US20210009659A1 - Multivalent heteromultimer scaffold design and constructs - Google Patents

Multivalent heteromultimer scaffold design and constructs Download PDF

Info

Publication number
US20210009659A1
US20210009659A1 US16/897,583 US202016897583A US2021009659A1 US 20210009659 A1 US20210009659 A1 US 20210009659A1 US 202016897583 A US202016897583 A US 202016897583A US 2021009659 A1 US2021009659 A1 US 2021009659A1
Authority
US
United States
Prior art keywords
heteromultimer
polypeptide
albumin
transporter
derived
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US16/897,583
Inventor
Surjit Bhimarao Dixit
Igor Edmondo Paolo D'angelo
David Kai Yuen Poon
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Zymeworks BC Inc
Original Assignee
Zymeworks Inc Canada
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 Zymeworks Inc Canada filed Critical Zymeworks Inc Canada
Priority to US16/897,583 priority Critical patent/US20210009659A1/en
Publication of US20210009659A1 publication Critical patent/US20210009659A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • 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
    • C07K14/76Albumins
    • C07K14/765Serum albumin, e.g. HSA
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • 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
    • 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
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • 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
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4721Lipocortins
    • 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
    • C07K14/475Growth factors; Growth regulators
    • C07K14/485Epidermal growth factor [EGF] (urogastrone)
    • 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
    • C07K14/575Hormones
    • C07K14/605Glucagons
    • 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
    • C07K14/76Albumins
    • 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
    • C07K14/79Transferrins, e.g. lactoferrins, ovotransferrins
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/283Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against Fc-receptors, e.g. CD16, CD32, CD64
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/32Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/48Hydrolases (3) acting on peptide bonds (3.4)
    • C12N9/50Proteinases, e.g. Endopeptidases (3.4.21-3.4.25)
    • C12N9/64Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue
    • C12N9/6421Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue from mammals
    • C12N9/6424Serine endopeptidases (3.4.21)
    • C12N9/6454Dibasic site splicing serine proteases, e.g. kexin (3.4.21.61); furin (3.4.21.75) and other proprotein convertases
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y304/00Hydrolases acting on peptide bonds, i.e. peptidases (3.4)
    • C12Y304/21Serine endopeptidases (3.4.21)
    • C12Y304/21061Kexin (3.4.21.61), i.e. proprotein convertase subtilisin/kexin type 9
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/31Fusion polypeptide fusions, other than Fc, for prolonged plasma life, e.g. albumin

Definitions

  • the field of the invention is the rational design of a scaffold for custom development of biotherapeutics.
  • HSA Human serum albumin
  • HA Human serum albumin
  • HSA Human serum albumin possesses many desirable characteristics. HSA is found throughout the body, but more specifically in the interstitial space and in blood at serum concentrations of 40 g/L which is equivalent to 0.7 mM (Yeh et al., Proc. Natl. Acad. Sci. USA, 89:1904-1908 (1992)). HSA is considered to be the most abundant protein of the serum and is responsible for maintaining osmolarity. HSA has favorable pharmacokinetic properties and is cleared very slowly by the liver and kidney displaying in vivo half-lives up to several weeks (Yeh et al., Proc. Natl. Acad. Sci. USA, 89:1904-1908 (1992); Waldmann, T.
  • HSA lacks enzymatic activity and antigenicity thereby eliminating potentially undesirable side effects. HSA acts as a carrier for endogenous as well as exogenous ligands. Combined, these features can be extended, at least partially, onto albumin based fusion protein. The poor pharmacokinetic properties displayed by therapeutic proteins can then be circumvented.
  • heteromultimers each heteromultimer comprising: at least a first monomer unit that comprises at least one cargo molecule, and a first transporter polypeptide; and at least a second monomer unit that comprises at least one cargo molecule and a second transporter polypeptide; wherein at least one transporter polypeptide is derived from a monomeric protein and wherein said transporter polypeptides self-assemble to form a quasi-native structure of said monomeric protein or analog thereof.
  • at least one cargo molecule is a drug, or a therapeutic agent.
  • at least one cargo molecule is a biomolecule.
  • the at least one biomolecule is a DNA, RNA, PNA or polypeptide.
  • at least one cargo molecule is a polypeptide.
  • each monomeric transporter polypeptide is unstable and preferentially forms a heteromultimer with at least one other transporter polypeptide.
  • each monomeric transporter polypeptide is stable and preferentially forms a heteromultimer with at least one other transporter polypeptide.
  • the heteromultimerization interface comprises at least one disulfide bond. In certain embodiments, the heteromultimerization interface does not comprise a disulfide bond.
  • heteromultimer that comprises: at least two monomers, wherein each monomer comprises at least one cargo molecule attached to a transporter polypeptide, such that said monomers self-assemble to form the heteromultimer.
  • a heteromultimer that comprises: at least two monomeric proteins, wherein each monomeric protein comprises at least one cargo polypeptide, attached to a transporter polypeptide, wherein at least one transporter polypeptide is derived from a monomeric protein and wherein said transporter polypeptides self-assemble to form a quasi-native structure of said monomeric protein or analog thereof.
  • heteromultimer that comprises: at least two monomeric proteins, wherein each monomeric protein comprises at least one cargo polypeptide attached to a transporter polypeptide, such that said monomeric proteins self-assemble via the transporter polypeptide to form the heteromultimer, and wherein at least one transporter polypeptide is derived from a monomeric protein and wherein said transporter polypeptides self-assemble to form a quasi-native structure of said monomeric protein or analog thereof.
  • the heteromultimer is a heterodimer.
  • the heteromultimer is bispecific.
  • the heteromultimer is multispecific.
  • the heteromltimer is bivalent.
  • the heteromultimer is multivalent.
  • the heteromultimer is multifunctional.
  • at least one transporter polypeptide is not derived from an antibody.
  • the transporter polypeptides are not derived from an antibody.
  • the transporter polypeptides are derivatives of albumin.
  • the transporter polypeptides are derived from human serum albumin (HSA or HA) of SEQ ID No. 1.
  • the transporter polypeptides are derived from alloalbumins (HAA).
  • the transporter polypeptides are derived from sequence homologous to the human serum albumin (HSA or HA) of SEQ ID No. 1.
  • the transporter polypeptides are derivatives of an annexin protein. In an embodiment, the transporter polypeptides are derived from different annexin proteins. In certain embodiments, the transporter polypeptides are derived from the same annexin protein. In an embodiment, at least one transporter polypeptide is derived from Annexin A1 or lipocortin I. In certain embodiments of the heteromultimer, all transporter polypeptides are derived from Annexin A1 of SEQ ID NO: 14. In certain embodiments of the heteromultimer, at least one transporter polypeptides is derived from a sequence homologous to SEQ ID NO: 14.
  • At least one transporter polypeptide is derived from Annexin A2 or annexin II. In certain embodiments of the heteromultimer, all transporter polypeptides are derived from Annexin A2 or lipocortin II. In an embodiment, at least one transporter polypeptide is derived from Annexin like protein. In certain embodiments of the heteromultimer, all transporter polypeptides are derived from Annexin like protein. In an embodiment, at least one transporter polypeptide is derived from the group comprising Annexin A1-Annexin A7. In an embodiment of the heteromultimer described herein, all transporter polypeptides are derived from the group comprising Annexin A1-Annexin A7. 14. In certain embodiments, the first annexin based transporter polypeptide has a sequence comprising SEQ ID NO:15, and the second annexin based transporter polypeptide has a sequence comprising SEQ ID NO: 16.
  • the transporter polypeptides are derivatives of transferrin. In an embodiment, at least one transporter polypeptide is derived from transferrin. In certain embodiments of the heteromultimer, at least one transporter polypeptides are derived from transferrin of SEQ ID NO: 19 or analog thereof. In certain embodiments of the heteromultimer, at least one transporter polypeptide is derived from a polypeptide sequence homologous to the transferrin. In certain embodiments of the heteromultimer described herein, at least one transporter polypeptide is derived from apo-transferrin. In certain embodiments, the first transferrin based transporter polypeptide has a sequence comprising SEQ ID NO:15 and the second transferrin based transporter polypeptide has a sequence comprising SEQ ID NO: 16.
  • At least one cargo molecule is a cargo polypeptide.
  • all cargo molecules are cargo polypeptides.
  • the cargo polypeptides are therapeutic proteins or fragments or variants thereof.
  • the cargo polypeptides are antigens or fragments or variants thereof.
  • the cargo polypeptides are antigen receptors or fragments or variants thereof.
  • the cargo polypeptide is an antibody, an antibody domain, a ligand or a receptor that binds a target polypeptide.
  • at least one cargo polypeptide is fused to the transporter polypeptide.
  • At least one cargo polypeptide is attached to the N-terminus of the transporter polypeptide. In some embodiments, at least one cargo polypeptide is attached to the C-terminus of the transporter polypeptide. In some embodiments, at least one cargo polypeptide is chemically linked to the transporter polypeptide. In some embodiments of the heteromultimers described herein, at least one cargo polypeptide comprises GLP-1 or fragment or variant thereof. In some embodiments, at least one cargo polypeptide comprises glucagon or fragment or variant thereof. In an embodiment, at least one cargo polypeptide comprises an EGF-A like domain.
  • heteromultimers each heteromultimer comprising: at least a first monomeric protein that comprises at least one cargo polypeptide and a first transporter polypeptide; and at least a second monomeric protein that comprises at least one cargo polypeptide and a second transporter polypeptide.
  • the heteromultimer is a heterodimer.
  • the heteromultimer is multispecific.
  • the heteromultimer is bispecific.
  • the transporter polypeptides are derivatives of the same protein.
  • the transporter polypeptides are derivatives of albumin.
  • the transporter polypeptides are derived from human serum albumin of SEQ ID No. 1.
  • the transporter polypeptides are derivatives of an annexin.
  • the transporter polypeptides are derivatives of Annexin A2.
  • the transporter polypeptides are derivatives of transferrin.
  • heteromultimers comprising: at least a first monomeric protein that comprises at least one cargo polypeptide and a first transporter polypeptide comprising a first segment of human serum albumin; and at least a second monomeric protein that comprises at least one cargo polypeptide, fragment and a second transporter polypeptide comprising a second segment of human serum albumin; wherein said transporter polypeptides self-assemble to form a quasi-native structure of albumin or analog thereof.
  • the first and second segments of human serum albumin are from non-overlapping regions of the protein.
  • the overlap is a 5% overlap.
  • heteromultimers each heteromultimer comprising: at least a first monomeric protein that comprises at least one cargo polypeptide and a first transporter polypeptide comprising a sequence of SEQ ID NO:2; and at least a second monomeric protein that comprises at least one cargo polypeptide, and a second transporter polypeptide comprising a sequence of SEQ ID NO: 3.
  • heteromultimers each heteromultimer comprising: at least a first monomeric protein that comprises at least one cargo polypeptide and a first transporter polypeptide comprising a sequence of SEQ ID NO:8; and at least a second monomeric protein that comprises at least one cargo polypeptide and a second transporter polypeptide comprising a sequence of SEQ ID NO: 10.
  • At least one transporter polypeptide is derived from alloalbumins. In certain embodiments, both transporter polypeptides are derived from alloalbumins. In certain embodiments, all transporter polypeptides are derivatives of the same alloalbumin. In some other embodiments, the transporter polypeptides are derivatives of different alloalbumins. In some embodiments, each transporter polypeptide is an alloalbumin derivative based on an alloalbumin selected from Table 2. In certain embodiments, the first monomeric protein comprises two cargo polypeptides. In some embodiments, the second monomeric protein comprises two cargo polypeptides. In some embodiment, at least one of the monomeric proteins is engineered by introducing mutations.
  • the introduced mutations improve the functionality of the monomeric protein as compared to the native, non-mutated form of the monomer. In certain embodiments the introduced mutations improve one or more of the stability, half-life and heteromultimer formation of the transporter polypeptide.
  • heteromultimers each heteromultimer comprising: at least a first monomeric protein that comprises at least one cargo polypeptide and a first transporter polypeptide; and at least a second monomeric protein that comprises at least one cargo polypeptide and a second transporter polypeptide.
  • at least one cargo polypeptide is selected from the proteins listed in Table 2 or fragments, variants or derivatives thereof.
  • at least one cargo polypeptide is selected from ligand, receptor, or antibody to one or more proteins listed in Table 2, or fragment, variant or derivative of said ligand, receptor or antibody.
  • heteromultimers comprising: at least a first monomeric protein that comprises at least one cargo polypeptide and a first transporter polypeptide; and at least a second monomeric protein that comprises at least one cargo polypeptide and a second transporter polypeptide, wherein at least one at least one cargo polypeptide is an antibody, or fragment or variant thereof.
  • all cargo polypeptides are antibodies or fragments or variants thereof.
  • the cargo polypeptide is an antibody that binds to a protein listed in Table 2.
  • the antibody fragment comprises antibody Fc or Fab or Fv region.
  • the cargo polypeptide is a non-antibody protein like nanobodies, affibody, maxibody, adnectins, domain antibody, evibody, ankyrin repeat proteins, anticalins, camlids or ligand protein or polypeptide binding to a therapeutically relavant target.
  • the antibody or its fragment is derived from an immunoglobulin selected from the group consisting of IgG, IgA, IgD, IgE, and IgM.
  • the IgG is of subtype selected from IgG1, IgG2a, IgG2b, IgG3 and IgG4.
  • the antibody is multispecific.
  • heteromultimers each heteromultimer comprising: at least a first monomeric protein that comprises at least one cargo polypeptide and a first transporter polypeptide; and at least a second monomeric protein that comprises at least one cargo polypeptide and a second transporter polypeptide, wherein at least one cargo polypeptide is a therapeutic antibody.
  • At least one cargo polypeptide is a therapeutic antibody or fragment or variant thereof, wherein the antibody is selected from antibody is selected from abagovomab, adalimumab, alemtuzumab, aurograb, bapineuzumab, basiliximab, belimumab, bevacizumab, briakinumab, canakinumab, catumaxomab, certolizumab pegol, certuximab, daclizumab, denosumab, efalizumab, galiximab, gemtuzumab ozagamicin, golimumab, ibritumomab tiuxetan, infliximab, ipilimumab, lumiliximab, mepolizumab, motavizumab, muromonab, mycograb, natalizumab, nimotuzumab,
  • the therapeutic antibody binds a disease related target antigen such as cancer antigen, inflammatory disease antigen or a metabolic disease antigen.
  • the target antigen could be a protein on a cell surface and the cell could belong to the group of B-cell, T-cell, stromal cell, endothelial cell, vascular cell, myeloid cell, hematopoietic cell or carcinoma cell.
  • heteromultimers each heteromultimer comprising: at least a first monomer that comprises at least one cargo molecule, fragment; and at least a second monomer that comprises at least one cargo molecule and a second transporter polypeptide, wherein at least one cargo polypeptide is an enzyme, enzyme inhibitor, hormone, therapeutic polypeptide, antigen, radiotoxin and chemotoxin inclusive of but not restricted to neurotoxins, interferons, cytokine fusion toxins and chemokine fusion toxins, cytokine, antibody fusion protein or variant or fragment thereof.
  • at least one cargo polypeptide comprises GLP-1 or fragment or variant thereof.
  • At least one cargo polypeptide comprises glucagon or fragment or variant thereof. In an embodiment, at least one cargo polypeptide comprises an EGF-A like domain.
  • the toxin is an immunotoxin such as Denileukin diftitox and Anti-CD22 immunotoxin such as CAT-3888 and CAT-8015. In certain embodiments, the toxin is saporin. In some embodiments, the toxin is a mitotoxin. In some embodiments, the toxin is a diphtheria toxin. In some embodiments, the toxin is botulinux toxin type A. In some embodiments, the toxin is ricin or a fragment there of. In some embodiments, the toxin is a toxin from RTX family of toxins.
  • heteromultimers each heteromultimer comprising: at least a first monomeric protein that comprises at least one cargo polypeptide and a first transporter polypeptide; and at least a second monomeric protein that comprises at least one cargo polypeptide and a second transporter polypeptide, wherein the cargo polypeptide is attached to the transporter polypeptide by chemical conjugation, native ligation, chemical ligation, a disulfide bond or direct fusion or fusion via a linker.
  • linkers for attaching cargo molecules such as cargo polypeptides to transporter polypeptides are selected from the linkers described in U.S. Pat. Nos. 5,482,858, 5,258,498 and 5,856,456, US2009060721, U.S.
  • host cells comprising nucleic acid encoding a heteromultimer described herein.
  • the nucleic acid encoding the first monomeric protein and the nucleic acid encoding the second monomeric protein are present in a single vector.
  • the nucleic acid encoding the first monomeric protein and the nucleic acid encoding the second monomeric protein are present in separate vectors.
  • the host cell is a prokaryotic cell or a eukaryotic cell.
  • the host cell is E. coli .
  • the host cell is yeast cell.
  • the yeast is S. cerevisiae .
  • the yeast is Pichia .
  • the yeast is Pichia pastoris .
  • the yeast is glycosylation deficient, and/or protease deficient.
  • a pharmaceutical composition that comprises a heteromultimer described herein and a pharmaceutically acceptable adjuvant.
  • methods of treating an individual suffering from a disease or disorder comprising administering to the individual an effective amount of a formulation or pharmaceutical composition described herein.
  • a method of treating cancer in a patient comprising administering to the patient a therapeutically effective amount of a heteromultimer described herein.
  • a method of treating an immune disorder in a patient comprising administering to the patient a therapeutically effective amount of a heteromultimer described herein.
  • a method of treating an infectious disease in a patient said method comprising administering to the patient a therapeutically effective amount of a heteromultimer described herein.
  • a method of treating a cardiovascular disorder in a patient comprising administering to the patient a therapeutically effective amount of a heteromultimer described herein.
  • a method of treating a respiratory disorder in a patient comprising administering to the patient a therapeutically effective amount of a heteromultimer described herein.
  • a method of treating a metabolic disorder in a patient comprising administering to the patient a therapeutically effective amount of a heteromultimer described herein.
  • In certain embodiments is a method of treating one or more of Congenital adrenal hyperplasia, Gaucher's disease, Hunter syndrome, Krabbe disease, Metachromatic leukodystrophy, Niemann-Pick disease, Phenylketonuria (PKU), Porphyria, Tay-Sachs disease, and Wilson's disease in a patient, said method comprising administering to the patient a therapeutically effective amount of a heteromultimer described herein.
  • heteromultimer proteins that comprise at least two monomeric proteins, wherein each monomeric protein comprises at least one cargo polypeptide, and an albumin based polypeptide, such that said monomeric proteins self-assemble to form the heteromultimer.
  • heteromultimer proteins that comprise at least two monomeric proteins, wherein each monomeric protein comprises at least one cargo polypeptide, and a transporter polypeptide, such that said transporter polypeptides self-assemble to form the heteromultimer.
  • each transporter polypeptide is an alloalbumin based polypeptide, such that said alloalbumin based polypeptides self-assemble to form the heteromultimer.
  • each transporter polypeptide is a transferrin based polypeptide.
  • each transporter polypeptide is an annexin based polypeptide.
  • each monomeric transporter polypeptide is unstable and preferentially forms a heteromultimer with at least one other transporter polypeptide.
  • a heteromultimer described herein is a heterodimer.
  • cargo polypeptide is an antibody, enzyme, hormone, therapeutic polypeptide, antigen, chemotoxin, radiotoxin, cytokine or variant or fragment thereof.
  • the cargo polypeptide of one monomeric protein functions in synergy with the cargo polypeptide of another monomeric protein.
  • heterodimer proteins that comprise at least two monomeric fusion proteins, wherein each monomeric fusion proteins comprises at least one cargo polypeptide fused to an albumin derived polypeptide, such that said albumin derived polypeptides self-assemble to form the multifunctional heterodimer.
  • heterodimeric proteins comprising a first monomer which comprises at least one cargo polypeptide fused to an albumin derived polypeptide; and a second monomer that comprises at least one cargo polypeptide fused to an albumin derived polypeptide.
  • the at least one cargo polypeptide of the first monomer is different from the at least one cargo polypeptide of the second monomer.
  • the at least one cargo polypeptide of the first monomer is the same as the at least one cargo polypeptide of the second monomer.
  • heteromultimer proteins that comprise at least two monomeric fusion proteins, wherein each monomeric fusion proteins comprises at least one cargo polypeptide fused to an alloalbumin derived polypeptide, such that said alloalbumin derived polypeptides self-assemble to form the multifunctional heteromultimer.
  • heteromultimer proteins that comprise at least two monomeric fusion proteins, wherein each monomeric fusion proteins comprises at least one cargo polypeptide fused to a transferrin derived polypeptide, such that said transferrin derived polypeptides self-assemble to form the heteromultimer.
  • heteromultimer proteins that comprise at least two monomeric fusion proteins, wherein each monomeric fusion proteins comprises at least one cargo polypeptide fused to an annexin derived polypeptide, such that said annexin derived polypeptides self-assemble to form the heteromultimer.
  • the annexin is Annexin A2.
  • heteromultimer proteins comprising a first monomer which comprises at least one cargo polypeptide fused to an alloalbumin derived polypeptide; and a second monomer that comprises at least one cargo polypeptide fused to an alloalbumin derived polypeptide.
  • the at least one cargo polypeptide of the first monomer is different from the at least one cargo polypeptide of the second monomer.
  • the at least one cargo polypeptide of the first monomer is the same as the at least one cargo polypeptide of the second monomer.
  • a heteromultimer that comprises: at least two monomers, each comprising a transporter polypeptide and optionally at least one cargo molecule attached to said transporter polypeptide, wherein each transporter polypeptide is obtained by segmentation of a whole protein such that said transporter polypeptides self-assemble to form quasi-native whole protein.
  • the heteromultimer is multispecific.
  • the transporter polypeptides are not derived from an antibody.
  • each monomer preferentially forms the heteromultimer as compared to a monomer or a homomultimer.
  • at least one cargo molecule is a therapeutic agent, or a biomolecule.
  • At least one cargo molecule is a biomolecule which is selected from a polypeptide, DNA, PNA, or RNA.
  • each transporter polypeptide is a derivate of albumin or alloalbumin.
  • each transporter polypeptide is a derivate of annexin.
  • each transporter polypeptide is a derivate of transferrin.
  • compositions that comprise an albumin-based and/or alloalbumin-based heteromultimeric protein described herein and a pharmaceutically acceptable diluent or carrier.
  • pharmaceutical formulations that comprise a transferrin-based heteromultimeric protein described herein and a pharmaceutically acceptable diluent or carrier.
  • pharmaceutical formulations that comprise an annexin-based heteromultimeric protein described herein and a pharmaceutically acceptable diluent or carrier.
  • a formulation described herein is provided as part of a kit or container.
  • the kit or container is packaged with instructions pertaining to extended shelf life of the therapeutic protein.
  • a heteromultimer described herein is used in a method of treating (e.g., ameliorating) preventing, or diagnosing a disease or disease symptom in an individual, comprising the step of administering said formulation to the individual.
  • fusion protein scaffolds with a known number of conjugation sites based on any transport protein of interest.
  • transgenic organisms modified to contain nucleic acid molecules described herein to encode and express monomeric fusion proteins described herein.
  • FIG. 1 depicts the structure of the Human Serum Albumin (HSA) molecule.
  • HSA Human Serum Albumin
  • FIG. 2 is a plot of buried solvent accessible surface area at the interface of two albumin-based polypeptides.
  • FIG. 3 depicts two albumin-based polypeptides expressed separately.
  • the two polypeptides are shown in light and dark grey respectively.
  • Each polypeptide comprises two fusion sites for functional cargo proteins and these sites are represented as spheres.
  • the disulphide residues in structure are shown as sticks.
  • FIG. 4 is a schematic representation of bispecific and other multifunctional therapeutics based on the multispecific heteromultimer described herein.
  • the albumin-based, or alloalbumin-based polypeptides are denoted A1 and A2.
  • Multifunctional heteromultimers are obtained by conjugating antigen binding motifs, cytokines and other forms of signaling molecules, chemotoxin, radiotoxins or other functionally relevant immunoconjugates to N and/or C terminal sites on A1 and A2 and this is represented by the + symbol.
  • FIG. 5 is a schematic of a bispecific antibody derived from a heterodimeric Fc domain.
  • Albumin or alloalbumin based polypeptides are connected to the C-terminal of the Fc to selectively drive the formation of heterodimers.
  • FIGS. 6A-6C show native gel electrophoresis profiles of full-length HSA and heterodimer scaffolds Albumin-based heteromultimer-1 (ABH1) and Albumin-based heteromultimer-2 (ABH2) formed by coexpression of HSA based transporter polypeptides.
  • FIG. 7 shows stability of wild type HSA and heterodimer scaffolds ABH1 and ABH2 stuided using Differential Scanning Calorimetry
  • FIGS. 8A-8B show equilibrium binding isotherms 3000 nM FcRN 3 ⁇ dilution series over 3000 RUs.
  • FIG. 8A shows Albumin and
  • FIG. 8B shows heteromultimer scaffold ABH1
  • FIG. 9 shows scheme for multivalent Albumin based heteromultimers comprising anti-Her2/neu and anti-CD16 scFv bioactive fusions
  • FIGS. 10A-10B contain a non-reducing SDS PAGE analysis of the heteromultimer ABH2 fusions described in table 8. The gel indicates all constructs form the correct complex with expected MW.
  • FIG. 11 shows structure of Annexin molecule based on the PDB structure 1MCX.
  • the two monomers that will be derived by splitting the Annexin molecule are color coded as light and dark grey units.
  • the sites of fusion for the cargo protein are represented as spheres.
  • FIG. 12 shows a plot of the buried solvent accessible surface area at the interface of Annexin based tranporter polypeptide-1, and Annexin based tranporter polypeptide-2.
  • FIG. 13 shows structure of transferrin molecule based on the PDB structure 1H76.
  • the two monomers derived by splitting the transferrin molecule are color coded as light and dark grey units.
  • the sites of fusion for the cargo protein are represented as spheres.
  • FIG. 14 shows a plot of the buried solvent accessible surface area at the interface of two transferrin based transporter polypeptides described herein.
  • FIGS. 15A-15SS shows sequences of multimers comprising transporter polypeptides based on human serum albumin.
  • bispecific molecules exhibit dual target specificities or are able to simultaneously perform multiple functional roles by providing the necessary spatiotemporal organization necessary for drug action.
