WO2006000448A2 - Fc-INTERFERON-BETA FUSION PROTEINS - Google Patents

Fc-INTERFERON-BETA FUSION PROTEINS Download PDF

Info

Publication number
WO2006000448A2
WO2006000448A2 PCT/EP2005/006925 EP2005006925W WO2006000448A2 WO 2006000448 A2 WO2006000448 A2 WO 2006000448A2 EP 2005006925 W EP2005006925 W EP 2005006925W WO 2006000448 A2 WO2006000448 A2 WO 2006000448A2
Authority
WO
WIPO (PCT)
Prior art keywords
ifn
fusion protein
interferon
immunoglobulin
region
Prior art date
Application number
PCT/EP2005/006925
Other languages
French (fr)
Other versions
WO2006000448A3 (en
Inventor
Stephen D. Gillies
Nigel John Watkins
Matthew Paul Baker
King-Ming Lo
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
Application filed by Merck Patent Gmbh filed Critical Merck Patent Gmbh
Priority to AU2005256519A priority Critical patent/AU2005256519B2/en
Priority to JP2007517216A priority patent/JP4808709B2/en
Priority to EP05766761A priority patent/EP1761559B1/en
Priority to AT05766761T priority patent/ATE443720T1/en
Priority to CA2572015A priority patent/CA2572015C/en
Priority to DE602005016799T priority patent/DE602005016799D1/en
Publication of WO2006000448A2 publication Critical patent/WO2006000448A2/en
Publication of WO2006000448A3 publication Critical patent/WO2006000448A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/46Hybrid immunoglobulins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • A61K47/6811Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug being a protein or peptide, e.g. transferrin or bleomycin
    • A61K47/6813Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug being a protein or peptide, e.g. transferrin or bleomycin the drug being a peptidic cytokine, e.g. an interleukin or interferon
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/555Interferons [IFN]
    • C07K14/565IFN-beta
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • 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

