated in growth medium containing 100 nM MTX.
BHK and NS/0 cells were grown in Dulbecco's modified Eagle's medium supplemented with 10% fetal bovine serum, 2 nM glutamine and penicillin/streptomycin. For routine characterization by gel electrophoresis, Fc fusion proteins in the conditioned media were captured on Protein A Sepharose (Repligen, Cambridge, MA) and then eluted by boiling in the protein sample buffer with or without 2- mercaptoethanol. After electrophoresis on a SDS-Gel, the protein bands were visualized by Coomassie staining. For purification, the fusion proteins bound on Protein A Sepharose were eluted in a sodium phosphate buffer (100 mM NaH2PO4, pH 3. And 150 mM NaCI). The eluate was then immediately neutralized with 0.1 volume of 2 M Tris-HCI, pH 8.
Example 3.
Carbohydrate characterization
Endoglycosidase-H was dissolved in 100 mM sodium acetate, pH 6.0, at a final concentration of 10 units/ml. N-glycanase was supplied as a 250 unit/ml suspension in 50% glycerol. Either human placental enzyme or fifty μl aliquot of decyl-agarose fraction containing GCR activity were adjusted to 0.5% SDS/1 M β- mercaptoethanol and boiled for two minutes. The samples were then diluted with appropriate buffer to either 200 mM sodium acetate, pH 6.0 (for endoglycosidase- H) or 200 mM sodium phosphate, pH 8.5 (for N-glycanase) to a final composition of 0.1% SDS, 0.7% NP-40, and 0.02M β-mercaptoethanol. The samples were again boiled for 1 min and then either endoglycosidase-H or N-glycanase added to final concentrations of 50 mu/ml or 20 U/ml, respectively. Digestions were for about 16 hours at 37° C. Carboxypeptidase Y was used as a control for both deglycosylation reactions.
Example 4
Amino acid sequence analysis:
Samples used for amino acid sequence analysis were electrophoretically fractionated on SDS-Gels as described above and then transferred to PVDF membranes as described by Matsudaira (J.B.C. 262:10035, 1987). Typically, after electrophoresis the gel was incubated in transfer buffer (0.1 M CAPS, 10% methanol, pH 11.0) for 10 minutes prior to transblotting (50 ma for 4 hours). The gel was then washed with HPLC grade water for 5 minutes, stained with 0.1 % Coomassie Blue R250 (in 50% methanol) for 5 minutes, and finally destained for 10 minutes with 50% methanol-10% acetic acid. The PVDF membrane was again washed with HPLC grade water, dried under a stream of nitrogen and stored in a sealing bag at -20° C until used for amino acid sequencing.
Amino acid sequence analysis was accomplished using an Applied Biosystems Model 470A gas-phase sequencer equipped with a Model 120A on-line PTH- amino acid analyzer. The program 03R PTH was used directly for sequencing without pretreatment of the membrane strip with polybrene. An approximately 2x8 mm piece of PVDF membrane containing the protein band of interest was excised, centered on the teflon seal, and placed in the cartridge block of the sequencer. Multiple strips of the PVDF membrane could be stacked in this manner, thus increasing the amount of protein available for sequencing. The initial and repetitive yields for sequencing recombinant GCR were calculated by comparison with the yields obtained after 100 picomoles of human placenta GCR were electrophoresed, transblotted to PVDF and subjected to ten cycles of amino acid sequence.
N-terminal amino acid sequence of mature human placental GCR was compared to N-terminal amino acid sequence of recombinant human GCR using the methods described in the text. The N-terminal amino acids determined by direct chemical sequencing of the mature human and recombinant GCR are identical indicating that the signal sequence in the recombinantly produced enzymes are correctly processed.
Example 5
GCR assays:
For pH profile and inhibition studies, GCR activity was measured using 100 mM potassium phosphate buffer containing 0.15% Triton X-100, 2.5 μl of β-D-1-14C- glucocerebroside (7.5 mg/ml in sodium taurocholate at 50 mg/ml), and the sample in the total volume of 200 μl. Preincubations with conduritol-B-epoxide were for 30 min at 37°C. For Km determination, β-glucosidase activity was assayed at pH 5.9 using the artificial substrate 4-methylumbellifery-β-D- glucopyranoside (4MUGP) in 100 mM potassium phosphate buffer containing 0.15% Triton X-100 and 0.125% sodium taurocholate. Purification of recombinant GCR was also monitored using 4MUGP.
It is understood that the examples and embodiments described herein are for illustrative purposes only and that various modifications or changes in light thereof will be suggested to persons skilled in the art and are to be included within the spirit and purview of this application and scope of the appended claims.
Other uses will be apparent to one skilled in the art in light of the present disclosures.