  • bispecific molecules are particularly interesting when the mode of therapeutic action involves retargeting of effector cells or molecules to a target such as a tumor cell [Muller D. and Kontermann R. E. (2010) Biodrugs 24, 89-98].
  • a target such as a tumor cell
  • the Fc portion of the antibody is involved in interactions with the neonatal Fc receptor (FcRn) which mediates an endocytic salvage pathway and this is attributed to improved serum half-life of the antibody molecule [Roopenian D. & Akilesh S. (2007) Nature Rev Immunol 7, 715-725].
  • FcRn neonatal Fc receptor
  • antibody based bispecific molecules have been problematic in clinical trials because of the strong cytokine responses as a result of the concurrent effector activity induced via the Fc portion of the bispecific antibody [Weiner L. M.; Alpaugh R. K. et al. (1996) Cancer Immunol Immunother 42, 141-150]. This highlights the needs for novel scaffolds that can aid in the design of bispecific and immunoconjugate molecules.
  • HSA human serum album
  • HSA (shown in FIG. 1 ) is a non-glycosylated 585-residue single polypeptide protein and the 3-dimensional structure of the protein was first observed using X-ray crystallography by Carter and coworkers [reviewed in Carter, D. C. & Ho, J. X. (1994) Adv Prot Chem 45, 153-203].
  • the HSA protein consists of three homologous domains: DI, DII, DIII, attributed to gene duplication, a feature common to the serum albumin in other species as well [Gray J. E. & Doolittle R. F. (1992) Protein Sci 1, 289-302]. Each of the three domains have been expressed and characterized separately and shown to be independently stable [Dockal M., Carter D.
  • Each domain is made up of 10 helical segments and based on the inter-helical organization each domain can be further classified into 2 sub-domains comprised of helix 1-6 and 7-10 respectively.
  • HSA has 17 disulphide bonds in total and all these cysteine pairs forming the linkages are within the individual domains. In general, HSA is a very stable due to the large number of disulphide bonds as well as the predominantly helical fold.
  • sequence identities of albumin molecules across a number of species is quite large, greater than 70% among albumin cDNA derived from humans, horse, bovine, rat, etc. [Carter, D. C. & Ho, J. X. (1994) Adv Prot Chem 45, 153-203].
  • split protein pairs have been used as sensors to understand protein-protein interactions in the area of functional proteomics.
  • the approach involves identifying suitable segments from a protein that can reconstitute to form an active native-like protein. Generating new split proteins is technically demanding.
  • the segmentation site has to yield two segments that efficiently reconstitute into the quasi-native protein when associated to each other.
  • the component protein segments should be soluble enough to stay in solution and selectively associate with the partner segments such that manufacture yields and purification will be economical. Deriving split protein segments that would recombine to form the quasi-native structure is quite challenging [Tafelmeyer P., Johnsson N. & Johnsson K. Chem & Biol 11, 681-689].
  • split proteins have not been used in the design of protein therapeutics, or as cargo delivery vehicles in the past.
  • a “heteromultimer” or “heteromultimeric polypeptide” is a molecule comprising at least a first monomer comprising a first transporter polypeptide and a second monomer comprising a second transporter polypeptide, wherein the second polypeptide differs in amino acid sequence from the first polypeptide by at least one amino acid residue.
  • the heteromultimer can comprise a “heterodimer” formed by the first and second transporter polypeptides.
  • the heteromultimer can form higher order tertiary structures such as, but not restricted to trimers and tetramers.
  • transporter polypeptides in addition to the first and second transporter polypeptides are present.
  • the assembly of transporter polypeptides to form the heteromultimer is driven by surface area burial.
  • the transporter polypeptides interact with each other by means of electrostatic interactions and/or salt-bridge interactions that drive heteromultimer formation by favoring heteromultimer formation and/or disfavoring homomultimer formation.
  • the transporter polypeptides inteact with each other by means of hydrophobic interactions that drive heteromultimer formation by favoring heteromultimer formation and/or disfavoring homomultimer formation.
  • the transporter polypeptides inteact with each other by means of covalent bond formation.
  • the covalent bonds are formed between naturally present or introduced cysteines that drive heteromultimer formation.
  • the transporter polypeptides inteact with each other by means of packing/size-complementarity/knobs-into-holes/protruberance-cavity type interactions that drive heteromultimer formation by favoring heteromultimer formation and/or disfavoring homomultimer formation. In some embodiments, the transporter polypeptides inteact with each other by means of cation-pi interactions that drive heteromultimer formation by favoring heteromultimer formation and/or disfavoring homomultimer formation. In certain embodiments the individual transporter polypeptides cannot exist as isolated monomers in solution. In certain embodiments, the heteromultimer is the preferred state of the individual transporter polypeptides as compared to the monomer.
  • bispecific is intended to include any agent, e.g., heteromultimer, monomer, protein, peptide, or protein or peptide complex, which has two different binding specificities.
  • the molecule may bind to, or interact with, (a) a cell surface target molecule and (b) an Fc receptor on the surface of an effector cell.
  • at least one monomer is bispecific formed by attaching to the same transporter polypeptide, two cargo molecules with different binding specificities.
  • the heteromultimer is itself bispecific formed by attaching to the transporter polypeptides, at least two cargo molecules with different specificities.
  • multispecific molecule or “heterospecific molecule” is intended to include any agent, e.g., a protein, peptide, or protein or peptide complex, which has more than two different binding specificities.
  • the molecule may bind to, or interact with, (a) a cell surface target molecule such as but not limited to cell surface antigens, (b) an Fc receptor on the surface of an effector cell, and (c) at least one other component.
  • a cell surface target molecule such as but not limited to cell surface antigens
  • Fc receptor on the surface of an effector cell
  • at least one other component e.g., cell surface target molecule such as but not limited to cell surface antigens, an Fc receptor on the surface of an effector cell, and (c) at least one other component.
  • embodiments of the heteromultimers described herein are inclusive of, but not limited to, bispecific, trispecific, tetraspecific, and other multispecific molecules.
  • these molecules are directed to cell surface antigens, such as
  • the expression “multivalent” is used throughout this specification to denote a heteromultimer comprising at least two sites of attachment for target molecules.
  • the multivalent heteromultimer is designed to have multiple binding sites for desired targets.
  • the binding sites are on at least one cargo molecules attached to a transporter polypeptide.
  • at least one binding site is on a transporter polypeptide.
  • the expression “bivalent” is used throughout this specification to denote a heteromultimer comprising two target binding sites. In certain embodiments of a bivalent heteromultimer, both binding sites are on the same monomer.
  • the expression “trivalent” is used throughout this specification to denote a heteromultimer comprising three target binding sites.
  • the expression “tetravalent” is used throughout this specification to denote a heteromultimer comprising four target binding sites.
  • Fusion proteins and polypeptides are created by joining two or more genes that originally code for separate polypeptides. Translation of this fusion gene results in a single polypeptide with functional properties derived from each of the original polypeptides.
  • at least one monomer may comprise a fusion protein formed by the fusion of at least one cargo polypeptide to the N- or C-terminus of a transporter polypeptide.
  • substantially purified refers to a heteromultimer described herein, or variant thereof that may be substantially or essentially free of components that normally accompany or interact with the protein as found in its naturally occurring environment, i.e. a native cell, or host cell in the case of recombinantly produced heteromultimer that in certain embodiments, is substantially free of cellular material includes preparations of protein having less than about 30%, less than about 25%, less than about 20%, less than about 15%, less than about 10%, less than about 5%, less than about 4%, less than about 3%, less than about 2%, or less than about 1% (by dry weight) of contaminating protein.
  • the protein in certain embodiments is present at about 30%, about 25%, about 20%, about 15%, about 10%, about 5%, about 4%, about 3%, about 2%, or about 1% or less of the dry weight of the cells.
  • the protein in certain embodiments, is present in the culture medium at about 5 g/L, about 4 g/L, about 3 g/L, about 2 g/L, about 1 g/L, about 750 mg/L, about 500 mg/L, about 250 mg/L, about 100 mg/L, about 50 mg/L, about 10 mg/L, or about 1 mg/L or less of the dry weight of the cells.
  • “substantially purified” heteromultimer produced by the methods described herein has a purity level of at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, specifically, a purity level of at least about 75%, 80%, 85%, and more specifically, a purity level of at least about 90%, a purity level of at least about 95%, a purity level of at least about 99% or greater as determined by appropriate methods such as SDS/PAGE analysis, RP-HPLC, SEC, and capillary electrophoresis.
  • a “recombinant host cell” or “host cell” refers to a cell that includes an exogenous polynucleotide, regardless of the method used for insertion, for example, direct uptake, transduction, f-mating, or other methods known in the art to create recombinant host cells.
  • the exogenous polynucleotide may be maintained as a nonintegrated vector, for example, a plasmid, or alternatively, may be integrated into the host genome.
  • the term “medium” or “media” includes any culture medium, solution, solid, semi-solid, or rigid support that may support or contain any host cell, including bacterial host cells, yeast host cells, insect host cells, plant host cells, eukaryotic host cells, mammalian host cells, CHO cells, prokaryotic host cells, E. coli , or Pseudomonas host cells, and cell contents.
  • the term may encompass medium in which the host cell has been grown, e.g., medium into which the protein has been secreted, including medium either before or after a proliferation step.
  • the term also may encompass buffers or reagents that contain host cell lysates, such as in the case where a heteromultimer described herein is produced intracellularly and the host cells are lysed or disrupted to release the heteromultimer.
  • Refolding describes any process, reaction or method which transforms disulfide bond containing polypeptides from an improperly folded or unfolded state to a native or properly folded conformation with respect to disulfide bonds.
  • Cofolding refers specifically to refolding processes, reactions, or methods which employ at least two monomeric polypeptides which interact with each other and result in the transformation of unfolded or improperly folded polypeptides to native, properly folded polypeptides.
  • the term “modulated serum half-life” means the positive or negative change in circulating half-life of a cargo polypeptide that is comprised by a heteromultimer described herein relative to its native form. Serum half-life is measured by taking blood samples at various time points after administration of heteromultimer, and determining the concentration of that molecule in each sample. Correlation of the serum concentration with time allows calculation of the serum half-life. Increased serum half-life desirably has at least about two-fold, but a smaller increase may be useful, for example where it enables a satisfactory dosing regimen or avoids a toxic effect. In some embodiments, the increase is at least about three-fold, at least about five-fold, or at least about ten-fold.
  • modulated therapeutic half-life means the positive or negative change in the half-life of the therapeutically effective amount of a cargo polypeptide comprised by a heteromultimer described herein, relative to its non-modified form.
  • Therapeutic half-life is measured by measuring pharmacokinetic and/or pharmacodynamic properties of the molecule at various time points after administration. Increased therapeutic half-life desirably enables a particular beneficial dosing regimen, a particular beneficial total dose, or avoids an undesired effect.
  • the increased therapeutic half-life results from increased potency, increased or decreased binding of the modified molecule to its target, increased or decreased breakdown of the molecule by enzymes such as proteases, or an increase or decrease in another parameter or mechanism of action of the non-modified molecule or an increase or decrease in receptor-mediated clearance of the molecule.
  • isolated when applied to a nucleic acid or protein, denotes that the nucleic acid or protein is free of at least some of the cellular components with which it is associated in the natural state, or that the nucleic acid or protein has been concentrated to a level greater than the concentration of its in vivo or in vitro production. It can be in a homogeneous state. Isolated substances can be in either a dry or semi-dry state, or in solution, including but not limited to, an aqueous solution. It can be a component of a pharmaceutical composition that comprises additional pharmaceutically acceptable carriers and/or excipients. Purity and homogeneity are typically determined using analytical chemistry techniques such as polyacrylamide gel electrophoresis or high performance liquid chromatography.
  • a protein which is the predominant species present in a preparation is substantially purified.
  • an isolated gene is separated from open reading frames which flank the gene and encode a protein other than the gene of interest.
  • the term “purified” denotes that a nucleic acid or protein gives rise to substantially one band in an electrophoretic gel. Particularly, it may mean that the nucleic acid or protein is at least 85% pure, at least 90% pure, at least 95% pure, at least 99% or greater pure.
  • nucleic acid refers to deoxyribonucleotides, deoxyribonucleosides, ribonucleosides, or ribonucleotides and polymers thereof in either single- or double-stranded form. Unless specifically limited, the term encompasses nucleic acids containing known analogues of natural nucleotides which have similar binding properties as the reference nucleic acid and are metabolized in a manner similar to naturally occurring nucleotides. Unless specifically limited otherwise, the term also refers to oligonucleotide analogs including PNA (peptidonucleic acid), analogs of DNA used in antisense technology (phosphorothioates, phosphoroamidates, and the like).
  • PNA peptidonucleic acid
  • analogs of DNA used in antisense technology phosphorothioates, phosphoroamidates, and the like.
  • nucleic acid sequence also implicitly encompasses conservatively modified variants thereof (including but not limited to, degenerate codon substitutions) and complementary sequences as well as the sequence explicitly indicated.
  • degenerate codon substitutions may be achieved by generating sequences in which the third position of one or more selected (or all) codons is substituted with mixed-base and/or deoxyinosine residues (Batzer et al., Nucleic Acid Res. 19:5081 (1991); Ohtsuka et al., J. Biol. Chem. 260:2605-2608 (1985); Rossolini et al., Mol. Cell. Probes 8:91-98 (1994)).
  • polypeptide “peptide” and “protein” are used interchangeably herein to refer to a polymer of amino acid residues. That is, a description directed to a polypeptide applies equally to a description of a peptide and a description of a protein, and vice versa.
  • the terms apply to naturally occurring amino acid polymers as well as amino acid polymers in which one or more amino acid residues is a non-naturally encoded amino acid.
  • the terms encompass amino acid chains of any length, including full length proteins, wherein the amino acid residues are linked by covalent peptide bonds.
  • amino acid refers to naturally occurring and non-naturally occurring amino acids, as well as amino acid analogs and amino acid mimetics that function in a manner similar to the naturally occurring amino acids.
  • Naturally encoded amino acids are the 20 common amino acids (alanine, arginine, asparagine, aspartic acid, cysteine, glutamine, glutamic acid, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, praline, serine, threonine, tryptophan, tyrosine, and valine) and pyrrolysine and selenocysteine.
  • Amino acid analogs refers to compounds that have the same basic chemical structure as a naturally occurring amino acid, i.e., an a carbon that is bound to a hydrogen, a carboxyl group, an amino group, and an R group, such as, homoserine, norleucine, methionine sulfoxide, methionine methyl sulfonium.
  • Such analogs have modified R groups (such as, norleucine) or modified peptide backbones, but retain the same basic chemical structure as a naturally occurring amino acid.
  • Reference to an amino acid includes, for example, naturally occurring proteogenic L-amino acids; D-amino acids, chemically modified amino acids such as amino acid variants and derivatives; naturally occurring non-proteogenic amino acids such as ⁇ -alanine, ornithine, etc.; and chemically synthesized compounds having properties known in the art to be characteristic of amino acids.
  • non-naturally occurring amino acids include, but are not limited to, ⁇ -methyl amino acids (e.g.
  • D-amino acids D-amino acids
  • histidine-like amino acids e.g., 2-amino-histidine, ⁇ -hydroxy-histidine, homohistidine
  • amino acids having an extra methylene in the side chain (“homo” amino acids)
  • amino acids having an extra methylene in the side chain (“homo” amino acids)
  • amino acids having an extra methylene in the side chain (“homo” amino acids)
  • amino acids in which a carboxylic acid functional group in the side chain is replaced with a sulfonic acid group e.g., cysteic acid.
  • the incorporation of non-natural amino acids, including synthetic non-native amino acids, substituted amino acids, or one or more D-amino acids into the proteins of the present invention may be advantageous in a number of different ways.
  • D-amino acid-containing peptides, etc. exhibit increased stability in vitro or in vivo compared to L-amino acid-containing counterparts.
  • the construction of peptides, etc., incorporating D-amino acids can be particularly useful when greater intracellular stability is desired or required.
  • D-peptides, etc. are resistant to endogenous peptidases and proteases, thereby providing improved bioavailability of the molecule, and prolonged lifetimes in vivo when such properties are desirable.
  • D-peptides, etc. cannot be processed efficiently for major histocompatibility complex class II-restricted presentation to T helper cells, and are therefore, less likely to induce humoral immune responses in the whole organism.
  • Amino acids may be referred to herein by either their commonly known three letter symbols or by the one-letter symbols recommended by the IUPAC-IUB Biochemical Nomenclature Commission. Nucleotides, likewise, may be referred to by their commonly accepted single-letter codes.
  • “Conservatively modified variants” applies to both amino acid and nucleic acid sequences. With respect to particular nucleic acid sequences, “conservatively modified variants” refers to those nucleic acids which encode identical or essentially identical amino acid sequences, or where the nucleic acid does not encode an amino acid sequence, to essentially identical sequences. Because of the degeneracy of the genetic code, a large number of functionally identical nucleic acids encode any given protein. For instance, the codons GCA, GCC, GCG and GCU all encode the amino acid alanine. Thus, at every position where an alanine is specified by a codon, the codon can be altered to any of the corresponding codons described without altering the encoded polypeptide.
  • nucleic acid variations are “silent variations,” which are one species of conservatively modified variations. Every nucleic acid sequence herein which encodes a polypeptide also describes every possible silent variation of the nucleic acid.
  • each codon in a nucleic acid except AUG, which is ordinarily the only codon for methionine, and TGG, which is ordinarily the only codon for tryptophan
  • TGG which is ordinarily the only codon for tryptophan
  • amino acid sequences one of ordinary skill in the art will recognize that individual substitutions, deletions or additions to a nucleic acid, peptide, polypeptide, or protein sequence which alters, adds or deletes a single amino acid or a small percentage of amino acids in the encoded sequence is a “conservatively modified variant” where the alteration results in the deletion of an amino acid, addition of an amino acid, or substitution of an amino acid with a chemically similar amino acid. Conservative substitution tables providing functionally similar amino acids are known to those of ordinary skill in the art. Such conservatively modified variants are in addition to and do not exclude polymorphic variants, interspecies homologs, and alleles of the invention.
  • nucleic acids or polypeptide sequences refer to two or more sequences or subsequences that are the same. Sequences are “substantially identical” if they have a percentage of amino acid residues or nucleotides that are the same (i.e., about 60% identity, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, or about 95% identity over a specified region), when compared and aligned for maximum correspondence over a comparison window, or designated region as measured using one of the following sequence comparison algorithms (or other algorithms available to persons of ordinary skill in the art) or by manual alignment and visual inspection. This definition also refers to the complement of a test sequence.
  • the identity can exist over a region that is at least about 50 amino acids or nucleotides in length, or over a region that is 75-100 amino acids or nucleotides in length, or, where not specified, across the entire sequence of a polynucleotide or polypeptide.
  • a polynucleotide encoding a polypeptide of the present invention, including homologs from species other than human, may be obtained by a process comprising the steps of screening a library under stringent hybridization conditions with a labeled probe having a polynucleotide sequence of the invention or a fragment thereof, and isolating full-length cDNA and genomic clones containing said polynucleotide sequence. Such hybridization techniques are well known to the skilled artisan.
  • sequence comparison typically one sequence acts as a reference sequence, to which test sequences are compared.
  • test and reference sequences are entered into a computer, subsequence coordinates are designated, if necessary, and sequence algorithm program parameters are designated. Default program parameters can be used, or alternative parameters can be designated.
  • sequence comparison algorithm then calculates the percent sequence identities for the test sequences relative to the reference sequence, based on the program parameters.
  • a “comparison window”, as used herein, includes reference to a segment of any one of the number of contiguous positions selected from the group consisting of from 20 to 600, usually about 50 to about 200, more usually about 100 to about 150 in which a sequence may be compared to a reference sequence of the same number of contiguous positions after the two sequences are optimally aligned.
  • Methods of alignment of sequences for comparison are known to those of ordinary skill in the art.
  • Optimal alignment of sequences for comparison can be conducted, including but not limited to, by the local homology algorithm of Smith and Waterman (1970) Adv. Appl. Math. 2:482c, by the homology alignment algorithm of Needleman and Wunsch (1970) J. Mol. Biol.
  • BLAST and BLAST 2.0 algorithms are described in Altschul et al. (1997) Nuc. Acids Res. 25:3389-3402, and Altschul et al. (1990) J. Mol. Biol. 215:403-410, respectively.
  • Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information available at the World Wide Web at ncbi.nlm.nih.gov.
  • the BLAST algorithm parameters W, T, and X determine the sensitivity and speed of the alignment.
  • W wordlength
  • E expectation
  • B BLOSUM62 scoring matrix
  • the BLAST algorithm also performs a statistical analysis of the similarity between two sequences (see, e.g., Karlin and Altschul (1993) Proc. Natl. Acad. Sci. USA 90:5873-5787).
  • One measure of similarity provided by the BLAST algorithm is the smallest sum probability (P(N)), which provides an indication of the probability by which a match between two nucleotide or amino acid sequences would occur by chance.
  • P(N) the smallest sum probability
  • a nucleic acid is considered similar to a reference sequence if the smallest sum probability in a comparison of the test nucleic acid to the reference nucleic acid is less than about 0.2, or less than about 0.01, or less than about 0.001.
  • the phrase “selectively (or specifically) hybridizes to” refers to the binding, duplexing, or hybridizing of a molecule only to a particular nucleotide sequence under stringent hybridization conditions when that sequence is present in a complex mixture (including but not limited to, total cellular or library DNA or RNA).
  • stringent hybridization conditions refers to hybridization of sequences of DNA, RNA, or other nucleic acids, or combinations thereof under conditions of low ionic strength and high temperature as is known in the art. Typically, under stringent conditions a probe will hybridize to its target subsequence in a complex mixture of nucleic acid (including but not limited to, total cellular or library DNA or RNA) but does not hybridize to other sequences in the complex mixture. Stringent conditions are sequence-dependent and will be different in different circumstances. Longer sequences hybridize specifically at higher temperatures.
  • the term “eukaryote” refers to organisms belonging to the phylogenetic domain Eucarya such as animals (including but not limited to, mammals, insects, reptiles, birds, etc.), ciliates, plants (including but not limited to, monocots, dicots, algae, etc.), fungi, yeasts, flagellates, microsporidia, protists, etc.
  • prokaryote refers to prokaryotic organisms.
  • a non-eukaryotic organism can belong to the Eubacteria (including but not limited to, Escherichia coli, Thermus thermophilus, Bacillus stearothermophilus, Pseudomonas fluorescens, Pseudomonas aeruginosa, Pseudomonas putida , etc.) phylogenetic domain, or the Archaea (including but not limited to, Methanococcus jannaschii, Methanobacterium thermoautotrophicum, Halobacterium such as Haloferax volcanii and Halobacterium species NRC-1, Archaeoglobus fulgidus, Pyrococcus furiosus, Pyrococcus horikoshii, Aeuropyrum pernix , etc.) phylogenetic domain.
  • Eubacteria including but not limited to, Escherichia coli, Thermus thermophil
  • subject refers to an animal, in some embodiments a mammal, and in other embodiments a human, who is the object of treatment, observation or experiment.
  • An animal may be a companion animal (e.g., dogs, cats, and the like), farm animal (e.g., cows, sheep, pigs, horses, and the like) or a laboratory animal (e.g., rats, mice, guinea pigs, and the like).
  • compositions containing the heteromultimer described herein can be administered for prophylactic, enhancing, and/or therapeutic treatments.
  • an “enhance” or “enhancing” means to increase or prolong either in potency or duration a desired effect.
  • the term “enhancing” refers to the ability to increase or prolong, either in potency or duration, the effect of other therapeutic agents on a system.
  • An “enhancing-effective amount,” as used herein, refers to an amount adequate to enhance the effect of another therapeutic agent in a desired system. When used in a patient, amounts effective for this use will depend on the severity and course of the disease, disorder or condition, previous therapy, the patient's health status and response to the drugs, and the judgment of the treating physician.
  • modified refers to any changes made to a given polypeptide, such as changes to the length of the polypeptide, the amino acid sequence, chemical structure, co-translational modification, or post-translational modification of a polypeptide.
  • modified means that the polypeptides being discussed are optionally modified, that is, the polypeptides under discussion can be modified or unmodified.
  • post-translationally modified refers to any modification of a natural or non-natural amino acid that occurs to such an amino acid after it has been incorporated into a polypeptide chain.
  • the term encompasses, by way of example only, co-translational in vivo modifications, co-translational in vitro modifications (such as in a cell-free translation system), post-translational in vivo modifications, and post-translational in vitro modifications.
  • segmentation refers to a precise internal splice of the original protein sequence which results in “segments” of the protein sequence that preferentially associate as heteromultimers to form a quasi-protein.
  • quasi-native proteins and/or ‘quasi-native structures’ present the native protein like functional and structural characteristics.
  • Proteins are naturally dynamics molecules and display an ensemble of structural configurations although we ascribe a native structure to it, such as the one obtained by X-ray crystallography.
  • the alternate structural configurations observed in the ensemble of geometries of that protein can be deemed to be quasi-native structures relative to each other or relative to the structure observed in the crystal.
  • homologous proteins sequences or proteins belonging to common structural families tend to fold into similar structural geometries. The member proteins belonging to this family can be deemed to achieve a quasi-native structure relative to each other.
  • Some of the unique sequences in the protein family could also exhibit similar functional attributes and hence can be referred to as quasi-native proteins relative to each other.
  • the transporter polypeptides assemble to form a quasi-native structure.
  • the reference native protein in this case is the protein from which the transporter polypeptide is derived and the reference native structure is the structure of the protein from which the transporter polypeptide is derived.
  • transporter polypeptide or “transporter polypeptide” or “transporter peptide” or “transporter peptide” or “transporter” refers to a polypeptide, such that said transporter polypeptide is capable of forming heteromultimeric proteins with other such transporter polypeptides in solution, and wherein said heteromultimeric proteins have a quasi-native structure of a monomeric protein from which at least one transporter polypeptide is derived.
  • all transporter polypeptides are derived from the same albumin or alloalbumin protein.
  • the heteromultimers are formed by transporter polypeptides derived from various albumin and alloalbumin proteins.