  • the invention relates to Fc-fusion proteins. More specifically, the invention relates to high-level expression and secretion of Fc-interferon-beta fusion proteins and variant forms thereof, and methods of making and using such proteins.
  • Interferons are single chain polypeptides secreted by most animal cells in response to a variety of stimuli, including viruses, mitogens and cytokines. Interferons participate in the regulation of cell functions and mediate antiproliferative, antiviral and immunomodulatory effects. Thus, they are of great interest therapeutically. Native interferons are divided into three major types, based on the cell types from which they are primarily derived, namely, interferon- ⁇ (from leukocytes), interferon- ⁇ (from fibroblasts), interferon- ⁇ (from immune cells).
  • Interferon- ⁇ exhibits various biological and immunological activities and as a result has potential applications in immunotherapy, antitumor, anticancer and antiviral therapies. Numerous investigations and clinical trials have been and are being conducted based on anticancer and antiviral properties of both wild-type and recombinant IFN- ⁇ . Clinical trials using recombinant IFN- ⁇ in the treatment of multiple sclerosis also have been conducted. Most cytokines, including native IFN- ⁇ , have relatively short circulating half- lives. Consequently, in order for IFN- ⁇ to be effective as a therapeutic agent, it must be administered in large and frequent doses to a patient; however, this often leads to toxic side effects.
  • Human IFN- ⁇ (huIFN- ⁇ ) is a glycoprotein of 166 amino acids and has a four helix-bundle structure. Recombinant huIFN- ⁇ may be commonly produced for use as a therapeutic in either a prokaryotic or a mammalian expression system.
  • proteins that are normally secreted such as huEFN- ⁇
  • huEFN- ⁇ proteins that are normally secreted
  • the effect of prokaryotic expression on protein folding and on potential post-translational modifications needs to be addressed.
  • ER endoplasmic reticulum
  • most proteins destined for the extracellular milieu are folded in the oxidizing environment of the endoplasmic reticulum (ER), which promotes the correct formation of disulfide bonds, hi contrast, the reducing environment of the prokaryotic cytosol interferes with the formation of cysteine bonds.
  • ER endoplasmic reticulum
  • proteins expressed in prokaryotic systems lack some post-translational modifications, such as N-linked glycosylation, which likely aid in the correct folding of the protein, increase the stability of the folded protein, and decrease the immunogenicity of the administered protein.
  • N-linked glycosylation a post-translational modification that likely aid in the correct folding of the protein, increase the stability of the folded protein, and decrease the immunogenicity of the administered protein.
  • proteins expressed in prokaryotic systems lack some post-translational modifications, such as N-linked glycosylation, which likely aid in the correct folding of the protein, increase the stability of the folded protein, and decrease the immunogenicity of the administered protein.
  • proteins expressed in prokaryotic systems lack some post-translational modifications, such as N-linked glycosylation, which likely aid in the correct folding of the protein, increase the stability of the folded protein, and decrease the immunogenicity of the administered protein.
  • proteins expressed in prokaryotic systems lack some post-translational modifications, such as N-linked glycosylation, which likely aid
  • glycosylation of IFN- ⁇ does not seem to be essential for its biological activity, the specific activity of glycosylated IFN- ⁇ in bioassays is greater than that of the unglycosylated form.
  • IFN- ⁇ produced in a eukaryotic expression system such as a mammalian expression system, is substantially non-aggregated, but does form aggregates when the glycan moiety is removed. Therefore, the glycosylated form of IFN- ⁇ is desirable for therapeutic use as its biophysical properties are closer to those of the native protein than the unglycosylated form.
  • the invention provides methods and compositions for expressing soluble, biologically active Fc-IFN- ⁇ fusion proteins and variants thereof (Fc-IFN- ⁇ so1 ).
  • the Fc- IFN- ⁇ so! fusion proteins of the invention demonstrate improved biological properties over unaltered Fc-IFN- ⁇ proteins such as increased solubility, prolonged circulating half-life, enhanced biological activity, and reduced immunogenicity.
  • the invention provides a fusion protein including an Fc-IFN- ⁇ fusion protein including an immunoglobulin Fc region and an IFN- ⁇ protein linked to the carboxy-terminus of the immunoglobulin Fc region.
  • the IFN- ⁇ protein includes an amino acid alteration at at least one of positions 17, 50, 57, 130, 131, 136, and 140, corresponding to native mature interferon- ⁇ .
  • the alteration to the amino acid can be a deletion, substitution or modification, hi one embodiment, the amino acid alteration substitutes either serine, alanine, valine or methionine in place of cysteine at position 17. hi another embodiment, the amino acid alteration substitutes histidine in place of phenylalanine at position 50. In yet another embodiment, the amino acid alteration substitutes alanine in place of leucine at position 57, while in a further embodiment, the amino acid alteration substitutes alanine in place of leucine at position 130.
  • a further embodiment allows an amino acid alteration substituting alanine in place of histidine at position 131, while an additional embodiment contemplates substituting alanine in place of lysine at position 136.
  • the amino acid alteration substitutes alanine or threonine in place of histidine at position 140.
  • the immunoglobulin Fc region can include an immunoglobulin hinge region and an immunoglobulin heavy chain constant region.
  • the Fc region is derived from IgG4, while in another it is derived from IgGl, and in yet another it is derived from IgG2.
  • the Fc region is derived from IgG4 but includes a hinge region from IgGl.
  • the Fc region is derived from IgG2 but includes a hinge region derived from IgGl.
  • the C-terminal lysine of the immunoglobulin Fc region can be replaced by an alanine residue.
  • a cysteine residue of the hinge region is mutated.
  • the invention provides different methods for joining the Fc moiety and the IFN- ⁇ moiety to create fusion proteins according to the invention. For example, in one embodiment the immunoglobulin Fc region and the interferon- ⁇ protein are fused together by a peptide bond.
  • the immunoglobulin Fc region and the interferon- ⁇ protein are connected by a peptide linker sequence to facilitate protein folding.
  • the linker sequence preferably is composed of glycine and serine residues.
  • the peptide linker sequence is Gly 4 SerGly 4 SerGly 3 SerGly (SEQ ID NO: 1).
  • the Fc-interferon- ⁇ fusion protein includes amino acid alterations at positions 17, 50, 131, and 140 to improve folding and reduce aggregation, hi one specific embodiment, the amino acid alterations are serine substituted in place of cysteine at position 17, histidine substituted in place of phenylalanine at position 50, alanine substituted in place of histidine at position 131, and threonine or alanine substituted in place of histidine at position 140.
  • the Fc region includes IgGl, IgG2, or IgG4.
  • the fusion protein can also include a polypeptide linker sequence connecting the interferon- ⁇ protein and the immunoglobulin Fc region, hi one embodiment, a cysteine residue of the hinge region is mutated.
  • the Fc-interferon- ⁇ fusion protein includes amino acid alterations at positions 17, 57, 131, and 140, improving folding and reducing aggregation of the expressed fusion protein, hi one specific embodiment, the amino acid alterations are serine substituted in place of cysteine at position 17, alanine substituted in place of leucine at position 57, alanine substituted in place of histidine at position 131, and threonine or alanine substituted in place of histidine at position 140.
  • the Fc region includes IgGl, IgG2, or IgG4.
  • the fusion protein can also include a polypeptide linker sequence connecting the interferon- ⁇ protein and the immunoglobulin Fc region.
  • a cysteine residue of the hinge region is mutated.
  • the invention also provides methods for encoding and expressing fusion proteins of the invention. For example, one aspect of the invention relates to nucleic acid molecules encoding any of the aforementioned Fc-interferon- ⁇ fusion proteins, while in another aspect, the invention relates to cells containing a nucleic acid encoding any of the aforementioned Fc-interferon- ⁇ fusion proteins.
  • the nucleic acid molecules of the invention can be incorporated in operative association into a replicable expression vector which can then be introduced, for example, by transfection, into a mammalian host cell competent to produce the immunoglobulin Fc-IFN- ⁇ so1 fusion protein.
  • the vector includes a nucleic acid molecule encoding any one of the aforementioned Fc-interferon- ⁇ fusion proteins.
  • the invention also encompasses a replicable expression vector for transfecting a mammalian cell.
  • the vector includes a nucleic acid molecule encoding any one of the aforementioned Fc-interferon- ⁇ fusion proteins.
  • the invention relates to methods of stabilizing Fc-interferon- ⁇ fusion proteins, hi one embodiment, the method includes the step of making any of the aforementioned Fc-interferon- ⁇ fusion proteins.
  • the stabilizing includes increasing the circulating half-life of the Fc-interferon- ⁇ fusion protein relative to an unaltered Fc-interferon- ⁇ fusion protein, hi yet another embodiment, the stabilizing includes decreasing the aggregation of the Fc-interferon- ⁇ fusion protein relative to an unaltered Fc-interferon- ⁇ fusion protein, while in a further embodiment, the stabilizing includes increasing the biological activity of the Fc-interferon- ⁇ fusion protein relative to an unaltered Fc-interferon- ⁇ fusion protein.
  • a further aspect of the invention relates to methods for treating a patient for a condition alleviated by the administration of interferon- ⁇ .
  • the treatment includes administering an effective amount of any of the aforementioned interferon- ⁇ fusion proteins to a mammal having the condition
  • the method includes administering a nucleic acid encoding any of the aforementioned interferon- ⁇ fusion proteins to a mammal having the condition
  • the method includes administering a cell encoding any of the aforementioned interferon- ⁇ fusion proteins to a mammal having the condition.
  • An Fc-interferon- ⁇ fusion protein comprising: (i) an immunoglobulin Fc region; and (ii) an interferon- ⁇ protein linked by peptide bond or a peptide linker sequence to the carboxy-terminus of the immunoglobulin Fc region, wherein the interferon- ⁇ protein comprises an amino acid alteration at at least one of positions 17, 50, 57, 130, 131, 136 and 140 corresponding to the native mature interferon- ⁇ .
  • a corresponding fusion protein, wherein the amino acid alteration is a substitution.
  • a corresponding fusion protein having amino acid substitutions within the interferon- ⁇ protein which are selcted from the group consisting of: C17S, C17A, C17V, C17M; F50H, L57A, L130A, H131A, K136A, H140A, H140T. • A corresponding fusion protein, having amino acid substitutions within the interferon- ⁇ protein at positions 17 and 50 and 131 and 140, or 17 and 57 and 131 and 140. • A corresponding fusion protein, having the following amino acid substitutions within the interferon- ⁇ protein: C17S and F50H and H131A and H140T or A, or C17S and L57A and H131A and H140T or A.
  • a corresponding fusion protein wherein the immunoglobulin Fc region comprises an immunoglobulin hinge region and an immunoglobulin heavy chain constant region.
  • a corresponding fusion protein wherein the immunoglobulin Fc region is derived • A corresponding fusion protein, wherein the immunoglobulin Fc region is derived from IgG4 and further comprises an immunoglobulin hinge region derived from IgGl.
  • a corresponding fusion protein wherein a cysteine residue of the hinge region has been mutated.
  • a corresponding fusion protein wherein the immunoglobulin Fc region is derived from IgGl, and an alanine residue is substituted in place of the C-terminal lysine of the immunoglobulin Fc region.
  • a corresponding fusion protein wherein the immunoglobulin Fc region is derived from IgG2 and further comprises an immunoglobulin hinge region derived from IgGl. • A corresponding fusion protein, wherein a cysteine residue of the hinge region has been mutated. • A corresponding fusion protein, wherein further an alanine residue is substituted in place of the C-terminal lysine of the immunoglobulin Fc region. • A corresponding fusion protein, wherein the peptide linker sequence is Gly 4 SerGly 4 SerGly 3 SerGly. • A nucleic acid molecule encoding any one of the Fc-interferon- ⁇ fusion proteins above.
  • a replicable expression vector for transfecting a mammalian cell comprising a nucleic acid molecule encoding any one of the Fc-interferon- ⁇ fusion proteins as cited above.
  • a method of stabilizing an Fc-interferon- ⁇ fusion protein comprising the step of making an Fc-interferon- ⁇ fusion protein, wherein the Fc-interferon- ⁇ fusion comprises any one Fc-interferon- ⁇ fusion proteinas specified above.
  • stabilizing comprises (i) increasing the circulating half-life of the Fc-interferon- ⁇ fusion protein relative to an unaltered Fc-interferon- ⁇ fusion protein, or (ii) decreasing the aggregation of the Fc-interferon- ⁇ fusion protein relative to an unaltered Fc-interferon- ⁇ fusion protein, or (iii) increasing the biological activity of the Fc-interferon- ⁇ fusion protein relative to an unaltered Fc-interferon- ⁇ fusion protein.
  • a use of a corresponding Fc-interferon- ⁇ fusion protein for the manufacture of a medicament for the treatment of a condition of a patient, which is alleviated by the administration of interferon- ⁇ .
  • FIGS. 1A-1C are schematic illustrations of non-limiting examples of Fc-IFN- ⁇ so1 fusion proteins constructed in accordance with the invention.
  • FIG. 2 is a photograph of an SDS-PAGE gel showing the migration patterns of HuFc- ⁇ 4-IFN- ⁇ and HuFc- ⁇ 4h-IFN- ⁇ fusion proteins without the C17S mutation and HuFc- ⁇ 4h-IFN- ⁇ (C 17S) fusion proteins in both reducing and non-reducing chemical environments.
  • FIG. 3 is the amino acid sequence for mature IFN- ⁇ (SEQ ID NO:2).
  • FIG. 4 is the amino acid sequence for mature human IFN- ⁇ (C17S) (SEQ ID NO:3).
  • FIG. 5 is the amino acid sequence for human Fc-IFN- ⁇ sol (C17S) of the ⁇ 4 isotype with a modified ⁇ l hinge (Fc ⁇ 4h-IFN- ⁇ so1 ) (SEQ ID NO:4).
  • FIG. 6 is the amino acid sequence for human Fc-(linker)-IFN- ⁇ , starting with the CH3 domain of the Fc ⁇ 4 isotype (SEQ E) NO:5).
  • FIG. 7 is the amino acid sequence for human Fc-(linker)-IFN- ⁇ sol (C17S), starting with the CH3 domain of the Fc ⁇ 4 isotype (SEQ ID NO:6).
  • FIG. 8 is the amino acid sequence for human Fc-(linker)-IFN- ⁇ sol (C17S L57A H131A H140T) starting with the CH3 domain of the Fc ⁇ 4 isotype (SEQ ID NO:7).
  • FIG. 9 is the amino acid sequence for human Fc-(linker)-IFN- ⁇ sol (C17S L57A H131A H140A) starting with the CH3 domain of the Fc ⁇ 4 isotype (SEQ ID NO:8).
  • FIG. 10 is the amino acid sequence for human Fc-(linker)-IFN- ⁇ sol (C17S F50A H131 A 5 H140A), starting with the CH3 domain of the Fc ⁇ 4 isotype (SEQ ID NO:9).
  • FIG. 11 is the amino acid sequence for human Fc-(linker)-IFN- ⁇ sol (Cl 7S F50A H131 A H140T), starting with the CH3 domain of the Fc ⁇ 4 isotype (SEQ ID NO: 10).
  • FIG. 12 is the amino acid sequence for mature mouse IFN- ⁇ (SEQ ID NO:11).
  • FIG. 13 is the amino acid sequence for mature mouse IFN- ⁇ (C17S) (SEQ ID NO: 12).
  • FIGS. 14 is the nucleic acid sequence encoding the fusion protein embodiment huFc ⁇ 4h-IFN- ⁇ sol (C17S) ( ⁇ 4 isotype with modified ⁇ l hinge wherein the first cysteine of the ⁇ l hinge is replaced by a serine), starting from the hinge region (SEQ ID NO: 13).
  • FIGS. 14 is the nucleic acid sequence encoding the fusion protein embodiment huFc ⁇ 4h-IFN- ⁇ sol (C17S) ( ⁇ 4 isotype with modified ⁇ l hinge wherein the first cysteine of the ⁇ l hinge is replaced by a serine), starting from the hinge region (SEQ ID NO: 13).
  • 15-1 through 15-3 show the linearized nucleic acid sequence of the pdCs vector containing huFc ⁇ 4h-(linker)-IFN- ⁇ sol (C17S) ( ⁇ 4 isotype with modified ⁇ l hinge wherein the first cysteine of the ⁇ l hinge is replaced by a serine), wherein the Fc region and the IFN- ⁇ moiety are attached via a linker polypeptide (SEQ ID NO: 14).
  • FIGS. 16 is the nucleic acid sequence encoding the fusion protein embodiment HuFc- ⁇ 4h-(linlcer)-IFN- ⁇ sol (C17S) ( ⁇ 4 isotype with modified ⁇ l hinge wherein the first cysteine of the ⁇ l hinge is replaced by a serine), starting from the hinge region, wherein the Fc region and the IFN- ⁇ moiety are attached via a linker polypeptide (SEQ ID NO: 15).
  • 17-1 through 17-3 show the linearized nucleic acid sequence of the pdCs vector containing huFc ⁇ 4h-(linker)-IFN- ⁇ sol (C17S L57A H131A H140A) ( ⁇ 4 isotype with modified ⁇ l hinge wherein the first cysteine of the ⁇ l hinge is replaced by a serine), wherein the Fc region and the IFN- ⁇ moiety are attached via a linker polypeptide (SEQ ID NO: 16).
  • SEQ ID NO: 16 linker polypeptide
  • FIG. 18 is the nucleic acid sequence of huFc ⁇ 4h-(linker)-IFN- ⁇ sol (C17S L57A H131 H140A) ( ⁇ 4 isotype with modified ⁇ l hinge wherein the first cysteine of the ⁇ l hinge is replaced by a serine), starting from the hinge, wherein the Fc region and the IFN- ⁇ moiety are attached via a linker polypeptide (SEQ ID NO: 17).
  • FIG. 18 is the nucleic acid sequence of huFc ⁇ 4h-(linker)-IFN- ⁇ sol (C17S L57A H131 H140A) ( ⁇ 4 isotype with modified ⁇ l hinge wherein the first cysteine of the ⁇ l hinge is replaced by a serine), starting from the hinge, wherein the Fc region and the IFN- ⁇ moiety are attached via a linker polypeptide (SEQ ID NO: 17).
  • SEQ ID NO: 17 linker polypeptide
  • 19-1 through 19-3 shows the linearized nucleic acid sequence of the pdCs vector containing huFc ⁇ 4h-(linker)-IFN- ⁇ sol (C17S E50H H131 A H140A) ( ⁇ 4 isotype with modified ⁇ l hinge wherein the first cysteine of the ⁇ l hinge is replaced by a serine), wherein the Fc region and the IFN- ⁇ moiety are attached via a linker polypeptide (SEQ ID NO: 18).
  • huFc ⁇ 4h-(linker)-IFN- ⁇ sol C 17S F50H Hl 31 A H 140A
  • ⁇ 4 isotype with modified ⁇ l hinge wherein the first cysteine of the ⁇ l hinge is replaced by a serine starting from the hinge, wherein the Fc region and the IFN- ⁇ moiety are attached via a linker polypeptide (SEQ ID NO: 19).
  • IFN- ⁇ mediates antiproliferative, antiviral and immunomodulatory effects and, in addition to its usefulness in treating multiple sclerosis, it is anticipated that many other conditions may be alleviated by IFN- ⁇ administration.
  • IFN- ⁇ its usefulness as a treatment for a variety of malignancies, such as acute myeloid leukemia, multiple myeloma, Hodgkin's disease, basal cell carcinoma, cervical dysplasia and osteosarcoma is under evaluation.
  • IFN- ⁇ is also being tested as a therapeutic agent against a variety of viral infections, including viral hepatitis, herpes zoster and genitalis, papilloma viruses, viral encephalitis, and cytomegalovirus pneumonia.
  • viral infections including viral hepatitis, herpes zoster and genitalis, papilloma viruses, viral encephalitis, and cytomegalovirus pneumonia.
  • recombinant mature IFN- ⁇ has a short circulating half-life, making it suboptimal for use in therapy. Therefore there is a need in the art to produce variants of TFN- ⁇ with improved pharmacokinetic properties, including improved serum half-life.
  • One method known in the art for prolonging the half-life of small proteins involves linking them to an immunoglobulin Fc region.
  • Fusions in which an Fc region is placed at the N-terminus of a ligand have a number of useful properties (Lq et al, U. S. Patent Nos. 5,726,044 and 5,541,087; Lo et al (1998) Protein Engineering 11: 495).
  • leptin is administered to a mouse as an Fc-leptin fusion molecule (See, for example, PCT patent application publication WO 00/40615)
  • the circulating half-life of leptin increases from about 18 minutes to more than 8 hours.
  • Fc-X fusion proteins Another useful property of Fc-X fusion proteins is that the Fc portion generally has the effect of increasing protein production significantly. This is believed to occur, in part, because the Fc moiety of the fusion protein, commonly referred to as the expression cassette, is designed for efficient secretion of the fusion protein and, in part, because the fusion proteins can be produced in and secreted from host mammalian cells that naturally express immunoglobulin such that the fusion protein is readily secreted from the host cell.
  • the invention provides (i) nucleic acid sequences which facilitate efficient production of immunoglobulin Fc-IFN- ⁇ so1 fusion proteins; (ii) nucleic acid constructs for rapid and efficient production and secretion of immunoglobulin Fc-IFN- ⁇ so1 fusion proteins in a variety of mammalian host cells; and (iii) methods for the production, secretion, and purification of recombinant variants of immunoglobulin Fc-IFN- ⁇ so! fusion proteins.
  • the present invention provides nucleic acid molecules, for example, DNA or RNA molecules, which encode serially in the 5 ' to 3 ' direction, a polypeptide including an immunoglobulin Fc region and an IFN- ⁇ so1 protein .
  • the nucleic acid molecules of the invention can be incorporated in operative association into a replicable expression vector which may then be introduced, for example, by transfection, into a mammalian host cell competent to produce the immunoglobulin Fc-IFN- ⁇ so1 fusion protein.
  • the invention also provides methods of stabilizing immunoglobulin Fc-IFN- ⁇ fusion proteins. Although many proteins have been successfully produced and purified as Fc fusions, including many four-helix bundle proteins such as IL-2 (huFc-IL2), it has been found that Fc-IFN- ⁇ fusion proteins, where IFN- ⁇ belongs to the class of four-helix bundle proteins, form aggregates at least partly due to aberrant disulfide bonds present in the protein ("covalent aggregation").
  • Fc-IFN- ⁇ proteins form aggregates through non-covalent interactions as well ("non-covalent aggregation").
  • the present invention alleviates aggregation by providing methods of stabilizing Fc-IFN- ⁇ fusion proteins including the step of making an Fc-IFN- ⁇ so1 fusion protein, where the fusion protein includes an IFN- ⁇ protein having at one or more amino acid alterations, linked to the carboxy-terminus of an immunoglobulin Fc region.
  • stabilizing includes increasing the solubility of the Fc- IFN- ⁇ so1 fusion protein relative to an unaltered Fc-IFN- ⁇ fusion protein, increasing the circulating half-life of the Fc-IFN- ⁇ so1 fusion protein relative to an unaltered Fc-IFN- ⁇ fusion protein, and/or enhancing the biological activity of the Fc-IFN- ⁇ so1 fusion protein relative to an unaltered Fc-IFN- ⁇ fusion protein.
  • Increased stabilization is achieved in part by the elimination of aberrant disulfide bonding in the fusion protein and in part by reducing the amount of non-covalent aggregation of the fusion protein.
  • the invention also provides methods for treating conditions alleviated by IFN- ⁇ , bioactive fragments or active variants thereof by administering to a mammal an effective amount of IFN- ⁇ produced by a method of the invention and/or an Fc-IFN- ⁇ so1 fusion protein of the invention.
  • the invention also provides methods for treating conditions alleviated by IFN- ⁇ or active variants thereof by administering a nucleic acid of the invention, for example, a "naked DNA," or a vector containing a DNA or RNA of the invention, to a mammal having the condition.
  • the invention provides fusion proteins and nucleic acid molecules encoding those proteins including an altered IFN- ⁇ protein linked to the C-terminus of an immunoglobulin Fc region.
  • the IFN- ⁇ moiety can include one ore more mutations to the amino acid structure of the IFN- ⁇ moiety and Fc-IFN- ⁇ construct to improve the protein folding properties of the fusion protein, to reduce aggregation, and to improve protein expression.
  • the IFN- ⁇ moiety of the soluble fusion protein Fc-IFN- ⁇ so1 can contain an alteration at position 17, corresponding to a cysteine in the native mature IFN- ⁇ linked to the carboxy-terminus of an immunoglobulin Fc region.
  • the amino acid sequence for native mature human IFN- ⁇ is shown in FIG. 3.
  • the amino acid alteration at position 17 of the IFN- ⁇ protein can be generated by an amino acid substitution, amino acid deletion or amino acid modification through methods known in the art.
  • Preferred alterations to the IFN- ⁇ moiety include substituting either a serine (C17S), valine (C17V), alanine (C17A) or methionine (C17M) in place of the cysteine at position 17.
  • An exemplary amino acid sequence of a soluble human Fc-IFN- ⁇ fusion protein containing the C17S mutation is shown in FIG.
  • the invention also includes huFc-IFN- ⁇ sol (C17V), huFc-IFN- ⁇ sol(C17A) and huFc-IFN- ⁇ soI (C17M) fusion protein constructs. In addition to an alteration at position 17 of the mature IFN- ⁇ moiety, the invention provides Fc-IFN- ⁇ fusion proteins with other altered residues.
  • the IFN- ⁇ moiety can be altered at one or more of positions 17, 50, 57, 130, 131, 136, and 140 corresponding to, respectively, a cysteine, a phenylalanine, a lysine, a leucine, a histidine, a lysine, and a histidine in the native mature IFN- ⁇ protein.
  • the IFN- ⁇ moiety is linked to the carboxy-terminus of an immunoglobulin Fc region.
  • Alterations to the amino acid structure at one or more of positions 17, 50, 57, 130, 131, 136, and 140 can include an amino acid substitution, amino acid deletion or amino acid modification and can be generated through methods known in the art.
  • the phenylalanine at position 50 is replaced by histidine (F50H).
  • the leucine at position 57 is replaced by alanine (L57A).
  • the histidine at position 131 is replaced by alanine (H131A)
  • the histidine at 140 is replaced by either alanine (H 140A) or threonine (H 140T).
  • the leucine at position 130 is replaced by alanine (L130A), while in yet another embodiment, the lysine at residue 136 is replaced by alanine (K136A).
  • any suitable amino acid capable of conferring the appropriate properties on the fusion protein may be substituted in place of the original amino acid residue at position 17, 50, 57, 130, 131, 136, and/or 140 of the IFN- ⁇ moiety.
  • the invention contemplates an IFN- ⁇ moiety of an Fc-IFN- ⁇ so1 fusion protein having any combination of one, two, three, four, five, six, or seven of the alterations to positions 17, 50, 57, 130, 131, 136 and/or 140 as disclosed herein.
  • the Fc- IFN- ⁇ so1 in one embodiment contains amino acid alterations at one or more of F50, Hl 31 and H140 of the mature form of IFN- ⁇ , optionally combined with a C17 alteration.
  • the IFN- ⁇ moiety of the Fc-IFN- ⁇ so1 fusion protein contains amino acid alterations at one or more of L57, H131 and H140 of the mature form of IFN- ⁇ , optionally combined with a Cl 7 alteration.
  • IFN- ⁇ moiety of the Fc-IFN- ⁇ so1 fusion protein includes the alterations C17S, F50H, H131A, and/or H140A.
  • the IFN- ⁇ moiety of the Fc-IFN- ⁇ so1 fusion protein includes the alterations C17S, F50H, H13 IA, and/or H140T.
  • the IFN- ⁇ moiety of the Fc-IFN- ⁇ so1 fusion protein includes the alterations C17S, L57A, H131A, and/or H140A, while in a further embodiment, the fusion protein includes the alterations C17S, L57A, H131 A, and/or H140T.
  • the Fc region is preferably a human Fc region.
  • Another embodiment of the invention includes nucleic acid sequences encoding Fc-IFN- ⁇ so1 variants with at least one codon substitution in the mature human IFN- ⁇ protein sequence, hi one embodiment, a codon substitution replaces the cysteine corresponding to position 17 in the mature human IFN- ⁇ sequence with a serine (C 17S). Expression of this nucleotide sequence, contained on an appropriate plasmid, in a mammalian cell culture system resulted in the efficient production of the fusion protein huFc-huIFN- ⁇ soI (C17S). In alternative embodiments, a codon substitution replaces the cysteine at position 17 with either an alanine, a valine, or a methionine.
  • nucleic acid sequence encoding a representative Fc-IFN- ⁇ so1 fusion protein huFc ⁇ 4h-IFN- ⁇ sol(C17S), starting from the hinge, is disclosed in FIG. 14 (SEQ ID NO: 13).
  • the invention also includes nucleic acid sequences encoding Fc- IFN- ⁇ so1 variants with codon substitutions replacing amino acids at one or more of positions 17, 50, 57, 130, 131, 136 and/or 140. Nucleic acids incorporating the altered codons of the invention can be created using methods known in the art.
  • the immunoglobulin Fc region and the IFN- ⁇ moiety of an Fc-IFN- ⁇ so1 fusion protein can be linked to one another in a variety of ways. While the C-terminus of the Fc moiety may be directly linked to the N-terminus of the IFN- ⁇ moiety via a peptide bond, the invention additionally includes connecting the Fc moiety and the IFN- ⁇ moiety via a linker peptide.
  • the linker peptide is located between the C-terminus of the Fc moiety and the N-terminus of the mature IFN- ⁇ moiety.
  • the invention also includes a nucleic acid sequence encoding the linker peptide.
  • the linker peptide is preferably composed of serine and glycine residues.
  • the linker has the amino acid sequence Gly 4 SerGly 4 SerGly 3 SerGly (SEQ ID NO:1), while in yet another embodiment a nucleic acid encoding an Fc-IFN- ⁇ so1 includes a nucleic acid sequence encoding the linker peptide Gly 4 SerGly 4 SerGly 3 SerGly (SEQ ID NO: 1).
  • Fc-linker-IFN- ⁇ so1 amino acid sequences of the invention are shown in FIGS. 6-11, while some exemplary Fc- linker-IFN- ⁇ so1 nucleic acid sequences of the invention are shown in FIGS. 14-16.
  • the Fc-linker-IFN- ⁇ so1 protein is huFc ⁇ 4-linker-IFN- ⁇ sol (C17S), wherein the Fc region is an IgG4 Fc region, and the linker is Gly 4 SerGly 4 SerGly 3 SerGly (SEQ ID NO:1).
  • Fc-IFN- ⁇ so1 and Fc-linker-IFN- ⁇ so1 nucleotide sequences such as those previously discussed, when contained on an appropriate plasmid, in a mammalian cell culture system will result in the efficient production of Fc-IFN- ⁇ so1 and Fc-linker-IFN- ⁇ so1 fusion proteins.
  • Fc-IFN- ⁇ so1 fusion proteins of the invention demonstrate unproved biological properties over unaltered Fc-IFN- ⁇ fusion proteins.
  • human Fc ⁇ 4h-IFN- ⁇ sol displayed folding properties that were different from, and improved over, the parent fusion protein Fc ⁇ 4-IFN- ⁇ so1 and Fc ⁇ 4h- IFN ⁇ so1 .
  • FIG. 2 it was found that predominantly a single species of the human Fc ⁇ 4h-IFN- ⁇ soI (C17S) fusion protein 3, 4 was seen when expressed in mammalian tissue culture cells, as ascertained by non-reducing SDS-PAGE gel analysis. This species corresponded to the correctly folded Fc ⁇ 4-IFN- ⁇ so1 fusion protein 3, 4.
  • the invention provides nucleic acid sequences encoding and amino acid sequences defining fusion proteins including an immunoglobulin Fc region and at least one target protein, referred to herein as IFN- ⁇ or variants thereof.
  • IFN- ⁇ or variants thereof Three exemplary embodiments of protein constructs embodying the invention are illustrated in the drawing as FIGS. IA-I C. Because dimeric constructs are preferred, all are illustrated as dimers cross-linked by a pair of disulfide bonds between cysteines in adjacent subunits.
  • the disulfide bonds 11, 12 are depicted as linking together the two immunoglobulin heavy chain Fc regions 1, 1' via an immunoglobulin hinge region within each heavy chain, and thus are characteristic of native forms of these molecules. While constructs including the hinge region of Fc are preferred and have shown promise as therapeutic agents, the invention contemplates that the crosslinking at other positions may be chosen as desired. Furthermore, under some circumstances, dimers or multimers useful in the practice of the invention may be produced by non- covalent association, for example, by hydrophobic interaction. Because homodimeric constructs are important embodiments of the invention, the drawings illustrate such constructs. It should be appreciated, however, that heterodimeric structures also are useful in the practice of the invention. FIG.
  • IA illustrates a dimeric construct, also termed a "unit dimer", produced in accordance with the principles set forth herein (see, for example, Example 1).
  • Each monomer of the homodimer includes an immunoglobulin Fc region 1 including a hinge region, a CH2 domain and a CH3 domain. Attached directly, i.e., via a polypeptide bond, to the C terminus of the Fc region is IFN- ⁇ so1 2. It should be understood that the Fc region may be attached to IFN- ⁇ so1 protein via a polypeptide linker (not shown).
  • FIGS. IB and 1C depict protein constructs of the invention which include as a target protein plural IFN- ⁇ proteins arranged in tandem and connected by a linker. In FIG.
  • the target protein includes full length IFN- ⁇ 2, a polypeptide linker made of glycine and serine residues 4, and an active variant of IFN- ⁇ 3.
  • This construct may be depicted by the formula Fc-X-X wherein the Xs represent different target proteins.
  • FIG. 1C differs from the construct of FIG. IB in that the most C-terminal protein domain includes a second, full length copy of IFN- ⁇ 2.
  • This construct may be depicted by the formula Fc-X-X, where the Xs represent identical target proteins.
  • the fusion protein may include a second target protein (Fc-X-X).
  • Fc-X-X a second target protein
  • the fusion protein may also include a second mature, full length IFN- ⁇ or an active IFN- ⁇ so1 variant or a bioactive fragment thereof.
  • the active variant is a variant in which one or more amino acid residues in the IFN- ⁇ moiety is substituted for another amino acid residue.
  • the first and second proteins can be the same protein, as in, for example, FIG. 1C, or they may be different proteins, as in, for example, FIG. IB.
  • the first and second proteins may be linked together, either directly or by means of a polypeptide linker. Alternatively, both proteins may be linked either directly or via a polypeptide linker, to the immunoglobulin Fc region.
  • the first protein can be connected to the N-terminus of the immunoglobulin Fc region and the second protein can be connected to the C-terminus of the immunoglobulin Fc region.
  • two fusion proteins may be linked to form dimers.
  • the two fusion proteins may associate, either covalently, for example, by a disulfide bond, a polypeptide bond or a crosslinking agent, or non-covalently, to produce a dimeric protein, hi a preferred embodiment, the two fusion proteins are associated covalently by means of at least one and more preferably two interchain disulfide bonds via cysteine residues, preferably located within immunoglobulin hinge regions disposed within the immunoglobulin Fc regions of each chain.
  • multivalent and multimeric forms of IFN- ⁇ fusion proteins and combinations thereof include multivalent and multimeric forms of IFN- ⁇ fusion proteins and combinations thereof.
  • multivalent refers to a recombinant molecule that incorporates two or more biologically active segments.
  • the protein fragments forming the multivalent molecule optionally may be linked through a polypeptide linker which attaches the constituent parts and permits each to function independently.
  • bivalent refers to a multivalent recombinant molecule having the configuration Fc-X, where X is an IFN- ⁇ protein.
  • the two proteins may be linked through a peptide linker. Constructs of the type shown can increase the apparent binding affinity between the protein and its receptor.
  • multimeric refers to the stable association of two or more polypeptide chains either covalently, for example, by means of a covalent interaction, for example, a disulfide bond, or non-covalently, for example, by hydrophobic interaction.
  • the term multimer is intended to encompass both homomultimers, wherein the subunits are the same, as well as, heteromultimers, wherein the subunits are different.
  • dimeric refers to a specific multimeric molecule where two polypeptide chains are stably associated through covalent or non-covalent interactions. Such constructions are shown schematically in FIG. IA.
  • the immunoglobulin Fc region including at least a portion of the hinge region, a CH2 domain and a CH3 domain, typically forms a dimer.
  • Many protein ligands are known to bind to their receptors as a dimer. If a protein ligand X dimerizes naturally, the X moiety in an Fc-X molecule will dimerize to a much greater extent, since the dimerization process is concentration dependent. The physical proximity of the two X moieties connected by Fc would make the dimerization an intramolecular process, greatly shifting the equilibrium in favor of the dimer and enhancing its binding to the receptor.
  • polypeptide linker is understood to mean a polypeptide sequence that can link together two proteins that in nature are not naturally linked together.
  • the polypeptide linker preferably includes a plurality of amino acids such as ' alanine, glycine and serine or combinations of such amino acids.
  • the polypeptide linker includes a series of glycine and serine peptides about 10-15 residues in length. See, for example, U.S. Patent Nos. 5,258,698 and 5,908,626.
  • a preferred linker polypeptide of the invention is Gly 4 SerGly 4 SerGly 3 SerGly (SEQ ID NO:1).
  • interferon- ⁇ or IFN- ⁇ is understood to mean not only full length mature interferon- ⁇ , for example, human IFN- ⁇ , but also homologs, variants and bioactive fragments or portions thereof. Known sequences of IFN- ⁇ may be found in GenBank.
  • interferon- ⁇ or "IFN- ⁇ ” also includes naturally occurring IFN- ⁇ and IFN- ⁇ -like proteins, moieties and molecules as well as IFN- ⁇ that is recombinantly produced or artificially synthesized.
  • bioactive fragment refers to any IFN- ⁇ protein fragment that has at least 5%, more preferably at least 10%, and most preferably at least 20% and optimally at least 30%, 40%, 50%, 60%, 70%, 80%, 90% or 100% of the biological activity of the template human IFN- ⁇ protein of SEQ ID NO:2, determined using the antiviral activity assay or cellular growth inhibition assays, as described in Examples 6 and 7.
  • variants includes species and allelic variants, as well as other naturally occurring or non-naturally occurring variants, for example, generated by genetic engineering protocols, that are at least 70% similar or 60% identical, more preferably at least 75% similar or 65% identical, and most preferably at least 80% similar or 70% identical to the mature human IFN- ⁇ protein disclosed in SEQ ID NO:2.
  • the candidate amino acid sequence and the reference amino acid sequence are first aligned using the dynamic programming algorithm described in Smith and Waterman (1981) J. MOL. BIOL. 147:195-197, in combination with the BLOSUM62 substitution matrix described in FIG.
  • the raw similarity score is the sum of the pair-wise similarity scores of the aligned amino acids.
  • the raw score then is normalized by dividing it by the number of amino acids in the smaller of the candidate or reference sequences. The normalized raw score is the percent similarity.
  • the aligned amino acids of each sequence again are compared sequentially. If the amino acids are non- identical, the pair-wise identity score is zero; otherwise the pair-wise identity score is 1.0.
  • the raw identity score is the sum of the identical aligned amino acids.
  • the raw score is then normalized by dividing it by the number of amino acids in the smaller of the candidate or reference sequences.