Claims

Patent Claims:
1. A fusion protein consisting essentially of an imunoglobuline molecule (Ig) or a fragment thereof and a non immunoglobulin molecule, wherein the non- immunoglobulin molecule is a protein having the biological activity of glucocerebrosidase (GCR-like protein).
2. A fusion protein of claim 1 wherein the Ig molecule has a specificity to a Fc receptor.
3. A fusion protein of claim 1 or 2, wherein the Ig molecule is covalently linked by its C-terminus to the N-terminus of the GCR-like protein.
4. A fusion protein of any of the claims 1 to 3 wherein a linker molecule is fused between the Ig molecule and the GCR-like protein.
5. A fusion protein of claim 4, wherein the linker molecule comprises a protease cleavage site.
6. A fusion protein of claim 5, wherein the protease cleavage site is specific for lysosomal proteases.
7. A fusion protein of any of the claims 1 to 6, wherein the GCR like protein is GCR.
8. A fusion protein of claim 7, wherein GCR is truncated (GCRtrUnc) or mutated (GCRm).
9. A fusion protein of claim 7 or 8, wherein GCR or the GCR-like protein has a modified glycosylation pattern or is non-glycosylated.
10. A fusion protein of any of the claims 1 to 9, wherein the Ig molecule is a Fc portion.
11. A fusion protein of any of the claims 1 to 9, wherein the Ig molecule is a whole antibody.
12. A fusion protein of any of the claims 1 to 11 wherein the Ig molecule or the fragment thereof is designed.
13. A fusion protein of claim 12, wherein the Ig molecule has a reduced affinity to a FcRn receptor.
14. A fusion protein of any of the claims 1 to 13, wherein the Ig molecule within the fusion protein is dimerized.
15. A DNA sequence encoding any of the fusion proteins of claims 1 to 14.
16. A DNA molecule encoding a fusion protein according to at least one of the claims 1 to 14 comprising:
(a) a signal / leader sequence
(b) an Ig molecule
(c) a target protein sequence having the biological activity of GCR.
17. An expression vector comprising a DNA of claim 15 or 16.
18. A host cell suitable for expressing an fusion protein as defined in at least one of the claims 1 to 14 comprising a vector of claim 17.
19. A method for producing a fusion protein of at least one of the claims 1 to 14, said method comprising:
(i) constructing a DNA encoding a precursor protein that comprises a leader sequence for secretion, the Ig molecule, the GCR, GCRm or GCR trunc portion and optionally the linker-sequence,
(ii) placing said fused DNA in an appropriate expression vector, (iii) expressing said fusion protein in a eukaryotic cell, and (iv) purifying said secreted fusion protein.
0. A pharmaceutical composition comprising a fusion protein according to at least one of the claims 1 to 14 and at least one pharmaceutically acceptable carrier, diluent or excipient.
21. A pharmaceutical composition of claim 20 containing at least one additional pharmaceutically effective drug and / or adjuvants.
22. Use of a fusion protein of any of claims 1 to 14 for the manufacture of a pharmaceutical composition for the treatment of glycolipid storage disorders.
23. The use of claim 22, wherein the glycolipid storage disorder is selectecd from the group consisting of Gaucher's, Fabry's and Tay-Sachs disease.
24. A method of treating glycolipid storage disorders comprising administering to a subject afflicted with said disease a pharmaceutical composition according to claim 16 or 17.
25. The method of claim 18 wherein the glycolipid storage disorder is selectecd from the group consisting of Gaucher's, Fabry's and Tay-Sachs disease.
PCT/EP2001/015328 2001-01-18 2001-12-27 Bifunctional fusion proteins with glucocerebrosidase activity WO2002057435A2 (en)