  • the transporter polypeptides are derived from transferrin. In certain embodiments of the heteromultimers described herein, all transporter polypeptides are derived from annexin proteins. In certain embodiments, the heteromultimers are formed by transporter polypeptides derived from the same annexin protein. In some embodiments, the heteromultimers are formed by transporter polypeptides derived from different annexin proteins. In an embodiment, the heteromultimers are formed by transporter polypeptides derived from annexin A2.
  • transporter polypeptides are segments of a whole protein, wherein said segments are capable of assembling to form a heteromultimer.
  • the transporter polypeptides are segments derived from a coiled coil protein.
  • the transporter polypeptides are segments derived from a leucine-zipper protein.
  • the transporter polypeptides are segments from a beta-barrel protein.
  • transporter polypeptides are segments obtained from a beta-propeller protein.
  • the transporter polypeptides are segments obtained from a helical bundle protein.
  • the transporter polypeptides are generated from for instance, but not restricted to proteins comprising a zinc finger motif, a helix-turn-helix motif or a beta-hairpin motif.
  • the transporter polypeptides are segments obtained from non-immunogenic proteins that are structurally stable, and have favorable biological properties.
  • albumin refers collectively to albumin protein or amino acid sequence, or an albumin segment or variant, having one or more functional activities (e.g., biological activities) of albumin.
  • albumin refers to human albumin or segments thereof (see for example, EP 201239, EP 322 094 WO 97/24445, WO95/23857) especially the mature form of human albumin as shown in FIG. 1 , or albumin from other vertebrates, or segments thereof, or analogs or variants of these molecules or fragments thereof.
  • albumin refers to a truncated version of albumin.
  • the term “quasi-albumin” refers to a heteromultimer molecule that has structure and/or function similar to the whole albumin, and wherein said heteromultimer molecule is formed by the assembly of two or more monomeric polypeptides designed based on the sequence of the whole albumin.
  • the monomeric polypeptides are “segments” that preferentially associate as heteromultimeric pairs to form a quasi-protein.
  • the quasi-albumin has 90% of the activity of the whole albumin.
  • the quasi-albumin has 75% of the activity of whole-albumin.
  • the quasi-albumin has 50% of the activity of whole albumin.
  • the quasi-albumin has 50-75% of the activity of whole albumin.
  • quasi-albumin has 80% of the activity of whole albumin. In some embodiments, the quasi-albumin has 90% of the structure of whole albumin as determined by molecular modeling. In some embodiments, the quasi-albumin has 80% of the structure of whole albumin as determined by molecular modeling. In some embodiments, the quasi-albumin has 70% of the structure of whole albumin as determined by molecular modeling. In some embodiments, the quasi-albumin has 50% of the structure of whole albumin as determined by molecular modeling. In some embodiments, the quasi-albumin has 50%-75% of the structure of whole albumin as determined by molecular modeling.
  • human serum albumin and human albumin (HA) are used interchangeably herein.
  • albumin and serum albumin are broader, and encompass human serum albumin (and fragments and variants thereof) as well as albumin from other species (and fragments and variants thereof).
  • each albumin-based monomer of the heteromultimeric proteins described herein is based on a variant of normal HA.
  • Each cargo polypeptide portion of the heteromultimeric proteins of the invention may also be variants of the Therapeutic proteins as described herein.
  • variants includes insertions, deletions and substitutions, either conservative or non conservative, where such changes do not substantially alter one or more of the oncotic, useful ligand-binding and non-immunogenic properties of albumin, or the active site, or active domain which confers the therapeutic activities of the Therapeutic proteins.
  • heteromultimeric proteins described herein include naturally occurring polymorphic variants of human albumin and fragments of human albumin, for example those fragments disclosed in EP 322 094 (namely HA (Pn), where n is 369 to 419).
  • the albumin is derived from any vertebrate, especially any mammal that includes but is not limited to human, cow, sheep, rat, mouse, rabbit, horse, dog or pig. In certain embodiments, the albumin is derived from non-mammalian albumins including, but are not limited to hen and salmon.
  • the sequence of human albumin is as shown:
  • An alloalbumin is a genetic variant of albumin.
  • the alloalbumin is human alloalbumin (HAA).
  • HAA human alloalbumin
  • Alloalbumins that differ in electrophoretic mobility from albumin have been identified through population genetics surveys in the course of clinical electrophoresis, or in blood donor surveys. As markers of mutation and migration, alloalbumins are of interest to geneticists, biochemists, and anthropologists, but most of these alloalbumin are not associated with disease (Minchioti et al. Human Mutations 29(8), 1007-1016(2008)).
  • annexin refers to a group of cellular proteins found in eukaryotic organisms. Annexin is also known as lipocortin. As used herein “annexin” may refer to any annexin protein, or to specific annexin proteins such as “annexin A1,” “annexin A2,” and “annexin A5.” Annexins are characterized by their calcium dependent ability to bind negatively charged phospholipids (i.e. membrane walls). Annexins are characterized by a repeat protein scaffold limited to 30-50 kDa in size with fairly ubiquitous tissue distribution. The basic structure of an annexin is composed of two domains: a structurally conserved C terminal “core” region and a divergent N terminal domain.
  • the core region binds the phospholipid cellular membrane in a Ca 2+ dependent manner.
  • the N terminal region binds cytoplasmic proteins.
  • Annexins are important in various cellular and physiological processes and provide a membrane scaffold.
  • the C terminal core is composed of four annexin repeats.
  • Annexin is characterized by its flexible repeat-like nature that influences its intrinsic membrane-sensing abilities. For instance, the affinity towards specific biomembranes can be controlled by the number of repeats. With the characteristic phospholipid sensing, annexin can be useful to sense/target intestinal junctions for drug delivery.
  • Another potential application for an annexin is targeting intestinal tight junctions and the Zonula Occludens region (ZO-1), which is known to be particularly difficult to traverse for larger protein therapeutics, significantly impairing drug absorption.
  • ZO-1 Zonula Occludens region
  • the term “quasi-annexin” refers to a heteromultimer molecule that has structure and/or function similar to the whole annexin, and wherein said heteromultimer molecule is formed by the assembly of two or more monomeric polypeptides designed based on the sequence of the whole annexin.
  • the monomeric polypeptides are “segments” that preferentially associate as heteromultimeric pairs to form a quasi-protein.
  • the quasi-annexin has 90% of the activity of the whole annexin.
  • the quasi-annexin has 75% of the activity of whole-annexin.
  • the quasi-annexin has 50% of the activity of whole annexin.
  • the quasi-annexin has 50-75% of the activity of whole annexin. In an embodiment, quasi-annexin has 80% of the activity of whole annexin. In some embodiments, the quasi-annexin has 90% of the structure of whole annexin as determined by molecular modeling. In some embodiments, the quasi-annexin has 80% of the structure of whole annexin as determined by molecular modeling. In some embodiments, the quasi-annexin has 70% of the structure of whole annexin as determined by molecular modeling. In some embodiments, the quasi-annexin has 50% of the structure of whole annexin as determined by molecular modeling. In some embodiments, the quasi-annexin has 50%-75% of the structure of whole annexin as determined by molecular modeling.
  • SEQ ID NO: 14 GSAVSPYPTFNPSSDVAALHKAIMVKGVDEATIIDILTKRNNAQRQQIKA AYLQETGKPLDETLKKALTGHLEEVVLALLKTPAQFDADELRAAMKGLGT DEDTLIEILASRTNKEIRDINRVYREELKRDLAKDITSDTSGDFRNALLS LAKGDRSEDFGVNEDLADSDARALYEAGERRKGTDVNVFNTILTTRSYPQ LRRVFQKYTKYSKHDMNKVLDLELKGDIEKCLTAIVKCATSKPAFFAEKL HQAMKGVGTRHKALIRIMVSRSEIDMNDIKAFYQKMYGISLCQAILDETK GDYEKILVALCGGN
  • Transferrins are monomeric proteins of about 76 kDa molecular weight present in all vertebrates and function as a iron-binding and transporting protein. Recombinant human transferrin and its fusions is being considered for the management of various diseases including thalassemia, atransferrinemia, age related macular degeneration, type 2 diabetes, during stem cell transplantation and in the treatment of acute infectious disease caused by the anthrax bacteria. Transferrin is stable in the gastrointestinal environment and a number of studies have shown that intact protein-transferrin conjugates can be orally delivered and remain bioactive.
  • the term “quasi-transferrin” refers to a heteromultimer molecule that has structure and/or function similar to the whole transferrin, and wherein said heteromultimer molecule is formed by the assembly of two or more monomeric polypeptides designed based on the sequence of the whole transferrin.
  • the monomeric polypeptides are “segments” that preferentially associate as heteromultimeric pairs to form a quasi-protein.
  • the quasi-transferrin has 90% of the activity of the whole transferrin.
  • the quasi-transferrin has 75% of the activity of whole-transferrin.
  • the quasi-transferrin has 50% of the activity of whole transferrin.
  • the quasi-transferrin has 50-75% of the activity of whole transferrin. In an embodiment, quasi-transferrin has 80% of the activity of whole transferrin. In some embodiments, the quasi-transferrin has 90% of the structure of whole transferrin as determined by molecular modeling. In some embodiments, the quasi-transferrin has 80% of the structure of whole transferrin as determined by molecular modeling. In some embodiments, the quasi-transferrin has 70% of the structure of whole transferrin as determined by molecular modeling. In some embodiments, the quasi-transferrin has 50% of the structure of whole transferrin as determined by molecular modeling. In some embodiments, the quasi-transferrin has 50%-75% of the structure of whole transferrin as determined by molecular modeling.
  • a heteromultimer described herein comprises monomers that comprise at least one cargo molecule, and at least one transporter polypeptide, said cargo molecule and transporter polypeptide associated with one another, by means inclusive of, but not restricted to genetic fusion or chemical conjugation.
  • at least one cargo molecule is a therapeutic agent.
  • the cargo molecule is a toxin.
  • the cargo molecule is an antigen, or analogs thereof.
  • the cargo molecule is a natural product, analog, or prodrug thereof.
  • the cargo molecule is a therapeutic agent such as a cytotoxin, e.g., a cytostatic or cytocidal agent, a therapeutic agent or a radioactive metal ion, e.g., alpha-emitters such as, for example, 213Bi.
  • a cytotoxin or cytotoxic agent includes any agent that is detrimental to cells.
  • Examples include paclitaxol, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicin, doxorubicin, daunorubicin, dihydroxy anthracin dione, mitoxantrone, mithramycin, actinomycin D, 1-dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, and puromycin and analogs or homologs thereof.
  • Therapeutic agents include, but are not limited to, antimetabolites (e.g., methotrexate, 6mercaptopurine, 6thioguanine, cytarabine, 5-fluorouracil decarbazine), alkylating agents (e.g., mechlorethamine, thioepa chlorambucil, melphalan, carmustine (BSNU) and lomustine (CCNU), cyclothosphamide, busulfan, dibromomannitol, streptozotocin, mitomycin C, and cis-dichlorodiamine platinum (II) (DDP) cisplatin), anthracyclines (e.g., daunorubicin (formerly daunomycin) and doxorubicin), antibiotics (e.g., dactinomycin (formerly actinomycin), bleomycin, mithramycin, and anthramycin (AMC)), and anti-mitotic agents (e.g.
  • the cargo molecule is a biomolecule.
  • the cargo molecule is a natural or synthetic nucleic acid.
  • at least one cargo molecule is one or more of a DNA, PNA, and/or RNA oligomer.
  • a heteromultimer described herein comprises monomeric proteins that comprise at least one cargo polypeptide, or fragments or variants thereof, and at least one transporter polypeptide, said cargo polypeptide and transporter polypeptide associated with one another, by means inclusive of, but not restricted to genetic fusion or chemical conjugation
  • Cargo polypeptide refers to proteins, polypeptides, antibodies, peptides or fragments or variants thereof, having one or more therapeutic and/or biological activities.
  • Cargo polypeptides encompassed by the invention include but are not limited to, proteins, polypeptides, peptides, antibodies, substrates or ligands to therapeutically relevant target proteins and biologics. (The terms peptides, proteins, and polypeptides are used interchangeably herein.)
  • Cargo polypeptide encompasses antibodies and fragments and variants thereof.
  • a heteromultimer described herein may contain at least a fragment or variant of a cargo polypeptide, and/or at least a fragment or variant of an antibody.
  • the term “Cargo polypeptide” refers to the endogenous or naturally occurring correlate of a cargo polypeptide.
  • a “Cargo biomolecule” is a biomolecule such as but not restricted to a protein, DNA, or RNA that is useful to treat, prevent or ameliorate a disease, condition or disorder.
  • a “Cargo polypeptide” may be one that binds specifically to a particular cell type (normal (e.g., lymphocytes) or abnormal e.g., (cancer cells)) and therefore may be used to target a compound (drug, or cytotoxic agent) to that cell type specifically.
  • a “Cargo molecule” is a molecule that has a biological, activity, and in particular, a biological activity that is useful for treating preventing or ameliorating a disease.
  • a non-inclusive list of biological activities that may be possessed by a Cargo molecule, for instance a Cargo polypeptide includes, enhancing the immune response, promoting angiogenesis, inhibiting angiogenesis, regulating hematopoietic functions, stimulating nerve growth, enhancing an immune response, inhibiting an immune response, or any one or more of the biological activities described herein.
  • Cargo polypeptides corresponding to a cargo polypeptide portion of a heteromultimer protein described herein, such as cell surface and secretory proteins, are often modified, by the attachment of one or more oligosaccharide groups.
  • the modification referred to as glycosylation, can dramatically affect the physical properties of proteins and can be important in protein stability, secretion, and localization. Glycosylation occurs at specific locations along the polypeptide backbone.
  • glycosylation characterized by O-linked oligosaccharides, which are attached to serine or threonine residues; and glycosylation characterized by N-linked oligosaccharides, which are attached to asparagine residues in an Asn-X-Ser/Thr sequence, where X can be any amino acid except proline.
  • N-acetylneuramic acid also known as sialic acid
  • Variables such as protein structure and cell type influence the number and nature of the carbohydrate units within the chains at different glycosylation sites. Glycosylation isomers are also common at the same site within a given cell type.
  • Table 2 provides a non-exhaustive list of Cargo polypeptides that correspond to a Cargo polypeptide portion of a heteromultimer described herein.
  • the “Cargo Polypeptide” column discloses Cargo polypeptide molecules followed by parentheses containing scientific and brand names that comprise, or alternatively consist of, that Cargo polypeptide molecule or a fragment or variant thereof.
  • the cargo molecule is a molecule that binds to a protein disclosed in the “Cargo polypeptide” column, or in Zhu et al. (Nucleic Acids Res. 38(1), D787-D791 (2009)); Wishart et al. (Nucleic Acids Res 36, D901-D906 (2008)); Ahmed et al. (Nucleic Acids Res 39, D960-D967 (2011)) incorporated by reference herein, or a protein that belongs in the class of therapeutic target molecules.
  • Cargo polypeptide as used herein may refer either to an individual Cargo polypeptide molecule (as defined by the amino acid sequence obtainable from the CAS and Genbank accession numbers), or to the entire group of Cargo polypeptide associated with a given Cargo polypeptide molecule disclosed in this column, or a Cargo polypeptide that binds to a polypeptide molecule disclosed in this column.
  • EPO Erythropoietin; Stimulates cellular Cell proliferation assay Anemia; Anemia in Renal Disease; Epoetin alfa; Epoetin differentiation of bone- using a erythroleukemic Anemia in Oncology Patients; beta; Gene- activated marrow stem cells at an cell line TF-1.
  • EPO Erythropoietin; Stimulates cellular Cell proliferation assay Anemia; Anemia in Renal Disease; Epoetin alfa; Epoetin differentiation of bone- using a erythroleukemic Anemia in Oncology Patients; beta; Gene- activated marrow stem cells at an cell line TF-1.
  • G-CSF G-CSF (Granulocyte Stimulates the Proliferation of murine Chemoprotection; Adjunct to colony- stimulating proliferation and NFS-60 cells (Weinstein Chemotherapy; Inflammatory factor; Granulokine; differentiation of the et al, Proc Natl Acad Sci disorders; Cancer; Leukemia; KRN 8601; Filgrastim; progenitor cells for USA 1986; 83, pp5010-4) Myelocytic leukemia; Neutropenia, Lenograstim; granulocytes and Primary neutropenias (e.g.; Meograstim; monocytes- Kostmann syndrome); Secondary Nartograstim; macrophages.
  • neutropenia Prevention of Neupogen; NOPIA; neutropenia; Prevention and Gran; GRANOCYTE; treatment of neutropenia in HIV- Granulokine; Neutrogin; infected patients; Prevention and Neu-up; Neutromax) treatment of neutropenia associated with chemotherapy; Infections associated with neutropenias; Myelopysplasia; Autoimmune disorders; Psoriasis; Mobilization of hematopoietic progenitor cells; Wound Healing; Autoimmune Disease; Transplants; Bone marrow transplants; Acute myelogeneous leukemia; Lymphoma, Non- Hodgkin's lymphoma; Acute lymphoblastic leukemia; Hodgkin's disease; Accelerated myeloid recovery; Glycogen storage disease.
  • GM-CSF Granulocyte- Regulates Colony Stimulating Bone Marrow Disorders; Bone macrophage colony- hematopoietic cell Assay: Testa, N. G., et marrow transplant; Chemoprotection; stimulating factor; differentiation, gene al., “Assays for Hepatitis C; HIV Infections; Cancer; rhuGM- CSF; BI 61012; expression, growth, and hematopoietic growth Lung Cancer; Melanoma; Malignant Prokine; Molgramostim; function.
  • Human growth hormone Binds to two GHR Ba/F3-hGHR Acromegaly; Growth failure; Growth (Pegvisamont; molecules and Induces proliferation assay, a hormone replacement; Growth Somatrem; Somatropin; signal transduction novel specific bioassay hormone deficiency; Pediatric TROVERT; through receptor for serum human growth Growth Hormone Deficiency; Adult PROTROPIN; BIO- dimerization hormone.
  • Insulin Human insulin; Stimulates glucose Insulin activity may be Hyperglycemia; Diabetes; Diabetes Insulin aspart; Insulin uptake and promotes assayed in vitro using a Insipidus; Diabetes mellitus; Type 1 Glargine; Insulin lispro; glycogenesis and [3-H]-glucose uptake diabetes; Type 2 diabetes; Insulin Lys-B28 Pro- B29; lipogenesis. assay.
  • Non- insulin dependent Diabetes B26- B30-insulin- B25- Mellitus NIDDM
  • Insulin- amide Insulin detemir
  • dependent Diabetes Mellitus LABI NOVOLIN
  • IDDM A Condition Associated NOVORAPID
  • HUMAHALE HUMAHALE; HUMAHALIN
  • Renal Cancer e.g., Renal consensus; recombinant synthetase
  • Lung Cancer e.g,. methionyl consensus expression of a GC-rich Non-Small Cell Lung Cancer or interferon; recombinant viral transcript. Mol Cell Small Cell Lung Cancer) Colon consensus interferon; Biol. 19(11): 7305-13.
  • Calcitonin (Salmon Regulates levels of Hypocalcemic Rat Bone Disorders; Fracture prevention; Calcitonin (Salcatonin); calcium and phosphate Bioassay, bone resorbing Hypercalcemia; Malignant Calcitonin human- in serum; causes a assay and the pit assay, hypercalcemia; Osteoporosis; Paget's salmon hybrid; reduction in serum CT receptor binding disease; Osteopenia, Forcaltonin; Fortical; calcium--an effect assay, CAMP stimulation Osteoclastogenesis; osteolysis; Calcitonin; Calcitonina opposite to that of assay: J Bone Miner Res osteomyelitis; osteonecrosis; Almirall; Calcitonina human parathyroid 1999 August; 14(8): periodontal bone loss; osteoarthritis; Rubber; Calcimar; hormone.
  • Growth hormone Acts on the anterior Growth hormone- Acromegaly; Growth failure; Growth releasing factor; Growth pituitary to stimulate releasing peptides hormone replacement; Growth hormone releasing the production and (GHRPs) are known to hormone deficiency; Pediatric hormone (Sermorelin secretion of growth release growth hormone Growth Hormone Deficiency; Adult acetate; Pralmorelin; hormone and exert a (GH) in vivo and in vitro Growth Hormone Deficiency; Somatorelin; trophic effect on the by a direct action on Idiopathic Growth Hormone Somatoliberin; Geref; gland. receptors in anterior Deficiency; Growth retardation; Gerel; Groliberin) pituitary cells.
  • GHRPs Growth hormone releasing the production and
  • Biological Prader- Willi Syndrome; Prader-Willi activity can be measured Syndrome in children 2 years or in cell lines expressing older; Growth deficiencies; Growth growth hormone failure associated with chronic renal releasing factor receptor insufficiency; Osteoporosis; (Mol Endocrinol 1992 Osteopenia, Osteoclastogenesis; October; 6(10): Postmenopausal osteoporosis; burns; 1734-44, Cachexia; Cancer Cachexia; Molecular Dwarfism; Metabolic Disorders; Endocrinology, Vol 7, Obesity; Renal failure; Turner's 77-84).
  • IL-2 (Aldesleukin; Promotes the growth of T cell proliferation assay Cancer; Solid Tumors; Metastatic interleukin-2 fusion B and T cells and “Biological activity of Renal Cell Carcinoma; Metastatic toxin; T cell growth augments NK cell and recombinant human Melanoma; Malignant Melanoma; factor; PROLEUKIN; CTL cell killing interleukin-2 produced in Melanoma; Renal Cell Carcinoma; IMMUNACE; activity.
  • Escherichia coli Escherichia coli .” Renal Cancer; Lung Cancer (e.g,. CELEUK; ONCOLIPIN Science 223: 1412-1415, Non-Small Cell Lung Cancer or 2; MACROLIN) 1984. natural killer (NK) Small Cell Lung Cancer); Colon cell and CTL cytotoxicity Cancer; Breast Cancer; Liver Cancer; assay “Control of Leukemia; Preleukemia; homeostasis of CD8+ Hematological Malignancies; memory T cells by Hematological Disorders; Acute opposing cytokines.
  • Lung Cancer e.g,. CELEUK; ONCOLIPIN Science 223: 1412-1415, Non-Small Cell Lung Cancer or 2; MACROLIN
  • NK natural killer
  • Parathyroid hormone Acts in conjuction with Adenylyl cyclase Bone Disorders; Fracture prevention; parathyrin (PTH; calcitonin to control stimulation in rat Hypercalcemia; Malignant Ostabolin; ALX1-11; calcium and phosphate osteosarcoma cells, hypercalcemia; Osteoporosis; Paget's hPTH 1-34; LY 333334; metabolism; elevates ovariectomized rat disease; Osteopenia, MN 10T; parathyroid blood calcium level; model of Osteoclastogenesis; osteolysis; hormone (1-31); stimulates the activity osteoporosis: IUBMB osteomyelitis; osteonecrosis; FORTEO; of osteocytes; enhances Life 2000 Februray; periodontal bone loss; osteoarthritis; PARATHAR) absorption of Ca+/Pi 49(2): 131-5 rheumatoid arthritis; osteopetrosis; from small intestine periodontal
  • Resistin Mediates insulin Ability of resistin to Hyperglycemia; Diabetes; Diabetes resistance in Type II influence type II diabetes Insipidus; Diabetes mellitus; Type 1 diabetes; inhibits can be determined using diabetes; Type 2 diabetes; Insulin insulin-stimulated assays known in the art: resistance; Insulin deficiency; glucose uptake Pontoglio et al., J Clin Hyperlipidemia; Hyperketonemia; Invest 1998 May 15; Non- insulin dependent Diabetes 101(10): 2215-22.
  • NIDDM Insulin-dependent Diabetes Mellitus
  • IDDM Insulin- dependent Diabetes Mellitus
  • TR6 DcR3; Decoy Inhibits Fas Ligand and Cellular apoptosis can Fas Ligand or LIGHT induced Receptor 3; FASTR)
  • AIM-2 T5, LIGHT
  • TUNEL failure including fulminant liver staining, measurement of failure
  • graft versus host disease caspase levels.
  • Inhibition graft rejection myelodysplastic of cell growth can also be syndrome; renal failure; insulin directly measured, for dependent diabetes mellitus; example by ALOMAR rheumatoid arthritis; inflammatory Blue staining.
  • Assay refs bowel disease; autoimmune disease; cytotoxicity assay on toxic epidermal necrolysis; multiple human fibrosarcoma sclerosis. (Epsevik and Nissen- Meyer, 1986, J. Immunol. methods).
  • DeCAF (D- SLAM; Inhibits proliferation
  • DeCAF activity can be B cell and/or T cell mediated immune BCM-like membrane and differentiation of B determined using assays disorders; Immunodeficiency (e.g., protein; BLAME (B cells; Antagonize BLyS known in the art, such as Common Variable lymphocyte activator activity for example, those Immunodeficiency, Selective IgA macrophage described in Examples Deficiency) expressed))x 32-33 of International Publication No. WO0111046.
  • BLyS B Lymphocyte Promotes proliferation
  • BLyS activity can be B cell and/or T cell mediated immune Stimulator; Neutrokine differentiation and determined using assays disorders, particularly immune alpha; TL7; BAFF; survival of B cells; known in the art, such as, system disorders associated with low TALL-1; THANK; Promotes for example, the B cell numbers or low serum radiolabeled BLyS) immunoglobulin costimulatory immunoglobulin; Immunodeficiency production by B cells. proliferation assay and (e.g., Common Variable other assays disclosed by Immunodeficiency, Selective IgA Moore et al., 1999, Deficiency).
  • lymphoma non-Hodgkins lymphoma, chronic lymphocytic leukemia, multiple myeloma.
  • Anti-BLyS single chain Agonize or antagonize BLyS agonist or B cell and/or T cell mediated immune antibody (scFvI116A01, BlyS activity. antagonist activity can be disorders; Autoimmune disorders, scFvI050B11, determined using assays particularly autoimmune diseases scFvI006D08) and known in the art, such as, associated with the production of others.
  • a modified autoantibodies for example, a modified autoantibodies; Rheumatoid Arthritis, version the costimulatory Systemic Lupus Erythmatosus; proliferation assay Sjogren's Syndrome, cancers disclosed by Moore et al., expressing Blys as an autocrine 1999, Science, growth factor, e.g. certain chronic 285(5425): 260-3, in lymphocytic leukemias. which BlyS is mixed or preincubated with the anti-BlyS antibody prior to being applied to the responder B lymphocytes.