  • the normalized raw score is the percent identity. Insertions and deletions are ignored for the purposes of calculating percent similarity and identity. Accordingly, gap penalties are not used in this calculation, although they are used in the initial alignment.
  • Variants may also include other IFN- ⁇ mutant proteins having IFN- ⁇ -like activity. Species and allelic variants, include, but are not limited to human and mouse IFN- ⁇ sequences. The human and mouse mature IFN- ⁇ proteins are depicted in SEQ ID NOs. :2 and 11, and in FIGS. 3 and 12 respectively.
  • the IFN- ⁇ sequence may include a portion or all of the consensus sequence set forth in SEQ ID NO:2, wherein the IFN- ⁇ has at least 5%, preferably at least 10%, more preferably at least 20%, 30% or 40%, most preferably at least 50%, and optimally 60%, 70%, 80%, 90% or 100% of the biological activity of the mature human IFN- ⁇ of SEQ ID NO:2, as determined using the antiviral activity assay or cellular growth inhibition assay of Examples 6 and 7.
  • the three-dimensional structure of IFN- ⁇ has been solved by X-ray crystallography (Karpusas et al, 1997, PNAS 94: 11813).
  • IFN- ⁇ is a dimer with a zinc ion at the dimer interface
  • IFN- ⁇ need not be a dimer in order to be active.
  • Structurally IFN- ⁇ contains an additional alpha-helical segment with respect to classical four helix bundle proteins, which is formed within the C-D loop, so that the canonical bundle structure is formed by helices A, B, C and E.
  • the structure also reveals a portion of the glycan moiety which is coupled to amino acid N80 at the start of helix C and is ordered along a portion of the protein, most likely shielding some of the surface-exposed hydrophobic amino acid residues from solvent.
  • the second interferon-beta moiety of the same Fc-Interferon-beta fusion protein may bind to a second receptor on the same cell with a much higher avidity (apparent affinity). This may occur because of the physical proximity of the second interferon-beta moiety to the receptor after the first interferon-beta moiety already is bound.
  • the apparent affinity may be increased by at least ten thousand-fold, i.e., 104.
  • Each protein subunit i.e., "X” has its own independent function so that in a multivalent molecule, the functions of the protein subunits may be additive or synergistic.
  • fusion of the normally dimeric Fc molecule to interferon-beta may increase the activity of interferon-beta.
  • constructs of the type shown in FIG. IA may increase the apparent binding affinity between interferon-beta and its receptor.
  • Fc Moiety The IFN- ⁇ fusion proteins disclosed herein are expressed as fusion proteins with an Fc region of an immunoglobulin. As is known, each immunoglobulin heavy chain constant region includes four or five domains.
  • the domains are named sequentially as follows: CHl-hinge-CH2-CH3(-CH4).
  • the DNA sequences of the heavy chain domains have cross-homology among the immunoglobulin classes, e.g., the CH2 domain of IgG is homologous to the CH2 domain of IgA and IgD 5 and to the CH3 domain of IgM and IgE.
  • immunoglobulin Fc region is understood to mean the carboxyl-terminal portion of an immunoglobulin chain constant region, preferably an immunoglobulin heavy chain constant region, or a portion thereof.
  • an immunoglobulin Fc region may include 1) a CH2 domain 2) a CH3 domain, 3) a CH4 domain 4) a CH2 domain and a CH3 domain, 5) a CH2 domain and a CH4 domain, 6) a CH3 domain and a CH4 domain or 7) a combination of an immunoglobulin hinge region and/or a CH2 domain and/or CH3 domain and/or a CH4 domain.
  • the immunoglobulin Fc region includes at least an immunoglobulin hinge region, while in another embodiment the immunoglobulin Fc region includes at least one immunoglobulin constant heavy region, for example, a CH2 domain or a CH3 domain, and depending on the type of immunoglobulin used to generate the Fc region, optionally a CH4 domain.
  • the Fc region includes a hinge region, a CH2 domain and a CH3 domain, and preferably lacks the CHl domain, while in another embodiment, the Fc region includes a hinge region and a CH2 domain.
  • the Fc region includes a hinge region and a CH3 domain.
  • the Fc region contains a functional binding site for the Fc protection receptor, FcRp.
  • the binding site for FcRp includes amino acids in both the CH2 and CH3 domains and the Fc- FcRp interaction contributes significantly to the extended serum half-life of Fc fusion proteins.
  • immunoglobulin Fc regions and component constant heavy domains may be from any immunoglobulin class, a preferred class of immunoglobulin for the Fc- IFN- ⁇ fusion proteins of the invention is IgG (Ig ⁇ ) ( ⁇ subclasses 1, 2, 3, or 4).
  • the nucleotide and amino acid sequences of human Fc ⁇ l are set forth in SEQ DD NOs: 78 and 79.
  • immunoglobulin IgA (Ig ⁇ ), IgD (Ig ⁇ ), IgE (Ig ⁇ ) and IgM (Ig ⁇ )
  • IgA immunoglobulin
  • IgD immunoglobulin
  • IgE immunoglobulin heavy chain constant region
  • IgM immunoglobulin M
  • the portion of the DNA construct encoding the immunoglobulin Fc region preferably includes at least a portion of a hinge domain, and preferably at least a portion of a CH3 domain of Fc ⁇ or the homologous domains in any of IgA, IgD, IgE, or IgM. It is contemplated that the Fc region used in the generation of the fusion proteins containing the IFN- ⁇ variants can be adapted to the specific application of the molecule, hi one embodiment, the Fc region is derived from an immunoglobulin ⁇ l isotype or variants thereof. The use of human Fc ⁇ l as the Fc region sequence has several advantages.
  • an Fc region derived from an immunoglobulin ⁇ l isotype can be used when targeting the fusion protein to the liver is desired.
  • the Fc ⁇ l domain may confer effector function activities to the fusion protein.
  • the effector function activities include the biological activities such as placental transfer and increased serum half-life.
  • the immunoglobulin Fc region also provides for detection by anti-Fc ELISA and purification through binding to Staphylococcus aureus protein A ("Protein A"). Ih certain applications, however, it may be desirable to delete specific effector functions from the immunoglobulin Fc region, such as Fc receptor binding and/or complement fixation.
  • Fc-IFN- ⁇ so1 fusion proteins use Fc regions derived from a different immunoglobulin ⁇ isotype i.e. ⁇ 2, ⁇ 3, or ⁇ 4, or variants thereof.
  • the Fc region can include a hinge region derived from a different immunoglobulin isotype than the Fc region itself.
  • Fc-IFN- ⁇ so1 fusion proteins contain a hinge region derived from an immunoglobulin ⁇ l or a variant thereof.
  • the immunoglobulin Fc region can be derived from an immunoglobulin ⁇ 2 isotype and include a hinge region derived from an immunoglobulin ⁇ l isotype or a variant thereof.
  • a cysteine residue of the ⁇ l hinge is modified, hi a further embodiment, the first cysteine of the ⁇ l l ⁇ nge is modified.
  • a serine is substituted in place of the first cysteine of the ⁇ l hinge.
  • the immunoglobulin ⁇ 2 isotype is ineffective in mediating effector functions and displays vastly reduced binding to Fc ⁇ receptor (Fc ⁇ R), it may be expected that this particular configuration of IFN- ⁇ fusion variant more closely mimics the biological activity of the free IFN- ⁇ molecule and in addition has the most enhanced circulating half-life when administered to a mammal.
  • Fc ⁇ R Fc ⁇ receptor
  • the Fc region of Fc-IFN- ⁇ so1 fusion proteins of the invention can be derived from an immunoglobulin ⁇ 4 isotype.
  • an immunoglobulin ⁇ 4 isotype is modified to contain a hinge region derived from an immunoglobulin ⁇ l isotype or a variant thereof, hi one embodiment, a cysteine residue of the ⁇ l hinge is modified, hi a further embodiment, the first cysteine of the ⁇ l hinge is modified, hi yet another embodiment, a serine is substituted in place of the first cysteine of the ⁇ l hinge.
  • immunoglobulin ⁇ 4 isotypes also exhibit lower affinity towards Fc ⁇ R and thus offer similar advantages in reducing immune effector functions.
  • Immunoglobulin ⁇ 4 is a preferred Fc region for making Fc-EFN- ⁇ so1 fusion proteins wherein the IFN- ⁇ moiety includes alterations to one of more amino acid residues at position 17, 50, 57, 130, 131, 136 and/or 140.
  • An exemplary amino acid sequence of an Fc-IFN- ⁇ so1 fusion protein of the invention which includes an Fc region of immunoglobulin ⁇ 4 isotype modified to contain a hinge region derived from an immunoglobulin ⁇ l is shown in FIG. 5 (SEQ ID NO:4).
  • the immunoglobulin Fc region used as a fusion partner in the DNA construct generally may be from any mammalian species. Where it is undesirable to elicit an immune response in the host cell or animal against the Fc region, the Fc region may be derived from the same species as the host cell or animal.
  • a human immunoglobulin Fc region can be used when the host animal or cell is human; likewise, a murine immunoglobulin Fc region can be used where the host animal or cell will be a mouse.
  • Nucleic acid sequences encoding, and amino acid sequences defining a human immunoglobulin Fc region useful in the practice of the invention are set forth in SEQ ID NO:78 and SEQ ID NO:79 respectively.
  • other immunoglobulin Fc region sequences useful in the practice of the invention may be found, for example, by those encoded by nucleotide sequences of the heavy chain constant region which includes the Fc region sequence as disclosed in the Genbank and/or EMBL databases, for example, AF045536.1 (Macaca fuscicularis, nucleotide sequence SEQ ID NO:20; amino acid sequence SEQ ID NO:21), AF045537.1 (Macaca mulatto, nucleotide sequence SEQ ID NO:22; amino acid sequence SEQ ID NO:23), AB016710 (Felis catus, nucleotide sequence SEQ ID NO:24; amino acid sequence SEQ E) NO:25), K00752 (Oryctolagus cuniculus,
  • substitution or deletion of amino acids within the immunoglobulin heavy chain constant regions may be useful in the practice of the invention.
  • One example may include introducing amino acid substitutions in the upper CH2 region to create an Fc variant with reduced affinity for Fc receptors (Cole et al. (1997) J. Immunol. 159:3613).
  • One of ordinary skill in the art can prepare such constructs using well known molecular biology techniques. It is understood that the present invention exploits conventional recombinant DNA methodologies for generating the Fc fusion proteins useful in the practice of the invention.
  • the Fc fusion constructs preferably are generated at the DNA level, and the resulting DNAs integrated into expression vectors, and expressed to produce the fusion proteins of the invention.
  • vector is understood to mean any nucleic acid including 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.
  • 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.
  • gene expression or "expression” of a target protein, is understood to mean the transcription of a DNA sequence, translation of the mRNA transcript, and secretion of an Fc fusion protein product.
  • a useful expression vector is pdCs (Lo et al. (1988) Protein Engineering 11 :495), in which the transcription of the Fc-X gene utilizes the enhancer/promoter of the human cytomegalovirus and the SV40 polyadenylation signal.
  • the enhancer and promoter sequence of the human cytomegalovirus used was derived from nucleotides -601 to +7 of the sequence provided in Boshart et al. (1985) Cell 41:521.
  • the vector also contains the mutant dihydrofolate reductase gene as a selection marker (Simonsen and Levinson (1983) Proc. Nat. Acad. Sci. USA 80:2495).
  • An appropriate host cell can be transformed or transfected with the DNA sequence of the invention, and utilized for the expression and/or secretion of the target protein.
  • Currently preferred host cells for use in the invention include immortal hybridoma cells, NS/0 myeloma cells, 293 cells, Chinese hamster ovary cells, HeLa cells, and COS cells.
  • One expression system that has been used to produce high level expression of fusion proteins in mammalian cells is a DNA construct encoding, in the 5' to 3' direction, a secretion cassette, including a signal sequence and an immunoglobulin Fc region, and a target protein such as IFN- ⁇ .
  • fusion proteins of the invention may or may not be include a signal sequence when expressed.
  • signal sequence is understood to mean a segment which directs the secretion of the IFN- ⁇ fusion protein and thereafter is cleaved following translation in the host cell.
  • the signal sequence of the invention is a polynucleotide which encodes an amino acid sequence which initiates transport of a protein across the membrane of the endoplasmic reticulum.
  • Signal sequences which are useful in the invention include antibody light chain signal sequences, e.g., antibody 14.18 (Gillies et. al. (1989) J. Immunol. Meth. 125:191), antibody heavy chain signal sequences, e.g., the M0PC141 antibody heavy chain signal sequence (Sakano et al. (1980) Nature 286:5774), and any other signal sequences which are known in the art (see, e.g., Watson (1984) Nucleic Acids Research 12:5145).
  • 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.
  • the signal peptide is usually cleaved within the lumen of the endoplasmic reticulum by cellular enzymes known as signal peptidases.
  • the suitability of a particular signal sequence for use in the secretion cassette may require some routine experimentation. Such experimentation will include determining the ability of the signal sequence to direct the secretion of an Fc fusion protein and also a determination of the optimal configuration, genomic or cDNA, of the sequence to be used in order to achieve efficient secretion of Fc fusion proteins. Additionally, one skilled in the art is capable of creating a synthetic signal peptide following the rules presented by von Heijne, referenced above, and testing for the efficacy of such a synthetic signal sequence by routine experimentation.
  • a signal sequence can also be referred to as a "signal peptide,” “leader sequence,” or “leader peptides.”
  • the fusion of the signal sequence and the immunoglobulin Fc region is sometimes referred to herein as secretion cassette.
  • An exemplary secretion cassette useful in the practice of the invention is a polynucleotide encoding, in a 5' to 3' direction, a signal sequence of an immunoglobulin light chain gene and an Fc ⁇ l region of the human immunoglobulin ⁇ l gene.
  • the Fc ⁇ l region of the immunoglobulin Fc ⁇ l gene preferably includes at least a portion of the immunoglobulin hinge domain and at least the CH3 domain, or more preferably at least a portion of the hinge domain, the CH2 domain and the CH3 domain.
  • the "portion" of the immunoglobulin hinge region is understood to mean a portion of the immunoglobulin hinge that contains at least one, preferably two cysteine residues capable of forming interchain disulfide bonds.
  • the DNA encoding the secretion cassette can be in its genomic configuration or its cDNA configuration. Under certain circumstances, it may be advantageous to produce the Fc region from human immunoglobulin Fc ⁇ 2 heavy chain sequences.
  • the DNA sequence encodes a proteolytic cleavage site interposed between the secretion cassette and the target protein.
  • a cleavage site provides for the proteolytic cleavage of the encoded fusion protein thus separating the Fc domain from the target protein.
  • proteolytic cleavage site is understood to mean amino acid sequences which are preferentially cleaved by a proteolytic enzyme or other proteolytic cleavage agents.
  • Useful proteolytic cleavage sites include amino acids sequences which are recognized by proteolytic enzymes such as trypsin, plasmin or enterokinase K. Many cleavage site/cleavage agent pairs are known (see, for example, U.S. Patent No. 5,726,044). Further, substitution or deletion of constructs of these constant regions, in which one or more amino acid residues of the constant region domains are substituted or deleted also would be useful. One example would be to introduce amino acid substitutions in the upper CH2 region to create an Fc variant with reduced affinity for Fc receptors (Cole et al. (1997) J. Immunol. 159: 3613). One of ordinary skill in the art can prepare such constructs using well known molecular biology techniques.
  • the fusion constructs disclosed herein produced high levels of Fc-IFN- ⁇ so1 .
  • the initial clones produced about 100 ⁇ g/mL of altered Fc-IFN- ⁇ so1 , which could be purified to homogeneity by Protein A affinity chromatography. Expression levels often can be increased several fold by subcloning.
  • Fc-IFN- ⁇ so1 fusion proteins of the invention including the mutation C17S, such as, for example Fc-(linker)-IFN- ⁇ so1 (C17S F50H H131A H140A) and Fc-(linker)-IFN- ⁇ sol (C17S L57A H131A H140T) are equally well expressed.
  • the Fc region is also glycosylated and highly charged at physiological pH. Therefore, the Fc region can help to solubilize hydrophobic proteins.
  • Fc-IFN- ⁇ fusion proteins exhibited greater bioactivity than the parental (un-modified) Fc-IFN- ⁇ fusion protein, as measured in a cell based anti-viral assay (Example 6), and were comparable to the bioactivity of a commercial preparation of IFN- ⁇ obtained from R&D Systems (Minneapolis, MN).
  • altered Fc-IFN- ⁇ fusion proteins exhibited longer serum half-lives compared to unaltered Fc-IFN- ⁇ fusion proteins.
  • the circulating half-life of Fc-IFN- ⁇ so1 including the mutation C17S is found to be significantly greater than that of the parent Fc-IFN- ⁇ fusion protein (see Example 8).
  • the fusion proteins of the invention provide several important clinical benefits.
  • Another embodiment of the present invention provides constructs having various structural conformations, e.g., bivalent or multivalent constructs, dimeric or multimeric constructs, and combinations thereof. Such functional conformations of molecules of the invention allow the synergistic effect of IFN- ⁇ and other anti- viral and anti-cancer proteins to be explored in animal models. 15 An important aspect of the invention is that the sequences and properties of various IFN- ⁇ proteins and encoding DNAs are quite similar.
  • the properties of IFN- ⁇ proteins and encoding DNAs are essentially identical, so that a common set of techniques can be used to generate any Fc-IFN- ⁇ DNA fusion, to express the fusion, to purify the fusion protein, and to administer the fusion protein for 0 therapeutic purposes.
  • the present invention also provides methods for the production of IFN- ⁇ of non- human species as Fc fusion proteins.
  • Non-human IFN- ⁇ fusion proteins are useful for preclinical studies of IFN- ⁇ because efficacy and toxicity studies of a protein drug must be performed in animal model systems before testing in human beings.
  • a human protein 25 may not work in a mouse model since the protein may elicit an immune response, and/or exhibit different pharmacokinetics skewing the test results. Therefore, the equivalent mouse protein is the best surrogate for the human protein for testing in a mouse model.
  • the present invention provides methods of treating various cancers, viral diseases, other diseases, related conditions and causes thereof by administering the DNA, RNA or 30 proteins of the invention to a mammal having such condition.
  • Related conditions may include, but are not limited to multiple sclerosis; a variety of malignancies, such as acute myeloid leukemia, multiple myeloma, Hodgldn's disease, basal cell carcinoma, cervical dysplasia and osteosarcoma; a variety of viral infections, including viral hepatitis, herpes zoster and genitalis, papilloma viruses, viral encephalitis, and cytomegalovirus pneumonia.
  • the present invention also provides methods for treating conditions alleviated by the administration of IFN- ⁇ .
  • a nucleic acid such as DNA or RNA
  • encoding an Fc-IFN- ⁇ so! fusion protein can be administered to a subject, preferably a mammal, as a therapeutic agent.
  • a cell containing a nucleic acid encoding an Fc-IFN- ⁇ so1 fusion protein can be administered to a subject, preferably a mammal, as a therapeutic agent.
  • an Fc-IFN- ⁇ so1 protein can be administered to a subject, preferably a mammal, as a therapeutic agent.
  • compositions of the present invention may be administered by any route which is compatible with the particular molecules. It is contemplated that the compositions of the present invention may be provided to an animal by any suitable means, directly (e.g. , locally, as by injection, implantation or topical administration to a tissue locus) or systemically (e.g., parenterally or orally).
  • the composition preferably includes part of an aqueous or physiologically compatible fluid suspension or solution.
  • the carrier or vehicle is physiologically acceptable so that in addition to delivery of the desired composition to the patient, it does not otherwise adversely affect the patient's electrolyte and/or volume balance.
  • the fluid medium for the agent thus can include normal physiologic saline.
  • the DNA constructs (or gene constructs) of the invention also can be used as a part of a gene therapy protocol to deliver nucleic acids encoding IFN- ⁇ or a fusion protein construct thereof.
  • the invention features expression vectors for in vivo transfection and expression of IFN- ⁇ or a fusion protein construct thereof in particular cell types so as to reconstitute or supplement the function of IFN- ⁇ .
  • Expression constructs of IFN- ⁇ , or fusion protein constructs thereof may be administered in any biologically effective carrier, e.g. any formulation or composition capable of effectively delivering the IFN- ⁇ gene or fusion protein construct thereof to cells in vivo.
  • Approaches include insertion of the subject gene in viral vectors including recombinant retroviruses, adenovirus, adeno- associated virus, and herpes simplex virus- 1, or recombinant bacterial or eukaryotic plasmids.
  • Preferred dosages per administration of nucleic acids encoding the fusion proteins of the invention are within the range of 1 ⁇ g/m2 to 100 mg/m2, more preferably 20 ⁇ g/m2 to 10 mg/m2, and most preferably 400 ⁇ g/m2 to 4 mg/m2 . It is contemplated that the optimal dosage and mode of administration may be determined by routine experimentation well within the level of skill in the art.
  • Preferred dosages of the fusion protein per administration are within the range of 0.1 mg/m2 - 100 mg/m2, more preferably, 1 mg/m2 - 20 mg/m2, and most preferably 2 mg/m2 - 6 mg/m2. It is contemplated that the optimal dosage, however, also depends upon the disease being treated and upon the existence of side effects. However, optimal dosages may be determined using routine experimentation. Administration of the fusion protein may be by periodic bolus injections, or by continuous intravenous or intraperitoneal administration from an external reservoir (for example, from an intravenous bag) or internal (for example, from a bioerodable implant).
  • fusion proteins of the invention also may be administered to the intended recipient together with a plurality of different biologically active molecules. It is contemplated, however, that the optimal combination of fusion protein and other molecules, modes of administration, dosages may be determined by routine experimentation well within the level of skill in the art. The invention is illustrated further by the following non-limiting examples.
  • Example 1 Cloning of huFc-huInterferon-beta (huFc-IFN- ⁇ ) and huFc-IFN- ⁇ S0 ' mutants.
  • Human interferon ⁇ (IFN- ⁇ ) cDNA was ordered from American Type Culture Collection (ATCC Number 31903). The sequence for the mature form was amplified by Polymerase Chain Reactions (PCR).
  • the forward primer used in the amplification reactions was 5' C CCG GGT ATG AGC TAC AAC TTG CTT (SEQ ID NO:45), where the sequence CCCGGGT encodes the carboxy terminus of the CH3 without the lysine codon, as well as the restriction endonuclease site Smal CCCGGG (Lo et al, Protein Engineering (1998) 11:495), and the sequence in bold encodes the N-terminus of the mature IFN- ⁇ coding sequence.
  • the reverse primer for this reaction was 5' CTC GAG TCA GTT TCG GAG GTA ACC TGT (SEQ ID NO:46), where TCA is the anticodon of the translation stop codon, and CTCGAG is the restriction site Xho I.
  • the amplified 450 bp PCR product was cloned into the pCRII vector (Invitrogen), and its sequence verified.
  • the Smal-Xhol restriction fragment with the completely correct mature IFN- ⁇ sequence was used for cloning into the expression vector pdCs-huFc, such that the coding sequence of mature IFN- ⁇ was fused in frame to the 3'-end of the Fc coding sequence.
  • the expression plasmid pdCs-huFc-IFN- ⁇ was constructed by ligating the Smal-Xhol restriction fragment containing the mature IFN- ⁇ cDNA with the Smal-Xhol restriction fragment of the pdCs-huFc vector according to Lo et al., (Protein Engineering (1998) 11 :495).
  • the huFc DNA corresponds to a sequence that when expressed produces the Fc fragment of the human immunoglobulin ⁇ 4 with a modified ⁇ l hinge sequence.
  • the amino acid sequence is shown in SEQ ID NO:77.
  • the same cloning strategy was used, while substituting the appropriate version of pdCs-huFc vector.
  • the Smal-Xhol restriction fragment of IFN- ⁇ was inserted into pdCS-huFc vector digested with Smal and Xhol, which encoded either an immunoglobulin ⁇ 4 isotype with a ⁇ 4-derived hinge region, or an immunoglobulin ⁇ 1 isotype, or an immunoglobulin ⁇ 2 isotype, or an immunoglobulin ⁇ 2 isotype but with an altered immunoglobulin ⁇ 1 -derived hinge region.
  • the protein sequence encoded by the nucleic acid sequence around the Smal site is LSLSPG (SEQ ED NO:53). Had the mutation been silent, the sequence would have present been LSLSLG (See e.g. FIG. 7, residues 101-106 or SEQ ID NO:76).
  • the cysteine 17 to serine (C 17S) mutation was introduced into the EFN- ⁇ nucleotide sequence by an overlapping PCR method (Daugherty et al., (1991) Nucleic Acids Res. 19:2471) using complementary mutagenic primers.
  • the forward primer sequence was: 5 ' AGA AGC AGC AAT TTT CAG AGT CAG AAG CTC CTG TGG CA (SEQ ED NO:47), where the underlined nucleotide indicates the introduced point mutation (TGT to AGT). Accordingly, the reverse primer was: 5' TG CCA CAG GAG CTT CTG ACT CTG AAA ATT GCT GCT TCT (SEQ ID NO:48).
  • the PCR fragment generated by the overlapping PCR method was ligated to the pCRII vector, the sequence verified, and the Smal-Xhol fragment ligated to any of the pdCs-huFc expression vectors as described above.
  • the amino acid sequence is shown as SEQ ID NO:3.
  • the sequence of the mouse counterpart with the mutation is depicted in SEQ ID NO:12.
  • the cysteine at position 17 is mutated to a serine in the Fc- IFN- ⁇ so1 protein that has the Fc portion including immunoglobulin ⁇ 4 with a modified ⁇ 1 hinge sequence.
  • the amino acid sequence is shown as SEQ ID NO:4.
  • a synthetic oligonucleotide duplex of the sequence 5' G GGT GCA GGG GGC GGG GGC AGC GGG GGC GGA GGA TCC GGC GGG GGC TC 3' was produced.
  • This blunt-ended, double-stranded duplex was inserted at the unique Smal site of the expression vector pdCs-huFc-IFN- ⁇ by ligation.
  • the orientation of the blunt- ended duplex in the resultant vector, pdCs-huFc-(GS linker)-IFN- ⁇ was confirmed by sequencing.
  • the amino acid sequence GAGGGGSGGGGSGGGS (SEQ ID NO:50) was added between the proline (codon CCG) and the glycine (codon GGT) residues encoded by the C CCG GGT (SEQ ID NO:51) sequence containing the Smal site.
  • FIG. 6 The amino acid sequence of a huFc-(GS linker) IFN- ⁇ starting with the CH3 domain of the Fc ⁇ 4 isotype is shown in FIG. 6 (SEQ ID NO:5).
  • LSLSPG SEQ ID NO:52
  • C-terminal amino acid sequence of immunoglobulin ⁇ 4 lacks the alanine residue present in the immunoglobulin ⁇ l, ⁇ 2 or ⁇ 3 C-terminal sequence LSLSPGA (SEQ ID NO:53).
  • the alanine is the result of mutating the native lysine residue.
  • Cl 7 may be altered to another amino acid, for instance alanine.
  • the forward primer is 5'AGA AGC AGC AAT TTT CAG GCT CAG AAG CTC CTG TGG CA 3', (SEQ ID NO:54)
  • the reverse primer is 5' TG CCA CAG GAG CTT CTG AGC CTG AAA ATT GCT GCT TCT 3', (SEQ ID NO:55), where the underlined nucleotides indicate the introduced mutations.
  • Further mutations in Fc-IFN- ⁇ so1 were introduced in the IFN- ⁇ moiety by overlap PCR.
  • Fc ⁇ 4h-(linker)-IFN- ⁇ sol C17S L57A H131A H140A
  • Fc ⁇ 4h-(linker)-IFN- ⁇ sol C17S F50H H131A H140A
  • C17S F50H H131A H140A are produced by starting with the template Fc ⁇ 4h-linker-IFN- ⁇ sol(C17S) prepared using methods previously described herein.
  • a first nucleic acid fragment is created by PCR using the forward primer sequence 5'CTC CCT GTC CCC GGG TGC AGG GGG (SEQ ID NO:56), which incorporates the restriction endonuclease Xmal site, and the reverse primer sequence 5' CTT GGC CTT CAG GTA GGC CAG AAT CCT CCC ATA ATA TC (SEQ ID NO:57), where GGC represents the Hl 31 A mutation.
  • a second fragment of the fusion protein is created by PCR using the forward primer sequence 5'GAT ATT ATG GGA GGA TTC TGG CCT ACC TGA AGG CCA AG (SEQ ID NO:58), where GGC represents the H131 A mutation, and the reverse primer sequence 5' CTT ATC ATG TCT GGA TCC CTC GAG (SEQ ID NO:59), which incorporates the BamHI restriction site.
  • the products from these reactions are purified on an electrophoretic gel according to standard methods.
  • the gel purified fragments are then together subjected to PCR using the forward primer sequence 5'CTC CCT GTC CCC GGG TGC AGG GGG (SEQ ID NO:60), which incorporates the Xmal restriction site, and the reverse primer sequence 5' CTT ATC ATG TCT GGA TCC CTC GAG (SEQ ID NO:61), which incorporates the BamHI restriction site.
  • the H140A mutation is introduced by subjecting the Fc ⁇ h-linker-IFN- ⁇ soI (C 17S H131A) to PCR to create a first fragment using the forward primer sequence 5'CTC CCT GTC CCC GGG TGC AGG GGG (SEQ ID NO:62), which incorporates the restriction endonuclease Xmal site, and the reverse primer sequence 5' GGT CCA GGC ACA GGC ACT GTA CTC CTT GGC (SEQ ID NO:63), where GGC represents the H140A mutation.
  • a second fragment of the fusion protein is created by PCR using the forward primer sequence 5 ' GGC AAG GAG TAC AGT GCC TGT GCC TGG ACC (SEQ ID NO:64), where GCC represents the H140A mutation.
  • the reverse primer sequence is 5' CTT ATC ATG TCT GGA TCC CTC GAG (SEQ ID NO:65), which incorporates the BamHI restriction site.
  • the products from these reactions are purified on an electrophoretic gel according to standard methods.
  • the gel purified fragments are then together subjected to PCR using the forward primer sequence 5'CTC CCT GTC CCC GGG TGC AGG GGG (SEQ ED NO:66), which incorporates the Xmal restriction site, and the reverse primer sequence 5' CTT ATC ATG TCT GGA TCC CTC GAG (SEQ ID NO: 67), which incorporates the BamHI restriction site.
  • this process may be followed to instead insert the H140T mutation of the invention by modifying the appropriate primers to express the threonine codon ACC.
  • a first nucleic acid fragment is created by PCR using the forward primer 5 'CTC CCT GTC CCC GGG TGC AGG GGG (SEQ ID NO:68), which incorporates the restriction endonuclease Xmal site, and either the reverse primer sequence 5' GAG CAT CTC ATA GAT GGT GGC TGC GGC GTC CT C (SEQ ID NO:69), where GGC represents the codon for creating theL57A mutation or the reverse primer sequence 5' GTCCTCCTTCTGATGCTGCTGCAGCTG(SEQIDNO:70),whereATG represents the codon creating the F50H mutation.
  • the template is subjected to PCR using the forward primer sequence 5' GAG GAC GCC GCA GCC ACC ATC TAT GAG ATG CTC (SEQ ID NO:71), where GCC represents the L57A mutation.
  • the template is subjected to PCR using the forward primer sequence 5' CAG CTG CAG CAG CAT CAG AAG GAG GAC (SEQ ID NO:72), where CAT represents the F50H mutation.
  • the reverse primer for production of the second fragment of either mutation is 5' CTT ATC ATG TCT GGA TCC CTC GAG (SEQ ID NO:73), which incorporates the BamHI restriction site.
  • the products from these reactions are purified on an electrophoretic gel according to standard methods.
  • the gel purified fragments are then used as the PCR to produce a nucleic acid encoding Fc ⁇ 4h-linker-IFN- ⁇ sol (C17S L57A H131A H140A) or Fc ⁇ 4h-linker-IFN- ⁇ sol (C17S F50H H131 A H140A).
  • the forward and reverse primers for this reaction are 5'CTC CCT GTC CCC GGG TGC AGG GGG (SEQ ID NO:74) and 5' CTT ATC ATG TCT GGA TCC CTC GAG (SEQ ID NO:75), respectively, as used in previous steps.
  • Example 2 Transfection and Expression of Fc-IFN- ⁇ Fusion Proteins.
  • the plasmid pdCs-huFc-IFN- ⁇ , pdCs- huFc-IFN- ⁇ sol (C17S) or other huFc fusion protein variants containing huIFN- ⁇ were introduced into human embryonic kidney HEK 293 cells (ATCC# CRL-1573) by transient transfection using lipofectamine (Invitr ⁇ gen).
  • lipofectamine Invitr ⁇ gen
  • the appropriate plasmid DNA was introduced into the mouse myeloma NS/0 cells by electroporation.
  • NS/0 cells were grown in Dulbecco's modified Eagle's medium supplemented with 10% heat-inactivated fetal bovine serum, 2 mM glutamine and penicillin/streptomycin. About 5x10 6 cells were washed once with PBS and resuspended in 0.5 ml PBS. 10 ⁇ g of linearized plasmid DNA were then incubated with the cells in a Gene Pulser Cuvette (0.4 cm electrode gap, BioRad) on ice for 10 min. Electroporation was performed using a Gene Pulser (BioRad, Hercules, CA) with settings at 0.25 V and 500 ⁇ F.
  • MTX methotrexate
  • the cells were fed every 3 days for two to three more times, and MTX-resistant clones appeared in 2 to 3 weeks.
  • Supernatants from clones were assayed by anti-Fc ELISA to identify high producers.
  • High producing clones were isolated and propagated in growth medium containing 100 nM MTX.
  • the growth medium typically used was H-SFM or CD medium (Life Technologies).
  • clones stably expressing huFc-IFN- ⁇ so1 fusion proteins were obtained in human embryonic kidney HEK 293 cells by methotrexate selection, by a method similar to the one described above.
  • HEK 293 clones were maintained in DMEM supplemented with 10% FBS.
  • Example 3 Characterization of huFc-IFN- ⁇ fusion proteins from cell supernatants.
  • the huFc-IFN- ⁇ fusion proteins were subsequently captured from the medium for further analysis.
  • the huFc-IFN- ⁇ fusion proteins secreted into the medium was captured on Protein A Sepharose beads (Repligen, Cambridge, MA) and then eluted by boiling the sample in protein sample buffer, with or without a reducing agent such as ⁇ -mercaptoethanol.
  • the samples were analyzed by SDS-PAGE and the protein bands were visualized by Coomassie staining.
  • IFN- ⁇ can be produced as a fusion protein of the form IFN- ⁇ -Fc. See, for example, U.S. Patent No. 5,908,626. A portion of these aggregates was stable to reducing agents, as additional bands to the expected 50 kDa band for huFc-IFN- ⁇ persisted in a reducing SDS-PAGE system.
  • Example 4 ELISA Procedures. The concentration of human Fc-containing protein products in the supernatants of MTX-resistant clones and other test samples were determined by anti-huFc ELISA. Standard procedures as described in detail below were essentially followed.
  • the substrate solution was added to the plate at 100 ⁇ L/well.
  • the substrate solution was prepared by dissolving 30 mg of OPD (o-phenylenediamine dihydrochloride (OPD) 3 (1 tablet) into 15 niL of 0.025 M Citric acid/0.05 M Na2HPO4 buffer, pH 5, which contained 0.03% of freshly added hydrogen peroxide.
  • OPD o-phenylenediamine dihydrochloride
  • the color was allowed to develop for 30 min. at room temperature in the dark.
  • the developing time is subject to change, depending on lot to lot variability of the coated plates, the secondary antibody, etc.
  • the reaction was stopped by adding 4N sulfuric acid, 100 ⁇ L/well.
  • the plate was read by a plate reader, which was set at both 490 and 650 nm and programmed to subtract the background OD at 650 nm from the OD at 490 nm.
  • Example 5 Purification and analysis of huFc-IFN- ⁇ proteins.
  • a standard purification of Fc-containing fusion proteins was performed based on the affinity of the Fc protein moiety for Protein A. Briefly, cell supernatants (from cells transfected with wildtype or mutant proteins) containing the fusion protein were loaded onto a pre-equilibrated (50 mM Sodium Phosphate, 150 mM NaCl at neutral pH) Protein A Sepharose Fast Flow column and the column was washed extensively in buffer (50 mM Sodium Phosphate, 150 mM NaCl at neutral pH).
  • Bound protein was eluted at a low pH (pH 2.5) in same buffer as above and fractions were immediately neutralized, optionally by eluting directly into a solution of IM Tris base, pH 11.
  • the Protein A Sepharose - purified huFc-IFN- ⁇ and huFc-IFN- ⁇ S0 ' fusion proteins were analyzed by analytical size exclusion chromatography (SEC), and the % non- aggregated material was quantified by calculating the area under the curve of chromatogram peaks. The integrity and purity of the fusion proteins was verified by SDS-PAGE electrophoresis. Table 1: Analytical SEC analysis of Fc-IFN- ⁇ fusion proteins
  • Example 6 Measurement of Antiviral Activity. Viral replication in cell culture often results in cytotoxicity, an effect known as cytopathic effect (CPE). Interferons, can inhibit viral proliferation and protect cells from CPE. The antiviral activity of IFN- ⁇ can be quantitated by cytopathic effect reduction (CPER), as described in "Lymphokines and Interferons: A Practical Approach", edited by MJ. Clemens, A.G. Morris, and AJ.H. Gearin, I.R.L. Press, Oxford, 1987.
  • CPER cytopathic effect reduction
  • the antiviral activities of purified huFc-IFN- ⁇ and huFc-IFN- ⁇ so1 were compared relative to a commercial huIFN- ⁇ standard (R&D Systems) or Betaferon (Serono) using the human epithelial lung carcinoma line A549 (ATCC # CCL-185) and the encephalomyocarditis virus (EMCV; ATCC # VR 129B) according to the CPER protocol described in the above reference.
  • the effective dose (ED50) was set as the amount of protein that led to 50% CPER (i.e. 50% of the cells being protected from lysis), determined relative to uninfected control cells.
  • the ED50 values were the average of at least three separate experiments.
  • Example 7 Cellular Growth Inhibition Assay.
  • the activity of the purified Fc-IFN- ⁇ fusion proteins was further determined in a cellular growth inhibition assay.
  • the proliferation of Daudi cells (ATCC # CCL-123), a B lymphoblast line derived from a patient with Burkitt's lymphoma, is normally inhibited by IFN- ⁇ . Accordingly, the antiproliferative effects of fusion proteins huFc- IFN- ⁇ and huFc-IFN- ⁇ sol (C17S) on Daudi cells were compared relative to a commercial human standard (Calbiochem).
  • a dilution series covering about a thousand fold concentration range was prepared in RPMI medium supplemented with 10 % fetal bovine serum, and 100 ⁇ l samples were aliquoted in wells of a 96 well plate. Daudi cells in growth phase were washed and resuspended at 2 x 10 5 cells/ml in the RPMI medium supplemented with 10 % fetal bovine serum, and 100 ⁇ l of the cells were aliquoted to each well containing the IFN- ⁇ dilutions. Further control wells contained either untreated cells or medium alone.
  • Example 8 Pharmacokinetics of huFc-IFN- ⁇ proteins.
  • Cells are removed by centrifugation in an Eppendorf high ⁇ speed microcentrifuge for 4 min at 12,500 g.
  • concentration of either Fc-huIFN- ⁇ or huFc-IFN- ⁇ so1 in the plasma is measured by anti-huFc ELISA and Western blot analysis using anti-huFc antibody.
  • an IFN- ⁇ ELISA maybe used.
  • the integrity of the circulating fusion protein is ascertained by an immunoblot of the serum probed with an anti-huFc antibody or with an anti-IFN- ⁇ antibody.
  • huFc-IFN- ⁇ so1 is greater than that of huFc-IFN- ⁇ , and at least 5-fold that of the free IFN- ⁇ . Furthermore, it is contemplated that the specific effects of Fc-IFN- ⁇ so1 are more pronounced in treatment of conditions and diseases such as multiple sclerosis, where administration of IFN- ⁇ is known to alleviate the condition.