Priority Applications (10)

Application Number Priority Date Filing Date Title
BR0116803-7A BR0116803A (en) 2001-01-18 2001-12-27 Bifunctional fusion proteins with glucocerebrosidase activity
CA002435037A CA2435037A1 (en) 2001-01-18 2001-12-27 Bifunctional fusion proteins with glucocerebrosidase activity
KR10-2003-7009521A KR20030067755A (en) 2001-01-18 2001-12-27 Bifunctional fusion proteins with glucocerebrosidase activity
HU0401300A HUP0401300A3 (en) 2001-01-18 2001-12-27 Bifunctional fusion proteins with glucocerebrosidase activity
MXPA03006294A MXPA03006294A (en) 2001-01-18 2001-12-27 Bifunctional fusion proteins with glucocerebrosidase activity.
PL01362394A PL362394A1 (en) 2001-01-18 2001-12-27 Bifunctional fusion proteins with glucocerebrosidase activity
JP2002558488A JP2004525621A (en) 2001-01-18 2001-12-27 Bifunctional fusion protein having glucocerebrosidase activity
US10/466,593 US20040043457A1 (en) 2001-01-18 2001-12-27 Bifunctional fusion proteins with glucocerebrosidase activity
EP01989057A EP1392826A2 (en) 2001-01-18 2001-12-27 Bifunctional fusion proteins with glucocerebrosidase activity
NO20033247A NO20033247D0 (en) 2001-01-18 2003-07-17 Bifunctional fusion proteins with glucocerebrosidase activity

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP01101056.8 2001-01-18
EP01101056 2001-01-18

Publications (2)

Publication Number Publication Date
WO2002057435A2 true WO2002057435A2 (en) 2002-07-25
WO2002057435A3 WO2002057435A3 (en) 2003-12-24

Family

ID=8176235

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2001/015328 WO2002057435A2 (en) 2001-01-18 2001-12-27 Bifunctional fusion proteins with glucocerebrosidase activity

Country Status (13)

Country Link
US (1) US20040043457A1 (en)
EP (1) EP1392826A2 (en)
JP (1) JP2004525621A (en)
KR (1) KR20030067755A (en)
CN (1) CN1630720A (en)
BR (1) BR0116803A (en)
CA (1) CA2435037A1 (en)
HU (1) HUP0401300A3 (en)
MX (1) MXPA03006294A (en)
NO (1) NO20033247D0 (en)
PL (1) PL362394A1 (en)
WO (1) WO2002057435A2 (en)
ZA (1) ZA200306333B (en)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007012188A1 (en) * 2005-07-27 2007-02-01 Qinghua Wang GLP/1/EXENDM 4 IgG Fc FUSION CONSTRUCTS FOR TREATMENT OF DIABETES
EP2256134A1 (en) * 2003-11-13 2010-12-01 Hanmi Pharmaceutical Co., Ltd. IgG Fc fragment for a drug carrier and method for the preparation thereof
WO2011107991A1 (en) * 2010-03-02 2011-09-09 Protalix Ltd. Glucocerebrosidase multimers and uses thereof
US8110665B2 (en) 2003-11-13 2012-02-07 Hanmi Holdings Co., Ltd. Pharmaceutical composition comprising an immunoglobulin FC region as a carrier
US8742079B2 (en) 2007-08-20 2014-06-03 Protalix Ltd. Saccharide-containing protein conjugates and uses thereof
US9194011B2 (en) 2009-11-17 2015-11-24 Protalix Ltd. Stabilized alpha-galactosidase and uses thereof
US9732333B2 (en) 2011-01-20 2017-08-15 Protalix Ltd. Nucleic acid construct for expression of alpha-galactosidase in plants and plant cells
US10604576B2 (en) 2016-06-20 2020-03-31 Kymab Limited Antibodies and immunocytokines
US11753479B2 (en) 2014-03-04 2023-09-12 Kymab Limited Nucleic acids encoding anti-OX40L antibodies
US11779604B2 (en) 2016-11-03 2023-10-10 Kymab Limited Antibodies, combinations comprising antibodies, biomarkers, uses and methods