  • MPIF-1 Myeloid Inhibits myeloid MPIF-1 activity can be Chemoprotection; Adjunct to Progenitor Inhibitory progenitor cells; and measured using the Chemotherapy; Inflammatory Factor; CK beta-8; activates monocytes myeloprotection assay disorders; Cancer; Leukemia; Mirostipen) and chemotaxis assay Myelocytic leukemia; Neutropenia, described in U.S. Pat. No. Primary neutropenias (e.g.; 6,001,606.
  • KDI Keratinocyte Inhibits bone marrow KDI activity can be Multiple sclerosis; Hepatitis; Cancer; Derived Interferon; proliferation; and shows measured using the Viral infections, HIV infections, Interferon Kappa antiviral activity. antiviral and cell Leukemia. Precursor) proliferation assays described in Examples 57-63 of International Publication No. WO0107608.
  • TNFR2 p75 Binds both TNF ⁇ and T-cell proliferation can Autoimmune disease; Rheumatoid (ENBREL) TNF ⁇ ; mediates T-cell be measured using assays Arthritis; Psoriatic arthritis; Still's proliferation by TNF; known in the art.
  • Tissue Wounds Oral Cavity Wounds, Diabetic Ulcers, Dermal Ulcers, Cubitus Ulcers, Arterial Ulcers, Venous Stasis Ulcers, Burns Resulting From Heat Exposure Or Chemicals, or Other Abnormal Wound Healing Conditions such as Uremia, Malnutrition, Vitamin Deficiencies or Complications Associated With Systemic Treatment With Steroids, Radiation Therapy or Antineoplastic Drugs or Antimetabolites; Promote Dermal Reestablishment Subsequent To Dermal Loss; Increase the Adherence Of Skin Grafts To A Wound Bed; Stimulate Re-Epithelialization from The Wound Bed; To Promote Skin Strength; Improve The Appearance Of Aged Skin; Proliferate Hepatocytes, Lung, Breast, Pancreas, Stomach, Bladder, Small Intestine, Large Intestine; Sebocytes, Hair Follicles, Type II Pneumocytes, Mucin- Producing Goblet Cells, or Other Epithelial Cells,
  • TR2 (and TR2sv1, Inhibits B cell Co-stimulation B-cell Herpes; immune disorders; TR2SV2; TNFRSF14; proliferation, and proliferation assay and Ig autoimmune disease; graft versus HVEM; Herpes Virus mediates and inhibits production assay (Moore host disease; graft rejection; variable Entry Mediator; ATAR) Herpes Simplex Virus et al., 1999, Science, immunodeficiency; (HSV) infection. 285(5425): 260-3.). immunodeficiency syndromes; HSV-1 and HSV-2 cancer. Infectivity Assay: International Publication No.
  • WO 97/ Macrophage derived Chemotactic for 04658 Chemokine Inflammatory diseases; wound chemokine, MDC monocyte-derived activities can be healing; (Ckbeta-13) dendritic cells and IL-2- determined using assays angiogenesis; AIDS infection. activated natural killer known in the art: cells. Methods in Molecular Biology, 2000, vol. 138: Chemokine Protocols. Edited by: A. E. I. Proudfoot, T. N. C. Wells, and C. A. Power.
  • GnRH Gonadotropin Promotes release of GnRH is known to cause Infertility; Kallmann's syndrome or Releasing Hormone
  • follicle-stimulating the release of follicle other forms of hypergonadotropic hormone and stimulating hormone hypergonadism (failure to go through luteinizing hormone (FSH) and/or luteinizing puberty naturally).
  • FSH luteinizing hormone
  • LH anterior pituitary gonadotropes.
  • GnRH activity can be determined by measuring FSH levels in the medium of cultured gonadotropes before and after GnRH supplementation. For example, Baker et al.
  • Teprotide Inhibits angiotensin Inhibition of ACE can be Hypertension; congestive heart converting enzyme determined using assays failure. (ACE). known in the art. For example, Anzenbacherova et al., J. Pharma Biomed Anal 2001 March; 24(5-6): 1151-6.
  • Human chemokine Involved in Chemokine activities can Autoimmune disorders; Immunity; HCC-1 (ckBeta-1; inflammation, allergy, be determined using Vascular and Inflammatory disorders; HWFBD) tissue rejection, viral assays known in the art: HIV; AIDS; infectious diseases. infection, and tumor Methods in Molecular biology; enhances Biology, 2000, vol.
  • vasoconstriction impaired production, arteriolar
  • in vitro cardiac output and/or hypertension smooth muscle using a proliferation Cardiovascular Disease; Cardiac vasoconstriction, and assay with rat cardiac Failure; Diabetes; Type II Diabetes; proliferation of cardiac fibroblasts as described in Proteinuria; Renal disorders, fibroblasts, Induces Naunyn Schmiedebergs congestive heart failure.
  • angiogenesis an Arch Pharmacol 1999 enzyme that converts May; 359(5): 394-9. angiotensin I to angiotensin1-9; also cleaves des-Arg, bradykinin and neurotensin.
  • TR1 (OCIF; Inhibits Coculture Assay for Osteoporosis; Paget's disease; Osteoclastogenesis osteoclastogenesis and Osteoclastogenesis, Bone osteopenia; osteolysis; osteomyelitis; inhibitory factor; bone resorption, and resorption assay using osteonecrosis; periodontal bone loss; osteoprotegerin, OPG; induces fibroblast fetal long-bone organ osteoarthritis; rheumatoid arthritis; tumor necrosis factor proliferation.
  • osteoclast member 11B precursor fibroblast proliferation differentiation inhibition
  • bone assays are each described disorders; bone healing and in Kwon et al., FASEB J. regeneration; organ calcification; 12: 845-854 (1998). vascular calcification.
  • E. I. Syndrome Lung Inflammation; chemotaxis; inhibits Proudfoot, T. N. C. Fibrotic Disorders; Atherosclerosis; proliferation of myeloid Wells, and C. A. Power. Periodontal diseases; Viral diseases; progenitors; chemotaxis ⁇ Humana Press Inc., Hepatitis. of PBMC's.
  • Totowa NJ Leptin Controls obesity in vivo modulation of Hyperglycemia; Diabetes; Diabetes through regulation of food intake, reduction in Insipidus; Diabetes mellitus; Type 1 appetite, reduction of body weight, and diabetes; Type 2 diabetes; Insulin body weight, and lowering of insulin and resistance; Insulin deficiency; lowering of insulin and glucose levels in ob/ob Hyperlipidemia; Hyperketonemia; glucose level. mice, radioimmunoassay Non-insulin dependent Diabetes (RIA) and activation of Mellitus (NIDDM); Insulin- the leptin receptor in a dependent Diabetes Mellitus cell-based assay.
  • RIA Non-insulin dependent Diabetes
  • NIDDM Mellitus
  • IDDM Immunosuppression Protein
  • IL-1 receptor antagonist Binds IL1 receptor 1) Competition for IL-1 Autoimmune Disease; Arthritis; (Anakinra; soluble without activating the binding to IL-1 receptors Rheumatoid Arthritis; Asthma; interleukin-1 receptor; target cells; inhibits the in YT-NCI or C3H/HeJ Diabetes; Diabetes Mellitus; GVHD; IRAP; KINERET; binding of IL1-alpha cells (Carter et al., Nature Inflammatory Bowel Disorders; ANTRIL) and IL1-beta; and 344: 633-638, 1990); 2) Chron's Disease; Ocular neutralizes the biologic Inhibition of IL-1- Inflammation; Psoriasis; Septic activity of IL1-alpha induced endothelial cell- Shock; Transplant Rejection; and IL1-beta.
  • TREM-1 Triggering Mediates activation of Secretion of cytokines, Inflammation; Sepsis; bacterial Receptor Expressed on neutrophil and chemokines, infection; autoimmune diseases; Monocytes 1 monocytes; Stimulates degranulation, and cell GVHD.
  • neutrophil and surface activation monocyte-mediated markers can be inflammatory response; determined using assays Promotes secretion of described in Bouchon et TNF, IL-8, and MCP-1; al, J Immunol 2000 May Induces neutrophil 15; 164(10): 4991-5.
  • T-cell FMAT can be used to Autoimmune disorders; Inflammation activation- expression measure T-cell surface of the gastrointestinal tract; Cancer; of CD152 marker; markers (CD69, CD152, Colon Cancer; Allergy; Crohn's Stimulates release of CD71, HLA-DR) and T- disease.
  • VEGF-2 Vascular Promotes endothelial VEGF activity can be Coronary artery disease; Critical limb Endothelial Growth cell proliferation. determined using assays ischemia; Vascular disease; Factor-2; VEGF-C) known in the art, such as proliferation of endothelial cells, both those disclosed in vascular and lymphatic.
  • Antagonists International Publication may be useful as anti-angiogenic No. WO0045835, for agents; Cancer.
  • HCHNF25 jumping Activates MIP1a Dendritic cell assays are Immune disorders; cancer. translocation Release in Dendritic well known in the art. For breakpoint) Cells. example, J. Immunol. 158: 2919-2925 (1997); J. Leukoc. Biol. 65: 822- 828 (1999).
  • HLDOU18 (Bone Activates L6/GSK3 Assays for activation of Hyperglycemia; Diabetes; Diabetes Morphogenic Protein 9 kinase assay.
  • GSK3 kinase activity are Insipidus; Diabetes mellitus; Type 1 (BMP9); Growth well known in the art.
  • Type 2 diabetes Insulin differentiation factor-2 example, Biol. Chem. resistance; Insulin deficiency; precursor (GDF-2 379(8-9): (1998) 1101- Hyperlipidemia; Hyperketonemia; precursor)) 1110.; Biochem J. 1993 Non- insulin dependent Diabetes Nov. 15; 296 (Pt 1): 15-9.
  • NIDDM Insulin-dependent Diabetes Mellitus
  • IDDM Insulin-dependent Diabetes Mellitus
  • Glucagon- Like-Peptide Stimulates the synthesis GLP1 activity may be Hyperglycemia; Diabetes; Diabetes 1 (GLP1; and release of insulin; assayed in vitro using a Insipidus; Diabetes mellitus; Type 1 Insulinotropin) enhances the sensitivity [3-H]-glucose uptake diabetes; Type 2 diabetes; Insulin of adipose, muscle, and assay.
  • Exendin-4 (AC-2993) Stimulates the synthesis Exendin-4 activity may Hyperglycemia; Diabetes; Diabetes and release of insulin; be assayed in vitro using Insipidus; Diabetes mellitus; Type 1 enhances the sensitivity a [3-H]-glucose uptake diabetes; Type 2 diabetes; Insulin of adipose, muscle, and assay. (J Biol Chem 1999 resistance; Insulin deficiency; liver tissues towards Oct. 22; 274(43): Hyperlipidemia; Hyperketonemia; insulin; stimulates 30864-30873).
  • T20 T20 HIV a peptide from residues Virus inhibition assays as HIV; AIDS; SIV (simian inhibitory peptide, 643-678 of the HIV described in Zhang et al., immunodeficiency virus) infection.
  • T1249 HIV a second generation Virus inhibition assays as HIV; AIDS; SIV (simian inhibitory peptide; HIV fusion inbitor described in Zhang et al., immunodeficiency virus) infection T1249 anti-HIV Sep. 26, 2002, peptide) Sciencexpress (www.sciencexpress.org).
  • Interferon Hybrids Confers a range of Anti-viral assay: Viral infections; HIV Infections; specifically preferred: cellular responses Rubinstein S, Familletti Hepatitis; Chronic Hepatitis; IFNalpha A/D hybrid including antiviral P C, Peslka S.
  • Renal Cancer e.g., Renal IFNalpha/beta hybrid synthetase.
  • Lung Cancer e.g., modulates MHC expression of a GC-rich Non-Small Cell Lung Cancer or antigen expression, NK viral transcript
  • B-type natriuretic stimulates smooth Inhibition of angiotensin Congestive heart failure; cardiac peptide (BNP, brain muscle relaxation and can be determined using volume overload; cardiac natriuretic peptide) vasodilation, assays known in the art, decompensation; Cardiac Failure; natriuresis, and for example using an in Left Ventricular Dysfunction; suppression of renin- vitro proliferation assay Dyspnea angiotensin and with rat cardiac endothelin. fibroblasts as described in Naunyn Schmiedebergs Arch Pharmacol 1999 May; 359(5): 394-9.
  • Vasodilation can be measured in animals by measuring the myogenic responses of small renal arteries in an isobaric arteriograph system (see Am J Physiol Regul Integr Comp Physiol 2002 August; 283(2): R349- R355). Natriuesis is determined by measuring the amount of sodium in the urine.
  • ⁇ -defensin including Suppression of HIV Virus inhibition assays as HIV, AIDS; ARC. alpha 1 defensin, alpha 2 replication; active described in Zhang et al., defensin, alpha 3 against bacteria, fungi, Sep. 26, 2002, defensin (myeloid- and enveloped viruses. Sciencexpress related defensin; (www.sciencexpress.org).
  • ADHR hypophosphatemic rickets/osteomalacia
  • TIO tumor- induced rickets/osteomalacia
  • P1pal-12 (pepducin, Regulation of protease- Platelet aggregation can Protection against systemic platelet PAR1-based pepducin) activated receptor be measured using activation, thrombus, heart attack, (PAR) signal methods known in the art stroke, and/or coagulation disorders. transduction and such as described in thrombin-mediated Nature Medicine 2002 aggregation of human October; 8(10): 1161-1165. platelets.
  • P4pal-10 (pepducin, Regulation of protease- Platelet aggregation can Protection against systemic platelet PAR4-based pepducin) activated receptor be measured using activation, thrombus, heart attack, (PAR) signal methods known in the art stroke, and/or coagulation disorders.
  • HRDFD27 Involved in the T-cell proliferation can Chemoprotection; Adjunct to proliferation of T cells; be measured using assays Chemotherapy; Inflammatory Production of known in the art. For disorders; Cancer; Leukemia; TNFgamma.
  • Tumors and/or cancers of the bone cancer, leukemia, and (osteochondromas, benign melanoma; chondromas, chondroblastomas, Overexpressed in colon, chondromyxoid fibromas, osteoid lung, breast and rectal osteomas, giant cell tumors, multiple tumors; Regulates myelomas, osteosarcomas, glucose and/or FFA fibrosarcomas, malignant fibrous update by adipocytes histiocytomas, chondrosarcomas, and skeletal muscle; Ewing's tumors, and/or malignant Induces lymphomas); Bone and joint redifferentiation of infections (osteomyelitits and/or chondrocytes infectious arthritis); Charcot's joints; Heel spurs; Sever's disease; Sport's injuries; Cancer; Solid Tumors; Melanoma; Malignant Melanoma; Renal Cancer (e.g., Renal Cell Carcinoma
  • Non- Small Cell Lung Cancer or Small Cell Lung Cancer Colon Cancer; Breast Cancer; Liver Cancer; Prostate Cancer; Bladder Cancer; Gastric Cancer; Sarcoma; AIDS-Related Kaposi's Sarcoma; Lymphoma; T Cell Lymphoma; Cutaneous T-Cell Lymphoma; Non-Hodgkin's Lymphoma; Brain Cancer; Glioma; Glioblastoma Multiforme; Cervical Dysplasia; Leukemia; Preleukemia; Bone Marrow Disorders; Bone Disorders; Hairy Cell Leukemia; Chronic Myelogeonus Leukemia; Hematological Malignancies; Hematological Disorders; Multiple Myeloma; Kidney diseases and disorders; Shonlein-Henoch purpura, Berger disease, celiac disease, dermatitis herpetiformis, Chron disease; Diabetes; Diabetes Insipidus; Diabetes mellitus; Type 1 diabetes; Type 2 diabetes; Insulin resistance; Insulin
  • HDPBQ71 hematomavirus disease
  • assays that may be Blood disorders and infection (e.g., and secretion of used or routinely viral infections, tuberculosis, IFN gamma; Activation modified to test infections associated with chronic of myeloid cells and/or immunomodulatory granulomatosus disease and hematopoietic cells activity of polypeptides malignant osteoporosis); of the invention
  • Autoimmunity disorders demyelination, systemic lupus erythematosis, drug induced hemolytic anemia, rheumatoid arthritis, Sjorgren's disease, scleroderma
  • HDPBQ71 Regulates production
  • Such assays that may be Blood disorders and infection (e.g., and secretion of used or routinely viral infections, tuberculosis, IFN gamma; Activation modified to test infections associated with chronic of myeloid cells and/or immunomodulatory granulomatosus disease and hem
  • Exp. Med. disorders Bone fractures and/or adaptive immune (2002) Jan. 21; 195(2): breaks; Osteoporosis responses 201-9. (postmenopausal, senile, or idiopathic juvenile); Gout and/or pseudogout; Paget's disease; Osteoarthritis; Tumors and/or cancers of the bone (osteochondromas, benign chondromas, chondroblastomas, chondromyxoid fibromas, osteoid osteomas, giant cell tumors, multiple myelomas, osteosarcomas, fibrosarcomas, malignant fibrous histiocytomas, chondrosarcomas, Ewing's tumors, and/or malignant lymphomas); Bone and joint infections (osteomyelitits and/or infectious arthritis); Charcot's joints; Heel spurs; Sever's disease; Sport's injuries Tumstatin (T5, T7 or T8 Inhibits angio
  • disorders associated with myelin and degradation Regulates Neurosci. (2002) Nov. 1; demyelination, such as, for example, calcium currents 22(21): 9221-7.
  • demyelination such as, for example, calcium currents 22(21): 9221-7.
  • ALS multiple sclerosis, Huntington's disease; Neuronal and spinal cord injuries;
  • Disorders of the eye such as, for example, retinitis pigmentosa, blindness, color-blindness, macular degeneration.
  • Somatostatin Inhibits growth Inhibition of growth Cancer; Metastatic carcinoid tumors; (Octreotide; octreotide hormone, glucagons hormone release in Vasoactive Intestinal Peptide acetate; Sandostating and insulin; Suppresses humans by somatostatin secreting adenomas; Diarrhea and LAR ®) LF response to GnRH; can be measured as Flushing; Prostatic disorders and Decreases splanchnic described in J. Clin. cancers; Breast cancer; blood flow; Inhibits Endocrinol. Metab.
  • Gastrointestinal disorders and release of serotonin (1973) October; 37(4): cancers; Cancers of the endocrine gastrin, vasoactive 632-4. system; Head and neck intestinal peptide, Inhibition of insulin paragangliomas; Liver disorders and secretin, motilin, and secretion by somatostatin cancers; Nasopharyngeal cancers; pancreatic polypeptide. can be measured as Thyroid disorders and cancers; described in the Lancet Acromegaly; Carcinoid Syndrome; (1973) Dec. 8; 2(7841): Gallbladder disorders, such as 1299-1301.
  • gallbladder contractility diseases and abnormal bile secretion Psoriasis; Diabetes; Diabetes Insipidus; Diabetes mellitus; Type 1 diabetes; Type 2 diabetes; Insulin resistance; Insulin deficiency; Hyperlipidemia; Hyperketonemia; Non- insulin dependent Diabetes Mellitus (NIDDM); Insulin-dependent Diabetes Mellitus (IDDM); A Condition Associated With Diabetes Including, But Not Limited To Obesity, Heart Disease, Hyperglycemia, Infections, Retinopathy, And/Or Ulcers; Metabolic Disorders; Immune Disorders; Obesity; Vascular Disorders; Suppression of Body Weight; Suppression of Appetite; Syndrome X; Kidney disorders; Neurological disorders and diseases, including Alzheimers Disease.
  • Parkinson's disease and dementia Neuropsychotic disorders, including Bipolar affective disorder; Rheumatoid arthritis; Hypertension; Intracranial hypertension; Esophageal varices; Graves' disease; Seizures; Epilepsy; Gastritis; Angiogenesis; IL-22 (IL22, interleukin- Stimulates glucose IL-22 activity may be Hyperglycemia; Diabetes; Diabetes 22; IL17D, 1L27) uptake in skeletal assayed in vitro using a Insipidus; Diabetes mellitus; Type 1 muscle cells; increases [3-H]-glucose uptake diabetes; Type 2 diabetes; Insulin skeletal muscle insulin assay. (J Biol Chem 1999 resistance; Insulin deficiency; sensitivity. Oct.
  • NIDDM Non- insulin dependent Diabetes Mellitus
  • IDDM Insulin- dependent Diabetes Mellitus
  • Metabolic Disorders Immune Disorders; Obesity; Vascular Disorders; Suppression of Body Weight; Suppression of Appetite; Syndrome X.
  • HCE1P80 Stimulates glucose HCE1P80 activity may Hyperglycemia; Diabetes; Diabetes uptake in; increases be assayed in vitro using Insipidus; Diabetes mellitus; Type 1 insulin sensitivity, a [3-H]-glucose uptake diabetes; Type 2 diabetes; Insulin assay. (J Biol Chem 1999 resistance; Insulin deficiency; Oct.
  • HDRMI82 Stimulates glucose HDRMI82 activity may Hyperglycemia; Diabetes; Diabetes uptake; increases be assayed in vitro using Insipidus; Diabetes mellitus; Type 1 insulin sensitivity.
  • HDALV07 Modulates insulin Insulin activity may be Diabetes; Diabetes Insipidus; (adiponectin; gelatin- action assayed in vitro using a Diabetes mellitus; Type 1 diabetes; binding 28k protein [3-H]-glucose uptake Type 2 diabetes; Insulin resistance; precursor; adipose most assay. (J Biol Chem 1999 Insulin deficiency; Hyperlipidemia; abundant gene Oct.
  • Other indications for antibodies and/or antagonists include treatment of weight loss; treatment of AIDS wasting; appetite stimulant; treatment of cachexia.
  • PYY Peptide YY
  • Treatment of including PYY 3-36 increases satiety; can be can be measured Obesity; treatment of Diabetes; (amino acid residues 31- decreases food intake. by methods known in the suppression of body weight gain; 64 of full length PYY, art (Batterham et al. suppression of appetite.
  • WNT10b Inhibits adipogenesis.
  • WNT10b activity can be Most Preferred: Treatment of measured using Obesity; suppression of body weight adipogenesis inhibition gain; suppression of appetite.
  • Other assays Ross et al., indications; Hyperglycemia; Science 2000; 289(5481): Diabetes; Diabetes Insipidus; 950-953 Diabetes mellitus; Type 1 diabetes; Type 2 diabetes; Insulin resistance; Insulin deficiency; Hyperlipidemia; Hyperketonemia; Non-insulin dependent Diabetes Mellitus (NIDDM); Insulin- dependent Diabetes Mellitus (IDDM).
  • WNT11 Promotes cardiogenesis.
  • WNT11 activity can be Treatment of Cardiovascular measured using assays disorders; Congestive Heart Failure; known in the art, Myocardial Infarction. including cardiogenesis assays (Eisenberg et al., Dev Dyn 1999 September; 216(1): 45-58).
  • Herstatin Inhibits cancer Herstatin activity can be Oncology; Cancer; Solid Tumors; proliferation. measured using cell Melanoma; Malignant Melanoma; proliferation assays Renal Cancer (e.g., Renal Cell known in the art (Doherty Carcinoma); Lung Cancer (e.g,. Non- et al., PNAS 1999; Small Cell Lung Cancer or Small 96(19): 10869-10874.
  • Adrenomedullin stimulates vasodilation; Vasodilation can be Treatment of Congestive Heart promotes bone growth. measured using assays Failure; Hypertension; Myocardial known in the art (Ashton Infarction; Septic Shock; et al. Pharmacology Osteoporosis; Postmenopausal 2000; 61(2): 101-105. osteoporosis; Osteopenia.
  • the promotion of bone growth can be measured using assays known in the art, such as the osteoblast proliferation assay (Cornish et al. Am J Physiol 1997 December; 273(6 Pt 1): E1113- 20).
  • Nogo Receptor Receptor for the axon The promotion of axon Treatment of Central Nervous growth inhibitor, Nogo. regeneration and growth System Damage; Spinal Cord Injury; can be measured using Peripheral Nerve Damage; assays known in the art Neurodegenerative Diseases; (Fournier et al. Nature Parkinson's Disease; Alzheimer's 2001; 409(6818): 341-346). Disease; Huntington's Disease; Amyotrophic Lateral Sclerosis; Progressive Supranuclear Palsy; Creutzfeld-Jacob Disease; Motor Neuron Disease.
  • CART Treatment of Amphetamine- fat storage; promotes can be can be measured Obesity; suppression of body weight Regulated Transcript) lipid oxidation. by methods known in the gain; suppression of appetite.
  • Other art Breastham et al. indications; Hyperglycemia; Nature 2002; 418: Diabetes; Diabetes Insipidus; 650654) Diabetes mellitus; Type 1 diabetes; Type 2 diabetes; Insulin resistance; Insulin deficiency; Hyperlipidemia; Hyperketonemia; Non-insulin dependent Diabetes Mellitus (NIDDM); Insulin- dependent Diabetes Mellitus (IDDM).
  • RegIV Cold Specific Stimulates glucose RegIV activity may be Hyperglycemia; Diabetes; Diabetes Gene; Colon Specific uptake; increases assayed in vitro using a Insipidus; Diabetes mellitus; Type 1 Protein) insulin sensitivity. [3-H]-glucose uptake diabetes; Type 2 diabetes; Insulin assay. (J Biol Chem 1999 resistance; Insulin deficiency; Oct. 22; 274(43): 30864- Hyperlipidemia; Hyperketonemia; 30873).
  • Non- insulin dependent Diabetes Mellitus NIDDM
  • Insulin-dependent Diabetes Mellitus IDDM
  • Pramlintide (Amylin) Slows gastric emptying; Appetite and food intake Treatment of Obesity; treatment of (CAS-151126-32-8) decreases food intake.
  • Terlipressin triglycyl Analog of vasopressin; Terlipressin activity can Variceal hemorrhage; cirrhosis; lycine vasopressin
  • Terlipressin activity can Variceal hemorrhage; cirrhosis; lycine vasopressin
  • Ularitide (CAS-118812- Stimulates natriuresis, Ularitide activity can be Excretory disorders; Acute renal 69-4) diuresis, and assessed by measuring failure; asthma; congestive heart vasodilation. cGMP accumulation in failure; hypertension; pulmonary rat renal cells. (Valentin hypertension; cardiovascular et al., Hypertension 1993 disorders April; 21(4): 432-8).