Abstract

Disclosed are Fc-interferon-beta (Fc-IFN-β) fusion proteins and nucleic acid molecules encoding them. The Fc- IFN-β fusion proteins include variants of the interferon-beta (IFN-β) protein that are altered to achieve enhanced biological activity, prolonged circulating half-life and greater solubility. Also disclosed are methods of producing the fusion proteins and methods of using the fusion proteins and/or nucleic acid molecules for treating diseases and conditions alleviated by the administration of interferon-beta.

Description

Fc-INTERFERON-BETA FUSION PROTEINS
FIELD OF THE INVENTION The invention relates to Fc-fusion proteins. More specifically, the invention relates to high-level expression and secretion of Fc-interferon-beta fusion proteins and variant forms thereof, and methods of making and using such proteins.
BACKGROUND OF THE INVENTION Interferons are single chain polypeptides secreted by most animal cells in response to a variety of stimuli, including viruses, mitogens and cytokines. Interferons participate in the regulation of cell functions and mediate antiproliferative, antiviral and immunomodulatory effects. Thus, they are of great interest therapeutically. Native interferons are divided into three major types, based on the cell types from which they are primarily derived, namely, interferon-α (from leukocytes), interferon-β (from fibroblasts), interferon-γ (from immune cells). Interferon-β (IFN-β) exhibits various biological and immunological activities and as a result has potential applications in immunotherapy, antitumor, anticancer and antiviral therapies. Numerous investigations and clinical trials have been and are being conducted based on anticancer and antiviral properties of both wild-type and recombinant IFN-β. Clinical trials using recombinant IFN-β in the treatment of multiple sclerosis also have been conducted. Most cytokines, including native IFN-β, have relatively short circulating half- lives. Consequently, in order for IFN-β to be effective as a therapeutic agent, it must be administered in large and frequent doses to a patient; however, this often leads to toxic side effects. Therefore, it is highly desirable to produce forms of IFN-β that have prolonged circulating half-lives compared to the native cytokine. Furthermore, for production purposes it is useful to produce forms of IFN-β that are easy to express and purify in large amounts. Human IFN-β (huIFN-β) is a glycoprotein of 166 amino acids and has a four helix-bundle structure. Recombinant huIFN-β may be commonly produced for use as a therapeutic in either a prokaryotic or a mammalian expression system. However, when proteins that are normally secreted, such as huEFN-β, in a mammalian environment are produced in a prokaryote, the effect of prokaryotic expression on protein folding and on potential post-translational modifications needs to be addressed. For example, in mammalian cells, most proteins destined for the extracellular milieu are folded in the oxidizing environment of the endoplasmic reticulum (ER), which promotes the correct formation of disulfide bonds, hi contrast, the reducing environment of the prokaryotic cytosol interferes with the formation of cysteine bonds. In addition, proteins expressed in prokaryotic systems lack some post-translational modifications, such as N-linked glycosylation, which likely aid in the correct folding of the protein, increase the stability of the folded protein, and decrease the immunogenicity of the administered protein. For example, when intact wild-type IFN-β is expressed in a prokaryotic expression system, it does not fold properly and forms aggregates. This can be overcome by mutating the free cysteine at position 17 of the mature IFN-β protein to, for example, a serine. This cysteine at position 17 is not involved in a disulfide bond. See, for example, U.S. Patent No. 4,737,462. hi contrast, when intact wild-type IFN-β is produced in a eukaryotic expression system, where the environment is appropriate for correct folding of the IFN-β protein, improper folding and aggregation are not observed. Because IFN-β protein appears to fold properly and not to aggregate when expressed in a eukaryotic expression system, this suggests that glycosylation plays an important role in proper folding of the IFN-β protein. Recombinant IFN-β produced in a eukaryotic expression system undergoes glycosylation, although it may not have the precise glycosylation pattern of the native IFN-β. See, for example, U.S. Patent No. 5,795,779. Whereas glycosylation of IFN-β does not seem to be essential for its biological activity, the specific activity of glycosylated IFN-β in bioassays is greater than that of the unglycosylated form. Indeed, IFN-β produced in a eukaryotic expression system, such as a mammalian expression system, is substantially non-aggregated, but does form aggregates when the glycan moiety is removed. Therefore, the glycosylated form of IFN- β is desirable for therapeutic use as its biophysical properties are closer to those of the native protein than the unglycosylated form. In addition, it has been found that linking a protein of interest "X" to an immunoglobulin Fc domain "Fc" to create an Fc-X fusion protein ("immunofusin") generally has the effect of increasing protein production significantly. This is believed to occur, in part, because the Fc moiety of the fusion protein, commonly referred to as the expression cassette, is designed for efficient secretion of the fusion protein, and in part because proteins are being produced and secreted from mammalian cells that are normally active for secretion. A further advantage of creating Fc-X fusion proteins is that the resultant inimunofusins exhibit an increased circulating half-life as compared to the free proteins of interest, which can be a significant therapeutic advantage. There is, therefore, a need in the art for biologically active immuiiofusins including an Fc moiety fused to an IFN-β moiety optimized to have biophysical properties that are close to those of native IFN-β.
SUMMARY OF THE INVENTION The invention provides methods and compositions for expressing soluble, biologically active Fc-IFN-β fusion proteins and variants thereof (Fc-IFN-βso1). The Fc- IFN-βso! fusion proteins of the invention demonstrate improved biological properties over unaltered Fc-IFN-β proteins such as increased solubility, prolonged circulating half-life, enhanced biological activity, and reduced immunogenicity. To improve the circulating half-life of IFN-β, the invention provides a fusion protein including an Fc-IFN-β fusion protein including an immunoglobulin Fc region and an IFN-β protein linked to the carboxy-terminus of the immunoglobulin Fc region. To improve folding and to reduce aggregation, the IFN-β protein includes an amino acid alteration at at least one of positions 17, 50, 57, 130, 131, 136, and 140, corresponding to native mature interferon-β. The alteration to the amino acid can be a deletion, substitution or modification, hi one embodiment, the amino acid alteration substitutes either serine, alanine, valine or methionine in place of cysteine at position 17. hi another embodiment, the amino acid alteration substitutes histidine in place of phenylalanine at position 50. In yet another embodiment, the amino acid alteration substitutes alanine in place of leucine at position 57, while in a further embodiment, the amino acid alteration substitutes alanine in place of leucine at position 130. A further embodiment allows an amino acid alteration substituting alanine in place of histidine at position 131, while an additional embodiment contemplates substituting alanine in place of lysine at position 136. In yet another embodiment, the amino acid alteration substitutes alanine or threonine in place of histidine at position 140. The immunoglobulin Fc region can include an immunoglobulin hinge region and an immunoglobulin heavy chain constant region. In one embodiment, the Fc region is derived from IgG4, while in another it is derived from IgGl, and in yet another it is derived from IgG2. Li another embodiment, the Fc region is derived from IgG4 but includes a hinge region from IgGl. In yet another embodiment, the Fc region is derived from IgG2 but includes a hinge region derived from IgGl. When the Fc region includes a CH3 domain, the C-terminal lysine of the immunoglobulin Fc region can be replaced by an alanine residue. In a further embodiment, a cysteine residue of the hinge region is mutated. The invention provides different methods for joining the Fc moiety and the IFN-β moiety to create fusion proteins according to the invention. For example, in one embodiment the immunoglobulin Fc region and the interferon-β protein are fused together by a peptide bond. In another embodiment, the immunoglobulin Fc region and the interferon-β protein are connected by a peptide linker sequence to facilitate protein folding. The linker sequence preferably is composed of glycine and serine residues. For example, in one embodiment, the peptide linker sequence is Gly4SerGly4SerGly3SerGly (SEQ ID NO: 1). hi one embodiment, the Fc-interferon-β fusion protein includes amino acid alterations at positions 17, 50, 131, and 140 to improve folding and reduce aggregation, hi one specific embodiment, the amino acid alterations are serine substituted in place of cysteine at position 17, histidine substituted in place of phenylalanine at position 50, alanine substituted in place of histidine at position 131, and threonine or alanine substituted in place of histidine at position 140. In certain embodiments, the Fc region includes IgGl, IgG2, or IgG4. The fusion protein can also include a polypeptide linker sequence connecting the interferon-β protein and the immunoglobulin Fc region, hi one embodiment, a cysteine residue of the hinge region is mutated. hi another embodiment, the Fc-interferon-β fusion protein includes amino acid alterations at positions 17, 57, 131, and 140, improving folding and reducing aggregation of the expressed fusion protein, hi one specific embodiment, the amino acid alterations are serine substituted in place of cysteine at position 17, alanine substituted in place of leucine at position 57, alanine substituted in place of histidine at position 131, and threonine or alanine substituted in place of histidine at position 140. In certain embodiments, the Fc region includes IgGl, IgG2, or IgG4. In another embodiment, the fusion protein can also include a polypeptide linker sequence connecting the interferon-β protein and the immunoglobulin Fc region. In a further embodiment, a cysteine residue of the hinge region is mutated. The invention also provides methods for encoding and expressing fusion proteins of the invention. For example, one aspect of the invention relates to nucleic acid molecules encoding any of the aforementioned Fc-interferon-β fusion proteins, while in another aspect, the invention relates to cells containing a nucleic acid encoding any of the aforementioned Fc-interferon-β fusion proteins. In a further aspect, the nucleic acid molecules of the invention can be incorporated in operative association into a replicable expression vector which can then be introduced, for example, by transfection, into a mammalian host cell competent to produce the immunoglobulin Fc-IFN-βso1 fusion protein. The vector includes a nucleic acid molecule encoding any one of the aforementioned Fc-interferon-β fusion proteins. The invention also encompasses a replicable expression vector for transfecting a mammalian cell. The vector includes a nucleic acid molecule encoding any one of the aforementioned Fc-interferon-β fusion proteins. In another aspect, the invention relates to methods of stabilizing Fc-interferon-β fusion proteins, hi one embodiment, the method includes the step of making any of the aforementioned Fc-interferon-β fusion proteins. In a further embodiment, the stabilizing includes increasing the circulating half-life of the Fc-interferon-β fusion protein relative to an unaltered Fc-interferon-β fusion protein, hi yet another embodiment, the stabilizing includes decreasing the aggregation of the Fc-interferon-β fusion protein relative to an unaltered Fc-interferon-β fusion protein, while in a further embodiment, the stabilizing includes increasing the biological activity of the Fc-interferon-β fusion protein relative to an unaltered Fc-interferon-β fusion protein. A further aspect of the invention relates to methods for treating a patient for a condition alleviated by the administration of interferon-β. In one embodiment, the treatment includes administering an effective amount of any of the aforementioned interferon-β fusion proteins to a mammal having the condition, hi another embodiment, the method includes administering a nucleic acid encoding any of the aforementioned interferon-β fusion proteins to a mammal having the condition, while in yet another embodiment, the method includes administering a cell encoding any of the aforementioned interferon-β fusion proteins to a mammal having the condition.
hi summary the inventions relates to the following subject matters: • An Fc-interferon-β fusion protein comprising: (i) an immunoglobulin Fc region; and (ii) an interferon-β protein linked by peptide bond or a peptide linker sequence to the carboxy-terminus of the immunoglobulin Fc region, wherein the interferon-β protein comprises an amino acid alteration at at least one of positions 17, 50, 57, 130, 131, 136 and 140 corresponding to the native mature interferon-β. • A corresponding fusion protein, wherein the amino acid alteration is a substitution. • A corresponding fusion protein, having amino acid substitutions within the interferon-β protein which are selcted from the group consisting of: C17S, C17A, C17V, C17M; F50H, L57A, L130A, H131A, K136A, H140A, H140T. • A corresponding fusion protein, having amino acid substitutions within the interferon-β protein at positions 17 and 50 and 131 and 140, or 17 and 57 and 131 and 140. • A corresponding fusion protein, having the following amino acid substitutions within the interferon-β protein: C17S and F50H and H131A and H140T or A, or C17S and L57A and H131A and H140T or A. • A corresponding fusion protein, wherein the immunoglobulin Fc region comprises an immunoglobulin hinge region and an immunoglobulin heavy chain constant region. • A corresponding fusion protein, wherein the immunoglobulin Fc region is derived
Figure imgf000007_0001
• A corresponding fusion protein, wherein the immunoglobulin Fc region is derived from IgG4 and further comprises an immunoglobulin hinge region derived from IgGl. • A corresponding fusion protein, wherein a cysteine residue of the hinge region has been mutated. • A corresponding fusion protein, wherein the immunoglobulin Fc region is derived from IgGl, and an alanine residue is substituted in place of the C-terminal lysine of the immunoglobulin Fc region. • A corresponding fusion protein, wherein the immunoglobulin Fc region is derived from IgG2 and further comprises an immunoglobulin hinge region derived from IgGl. • A corresponding fusion protein, wherein a cysteine residue of the hinge region has been mutated. • A corresponding fusion protein, wherein further an alanine residue is substituted in place of the C-terminal lysine of the immunoglobulin Fc region. • A corresponding fusion protein, wherein the peptide linker sequence is Gly4SerGly4SerGly3SerGly. • A nucleic acid molecule encoding any one of the Fc-interferon-β fusion proteins above. • A replicable expression vector for transfecting a mammalian cell, the vector comprising a nucleic acid molecule encoding any one of the Fc-interferon-β fusion proteins as cited above. • A cell containing a corresponding nucleic acid molecule. • A method of stabilizing an Fc-interferon-β fusion protein comprising the step of making an Fc-interferon-β fusion protein, wherein the Fc-interferon-β fusion comprises any one Fc-interferon-β fusion proteinas specified above. • A corresponding method, wherein stabilizing comprises (i) increasing the circulating half-life of the Fc-interferon-β fusion protein relative to an unaltered Fc-interferon-β fusion protein, or (ii) decreasing the aggregation of the Fc-interferon-β fusion protein relative to an unaltered Fc-interferon-β fusion protein, or (iii) increasing the biological activity of the Fc-interferon-β fusion protein relative to an unaltered Fc-interferon-β fusion protein. • A use of a corresponding Fc-interferon-β fusion protein for the manufacture of a medicament for the treatment of a condition of a patient, which is alleviated by the administration of interferon-β.
The foregoing and other objects, features and advantages of the invention will be apparent from the description, drawings, and claims that follow.
BRIEF DESCRIPTION OF THE DRAWINGS
FIGS. 1A-1C are schematic illustrations of non-limiting examples of Fc-IFN-βso1 fusion proteins constructed in accordance with the invention. FIG. 2 is a photograph of an SDS-PAGE gel showing the migration patterns of HuFc-γ4-IFN-β and HuFc-γ4h-IFN-β fusion proteins without the C17S mutation and HuFc-γ4h-IFN-β(C 17S) fusion proteins in both reducing and non-reducing chemical environments. FIG. 3 is the amino acid sequence for mature IFN-β (SEQ ID NO:2). FIG. 4 is the amino acid sequence for mature human IFN-β(C17S) (SEQ ID NO:3). FIG. 5 is the amino acid sequence for human Fc-IFN-βsol(C17S) of the γ4 isotype with a modified γl hinge (Fcγ4h-IFN-βso1) (SEQ ID NO:4). FIG. 6 is the amino acid sequence for human Fc-(linker)-IFN-β, starting with the CH3 domain of the Fcγ4 isotype (SEQ E) NO:5). FIG. 7 is the amino acid sequence for human Fc-(linker)-IFN-βsol(C17S), starting with the CH3 domain of the Fcγ4 isotype (SEQ ID NO:6). FIG. 8 is the amino acid sequence for human Fc-(linker)-IFN-βsol(C17S L57A H131A H140T) starting with the CH3 domain of the Fcγ4 isotype (SEQ ID NO:7). FIG. 9 is the amino acid sequence for human Fc-(linker)-IFN-βsol(C17S L57A H131A H140A) starting with the CH3 domain of the Fcγ4 isotype (SEQ ID NO:8). FIG. 10 is the amino acid sequence for human Fc-(linker)-IFN-βsol(C17S F50A H131 A5 H140A), starting with the CH3 domain of the Fcγ4 isotype (SEQ ID NO:9). FIG. 11 is the amino acid sequence for human Fc-(linker)-IFN-βsol(Cl 7S F50A H131 A H140T), starting with the CH3 domain of the Fcγ4 isotype (SEQ ID NO: 10). FIG. 12 is the amino acid sequence for mature mouse IFN-β (SEQ ID NO:11). FIG. 13 is the amino acid sequence for mature mouse IFN-β(C17S) (SEQ ID NO: 12). FIG. 14 is the nucleic acid sequence encoding the fusion protein embodiment huFcγ4h-IFN-βsol(C17S) (γ4 isotype with modified γl hinge wherein the first cysteine of the γl hinge is replaced by a serine), starting from the hinge region (SEQ ID NO: 13). FIGS. 15-1 through 15-3 show the linearized nucleic acid sequence of the pdCs vector containing huFcγ4h-(linker)-IFN-βsol(C17S) (γ4 isotype with modified γl hinge wherein the first cysteine of the γl hinge is replaced by a serine), wherein the Fc region and the IFN-β moiety are attached via a linker polypeptide (SEQ ID NO: 14). FIG. 16 is the nucleic acid sequence encoding the fusion protein embodiment HuFc-γ4h-(linlcer)-IFN-βsol(C17S) (γ4 isotype with modified γl hinge wherein the first cysteine of the γl hinge is replaced by a serine), starting from the hinge region, wherein the Fc region and the IFN-β moiety are attached via a linker polypeptide (SEQ ID NO: 15). FIGS. 17-1 through 17-3 show the linearized nucleic acid sequence of the pdCs vector containing huFcγ4h-(linker)-IFN-βsol(C17S L57A H131A H140A) (γ4 isotype with modified γl hinge wherein the first cysteine of the γl hinge is replaced by a serine), wherein the Fc region and the IFN-β moiety are attached via a linker polypeptide (SEQ ID NO: 16). FIG. 18 is the nucleic acid sequence of huFcγ4h-(linker)-IFN-βsol(C17S L57A H131 H140A) (γ4 isotype with modified γl hinge wherein the first cysteine of the γl hinge is replaced by a serine), starting from the hinge, wherein the Fc region and the IFN-β moiety are attached via a linker polypeptide (SEQ ID NO: 17). FIG. 19-1 through 19-3 shows the linearized nucleic acid sequence of the pdCs vector containing huFcγ4h-(linker)-IFN-βsol(C17S E50H H131 A H140A) (γ4 isotype with modified γl hinge wherein the first cysteine of the γl hinge is replaced by a serine), wherein the Fc region and the IFN-β moiety are attached via a linker polypeptide (SEQ ID NO: 18). FIG. 20 is the nucleic acid sequence of huFcγ4h-(linker)-IFN-βsol(C 17S F50H Hl 31 A H 140A) (γ4 isotype with modified γl hinge wherein the first cysteine of the γl hinge is replaced by a serine) starting from the hinge, wherein the Fc region and the IFN-β moiety are attached via a linker polypeptide (SEQ ID NO: 19).
DETAILED DESCRIPTION OF THE INVENTION IFN-β mediates antiproliferative, antiviral and immunomodulatory effects and, in addition to its usefulness in treating multiple sclerosis, it is anticipated that many other conditions may be alleviated by IFN-β administration. For example, its usefulness as a treatment for a variety of malignancies, such as acute myeloid leukemia, multiple myeloma, Hodgkin's disease, basal cell carcinoma, cervical dysplasia and osteosarcoma is under evaluation. IFN-β is also being tested as a therapeutic agent against a variety of viral infections, including viral hepatitis, herpes zoster and genitalis, papilloma viruses, viral encephalitis, and cytomegalovirus pneumonia. However, when administered to a patient, recombinant mature IFN-β has a short circulating half-life, making it suboptimal for use in therapy. Therefore there is a need in the art to produce variants of TFN-β with improved pharmacokinetic properties, including improved serum half-life. One method known in the art for prolonging the half-life of small proteins involves linking them to an immunoglobulin Fc region. Fusions in which an Fc region is placed at the N-terminus of a ligand (termed 'immunofusins' or 1Fc-X' fusions, where X is a ligand such as IFN-β) have a number of useful properties (Lq et al, U. S. Patent Nos. 5,726,044 and 5,541,087; Lo et al (1998) Protein Engineering 11: 495). For instance, if leptin is administered to a mouse as an Fc-leptin fusion molecule (See, for example, PCT patent application publication WO 00/40615), the circulating half-life of leptin increases from about 18 minutes to more than 8 hours. Similarly, the half-life of IL-2 in a mouse is increased from a few minutes to a few hours when it is administered as an Fc-IL2 fusion protein. Another useful property of Fc-X fusion proteins is that the Fc portion generally has the effect of increasing protein production significantly. This is believed to occur, in part, because the Fc moiety of the fusion protein, commonly referred to as the expression cassette, is designed for efficient secretion of the fusion protein and, in part, because the fusion proteins can be produced in and secreted from host mammalian cells that naturally express immunoglobulin such that the fusion protein is readily secreted from the host cell. While it may be possible to produce these fusion proteins in a prokaryotic expression system, a eukaryotic expression system is preferred and a mammalian expression system is most preferred. Surprisingly, it was found that when an unaltered Fc-IFN-β immunofusin was produced in a eukaryotic expression system, it was poorly expressed, misfolded and substantially aggregated, hi contrast, recombinant IFN-β proteins produced in a eukaryotic expression system are soluble and 98 % monomelic (Runkel et al. (1998), Pharmaceutical Research 15:641). Thus it appeared that the placement of the Fc moiety at the N-terminus of the IFN-β moiety affected the ability of the fusion protein to fold correctly as no aggregation is observed when IFN-β is produced as a fusion protein where the IFN-β moiety precedes the Fc domain (See U.S. Patent No. 5,908,626). Therefore, there is a need in the art to create Fc-IFN-β fusion proteins that fold correctly and are substantially non-aggregated. Consequently, the invention provides (i) nucleic acid sequences which facilitate efficient production of immunoglobulin Fc-IFN-βso1 fusion proteins; (ii) nucleic acid constructs for rapid and efficient production and secretion of immunoglobulin Fc-IFN-βso1 fusion proteins in a variety of mammalian host cells; and (iii) methods for the production, secretion, and purification of recombinant variants of immunoglobulin Fc-IFN-βso! fusion proteins. hi particular, the present invention provides nucleic acid molecules, for example, DNA or RNA molecules, which encode serially in the 5 ' to 3 ' direction, a polypeptide including an immunoglobulin Fc region and an IFN-βso1 protein . The nucleic acid molecules of the invention can be incorporated in operative association into a replicable expression vector which may then be introduced, for example, by transfection, into a mammalian host cell competent to produce the immunoglobulin Fc-IFN-βso1 fusion protein. The invention also provides methods of stabilizing immunoglobulin Fc-IFN-β fusion proteins. Although many proteins have been successfully produced and purified as Fc fusions, including many four-helix bundle proteins such as IL-2 (huFc-IL2), it has been found that Fc-IFN-β fusion proteins, where IFN-β belongs to the class of four-helix bundle proteins, form aggregates at least partly due to aberrant disulfide bonds present in the protein ("covalent aggregation"). In addition, it has been found that Fc-IFN-β proteins form aggregates through non-covalent interactions as well ("non-covalent aggregation"). The present invention alleviates aggregation by providing methods of stabilizing Fc-IFN-β fusion proteins including the step of making an Fc-IFN-βso1 fusion protein, where the fusion protein includes an IFN-β protein having at one or more amino acid alterations, linked to the carboxy-terminus of an immunoglobulin Fc region. In embodiments of the invention, stabilizing includes increasing the solubility of the Fc- IFN-βso1 fusion protein relative to an unaltered Fc-IFN-β fusion protein, increasing the circulating half-life of the Fc-IFN-βso1 fusion protein relative to an unaltered Fc-IFN-β fusion protein, and/or enhancing the biological activity of the Fc-IFN-βso1 fusion protein relative to an unaltered Fc-IFN-β fusion protein. Increased stabilization is achieved in part by the elimination of aberrant disulfide bonding in the fusion protein and in part by reducing the amount of non-covalent aggregation of the fusion protein. The invention also provides methods for treating conditions alleviated by IFN-β, bioactive fragments or active variants thereof by administering to a mammal an effective amount of IFN-β produced by a method of the invention and/or an Fc-IFN-βso1 fusion protein of the invention. The invention also provides methods for treating conditions alleviated by IFN-β or active variants thereof by administering a nucleic acid of the invention, for example, a "naked DNA," or a vector containing a DNA or RNA of the invention, to a mammal having the condition.
IFN-β Moiety The invention provides fusion proteins and nucleic acid molecules encoding those proteins including an altered IFN-β protein linked to the C-terminus of an immunoglobulin Fc region. The IFN-β moiety can include one ore more mutations to the amino acid structure of the IFN-β moiety and Fc-IFN-β construct to improve the protein folding properties of the fusion protein, to reduce aggregation, and to improve protein expression. For example, the IFN-β moiety of the soluble fusion protein Fc-IFN-βso1 can contain an alteration at position 17, corresponding to a cysteine in the native mature IFN- β linked to the carboxy-terminus of an immunoglobulin Fc region. The amino acid sequence for native mature human IFN-β is shown in FIG. 3. The amino acid alteration at position 17 of the IFN-β protein can be generated by an amino acid substitution, amino acid deletion or amino acid modification through methods known in the art. Preferred alterations to the IFN-β moiety include substituting either a serine (C17S), valine (C17V), alanine (C17A) or methionine (C17M) in place of the cysteine at position 17. An exemplary amino acid sequence of a soluble human Fc-IFN-β fusion protein containing the C17S mutation (huFc-IFN-βsol(C17S)) is shown in FIG. 5 (SEQ ID NO:4), while the amino acid sequence for an IFN-β moiety including the Cl 7S mutation is shown in FIG. 4 (SEQ ID NO:3). The invention also includes huFc-IFN-βsol(C17V), huFc-IFN- βsol(C17A) and huFc-IFN-βsoI(C17M) fusion protein constructs. In addition to an alteration at position 17 of the mature IFN-β moiety, the invention provides Fc-IFN-β fusion proteins with other altered residues. For example, the IFN-β moiety can be altered at one or more of positions 17, 50, 57, 130, 131, 136, and 140 corresponding to, respectively, a cysteine, a phenylalanine, a lysine, a leucine, a histidine, a lysine, and a histidine in the native mature IFN-β protein. The IFN-β moiety is linked to the carboxy-terminus of an immunoglobulin Fc region. Alterations to the amino acid structure at one or more of positions 17, 50, 57, 130, 131, 136, and 140 can include an amino acid substitution, amino acid deletion or amino acid modification and can be generated through methods known in the art. Alterations introduced at these residues are believed to alleviate the causes of non-covalent aggregation. Ih one embodiment, the phenylalanine at position 50 is replaced by histidine (F50H). Li another embodiment, the leucine at position 57 is replaced by alanine (L57A). In a further embodiment, the histidine at position 131 is replaced by alanine (H131A), while in yet another embodiment, the histidine at 140 is replaced by either alanine (H 140A) or threonine (H 140T). In another embodiment, the leucine at position 130 is replaced by alanine (L130A), while in yet another embodiment, the lysine at residue 136 is replaced by alanine (K136A). While certain amino acid substitutions have been enumerated herein, the invention is not limited to these enumerated alterations. Any suitable amino acid capable of conferring the appropriate properties on the fusion protein may be substituted in place of the original amino acid residue at position 17, 50, 57, 130, 131, 136, and/or 140 of the IFN-β moiety. The invention contemplates an IFN-β moiety of an Fc-IFN-βso1 fusion protein having any combination of one, two, three, four, five, six, or seven of the alterations to positions 17, 50, 57, 130, 131, 136 and/or 140 as disclosed herein. For example, the Fc- IFN-βso1 in one embodiment contains amino acid alterations at one or more of F50, Hl 31 and H140 of the mature form of IFN-β, optionally combined with a C17 alteration. In another embodiment, the IFN-β moiety of the Fc-IFN-βso1 fusion protein contains amino acid alterations at one or more of L57, H131 and H140 of the mature form of IFN- β, optionally combined with a Cl 7 alteration. In another embodiment, IFN-β moiety of the Fc-IFN-βso1 fusion protein includes the alterations C17S, F50H, H131A, and/or H140A. FIGS. 8-11 show exemplary amino acid sequences of embodiments of Fc-IFN-βso1 fusion proteins incorporating various combinations of these mutations. In yet another embodiment, the IFN-β moiety of the Fc-IFN-βso1 fusion protein includes the alterations C17S, F50H, H13 IA, and/or H140T. hi yet another embodiment, the IFN-β moiety of the Fc-IFN-βso1 fusion protein includes the alterations C17S, L57A, H131A, and/or H140A, while in a further embodiment, the fusion protein includes the alterations C17S, L57A, H131 A, and/or H140T. The Fc region is preferably a human Fc region. Another embodiment of the invention includes nucleic acid sequences encoding Fc-IFN-βso1 variants with at least one codon substitution in the mature human IFN-β protein sequence, hi one embodiment, a codon substitution replaces the cysteine corresponding to position 17 in the mature human IFN-β sequence with a serine (C 17S). Expression of this nucleotide sequence, contained on an appropriate plasmid, in a mammalian cell culture system resulted in the efficient production of the fusion protein huFc-huIFN-βsoI(C17S). In alternative embodiments, a codon substitution replaces the cysteine at position 17 with either an alanine, a valine, or a methionine. Similarly, expression from any of these nucleotide sequences, contained on an appropriate plasmid, in a mammalian cell culture system will result in the efficient production of fusion protein huFc-huIFN-βsol(C17A), huFc-huIFN-βsol(C17V), or huFc-huIFN-βsol(C17M). In one embodiment, a nucleic acid sequence encoding a representative Fc-IFN-βso1 fusion protein huFcγ4h-IFN-βsol(C17S), starting from the hinge, is disclosed in FIG. 14 (SEQ ID NO: 13). The invention also includes nucleic acid sequences encoding Fc- IFN-βso1 variants with codon substitutions replacing amino acids at one or more of positions 17, 50, 57, 130, 131, 136 and/or 140. Nucleic acids incorporating the altered codons of the invention can be created using methods known in the art. The immunoglobulin Fc region and the IFN-β moiety of an Fc-IFN-βso1 fusion protein can be linked to one another in a variety of ways. While the C-terminus of the Fc moiety may be directly linked to the N-terminus of the IFN-β moiety via a peptide bond, the invention additionally includes connecting the Fc moiety and the IFN-β moiety via a linker peptide. The linker peptide is located between the C-terminus of the Fc moiety and the N-terminus of the mature IFN-β moiety. The invention also includes a nucleic acid sequence encoding the linker peptide. The linker peptide is preferably composed of serine and glycine residues. In one embodiment, the linker has the amino acid sequence Gly4SerGly4SerGly3SerGly (SEQ ID NO:1), while in yet another embodiment a nucleic acid encoding an Fc-IFN-βso1 includes a nucleic acid sequence encoding the linker peptide Gly4SerGly4SerGly3SerGly (SEQ ID NO: 1). Some exemplary Fc-linker-IFN-βso1 amino acid sequences of the invention are shown in FIGS. 6-11, while some exemplary Fc- linker-IFN-βso1 nucleic acid sequences of the invention are shown in FIGS. 14-16. For example, in one embodiment, the Fc-linker-IFN-βso1 protein is huFcγ4-linker-IFN- βsol(C17S), wherein the Fc region is an IgG4 Fc region, and the linker is Gly4SerGly4SerGly3SerGly (SEQ ID NO:1). Expression of fusion proteins of the invention from Fc-IFN-βso1 and Fc-linker-IFN-βso1 nucleotide sequences, such as those previously discussed, when contained on an appropriate plasmid, in a mammalian cell culture system will result in the efficient production of Fc-IFN-βso1 and Fc-linker-IFN-βso1 fusion proteins. As previously mentioned, Fc-IFN-βso1 fusion proteins of the invention demonstrate unproved biological properties over unaltered Fc-IFN-β fusion proteins. For example, it was found that human Fcγ4h-IFN-βsol(C17S) displayed folding properties that were different from, and improved over, the parent fusion protein Fcγ4-IFN-βso1 and Fcγ4h- IFNβso1. hi particular, as demonstrated in FIG. 2, it was found that predominantly a single species of the human Fcγ4h-IFN-βsoI(C17S) fusion protein 3, 4 was seen when expressed in mammalian tissue culture cells, as ascertained by non-reducing SDS-PAGE gel analysis. This species corresponded to the correctly folded Fcγ4-IFN-βso1 fusion protein 3, 4. In contrast, for the parent molecule Fcγ4-IFN-βso1 1 and for Fcγ4h-IFN-βso1 2, many high molecular weight species were observed, as evidenced by an unresolved trail of high molecular weight proteins on a non-reducing SDS-PAGE gel 1, 2. On a reducing SDS- PAGE gel system, this trail resolved to a significant extent into a single band for both human Fcγ4-IFN-βso1 5 and human Fcγ4h-IFN-βso1 6, suggesting that the aggregation was largely driven by the presence of covalent disulfide bonds. Therefore, the introduction of the single point mutation C17S into the human Fcγ4h-IFN-βso1 fusion protein 7 restored proper folding of the protein 7. Moreover, it was found by analytical size exclusion chromatography (SEC), that, whereas non- aggregated protein of the parent molecule could not be obtained, at least 10% of Fc-IFN-βsol(C17S) was non-aggregated after purification with Protein A. Therefore, the introduction of the single point mutation C17S into the Fc-IFN-βso1 fusion protein facilitated the production of non-aggregated material. Furthermore, introduction of a linker peptide at the junction between the Fc region and the IFN-β moiety resulted in about a two-fold increase in yield of non-aggregated material over Fc-IFN-βsol(C17S) without the linker. Expression from, for example, a nucleotide sequence encoding the fusion protein Fc-linker-IFN-βsol(C17S F50H H131A H140A) wherein the linker is Gly4SerGly4SerGly3SerGly (SEQ ID NO:1), as shown in FIGS. 19-1 through 19-3, contained on an appropriate plasmid, in a mammalian cell culture system resulted in the efficient production of the fusion protein Fc-linker-IFN-βso!(C17S F50H H131A H140A). It was found that this fusion protein product contained about 50% non-aggregated material after purification by Protein A, as assessed by analytical SEC, which represents a considerable further improvement over the results obtained with Fc-IFN-βs0' protein containing a single point mutation in IFN-β, Fc-linker-IFN-βso!(C17S). A similar further increase in expression characteristics was seen with the Fc-IFN-βso1 protein Fc-linker- IFN-βsol(C17S L57A H131A H140T). As previously mentioned, the invention provides nucleic acid sequences encoding and amino acid sequences defining fusion proteins including an immunoglobulin Fc region and at least one target protein, referred to herein as IFN-β or variants thereof. Three exemplary embodiments of protein constructs embodying the invention are illustrated in the drawing as FIGS. IA-I C. Because dimeric constructs are preferred, all are illustrated as dimers cross-linked by a pair of disulfide bonds between cysteines in adjacent subunits. In the drawings, the disulfide bonds 11, 12 are depicted as linking together the two immunoglobulin heavy chain Fc regions 1, 1' via an immunoglobulin hinge region within each heavy chain, and thus are characteristic of native forms of these molecules. While constructs including the hinge region of Fc are preferred and have shown promise as therapeutic agents, the invention contemplates that the crosslinking at other positions may be chosen as desired. Furthermore, under some circumstances, dimers or multimers useful in the practice of the invention may be produced by non- covalent association, for example, by hydrophobic interaction. Because homodimeric constructs are important embodiments of the invention, the drawings illustrate such constructs. It should be appreciated, however, that heterodimeric structures also are useful in the practice of the invention. FIG. IA illustrates a dimeric construct, also termed a "unit dimer", produced in accordance with the principles set forth herein (see, for example, Example 1). Each monomer of the homodimer includes an immunoglobulin Fc region 1 including a hinge region, a CH2 domain and a CH3 domain. Attached directly, i.e., via a polypeptide bond, to the C terminus of the Fc region is IFN-βso1 2. It should be understood that the Fc region may be attached to IFN-βso1 protein via a polypeptide linker (not shown). FIGS. IB and 1C depict protein constructs of the invention which include as a target protein plural IFN-β proteins arranged in tandem and connected by a linker. In FIG. IB, the target protein includes full length IFN-β 2, a polypeptide linker made of glycine and serine residues 4, and an active variant of IFN-β 3. This construct may be depicted by the formula Fc-X-X wherein the Xs represent different target proteins. FIG. 1C differs from the construct of FIG. IB in that the most C-terminal protein domain includes a second, full length copy of IFN-β 2. This construct may be depicted by the formula Fc-X-X, where the Xs represent identical target proteins. Although FIGS. IA-I C represent Fc-X constructs, where X is the target protein, it is contemplated that useful proteins of the invention may also be depicted by the formula X-Fc-X, wherein the Xs may represent the same or different target proteins. As shown in FIGS. IB and 1C, the fusion protein may include a second target protein (Fc-X-X). For example, in addition to a fusion protein having a first IFN-β target protein, the fusion protein may also include a second mature, full length IFN-β or an active IFN-βso1 variant or a bioactive fragment thereof. In one aspect, the active variant is a variant in which one or more amino acid residues in the IFN-β moiety is substituted for another amino acid residue. Several IFN-β substitution variants were discussed previously. For example, a cysteine at position 17, corresponding to the native mature IFN-β may be replaced with a serine, a valine, an alanine or a methionine. In this type of construct, the first and second proteins can be the same protein, as in, for example, FIG. 1C, or they may be different proteins, as in, for example, FIG. IB. The first and second proteins may be linked together, either directly or by means of a polypeptide linker. Alternatively, both proteins may be linked either directly or via a polypeptide linker, to the immunoglobulin Fc region. In a further embodiment, the first protein can be connected to the N-terminus of the immunoglobulin Fc region and the second protein can be connected to the C-terminus of the immunoglobulin Fc region. In one embodiment, two fusion proteins may be linked to form dimers. The two fusion proteins may associate, either covalently, for example, by a disulfide bond, a polypeptide bond or a crosslinking agent, or non-covalently, to produce a dimeric protein, hi a preferred embodiment, the two fusion proteins are associated covalently by means of at least one and more preferably two interchain disulfide bonds via cysteine residues, preferably located within immunoglobulin hinge regions disposed within the immunoglobulin Fc regions of each chain. Other embodiments of the invention include multivalent and multimeric forms of IFN-β fusion proteins and combinations thereof. As used herein, the term "multivalent" refers to a recombinant molecule that incorporates two or more biologically active segments. The protein fragments forming the multivalent molecule optionally may be linked through a polypeptide linker which attaches the constituent parts and permits each to function independently. As used herein, the term "bivalent" refers to a multivalent recombinant molecule having the configuration Fc-X, where X is an IFN-β protein. The two proteins may be linked through a peptide linker. Constructs of the type shown can increase the apparent binding affinity between the protein and its receptor. As used herein, the term "multimeric" refers to the stable association of two or more polypeptide chains either covalently, for example, by means of a covalent interaction, for example, a disulfide bond, or non-covalently, for example, by hydrophobic interaction. The term multimer is intended to encompass both homomultimers, wherein the subunits are the same, as well as, heteromultimers, wherein the subunits are different. As used herein, the term "dimeric" refers to a specific multimeric molecule where two polypeptide chains are stably associated through covalent or non-covalent interactions. Such constructions are shown schematically in FIG. IA. It should be understood that the immunoglobulin Fc region including at least a portion of the hinge region, a CH2 domain and a CH3 domain, typically forms a dimer. Many protein ligands are known to bind to their receptors as a dimer. If a protein ligand X dimerizes naturally, the X moiety in an Fc-X molecule will dimerize to a much greater extent, since the dimerization process is concentration dependent. The physical proximity of the two X moieties connected by Fc would make the dimerization an intramolecular process, greatly shifting the equilibrium in favor of the dimer and enhancing its binding to the receptor. As used herein, the tenn "polypeptide linker" is understood to mean a polypeptide sequence that can link together two proteins that in nature are not naturally linked together. The polypeptide linker preferably includes a plurality of amino acids such as ' alanine, glycine and serine or combinations of such amino acids. Preferably, the polypeptide linker includes a series of glycine and serine peptides about 10-15 residues in length. See, for example, U.S. Patent Nos. 5,258,698 and 5,908,626. A preferred linker polypeptide of the invention is Gly4SerGly4SerGly3SerGly (SEQ ID NO:1). However, it is contemplated, that the optimal linker length and amino acid composition may be determined by routine experimentation by methods well known in the art. As used herein, the term "interferon-β or IFN-β" is understood to mean not only full length mature interferon-β, for example, human IFN-β, but also homologs, variants and bioactive fragments or portions thereof. Known sequences of IFN-β may be found in GenBank. The term "interferon-β" or "IFN-β" also includes naturally occurring IFN-β and IFN-β-like proteins, moieties and molecules as well as IFN-β that is recombinantly produced or artificially synthesized. The term "bioactive fragment" or portion refers to any IFN-β protein fragment that has at least 5%, more preferably at least 10%, and most preferably at least 20% and optimally at least 30%, 40%, 50%, 60%, 70%, 80%, 90% or 100% of the biological activity of the template human IFN-β protein of SEQ ID NO:2, determined using the antiviral activity assay or cellular growth inhibition assays, as described in Examples 6 and 7. The term "variants" includes species and allelic variants, as well as other naturally occurring or non-naturally occurring variants, for example, generated by genetic engineering protocols, that are at least 70% similar or 60% identical, more preferably at least 75% similar or 65% identical, and most preferably at least 80% similar or 70% identical to the mature human IFN-β protein disclosed in SEQ ID NO:2. In order to determine whether a candidate polypeptide has the requisite percentage similarity or identity to a reference polypeptide, the candidate amino acid sequence and the reference amino acid sequence are first aligned using the dynamic programming algorithm described in Smith and Waterman (1981) J. MOL. BIOL. 147:195-197, in combination with the BLOSUM62 substitution matrix described in FIG. 2 of Henikoff and Henikoff (1992), "Amino acid substitution matrices from protein blocks", PROC. NATL. ACAD. SCI. USA 89:10915-10919. For the present invention, an appropriate value for the gap insertion penalty is -12, and an appropriate value for the gap extension penalty is -4. Computer programs performing alignments using the algorithm of Smith- Waterman and the BLOSUM62 matrix, such as the GCG program suite (Oxford Molecular Group, Oxford, England), are commercially available and widely used by those skilled in the art. Once the alignment between the candidate and reference sequence is made, a percent similarity score may be calculated. The individual amino acids of each sequence are compared sequentially according to their similarity to each other. If the value in the BLOSUM62 matrix corresponding to the two aligned amino acids is zero or a negative number, the pair-wise similarity score is zero; otherwise the pair-wise similarity score is 1.0. The raw similarity score is the sum of the pair-wise similarity scores of the aligned amino acids. The raw score then is normalized by dividing it by the number of amino acids in the smaller of the candidate or reference sequences. The normalized raw score is the percent similarity. Alternatively, to calculate a percent identity, the aligned amino acids of each sequence again are compared sequentially. If the amino acids are non- identical, the pair-wise identity score is zero; otherwise the pair-wise identity score is 1.0. The raw identity score is the sum of the identical aligned amino acids. The raw score is then normalized by dividing it by the number of amino acids in the smaller of the candidate or reference sequences. The normalized raw score is the percent identity. Insertions and deletions are ignored for the purposes of calculating percent similarity and identity. Accordingly, gap penalties are not used in this calculation, although they are used in the initial alignment. Variants may also include other IFN-β mutant proteins having IFN-β-like activity. Species and allelic variants, include, but are not limited to human and mouse IFN-β sequences. The human and mouse mature IFN-β proteins are depicted in SEQ ID NOs. :2 and 11, and in FIGS. 3 and 12 respectively. Furthermore, the IFN-β sequence may include a portion or all of the consensus sequence set forth in SEQ ID NO:2, wherein the IFN-β has at least 5%, preferably at least 10%, more preferably at least 20%, 30% or 40%, most preferably at least 50%, and optimally 60%, 70%, 80%, 90% or 100% of the biological activity of the mature human IFN-β of SEQ ID NO:2, as determined using the antiviral activity assay or cellular growth inhibition assay of Examples 6 and 7. The three-dimensional structure of IFN-β has been solved by X-ray crystallography (Karpusas et al, 1997, PNAS 94: 11813). Although in the crystallized state, IFN-β molecule is a dimer with a zinc ion at the dimer interface, it is thought that IFN-β need not be a dimer in order to be active. Structurally IFN-β contains an additional alpha-helical segment with respect to classical four helix bundle proteins, which is formed within the C-D loop, so that the canonical bundle structure is formed by helices A, B, C and E. Interestingly, the structure also reveals a portion of the glycan moiety which is coupled to amino acid N80 at the start of helix C and is ordered along a portion of the protein, most likely shielding some of the surface-exposed hydrophobic amino acid residues from solvent. Glycosylation of IFN-β has been shown to be important for the solubility and stability of the molecule, and this could explain the propensity of the non- glycosylated IFN-β molecule to aggregate. The free cysteine at position 17 in helix A appears proximal to the surface but buried, and, without wishing to be bound by theory, it is possible that scrambled disulfide bonds may in turn prevent the correct glycosylation of the protein. Dimerization of a ligand can increase the apparent binding affinity between the ligand and its receptor. For instance, if one interferon-beta moiety of an Fc-interferon- beta fusion protein can bind to a receptor on a cell with a certain affinity, the second interferon-beta moiety of the same Fc-Interferon-beta fusion protein may bind to a second receptor on the same cell with a much higher avidity (apparent affinity). This may occur because of the physical proximity of the second interferon-beta moiety to the receptor after the first interferon-beta moiety already is bound. In the case of an antibody binding to an antigen, the apparent affinity may be increased by at least ten thousand-fold, i.e., 104. Each protein subunit, i.e., "X," has its