Families Citing this family (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ATE267215T1 (en) 1997-12-08 2004-06-15 Lexigen Pharm Corp HETERODIMARY FUSION PROTEINS FOR USE FOR TARGETED IMMUNTHERAPY AND GENERAL IMMUNE EXCITATION
JP2002511432A (en) * 1998-04-15 2002-04-16 レキシジェン ファーマシューティカルズ コーポレイション Enhancement of antibody-cytokine fusion protein-mediated immune response by co-administration of an angiogenesis inhibitor
SK782002A3 (en) 1999-07-21 2003-08-05 Lexigen Pharm Corp FC fusion proteins for enhancing the immunogenicity of protein and peptide antigens
HUP0202442A3 (en) * 1999-08-09 2005-01-28 Lexigen Pharmaceuticals Corp L Multiple cytokine-antibody complexes
US20050202538A1 (en) * 1999-11-12 2005-09-15 Merck Patent Gmbh Fc-erythropoietin fusion protein with improved pharmacokinetics
ATE336514T1 (en) * 2000-02-11 2006-09-15 Merck Patent Gmbh INCREASE THE CIRCULATION HALF-LIFE OF ANTIBODIES-BASED FUSION PROTEINS
PT1294401E (en) * 2000-06-29 2007-11-09 Merck Patent Gmbh Enhancement of antibody-cytokine fusion protein mediated immune responses by combined treatment with immunocytokine uptake enhancing agents
KR20090010127A (en) * 2001-03-07 2009-01-28 메르크 파텐트 게엠베하 Expression technology for proteins containing a hybrid isotype antibody moiety
WO2002079415A2 (en) * 2001-03-30 2002-10-10 Lexigen Pharmaceuticals Corp. Reducing the immunogenicity of fusion proteins
DK1383785T3 (en) * 2001-05-03 2011-05-23 Merck Patent Gmbh Recombinant tumor-specific antibody and its use
ATE542137T1 (en) * 2001-12-04 2012-02-15 Merck Patent Gmbh IMMUNOCYTOKINE WITH MODULATED SELECTIVITY
WO2004055056A1 (en) * 2002-12-17 2004-07-01 Merck Patent Gmbh Humanized antibody (h14.18) of the mouse 14.18 antibody binding to gd2 and its fusion with il-2
US20050069521A1 (en) * 2003-08-28 2005-03-31 Emd Lexigen Research Center Corp. Enhancing the circulating half-life of interleukin-2 proteins
DE602004013372T2 (en) 2003-12-30 2009-07-02 Merck Patent Gmbh IL-7 FUSION PROTEINS WITH ANTIBODY PORTIONS, THEIR PREPARATION AND THEIR USE
RU2370276C2 (en) * 2003-12-31 2009-10-20 Мерк Патент Гмбх Fc-ERYTHROPOIETIN FUSED PROTEIN WITH IMPROVED PHARMACOKINETICS
ES2330860T3 (en) 2004-01-22 2009-12-16 Merck Patent Gmbh ANTIBODY ANTIBODIES WITH FIXING THE REDUCED COMPLEMENT.
US7670595B2 (en) * 2004-06-28 2010-03-02 Merck Patent Gmbh Fc-interferon-beta fusion proteins
CN101072793B (en) * 2004-12-09 2012-06-20 默克专利有限公司 Il-7 variants with reduced immunogenicity
JP2007063225A (en) * 2005-09-01 2007-03-15 Takeda Chem Ind Ltd Imidazopyridine compound
US20070104689A1 (en) * 2005-09-27 2007-05-10 Merck Patent Gmbh Compositions and methods for treating tumors presenting survivin antigens
EP1948614A2 (en) * 2005-11-18 2008-07-30 Takeda San Diego, Inc. Glucokinase activators
PT1966238E (en) 2005-12-30 2012-07-31 Merck Patent Gmbh Interleukin-12p40 variants with improved stability
KR101397290B1 (en) * 2005-12-30 2014-05-21 메르크 파텐트 게엠베하 Anti-cd19 antibodies with reduced immunogenicity
PT1986612E (en) 2006-02-07 2012-11-06 Shire Human Genetic Therapies Stabilized composition of glucocerebrosidase
EP2001875A2 (en) 2006-03-08 2008-12-17 Takeda San Diego, Inc. Glucokinase activators
US8748567B2 (en) * 2006-05-22 2014-06-10 Children's Medical Center Corporation Method for delivery across the blood brain barrier
WO2007143434A2 (en) 2006-05-31 2007-12-13 Takeda San Diego, Inc. Indazole and isoindole derivatives as glucokinase activating agents
EP2091947A2 (en) 2006-12-20 2009-08-26 Takeda San Diego, Inc. Glucokinase activators
US8173645B2 (en) * 2007-03-21 2012-05-08 Takeda San Diego, Inc. Glucokinase activators
EA201171259A1 (en) * 2009-04-22 2012-05-30 Мерк Патент Гмбх ANTIBODY HYBRID PROTEINS WITH MODIFIED FCRN BINDING SITES
US8889621B2 (en) 2009-10-30 2014-11-18 New York University Inhibiting binding of FGF23 to the binary FGFR-Klotho complex for the treatment of hypophosphatemia
BR112012012947B8 (en) 2009-11-27 2021-05-25 Genzyme Corp hemitartrate salt and pharmaceutical composition comprising the same
US9657075B2 (en) 2012-06-07 2017-05-23 New York University Chimeric fibroblast growth factor 23 proteins and methods of use
US9550820B2 (en) 2013-02-22 2017-01-24 New York University Chimeric fibroblast growth factor 23/fibroblast growth factor 19 proteins and methods of use
JOP20140087B1 (en) 2013-03-13 2021-08-17 Amgen Inc Proteins specific for baff and b7rp1 and uses thereof
US9458246B2 (en) 2013-03-13 2016-10-04 Amgen Inc. Proteins specific for BAFF and B7RP1
WO2015009052A1 (en) * 2013-07-16 2015-01-22 일동제약 주식회사 Fusion protein of immunoglobulin hybrid fc and enzyme
CA2943361A1 (en) * 2014-03-28 2015-10-01 New York University Fgf23 fusion proteins
CN111094559A (en) * 2017-07-07 2020-05-01 韩美药品株式会社 Novel therapeutic enzyme fusion proteins and uses thereof