  • Aprotinin (Trasylol) Serine protease Inhibition of thrombin- Inhibition of fibrinolysis; reduction (CAS-9087-70-1; CAS- inhibitor; attenuates induced platelet of blood loss during surgery; 11061-94-2; CAS- Systemic Inflammatory aggregation can be Treatment of Inflammation and 12407- 79-3) Response, fibrinolysis measured using methods immune Disorders. and thrombin-induced known in the art. (Poullis platelet aggregation. et al., J Thorac Cardiovasc Surg 2000 August; 120(2): 370-8).
  • Aspartocin (CAS-4117- Antibacteria Aspartocin activity can Treatment of Infectious Diseases; 65-1; CAS- 1402-89-7) be assessed using in vitro treatment of bacterial infections. antibacterial assays known in the art. (Zasloff et al., Antimicrobial Agents and Chemotherapy 1999, 43: 782-788).
  • Calcitonin (Calcimar) Regulates levels of Hypocalcemic Rat Musculoskeletal; Osteroporosis; (CAS-21215-62-3) calcium and phosphate Bioassay, bone resorbing Paget's disease; hypercalcemia; Bone in serum; causes a assay and the pit assay, Disorders; Fracture prevention; reduction in serum CT receptor binding Malignant hypercalcemia; calcium--an effect assay, CAMP stimulation Osteopenia, Osteoclastogenesis; opposite to that of assay: J Bone Miner Res osteolysis; osteomyelitis; human parathyroid 1999 August; 14(8): osteonecrosis; periodontal bone loss; hormone.
  • Carperitide (HANP; Stimulates natriuresis, Carperitide activity can Treatment of Heart Failure; recombinant human diuresis, and be assessed in vitro by Cardiovascular disorders; Respiratory atrial natriuretic peptide) vasodilation. measuring cGMP disorders; Acute respiratory distress (CAS-89213- 87-6) accumulation in a number syndrome. of cell lines, including PC12 cells and cultured human glomerular cells. (Medvede et al., Life Sci 2001 Aug.
  • Desirudin recombinant Inhibits thrombin; Desirudin activity can be Blood-related disorder; Thrombosis; hirudin; Revasc) (CAS- inhibits blood clotting. assessed using blood thrombocytopenia; hemorrhages. 120993- 53-5) clotting assays known in the art, such as in vitro platelet aggregation assays. (Glusa, Haemostasis 1991; 21 Suppl 1: 116-20).
  • Emoctakin interleukin proinflammatory Treatment of Inflammation, Immune 8) (CAS-142298- 00-8) cytokine disorders.
  • RSV infection Felypressin (CAS-56- Derivative of Felypressin Treatment of pain; to induce local 59-7) Vasopressin; Stimulates vasoconstriction activity anesthesia. vasoconstriction; can be measured using Induces local assays of anesthesia. vasoconstriction, such as the isolated arterial ring preparation. (Landstrom et al., Hum Reprod 1999 January; 14(1): 151-5).
  • Glucagon CAS-16941- Induces hyperglycemia.
  • Glucagon activity may be Hypoglycemia; Diabetes; Diabetes 32-5) assayed in vitro using a Insipidus; Diabetes mellitus; Type 1 [3-H]-glucose uptake diabetes; Type 2 diabetes; Insulin assay. (J Biol Chem 1999 resistance; Insulin deficiency; Oct. 22; 274(43): 30864- Hyperlipidemia; Hyperketonemia; 30873).
  • Non- insulin dependent Diabetes Mellitus NIDDM
  • Insulin-dependent Diabetes Mellitus IDDM
  • Non- insulin dependent Diabetes Mellitus NIDDM
  • Insulin-dependent Diabetes Mellitus IDDM
  • Becaplermin (Regranex; Promotes wound Becaplermin activity can Stimulate Epithelial Cell recombinant PDGF-BB) healing. be assessed using animal Proliferation; Stimulate Basal (CAS-165101- 51-9) wound healing models Keratinocytes; Promote Wound known in the art.
  • NP_008939 amino demyelinating disorders; neural acids 62-101)
  • neural cell e.g., neuron, glial cell, and schwann cell
  • Gastric inhibitory Increases nutrient Nutrient uptake and Most Preferred: loss of body weight, polypeptide (GIP), uptake and tryglyceride tryglyceride AIDS wasting, cachexia, and loss of including GIP fragments accumulation in accumulation can be appetite.
  • GIP polypeptide
  • Other Obesity; Diabetes; (Genbank Accession adipocytes, which leads measured by methods insulin resistance; body weight gain; No. NM_004123) to obesity and insulin described in Miyawaki et excessive appetite. resistance. al., Nat.
  • POMC Activity of POMC- Preferred resistance to stress; anti- (proopiomelanocortin), derived fragments are inflammatory activity; analgesic including fragments or diverse, and well- activity; increased skin pigmentation; variants (such as, for known in the art. See, increased protein catabolism; example, alpha- for example, Hadley et increased gluconeogenesis; obesity; melanocyte stimulating al., Ann NY Acad Sci diabetes. Other: decreased protein hormone, ⁇ MSH, 1999 Oct.
  • 6,350,430 6,350,430 6,350,430 inflammatory activity; analgesic activity; increased skin pigmentation; increased protein catabolism; increased gluconeogenesis.
  • NDP U.S. Pat. No. See U.S. Pat. No. See U.S. Pat. No. See U.S. Pat. No. Resistance to stress; anti- 6,350,430) 6,350,430 6,350,430 inflammatory activity; analgesic activity; increased skin pigmentation; increased protein catabolism; increased gluconeogenesis.
  • Interleukin-21 Immunomodulator; IL-21 activity can be Autoimmune disorders; inhibits interferon assessed by measuring Inflammatory disorders; Treatment of gamma production by interferon gamma Psoriasis; Rheumatoid Arthritis; Th1 cells, production in Th1 cells. Inflammatory bowel disease. (Wurster et al.,: J Exp Med 2002 Oct.
  • Interleukin-4 Interleukin-4 (IL-4) Immunomodulator; IL-4 activity can be Treatment of Psoriasis; Autoimmune promotes the assessed by measuring disorders; Rheumatoid Arthritis; differentiation of T cells Th1/Th2 cytokine Inflammatory bowel disease; into Th2 phenotype. responses of isolated Inflammatory disorders. spleen cells in vitro. (Waltz et al., Horm Metab Res 2002 October; 34(10): 561-9). Osteoclast Inhibitory Inhibits osteoclast Osteoclast Inhibitory Treatment of Bone Disorders; Lectin (OCIL) formation.
  • OCIL Lectin
  • Lectin activity can be Osteoporosis; Fracture prevention; assessed using osteoclast Hypercalcemia; Malignant formation assays known hypercalcemia; Paget's disease; in the art.
  • PCSK9 Inhibitor Inhibits the interaction Further LDL lowering Treatment of coronary heart disease. of PCSK9 with LDL through targeting PCSK9 Receptor. for coronary artery disease. (Cao et al. Endocrine, Metabolic & Immune Disorders-Drug Targets 2008, 8, 238-243)
  • a polypeptide having functional activity refers to a polypeptide capable of displaying one or more known functional activities associated with the full-length, pro-protein, and/or mature form of a cargo polypeptide.
  • Such functional activities include, but are not limited to, biological activity, antigenicity [ability to bind (or compete with a polypeptide for binding) to an anti-polypeptide antibody], immunogenicity (ability to generate antibody which binds to a specific polypeptide described herein), ability to form multimers with polypeptides described herein, and ability to bind to a receptor or ligand for a polypeptide.
  • the functional activity includes the ability to improve the expression and stability of a partner protein.
  • a polypeptide having biological activity refers to a polypeptide exhibiting activity similar to, but not necessarily identical to, an activity of a therapeutic protein described herein, including mature forms, as measured in a particular biological assay, with or without dose dependency. In the case where dose dependency does exist, it need not be identical to that of the polypeptide, but rather substantially similar to the dose-dependence in a given activity as compared to the polypeptide described herein (i.e., the candidate polypeptide will exhibit greater activity or not more than about 25-fold less, or not more than about tenfold less activity, or not more than about three-fold less activity relative to a polypeptide described herein, or presented in Table 2).
  • a heteromultimer described herein has at least one biological and/or therapeutic activity associated with the cargo molecule when said cargo molecule is not linked to the transporter polypeptide. In certain embodiments, a heteromultimer described herein has at least one biological and/or therapeutic activity associated with the cargo polypeptide when said cargo polypeptide is not linked to the transporter polypeptide. In certain embodiments, a heteromultimeric protein described herein has at least one biological and/or therapeutic activity associated with the cargo polypeptide portion (or fragment or variant thereof) when said cargo polypeptide is not linked to the albumin or alloalbumin based polypeptide.
  • heteromultimeric proteins described herein can be assayed for functional activity (e.g., biological activity) using or routinely modifying assays known in the art, as well as assays described herein. Additionally, one of skill in the art may routinely assay fragments of a protein corresponding to a cargo protein portion of an albumin or alloalbumin based monomeric polypeptide, for activity using assays referenced in its corresponding row of Table 2 (e.g., in column 3 of Table 2). In certain embodiments, are assay of fragments of an albumin protein corresponding to an albumin protein portion of a heteromultimer, for activity using assays known in the art and/or as described in the Examples section below.
  • various immunoassays known in the art can be used, including but not limited to, competitive and non-competitive assay systems using techniques such as radioimmunoassays, ELISA (enzyme linked immunosorbent assay), “sandwich” immunoassays, immunoradiometric assays, gel diffusion precipitation reactions, immunodiffusion assays, in situ immunoassays (using colloidal gold, enzyme or radioisotope labels, for example), western blots, precipitation reactions, agglutination assays (e.g., gel agglutination assays, hemagglutination assays), complement fixation assays, immunofluorescence assays, protein A assays, and immunoelectrophoresis assays,
  • antibody binding is detected by detecting a label on the primary antibody.
  • the primary antibody is detected by detecting binding of a secondary antibody or reagent to the primary antibody.
  • the secondary antibody is labeled. Many means are known in the art for detecting binding in an immunoassay and are within the scope of the present invention.
  • a binding partner e.g., a receptor or a ligand
  • binding to that binding partner by a heteromultimer described herein is assayed, e.g., by means well-known in the art, such as, for example, reducing and non-reducing gel chromatography, protein affinity chromatography, and affinity blotting. See generally, Phizicky et al., Microbiol. Rev. 59:94-123 (1995).
  • the ability of physiological correlates of a heteromultimeric protein to bind to a substrate(s) of polypeptides corresponding to the cargo protein portion of the heteromultimer can be routinely assayed using techniques known in the art.
  • heteromultimers described herein are used in assays to test for one or more biological activities. If a heteromultimer exhibits an activity in a particular assay, it is likely that at least one cargo protein comprised by one or more monomers of the heteromultimer is implicated in the diseases associated with the biological activity. Thus, the heteromultimer is of use in a treatment of the associated disease.
  • a method of treating a disease or disorder comprising administering to a patient in which such treatment, prevention or amelioration is desired, a heteromultimer described herein, in an amount effective to treat, prevent or ameliorate the disease or disorder.
  • monomeric albumin or alloalbumin based fusion proteins produced by a cell, wherein said proteins are encoded by polynucleotides, wherein said monomeric proteins comprise at least one cargo protein, and an albumin or alloalbumin derived polypeptide, such that said monomers form heteromultimers in solution.
  • the polynucleotides when used to express the encoded protein from a cell, the cell's natural secretion and processing steps produces a protein that lacks at least one signal sequence.
  • the specific amino acid sequence of the signal sequence is well known in the art.
  • heteromultimers described herein are used in the diagnosis, prognosis, prevention and/or treatment of diseases and/or disorders of the endocrine system. In some embodiments, heteromultimers described herein are used in the diagnosis, prognosis, prevention and/or treatment of diseases and/or disorders of the nervous system.
  • heteromultimers described herein are used in the diagnosis, prognosis, prevention and/or treatment of diseases and/or disorders of the immune system. In certain embodiments, heteromultimers described herein are used in the diagnosis, prognosis, prevention and/or treatment of diseases and/or disorders of the respiratory system.
  • heteromultimers described herein are used in the diagnosis, prognosis, prevention and/or treatment of diseases and/or disorders of the cardiovascular system. In some embodiments, heteromultimers described herein are used in the diagnosis, prognosis, prevention and/or treatment of diseases and/or disorders of the reproductive system.
  • heteromultimers described herein are used in the diagnosis, prognosis, prevention and/or treatment of diseases and/or disorders of the digestive system.
  • heteromultimer proteins described herein are used in the diagnosis, prognosis, prevention and/or treatment of diseases or disorders relating to the blood.
  • heteromultimers described herein are used in the diagnosis and/or prognosis of diseases and/or disorders associated with at least one tissue(s) in which at least one gene of interest is expressed, wherein a heteromultimer described herein comprises a cargo molecule that binds said at least one gene of interest.
  • heteromultimers described herein and/or polynucleotides encoding the albumin/alloalbumin based monomers that associate to form heteromultimers described herein are used in the diagnosis, detection and/or treatment of diseases and/or disorders associated with activities that include, but are not limited to, prohormone activation, neurotransmitter activity, cellular signaling, cellular proliferation, cellular differentiation, and cell migration.
  • heteromultimers described herein are directed to antibody-based therapies which involve administering heteromultimers described comprising cargo polypeptide(s) which is an antibody, a fragment or variant of an antibody, to a patient for treating one or more of the disclosed diseases, disorders, or conditions.
  • Therapeutic compounds described herein include, but are not limited to, heteromultimers described herein, nucleic acids encoding heteromultimers described herein.
  • antibody-based therapies which involve administering heteromultimers described herein comprising at least a fragment or variant of an antibody to a patient for treating one or more diseases, disorders, or conditions, including but not limited to: neural disorders, immune system disorders, muscular disorders, reproductive disorders, gastrointestinal disorders, pulmonary disorders, cardiovascular disorders, renal disorders, proliferative disorders, and/or cancerous diseases and conditions, and/or as described elsewhere herein.
  • heteromultimer proteins of the invention comprising at least a fragment or variant of an antibody are used therapeutically includes binding locally or systemically in the body or by direct cytotoxicity of the antibody, e.g. as mediated by complement (CDC) or by effector cells (ADCC). Some of these approaches are described in more detail below.
  • heteromultimers described herein, comprising at least a fragment or variant of an antibody may be administered alone or in combination with other types of treatments (e.g., radiation therapy, chemotherapy, hormonal therapy, immunotherapy and anti-tumor agents).
  • treatments e.g., radiation therapy, chemotherapy, hormonal therapy, immunotherapy and anti-tumor agents.
  • administration of products of a species origin or species reactivity in the case of antibodies
  • human antibodies, fragments derivatives, analogs, or nucleic acids are administered to a human patient for therapy or prophylaxis.
  • nucleic acids comprising sequences encoding heteromultimer proteins described herein are administered to treat, inhibit or prevent a disease or disorder associated with aberrant expression and/or activity of a protein, by way of gene therapy.
  • Gene therapy refers to therapy performed by the administration to a subject of an expressed or expressible nucleic acid.
  • the nucleic acids produce their encoded protein that mediates a therapeutic effect. Any of the methods for gene therapy available in the art can be used.
  • in vitro assays to demonstrate the therapeutic or prophylactic utility of a compound or pharmaceutical composition include, the effect of a compound on a cell line or a patient tissue sample.
  • the effect of the compound or composition on the cell line and/or tissue sample can be determined utilizing techniques known to those of skill in the art including, but not limited to, rosette formation assays and cell lysis assays.
  • in vitro assays which can be used to determine whether administration of a specific compound is indicated, include in vitro cell culture assays in which a patient tissue sample is grown in culture, and exposed to or otherwise administered a heteromultimer, and the effect of such heteromultimer upon the tissue sample is observed.
  • the heteromultimer is substantially purified (e.g., substantially free from substances that limit its effect or produce undesired side-effects).
  • the subject is an animal, including but not limited to animals such as cows, pigs, horses, chickens, cats, dogs, etc., and in certain embodiments, a mammal, and most preferably human.
  • a heteromultimer formulation described herein e.g., encapsulation in liposomes, microparticles, microcapsules, recombinant cells capable of expressing the compound, receptor-mediated endocytosis (see, e.g., Wu and Wu, J. Biol. Chem. 262:4429-4432 (1987)), construction of a nucleic acid as part of a retroviral or other vector, etc.
  • Methods of introduction include but are not limited to intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural, and oral routes.
  • the compounds or compositions may be administered by any convenient route, for example by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa, etc.) and may be administered together with other biologically active agents. Administration can be systemic or local.
  • Pulmonary administration can also be employed, e.g., by use of an inhaler or nebulizer, and formulation with an aerosolizing agent.
  • the heteromultimers, or compositions described herein may be administered locally to the area in need of treatment; this may be achieved by, for example, and not by way of limitation, local infusion during surgery, topical application, e.g., in conjunction with a wound dressing after surgery, by injection, by means of a catheter, by means of a suppository, or by means of an implant, said implant being of a porous, non-porous, or gelatinous material, including membranes, such as sialastic membranes, or fibers.
  • a protein, including an antibody, of the invention care must be taken to use materials to which the protein does not absorb.
  • the heteromultimers or composition can be delivered in a vesicle, in particular a liposome (see Langer, Science 249:1527-1533 (1990); Treat et al., in Liposomes in the Therapy of Infectious Disease and Cancer, Lopez-Berestein and Fidler (eds.), Liss, New York, pp. 353-365 (1989); Lopez-Berestein, ibid., pp. 317-327; see generally ibid.)
  • the heteromultimers or composition can be delivered in a controlled release system.
  • a pump may be used (see Langer, supra; Sefton, CRC Crit. Ref. Biomed. Eng. 14:201 (1987); Buchwald et al., Surgery 88:507 (1980); Saudek et al., N. Engl. J. Med. 321:574 (1989)).
  • polymeric materials can be used (see Medical Applications of Controlled Release, Langer and Wise (eds.), CRC Pres., Boca Raton, Fla.
  • a controlled release system can be placed in proximity of the therapeutic target, e.g., the brain, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, in Medical Applications of Controlled Release, supra, vol. 2, pp. 115-138 (1984)).
  • the nucleic acid in a specific embodiment comprising a nucleic acid encoding a heteromultimer described herein, can be administered in vivo to promote expression of its encoded protein, by constructing it as part of an appropriate nucleic acid expression vector and administering it so that it becomes intracellular, e.g., by use of a retroviral vector (see U.S. Pat. No.
  • a nucleic acid can be introduced intracellularly and incorporated within host cell DNA for expression, by homologous recombination.
  • compositions comprise a therapeutically effective amount of a compound, and a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans.
  • carrier refers to a diluent, adjuvant, excipient, or vehicle with which the therapeutic is administered.
  • Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like.
  • Water is a preferred carrier when the pharmaceutical composition is administered intravenously.
  • Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions.
  • Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like.
  • the composition if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents.
  • compositions can take the form of solutions, suspensions, emulsion, tablets, pills, capsules, powders, sustained-release formulations and the like.
  • the composition can be formulated as a suppository, with traditional binders and carriers such as triglycerides.
  • Oral formulation can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc. Examples of suitable pharmaceutical carriers are described in “Remington's Pharmaceutical Sciences” by E. W. Martin.
  • Such compositions will contain a therapeutically effective amount of the compound, preferably in purified form, together with a suitable amount of carrier so as to provide the form for proper administration to the patient.
  • the formulation should suit the mode of administration.
  • the composition comprising the heteromultimer is formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous administration to human beings.
  • compositions for intravenous administration are solutions in sterile isotonic aqueous buffer.
  • the composition may also include a solubilizing agent and a local anesthetic such as lignocaine to ease pain at the site of the injection.
  • the ingredients are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent.
  • composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline.
  • an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.
  • compositions described herein are formulated as neutral or salt forms.
  • Pharmaceutically acceptable salts include those formed with anions such as those derived from hydrochloric, phosphoric, acetic, oxalic, tartaric acids, etc., and those formed with cations such as those derived from sodium, potassium, ammonium, calcium, ferric hydroxide isopropylamine, triethylamine, 2-ethylamino ethanol, histidine, procaine, etc.
  • the amount of the composition described herein which will be effective in the treatment, inhibition and prevention of a disease or disorder associated with aberrant expression and/or activity of a Therapeutic protein can be determined by standard clinical techniques.
  • in vitro assays may optionally be employed to help identify optimal dosage ranges.
  • the precise dose to be employed in the formulation will also depend on the route of administration, and the seriousness of the disease or disorder, and should be decided according to the judgment of the practitioner and each patient's circumstances. Effective doses are extrapolated from dose-response curves derived from in vitro or animal model test systems.
  • heteromultimers produced as recombinant molecules by secretion from yeast, a microorganism such as a bacterium, or a human or animal cell line.
  • the polypeptides are secreted from the host cells.
  • Embodiments include a cell, such as a yeast cell transformed to express a heteromultimer protein described herein.
  • a cell such as a yeast cell transformed to express a heteromultimer protein described herein.
  • culture of those cells preferably a monoclonal (clonally homogeneous) culture, or a culture derived from a monoclonal culture, in a nutrient medium. If the polypeptide is secreted, the medium will contain the polypeptide, with the cells, or without the cells if they have been filtered or centrifuged away.
  • Many expression systems are known and may be used, including bacteria (for example E.
  • yeasts for example Saccharomyces cerevisiae, Kluyveromyces lactis and Pichia pastoris
  • filamentous fungi for example Aspergillus
  • plant cells animal cells and insect cells.
  • a heteromultimer described herein is produced in conventional ways, for example from a coding sequence inserted in the host chromosome or on a free plasmid.
  • the yeasts are transformed with a coding sequence for the desired protein in any of the usual ways, for example electroporation. Methods for transformation of yeast by electroporation are disclosed in Becker & Guarente (1990) Methods Enzymol. 194, 182.
  • Successfully transformed cells i.e., cells that contain a DNA construct of the present invention
  • cells resulting from the introduction of an expression construct can be grown to produce the desired polypeptide.
  • Cells can be harvested and lysed and their DNA content examined for the presence of the DNA using a method such as that described by Southern (1975) J. Mol. Biol. 98, 503 or Berent et al. (1985) Biotech. 3, 208.
  • the presence of the protein in the supernatant can be detected using antibodies.
  • Useful yeast plasmid vectors include pRS403-406 and pRS413-416 and are generally available from Stratagene Cloning Systems, La Jolla, Calif. 92037, USA.
  • Plasmids pRS403, pRS404, pRS405 and pRS406 are Yeast Integrating plasmids (YIps) and incorporate the yeast selectable markers HIS3, 7RP1, LEU2 and URA3.
  • Plasmids pRS413-416 are Yeast Centromere plasmids (Ycps).
  • a variety of methods have been developed to operably link DNA to vectors via complementary cohesive termini. For instance, complementary homopolymer tracts can be added to the DNA segment to be inserted to the vector DNA. The vector and DNA segment are then joined by hydrogen bonding between the complementary honmopolymeric tails to form recombinant DNA molecules.
  • Synthetic linkers containing one or more restriction sites provide an alternative method of joining the DNA segment to vectors.
  • the DNA segment generated by endonuclease restriction digestion, is treated with bacteriophage T4 DNA polymerase or E. coli DNA polymerase 1, enzymes that remove protruding, -single-stranded termini with their 3′ 5′-exonucleolytic activities, and fill in recessed 3′-ends with their polymerizing activities.
  • blunt-ended DNA segments are then incubated with a large molar excess of linker molecules in the presence of an enzyme that is able to catalyze the ligation of blunt-ended DNA molecules, such as bacteriophage T4 DNA ligase. Tus, the products of the reaction are DNA segments carrying polymeric linker sequences at their ends. These DNA segments are then cleaved with the appropriate restriction enzyme and ligated to an expression vector that has been cleaved with an enzyme that produces termini compatible with those of the DNA segment.
  • Synthetic linkers containing a variety of restriction endonuclease sites are commercially available from a number of sources including International Biotechnologies Inc, New Haven, Conn., USA.
  • Exemplary genera of yeast contemplated to be useful in the practice of the present invention as hosts for expressing the albumin, fusion proteins are Pichua (formerly classified as Hansenula ), Saccharomyces, Kluyveromyces, Aspergillus, Candida, Torulopsis, Torulaspora, Schizosaccharomyces, Citeromyces, Pachysolen, Zygosaccharomyces, Debaromyces, Trichoderma, Cephalosporium, Humicola, Mucor, Neurospora, Yarrowia, Metschunikowia, Rhodosporidium, Leucosporidium, Botryoascus, Sporidiobolus, Endomycopsis , and the like.
  • Saccharomyces spp. are S. cerevisiae, S. italicus and S. rouxii.
  • Kluyveromyces spp. are K. fragilis, K. lactis and K. marxianus .
  • a suitable Torulaspora species is T. delbrueckii .
  • Examples of Pichia ( Hansenula ) spp. are P. angusta (formerly H. polymorpha ), P. anomala (formerly H. anomala ) and P. pastoris .
  • Methods for the transformation of S. cerevisiae are taught generally in EP 251744, EP 258 067 and WO 90/01063, all of which are incorporated herein by reference.
  • Preferred exemplary species of Saccharomyces include S. cerevisiae, S. italicus, S. diastaticus , and Zygosaccharomyces rouxii .
  • Preferred exemplary species of Kluyveromyces include K. fragilis and K. lactis .
  • Preferred exemplary species of Hansenula include H. polymorpha (now Pichia angusta ), H. anomala (now Pichia anomala ), and Pichia capsulata .
  • Additional preferred exemplary species of Pichia include P. pastoris .
  • Preferred exemplary species of Aspergillus include A. niger and A. nidulans .
  • Preferred exemplary species of Yarrowia include Y. lipolytica .
  • yeast species are available from the ATCC.
  • the following preferred yeast species are available from the ATCC and are useful in the expression of albumin fusion proteins: Saccharomyces cerevisiae , Hansen, teleomorph strain BY4743 yap3 mutant (ATCC Accession No. 4022731); Saccharomyces cerevisiae Hansen, teleomorph strain BY4743 hsp150 mutant (ATCC Accession No. 4021266); Saccharomyces cerevisiae Hansen, teleomorph strain BY4743 pmt1 mutant (ATCC Accession No. 4023792); Saccharomyces cerevisiae Hansen, teleomorph (ATCC Accession Nos.