own independent function so that in a multivalent molecule, the functions of the protein subunits may be additive or synergistic. Thus, fusion of the normally dimeric Fc molecule to interferon-beta may increase the activity of interferon-beta.' Accordingly, constructs of the type shown in FIG. IA may increase the apparent binding affinity between interferon-beta and its receptor. Fc Moiety The IFN-β fusion proteins disclosed herein are expressed as fusion proteins with an Fc region of an immunoglobulin. As is known, each immunoglobulin heavy chain constant region includes four or five domains. The domains are named sequentially as follows: CHl-hinge-CH2-CH3(-CH4). The DNA sequences of the heavy chain domains have cross-homology among the immunoglobulin classes, e.g., the CH2 domain of IgG is homologous to the CH2 domain of IgA and IgD5 and to the CH3 domain of IgM and IgE. As used herein, the term, "immunoglobulin Fc region" is understood to mean the carboxyl-terminal portion of an immunoglobulin chain constant region, preferably an immunoglobulin heavy chain constant region, or a portion thereof. For example, an immunoglobulin Fc region may include 1) a CH2 domain 2) a CH3 domain, 3) a CH4 domain 4) a CH2 domain and a CH3 domain, 5) a CH2 domain and a CH4 domain, 6) a CH3 domain and a CH4 domain or 7) a combination of an immunoglobulin hinge region and/or a CH2 domain and/or CH3 domain and/or a CH4 domain. In one embodiment, the immunoglobulin Fc region includes at least an immunoglobulin hinge region, while in another embodiment the immunoglobulin Fc region includes at least one immunoglobulin constant heavy region, for example, a CH2 domain or a CH3 domain, and depending on the type of immunoglobulin used to generate the Fc region, optionally a CH4 domain. In another embodiment, the Fc region includes a hinge region, a CH2 domain and a CH3 domain, and preferably lacks the CHl domain, while in another embodiment, the Fc region includes a hinge region and a CH2 domain. In yet another embodiment, the Fc region includes a hinge region and a CH3 domain. Ih a further embodiment, the Fc region contains a functional binding site for the Fc protection receptor, FcRp. The binding site for FcRp includes amino acids in both the CH2 and CH3 domains and the Fc- FcRp interaction contributes significantly to the extended serum half-life of Fc fusion proteins. Although immunoglobulin Fc regions and component constant heavy domains may be from any immunoglobulin class, a preferred class of immunoglobulin for the Fc- IFN-β fusion proteins of the invention is IgG (Igγ) (γ subclasses 1, 2, 3, or 4). The nucleotide and amino acid sequences of human Fcγl are set forth in SEQ DD NOs: 78 and 79. Other classes of immunoglobulin, IgA (Igα), IgD (Igδ), IgE (Igε) and IgM (Igμ), can also be used. The choice of appropriate immunoglobulin heavy chain constant regions is discussed in detail in U.S. Patent Nos. 5,541,087, and 5,726,044. The choice of particular immunoglobulin heavy chain constant region sequences from certain immunoglobulin classes and subclasses to achieve a particular result is considered to be within the level of skill in the art. The portion of the DNA construct encoding the immunoglobulin Fc region preferably includes at least a portion of a hinge domain, and preferably at least a portion of a CH3 domain of Fcγ or the homologous domains in any of IgA, IgD, IgE, or IgM. It is contemplated that the Fc region used in the generation of the fusion proteins containing the IFN-β variants can be adapted to the specific application of the molecule, hi one embodiment, the Fc region is derived from an immunoglobulin γl isotype or variants thereof. The use of human Fcγl as the Fc region sequence has several advantages. For example, an Fc region derived from an immunoglobulin γl isotype can be used when targeting the fusion protein to the liver is desired. Additionally, if the Fc fusion protein is to be used as a biophaπnaceutical, the Fcγl domain may confer effector function activities to the fusion protein. The effector function activities include the biological activities such as placental transfer and increased serum half-life. The immunoglobulin Fc region also provides for detection by anti-Fc ELISA and purification through binding to Staphylococcus aureus protein A ("Protein A"). Ih certain applications, however, it may be desirable to delete specific effector functions from the immunoglobulin Fc region, such as Fc receptor binding and/or complement fixation. When an Fc region derived from immunoglobulin γl is used, a lysine at the carboxy terminus of the immunoglobulin Fc region is typically replaced with an alanine. This improves the circulating half life of the Fc-IFN-βso1 fusion protein. Other embodiments of Fc-IFN-βso1 fusion proteins use Fc regions derived from a different immunoglobulin γ isotype i.e. γ2, γ3, or γ4, or variants thereof. The Fc region can include a hinge region derived from a different immunoglobulin isotype than the Fc region itself. For example, some embodiments of Fc-IFN-βso1 fusion proteins contain a hinge region derived from an immunoglobulin γl or a variant thereof. For instance, the immunoglobulin Fc region can be derived from an immunoglobulin γ2 isotype and include a hinge region derived from an immunoglobulin γl isotype or a variant thereof. In one embodiment, a cysteine residue of the γl hinge is modified, hi a further embodiment, the first cysteine of the γl lήnge is modified. In yet another embodiment, a serine is substituted in place of the first cysteine of the γl hinge. Because the immunoglobulin γ2 isotype is ineffective in mediating effector functions and displays vastly reduced binding to Fcγ receptor (FcγR), it may be expected that this particular configuration of IFN-β fusion variant more closely mimics the biological activity of the free IFN-β molecule and in addition has the most enhanced circulating half-life when administered to a mammal. Just as with γl, it is preferable to mutate the carboxy-terminal lysine of the Fc region to alanine in order to improve the circulating half life of the Fc- IFN-βso1 fusion protein. As previously stated, the Fc region of Fc-IFN-βso1 fusion proteins of the invention can be derived from an immunoglobulin γ4 isotype. In some embodiments of the invention, an immunoglobulin γ4 isotype is modified to contain a hinge region derived from an immunoglobulin γl isotype or a variant thereof, hi one embodiment, a cysteine residue of the γl hinge is modified, hi a further embodiment, the first cysteine of the γl hinge is modified, hi yet another embodiment, a serine is substituted in place of the first cysteine of the γl hinge. Like immunoglobulin γ2 isotypes, immunoglobulin γ4 isotypes also exhibit lower affinity towards FcγR and thus offer similar advantages in reducing immune effector functions. When an Fc region derived from γ 1, 2, 3 or 4 is used, a lysine at the carboxy-terminus of the immunoglobulin Fc region is typically replaced with an alanine. Immunoglobulin γ4 is a preferred Fc region for making Fc-EFN-βso1 fusion proteins wherein the IFN-β moiety includes alterations to one of more amino acid residues at position 17, 50, 57, 130, 131, 136 and/or 140. An exemplary amino acid sequence of an Fc-IFN-βso1 fusion protein of the invention which includes an Fc region of immunoglobulin γ4 isotype modified to contain a hinge region derived from an immunoglobulin γl is shown in FIG. 5 (SEQ ID NO:4). Depending on the application, constant region genes from species other than human, for example, mouse or rat, may be used. The immunoglobulin Fc region used as a fusion partner in the DNA construct generally may be from any mammalian species. Where it is undesirable to elicit an immune response in the host cell or animal against the Fc region, the Fc region may be derived from the same species as the host cell or animal. For example, a human immunoglobulin Fc region can be used when the host animal or cell is human; likewise, a murine immunoglobulin Fc region can be used where the host animal or cell will be a mouse. Nucleic acid sequences encoding, and amino acid sequences defining a human immunoglobulin Fc region useful in the practice of the invention are set forth in SEQ ID NO:78 and SEQ ID NO:79 respectively. However, it is contemplated that other immunoglobulin Fc region sequences useful in the practice of the invention may be found, for example, by those encoded by nucleotide sequences of the heavy chain constant region which includes the Fc region sequence as disclosed in the Genbank and/or EMBL databases, for example, AF045536.1 (Macaca fuscicularis, nucleotide sequence SEQ ID NO:20; amino acid sequence SEQ ID NO:21), AF045537.1 (Macaca mulatto, nucleotide sequence SEQ ID NO:22; amino acid sequence SEQ ID NO:23), AB016710 (Felis catus, nucleotide sequence SEQ ID NO:24; amino acid sequence SEQ E) NO:25), K00752 (Oryctolagus cuniculus, nucleotide sequence SEQ ID NO:26; amino acid sequence SEQ ID NO:27), U03780 (Sus scrofa, nucleotide sequence SEQ E) NO:28; amino acid sequence SEQ E) NO:29), Z48947 (Camelus dromedaries, nucleotide sequence SEQ E) NO:30), (Bos taurus, nucleotide sequence SEQ E) NO:31; amino acid sequence SEQ E) NO:32), L07789 (Mustela vison, nucleotide sequence SEQ E) NO:33; amino acid sequence SEQ E) NO:34), X69797 (Ovis aries, nucleotide sequence SEQ E) NO:35; amino acid sequence SEQ E) NO:36), U17166 (Cricetulus migratorius, nucleotide sequence SEQ E) NO:37; amino acid sequence SEQ E) NO:38), X07189 (Rattus rattus, nucleotide sequence SEQ E) NO:39; amino acid sequence SEQ TD NO:40), AF157619.1 (Trichosurus vulpecula, nucleotide sequence SEQ E) NO:41; amino acid sequence SEQ E) NO:42), or AF035195 (Monodelphis domestica, nucleotide sequence SEQ E) NO:43; amino acid sequence SEQ E) NO:44). Furthermore, it is contemplated that substitution or deletion of amino acids within the immunoglobulin heavy chain constant regions may be useful in the practice of the invention. One example may include introducing amino acid substitutions in the upper CH2 region to create an Fc variant with reduced affinity for Fc receptors (Cole et al. (1997) J. Immunol. 159:3613). One of ordinary skill in the art can prepare such constructs using well known molecular biology techniques. It is understood that the present invention exploits conventional recombinant DNA methodologies for generating the Fc fusion proteins useful in the practice of the invention. The Fc fusion constructs preferably are generated at the DNA level, and the resulting DNAs integrated into expression vectors, and expressed to produce the fusion proteins of the invention. As used herein, the term "vector" is understood to mean any nucleic acid including 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, the term "gene expression" or "expression" of a target protein, is understood to mean the transcription of a DNA sequence, translation of the mRNA transcript, and secretion of an Fc fusion protein product. A useful expression vector is pdCs (Lo et al. (1988) Protein Engineering 11 :495), in which the transcription of the Fc-X gene utilizes the enhancer/promoter of the human cytomegalovirus and the SV40 polyadenylation signal. The enhancer and promoter sequence of the human cytomegalovirus used was derived from nucleotides -601 to +7 of the sequence provided in Boshart et al. (1985) Cell 41:521. The vector also contains the mutant dihydrofolate reductase gene as a selection marker (Simonsen and Levinson (1983) Proc. Nat. Acad. Sci. USA 80:2495). An appropriate host cell can be transformed or transfected with the DNA sequence of the invention, and utilized for the expression and/or secretion of the target protein. Currently preferred host cells for use in the invention include immortal hybridoma cells, NS/0 myeloma cells, 293 cells, Chinese hamster ovary cells, HeLa cells, and COS cells. One expression system that has been used to produce high level expression of fusion proteins in mammalian cells is a DNA construct encoding, in the 5' to 3' direction, a secretion cassette, including a signal sequence and an immunoglobulin Fc region, and a target protein such as IFN-β. Several target proteins have been expressed successfully in such a system and include, for example, IL2, CD26, Tat, Rev, OSF-2, βIG-H3, IgE Receptor, PSMA, and gpl20. These expression constructs are disclosed in U.S. Patent Nos. 5,541,087 and 5,726,044 to Lo et al. The fusion proteins of the invention may or may not be include a signal sequence when expressed. As used herein, the term "signal sequence" is understood to mean a segment which directs the secretion of the IFN-β fusion protein and thereafter is cleaved following translation in the host cell. The signal sequence of the invention is a polynucleotide which encodes an amino acid sequence which initiates transport of a protein across the membrane of the endoplasmic reticulum. Signal sequences which are useful in the invention include antibody light chain signal sequences, e.g., antibody 14.18 (Gillies et. al. (1989) J. Immunol. Meth. 125:191), antibody heavy chain signal sequences, e.g., the M0PC141 antibody heavy chain signal sequence (Sakano et al. (1980) Nature 286:5774), and any other signal sequences which are known in the art (see, e.g., Watson (1984) Nucleic Acids Research 12:5145). Signal sequences have been well characterized 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 signal sequence may be cleaved from the amino-terminus of the fusion protein during secretion. This results in the secretion of an Fc fusion protein consisting of the immunoglobulin Fc region and the target protein. A detailed discussion of signal peptide sequences is provided by von Heijne (1986) Nucleic Acids Res. 14:4683. As would be apparent to one of skill in the art, the suitability of a particular signal sequence for use in the secretion cassette may require some routine experimentation. Such experimentation will include determining the ability of the signal sequence to direct the secretion of an Fc fusion protein and also a determination of the optimal configuration, genomic or cDNA, of the sequence to be used in order to achieve efficient secretion of Fc fusion proteins. Additionally, one skilled in the art is capable of creating a synthetic signal peptide following the rules presented by von Heijne, referenced above, and testing for the efficacy of such a synthetic signal sequence by routine experimentation. A signal sequence can also be referred to as a "signal peptide," "leader sequence," or "leader peptides." The fusion of the signal sequence and the immunoglobulin Fc region is sometimes referred to herein as secretion cassette. An exemplary secretion cassette useful in the practice of the invention is a polynucleotide encoding, in a 5' to 3' direction, a signal sequence of an immunoglobulin light chain gene and an Fcγl region of the human immunoglobulin γl gene. The Fcγl region of the immunoglobulin Fcγl gene preferably includes at least a portion of the immunoglobulin hinge domain and at least the CH3 domain, or more preferably at least a portion of the hinge domain, the CH2 domain and the CH3 domain. As used herein, the "portion" of the immunoglobulin hinge region is understood to mean a portion of the immunoglobulin hinge that contains at least one, preferably two cysteine residues capable of forming interchain disulfide bonds. The DNA encoding the secretion cassette can be in its genomic configuration or its cDNA configuration. Under certain circumstances, it may be advantageous to produce the Fc region from human immunoglobulin Fcγ2 heavy chain sequences. Although Fc fusions based on human immunoglobulin γl and γ2 sequences behave similarly in mice, the Fc fusions based on the γ2 sequences can display superior pharmacokinetics in humans. In another embodiment, the DNA sequence encodes a proteolytic cleavage site interposed between the secretion cassette and the target protein. A cleavage site provides for the proteolytic cleavage of the encoded fusion protein thus separating the Fc domain from the target protein. As used herein, "proteolytic cleavage site" is understood to mean amino acid sequences which are preferentially cleaved by a proteolytic enzyme or other proteolytic cleavage agents. Useful proteolytic cleavage sites include amino acids sequences which are recognized by proteolytic enzymes such as trypsin, plasmin or enterokinase K. Many cleavage site/cleavage agent pairs are known (see, for example, U.S. Patent No. 5,726,044). Further, substitution or deletion of constructs of these constant regions, in which one or more amino acid residues of the constant region domains are substituted or deleted also would be useful. One example would be to introduce amino acid substitutions in the upper CH2 region to create an Fc variant with reduced affinity for Fc receptors (Cole et al. (1997) J. Immunol. 159: 3613). One of ordinary skill in the art can prepare such constructs using well known molecular biology techniques. The fusion constructs disclosed herein produced high levels of Fc-IFN-βso1. The initial clones produced about 100 μg/mL of altered Fc-IFN-βso1, which could be purified to homogeneity by Protein A affinity chromatography. Expression levels often can be increased several fold by subcloning. As stated above, it was found that when IFN-β with the cysteine at position 17 replaced with a serine, an alanine, a valine or a methionine is expressed as Fc fusion molecules, high levels of expression were obtained, presumably because the amino acid substitution at position 17 of the IFN-βso1 protein prevents aberrant disulfide bond formation in the protein and the Fc region acts as a carrier, helping the polypeptide to fold correctly and to be secreted efficiently. Similarly, other Fc-IFN-βso1 fusion proteins of the invention including the mutation C17S, such as, for example Fc-(linker)-IFN-βso1 (C17S F50H H131A H140A) and Fc-(linker)-IFN-βsol(C17S L57A H131A H140T) are equally well expressed. Moreover, the Fc region is also glycosylated and highly charged at physiological pH. Therefore, the Fc region can help to solubilize hydrophobic proteins. In addition to high levels of expression, Fc-IFN-βso! proteins exhibited greater bioactivity than the parental (un-modified) Fc-IFN-β fusion protein, as measured in a cell based anti-viral assay (Example 6), and were comparable to the bioactivity of a commercial preparation of IFN-β obtained from R&D Systems (Minneapolis, MN). In addition to the high levels of expression, altered Fc-IFN-β fusion proteins exhibited longer serum half-lives compared to unaltered Fc-IFN-β fusion proteins. For 5 example, the circulating half-life of Fc-IFN-βso1 including the mutation C17S is found to be significantly greater than that of the parent Fc-IFN-β fusion protein (see Example 8). The fusion proteins of the invention provide several important clinical benefits. As demonstrated in the tests of biological assays in Examples 6 and 7, the biological activity of altered Fc-IFN-βso1 is significantly higher than that of unaltered Fc-IFN-β. l o Another embodiment of the present invention provides constructs having various structural conformations, e.g., bivalent or multivalent constructs, dimeric or multimeric constructs, and combinations thereof. Such functional conformations of molecules of the invention allow the synergistic effect of IFN-β and other anti- viral and anti-cancer proteins to be explored in animal models. 15 An important aspect of the invention is that the sequences and properties of various IFN-β proteins and encoding DNAs are quite similar. In the context of Fc-X fusions, the properties of IFN-β proteins and encoding DNAs are essentially identical, so that a common set of techniques can be used to generate any Fc-IFN-β DNA fusion, to express the fusion, to purify the fusion protein, and to administer the fusion protein for 0 therapeutic purposes. The present invention also provides methods for the production of IFN-β of non- human species as Fc fusion proteins. Non-human IFN-β fusion proteins are useful for preclinical studies of IFN-β because efficacy and toxicity studies of a protein drug must be performed in animal model systems before testing in human beings. A human protein 25 may not work in a mouse model since the protein may elicit an immune response, and/or exhibit different pharmacokinetics skewing the test results. Therefore, the equivalent mouse protein is the best surrogate for the human protein for testing in a mouse model. The present invention provides methods of treating various cancers, viral diseases, other diseases, related conditions and causes thereof by administering the DNA, RNA or 30 proteins of the invention to a mammal having such condition. Related conditions may include, but are not limited to multiple sclerosis; a variety of malignancies, such as acute myeloid leukemia, multiple myeloma, Hodgldn's disease, basal cell carcinoma, cervical dysplasia and osteosarcoma; a variety of viral infections, including viral hepatitis, herpes zoster and genitalis, papilloma viruses, viral encephalitis, and cytomegalovirus pneumonia. In view of the broad roles played by IFN-β in modulating immune responses, the present invention also provides methods for treating conditions alleviated by the administration of IFN-β. These methods include administering to a mammal having the condition, which may or may not be directly related to viral infection or cancer, an effective amount of a composition of the invention. For example, a nucleic acid, such as DNA or RNA, encoding an Fc-IFN-βso! fusion protein can be administered to a subject, preferably a mammal, as a therapeutic agent. Additionally, a cell containing a nucleic acid encoding an Fc-IFN-βso1 fusion protein can be administered to a subject, preferably a mammal, as a therapeutic agent. Furthermore, an Fc-IFN-βso1 protein can be administered to a subject, preferably a mammal, as a therapeutic agent. The proteins of the invention not only are useful as therapeutic agents, but one skilled in the art recognizes that the proteins are useful in the production of antibodies for diagnostic use. Likewise, appropriate administration of the DNA or RNA, e.g., in a vector or other delivery system for such uses, is included in methods of use of the invention. Compositions of the present invention may be administered by any route which is compatible with the particular molecules. It is contemplated that the compositions of the present invention may be provided to an animal by any suitable means, directly (e.g. , locally, as by injection, implantation or topical administration to a tissue locus) or systemically (e.g., parenterally or orally). Where the composition is to be provided parenterally, such as by intravenous, subcutaneous, ophthalmic, intraperitoneal, intramuscular, buccal, rectal, vaginal, intraorbital, intracerebral, intracranial, intraspinal, intraventricular, intrathecal, intracisternal, intracapsular, intranasal or by aerosol administration, the composition preferably includes part of an aqueous or physiologically compatible fluid suspension or solution. Thus, the carrier or vehicle is physiologically acceptable so that in addition to delivery of the desired composition to the patient, it does not otherwise adversely affect the patient's electrolyte and/or volume balance. The fluid medium for the agent thus can include normal physiologic saline. The DNA constructs (or gene constructs) of the invention also can be used as a part of a gene therapy protocol to deliver nucleic acids encoding IFN-β or a fusion protein construct thereof. The invention features expression vectors for in vivo transfection and expression of IFN-β or a fusion protein construct thereof in particular cell types so as to reconstitute or supplement the function of IFN-β. Expression constructs of IFN-β, or fusion protein constructs thereof, may be administered in any biologically effective carrier, e.g. any formulation or composition capable of effectively delivering the IFN-β gene or fusion protein construct thereof to cells in vivo. Approaches include insertion of the subject gene in viral vectors including recombinant retroviruses, adenovirus, adeno- associated virus, and herpes simplex virus- 1, or recombinant bacterial or eukaryotic plasmids. Preferred dosages per administration of nucleic acids encoding the fusion proteins of the invention are within the range of 1 μg/m2 to 100 mg/m2, more preferably 20 μg/m2 to 10 mg/m2, and most preferably 400 μg/m2 to 4 mg/m2 . It is contemplated that the optimal dosage and mode of administration may be determined by routine experimentation well within the level of skill in the art. Preferred dosages of the fusion protein per administration are within the range of 0.1 mg/m2 - 100 mg/m2, more preferably, 1 mg/m2 - 20 mg/m2, and most preferably 2 mg/m2 - 6 mg/m2. It is contemplated that the optimal dosage, however, also depends upon the disease being treated and upon the existence of side effects. However, optimal dosages may be determined using routine experimentation. Administration of the fusion protein may be by periodic bolus injections, or by continuous intravenous or intraperitoneal administration from an external reservoir (for example, from an intravenous bag) or internal (for example, from a bioerodable implant). Furthermore, it is contemplated that the fusion proteins of the invention also may be administered to the intended recipient together with a plurality of different biologically active molecules. It is contemplated, however, that the optimal combination of fusion protein and other molecules, modes of administration, dosages may be determined by routine experimentation well within the level of skill in the art. The invention is illustrated further by the following non-limiting examples.
EXAMPLES
Example 1. Cloning of huFc-huInterferon-beta (huFc-IFN-β) and huFc-IFN-βS0' mutants. Human interferon β (IFN-β) cDNA was ordered from American Type Culture Collection (ATCC Number 31903). The sequence for the mature form was amplified by Polymerase Chain Reactions (PCR). The forward primer used in the amplification reactions was 5' C CCG GGT ATG AGC TAC AAC TTG CTT (SEQ ID NO:45), where the sequence CCCGGGT encodes the carboxy terminus of the CH3 without the lysine codon, as well as the restriction endonuclease site Smal CCCGGG (Lo et al, Protein Engineering (1998) 11:495), and the sequence in bold encodes the N-terminus of the mature IFN-β coding sequence. The reverse primer for this reaction was 5' CTC GAG TCA GTT TCG GAG GTA ACC TGT (SEQ ID NO:46), where TCA is the anticodon of the translation stop codon, and CTCGAG is the restriction site Xho I. The amplified 450 bp PCR product was cloned into the pCRII vector (Invitrogen), and its sequence verified. The Smal-Xhol restriction fragment with the completely correct mature IFN-β sequence was used for cloning into the expression vector pdCs-huFc, such that the coding sequence of mature IFN-β was fused in frame to the 3'-end of the Fc coding sequence. The expression plasmid pdCs-huFc-IFN-β was constructed by ligating the Smal-Xhol restriction fragment containing the mature IFN-β cDNA with the Smal-Xhol restriction fragment of the pdCs-huFc vector according to Lo et al., (Protein Engineering (1998) 11 :495). The huFc DNA corresponds to a sequence that when expressed produces the Fc fragment of the human immunoglobulin γ4 with a modified γl hinge sequence. The amino acid sequence is shown in SEQ ID NO:77. To generate further fusion proteins including the IFN-β fused to Fc moieties of a different isotype or containing other alterations, the same cloning strategy was used, while substituting the appropriate version of pdCs-huFc vector. Thus, the Smal-Xhol restriction fragment of IFN-β was inserted into pdCS-huFc vector digested with Smal and Xhol, which encoded either an immunoglobulin γ 4 isotype with a γ 4-derived hinge region, or an immunoglobulin γ 1 isotype, or an immunoglobulin γ 2 isotype, or an immunoglobulin γ2 isotype but with an altered immunoglobulin γ 1 -derived hinge region. Because the introduction of the Smal cloning site into the vector encoding an immunoglobulin γ 4 isotype does not result in a silent mutation in the expressed protein of the Fc moiety, the protein sequence encoded by the nucleic acid sequence around the Smal site is LSLSPG (SEQ ED NO:53). Had the mutation been silent, the sequence would have present been LSLSLG (See e.g. FIG. 7, residues 101-106 or SEQ ID NO:76). The cysteine 17 to serine (C 17S) mutation was introduced into the EFN-β nucleotide sequence by an overlapping PCR method (Daugherty et al., (1991) Nucleic Acids Res. 19:2471) using complementary mutagenic primers. The forward primer sequence was: 5 ' AGA AGC AGC AAT TTT CAG AGT CAG AAG CTC CTG TGG CA (SEQ ED NO:47), where the underlined nucleotide indicates the introduced point mutation (TGT to AGT). Accordingly, the reverse primer was: 5' TG CCA CAG GAG CTT CTG ACT CTG AAA ATT GCT GCT TCT (SEQ ID NO:48). The PCR fragment generated by the overlapping PCR method was ligated to the pCRII vector, the sequence verified, and the Smal-Xhol fragment ligated to any of the pdCs-huFc expression vectors as described above. The amino acid sequence is shown as SEQ ID NO:3. The sequence of the mouse counterpart with the mutation is depicted in SEQ ID NO:12. As discussed above, the cysteine at position 17 is mutated to a serine in the Fc- IFN-βso1 protein that has the Fc portion including immunoglobulin γ 4 with a modified γ 1 hinge sequence. The amino acid sequence is shown as SEQ ID NO:4. To introduce a flexible linker sequence between the huFc moiety and the IFN-β moiety, a synthetic oligonucleotide duplex of the sequence 5' G GGT GCA GGG GGC GGG GGC AGC GGG GGC GGA GGA TCC GGC GGG GGC TC 3' (SEQ ID NO:49) was produced. This blunt-ended, double-stranded duplex was inserted at the unique Smal site of the expression vector pdCs-huFc-IFN-β by ligation. The orientation of the blunt- ended duplex in the resultant vector, pdCs-huFc-(GS linker)-IFN-β was confirmed by sequencing. As a result, the amino acid sequence GAGGGGSGGGGSGGGS (SEQ ID NO:50) was added between the proline (codon CCG) and the glycine (codon GGT) residues encoded by the C CCG GGT (SEQ ID NO:51) sequence containing the Smal site. The amino acid sequence of a huFc-(GS linker) IFN-β starting with the CH3 domain of the Fcγ4 isotype is shown in FIG. 6 (SEQ ID NO:5). When using this linker with immunoglobulin γ4 constructs of the invention, it is important to note that LSLSPG (SEQ ID NO:52) C-terminal amino acid sequence of immunoglobulin γ4 lacks the alanine residue present in the immunoglobulin γl, γ2 or γ3 C-terminal sequence LSLSPGA (SEQ ID NO:53). As stated earlier, the alanine is the result of mutating the native lysine residue. When the linker is inserted in the γl, γ2 or γ3 construct, terminal glycine and alanine residues are identically substituted by a glycine and alanine of the linker. Thus, when the linker is inserted into immunoglobulin γ4 Fc-IFN-β, the amino acid sequence gains an additional alanine residue when the C-terminal glycine is replaced by glycine and alanine. This is exemplified by comparing, for example, FIG. 5, residues 226-231 (SEQ ID NO:4) and FIG. 6, beginning at residue 101 (SEQ ID NO:5). Further Fc-IFN-βso1 protein variants can be produced that contain mutations in the IFN-β moiety. For example, Cl 7 may be altered to another amino acid, for instance alanine. In order to introduce the C17A mutation, the following mutagenic oligonucleotides are used: the forward primer is 5'AGA AGC AGC AAT TTT CAG GCT CAG AAG CTC CTG TGG CA 3', (SEQ ID NO:54), and the reverse primer is 5' TG CCA CAG GAG CTT CTG AGC CTG AAA ATT GCT GCT TCT 3', (SEQ ID NO:55), where the underlined nucleotides indicate the introduced mutations. Further mutations in Fc-IFN-βso1 were introduced in the IFN-β moiety by overlap PCR. Preferred IFN-β fusion proteins of the invention, Fcγ4h-(linker)-IFN-βsol(C17S L57A H131A H140A) and Fcγ4h-(linker)-IFN-βsol(C17S F50H H131A H140A), are produced by starting with the template Fcγ4h-linker-IFN-βsol(C17S) prepared using methods previously described herein. To introduce the H131A mutation to the Fcγ4h-(linker)-IFN-βsol(C17S) template, a first nucleic acid fragment is created by PCR using the forward primer sequence 5'CTC CCT GTC CCC GGG TGC AGG GGG (SEQ ID NO:56), which incorporates the restriction endonuclease Xmal site, and the reverse primer sequence 5' CTT GGC CTT CAG GTA GGC CAG AAT CCT CCC ATA ATA TC (SEQ ID NO:57), where GGC represents the Hl 31 A mutation. A second fragment of the fusion protein is created by PCR using the forward primer sequence 5'GAT ATT ATG GGA GGA TTC TGG CCT ACC TGA AGG CCA AG (SEQ ID NO:58), where GGC represents the H131 A mutation, and the reverse primer sequence 5' CTT ATC ATG TCT GGA TCC CTC GAG (SEQ ID NO:59), which incorporates the BamHI restriction site. The products from these reactions are purified on an electrophoretic gel according to standard methods. The gel purified fragments are then together subjected to PCR using the forward primer sequence 5'CTC CCT GTC CCC GGG TGC AGG GGG (SEQ ID NO:60), which incorporates the Xmal restriction site, and the reverse primer sequence 5' CTT ATC ATG TCT GGA TCC CTC GAG (SEQ ID NO:61), which incorporates the BamHI restriction site. This results in a nucleic acid encoding Fcγ4h-linker-IFN-βsol(C17S Hl 3 IA). Next, the H140A mutation is introduced by subjecting the Fcγh-linker-IFN- βsoI(C 17S H131A) to PCR to create a first fragment using the forward primer sequence 5'CTC CCT GTC CCC GGG TGC AGG GGG (SEQ ID NO:62), which incorporates the restriction endonuclease Xmal site, and the reverse primer sequence 5' GGT CCA GGC ACA GGC ACT GTA CTC CTT GGC (SEQ ID NO:63), where GGC represents the H140A mutation. A second fragment of the fusion protein is created by PCR using the forward primer sequence 5 ' GGC AAG GAG TAC AGT GCC TGT GCC TGG ACC (SEQ ID NO:64), where GCC represents the H140A mutation. The reverse primer sequence is 5' CTT ATC ATG TCT GGA TCC CTC GAG (SEQ ID NO:65), which incorporates the BamHI restriction site. The products from these reactions are purified on an electrophoretic gel according to standard methods. The gel purified fragments are then together subjected to PCR using the forward primer sequence 5'CTC CCT GTC CCC GGG TGC AGG GGG (SEQ ED NO:66), which incorporates the Xmal restriction site, and the reverse primer sequence 5' CTT ATC ATG TCT GGA TCC CTC GAG (SEQ ID NO: 67), which incorporates the BamHI restriction site. This results in a nucleic acid encoding Fcγ4h-linker-IFN-βsol(C17S H131 A H140A). Alternatively, this process may be followed to instead insert the H140T mutation of the invention by modifying the appropriate primers to express the threonine codon ACC. Finally, to introduce either the F50H mutation or the L57A mutation to the template Fcγ4h-linker-IFN-βsol(C17S H131 A H140A) template prepared in the previous step, a first nucleic acid fragment is created by PCR using the forward primer 5 'CTC CCT GTC CCC GGG TGC AGG GGG (SEQ ID NO:68), which incorporates the restriction endonuclease Xmal site, and either the reverse primer sequence 5' GAG CAT CTC ATA GAT GGT GGC TGC GGC GTC CT C (SEQ ID NO:69), where GGC represents the codon for creating theL57A mutation or the reverse primer sequence 5' GTCCTCCTTCTGATGCTGCTGCAGCTG(SEQIDNO:70),whereATG represents the codon creating the F50H mutation. To create the second fragment of the fusion protein for the L57A mutation, the template is subjected to PCR using the forward primer sequence 5' GAG GAC GCC GCA GCC ACC ATC TAT GAG ATG CTC (SEQ ID NO:71), where GCC represents the L57A mutation. To create the second fragment of the fusion protein for introducing the F50H mutation, the template is subjected to PCR using the forward primer sequence 5' CAG CTG CAG CAG CAT CAG AAG GAG GAC (SEQ ID NO:72), where CAT represents the F50H mutation. The reverse primer for production of the second fragment of either mutation is 5' CTT ATC ATG TCT GGA TCC CTC GAG (SEQ ID NO:73), which incorporates the BamHI restriction site. The products from these reactions are purified on an electrophoretic gel according to standard methods. The gel purified fragments are then used as the PCR to produce a nucleic acid encoding Fcγ4h-linker-IFN-βsol(C17S L57A H131A H140A) or Fcγ4h-linker-IFN- βsol(C17S F50H H131 A H140A). The forward and reverse primers for this reaction are 5'CTC CCT GTC CCC GGG TGC AGG GGG (SEQ ID NO:74) and 5' CTT ATC ATG TCT GGA TCC CTC GAG (SEQ ID NO:75), respectively, as used in previous steps.
Example 2. Transfection and Expression of Fc-IFN-β Fusion Proteins. For rapid analysis of protein expression, the plasmid pdCs-huFc-IFN-β, pdCs- huFc-IFN-βsol(C17S) or other huFc fusion protein variants containing huIFN-β were introduced into human embryonic kidney HEK 293 cells (ATCC# CRL-1573) by transient transfection using lipofectamine (Invitrόgen). To obtain stably transfected clones which express huFc-IFN-βsol(C17S), for example, the appropriate plasmid DNA was introduced into the mouse myeloma NS/0 cells by electroporation. NS/0 cells were grown in Dulbecco's modified Eagle's medium supplemented with 10% heat-inactivated fetal bovine serum, 2 mM glutamine and penicillin/streptomycin. About 5x106 cells 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, BioRad) on ice for 10 min. Electroporation was performed using a Gene Pulser (BioRad, Hercules, CA) with settings at 0.25 V and 500 μF. Cells were allowed to recover for 10 min on ice, after which they were resuspended in growth medium and plated onto two 96 well plates. Stably transfected clones were selected by their growth in the presence of 100 nM methotrexate (MTX), which was added to the growth medium two days post-transfection. The cells were fed every 3 days for two to three more times, and MTX-resistant clones appeared in 2 to 3 weeks. Supernatants from clones were assayed by anti-Fc ELISA to identify high producers. High producing clones were isolated and propagated in growth medium containing 100 nM MTX. The growth medium typically used was H-SFM or CD medium (Life Technologies). Alternatively, clones stably expressing huFc-IFN-βso1 fusion proteins were obtained in human embryonic kidney HEK 293 cells by methotrexate selection, by a method similar to the one described above. HEK 293 clones were maintained in DMEM supplemented with 10% FBS.
Example 3. Characterization of huFc-IFN-β fusion proteins from cell supernatants. The huFc-IFN-β fusion proteins were subsequently captured from the medium for further analysis. For routine characterization by gel electrophoresis, the huFc-IFN-β fusion proteins secreted into the medium was captured on Protein A Sepharose beads (Repligen, Cambridge, MA) and then eluted by boiling the sample in protein sample buffer, with or without a reducing agent such as β-mercaptoethanol. The samples were analyzed by SDS-PAGE and the protein bands were visualized by Coomassie staining. When huFc-IFN-β protein containing an immunoglobulin γ 4 isotype was analyzed by SDS-PAGE, it was found that the protein was not expressed in mammalian tissue culture cells as a uniform species. As shown in FIG. 2, under non-reducing conditions, in addition to a major band at 100 kDa which represented the huFc-IFN-β, multiple other bands were clearly visible, as well as an unresolved trail of higher molecular weight proteins. These results indicated that when expressed as an Fc fusion protein, the wildtype IFN-β formed aggregates. This finding was in contrast to what is generally found with unmodified IFN-β; when the wildtype sequence is cloned into an expression vector, and expressed and secreted in mammalian cell culture it is found to be 98% monomeric by size exclusion chromatography (Runkel et al, (1998), Pharmaceutical Research 15:641). This result was further unexpected in light of the fact that IFN-β can be produced as a fusion protein of the form IFN-β-Fc. See, for example, U.S. Patent No. 5,908,626. A portion of these aggregates was stable to reducing agents, as additional bands to the expected 50 kDa band for huFc-IFN-β persisted in a reducing SDS-PAGE system. However, the amount of material exhibiting abnormal migration was vastly diminished. This result suggested that to a significant extent the aggregation was due to scrambled disulfide bond formation. An Fc-IFN-β variant which contained a substitution of the hinge region with one derived from immunoglobulin γ 1 was analyzed. It was found that this substitution had no impact on the behavior of the fusion protein, although it did not contain four disulfide bonds like the immunoglobulin γ 4 hinge region. Similarly, using an Fc isotype derived from an immunoglobulin γ 1 in the fusion construct also had no effect. Thus, while the aggregation appeared to be due to the presence of the Fc moiety, the aggregation could not be alleviated by alterations in the Fc moiety. It has been reported that when EFN-β is fused to the N-terminal region of Fc, the introduction of a linker sequence is useful. See, for example, U.S. Patent No. 5,908,626. Similar to the Fc-IFN-β fusion proteins with either the altered hinge regions or altered Fc regions, an Fc- IFN-β fusion protein containing a Gly-Ser linker region, which separates the Fc region from the IFN-β moiety also yields the same result as above. hi contrast, SDS-PAGE analysis of huFc-IFN-β(C17S) revealed that this protein was substantially non-aggregated. Under non-reducing conditions, the band of 100 kDa corresponding to huFc-IFN-β (C17S) represented practically the only visible band on the gel. Moreover, under reducing conditions, the more prominent band representing aggregated fusion protein, most probably due to interaction of exposed hydrophobic patches, was also absent. Therefore, the introduction of a cysteine substitution at position 17 of the mature sequence of IFN-β promoted the correct folding of the fusion protein. This result was surprising on two counts: for one, the presence of a free cysteine in the "X" portion of an Fc-X protein had not presented a problem in other fusion proteins, such as Fc-IL2; and the presence of the free cysteine in EFN-β had not presented a problem either when the free protein or when an IFN-β-Fc protein were expressed in a mammalian expression system.
Example 4: ELISA Procedures. The concentration of human Fc-containing protein products in the supernatants of MTX-resistant clones and other test samples were determined by anti-huFc ELISA. Standard procedures as described in detail below were essentially followed. A. Coating plates ELISA plates were coated with AffmiPure Goat anti-Human IgG (H+L) (Jackson Immuno Research Laboratories, West Grove, PA) at 5 μg/mL in PBS, and 100 μL/well in 96-well plates (Nunc-Immuno plate Maxisorp). Coated plates were covered and incubated at 4°C overnight. Plates then were washed 4 times with 0.05% Tween (Tween 20) in PBS, and blocked with 1% BSA/1% goat serum in PBS, 200 μL/well. After incubation with the blocking buffer at 370C for 2 hrs, the plates were washed 4 times with 0.05% Tween in PBS and tapped dry on paper towels. B. Incubation with test samples and secondary antibody Test samples were diluted as appropriate in sample buffer (1% BSA/1% goat serum/0.05% Tween in PBS). A standard curve was prepared using a chimeric antibody (with a human Fc), the concentration of which was known. To prepare a standard curve, serial dilutions were made in the sample buffer to give a standard curve ranging from 125 ng/niL to 3.9 ng/niL. The diluted samples and standards were added to the plate, 100 μL/well and the plate incubated at 37°C for 2 hr. After incubation, the plate was washed 8 times with 0.05% Tween in PBS. To each well was then added 100 μL of the secondary antibody, the horseradish peroxidase-conjugated anti-human IgG (Jackson Immuno Research), diluted around 1:120,000 in the sample buffer. The exact dilution of the secondary antibody has to be determined for each lot of the HRP-conjugated anti- human IgG. After incubation at 370C for 2 hr, the plate was washed 8 times with 0.05% Tween in PBS. C. Development The substrate solution was added to the plate at 100 μL/well. The substrate solution was prepared by dissolving 30 mg of OPD (o-phenylenediamine dihydrochloride (OPD)3 (1 tablet) into 15 niL of 0.025 M Citric acid/0.05 M Na2HPO4 buffer, pH 5, which contained 0.03% of freshly added hydrogen peroxide. The color was allowed to develop for 30 min. at room temperature in the dark. The developing time is subject to change, depending on lot to lot variability of the coated plates, the secondary antibody, etc. The reaction was stopped by adding 4N sulfuric acid, 100 μL/well. The plate was read by a plate reader, which was set at both 490 and 650 nm and programmed to subtract the background OD at 650 nm from the OD at 490 nm.
Example 5. Purification and analysis of huFc-IFN-β proteins. A standard purification of Fc-containing fusion proteins was performed based on the affinity of the Fc protein moiety for Protein A. Briefly, cell supernatants (from cells transfected with wildtype or mutant proteins) containing the fusion protein were loaded onto a pre-equilibrated (50 mM Sodium Phosphate, 150 mM NaCl at neutral pH) Protein A Sepharose Fast Flow column and the column was washed extensively in buffer (50 mM Sodium Phosphate, 150 mM NaCl at neutral pH). Bound protein was eluted at a low pH (pH 2.5) in same buffer as above and fractions were immediately neutralized, optionally by eluting directly into a solution of IM Tris base, pH 11. The Protein A Sepharose - purified huFc-IFN-β and huFc-IFN-βS0' fusion proteins were analyzed by analytical size exclusion chromatography (SEC), and the % non- aggregated material was quantified by calculating the area under the curve of chromatogram peaks. The integrity and purity of the fusion proteins was verified by SDS-PAGE electrophoresis. Table 1: Analytical SEC analysis of Fc-IFN-β fusion proteins
Figure imgf000039_0001
hi a second purification step, neutralized Protein A Sepharose eluates containing Fc-IFN-βso1 fusion proteins were applied to a preparative SEC column and peak fractions were collected, yielding Fc-IFN-βso1 protein preparations consisting of at least 90% non- aggregated material. While the yield of purified product for Fc-γ4h-linker-IFN-β(C17S) was about 10%, for Fc-γ4h-linker-IFN-βsol(C17S L57A H131 A H140T) it was about 75%. This result indicated that the combination of mutations C17S with, for example L57A, H131 A, and H140T in the IFN-β moiety significantly promoted the solubility characteristics of the Fc-IFN-β fusion proteins.
Example 6. Measurement of Antiviral Activity. Viral replication in cell culture often results in cytotoxicity, an effect known as cytopathic effect (CPE). Interferons, can inhibit viral proliferation and protect cells from CPE. The antiviral activity of IFN-β can be quantitated by cytopathic effect reduction (CPER), as described in "Lymphokines and Interferons: A Practical Approach", edited by MJ. Clemens, A.G. Morris, and AJ.H. Gearin, I.R.L. Press, Oxford, 1987. The antiviral activities of purified huFc-IFN-β and huFc-IFN-βso1 were compared relative to a commercial huIFN-β standard (R&D Systems) or Betaferon (Serono) using the human epithelial lung carcinoma line A549 (ATCC # CCL-185) and the encephalomyocarditis virus (EMCV; ATCC # VR 129B) according to the CPER protocol described in the above reference. The effective dose (ED50) was set as the amount of protein that led to 50% CPER (i.e. 50% of the cells being protected from lysis), determined relative to uninfected control cells. The ED50 values were the average of at least three separate experiments. It was found that the effective doses that gave 50% CPER were 50 pg/ml for huFc-IFN-β 70 pg/ml for huFc-IFN-βsol(C17S), 14 pg/ml for huFc-IFN-βsol(C17S, F50H, H131A, H140A) and 17 pg/ml for huFc-IFN-βsol(C17S, L57A, H131A, H140T). These values, which had been normalized to the amount of IFN-β in the fusion protein, correlated well with the ED50 of 90 pg/ml or 40 pg/ml found with the commercial standard or Betaferon, respectively. Therefore, the IFN-β fusion proteins retained substantial anti- viral activity in a CPER assay, and the huFc-IFN-βso1 fusion proteins had an ED50 about equivalent to that of the free huIFN-β.
Example 7: Cellular Growth Inhibition Assay. The activity of the purified Fc-IFN-β fusion proteins was further determined in a cellular growth inhibition assay. The proliferation of Daudi cells (ATCC # CCL-123), a B lymphoblast line derived from a patient with Burkitt's lymphoma, is normally inhibited by IFN-β. Accordingly, the antiproliferative effects of fusion proteins huFc- IFN-β and huFc-IFN-βsol(C17S) on Daudi cells were compared relative to a commercial human standard (Calbiochem). To set up the assay for each of these proteins, a dilution series covering about a thousand fold concentration range was prepared in RPMI medium supplemented with 10 % fetal bovine serum, and 100 μl samples were aliquoted in wells of a 96 well plate. Daudi cells in growth phase were washed and resuspended at 2 x 105 cells/ml in the RPMI medium supplemented with 10 % fetal bovine serum, and 100 μl of the cells were aliquoted to each well containing the IFN-β dilutions. Further control wells contained either untreated cells or medium alone. After incubation for an additional 72 hours proliferation was measured by mitochondrial dehydrogenase activity, using the chromogenic enzyme substrate MTS (Promega # G5421) in the presence of the electron donor PMS (Sigma # P 5812). The ED50 values, determined from activity curves, were found to be around 3 ng/ml to 3.5 ng/ml for each of the fusion proteins as well as for the commercial IFN-β protein. It was therefore concluded that the IFN-β fusion proteins were as effective as the free IFN-β in inhibiting Daudi cell growth.
Example 8. Pharmacokinetics of huFc-IFN-β proteins. The pharmacokinetics of huFc-IFN-β and huFc-IFN-βso! fusion proteins are determined in a group of 4 Balb/c mice, for each protein. Twenty-five milligrams of the fusion protein is injected into the tail vein of each mouse. Blood is obtained by retro- orbital bleeding immediately after injection (i.e., at t = 0 min), and at 30 min, 1 hr, 2 hrs, 4 hrs, 8 hrs, and 24 hrs post-injection. Blood samples are collected in tubes containing heparin to prevent clotting. Cells are removed by centrifugation in an Eppendorf high¬ speed microcentrifuge for 4 min at 12,500 g. The concentration of either Fc-huIFN-β or huFc-IFN-βso1 in the plasma is measured by anti-huFc ELISA and Western blot analysis using anti-huFc antibody. Alternatively, an IFN-β ELISA maybe used. The integrity of the circulating fusion protein is ascertained by an immunoblot of the serum probed with an anti-huFc antibody or with an anti-IFN-β antibody. It is found that the circulating half- life of huFc-IFN-βso1 is greater than that of huFc-IFN-β, and at least 5-fold that of the free IFN-β. Furthermore, it is contemplated that the specific effects of Fc-IFN-βso1 are more pronounced in treatment of conditions and diseases such as multiple sclerosis, where administration of IFN-β is known to alleviate the condition.