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5428130A (en) * 1989-02-23 1995-06-27 Genentech, Inc. Hybrid immunoglobulins
US5650150A (en) * 1990-11-09 1997-07-22 Gillies; Stephen D. Recombinant antibody cytokine fusion proteins
WO1998022577A1 (en) * 1996-11-15 1998-05-28 Maria Grazia Masucci Fusion proteins having increased half-lives
WO1998046257A1 (en) * 1997-04-17 1998-10-22 Amgen Inc. Compositions comprising conjugates of stable, active, human ob protein with antibody fc chain and methods
WO2001060412A2 (en) * 2000-02-15 2001-08-23 Genzyme Corporation Modification of biopolymers for improved drug delivery

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6475486B1 (en) * 1990-10-18 2002-11-05 Aventis Pharma Deutschland Gmbh Glycosyl-etoposide prodrugs, a process for preparation thereof and the use thereof in combination with functionalized tumor-specific enzyme conjugates
AU772153B2 (en) * 1999-02-12 2004-04-08 Molecular Insight Pharmaceuticals, Inc. Matrices for drug delivery and methods for making and using the same
DE10102053A1 (en) * 2001-01-17 2002-07-18 Merck Patent Gmbh Piperazinylcarbonyl-quinoline and piperazinylcarbonyl-isoquinoline derivatives useful for treatment of e.g. schizophrenia, psychoses, depression, Parkinson's disease and Alzheimer's disease