  • Suitable promoters for S. cerevisiae include those associated with the PGKI gene, GAL1 or GAL10 genes, CYCI, PH05, TRP1, ADH1, ADH2, the genes for glyceraldehyde-3-phosphate dehydrogenase, hexokinase, pyruvate decarboxylase, phosphofructokinase, triose phosphate isomerase, phosphoglucose isomerase, glucokinase, alpha-mating factor pheromone, [a mating factor pheromone], the PRBI promoter, the GUT2 promoter, the GPDI promoter, and hybrid promoters involving hybrids of parts of 5′ regulatory regions with parts of 5′ regulatory regions of other promoters or with upstream activation sites (e.g. the promoter of EP-A-258 067).
  • Convenient regulatable promoters for use in Schizosaccharomyces pombe are the thiamine-repressible promoter from the nmt gene as described by Maundrell (1990) J. Biol. Chem. 265, 10857-10864 and the glucose repressible jbpl gene promoter as described by Hoffman & Winston (1990) Genetics 124, 807-816.
  • Pichia expression kits are commercially available from Invitrogen BV, Leek, Netherlands, and Invitrogen Corp., San Diego, Calif.
  • Suitable promoters include AOX1 and AOX2.
  • Gleeson et al. (1986) J. Gen. Microbiol. 132, 3459-3465 include information on Hansenula vectors and transformation, suitable promoters being MOX1 and FMD1; whilst EP 361991, Fleer et al. (1991) and other publications from Rhone-Poulenc Rorer teach how to express foreign proteins in Kluyveromyces spp., a suitable promoter being PGKI.
  • the transcription termination signal is preferably the 3′ flanking sequence of a eukaryotic gene which contains proper signals for transcription termination and polyadenylation.
  • Suitable 3′ flanking sequences may, for example, be those of the gene naturally linked to the expression control sequence used, i.e. may correspond to the promoter. Alternatively, they may be different in which case the termination signal of the S. cerevisiae ADHI gene is preferred.
  • the desired heteromultimer protein is initially expressed with a secretion leader sequence, which may be any leader effective in the yeast chosen.
  • leader sequence useful in S. cerevisiae include that from the mating factor alpha polypeptide (MF ⁇ -1) and the hybrid leaders of EP-A-387 319. Such leaders (or signals) are cleaved by the yeast before the mature albumin is released into the surrounding medium. Further such leaders include those of S. cerevisiae invertase (SUC2) disclosed in JP 62-096086 (granted as 911036516), acid phosphatase (PH05), the pre-sequence of MF ⁇ -1, 0 glucanase (BGL2) and killer toxin; S. diastaticus glucoamylase Il; S. carlsbergensis ⁇ -galactosidase (MEL1); K. lactis killer toxin; and Candida glucoamylase.
  • SUC2 S. cerevisiae invertase
  • vectors containing a polynucleotide encoding a heteromultimer protein described herein, host cells, and the production of the heteromultimer proteins by synthetic and recombinant techniques may be, for example, a phage, plasmid, viral, or retroviral vector. Retroviral vectors may be replication competent or replication defective. In the latter case, viral propagation generally will occur only in complementing host cells.
  • the polynucleotides encoding heteromultimer proteins described herein are joined to a vector containing a selectable marker for propagation in a host.
  • a plasmid vector is introduced in a precipitate, such as a calcium phosphate precipitate, or in a complex with a charged lipid. If the vector is a virus, it may be packaged in vitro using an appropriate packaging cell line and then transduced into host cells.
  • the polynucleotide insert is operatively linked to an appropriate promoter, such as the phage lambda PL promoter, the E. coli lac, trp, phoA and rac promoters, the SV40 early and late promoters and promoters of retroviral LTRs, to name a few.
  • an appropriate promoter such as the phage lambda PL promoter, the E. coli lac, trp, phoA and rac promoters, the SV40 early and late promoters and promoters of retroviral LTRs, to name a few.
  • Other suitable promoters will be known to the skilled artisan.
  • the expression constructs will further contain sites for transcription initiation, termination, and, in the transcribed region, a ribosome binding site for translation.
  • the coding portion of the transcripts expressed by the constructs will preferably include a translation initiating codon at the beginning and a termination codon (UAA, UGA or UAG) appropriately
  • the expression vectors will preferably include at least one selectable marker.
  • markers include dihydrofolate reductase, G418, glutamine synthase, or neomycin resistance for eukaryotic cell culture, and tetracycline, kanamycin or ampicillin resistance genes for culturing in E. coli and other bacteria.
  • Representative examples of appropriate hosts include, but are not limited to, bacterial cells, such as E. coli, Streptomyces and Salmonella typhimurium cells; fungal cells, such as yeast cells (e.g., Saccharomyces cerevisiae or Pichia pastoris (ATCC Accession No.
  • insect cells such as Drosophila S2 and Spodoptera Sf cells
  • animal cells such as CHO, COS, NSO, 293, and Bowes melanoma cells
  • plant cells Appropriate culture mediums and conditions for the above-described host cells are known in the art.
  • vectors preferred for use in bacteria include pQE70, pQE60 and pQE-9, available from QIAGEN, Inc.; pBluescript vectors, Phagescript vectors, pNH8A, pNH16a, pNH18A; pNH46A, available from Stratagene Cloning Systems, Inc.; and ptrc99a, pKK223-3, pKK233-3, pDR540, pRIT5 available from Pharmacia Biotech, Inc.
  • preferred eukaryotic vectors are pWLNEO, pSV2CAT, pOG44, pXTI and pSG available from Stratagene; and pSVK3, pBPV, pMSG and pSVL available from Pharmacia.
  • Preferred expression vectors for use in yeast systems include, but are not limited to pYES2, pYDI, pTEFI/Zeo, pYES2/GS, pPICZ, pGAPZ, pGAPZalph, pPIC9, pPIC3.5, pHIL-D2, pHIL-S1, pPIC3.5K, pPIC9K, and PA0815 (all available from Invitrogen, Carlbad, Calif.).
  • Other suitable vectors will be readily apparent to the skilled artisan.
  • polynucleotides encoding a heteromultimer protein described herein are fused to signal sequences that will direct the localization of a protein of the invention to particular compartments of a prokaryotic or eukaryotic cell and/or direct the secretion of a protein of the invention from a prokaryotic or eukaryotic cell.
  • signal sequences that will direct the localization of a protein of the invention to particular compartments of a prokaryotic or eukaryotic cell and/or direct the secretion of a protein of the invention from a prokaryotic or eukaryotic cell.
  • E. coli one may wish to direct the expression of the protein to the periplasmic space.
  • Examples of signal sequences or proteins (or fragments thereof) to which the heteromultimeric proteins are fused in order to direct the expression of the polypeptide to the periplasmic space of bacteria include, but are not limited to, the pelB signal sequence, the maltose binding protein (MBP) signal sequence, MBP, the ompA signal sequence, the signal sequence of the periplasmic E. coli heat-labile enterotoxin B-subunit, and the signal sequence of alkaline phosphatase.
  • MBP maltose binding protein
  • ompA the signal sequence of the periplasmic E. coli heat-labile enterotoxin B-subunit
  • alkaline phosphatase Several vectors are commercially available for the construction of fusion proteins which will direct the localization of a protein, such as the pMAL series of vectors (particularly the pMAL-.rho. series) available from New England Biolabs.
  • polynucleotides albumin fusion proteins of the invention may be fused to the pelB pectate lyase signal sequence to increase the efficiency of expression and purification of such polypeptides in Gram-negative bacteria. See, U.S. Pat. Nos. 5,576,195 and 5,846,818, the contents of which are herein incorporated by reference in their entireties.
  • MPIF-1 signal sequence e.g., amino acids 1-21 of GenBank Accession number AAB51134
  • MLQNSAVLLLLVISASA stanniocalcin signal sequence
  • MPTWAWWLFLVLLLALWAPARG consensus signal sequence
  • a suitable signal sequence that may be used in conjunction with baculoviral expression systems is the gp67 signal sequence (e.g., amino acids 1-19 of GenBank Accession Number AAA72759).
  • Glutaminase GS
  • DHFR DHFR
  • An advantage of glutamine synthase based vectors are the availability of cell lines (e.g., the murine myeloma cell line, NSO) which are glutamine synthase negative.
  • Glutamine synthase expression systems can also function in glutamine synthase expressing cells (e.g., Chinese Hamster Ovary (CHO) cells) by providing additional inhibitor to prevent the functioning of the endogenous gene.
  • glutamine synthase expression system and components thereof are detailed in PCT publications: WO87/04462; WO86/05807; WO89/10036; WO89/10404; and WO91/06657, which are hereby incorporated in their entireties by reference herein. Additionally, glutamine synthase expression vectors can be obtained from Lonza Biologics, Inc. (Portsmouth, N.H.). Expression and production of monoclonal antibodies using a GS expression system in murine myeloma cells is described in Bebbington et al., Bio/technology 10:169(1992) and in Biblia and Robinson Biotechnol. Prog. 11:1(1995) which are herein incorporated by reference.
  • host cells containing vector constructs described herein and additionally host cells containing nucleotide sequences that are operably associated with one or more heterologous control regions (e.g., promoter and/or enhancer) using techniques known of in the art.
  • the host cell can be a higher eukaryotic cell, such as a mammalian cell (e.g., a human derived cell), or a lower eukaryotic cell, such as a yeast cell, or the host cell can be a prokaryotic cell, such as a bacterial cell.
  • a host strain may be chosen which modulates the expression of the inserted gene sequences, or modifies and processes the gene product in the specific fashion desired.
  • Expression from certain promoters can be elevated in the presence of certain inducers; thus expression of the genetically engineered polypeptide may be controlled.
  • different host cells have characteristics and specific mechanisms for the translational and post-translational processing and modification (e.g., phosphorylation, cleavage) of proteins. Appropriate cell lines can be chosen to ensure the desired modifications and processing of the foreign protein expressed.
  • nucleic acids and nucleic acid constructs of the invention into the host cell can be effected by calcium phosphate transfection, DEAE-dextran mediated transfection, cationic lipid-mediated transfection, electroporation, transduction, infection, or other methods. Such methods are described in many standard laboratory manuals, such as Davis et al., Basic Methods In Molecular Biology (1986). It is specifically contemplated that the polypeptides of the present invention may in fact be expressed by a host cell lacking a recombinant vector.
  • the invention also encompasses primary, secondary, and immortalized host cells of vertebrate origin, particularly mammalian origin, that have been engineered to delete or replace endogenous genetic material (e.g., the coding sequence corresponding to a Cargo polypeptide is replaced with a heteromultimer protein corresponding to the Cargo polypeptide), and/or to include genetic material.
  • endogenous genetic material e.g., the coding sequence corresponding to a Cargo polypeptide is replaced with a heteromultimer protein corresponding to the Cargo polypeptide
  • the genetic material operably associated with the endogenous polynucleotide may activate, alter, and/or amplify endogenous polynucleotides.
  • heterologous polynucleotides e.g., polynucleotides encoding an albumin protein, or a fragment or variant thereof
  • heterologous control regions e.g., promoter and/or enhancer
  • endogenous polynucleotide sequences encoding a Therapeutic protein via homologous recombination (see, e.g., U.S. Pat. No. 5,641,670, issued Jun. 24, 1997; International Publication Number WO 96/29411; International Publication Number WO 94/12650; Koller et al., Proc. Natl. Acad. Sci. USA 86:8932-8935 (1989); and Zijlstra et al., Nature 342:435-438 (1989), the disclosures of each of which are incorporated by reference in their entireties).
  • Heteromultimer proteins described herein can be recovered and purified from recombinant cell cultures by well-known methods including ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography, hydrophobic charge interaction chromatography and lectin chromatography. Most preferably, high performance liquid chromatography (“HPLC”) is employed for purification.
  • HPLC high performance liquid chromatography
  • heteromultimer proteins of the invention are purified using Anion Exchange Chromatography including, but not limited to, chromatography on Q-sepharose, DEAE sepharose, poros HQ, poros DEAF, Toyopearl Q, Toyopearl QAE, Toyopearl DEAE, Resource/Source Q and DEAE, Fractogel Q and DEAE columns.
  • Anion Exchange Chromatography including, but not limited to, chromatography on Q-sepharose, DEAE sepharose, poros HQ, poros DEAF, Toyopearl Q, Toyopearl QAE, Toyopearl DEAE, Resource/Source Q and DEAE, Fractogel Q and DEAE columns.
  • the proteins described herein are purified using Cation Exchange Chromatography including, but not limited to, SP-sepharose, CM sepharose, poros HS, poros CM, Toyopearl SP, Toyopearl CM, Resource/Source S and CM, Fractogel S and CM columns and their equivalents and comparables.
  • heteromultimer proteins described herein can be chemically synthesized using techniques known in the art (e.g., see Creighton, 1983, Proteins: Structures and Molecular Principles, W. H. Freeman & Co., N.Y and Hunkapiller et al., Nature, 310:105-111 (1984)).
  • a polypeptide corresponding to a fragment of a polypeptide can be synthesized by use of a peptide synthesizer.
  • nonclassical amino acids or chemical amino acid analogs can be introduced as a substitution or addition into the polypeptide sequence.
  • Non-classical amino acids include, but are not limited to, to the D-isomers of the common amino acids, 2,4diaminobutyric acid, alpha-amino isobutyric acid, 4aminobutyric acid, Abu, 2-amino butyric acid, g-Abu, e-Ahx, 6amino hexanoic acid, Aib, 2-amino isobutyric acid, 3-amino propionic acid, omithine, norleucine, norvaline, hydroxyproline, sarcosine, citrulline, homocitrulline, cysteic acid, t-butylglycine, t-butylalanine, phenylglycine, cyclohexylalanine, ⁇ -alanine, fluoro-amino acids, designer amino acids such as ⁇ -methyl amino acids, C ⁇ -methyl amino acids, N ⁇ -methyl amino acids, and amino acid analogs in general. Furthermore, the amino
  • heteromultimers which are differentially modified during or after translation, e.g., by glycosylation, acetylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, linkage to an antibody molecule or other cellular ligand, etc. Any of numerous chemical modifications may be carried out by known techniques, including but not limited, to specific chemical cleavage by cyanogen bromide, trypsin, chymotrypsin, papain, V8 protease, NaBH 4 ; acetylation, formylation, oxidation, reduction; metabolic synthesis in the presence of tunicamycin; etc.
  • Additional post-translational modifications encompassed herein include, for example, e.g., N-linked or O-linked carbohydrate chains, processing of N-terminal or C-terminal ends), attachment of chemical moieties to the amino acid backbone, chemical modifications of N-linked or O-linked carbohydrate chains, and addition or deletion of an N-terminal methionine residue as a result of procaryotic host cell expression.
  • the heteromultimer proteins are modified with a detectable label, such as an enzymatic, fluorescent, isotopic or affinity label to allow for detection and isolation of the protein.
  • suitable enzymes include horseradish peroxidase, alkaline phosphatase, beta-galactosidase, or acetylcholinesterase
  • suitable prosthetic group complexes include streptavidin biotin and avidin/biotin
  • suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin
  • an example of a luminescent material includes luminol
  • examples of bioluminescent materials include luciferase, luciferin, and aequorin
  • suitable radioactive material include iodine, carbon, sulfur, tritium, indium, technetium, thallium, gallium, palladium, molybdenum, xenon, fluorine.
  • heteromultimer proteins or fragments or variants thereof are attached to macrocyclic chelators that associate with radiometal ions.
  • heteromultimer described herein is modified by either natural processes, such as post-translational processing, or by chemical modification techniques which are well known in the art. It will be appreciated that the same type of modification may be present in the same or varying degrees at several sites in a given polypeptide.
  • Polypeptides of the invention may be branched, for example, as a result of ubiquitination, and they may be cyclic, with or without branching. Cyclic, branched, and branched cyclic polypeptides may result from posttranslation natural processes or may be made by synthetic methods.
  • Modifications include acetylation, acylation, ADP-ribosylation, amidation, covalent attachment of flavin, covalent attachment of a heme moiety, covalent attachment of a nucleotide or nucleotide derivative, covalent attachment of a lipid or lipid derivative, covalent attachment of phosphotidylinositol, cross-linking, cyclization, disulfide bond formation, demethylation, formation of covalent cross-links, formation of cysteine, formation of pyroglutamate, formylation, gamma-carboxylation, glycosylation, GPI anchor formation, hydroxylation, iodination, methylation, myristylation, oxidation, pegylation, proteolytic processing, phosphorylation, prenylation, racemization, selenoylation, sulfation, transfer-RNA mediated addition of amino acids to proteins such as arginylation, and ubiquitination.
  • heteromultimeric proteins may also be attached to solid supports, which are particularly useful for immunoassays or purification of polypeptides that are bound by, that bind to, or associate with albumin fusion proteins of the invention.
  • solid supports include, but are not limited to, glass, cellulose, polyacrylamide, nylon, polystyrene, polyvinyl chloride or polypropylene.
  • heteromultimeric protein comprises only the VH domain of an antibody
  • heteromultimeric protein comprises only the VL domain of an antibody
  • the chemical moieties for derivitization may be selected from water soluble polymers such as polyethylene glycol, ethylene glycol/propylene glycol copolymers, carboxymethylcellulose, dextran, polyvinyl alcohol and the like.
  • the proteins may be modified at random positions within the molecule, or at predetermined positions within the molecule and may include one, two, three or more attached chemical moieties.
  • the polymer may be of any molecular weight, and may be branched or unbranched.
  • the preferred molecular weight is between about 1 kDa and about 100 kDa (the term “about” indicating that in preparations of polyethylene glycol, some molecules will weigh more, some less, than the stated molecular weight) for ease in handling and manufacturing.
  • Other sizes may be used, depending on the desired therapeutic profile (e.g., the duration of sustained release desired, the effects, if any on biological activity, the ease in handling, the degree or lack of antigenicity and other known effects of the polyethylene glycol to a Therapeutic protein or analog).
  • the polyethylene glycol may have an average molecular weight of about 200, 500, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 5500, 6000, 6500, 7000, 7500, 8000, 8500, 9000, 9500, 10,000, 10,500, 11,000, 11,500, 12,000, 12,500, 13,000, 13,500, 14,000, 14,500, 15,000, 105,500, 16,000, 16,500, 17,000, 17,500, 18,000, 18,500, 19,000, 19,500, 20,000, 25,000, 30,000, 35,000, 40,000, 45,000, 50,000, 55,000, 60,000, 65,000, 70,000, 75,000, 80,000, 85,000, 90,000, 95,000, or 100,000 kDa.
  • the presence and quantity of heteromultimer proteins described herein may be determined using ELISA, a well known immunoassay known in the art.
  • ELISA protocol that would be useful for detecting/quantifying heteromultimers described herein, comprises the steps of coating an ELISA plate with an anti-human serum albumin antibody, blocking the plate to prevent non-specific binding, washing the ELISA plate, adding a solution containing the protein described herein (at one or more different concentrations), adding a secondary anti-cargo polypeptide specific antibody coupled to a detectable label (as described herein or otherwise known in the art), and detecting the presence of the secondary antibody.
  • the ELISA plate might be coated with the anti-cargo polypeptide specific antibody and the labeled secondary reagent might be the anti-human albumin specific antibody.
  • multifunctional heteromultimers that comprise: at least two monomers, wherein at least one monomer comprises at least one cargo molecule attached to a transporter polypeptide, such that said monomers associate to form the heteromultimer; wherein at least one transporter polypeptide is derived from a monomeric protein and wherein said transporter polypeptides self-assemble to form a quasi-native structure of said monomeric protein or analog thereof.
  • the cargo molecule is a biomolecule.
  • a heteromultimer that comprises: at least two monomeric proteins, wherein each monomeric protein comprises at least one cargo polypeptide, attached to a transporter polypeptide, such that said monomeric proteins self-assemble to form the heteromultimer.
  • the heteromultimer is a heterodimer. In an embodiment, the heteromultimer is bispecific. In an embodiment, the heteromultimer is multispecific. In certain embodiments, at least one transporter polypeptide is not derived from an antibody. In certain embodiments, the transporter polypeptides are not derived from an antibody. In an embodiment, the heteromultimer is multifunctional. In certain embodiments, the transporter polypeptides are derivatives of albumin. In certain embodiments of the heteromultimer described herein, the transporter polypeptides are derived from human serum albumin of SEQ ID No. 1. In certain embodiments of the heteromultimer described herein, the transporter polypeptides are derived from alloalbumins.
  • the cargo polypeptides are therapeutic proteins described herein, or fragments or variants thereof. In some embodiments, at least one cargo polypeptide is fused to the transporter polypeptide. In certain embodiments, at least one cargo polypeptide is attached to the N-terminus of the transporter polypeptide. In some embodiments, at least one cargo polypeptide is attached to the C-terminus of the transporter polypeptide.
  • heteromultimers each heteromultimer comprising: at least a first monomer that comprises at least one cargo molecule, and a first transporter polypeptide; and at least a second monomer that comprises at least one cargo molecule and a second transporter polypeptide wherein at least one transporter polypeptide is derived from a monomeric protein and wherein said transporter polypeptides self-assemble to form a quasi-native structure of said monomeric protein or analog thereof.
  • at least one cargo molecule is a therapeutic agent described herein.
  • at least one cargo molecule is a biomolecule described herein.
  • heteromultimers each heteromultimer comprising: at least a first monomeric protein that comprises at least one cargo polypeptide and a first transporter polypeptide; and at least a second monomeric protein that comprises at least one cargo polypeptide and a second transporter polypeptide.
  • the heteromultimer is a heterodimer.
  • the heteromultimer is multivalent.
  • the heteromultimer is bivalent.
  • the heteromultimer is multispecific.
  • the heteromultimer is bispecific.
  • the transporter polypeptides are derivatives of albumin.
  • the transporter polypeptides are derived from human serum albumin of SEQ ID No. 1.
  • heteromultimers each heteromultimer comprising: at least a first monomeric protein that comprises at least one cargo polypeptide and a first transporter polypeptide comprising a sequence of SEQ ID NO:2; and at least a second monomeric protein that comprises at least one cargo polypeptide and a second transporter polypeptide comprising a sequence of SEQ ID NO: 3.
  • at least one transporter polypeptide is derived from alloalbumins.
  • both transporter polypeptides are derived from alloalbumins.
  • all transporter polypeptides are derivatives of the same alloalbumin.
  • the transporter polypeptides are derivatives of different alloalbumins.
  • each transporter polypeptide is an alloalbumin derivative based on an alloalbumin selected from Table 2.
  • the first monomeric protein comprises two cargo polypeptides.
  • the second monomeric protein comprises two cargo polypeptides.
  • the transporter polypeptides are derivatives of an annexin protein. In an embodiment, the transporter polypeptides are derived from different annexin proteins. In certain embodiments, the transporter polypeptides are derived from the same annexin protein. In an embodiment, at least one transporter polypeptide is derived from Annexin A1 or lipocortin I. In certain embodiments of the heteromultimer, all transporter polypeptides are derived from Annexin A1 of SEQ ID NO: 14. In certain embodiments of the heteromultimer, at least one transporter polypeptides is derived from a sequence homologous to SEQ ID NO: 14.
  • At least one transporter polypeptide is derived from Annexin A2 or annexin II. In certain embodiments of the heteromultimer, all transporter polypeptides are derived from Annexin A2 or lipocortin II. In an embodiment, at least one transporter polypeptide is derived from Annexin like protein. In certain embodiments of the heteromultimer, all transporter polypeptides are derived from Annexin like protein. In an embodiment, at least one transporter polypeptide is derived from the group comprising Annexin A1-Annexin A7. In an embodiment of the heteromultimer described herein, all transporter polypeptides are derived from the group comprising Annexin A1-Annexin A7. 14. In certain embodiments, the first annexin based transporter polypeptide has a sequence comprising SEQ ID NO:15, and the second annexin based transporter polypeptide has a sequence comprising SEQ ID NO: 16.
  • the transporter polypeptides are derivatives of transferrin. In an embodiment, at least one transporter polypeptide is derived from transferrin. In certain embodiments of the heteromultimer, at least one transporter polypeptides are derived from transferrin of SEQ ID NO: 19 or analog thereof. In certain embodiments of the heteromultimer, at least one transporter polypeptide is derived from a polypeptide sequence homologous to the transferrin. In certain embodiments of the heteromultimer described herein, at least one transporter polypeptide is derived from apo-transferrin.
  • the first transferrin based transporter polypeptide has a sequence comprising SEQ ID NO:15 and the second transferrin based transporter polypeptide has a sequence comprising SEQ ID NO: 16.
  • heteromultimers each heteromultimer comprising: at least a first monomeric protein that comprises at least one cargo polypeptide and a first transporter polypeptide; and at least a second monomeric protein that comprises at least one cargo polypeptide and a second transporter polypeptide, wherein said cargo polypeptides are selected from the proteins listed in Table 2, and wherein at least one transporter polypeptide is derived from a monomeric protein and wherein said transporter polypeptides self-assemble to form a quasi-native structure of said monomeric protein or analog thereof.
  • heteromultimers comprising: at least a first monomeric protein that comprises at least one cargo polypeptide and a first transporter polypeptide; and at least a second monomeric protein that comprises at least one cargo polypeptide and a second transporter polypeptide, wherein at least one at least one cargo polypeptide is an antibody, or fragment or variant thereof.
  • all cargo polypeptides are antibodies or fragments or variants thereof.
  • at least one cargo molecule attached to the first transporter polypeptide is the same as at least one cargo molecule attached to the second transporter polypeptide.
  • the cargo molecules attached to the first transporter polypeptide are different from the cargo molecule on the second transporter polypeptide.
  • the cargo molecules attached to the first transporter polypeptide are the same. In certain embodiments at least two cargo molecules attached to the first transporter polypeptide are different from each other. In certain embodiments at least two cargo molecules attached to the second transporter polypeptide are the same. In certain embodiments at least two cargo molecules attached to the second transporter polypeptide are different.
  • the antibody fragment comprises antibody Fc region. In some embodiments, the antibody is an immunoglobulin selected from the group consisting of IgG, IgA, IgD, IgE, and IgM.
  • the IgG is of subtype selected from IgG1, IgG2a, IgG2b, IgG3 and IgG4.
  • the antibody is a multispecific antibody. In some embodiments, the multispecific antibody is a bispecific antibody.