Claims

PATENT CLAIMS;
1. An Fc-interferon-β fusion protein comprising: (i) an immunoglobulin Fc region; and (ii) an interferon-β protein linked by peptide bond or a peptide linker sequence to the carboxy-terminus of the immunoglobulin Fc region, wherein the interferon-β protein comprises an amino acid alteration at at least one of positions 17, 50, 57, 130, 131, 136 and 140 corresponding to the native mature interferon-β.
2. The fusion protein of claim 1, wherein the amino acid alteration is a substitution.
3. The fusion protein of claim 2, having amino acid substitutions within the interferon-β protein which are selected from the group consisting of: C17S, C17A, C17V, C17M; F50H, L57A, Ll 30A5 H131A, K136A, H140A, H140T.
4. A fusion protein according to claim 2, having amino acid substitutions within the interferon-β protein at positions 17 and 50 and 131 and 140, or 17 and 57 and 131 and 140.
5. A fusion protein of claim 4, having the following amino acid substitutions within the interferon-β protein: C17S and F50H and H131A and H140T or A, or C17S and L57A and H131 A and H140T or A.
6. A fusion protein according to any of the claims 1 - 5, wherein the immunoglobulin Fc region comprises an immunoglobulin hinge region and an immunoglobulin heavy chain constant region.
7. A fusion protein according to any of the claims 1 - 6, wherein the immunoglobulin Fc region is derived from IgG4, IgG2 or IgGl.
8. A fusion protein of claim 7, wherein the immunoglobulin Fc region is derived from IgG4 and further comprises an immunoglobulin hinge region derived from IgGl.
9. A fusion protein of claim 8, wherein a cysteine residue of the hinge region has been mutated.
10. The fusion protein of claim 7, wherein the immunoglobulin Fc region is derived from IgGl, and an alanine residue is substituted in place of the C-terminal lysine of the immunoglobulin Fc region.
11. A fusion protein according to claim 7, wherein the immunoglobulin Fc region is derived from IgG2 and further comprises an immunoglobulin hinge region derived from IgGl.
12. A fusion protein of claim 11, wherein a cysteine residue of the hinge region has been mutated.
13. A fusion protein of claim 11 or 12, wherein further an alanine residue is substituted in place of the C-terminal lysine of the immunoglobulin Fc region.
14. A fusion protein according to any of the claims 1 - 13, wherein the peptide linker sequence is Gly4SerGly4SerGly3SerGly.
15. A nucleic acid molecule encoding any one of the Fc-interferon-β fusion proteins of claims 1-14.
16. A replicable expression vector for transfecting a mammalian cell, the vector comprising a nucleic acid molecule encoding any one of the Fc-interferon-β fusion proteins of claims 1-14.
17. A cell containing a nucleic acid molecule of claim 15 or 16.
18. A method of stabilizing an Fc-interferon-β fusion protein comprising the step of making an Fc-interferon-β fusion protein, wherein the Fc-interferon-β fusion comprises any one Fc-interferon-β fusion protein of claims 1-14.
19. The method of claim 18, wherein stabilizing comprises (i) increasing the circulating half-life of the Fc-interferon-β fusion protein relative to an unaltered Fc-interferon-β fusion protein, or (ii) decreasing the aggregation of the Fc-interferon-β fusion protein relative to an unaltered Fc-interferon-β fusion protein, or (iii) increasing the biological activity of the Fc-interferon-β fusion protein relative to an unaltered Fc-interferon-β fusion protein.
20. Use of a Fc-interferon-β fusion protein of any of the claims 1 - 14 for the manufacture of a medicament for the treatment of a condition of a patient, which is alleviated by the administration of interferon-β.
PCT/EP2005/006925 2004-06-28 2005-06-28 Fc-INTERFERON-BETA FUSION PROTEINS WO2006000448A2 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
AU2005256519A AU2005256519B2 (en) 2004-06-28 2005-06-28 Fc-interferon-beta fusion proteins
JP2007517216A JP4808709B2 (en) 2004-06-28 2005-06-28 Fc-interferon-beta fusion protein
EP05766761A EP1761559B1 (en) 2004-06-28 2005-06-28 Fc-INTERFERON-BETA FUSION PROTEINS
AT05766761T ATE443720T1 (en) 2004-06-28 2005-06-28 FC-INTERFERON-BETA FUSION PROTEINS
CA2572015A CA2572015C (en) 2004-06-28 2005-06-28 Fc-interferon-beta fusion proteins
DE602005016799T DE602005016799D1 (en) 2004-06-28 2005-06-28 FC INTERFERON BETA FUSION PROTEINS