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5428130A (en) * 1989-02-23 1995-06-27 Genentech, Inc. Hybrid immunoglobulins
US5650150A (en) * 1990-11-09 1997-07-22 Gillies; Stephen D. Recombinant antibody cytokine fusion proteins
WO1998022577A1 (en) * 1996-11-15 1998-05-28 Maria Grazia Masucci Fusion proteins having increased half-lives
WO1998046257A1 (en) * 1997-04-17 1998-10-22 Amgen Inc. Compositions comprising conjugates of stable, active, human ob protein with antibody fc chain and methods
WO2001060412A2 (en) * 2000-02-15 2001-08-23 Genzyme Corporation Modification of biopolymers for improved drug delivery

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
CAPON D J ET AL: "DESIGNING CD4 IMMUNOADHESINS FOR AIDS THERAPY" NATURE, MACMILLAN JOURNALS LTD. LONDON, GB, vol. 337, 9 February 1989 (1989-02-09), pages 525-531, XP002935537 ISSN: 0028-0836 *
RUSSELL CHRIS S ET AL: "Recombinant proteins for genetic disease." CLINICAL GENETICS, vol. 55, no. 6, June 1999 (1999-06), pages 389-394, XP002231580 ISSN: 0009-9163 *

Cited By (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9750820B2 (en) 2003-11-13 2017-09-05 Hanmi Science Co., Ltd. IgG Fc fragment for a drug carrier and method for the preparation thereof
EP2256134A1 (en) * 2003-11-13 2010-12-01 Hanmi Pharmaceutical Co., Ltd. IgG Fc fragment for a drug carrier and method for the preparation thereof
US11058776B2 (en) 2003-11-13 2021-07-13 Hanmi Science Co., Ltd. IgG Fc fragment for a drug carrier and method for the preparation thereof
US8029789B2 (en) 2003-11-13 2011-10-04 Hanmi Holdings Co., Ltd. Method for the mass production of immunoglobulin constant region
US8110665B2 (en) 2003-11-13 2012-02-07 Hanmi Holdings Co., Ltd. Pharmaceutical composition comprising an immunoglobulin FC region as a carrier
US8822650B2 (en) 2003-11-13 2014-09-02 Hanmi Science Co., Ltd Method for the mass production of immunoglobulin constant region
US8846874B2 (en) 2003-11-13 2014-09-30 Hanmi Science Co., Ltd IgG Fc fragment for a drug carrier and method for the preparation thereof
US10272159B2 (en) 2003-11-13 2019-04-30 Hanmi Science Co., Ltd. IgG Fc fragment for a drug carrier and method for the preparation thereof
US10071166B2 (en) 2003-11-13 2018-09-11 Hanmi Science Co., Ltd. Protein complex using an immunoglobulin fragment and method for the preparation thereof
US8658174B2 (en) 2005-07-27 2014-02-25 Qinghua Wang GLP/1/exendin 4 IgG Fc fusion constructs for treatment of diabetes
WO2007012188A1 (en) * 2005-07-27 2007-02-01 Qinghua Wang GLP/1/EXENDM 4 IgG Fc FUSION CONSTRUCTS FOR TREATMENT OF DIABETES
US8742079B2 (en) 2007-08-20 2014-06-03 Protalix Ltd. Saccharide-containing protein conjugates and uses thereof
US9708595B2 (en) 2009-11-17 2017-07-18 Protalix Ltd. Stabilized alpha-galactosidase and uses thereof
US9194011B2 (en) 2009-11-17 2015-11-24 Protalix Ltd. Stabilized alpha-galactosidase and uses thereof
US10280414B2 (en) 2009-11-17 2019-05-07 Protalix Ltd. Stabilized α-galactosidase and uses thereof
US10870842B2 (en) 2009-11-17 2020-12-22 Protalix Ltd. Stabilized alpha-galactosidase and uses thereof
WO2011107991A1 (en) * 2010-03-02 2011-09-09 Protalix Ltd. Glucocerebrosidase multimers and uses thereof
US9732333B2 (en) 2011-01-20 2017-08-15 Protalix Ltd. Nucleic acid construct for expression of alpha-galactosidase in plants and plant cells
US11753479B2 (en) 2014-03-04 2023-09-12 Kymab Limited Nucleic acids encoding anti-OX40L antibodies
US11773175B2 (en) 2014-03-04 2023-10-03 Kymab Limited Antibodies, uses and methods
US10604576B2 (en) 2016-06-20 2020-03-31 Kymab Limited Antibodies and immunocytokines
US11779604B2 (en) 2016-11-03 2023-10-10 Kymab Limited Antibodies, combinations comprising antibodies, biomarkers, uses and methods