  • heteromultimers each heteromultimer comprising: at least a first monomeric protein that comprises at least one cargo polypeptide and a first transporter polypeptide; and at least a second monomeric protein that comprises at least one cargo polypeptide and a second transporter polypeptide, wherein at least one cargo polypeptide is a therapeutic antibody.
  • At least one cargo polypeptide is a therapeutic antibody or fragment or variant thereof, wherein the antibody is selected from antibody is selected from abagovomab, adalimumab, alemtuzumab, aurograb, bapineuzumab, basiliximab, belimumab, bevacizumab, briakinumab, canakinumab, catumaxomab, certolizumab pegol, certuximab, daclizumab, denosumab, efalizumab, galiximab, gemtuzumab ozagamicin, golimumab, ibritumomab tiuxetan, infliximab, ipilimumab, lumiliximab, mepolizumab, motavizumab, muromonab, mycograb, natalizumab, nimotuzumab,
  • heteromultimers each heteromultimer comprising: at least a first monomeric protein that comprises at least one cargo polypeptide and a first transporter polypeptide; and at least a second monomeric protein that comprises at least one cargo polypeptide and a second transporter polypeptide, wherein at least one cargo polypeptide is an enzyme, hormone, therapeutic polypeptide, antigen, chemotoxin, radiotoxin, cytokine or variant or fragment thereof.
  • heteromultimers each heteromultimer comprising: at least a first monomeric protein that comprises at least one cargo polypeptide and a first transporter polypeptide; and at least a second monomeric protein that comprises at least one cargo polypeptide and a second transporter polypeptide, wherein the cargo polypeptide is attached to the transporter polypeptide by chemical conjugation, native ligation, chemical ligation, a disulfide bond or fusion.
  • host cells comprising nucleic acid encoding a heteromultimer described herein.
  • the nucleic acid encoding the first monomeric protein and the nucleic acid encoding the second monomeric protein are present in a single vector.
  • the nucleic acid encoding the first monomeric protein and the nucleic acid encoding the second monomeric protein are present in separate vectors.
  • the host cell is a prokaryotic cell or a eukaryotic cell.
  • the host cell is yeast cell.
  • the yeast is S. cerevisiae .
  • the yeast is glycosylation deficient, and/or protease deficient.
  • the host cell is a bacterial cell.
  • the host cell expressing a heteromultimer descried herein is a mammalian cell.
  • the mammalian cell is a CHO cell, a BHK cell, NSO cell, COS cell or a human cell.
  • a pharmaceutical composition that comprises a heteromultimer described herein and a pharmaceutically acceptable adjuvant.
  • methods of treating an individual suffering from a disease or disorder comprising administering to the individual an effective amount of a formulation or pharmaceutical composition described herein.
  • a method of treating cancer in a patient comprising administering to the patient a therapeutically effective amount of a heteromultimer described herein.
  • a method of treating an immune disorder in a patient comprising administering to the patient a therapeutically effective amount of a heteromultimer described herein.
  • a method of treating an infectious disease in a patient said method comprising administering to the patient a therapeutically effective amount of a heteromultimer described herein.
  • a method of treating a cardiovascular disorder in a patient comprising administering to the patient a therapeutically effective amount of a heteromultimer described herein.
  • a method of treating a respiratory disorder in a patient comprising administering to the patient a therapeutically effective amount of a heteromultimer described herein.
  • kits for detecting the presence of a biomarker of interest in an individual comprising (a) an amount of a heteromultimer described herein, wherein said heteromultimer comprises at least one cargo polypeptide such that said cargo polypeptide is capable of binding to the biomarker of interest; and (b) instructions for use.
  • heteromultimer proteins that comprise at least two monomeric proteins, wherein each monomeric protein comprises at least one cargo polypeptide, and an albumin based polypeptide, such that said monomeric proteins self-assemble to form the heteromultimer.
  • the cargo polypeptide is fused to the albumin or alloalbumin based polypeptide. In some embodiments, the cargo polypeptide is chemically conjugated to the albumin or alloalbumin based polypeptide. In certain embodiments, the cargo polypeptide is attached to the albumin or alloalbumin based polypeptide by means of chemical ligation or a disulfide bond.
  • heteromultimer proteins that comprise at least two monomeric proteins, wherein each monomeric protein comprises at least one cargo polypeptide, and an alloalbumin based polypeptide, such that said alloalbumin based polypeptides self-assemble to form the heteromultimer with a quasi-native structure of said alloalbumin or analog thereof.
  • a heteromultimer described herein is a heterodimer.
  • cargo polypeptide is an antibody, enzyme, hormone, therapeutic polypeptide, antigen, chemotoxin, radiotoxin, cytokine or variant or fragment thereof.
  • the cargo polypeptide of one monomeric protein functions in synergy with the cargo polypeptide of another monomeric protein.
  • HSA human serum albumin
  • this strategy is also used to design heteromultimers comprising monomeric polypeptides that comprise transporter polypeptides that are derivatives of HSA variants, alloalbumins other homologous albumin molecules from other species and also Annexin and Transferrin.
  • the monomers described herein can be engineered using a variety of strategies to improve biophysical characteristics such as the stability of the individual transporter polypeptides or their associated complex.
  • transporter polypeptide which is a HAS, HAA, Annexin or Transferrin derived scaffold that can be conjugated or fused with cargo polypeptides such as other functional domains such as antigen binding protein units, target substrates or inhibitors or payloads such as chemotoxins, radiotoxins, cytokines, etc. to achieve a multispecific or multifunctional therapeutic protein.
  • Described herein are fusions of heterodimeric Fc with transporter polypeptides based on HSA to yield bispecific antibody based therapeutics with sufficient purity and stability for pharmaceutical applications.
  • a method of deriving a multispecific or multifunctional protein comprising self-assembling monomers that comprise transporter polypeptides based on HSA, such that, the protein has a number of favorable pharmacokinetic properties including improved half-life, improved stability, low immunogenicity, etc.
  • heterodimer proteins that comprise at least two monomeric fusion proteins, wherein each monomeric fusion proteins comprises at least one cargo polypeptide fused to an albumin derived polypeptide, such that said albumin derived polypeptides self-assemble to form the multifunctional heterodimer with a quasi-native structure of albumin or an analog thereof.
  • heterodimer proteins that comprise at least two monomeric fusion proteins, wherein each monomeric fusion proteins comprises at least one cargo polypeptide fused to an alloalbumin derived polypeptide, such that said alloalbumin derived polypeptides self-assemble to form the multifunctional heterodimer.
  • heteromultimer proteins that comprise at least two monomeric fusion proteins, wherein each monomeric fusion proteins comprises at least one cargo polypeptide fused to an alloalbumin derived polypeptide, such that said alloalbumin derived polypeptides self-assemble to form the multifunctional heterodimer.
  • heterodimeric proteins comprising a first monomer which comprises at least one cargo polypeptide fused to an alloalbumin derived polypeptide; and a second monomer that comprises at least one cargo polypeptide fused to an alloalbumin derived polypeptide.
  • the at least one cargo polypeptide of the first monomer is different from the at least one cargo polypeptide of the second monomer.
  • a heteromultimer that comprises: at least two monomers, each comprising a transporter polypeptide and optionally at least one cargo molecule attached to said transporter polypeptide, wherein each transporter polypeptide is obtained by segmentation of a whole protein such that said transporter polypeptides self-assemble to form quasi-native whole protein.
  • the heteromultimer is multispecific.
  • the transporter polypeptides are not derived from an antibody.
  • each monomer preferentially forms the heteromultimer as compared to a monomer or a homomultimer.
  • at least one cargo molecule is a therapeutic agent, or a biomolecule.
  • At least one cargo molecule is a biomolecule which is selected from a polypeptide, DNA, PNA, or RNA.
  • each transporter polypeptide is a derivate of albumin or alloalbumin.
  • each transporter polypeptide is a derivate of annexin.
  • each transporter polypeptide is a derivate of transferrin.
  • a formulation described herein is provided as part of a kit or container.
  • the kit or container is packaged with instructions pertaining to extended shelf life of the therapeutic protein.
  • a heteromultimer described herein is used in a method of treating (e.g., ameliorating) preventing, or diagnosing a disease or disease symptom in an individual, comprising the step of administering said formulation to the individual.
  • transgenic organisms modified to contain nucleic acid molecules described herein to encode and express monomeric fusion proteins described herein.
  • Electrostatic interactions involve the formation of salt bridges, hydrogen bonds and the pervasive dispersion interactions. Solvent exclusion and reorganization around non-polar atomic groups at the interface and its associated entropic effects play a role in the hydrophobic component of the binding thermodynamics. Residues with geometries that are optimized for hydrophobic interaction with one another will form contacts (i.e. stacking, pi-pi, cation-pi contacts favorable for stabilizing a protein-protein interface.
  • thermodynamic effects control multi-step protein folding processes that involve the pre-organization of secondary structural units and tertiary domains, which is followed by their association to form the folded quaternary state of the protein.
  • An alternate mechanism to protein folding and binding involves a coupled protein folding and binding process that ultimately results in the quaternary state of the protein.
  • the individual protein components need to be co-expressed or be present in the same medium and each of the components or monomers will stably fold into its final structural state only on association with its obligate partner. ( FIG. 6 )
  • a split protein involves recognizing a segmentation site in the native protein, using information from sequence, secondary structure and fold that will yield at least two transporter polypeptides that efficiently form the quasi-native protein structure by self-assembling to form a heteromultimer together. For example, these split protein transporter polypeptides selectively self-assemble and form the quasi-native state when co-expressed. While generating a split protein complementary pair of transporter polypeptides, in a way, the attempt is to emulate a number of naturally occurring obligate protein-protein complexes that exhibit their functionality as a complex while being non-functional in their uncomplexed state.
  • a successful implementation of the strategy results in polypeptides that selectively self-assemble to form heteromultimers with each other, are soluble as individual entities and for functional relevance, do not impair the folding, binding and activity of other components in the environment.
  • the intrinsic nature of the polypeptides to reconstitute with each other has applications in area of creating heteromultimeric fusion entities out of cargo molecules that are not efficient at forming multimers by themselves.
  • the functional role of the split protein segments is to act as transporter polypeptides that drive heteromultimerization.
  • One of the critical requirements for such stable association is the formation of a large buried area of surface complementarity at the interface between the polypeptide chains.
  • the native fold of the original protein provides indication of the natural complementarity of regions within the protein.
  • FIG. 2 shows the solvent accessible surface area buried at the interface of two albumin-based polypeptides that would ideally fold into the quasi-native structure of HSA, when the segmentation point is moved along the protein sequence.
  • the analysis indicates that a large surface area, of the order of about 2000 ⁇ 2 is buried when the split segmentation is introduced anywhere between residues 30 and 520 with a few exceptions.
  • Albumin has an exceptionally large number of disulphides bridges that contributes to the stability of the native protein structure. Section of the protein near residues 110, 190, 300, 390 and 500 provide sites for segmentation that do not split the residues involved in a disulphide link across the two transporter polypeptides.
  • FIG. 3 presents a model representation of one such quasi-native albumin structure derived by removal of loop from residues 294 to 303 in the HSA sequence.
  • the total buried surface area for the two albumin based polypeptides of SEQ ID No. 2, and SEQ ID No: 3 shown herein is approximately 2500 ⁇ 2 . This is within the average range of 1910-3880 ⁇ 2 observed in a number of protein-protein heterodimeric and homodimeric co-complex structures [Bahadhur R P. & Zacharias M. (2008) Cell Mol Life Sci 65, 1059-1072]. his suggests that there is a strong likelihood for the two polypeptides to selectively associate with each other if the folding pathway of the two polypeptides is fairly independent of each other.
  • a stable quasi-native structure with the two polypeptides gives us the opportunity to employ these polypeptides to drive the formation of bispecific or other multifunctional molecules after fusing the appropriate cargo proteins of interest to the N or C terminus of the albumin based polypeptides employed as transporter polypeptides.
  • a number of other alternate segmentation patterns resulting in transportation polypeptide pair heterodimer can be designed.
  • the fused cargo proteins can be antigen binding domains or other payloads such as chemotoxins, radiotoxins or cytokines (as represented in FIG. 4 ).
  • the resulting heterodimers have many of the favorable properties intrinsic to HSA including properties like improved half-life, stability and low immunogenicity.
  • Traditional linkers such as (Gly 4 Ser) x can be used for the association of the cargo protein with the transporter polypeptide.
  • each of the HSA based transporter polypeptides is fused independently to the C-terminus of two heavy chains in a bispecific Fc molecule (as represented in FIG. 5 ).
  • the strong and selective pairing of the two transporter polypeptides (such as SEQ ID No. 2, and SEQ ID No. 3) drives the selectively heterodimerization of the Fc and also contribute to its stability and other valuable pharmacokinetic properties.
  • Residue 1-29 (EFATMAVMAPRTLVLLLSGALALTQTWAG) is the N-terminal export signal sequence region that gets cleaved. his sequence fulfills the same role as the natural signal sequence but it's optimized for mammalian and CHO cell lines.
  • SEQ ID No. 1 gi
  • albumin based polypeptides useful as transporter polypeptides are as follows:
  • the genes encoding the full length WT HA and the HA based transporter polypeptide monomers were constructed via gene synthesis using codons optimized for human/mammalian expression.
  • the constructs were designed from known full-length Human Serum Albumin Preprotein (GENEBANK: NP_000468.1), after exclusion of the signal sequence EFATMAVMAPRTLVLLLSGALALTQTWAG.
  • the final gene products were subcloned into the mammalian expression vector pTT5 (NRC-BRI, Canada) (Durocher et al). High level and high-throughput recombinant protein production by transient transfection of suspension-growing human CHO-3E7 was performed.
  • FIG. 6A provides SDS-PAGE (non-reducing) gel analysis of the two heteromultimer constructs (ABH1 and ABH2), after co-expression (different DNA transfection ratios are shown). WT full-length HSA is shown as control. As expected, ABH2 retains the disulfide linkage in non-reducing SDS-PAGE, with a MW roughly double the non-disulfide linked ABH1I.
  • FIG. 6A provides SDS-PAGE (non-reducing) gel analysis of the two heteromultimer constructs (ABH1 and ABH2), after co-expression (different DNA transfection ratios are shown). WT full-length HSA is shown as control. As expected, ABH2 retains the disulfide linkage in non-reducing SDS-PAGE, with a MW roughly double the non-disulfide linked ABH1I.
  • ABH1 and ABH2 both form a complex of expected mass, comparable to the full-length WT HSA. Furthermore, upon expression, neither the transporter polypeptides forming ABH1 nor the ones forming ABH2 homodimerize; rather they preferably form a stable hetercomplex. See Table 3 below for details.
  • WT-HSA and the two Albumin based heteromultimers were expressed in CHO-3E7 cell line grown in suspension in FreeStyle F17 medium (Invitrogen) supplemented with 0.1% w/v pluronic and 4 mM glutamine.
  • the day of transfection cell density should be around 1.5-2 million cells/ml and viability must be greater than 97%.
  • Transfection is done according to patent application WO 2009/137911 using a mixture of plasmid DNA made of 5% pTro-GFP plasmid (green fluorescent protein to determine transfection efficiency, Table 4), 15% pTT22-AKT plasmid, 21% HSA plasmids (10.63% of each), 68.37% of Salmon Sperm DNA.
  • the shake flask containing cells is then placed on an orbital shaker set to 120 rpm in a humidified incubator with 5% CO2 at 37° C.
  • Twenty-four hours post-transfection 1% w/v TN1 and 0.5 mM VPA (Valproic acid) are added to the cultures.
  • the cultures are then transferred on an orbital shaker (120 rpm) placed in a humidified incubator with 5% CO2 set at 32° C.
  • GFP positive cells should be between 30-60% as determined by flow cytometry.
  • Cells were harvested 7 days post-transfection and spun at 4,000 rpm for 20 minutes. The supernatant was filter-sterlized (clarified) using a 0.45 ⁇ m filter (Millipore).
  • FIG. 6C shows SDS-PAGE (non-reducing) analysis of the ABH2 heteromultimer and WT HSA, both after the final stage of purification. Both constructs show the expected MW.
  • Multivalent heteromultimer ABH2 was generated by expressing its single monomeric transporter polypeptides, SEQ ID NO: 8 and SEQ ID NO: 10, fused at one or both termini to cargo polypeptides that are either antiHer2scFv (4D5) and/or anti-CD16 scFv (NM3E). These form a set of 8 base construct monomers based off transporter polypeptide 1 and 8 base construct monomers based off transporter polypeptide 2. Different combinations of these base constructs were combined upon co-expression to form heteromultimers displaying all combination of the two cargo polypeptides at any of the four terminal positions of the two transporter polypeptides, ranging from monovalent to tetravalent.
  • bioactive cargo polypeptides were fused to the heteromultimer transporter polypeptides via a GGSG linker, for the N terminus of one monomer and a longer (GGS)4GG linker for all other termini in the other monomer.
  • Multivalent constructs were generated as outlined in Example 2 using heteromultimer ABH2.
  • the final gene products were subcloned into the mammalian expression vector pTT5 (NRC-BRI, Canada) (Durocher et al). High level and high-throughput recombinant protein production by transient transfection of suspension-growing human CHO-3E7 was performed.
  • Purification was performed by application of the cellular supernatant with expressed protein to a QIAGEN-tip 500 column packed with Blue Sepharose matrix (GE Healthcare) coupled to an AKTA Express system (GE Healthcare) using a flow rate of 1 ml/ml.
  • the column was equilibrated with equilibrated with sample buffer composed of 20 ml of PBS pH 7.2, 300 mM NaCl.
  • sample buffer composed of 20 ml of PBS pH 7.2, 300 mM NaCl.
  • the sample was loaded at a flow rate of 5 ml/min and subsequently washed with sample buffer.
  • the protein was eluted by application of NaCl gradient ranging from 300 mM to 2000 mM.
  • FIGS. 10A-10B shows SDS-PAGE (non-reducing) analysis of the ABH2 heteromultimer fused to different cargo polypeptides. The position of those polypeptides in the heteromultimer relative to the transporter polypeptides is outlined in table 8below. All constructs showed the expected molecular weight.
  • GLM sensorchips the Biorad ProteOn amine coupling kit (EDC, sNHS and ethanolamine), and 10 mM sodium acetate buffers were purchased from Bio-Rad Laboratories (Canada) Ltd. (Mississauga, ON). Recombinant Her-2 protein was purchased from eBioscience (San Diego, Calif.). HEPES buffer, EDTA, and NaCl were purchased from Sigma-Aldrich (Oakville, ON). 10% Tween 20 solution was purchased from Teknova (Hollister, Calif.).
  • a 4.0 ⁇ g/mL solution of CD16 in 10 mM NaOAc pH 4.5 was injected in the ligand (vertical) direction at a flow rate of 25 ⁇ L/min until approximately 3000 resonance units (RUs) were immobilized.
  • Remaining active groups were quenched by a 300 s injection of 1M ethanolamine at 30 ⁇ L/min in the analyte direction, and this also ensures mock-activated interspots are created for blank referencing.
  • a 500 nM 3-fold dilution series of V515 was injected over 3000 RUs CD16aWT (L6) compared to blank (L5).
  • Injections were repeated in standard running buffer (DPBS/3.4 mM EDTA/0.05% Tween20) and running buffer with an additional 350 mM NaCl.
  • Sensorgrams were aligned and double-referenced using the buffer blank injection and interspots, and the resulting sensorgrams were analyzed using ProteOn Manager software v3.0.
  • K D values were determined from binding isotherms using the Equilibrium Fit model.
  • kinetic and affinity values were additionally determined by fitting the referenced sensorgrams to the 1:1 Langmuir binding model using local R max , and affinity constants (K D M) were derived from the resulting rate constants (k d s ⁇ 1 /k a M ⁇ 1 s ⁇ 1 ). All K D values are reported as the mean and standard deviation from three independent runs.
  • ABH2 heteromultimer fused to a single antiCD16scFv has full activity and binds its target with good reproducibility and K D similar to the free anti CD16 scFv (NM3E).
  • the peptide sequence of the EGF-A domain in PCSK9 protein or another polypeptide sequence homologous to the EGF-A domain, capable of specifically binding the low density lipoprotein receptor (LDL-R) is derived by sequencing or from a database such as GenBank.
  • the cDNA for the cargo polypeptide comprising EGF-A like domain is isolated by a variety of means including but not exclusively, from cDNA libraries, by RT-PCR and by PCR using a series of overlapping synthetic oligonucleotide primers, all using standard methods.
  • the cargo protein is engineered to improve stability, target binding features or other biophysical or therapeutically relevant properties.
  • the polypeptide is employed as the cargo protein in the creation of a heteromultimer with application in the treatment of hypercholesterolemia.
  • the first and second monomeric fusion polypeptide sequence is derived by fusing the cargo protein sequence directly or with an intermediate linker peptide to the N-terminus and/or C-terminus of HA or HAA based transporter polypeptide such as SEQ ID No: 2, SEQ ID NO: 3, SEQ ID NO: 8 or SEQ ID NO: 10.
  • This monomeric fusion protein sequence is reverse translated to its corresponding DNA sequence to be introduced in an expression vector, sequence optimized for expression in a particular cell line of interest.
  • the first and second monomeric fusion proteins are transfected and coexpressed in the cell line of interest. In certain cases, the transfection is in 1:1 ratio for the two vectors. In some examples, the ratio is selected from 1.5:1, 2:1, 1:1.5, 1:2 etc.
  • the peptide sequence of GLP-1 or another polypeptide sequence homologous to this peptide, capable of specifically binding the GLP-1 receptor or acting as a GLP-1 agonist is derived by sequencing or from a database such as GenBank.
  • the peptide sequence of Glucagon or another polypeptide sequence homologous to this peptide, capable of specifically binding the Glucagon receptor or acting as a Glucagon receptor agonist is derived by sequencing or from a database such as GenBank.
  • each cargo polypeptide comprising GLP-1 or Glucagon is isolated by a variety of means including but not exclusively, from cDNA libraries, by RT-PCR and by PCR using a series of overlapping synthetic oligonucleotide primers, all using standard methods.
  • these GLP-1 or Glucagon based cargo polypeptides are engineered to improve stability, target binding features or other biophysical or therapeutically relevant properties.
  • These GLP-1 and Glucagon based polypeptides are employed as one or more cargo molecules in the creation of a heteromultimer with application in the treatment of type-2 diabetes or another disease related to glucose metabolism.
  • the first and second monomeric fusion polypeptide sequence is derived by fusing the cargo protein sequence directly or with an intermediate linker peptide to the N-terminus and/or C-terminus of HA or HAA based transporter polypeptide such as SEQ ID No: 2, SEQ ID NO: 3, SEQ ID NO: 8 or SEQ ID NO: 10.
  • the fusion proteins can be monospecific with either GLP-1 or Glucagon like polypeptides or be bispecific (coagonist) with both the GLP-1 and Glucagon like polypeptides.
  • Each monomeric fusion protein sequence is reverse translated to its corresponding DNA sequence to be introduced in an expression vector, sequence optimized for expression in a particular cell line of interest.
  • the first and second monomeric fusion proteins are transfected and coexpressed in the cell line of interest. In certain cases, the transfection is in 1:1 ratio for the two vectors. In some examples, the ratio is selected from 1.5:1, 2:1, 1:1.5, 1:2 etc
  • Annexin is split with an extensive interface to generate a multivalent heteromultimer scaffold comprising two transporter polypeptides.
  • Annexin is a 346 residue protein (PDB ID 1MCX).
  • PDB ID 1MCX 346 residue protein
  • Heteromultimer comprising two transporter polypeptides based on annexin split in the region between residue 186 and 194 is shown in FIG. 11 .
  • the large interfacial area between the two transporter polypeptides leads to self-assembly of the heterodimer.
  • the self-assembly of the two units allows for the design of multivalent construct with transporter polypeptides based on the annexin core. Two structures are available, Pig and Human.
  • the two structures are superimposable with an rmsd of 0.6 A.
  • the following stretch of sequence can be removed from the human Annexin sequence DRSEDF (residues 160 through 165).
  • the truncation does not break any secondary structure element and does not involve introducing or removing any Proline residue.
  • FIG. 12 shows a plot of the buried solvent accessible surface area at the interface of Annexin based transporter polypeptide-1 (ABT-1), and Annexin based transporter polypeptide-2 (ABT-2).
  • a split annexin near residue position 186 forms a heterodimer with about 3200 ⁇ 2 of buried surface area.
  • the transporter polypeptides such as ABT-1 and ABT-2 based on Annexin can be used to attach cargo biomolecules using the same methods as described above for albumin based transporter polypeptides.
  • transferrin Based on the large number of therapeutically relevant properties of transferrin, this protein presents itself as an interesting scaffold molecule for the design of multivalent protein fusion drugs following the creation of a self-assembling protein and its split component parts.
  • the structure of transferrin is shown in FIG. 13 based on the crystal structure (1H76) available in the protein data bank [Hall D R et al. Acta Crystallogr D 2002, 58, 70-80].
  • the transferrin molecule is composed of two structurally similar lobes, the N and C terminal lobes, connected by a short peptide linker between residues 333 and 342.
  • a heterodimer is designed based on transferrin protein, said heterodimer comprising a first transporter polypeptide involving residues 1-333 of transferrin and a second transporter polypeptide composed of residues from 342 to the C-terminus of the original transferrin sequence.
  • the two transporter polypeptides fold independently and pair to form a quasi-transferrin scaffold capable of maintaining its therapeutically relevant properties.
  • a Transferrin scaffold allows for the production of multivalent fusion molecules, e.g. a multivalent GLP-1 fusion with transporter polypeptides based on transferring. These fusions can be similar to the GLP-1-fusion polypeptides with Albumin based transporter polypeptides.
  • FIG. 13 provides structure of transferrin molecule based on the PDB structure 1H76.
  • the two monomeric transporter polypeptides derived by splitting the transferrin molecule are color coded as light and dark grey units.
  • the sites of fusion for the cargo molecules are represented as spheres.
  • FIG. 14 shows a plot of the buried solvent accessible surface area at the interface of two transferrin based polypeptides.
  • a split transferrin near residue position 330 such as the two transporter polypeptides shown below, forms a heterodimer with about 1800 A2 of buried surface area.