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US58338904P 2004-06-28 2004-06-28
US60/583,389 2004-06-28

Publications (2)

Publication Number Publication Date
WO2006000448A2 true WO2006000448A2 (en) 2006-01-05
WO2006000448A3 WO2006000448A3 (en) 2006-03-02

Family

ID=35429166

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2005/006925 WO2006000448A2 (en) 2004-06-28 2005-06-28 Fc-INTERFERON-BETA FUSION PROTEINS

Country Status (9)

Country Link
US (3) US7670595B2 (en)
EP (1) EP1761559B1 (en)
JP (1) JP4808709B2 (en)
AT (1) ATE443720T1 (en)
AU (1) AU2005256519B2 (en)
CA (1) CA2572015C (en)
DE (1) DE602005016799D1 (en)
ES (1) ES2332924T3 (en)
WO (1) WO2006000448A2 (en)

Cited By (70)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006127757A2 (en) * 2005-05-26 2006-11-30 Schering Corporation Interferon-igg fusion
WO2008077616A1 (en) 2006-12-22 2008-07-03 Csl Behring Gmbh Modified coagulation factors with prolonged in vivo half-life
WO2009053368A1 (en) * 2007-10-22 2009-04-30 Merck Serono S.A. Single ifn-beta fused to a mutated igg fc fragment
JP2010529835A (en) * 2007-05-25 2010-09-02 ビオマリン プハルマセウトイカル インコーポレイテッド Prokaryotic phenylalanine ammonia lyase composition and method of using the composition
WO2010121766A1 (en) * 2009-04-22 2010-10-28 Merck Patent Gmbh Antibody fusion proteins with modified fcrn binding sites
WO2011098644A2 (en) 2010-02-09 2011-08-18 Proyecto De Biomedicina Cima, S.L. Compositions for the treatment of infectious and tumoural diseases
EP2371857A1 (en) 2010-04-01 2011-10-05 CSL Behring GmbH Factor XII inhibitors for treating interstitial lung disease
US8114630B2 (en) 2007-05-02 2012-02-14 Ambrx, Inc. Modified interferon beta polypeptides and their uses
CN102585013A (en) * 2011-01-07 2012-07-18 中国人民解放军军事医学科学院生物工程研究所 Fusion protein containing omega interferon and method for preparing same
EP2497489A1 (en) 2011-03-09 2012-09-12 CSL Behring GmbH Factor XII inhibitors for the treatment of silent brain ischemia and ischemia of other organs
WO2012120128A1 (en) 2011-03-09 2012-09-13 Csl Behring Gmbh Factor xii inhibitors for the administration with medical procedures comprising contact with artificial surfaces
US8283319B2 (en) 2007-02-12 2012-10-09 Csl Behring Gmbh Therapeutic application of Kazal-type serine protease inhibitors
EP2572734A3 (en) * 2006-10-31 2013-06-19 East Carolina University Cytokine-based fusion proteins for treatment of immune disorders
WO2013119903A1 (en) 2012-02-10 2013-08-15 Research Corporation Technologies, Inc. Fusion proteins comprising immunoglobulin constant domain-derived scaffolds
WO2013120939A1 (en) 2012-02-15 2013-08-22 Csl Behring Gmbh Von willebrand factor variants having improved factor viii binding affinity
US8575104B2 (en) 2008-06-24 2013-11-05 Csl Behring Gmbh Factor VIII, von willebrand factor or complexes thereof with prolonged in vivo half-life
CN103570836A (en) * 2013-10-25 2014-02-12 江苏众红生物工程创药研究院有限公司 Recombinant porcine interferon beta1-Fc fusion protein as well as encoding gene and expressing method thereof
US8765915B2 (en) 2006-02-06 2014-07-01 Csl Behring Gmbh Modified coagulation factor VIIa with extended half-life
WO2014152940A1 (en) * 2013-03-14 2014-09-25 Shire Human Genetic Therapies, Inc. Mrna therapeutic compositions and use to treat diseases and disorders
US8945528B2 (en) 2010-08-31 2015-02-03 Generon (Shanghai) Corporation Ltd. Use of interleukin-22 in treating viral hepatitis
WO2015017710A1 (en) * 2013-07-31 2015-02-05 Amgen Inc. Growth differentiation factor 15 (gdf-15) constructs
US9181321B2 (en) 2013-03-14 2015-11-10 Shire Human Genetic Therapies, Inc. CFTR mRNA compositions and related methods and uses
WO2016000039A1 (en) 2014-07-02 2016-01-07 Csl Limited Modified von willebrand factor
US9308281B2 (en) 2011-06-08 2016-04-12 Shire Human Genetic Therapies, Inc. MRNA therapy for Fabry disease
US9352024B2 (en) 2011-12-27 2016-05-31 Generon (Shanghai) Corporation Ltd. Uses of interleukin-22(IL-22) in treating and preventing nerve damage diseases or neurodegenerative diseases
WO2016188905A1 (en) 2015-05-22 2016-12-01 Csl Behring Recombinant Facility Ag Methods for preparing modified von willebrand factor
WO2016188907A1 (en) 2015-05-22 2016-12-01 Csl Behring Recombinant Facility Ag Truncated von willebrand factor polypeptides for treating hemophilia
US9522176B2 (en) 2013-10-22 2016-12-20 Shire Human Genetic Therapies, Inc. MRNA therapy for phenylketonuria
US9550819B2 (en) 2012-03-27 2017-01-24 Ngm Biopharmaceuticals, Inc. Compositions and methods of use for treating metabolic disorders
JP2017031215A (en) * 2007-09-21 2017-02-09 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア Interferon-antibody fusion proteins demonstrating potent apoptotic and anti-tumor activities
EP3184149A1 (en) 2015-12-23 2017-06-28 Julius-Maximilians-Universität Würzburg Soluble glycoprotein v for treating thrombotic diseases
WO2017117631A1 (en) 2016-01-07 2017-07-13 Csl Limited Mutated truncated von willebrand factor
US9828415B2 (en) 2013-01-30 2017-11-28 Ngm Biopharmaceuticals, Inc. Compositions and methods of use for treating metabolic disorders
US9827291B2 (en) 2013-01-30 2017-11-28 Ngm Biopharmaceuticals, Inc. Compositions and methods of use for treating metabolic disorders
US9834586B2 (en) 2014-07-30 2017-12-05 Ngm Biopharmaceuticals, Inc. Compositions and methods of use for treating metabolic disorders
US9850269B2 (en) 2014-04-25 2017-12-26 Translate Bio, Inc. Methods for purification of messenger RNA
US9878017B2 (en) 2013-04-22 2018-01-30 Csl Ltd. Covalent complex of von Willebrand Factor and factor VIII, compositions, and uses relating thereto
US9920118B2 (en) 2014-10-31 2018-03-20 Ngm Biopharmaceuticals, Inc. Compositions and methods of use for treating metabolic disorders
EP3127923A4 (en) * 2014-03-31 2018-04-18 Hanmi Pharm. Co., Ltd. Method for improving solubility of protein and peptide by using immunoglobulin fc fragment linkage
US9957499B2 (en) 2013-03-14 2018-05-01 Translate Bio, Inc. Methods for purification of messenger RNA
WO2018087271A1 (en) 2016-11-11 2018-05-17 Csl Behring Recombinant Facility Ag Truncated von willebrand factor polypeptides for extravascular administration in the treatment or prophylaxis of a blood coagulation disorder
WO2018087267A1 (en) 2016-11-11 2018-05-17 Csl Behring Recombinant Facility Ag Truncated von willebrand factor polypeptides for treating hemophilia
EP3266801A4 (en) * 2015-03-03 2018-09-12 Abion Inc. Human interferon-beta variant conjugated immunocytokine and method for preparing same
US10093745B2 (en) 2013-05-29 2018-10-09 The Regents Of The University Of California Anti-CSPG4 fusions with interferon for the treatment of malignancy
WO2018234518A1 (en) 2017-06-22 2018-12-27 CSL Behring Lengnau AG Modulation of fviii immunogenicity by truncated vwf
US10286047B2 (en) 2013-03-08 2019-05-14 Csl Behring Gmbh Treatment and prevention of remote ischemia-reperfusion injury
US20190185545A1 (en) * 2016-08-15 2019-06-20 Children's Medical Center Corporation Apom-fc fusion proteins, complexes thereof with sphingosine 1-phosphate (s1p), and methods for treating vascular and non-vascular diseases
US10336798B2 (en) 2012-01-26 2019-07-02 Amgen Inc. Growth differentiation factor 15 (GDF-15) polypeptides
US10363306B2 (en) 2009-03-31 2019-07-30 East Carolina University Cytokines and neuroantigens for treatment of immune disorders
US10543169B2 (en) 2013-11-07 2020-01-28 Generon (Shanghai) Corporation Ltd. Use of IL-22 dimer in manufacture of a medicament for intravenous administration
US10576166B2 (en) 2009-12-01 2020-03-03 Translate Bio, Inc. Liver specific delivery of messenger RNA
US10752664B2 (en) 2011-04-08 2020-08-25 Amgen Inc. Method of treating or ameliorating metabolic disorders using growth differentiation factor 15 (GDF-15)
US10786551B2 (en) 2007-08-06 2020-09-29 Generon (Shanghai) Corporation Ltd. Use of interleukin-22 in the treatment of fatty liver disease
US10808023B2 (en) 2016-01-07 2020-10-20 CSL Behring Lengnau AG Mutated von Willebrand factor
WO2020234195A1 (en) 2019-05-17 2020-11-26 Universitaet Zuerich Haptoglobin for use in treating an adverse secondary neurological outcome following a haemorrhagic stroke
WO2021001522A1 (en) 2019-07-04 2021-01-07 CSL Behring Lengnau AG A truncated von willebrand factor (vwf) for increasing the in vitro stability of coagulation factor viii
US10975158B2 (en) 2012-12-07 2021-04-13 The Regents Of The University Of California CD138-targeted interferon demonstrates potent apoptotic and anti-tumor activities
WO2021094344A1 (en) 2019-11-11 2021-05-20 CSL Behring Lengnau AG Polypeptides for inducing tolerance to factor viii
US11161889B2 (en) 2018-04-09 2021-11-02 Amgen Inc. Growth differentiation factor 15 fusion proteins
US11168109B2 (en) 2012-03-08 2021-11-09 Hanmi Science Co., Ltd. Process for preparation of physiologically active polypeptide complex
US11173190B2 (en) 2017-05-16 2021-11-16 Translate Bio, Inc. Treatment of cystic fibrosis by delivery of codon-optimized mRNA encoding CFTR
US11174500B2 (en) 2018-08-24 2021-11-16 Translate Bio, Inc. Methods for purification of messenger RNA
US11224642B2 (en) 2013-10-22 2022-01-18 Translate Bio, Inc. MRNA therapy for argininosuccinate synthetase deficiency
US11253605B2 (en) 2017-02-27 2022-02-22 Translate Bio, Inc. Codon-optimized CFTR MRNA
US11254936B2 (en) 2012-06-08 2022-02-22 Translate Bio, Inc. Nuclease resistant polynucleotides and uses thereof
WO2022162218A1 (en) 2021-02-01 2022-08-04 Csl Behring Ag Method of treating or preventing an adverse secondary neurological outcome following a haemorrhagic stroke
US11434271B2 (en) 2011-11-04 2022-09-06 Hanmi Science Co., Ltd. Method for preparing physiologically active polypeptide complex
WO2022234070A1 (en) 2021-05-07 2022-11-10 Csl Behring Ag Expression system for producing a recombinant haptoglobin (hp) beta chain
US11510966B2 (en) 2016-04-15 2022-11-29 Evive Biotechnology (Shanghai) Ltd Use of IL-22 in treating necrotizing enterocolitis
WO2024047219A1 (en) 2022-09-02 2024-03-07 Csl Behring Ag Haptoglobin for use in treating or preventing exaggerated erectile response or erectile dysfunction

Families Citing this family (47)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2399832C (en) 2000-02-11 2011-09-20 Stephen D. Gillies Enhancing the circulating half-life of antibody-based fusion proteins
CA2440221C (en) 2001-03-07 2013-02-05 Merck Patent Gesellschaft Mit Beschraenkter Haftung 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
BR0209177A (en) 2001-05-03 2004-10-05 Merck Patent Gmbh Recombinant tumor specific antibody and use
ES2381025T3 (en) 2001-12-04 2012-05-22 Merck Patent Gmbh Immunocytokines with modulated selectivity
WO2005063820A2 (en) * 2003-12-30 2005-07-14 Merck Patent Gmbh Il-7 fusion proteins
US7670595B2 (en) * 2004-06-28 2010-03-02 Merck Patent Gmbh Fc-interferon-beta fusion proteins
DE602005020837D1 (en) * 2004-12-09 2010-06-02 Merck Patent Gmbh IL-7 VARIANTS WITH REDUCED IMMUNOGENITY
US7566456B2 (en) * 2005-06-23 2009-07-28 Haiming Chen Allergen vaccine proteins for the treatment and prevention of allergic diseases
CA2635618C (en) * 2005-12-30 2015-10-06 Merck Patent Gesellschaft Mit Beschraekter Haftung Interleukin-12p40 variants with improved stability
PT2270050E (en) 2005-12-30 2013-09-12 Merck Patent Gmbh Anti-cd19 antibodies with reduced immunogenicity
FR2907454B1 (en) * 2006-10-18 2009-01-23 Biomethodes Sa IMPROVED VARIANTS OF INTERFERON BETA HUMAIN
AR080993A1 (en) * 2010-04-02 2012-05-30 Hanmi Holdings Co Ltd FORMULATION OF PROLONGED BETA INTERFERTION ACTION WHERE AN IMMUNOGLOBULIN FRAGMENT IS USED
WO2012065059A2 (en) * 2010-11-12 2012-05-18 The General Hospital Corporation Treatment of atherosclerosis with bmp-alk3 antagonists
CN104203982B (en) 2011-10-28 2018-08-31 特瓦制药澳大利亚私人有限公司 Polypeptide construct and application thereof
US8936782B2 (en) * 2011-11-22 2015-01-20 Cedars-Sinai Medical Center Interferon beta as antibacterial agents
WO2013103981A1 (en) * 2012-01-06 2013-07-11 Ember Therapeutics, Inc. Compositions and methods of use of fndc5 polypeptides
AU2013359907B2 (en) 2012-12-11 2018-01-18 Albert Einstein College Of Medicine, Inc. Methods for high throughput receptor:ligand identification
WO2014124217A1 (en) * 2013-02-07 2014-08-14 Albert Einstein College Of Medicine Of Yeshiva University A selective high-affinity immune stimulatory reagent and uses thereof
US20150366944A1 (en) 2013-02-20 2015-12-24 Yeda Research And Development Co. Ltd. Interferon treatment targeting mutant p53 expressing cells
WO2014159813A1 (en) * 2013-03-13 2014-10-02 Moderna Therapeutics, Inc. Long-lived polynucleotide molecules
US11117975B2 (en) 2013-04-29 2021-09-14 Teva Pharmaceuticals Australia Pty Ltd Anti-CD38 antibodies and fusions to attenuated interferon alpha-2B
SG11201508923VA (en) 2013-04-29 2015-11-27 Teva Pharmaceuticals Australia Pty Ltd Anti-cd38 antibodies and fusions to attenuated interferon alpha-2b
DK3105246T3 (en) 2014-02-10 2021-06-14 Merck Patent Gmbh Targeted TGF-beta inhibition
US9738702B2 (en) 2014-03-14 2017-08-22 Janssen Biotech, Inc. Antibodies with improved half-life in ferrets
UA119352C2 (en) 2014-05-01 2019-06-10 Тева Фармасьютикалз Острейліа Пті Лтд Combination of lenalidomide or pomalidomide and cd38 antibody-attenuated interferon-alpha constructs, and the use thereof
PL3157552T3 (en) * 2014-06-18 2020-05-18 Albert Einstein College Of Medicine Syntac polypeptides and uses thereof
WO2016044676A2 (en) * 2014-09-18 2016-03-24 AskGene Pharma, Inc. Novel feline erythropoietin receptor agonists
KR102639037B1 (en) 2014-10-29 2024-02-20 테바 파마슈티컬즈 오스트레일리아 피티와이 엘티디 INTERFERON α2b VARIANTS
CN104530213A (en) * 2014-11-24 2015-04-22 广西大学 Viral hepatitis treatment drug interferon production technology
US11820807B2 (en) 2015-06-12 2023-11-21 Ubi Pharma Inc Immunoglobulin fusion proteins and uses thereof
AU2016335912B2 (en) * 2015-10-05 2023-08-31 Circle33 Llc Antibodies with improved stability to intestinal digestion
IL262606B2 (en) 2016-05-18 2023-04-01 Albert Einstein College Medicine Inc Variant pd-l1 polypeptides, t-cell modulatory multimeric polypeptides, and methods of use thereof
BR112018073606A2 (en) 2016-05-18 2019-02-26 Cue Biopharma, Inc. T-cell modulating multimeric polypeptides and methods of using them
US11129906B1 (en) 2016-12-07 2021-09-28 David Gordon Bermudes Chimeric protein toxins for expression by therapeutic bacteria
IL297617B2 (en) 2016-12-22 2023-11-01 Cue Biopharma Inc T-cell modulatory multimeric polypeptides and methods of use thereof
WO2018129474A1 (en) 2017-01-09 2018-07-12 Cue Biopharma, Inc. T-cell modulatory multimeric polypeptides and methods of use thereof
SG11201908391XA (en) 2017-03-15 2019-10-30 Cue Biopharma Inc Methods for modulating an immune response
EP3604538A4 (en) 2017-03-29 2020-12-30 University of Miyazaki Long-acting adrenomedullin derivative
EP3624820A1 (en) * 2017-04-21 2020-03-25 H. Hoffnabb-La Roche Ag Use of klk5 antagonists for treatment of a disease
CN111886241A (en) 2018-01-09 2020-11-03 库尔生物制药有限公司 Multimeric T cell modulating polypeptides and methods of use thereof
WO2021110071A1 (en) 2019-12-02 2021-06-10 The Hong Kong Polytechnic University Methods for inducing intermittent fasting and modulating autophagy
CN115989239A (en) * 2020-04-29 2023-04-18 瑷备恩有限公司 Human interferon beta variants with double mutations and methods for improving the stability of human interferon beta variants
WO2021221470A1 (en) * 2020-04-29 2021-11-04 (주) 제노팜 Recombinant protein having fused interferon-beta mutein and antibody, and pharmaceutical composition comprising same
MX2022013208A (en) 2020-05-12 2022-11-14 Cue Biopharma Inc Multimeric t-cell modulatory polypeptides and methods of use thereof.
WO2022177018A1 (en) * 2021-02-22 2022-08-25 国立大学法人宮崎大学 Method for producing long-acting adrenomedullin derivative
WO2023039242A2 (en) * 2021-09-13 2023-03-16 Achelois Biopharma, Inc. Multivalent interferon particles compositions and methods of use

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4737462A (en) * 1982-10-19 1988-04-12 Cetus Corporation Structural genes, plasmids and transformed cells for producing cysteine depleted muteins of interferon-β
WO2000023472A2 (en) * 1998-10-16 2000-04-27 Biogen, Inc. Interferon-beta fusion proteins and uses
WO2000069913A1 (en) * 1999-05-19 2000-11-23 Lexigen Pharmaceuticals Corp. EXPRESSION AND EXPORT OF INTERFERON-ALPHA PROTEINS AS Fc FUSION PROTEINS
WO2001003737A1 (en) * 1999-07-13 2001-01-18 Bolder Biotechnology Inc. Immunoglobulin fusion proteins
WO2002074783A2 (en) * 2001-03-15 2002-09-26 Merck Patent Gmbh Modified interferon beta with reduced immunogenicity

Family Cites Families (89)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4469797A (en) 1982-09-23 1984-09-04 Miles Laboratories, Inc. Digoxigenin immunogens, antibodies, labeled conjugates, and related derivatives
US4966843A (en) 1982-11-01 1990-10-30 Cetus Corporation Expression of interferon genes in Chinese hamster ovary cells
US5679543A (en) 1985-08-29 1997-10-21 Genencor International, Inc. DNA sequences, vectors and fusion polypeptides to increase secretion of desired polypeptides from filamentous fungi
EP0237019A3 (en) 1986-03-14 1988-03-09 Toray Industries, Inc. Interferon conjugate and production thereof using recombinant gene
US5019368A (en) 1989-02-23 1991-05-28 Cancer Biologics, Inc. Detection of necrotic malignant tissue and associated therapy
DE3856559T2 (en) 1987-05-21 2004-04-29 Micromet Ag Multifunctional proteins with predetermined objectives
ATE122238T1 (en) 1987-06-10 1995-05-15 Dana Farber Cancer Inst Inc BIFUNCTIONAL ANTIBODY CONSTRUCTIONS AND METHOD FOR SELECTIVE KILLING OF CELLS.
US5064646A (en) 1988-08-02 1991-11-12 The University Of Maryland Novel infectious bursal disease virus
ES2054753T3 (en) 1987-09-02 1994-08-16 Ciba Geigy Ag CONJUGATES OF CYTOKINES WITH IMMUNOGLOBULINS.
EP0308936B1 (en) 1987-09-23 1994-07-06 Bristol-Myers Squibb Company Antibody heteroconjugates for the killing of HIV-infected cells
NZ226414A (en) 1987-10-02 1992-07-28 Genentech Inc Cd4 peptide adhesion variants and their preparation and use
WO1989006692A1 (en) 1988-01-12 1989-07-27 Genentech, Inc. Method of treating tumor cells by inhibiting growth factor receptor function
CA1341588C (en) 1988-01-26 2009-01-06 Michel Revel Human ifn-beta2/i1-6, its purification and use
US5120525A (en) 1988-03-29 1992-06-09 Immunomedics, Inc. Radiolabeled antibody cytotoxic therapy of cancer
IE62463B1 (en) 1988-07-07 1995-02-08 Res Dev Foundation Immunoconjugates for cancer diagnosis and therapy
US5601819A (en) 1988-08-11 1997-02-11 The General Hospital Corporation Bispecific antibodies for selective immune regulation and for selective immune cell binding
US5242824A (en) 1988-12-22 1993-09-07 Oncogen Monoclonal antibody to human carcinomas
US6750329B1 (en) 1989-05-05 2004-06-15 Research Development Foundation Antibody delivery system for biological response modifiers
ZA902949B (en) 1989-05-05 1992-02-26 Res Dev Foundation A novel antibody delivery system for biological response modifiers
ATE123065T1 (en) 1989-07-07 1995-06-15 Takeda Chemical Industries Ltd PROTEINS AND THEIR PRODUCTION.
US5314995A (en) 1990-01-22 1994-05-24 Oncogen Therapeutic interleukin-2-antibody based fusion proteins
US5349053A (en) 1990-06-01 1994-09-20 Protein Design Labs, Inc. Chimeric ligand/immunoglobulin molecules and their uses
US7253264B1 (en) 1990-06-28 2007-08-07 Sanofi-Arentideutschland GmbH Immunoglobulin fusion proteins, their production and use
US5650150A (en) 1990-11-09 1997-07-22 Gillies; Stephen D. Recombinant antibody cytokine fusion proteins
US5709859A (en) 1991-01-24 1998-01-20 Bristol-Myers Squibb Company Mixed specificity fusion proteins
US6072039A (en) 1991-04-19 2000-06-06 Rohm And Haas Company Hybrid polypeptide comparing a biotinylated avidin binding polypeptide fused to a polypeptide of interest
ATE173763T1 (en) 1991-08-30 1998-12-15 Hutchinson Fred Cancer Res HYBRID CYTOKINE
US20020037558A1 (en) 1991-10-23 2002-03-28 Kin-Ming Lo E.coli produced immunoglobulin constructs
US6627615B1 (en) 1991-12-17 2003-09-30 The Regents Of The University Of California Methods and compositions for in vivo gene therapy
DK0615451T3 (en) 1992-05-26 2006-04-24 Immunex Corp Hitherto unknown cytokine that binds to CD30
CA2134773A1 (en) 1992-06-04 1993-12-09 Robert J. Debs Methods and compositions for in vivo gene therapy
US5614184A (en) 1992-07-28 1997-03-25 New England Deaconess Hospital Recombinant human erythropoietin mutants and therapeutic methods employing them
CA2142007C (en) 1992-08-11 2007-10-30 Robert Glen Urban Immunomodulatory peptides
DE4228839A1 (en) 1992-08-29 1994-03-03 Behringwerke Ag Methods for the detection and determination of mediators
US5554512A (en) 1993-05-24 1996-09-10 Immunex Corporation Ligands for flt3 receptors
US5541087A (en) 1994-09-14 1996-07-30 Fuji Immunopharmaceuticals Corporation Expression and export technology of proteins as immunofusins
PT706799E (en) 1994-09-16 2002-05-31 Merck Patent Gmbh IMUNOCONJUGADOS II
US6485726B1 (en) 1995-01-17 2002-11-26 The Brigham And Women's Hospital, Inc. Receptor specific transepithelial transport of therapeutics
US6086875A (en) 1995-01-17 2000-07-11 The Brigham And Women's Hospital, Inc. Receptor specific transepithelial transport of immunogens
ES2225874T3 (en) 1995-03-10 2005-03-16 Genentech, Inc. RECEIVER ACTIVATION THROUGH GAS.
US6620413B1 (en) 1995-12-27 2003-09-16 Genentech, Inc. OB protein-polymer chimeras
US5723125A (en) 1995-12-28 1998-03-03 Tanox Biosystems, Inc. Hybrid with interferon-alpha and an immunoglobulin Fc linked through a non-immunogenic peptide
US6750334B1 (en) 1996-02-02 2004-06-15 Repligen Corporation CTLA4-immunoglobulin fusion proteins having modified effector functions and uses therefor
ATE218143T1 (en) 1996-09-03 2002-06-15 Gsf Forschungszentrum Umwelt USE OF BI- AND TRISPECIFIC ANTIBODIES TO INDUCE TUMOR IMMUNITY
US6100387A (en) 1997-02-28 2000-08-08 Genetics Institute, Inc. Chimeric polypeptides containing chemokine domains
ATE267215T1 (en) 1997-12-08 2004-06-15 Lexigen Pharm Corp HETERODIMARY FUSION PROTEINS FOR USE FOR TARGETED IMMUNTHERAPY AND GENERAL IMMUNE EXCITATION
US20030105294A1 (en) 1998-02-25 2003-06-05 Stephen Gillies Enhancing the circulating half life of antibody-based fusion proteins
PL343462A1 (en) 1998-04-15 2001-08-13 Lexigen Pharm Corp Enhancement of antibody-cytokine fusion protein mediated immune responses by co-administration with angiogenesis inhibitor
EP1071469A2 (en) 1998-04-17 2001-01-31 Lexigen Pharmaceuticals Corp. Enhancement of antibody-cytokine fusion protein mediated immune responses by co-administration with prostaglandin inhibitor
WO1999058662A1 (en) 1998-05-14 1999-11-18 Merck Patent Gmbh Fused protein
US20020142374A1 (en) 1998-08-17 2002-10-03 Michael Gallo Generation of modified molecules with increased serum half-lives
CA2339331C (en) 1998-08-25 2011-03-01 Lexigen Pharmaceuticals Corporation Expression and export of angiogenesis inhibitors as immunofusins
US6646113B1 (en) 1998-09-17 2003-11-11 The Trustees Of The University Of Pennsylvania Nucleic acid molecule encoding human survival of motor neuron-interacting protein 1 (SIP1) deletion mutants
US6335176B1 (en) 1998-10-16 2002-01-01 Pharmacopeia, Inc. Incorporation of phosphorylation sites
KR20020007287A (en) 1999-01-07 2002-01-26 추후보정 EXPRESSION AND EXPORT OF ANTI-OBESITY PROTEINS AS Fc FUSION PROTEINS
KR100922809B1 (en) 1999-05-06 2009-10-21 웨이크 포리스트 유니버시티 Compositions And Methods For Identifying Antigens Which Elicit An Immune Response
SK782002A3 (en) 1999-07-21 2003-08-05 Lexigen Pharm Corp FC fusion proteins for enhancing the immunogenicity of protein and peptide antigens
US7067110B1 (en) 1999-07-21 2006-06-27 Emd Lexigen Research Center Corp. Fc fusion proteins for enhancing the immunogenicity of protein and peptide antigens
WO2001010912A1 (en) 1999-08-09 2001-02-15 Lexigen Pharmaceuticals Corp. Multiple cytokine-antibody complexes
US20050202538A1 (en) 1999-11-12 2005-09-15 Merck Patent Gmbh Fc-erythropoietin fusion protein with improved pharmacokinetics
WO2001036489A2 (en) 1999-11-12 2001-05-25 Merck Patent Gmbh Erythropoietin forms with improved properties
CA2399832C (en) 2000-02-11 2011-09-20 Stephen D. Gillies Enhancing the circulating half-life of antibody-based fusion proteins
PT1257297E (en) 2000-02-24 2006-12-29 Philogen Spa Compositions and methods for treatemnt of angiogenesis in pathological lesions
MXPA02011016A (en) 2000-05-12 2004-03-16 Neose Technologies Inc In vitro.
ES2288967T3 (en) 2000-06-29 2008-02-01 Merck Patent Gmbh REINFORCEMENT OF IMMUNE ANSWERS MEDIATED BY THE ANTIBODY-CYTOKIN FUSION PROTEIN THROUGH THE TREATMENT COMBINED BY AGENTS THAT IMPROVE THE INCORPORATION OF IMMUNOCITOQUINE.
JP2004525621A (en) 2001-01-18 2004-08-26 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフトング Bifunctional fusion protein having glucocerebrosidase activity
WO2002069232A2 (en) 2001-02-19 2002-09-06 Merck Patent Gmbh Method for identification of t-cell epitopes and use for preparing molecules with reeduced immunogenicity
JP2004532620A (en) 2001-02-19 2004-10-28 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフトング Artificial proteins with reduced immunogenicity
CA2440221C (en) 2001-03-07 2013-02-05 Merck Patent Gesellschaft Mit Beschraenkter Haftung 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
DE10119338A1 (en) * 2001-04-20 2002-10-24 Clariant Gmbh Use of copolymers based on acrylamidoalkylsulfonic acids as thickeners in preparations containing organic solvents
BR0209177A (en) 2001-05-03 2004-10-05 Merck Patent Gmbh Recombinant tumor specific antibody and use
ES2381025T3 (en) 2001-12-04 2012-05-22 Merck Patent Gmbh Immunocytokines with modulated selectivity
MXPA04008150A (en) * 2002-02-21 2005-06-17 Wyeth Corp A follistatin domain containing protein.
US6938440B2 (en) * 2002-11-01 2005-09-06 Michele Smith Designs, Inc. Jewelry distribution and display combination and kit
ES2346205T3 (en) 2002-12-17 2010-10-13 Merck Patent Gmbh HUMANIZED ANTIBODY (H14.18) OF ANTIBODY 14.18 OF MOUSE THAT LINKS 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
US7435788B2 (en) * 2003-12-19 2008-10-14 Advanced Cardiovascular Systems, Inc. Biobeneficial polyamide/polyethylene glycol polymers for use with drug eluting stents
WO2005063820A2 (en) 2003-12-30 2005-07-14 Merck Patent Gmbh Il-7 fusion proteins
KR20060124656A (en) 2003-12-31 2006-12-05 메르크 파텐트 게엠베하 Fc-erythropoietin fusion protein with improved pharmacokinetics
CA2553883C (en) 2004-01-22 2013-04-02 Merck Patent Gesellschaft Mit Beschraenkter Haftung Anti-cancer antibodies with reduced complement fixation
US7670595B2 (en) 2004-06-28 2010-03-02 Merck Patent Gmbh Fc-interferon-beta fusion proteins
DE602005020837D1 (en) 2004-12-09 2010-06-02 Merck Patent Gmbh IL-7 VARIANTS WITH REDUCED IMMUNOGENITY
US20070104689A1 (en) 2005-09-27 2007-05-10 Merck Patent Gmbh Compositions and methods for treating tumors presenting survivin antigens
KR20080090484A (en) 2005-12-30 2008-10-08 메르크 파텐트 게엠베하 Anti-il-6 antibodies preventing the binding of il-6 complexed with il-6ralpha to gp130
PT2270050E (en) 2005-12-30 2013-09-12 Merck Patent Gmbh Anti-cd19 antibodies with reduced immunogenicity
CA2635618C (en) 2005-12-30 2015-10-06 Merck Patent Gesellschaft Mit Beschraekter Haftung Interleukin-12p40 variants with improved stability
AU2007229698B9 (en) 2006-03-24 2012-11-08 Merck Patent Gmbh Engineered heterodimeric protein domains
EP2038417A2 (en) 2006-07-06 2009-03-25 Merck Patent GmbH Compositions and methods for enhancing the efficacy of il-2 mediated immune responses