Also Published As

Publication number Publication date
ZA200306333B (en) 2004-11-17
HUP0401300A2 (en) 2004-09-28
HUP0401300A3 (en) 2005-06-28
JP2004525621A (en) 2004-08-26
PL362394A1 (en) 2004-11-02
EP1392826A2 (en) 2004-03-03
MXPA03006294A (en) 2003-09-16
KR20030067755A (en) 2003-08-14
CA2435037A1 (en) 2002-07-25
NO20033247L (en) 2003-07-17
CN1630720A (en) 2005-06-22
US20040043457A1 (en) 2004-03-04
WO2002057435A3 (en) 2003-12-24
NO20033247D0 (en) 2003-07-17
BR0116803A (en) 2004-02-17

Similar Documents

Publication Publication Date Title
US20040043457A1 (en) Bifunctional fusion proteins with glucocerebrosidase activity
EP1107989B1 (en) Expression and export of angiostatin and endostatin as immunofusins
US7211253B1 (en) Erythropoietin forms with improved properties
ES2068791T5 (en) METHODS AND DEOXIRRIBONUCLEIC ACID FOR THE PREPARATION OF TISSULAR FACTOR PROTEIN.
ES2581828T3 (en) Purification process of alpha-galactosidase A
US7122354B2 (en) Nucleic acid encoding a chimeric polypeptide
US5455337A (en) DNA encoding chimeric polypeptides comprising the interleukin-5 receptor α-chain fused to immunoglobulin heavy chain constant regions
US20040053366A1 (en) Expression and export of anti-obesity proteins as Fc fusion proteins
WO1998011206A9 (en) THERAPY FOR α-GALACTOSIDASE A DEFICIENCY
WO1996011955A1 (en) Antigen-binding fusion proteins
JPH10502810A (en) Novel cytokine named LERK-5
AU2002242643A1 (en) Bifunctional fusion proteins with glucocerebrosidase activity
JP2012511309A (en) EC-SOD carboxyl terminal apoptin protein transduction domain fusion protein
AU762400B2 (en) Therapy for alpha-galactosidase a deficiency
AU2008200265A1 (en) Therapy for alpha-galactosidase A deficiency

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

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

AL Designated countries for regional patents

Kind code of ref document: A2

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

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

Ref document number: 2001989057

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: PA/a/2003/006294

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 2002558488

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 1020037009521

Country of ref document: KR

Ref document number: 2435037

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 10466593

Country of ref document: US

Ref document number: 018220797

Country of ref document: CN

WWE Wipo information: entry into national phase

Ref document number: 2002242643

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2003/06333

Country of ref document: ZA

Ref document number: 200306333

Country of ref document: ZA

WWP Wipo information: published in national office

Ref document number: 1020037009521

Country of ref document: KR

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWP Wipo information: published in national office

Ref document number: 2001989057

Country of ref document: EP

WWW Wipo information: withdrawn in national office

Ref document number: 2001989057

Country of ref document: EP

DPE2 Request for preliminary examination filed before expiration of 19th month from priority date (pct application filed from 20040101)