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4737462A (en) * 1982-10-19 1988-04-12 Cetus Corporation Structural genes, plasmids and transformed cells for producing cysteine depleted muteins of interferon-β
WO2000023472A2 (en) * 1998-10-16 2000-04-27 Biogen, Inc. Interferon-beta fusion proteins and uses
WO2000069913A1 (en) * 1999-05-19 2000-11-23 Lexigen Pharmaceuticals Corp. EXPRESSION AND EXPORT OF INTERFERON-ALPHA PROTEINS AS Fc FUSION PROTEINS
WO2001003737A1 (en) * 1999-07-13 2001-01-18 Bolder Biotechnology Inc. Immunoglobulin fusion proteins
WO2002074783A2 (en) * 2001-03-15 2002-09-26 Merck Patent Gmbh Modified interferon beta with reduced immunogenicity

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
DAY C ET AL: "ENGINEERED DISULFIDE BOND GREATLY INCREASES SPECIFIC ACTIVITY OF RECOMBINANT MURINE INTERFERON- BETA" JOURNAL OF INTERFERON RESEARCH, MARY ANN LIEBERT, INC., NEW YORK, NY, US, vol. 12, no. 2, 1 April 1992 (1992-04-01), pages 139-143, XP000647532 ISSN: 0197-8357 *
LO K-M ET AL: "High level expression and secretion of Fc-X fusion proteins in mammalian cells" PROTEIN ENGINEERING, OXFORD UNIVERSITY PRESS, SURREY, GB, vol. 11, no. 6, June 1998 (1998-06), pages 495-500, XP002125745 ISSN: 0269-2139 cited in the application *
RUNKEL LAURA ET AL: "Systematic mutational mapping of sites on human interferon-beta-1a that are important for receptor binding and functional activity" BIOCHEMISTRY, AMERICAN CHEMICAL SOCIETY. EASTON, PA, US, vol. 39, no. 10, 14 March 2000 (2000-03-14), pages 2538-2551, XP002160244 ISSN: 0006-2960 *
STEWART A G ET AL: "CHEMICAL MUTAGENESIS OF HUMAN INTERFERON-BETA: CONSTRUCTION, EXPRESSION IN E. COLI, AND BIOLOGICAL ACTIVITY OF SODIUM BISULFITE-INDUCED MUTATIONS" DNA, MARY ANN LIEBERT, NEW YORK, NY, US, vol. 6, no. 2, April 1987 (1987-04), pages 119-128, XP009004161 ISSN: 0198-0238 *

Cited By (150)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006127757A2 (en) * 2005-05-26 2006-11-30 Schering Corporation Interferon-igg fusion
WO2006127757A3 (en) * 2005-05-26 2007-03-15 Schering Corp Interferon-igg fusion
JP2008545409A (en) * 2005-05-26 2008-12-18 シェーリング コーポレイション Interferon-IgG fusion
US8765915B2 (en) 2006-02-06 2014-07-01 Csl Behring Gmbh Modified coagulation factor VIIa with extended half-life
US9745361B2 (en) 2006-10-31 2017-08-29 East Carolina University Cytokine-based fusion proteins for treatment of immune disorders
US8920808B2 (en) 2006-10-31 2014-12-30 East Carolina University Cytokine-based fusion proteins for treatment of multiple sclerosis
EP2572734A3 (en) * 2006-10-31 2013-06-19 East Carolina University Cytokine-based fusion proteins for treatment of immune disorders
US10273284B2 (en) 2006-10-31 2019-04-30 East Carolina University Cytokine-based fusion proteins for treatment of immune disorders
WO2008077616A1 (en) 2006-12-22 2008-07-03 Csl Behring Gmbh Modified coagulation factors with prolonged in vivo half-life
EP3231440A1 (en) 2006-12-22 2017-10-18 CSL Behring GmbH Modified coagulation factors with prolonged in vivo half-life
US8754194B2 (en) 2006-12-22 2014-06-17 Csl Behring Gmbh Modified coagulation factors with prolonged in vivo half-life
US8283319B2 (en) 2007-02-12 2012-10-09 Csl Behring Gmbh Therapeutic application of Kazal-type serine protease inhibitors
EP2526962A1 (en) 2007-02-12 2012-11-28 CSL Behring GmbH Therapeutic application of Kazal-type serine protease inhibitors
US8114630B2 (en) 2007-05-02 2012-02-14 Ambrx, Inc. Modified interferon beta polypeptides and their uses
JP2010529835A (en) * 2007-05-25 2010-09-02 ビオマリン プハルマセウトイカル インコーポレイテッド Prokaryotic phenylalanine ammonia lyase composition and method of using the composition
US10786551B2 (en) 2007-08-06 2020-09-29 Generon (Shanghai) Corporation Ltd. Use of interleukin-22 in the treatment of fatty liver disease
JP2017031215A (en) * 2007-09-21 2017-02-09 ザ リージェンツ オブ ザ ユニバーシティ オブ カリフォルニア Interferon-antibody fusion proteins demonstrating potent apoptotic and anti-tumor activities
US10182984B2 (en) 2007-09-21 2019-01-22 The Regents Of The University Of California Targeted interferons demonstrate potent apoptotic and anti-tumor activities
WO2009053368A1 (en) * 2007-10-22 2009-04-30 Merck Serono S.A. Single ifn-beta fused to a mutated igg fc fragment
EP2865760A1 (en) 2008-06-24 2015-04-29 CSL Behring GmbH Factor VIII, von Willebrand factor or complexes thereof with prolonged in vivo half-life
US8575104B2 (en) 2008-06-24 2013-11-05 Csl Behring Gmbh Factor VIII, von willebrand factor or complexes thereof with prolonged in vivo half-life
US9290561B2 (en) 2008-06-24 2016-03-22 Csl Behring Gmbh Factor VIII, von Willebrand factor or complexes thereof with prolonged in vivo half-life
US10363306B2 (en) 2009-03-31 2019-07-30 East Carolina University Cytokines and neuroantigens for treatment of immune disorders
WO2010121766A1 (en) * 2009-04-22 2010-10-28 Merck Patent Gmbh Antibody fusion proteins with modified fcrn binding sites
US8907066B2 (en) 2009-04-22 2014-12-09 Merck Patent Gmbh Antibody fusion proteins with a modified FcRn binding site
CN102405230A (en) * 2009-04-22 2012-04-04 默克专利有限公司 Antibody fusion proteins with modified fcrn binding sites
US10576166B2 (en) 2009-12-01 2020-03-03 Translate Bio, Inc. Liver specific delivery of messenger RNA
WO2011098644A2 (en) 2010-02-09 2011-08-18 Proyecto De Biomedicina Cima, S.L. Compositions for the treatment of infectious and tumoural diseases
EP2371857A1 (en) 2010-04-01 2011-10-05 CSL Behring GmbH Factor XII inhibitors for treating interstitial lung disease
WO2011121123A1 (en) 2010-04-01 2011-10-06 Csl Behring Gmbh Factor xii inhibitors for treating interstitial lung disease
US9150638B2 (en) 2010-04-01 2015-10-06 Justus-Liebig-Universität Giessen Factor XII inhibitors for treating interstitial lung disease
US8945528B2 (en) 2010-08-31 2015-02-03 Generon (Shanghai) Corporation Ltd. Use of interleukin-22 in treating viral hepatitis
US9629898B2 (en) 2010-08-31 2017-04-25 Generon (Shanghai) Corporation, Ltd. Use of interleukin-22 in treating viral hepatitis
CN102585013A (en) * 2011-01-07 2012-07-18 中国人民解放军军事医学科学院生物工程研究所 Fusion protein containing omega interferon and method for preparing same
US9803210B2 (en) 2011-02-11 2017-10-31 Research Corporation Technologies, Inc. Fusion proteins comprising immunoglobulin constant domain-derived scaffolds
US9624307B2 (en) 2011-03-09 2017-04-18 The General Hospital Corporation Factor XII inhibitors for the treatment of silent brain ischemia and ischemia of other organs
US9352016B2 (en) 2011-03-09 2016-05-31 Csl Behring Gmbh Factor XII inhibitors for the administration with medical procedures comprising contact with artificial surfaces
US9987328B2 (en) 2011-03-09 2018-06-05 Csl Behring Gmbh Factor XII inhibitors for the administration with medical procedures comprising contact with artificial surfaces
EP2497489A1 (en) 2011-03-09 2012-09-12 CSL Behring GmbH Factor XII inhibitors for the treatment of silent brain ischemia and ischemia of other organs
WO2012120124A1 (en) 2011-03-09 2012-09-13 Csl Behring Gmbh Factor xii inhibitors for the treatment of silent brain ischemia and ischemia of other organs
WO2012120128A1 (en) 2011-03-09 2012-09-13 Csl Behring Gmbh Factor xii inhibitors for the administration with medical procedures comprising contact with artificial surfaces
US10752664B2 (en) 2011-04-08 2020-08-25 Amgen Inc. Method of treating or ameliorating metabolic disorders using growth differentiation factor 15 (GDF-15)
US11052159B2 (en) 2011-06-08 2021-07-06 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US10507249B2 (en) 2011-06-08 2019-12-17 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US11185595B2 (en) 2011-06-08 2021-11-30 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US11547764B2 (en) 2011-06-08 2023-01-10 Translate Bio, Inc. Lipid nanoparticle compositions and methods for MRNA delivery
US9597413B2 (en) 2011-06-08 2017-03-21 Shire Human Genetic Therapies, Inc. Pulmonary delivery of mRNA
US11730825B2 (en) 2011-06-08 2023-08-22 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US10350303B1 (en) 2011-06-08 2019-07-16 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US10413618B2 (en) 2011-06-08 2019-09-17 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US9308281B2 (en) 2011-06-08 2016-04-12 Shire Human Genetic Therapies, Inc. MRNA therapy for Fabry disease
US10888626B2 (en) 2011-06-08 2021-01-12 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US11951179B2 (en) 2011-06-08 2024-04-09 Translate Bio, Inc. Lipid nanoparticle compositions and methods for MRNA delivery
US11951180B2 (en) 2011-06-08 2024-04-09 Translate Bio, Inc. Lipid nanoparticle compositions and methods for MRNA delivery
US11951181B2 (en) 2011-06-08 2024-04-09 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US11291734B2 (en) 2011-06-08 2022-04-05 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US11338044B2 (en) 2011-06-08 2022-05-24 Translate Bio, Inc. Lipid nanoparticle compositions and methods for mRNA delivery
US10238754B2 (en) 2011-06-08 2019-03-26 Translate Bio, Inc. Lipid nanoparticle compositions and methods for MRNA delivery
US11434271B2 (en) 2011-11-04 2022-09-06 Hanmi Science Co., Ltd. Method for preparing physiologically active polypeptide complex
US9352024B2 (en) 2011-12-27 2016-05-31 Generon (Shanghai) Corporation Ltd. Uses of interleukin-22(IL-22) in treating and preventing nerve damage diseases or neurodegenerative diseases
US11739131B2 (en) 2012-01-26 2023-08-29 Amgen Inc. Growth differentiation factor 15 (GDF-15) polypeptides
US10336798B2 (en) 2012-01-26 2019-07-02 Amgen Inc. Growth differentiation factor 15 (GDF-15) polypeptides
WO2013119903A1 (en) 2012-02-10 2013-08-15 Research Corporation Technologies, Inc. Fusion proteins comprising immunoglobulin constant domain-derived scaffolds
EP2812432A4 (en) * 2012-02-10 2016-02-17 Res Corp Technologies Inc Fusion proteins comprising immunoglobulin constant domain-derived scaffolds
WO2013120939A1 (en) 2012-02-15 2013-08-22 Csl Behring Gmbh Von willebrand factor variants having improved factor viii binding affinity
US9458223B2 (en) 2012-02-15 2016-10-04 Csl Behring Gmbh Von willebrand factor variants having improved factor VIII binding affinity
US11168109B2 (en) 2012-03-08 2021-11-09 Hanmi Science Co., Ltd. Process for preparation of physiologically active polypeptide complex
US10195250B2 (en) 2012-03-27 2019-02-05 Ngm Biopharmaceuticals, Inc. Compositions and methods of use for treating metabolic disorders
US10869909B2 (en) 2012-03-27 2020-12-22 Ngm Biopharmaceuticals, Inc. Compositions and methods of use for treating metabolic disorders
US9550819B2 (en) 2012-03-27 2017-01-24 Ngm Biopharmaceuticals, Inc. Compositions and methods of use for treating metabolic disorders
US11254936B2 (en) 2012-06-08 2022-02-22 Translate Bio, Inc. Nuclease resistant polynucleotides and uses thereof
US10975158B2 (en) 2012-12-07 2021-04-13 The Regents Of The University Of California CD138-targeted interferon demonstrates potent apoptotic and anti-tumor activities
US10323075B2 (en) 2013-01-30 2019-06-18 Ngm Biopharmaceuticals, Inc. Compositions and methods of use for treating metabolic disorders
US9828415B2 (en) 2013-01-30 2017-11-28 Ngm Biopharmaceuticals, Inc. Compositions and methods of use for treating metabolic disorders
US10610568B2 (en) 2013-01-30 2020-04-07 Ngm Biopharmaceuticals, Inc. Compositions and methods of use for treating metabolic disorders
US9827291B2 (en) 2013-01-30 2017-11-28 Ngm Biopharmaceuticals, Inc. Compositions and methods of use for treating metabolic disorders
US10286047B2 (en) 2013-03-08 2019-05-14 Csl Behring Gmbh Treatment and prevention of remote ischemia-reperfusion injury
US10973891B2 (en) 2013-03-08 2021-04-13 Csl Behring Gmbh Treatment and prevention of remote ischemia-reperfusion injury
US9713626B2 (en) 2013-03-14 2017-07-25 Rana Therapeutics, Inc. CFTR mRNA compositions and related methods and uses
WO2014152940A1 (en) * 2013-03-14 2014-09-25 Shire Human Genetic Therapies, Inc. Mrna therapeutic compositions and use to treat diseases and disorders
US11820977B2 (en) 2013-03-14 2023-11-21 Translate Bio, Inc. Methods for purification of messenger RNA
US10876104B2 (en) 2013-03-14 2020-12-29 Translate Bio, Inc. Methods for purification of messenger RNA
US9181321B2 (en) 2013-03-14 2015-11-10 Shire Human Genetic Therapies, Inc. CFTR mRNA compositions and related methods and uses
EP3431592A1 (en) * 2013-03-14 2019-01-23 Translate Bio, Inc. Mrna therapeutic compositions and use to treat diseases and disorders
US11510937B2 (en) 2013-03-14 2022-11-29 Translate Bio, Inc. CFTR MRNA compositions and related methods and uses
US10420791B2 (en) 2013-03-14 2019-09-24 Translate Bio, Inc. CFTR MRNA compositions and related methods and uses
US11692189B2 (en) 2013-03-14 2023-07-04 Translate Bio, Inc. Methods for purification of messenger RNA
US9957499B2 (en) 2013-03-14 2018-05-01 Translate Bio, Inc. Methods for purification of messenger RNA
US9878017B2 (en) 2013-04-22 2018-01-30 Csl Ltd. Covalent complex of von Willebrand Factor and factor VIII, compositions, and uses relating thereto
US10822427B2 (en) 2013-05-29 2020-11-03 The Regents Of The University Of California Anti-CSPG4 fusions with interferon for the treatment of malignancy
US10093745B2 (en) 2013-05-29 2018-10-09 The Regents Of The University Of California Anti-CSPG4 fusions with interferon for the treatment of malignancy
EA037355B1 (en) * 2013-07-31 2021-03-17 Эмджен Инк. Growth differentiation factor 15 (gdf-15) constructs
US9862752B2 (en) 2013-07-31 2018-01-09 Amgen Inc. Growth differentiation factor 15 (GDF-15) constructs
US10894814B2 (en) 2013-07-31 2021-01-19 Amgen Inc. Growth differentiation factor 15 (GDF-15) constructs
CN105980400B (en) * 2013-07-31 2021-05-07 美国安进公司 Growth differentiation factor 15(GDF-15) constructs
WO2015017710A1 (en) * 2013-07-31 2015-02-05 Amgen Inc. Growth differentiation factor 15 (gdf-15) constructs
CN105980400A (en) * 2013-07-31 2016-09-28 美国安进公司 Growth differentiation factor 15 (GDF-15) constructs
US11224642B2 (en) 2013-10-22 2022-01-18 Translate Bio, Inc. MRNA therapy for argininosuccinate synthetase deficiency
US9522176B2 (en) 2013-10-22 2016-12-20 Shire Human Genetic Therapies, Inc. MRNA therapy for phenylketonuria
US10208295B2 (en) 2013-10-22 2019-02-19 Translate Bio, Inc. MRNA therapy for phenylketonuria
US11377642B2 (en) 2013-10-22 2022-07-05 Translate Bio, Inc. mRNA therapy for phenylketonuria
CN103570836A (en) * 2013-10-25 2014-02-12 江苏众红生物工程创药研究院有限公司 Recombinant porcine interferon beta1-Fc fusion protein as well as encoding gene and expressing method thereof
US11654104B2 (en) 2013-11-07 2023-05-23 Evive Biotechnology (Shanghai) Ltd Use of IL-22 dimer in manufacture of a medicament for intravenous administration
US10543169B2 (en) 2013-11-07 2020-01-28 Generon (Shanghai) Corporation Ltd. Use of IL-22 dimer in manufacture of a medicament for intravenous administration
AU2015242657B2 (en) * 2014-03-31 2020-05-21 Hanmi Pharm. Co., Ltd. Method for improving solubility of protein and peptide by using immunoglobulin Fc fragment linkage
EP3127923A4 (en) * 2014-03-31 2018-04-18 Hanmi Pharm. Co., Ltd. Method for improving solubility of protein and peptide by using immunoglobulin fc fragment linkage
EP3889185A3 (en) * 2014-03-31 2022-01-26 Hanmi Pharm. Co., Ltd. Composition for improving the solubility of a protein or peptide by using immunoglobulin fc fragment linkage
US11059841B2 (en) 2014-04-25 2021-07-13 Translate Bio, Inc. Methods for purification of messenger RNA
US11884692B2 (en) 2014-04-25 2024-01-30 Translate Bio, Inc. Methods for purification of messenger RNA
US10155785B2 (en) 2014-04-25 2018-12-18 Translate Bio, Inc. Methods for purification of messenger RNA
US9850269B2 (en) 2014-04-25 2017-12-26 Translate Bio, Inc. Methods for purification of messenger RNA
WO2016000039A1 (en) 2014-07-02 2016-01-07 Csl Limited Modified von willebrand factor
US10253088B2 (en) 2014-07-02 2019-04-09 CSL Behring Lengnau AG Modified von Willebrand Factor
US9834586B2 (en) 2014-07-30 2017-12-05 Ngm Biopharmaceuticals, Inc. Compositions and methods of use for treating metabolic disorders
US11358995B2 (en) 2014-07-30 2022-06-14 Ngm Biopharmaceuticals, Inc. Compositions and methods of use for treating metabolic disorders
US9920118B2 (en) 2014-10-31 2018-03-20 Ngm Biopharmaceuticals, Inc. Compositions and methods of use for treating metabolic disorders
US10562965B2 (en) 2014-10-31 2020-02-18 Ngm Biopharmaceuticals, Inc. Compositions and methods of use for treating metabolic disorders
US11530260B2 (en) 2014-10-31 2022-12-20 Ngm Biopharmaceuticals, Inc. Compositions and methods of use for treating metabolic disorders
EP3266801A4 (en) * 2015-03-03 2018-09-12 Abion Inc. Human interferon-beta variant conjugated immunocytokine and method for preparing same
EP4089109A2 (en) 2015-05-22 2022-11-16 CSL Behring Lengnau AG Methods for preparing modified von willebrand factor
US10905747B2 (en) 2015-05-22 2021-02-02 CSL Behring Lengnau AG Methods for preparing modified von Willebrand factor
US10688157B2 (en) 2015-05-22 2020-06-23 CSL Behring Lengnau AG Truncated von Willebrand factor polypeptides for treating hemophilia
US10772936B2 (en) 2015-05-22 2020-09-15 CSL Behring Lengnau AG Methods for preparing modified von Willebrand factor
WO2016188907A1 (en) 2015-05-22 2016-12-01 Csl Behring Recombinant Facility Ag Truncated von willebrand factor polypeptides for treating hemophilia
US11564976B2 (en) 2015-05-22 2023-01-31 CSL Behring Lengnau AG Methods for preparing modified von Willebrand Factor
WO2016188905A1 (en) 2015-05-22 2016-12-01 Csl Behring Recombinant Facility Ag Methods for preparing modified von willebrand factor
US11028144B2 (en) 2015-12-23 2021-06-08 Julius-Maximilians-Universität Würzburg Soluble glycoprotein V for treating thrombotic diseases
EP3184149A1 (en) 2015-12-23 2017-06-28 Julius-Maximilians-Universität Würzburg Soluble glycoprotein v for treating thrombotic diseases
WO2017117631A1 (en) 2016-01-07 2017-07-13 Csl Limited Mutated truncated von willebrand factor
US10808023B2 (en) 2016-01-07 2020-10-20 CSL Behring Lengnau AG Mutated von Willebrand factor
US10806774B2 (en) 2016-01-07 2020-10-20 CSL Behring Lengnau AG Mutated truncated von Willebrand Factor
US11510966B2 (en) 2016-04-15 2022-11-29 Evive Biotechnology (Shanghai) Ltd Use of IL-22 in treating necrotizing enterocolitis
US10870689B2 (en) * 2016-08-15 2020-12-22 The Children's Medical Center Corporation ApoM-Fc fusion proteins, complexes thereof with sphingosine 1-phosphate (S1P), and methods for treating vascular and non-vascular diseases
US20190185545A1 (en) * 2016-08-15 2019-06-20 Children's Medical Center Corporation Apom-fc fusion proteins, complexes thereof with sphingosine 1-phosphate (s1p), and methods for treating vascular and non-vascular diseases
WO2018087271A1 (en) 2016-11-11 2018-05-17 Csl Behring Recombinant Facility Ag Truncated von willebrand factor polypeptides for extravascular administration in the treatment or prophylaxis of a blood coagulation disorder
US11890327B2 (en) 2016-11-11 2024-02-06 CSL Behring Lengnau AG Truncated von Willebrand factor polypeptides for extravascular administration in the treatment or prophylaxis of a blood coagulation disorder
WO2018087267A1 (en) 2016-11-11 2018-05-17 Csl Behring Recombinant Facility Ag Truncated von willebrand factor polypeptides for treating hemophilia
US11814421B2 (en) 2016-11-11 2023-11-14 CSL Behring Lengnau AG Truncated von Willebrand Factor polypeptides for treating hemophilia
US11253605B2 (en) 2017-02-27 2022-02-22 Translate Bio, Inc. Codon-optimized CFTR MRNA
US11173190B2 (en) 2017-05-16 2021-11-16 Translate Bio, Inc. Treatment of cystic fibrosis by delivery of codon-optimized mRNA encoding CFTR
US11141466B2 (en) 2017-06-22 2021-10-12 CSL Behring Lengnau AG Modulation of FVIII immunogenicity by truncated VWF
WO2018234518A1 (en) 2017-06-22 2018-12-27 CSL Behring Lengnau AG Modulation of fviii immunogenicity by truncated vwf
US11161889B2 (en) 2018-04-09 2021-11-02 Amgen Inc. Growth differentiation factor 15 fusion proteins
US11174500B2 (en) 2018-08-24 2021-11-16 Translate Bio, Inc. Methods for purification of messenger RNA
WO2020234195A1 (en) 2019-05-17 2020-11-26 Universitaet Zuerich Haptoglobin for use in treating an adverse secondary neurological outcome following a haemorrhagic stroke
WO2021001522A1 (en) 2019-07-04 2021-01-07 CSL Behring Lengnau AG A truncated von willebrand factor (vwf) for increasing the in vitro stability of coagulation factor viii
WO2021094344A1 (en) 2019-11-11 2021-05-20 CSL Behring Lengnau AG Polypeptides for inducing tolerance to factor viii
WO2022162218A1 (en) 2021-02-01 2022-08-04 Csl Behring Ag Method of treating or preventing an adverse secondary neurological outcome following a haemorrhagic stroke
WO2022234070A1 (en) 2021-05-07 2022-11-10 Csl Behring Ag Expression system for producing a recombinant haptoglobin (hp) beta chain
WO2024047219A1 (en) 2022-09-02 2024-03-07 Csl Behring Ag Haptoglobin for use in treating or preventing exaggerated erectile response or erectile dysfunction

Also Published As

Publication number Publication date
EP1761559A2 (en) 2007-03-14
CA2572015A1 (en) 2006-01-05
WO2006000448A3 (en) 2006-03-02
EP1761559B1 (en) 2009-09-23
US7670595B2 (en) 2010-03-02
AU2005256519B2 (en) 2011-10-13
CA2572015C (en) 2014-04-22
US8557232B2 (en) 2013-10-15
ES2332924T3 (en) 2010-02-15
JP4808709B2 (en) 2011-11-02
US20140005361A1 (en) 2014-01-02
US20090191154A1 (en) 2009-07-30
US20060228332A1 (en) 2006-10-12
AU2005256519A1 (en) 2006-01-05
JP2008508862A (en) 2008-03-27
DE602005016799D1 (en) 2009-11-05
ATE443720T1 (en) 2009-10-15

Similar Documents

Publication Publication Date Title
EP1761559B1 (en) Fc-INTERFERON-BETA FUSION PROTEINS
AU777963B2 (en) Expression and export of interferon-alpha proteins as Fc fusion proteins
EP1366067B1 (en) Expression technology for proteins containing a hybrid isotype antibody moiety
US7232668B2 (en) Fc fusion proteins of human granulocyte colony-stimulating factor with increased biological activities
EP0888122B1 (en) HYBRID WITH INTERFERON-alpha AND AN IMMUNOGLOBULIN Fc LINKED THROUGH A NON-IMMUNOGENIC PEPTIDE
US8907066B2 (en) Antibody fusion proteins with a modified FcRn binding site
AU2002248571A1 (en) Expression technology for proteins containing a hybrid isotype antibody moiety
US8637637B2 (en) Fc fusion proteins of human growth hormone
JP7136690B2 (en) Immunoglobulin fusion proteins and uses thereof

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

AL Designated countries for regional patents

Kind code of ref document: A2

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

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2005766761

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2572015

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2007517216

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

WWW Wipo information: withdrawn in national office

Country of ref document: DE

WWE Wipo information: entry into national phase

Ref document number: 2005256519

Country of ref document: AU

ENP Entry into the national phase

Ref document number: 2005256519

Country of ref document: AU

Date of ref document: 20050628

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2005256519

Country of ref document: AU

WWP Wipo information: published in national office

Ref document number: 2005766761

Country of ref document: EP