  • Transferrin based transporter polypeptide-1 SEQ ID NO: 17: MRLAVGALLV CAVLGLCLAV PDKTVRWCAV SEHEATKCQS FRDHMKSVIP SDGPSVACVK KASYLDCIRA IAANEADAVT LDAGLVYDAY LAPNNLKPVV AEFYGSKEDP QTFYYAVAVV KKDSGFQMNQ LRGKKSCHTG LGRSAGWNIP IGLLYCDLPE PRKPLEKAVA NFFSGSCAPC ADGTDFPQLC QLCPGCGCST LNQYFGYSGA FKCLKDGAGD VAFVKHSTIF ENLANKADRD QYELLCLDNT RKPVDEYKDC HLAQVPSHTV VARSMGGKED LIWELLNQAQ EHFGKDKSKE FQLFSSPHGK DLLFKDSAHG FLKVPPRMDA KMYLGYEYVT AIRNLREG.
  • Transferrin based transporter polypeptide-2 SEQ ID NO: 18: ECKPVKWCALSHHE RLKCDEWSVN SVGKIECVSA ETTEDCIAKI MNGEADAMSL DGGFVYIAGK CGLVPVLAEN YNKSDNCEDT PEAGYFAVAV VKKSASDLTW DNLKGKKSCH TAVGRTAGWN IPMGLLYNKI NHCRFDEFFS EGCAPGSKKD SSLCKLCMGS GLNLCEPNNK EGYYGYTGAF RCLVEKGDVA FVKHQTVPQN TGGKNPDPWA KNLNEKDYEL LCLDGTRKPV EEYANCHLAR APNHAVVTRK DKEACVHKIL RQQQHLFGSN VTDCSGNFCL FRSETKDLLF RDDTVCLAKL HDRNTYEKYL GEEYVKAVGN LRKCSTSSLL EACTFRRP.
  • heteromultimer proteins described herein may additionally be fused to other proteins to generate “multifusion proteins”.
  • These multifusion proteins can be used for a variety of applications. For example, fusion of the proteins described herein to His-tag IgG domains, and maltose binding protein facilitates purification. (See e.g EP A 394,827; Traunecker et al., Nature 331:84-86 (1988)).
  • Nuclear localization signals fused to the polypeptides can target the protein to a specific subcellular localization.
  • heteromultimer proteins described herein may further increase the solubility and/or stability of the heteromultimer.
  • the heteromultimer proteins described above can be made using or routinely modifying techniques known in the art and/or by modifying the following protocol, which outlines the fusion of a polypeptide to an IgG molecule.
  • the human Fc portion of the IgG molecule can be PCR amplified, using primers that span the 5′ and 3′ ends of the sequence described below. These primers also should have convenient restriction enzyme sites that will facilitate cloning into an expression vector, preferably a mammalian or yeast expression vector.
  • the human Fc portion can be ligated into the BamHI cloning site. Note that the 3′ BamHI site should be destroyed.
  • the vector containing the human Fc portion is re-restricted with BamHI, linearizing the vector, and a polynucleotide encoding a heteromultimeric protein described herein (generated and isolated using techniques known in the art), is ligated into this BamHI site. Note that the polynucleotide encoding the fusion protein of the invention is cloned without a stop codon; otherwise an Fc containing fusion protein will not be produced.
  • pC4 does not need a second signal peptide.
  • the vector can be modified to include a heterologous signal sequence. (See, e.g., International Publication No. WO 96/34891.)

Abstract

Provided herein are multifunctional heteromer proteins. In specific embodiments is a heteromultimer that comprises: at least two monomeric proteins, wherein each monomeric protein comprises at least one cargo polypeptide, attached to a transporter polypeptide, such that said monomeric proteins associate to form the heteromultimer. These therapeutically novel molecules comprise monomers that function as scaffolds for the conjugation or fusion of therapeutic molecular entities resulting in the creation of bispecific or multivalent molecular species.

Description

    CROSS REFERENCE TO RELATED APPLICATIONS
  • This application is a continuation of U.S. application Ser. No. 13/411,353, filed Mar. 2, 2012, now U.S. Pat. No. 9,499,605, which claims the benefit of U.S. Provisional Patent Application No. 61/449,016, filed Mar. 3, 2011, which are incorporated herein by reference in their entireties.
  • FIELD OF INVENTION
  • The field of the invention is the rational design of a scaffold for custom development of biotherapeutics.
  • DESCRIPTION OF RELATED ART
  • In the realm of therapeutic proteins, antibodies with their multivalent target binding features are excellent scaffolds for the design of drug candidates. Advancing these features further, designed bispecific antibodies and other fused multispecific therapeutics exhibit dual or multiple target specificities and an opportunity to create drugs with novel modes of action. The development of such multivalent and multispecific therapeutic proteins with favorable pharmacokinetics and functional activity has been a challenge.
  • Human serum albumin (HSA, or HA), a protein of 585 amino acids in its mature form is responsible for a significant proportion of the osmotic pressure of serum and also functions as a carrier of endogenous and exogenous ligands. The role of albumin as a carrier molecule and its stable nature are desirable properties for use as a carrier and transporter of polypeptides in vivo.
  • Human serum albumin possesses many desirable characteristics. HSA is found throughout the body, but more specifically in the interstitial space and in blood at serum concentrations of 40 g/L which is equivalent to 0.7 mM (Yeh et al., Proc. Natl. Acad. Sci. USA, 89:1904-1908 (1992)). HSA is considered to be the most abundant protein of the serum and is responsible for maintaining osmolarity. HSA has favorable pharmacokinetic properties and is cleared very slowly by the liver and kidney displaying in vivo half-lives up to several weeks (Yeh et al., Proc. Natl. Acad. Sci. USA, 89:1904-1908 (1992); Waldmann, T. A., Albumin Structure, Function and Uses, pp. 255-273 (1977); Sarav et al., J Am Soc Nephrol 20:1941-1952(2009)). HSA lacks enzymatic activity and antigenicity thereby eliminating potentially undesirable side effects. HSA acts as a carrier for endogenous as well as exogenous ligands. Combined, these features can be extended, at least partially, onto albumin based fusion protein. The poor pharmacokinetic properties displayed by therapeutic proteins can then be circumvented.
  • SUMMARY OF THE INVENTION
  • Provided herein are multifunctional heteromultimers and methods to design them. In certain embodiments are heteromultimers, each heteromultimer comprising: at least a first monomer unit that comprises at least one cargo molecule, and a first transporter polypeptide; and at least a second monomer unit that comprises at least one cargo molecule and a second transporter polypeptide; wherein at least one transporter polypeptide is derived from a monomeric protein and wherein said transporter polypeptides self-assemble to form a quasi-native structure of said monomeric protein or analog thereof. In certain embodiments, at least one cargo molecule is a drug, or a therapeutic agent. In certain embodiments, at least one cargo molecule is a biomolecule. In an embodiment, the at least one biomolecule is a DNA, RNA, PNA or polypeptide. In an embodiment, at least one cargo molecule is a polypeptide. In certain embodiments, each monomeric transporter polypeptide is unstable and preferentially forms a heteromultimer with at least one other transporter polypeptide. In certain embodiments, each monomeric transporter polypeptide is stable and preferentially forms a heteromultimer with at least one other transporter polypeptide. In certain embodiments, the heteromultimerization interface comprises at least one disulfide bond. In certain embodiments, the heteromultimerization interface does not comprise a disulfide bond.
  • In specific embodiments is a heteromultimer that comprises: at least two monomers, wherein each monomer comprises at least one cargo molecule attached to a transporter polypeptide, such that said monomers self-assemble to form the heteromultimer. In certain embodiments is a heteromultimer that comprises: at least two monomeric proteins, wherein each monomeric protein comprises at least one cargo polypeptide, attached to a transporter polypeptide, wherein at least one transporter polypeptide is derived from a monomeric protein and wherein said transporter polypeptides self-assemble to form a quasi-native structure of said monomeric protein or analog thereof. In certain embodiments is a heteromultimer that comprises: at least two monomeric proteins, wherein each monomeric protein comprises at least one cargo polypeptide attached to a transporter polypeptide, such that said monomeric proteins self-assemble via the transporter polypeptide to form the heteromultimer, and wherein at least one transporter polypeptide is derived from a monomeric protein and wherein said transporter polypeptides self-assemble to form a quasi-native structure of said monomeric protein or analog thereof. In certain embodiments, the heteromultimer is a heterodimer. In an embodiment, the heteromultimer is bispecific. In an embodiment, the heteromultimer is multispecific. In certain embodiments, the heteromltimer is bivalent. In an embodiment the heteromultimer is multivalent. In an embodiment, the heteromultimer is multifunctional. In certain embodiments, at least one transporter polypeptide is not derived from an antibody. In certain embodiments, the transporter polypeptides are not derived from an antibody. In certain embodiments, the transporter polypeptides are derivatives of albumin. In certain embodiments of the hetermultimer described herein, the transporter polypeptides are derived from human serum albumin (HSA or HA) of SEQ ID No. 1. In certain embodiments of the hetermultimer described herein, the transporter polypeptides are derived from alloalbumins (HAA). In certain embodiments of the hetermultimer described herein, the transporter polypeptides are derived from sequence homologous to the human serum albumin (HSA or HA) of SEQ ID No. 1.
  • In some embodiments of the heteromultimer described herein, the transporter polypeptides are derivatives of an annexin protein. In an embodiment, the transporter polypeptides are derived from different annexin proteins. In certain embodiments, the transporter polypeptides are derived from the same annexin protein. In an embodiment, at least one transporter polypeptide is derived from Annexin A1 or lipocortin I. In certain embodiments of the heteromultimer, all transporter polypeptides are derived from Annexin A1 of SEQ ID NO: 14. In certain embodiments of the heteromultimer, at least one transporter polypeptides is derived from a sequence homologous to SEQ ID NO: 14. In an embodiment, at least one transporter polypeptide is derived from Annexin A2 or annexin II. In certain embodiments of the heteromultimer, all transporter polypeptides are derived from Annexin A2 or lipocortin II. In an embodiment, at least one transporter polypeptide is derived from Annexin like protein. In certain embodiments of the heteromultimer, all transporter polypeptides are derived from Annexin like protein. In an embodiment, at least one transporter polypeptide is derived from the group comprising Annexin A1-Annexin A7. In an embodiment of the heteromultimer described herein, all transporter polypeptides are derived from the group comprising Annexin A1-Annexin A7. 14. In certain embodiments, the first annexin based transporter polypeptide has a sequence comprising SEQ ID NO:15, and the second annexin based transporter polypeptide has a sequence comprising SEQ ID NO: 16.
  • In some embodiments of the heteromultimer described herein, the transporter polypeptides are derivatives of transferrin. In an embodiment, at least one transporter polypeptide is derived from transferrin. In certain embodiments of the heteromultimer, at least one transporter polypeptides are derived from transferrin of SEQ ID NO: 19 or analog thereof. In certain embodiments of the heteromultimer, at least one transporter polypeptide is derived from a polypeptide sequence homologous to the transferrin. In certain embodiments of the heteromultimer described herein, at least one transporter polypeptide is derived from apo-transferrin. In certain embodiments, the first transferrin based transporter polypeptide has a sequence comprising SEQ ID NO:15 and the second transferrin based transporter polypeptide has a sequence comprising SEQ ID NO: 16.
  • In certain embodiments of the heteromultimer, at least one cargo molecule is a cargo polypeptide. In an embodiment of the heteromultimer described herein, all cargo molecules are cargo polypeptides. In certain embodiments, the cargo polypeptides are therapeutic proteins or fragments or variants thereof. In certain embodiments, the cargo polypeptides are antigens or fragments or variants thereof. In certain embodiments, the cargo polypeptides are antigen receptors or fragments or variants thereof. In some embodiments, the cargo polypeptide is an antibody, an antibody domain, a ligand or a receptor that binds a target polypeptide. In some embodiments, at least one cargo polypeptide is fused to the transporter polypeptide. In certain embodiments, at least one cargo polypeptide is attached to the N-terminus of the transporter polypeptide. In some embodiments, at least one cargo polypeptide is attached to the C-terminus of the transporter polypeptide. In some embodiments, at least one cargo polypeptide is chemically linked to the transporter polypeptide. In some embodiments of the heteromultimers described herein, at least one cargo polypeptide comprises GLP-1 or fragment or variant thereof. In some embodiments, at least one cargo polypeptide comprises glucagon or fragment or variant thereof. In an embodiment, at least one cargo polypeptide comprises an EGF-A like domain.
  • Provided herein are heteromultimers, each heteromultimer comprising: at least a first monomeric protein that comprises at least one cargo polypeptide and a first transporter polypeptide; and at least a second monomeric protein that comprises at least one cargo polypeptide and a second transporter polypeptide. In certain embodiments, the heteromultimer is a heterodimer. In an embodiment, the heteromultimer is multispecific. In an embodiment, the heteromultimer is bispecific. In certain embodiments of the heteromultimer, the transporter polypeptides are derivatives of the same protein. In certain embodiments, the transporter polypeptides are derivatives of albumin. In certain embodiments of the hetermultimer described herein, the transporter polypeptides are derived from human serum albumin of SEQ ID No. 1. In certain embodiments, the transporter polypeptides are derivatives of an annexin. In an embodiment, the transporter polypeptides are derivatives of Annexin A2. In some embodiments, the transporter polypeptides are derivatives of transferrin.
  • In certain embodiments, are heteromultimers, each heteromultimer comprising: at least a first monomeric protein that comprises at least one cargo polypeptide and a first transporter polypeptide comprising a first segment of human serum albumin; and at least a second monomeric protein that comprises at least one cargo polypeptide, fragment and a second transporter polypeptide comprising a second segment of human serum albumin; wherein said transporter polypeptides self-assemble to form a quasi-native structure of albumin or analog thereof. In certain embodiments, the first and second segments of human serum albumin are from non-overlapping regions of the protein. In certain embodiments, there is an overlap between the sequences of the first and second segments of human serum albumin. In some embodiments, the overlap is a 5% overlap. In an embodiment, the overlap is a 10% overlap. In certain embodiments, the first segment of human serum albumin comprises a sequence of SEQ ID NO:2, and the second segment of human serum albumin comprises a sequence of SEQ ID NO: 3. In certain embodiments, the first segment of human serum albumin comprises a sequence of SEQ ID NO:8, and the second segment of human serum albumin comprises a sequence of SEQ ID NO: 10.
  • In certain embodiments, are heteromultimers, each heteromultimer comprising: at least a first monomeric protein that comprises at least one cargo polypeptide and a first transporter polypeptide comprising a sequence of SEQ ID NO:2; and at least a second monomeric protein that comprises at least one cargo polypeptide, and a second transporter polypeptide comprising a sequence of SEQ ID NO: 3. In certain embodiments, are heteromultimers, each heteromultimer comprising: at least a first monomeric protein that comprises at least one cargo polypeptide and a first transporter polypeptide comprising a sequence of SEQ ID NO:8; and at least a second monomeric protein that comprises at least one cargo polypeptide and a second transporter polypeptide comprising a sequence of SEQ ID NO: 10. In certain embodiments of the hetermultimer described herein, at least one transporter polypeptide is derived from alloalbumins. In certain embodiments, both transporter polypeptides are derived from alloalbumins. In certain embodiments, all transporter polypeptides are derivatives of the same alloalbumin. In some other embodiments, the transporter polypeptides are derivatives of different alloalbumins. In some embodiments, each transporter polypeptide is an alloalbumin derivative based on an alloalbumin selected from Table 2. In certain embodiments, the first monomeric protein comprises two cargo polypeptides. In some embodiments, the second monomeric protein comprises two cargo polypeptides. In some embodiment, at least one of the monomeric proteins is engineered by introducing mutations. In certain embodiments, the introduced mutations improve the functionality of the monomeric protein as compared to the native, non-mutated form of the monomer. In certain embodiments the introduced mutations improve one or more of the stability, half-life and heteromultimer formation of the transporter polypeptide.
  • Provided herein are heteromultimers, each heteromultimer comprising: at least a first monomeric protein that comprises at least one cargo polypeptide and a first transporter polypeptide; and at least a second monomeric protein that comprises at least one cargo polypeptide and a second transporter polypeptide. In certain embodiments, at least one cargo polypeptide is selected from the proteins listed in Table 2 or fragments, variants or derivatives thereof. In certain embodiments, at least one cargo polypeptide is selected from ligand, receptor, or antibody to one or more proteins listed in Table 2, or fragment, variant or derivative of said ligand, receptor or antibody. In certain embodiments, at least one cargo polypeptide targets a cell surface antigen from the group consisting of CD19, CD20, CD22, CD25, CD30, CD33, CD40, CD56, CD64, CD70, CD74, CD79, CD105, Cd138, CD174, CD205, CD227, CD326, CD340, MUC16, GPNMB, PSMA, Cripto, ED-B, TMEFF2, EphB2, EphA2, FAP, integrin, Mesothelin, EGFR, TAG-72, GD2, CAIX, 5T4. In certain embodiments, are heteromultimers, each heteromultimer comprising: at least a first monomeric protein that comprises at least one cargo polypeptide and a first transporter polypeptide; and at least a second monomeric protein that comprises at least one cargo polypeptide and a second transporter polypeptide, wherein at least one at least one cargo polypeptide is an antibody, or fragment or variant thereof. In certain embodiments, all cargo polypeptides are antibodies or fragments or variants thereof. In some embodiments, the cargo polypeptide is an antibody that binds to a protein listed in Table 2. In some embodiments, the antibody fragment comprises antibody Fc or Fab or Fv region. In some embodiment the cargo polypeptide is a non-antibody protein like nanobodies, affibody, maxibody, adnectins, domain antibody, evibody, ankyrin repeat proteins, anticalins, camlids or ligand protein or polypeptide binding to a therapeutically relavant target. In some embodiments, the antibody or its fragment is derived from an immunoglobulin selected from the group consisting of IgG, IgA, IgD, IgE, and IgM. In certain embodiments, the IgG is of subtype selected from IgG1, IgG2a, IgG2b, IgG3 and IgG4. In certain embodiments, the antibody is multispecific.
  • Provided herein are heteromultimers, each heteromultimer comprising: at least a first monomeric protein that comprises at least one cargo polypeptide and a first transporter polypeptide; and at least a second monomeric protein that comprises at least one cargo polypeptide and a second transporter polypeptide, wherein at least one cargo polypeptide is a therapeutic antibody. In some embodiments of the heteromultimers described herein, at least one cargo polypeptide is a therapeutic antibody or fragment or variant thereof, wherein the antibody is selected from antibody is selected from abagovomab, adalimumab, alemtuzumab, aurograb, bapineuzumab, basiliximab, belimumab, bevacizumab, briakinumab, canakinumab, catumaxomab, certolizumab pegol, certuximab, daclizumab, denosumab, efalizumab, galiximab, gemtuzumab ozagamicin, golimumab, ibritumomab tiuxetan, infliximab, ipilimumab, lumiliximab, mepolizumab, motavizumab, muromonab, mycograb, natalizumab, nimotuzumab, ocrelizumab, ofatumumab, omalizumab, palivizumab, panitumumab, pertuzumab, ranizumab, reslizumab, rituximab, teplizumab, toclizumab, tositumomab, trastuzumab, Proxinium, Rencarex, ustekinumab, and zalutumumab. In certain embodiments, the therapeutic antibody binds a disease related target antigen such as cancer antigen, inflammatory disease antigen or a metabolic disease antigen. In certain embodiments, the target antigen could be a protein on a cell surface and the cell could belong to the group of B-cell, T-cell, stromal cell, endothelial cell, vascular cell, myeloid cell, hematopoietic cell or carcinoma cell.
  • Provided herein are heteromultimers, each heteromultimer comprising: at least a first monomer that comprises at least one cargo molecule, fragment; and at least a second monomer that comprises at least one cargo molecule and a second transporter polypeptide, wherein at least one cargo polypeptide is an enzyme, enzyme inhibitor, hormone, therapeutic polypeptide, antigen, radiotoxin and chemotoxin inclusive of but not restricted to neurotoxins, interferons, cytokine fusion toxins and chemokine fusion toxins, cytokine, antibody fusion protein or variant or fragment thereof. In some embodiments of the heteromultimers described herein, at least one cargo polypeptide comprises GLP-1 or fragment or variant thereof. In some embodiments, at least one cargo polypeptide comprises glucagon or fragment or variant thereof. In an embodiment, at least one cargo polypeptide comprises an EGF-A like domain. In certain embodiments, the toxin is an immunotoxin such as Denileukin diftitox and Anti-CD22 immunotoxin such as CAT-3888 and CAT-8015. In certain embodiments, the toxin is saporin. In some embodiments, the toxin is a mitotoxin. In some embodiments, the toxin is a diphtheria toxin. In some embodiments, the toxin is botulinux toxin type A. In some embodiments, the toxin is ricin or a fragment there of. In some embodiments, the toxin is a toxin from RTX family of toxins.
  • Provided herein are heteromultimers, each heteromultimer comprising: at least a first monomeric protein that comprises at least one cargo polypeptide and a first transporter polypeptide; and at least a second monomeric protein that comprises at least one cargo polypeptide and a second transporter polypeptide, wherein the cargo polypeptide is attached to the transporter polypeptide by chemical conjugation, native ligation, chemical ligation, a disulfide bond or direct fusion or fusion via a linker. In certain embodiments, linkers for attaching cargo molecules such as cargo polypeptides to transporter polypeptides are selected from the linkers described in U.S. Pat. Nos. 5,482,858, 5,258,498 and 5,856,456, US2009060721, U.S. Pat. Nos. 6,492,123, 4,946,778, 5,869,620, 7,385,032, 5,073,627, 5,108,910, 7,977,457, 5,856,456, 7,138,497, U.S. Pat. Nos. 5,837,846, 5,990,275, EP1088888 incorporated by reference herein.
  • Provided herein are host cells comprising nucleic acid encoding a heteromultimer described herein. In certain embodiments, the nucleic acid encoding the first monomeric protein and the nucleic acid encoding the second monomeric protein are present in a single vector. In certain embodiments, the nucleic acid encoding the first monomeric protein and the nucleic acid encoding the second monomeric protein are present in separate vectors.
  • Provided herein is a method of making a heteromultimer, wherein said method comprises: culturing a host cell described herein such that the nucleic acid encoding a heteromultimer described herein is expressed; and recovering the heteromultimer from the cell culture. In some embodiments, the host cell is a prokaryotic cell or a eukaryotic cell. In some embodiments, the host cell is E. coli. In certain embodiments, the host cell is yeast cell. In some embodiments, the yeast is S. cerevisiae. In some embodiments, the yeast is Pichia. In a certain embodiment, the yeast is Pichia pastoris. In some embodiments, the yeast is glycosylation deficient, and/or protease deficient. In some embodiments, the host cell is a bacterial cell. In some embodiments, the host cell expressing a heteromultimer descried herein is a mammalian cell. In certain embodiments, the mammalian cell is a CHO cell, a BHK cell, NSO cell, COS cell or a human cell.
  • Provided is a pharmaceutical composition that comprises a heteromultimer described herein and a pharmaceutically acceptable adjuvant. Also provided are methods of treating an individual suffering from a disease or disorder, said method comprising administering to the individual an effective amount of a formulation or pharmaceutical composition described herein. In certain embodiments is a method of treating cancer in a patient, said method comprising administering to the patient a therapeutically effective amount of a heteromultimer described herein. In some embodiments is a method of treating an immune disorder in a patient, said method comprising administering to the patient a therapeutically effective amount of a heteromultimer described herein. Also provided is a method of treating an infectious disease in a patient, said method comprising administering to the patient a therapeutically effective amount of a heteromultimer described herein. In certain embodiments is a method of treating a cardiovascular disorder in a patient, said method comprising administering to the patient a therapeutically effective amount of a heteromultimer described herein. In certain embodiments is a method of treating a respiratory disorder in a patient, said method comprising administering to the patient a therapeutically effective amount of a heteromultimer described herein. In certain embodiments is a method of treating a metabolic disorder in a patient, said method comprising administering to the patient a therapeutically effective amount of a heteromultimer described herein. In certain embodiments is a method of treating one or more of Congenital adrenal hyperplasia, Gaucher's disease, Hunter syndrome, Krabbe disease, Metachromatic leukodystrophy, Niemann-Pick disease, Phenylketonuria (PKU), Porphyria, Tay-Sachs disease, and Wilson's disease in a patient, said method comprising administering to the patient a therapeutically effective amount of a heteromultimer described herein.
  • Provided is a kit for detecting the presence of a biomarker of interest in an individual, said kit comprising (a) an amount of a heteromultimer described herein, wherein said heteromultimer comprises at least one cargo polypeptide such that said cargo polypeptide is capable of binding to the biomarker of interest; and (b) instructions for use.
  • Provided herein are heteromultimer proteins that comprise at least two monomeric proteins, wherein each monomeric protein comprises at least one cargo polypeptide, and an albumin based polypeptide, such that said monomeric proteins self-assemble to form the heteromultimer.
  • In certain embodiments, the cargo polypeptide is fused to the albumin or alloalbumin based transporter polypeptide. In some embodiments, the cargo polypeptide is fused to the transferrin based transporter polypeptide. In certain embodiments, the cargo polypeptide is fused to the annexin based transporter polypeptide. In some embodiments, the fusion is at the N terminus of the transporter polypeptide. In certain embodiments, the fusion is at the C terminus of the transporter polypeptide. In some embodiments, the fusion involves a bridging linker or spacer molecule. In some embodiments, the cargo polypeptide is chemically conjugated to the transporter polypeptide. In certain embodiments, the cargo polypeptide is attached to the transporter polypeptide by means of chemical ligation or a disulfide bond.
  • Provided herein are heteromultimer proteins that comprise at least two monomeric proteins, wherein each monomeric protein comprises at least one cargo polypeptide, and a transporter polypeptide, such that said transporter polypeptides self-assemble to form the heteromultimer. In some embodiments, each transporter polypeptide is an alloalbumin based polypeptide, such that said alloalbumin based polypeptides self-assemble to form the heteromultimer. In some embodiments, each transporter polypeptide is a transferrin based polypeptide. In some embodiments, each transporter polypeptide is an annexin based polypeptide. In certain embodiments, each monomeric transporter polypeptide is unstable and preferentially forms a heteromultimer with at least one other transporter polypeptide.
  • In some embodiments, a heteromultimer described herein is a heterodimer. In some embodiments cargo polypeptide is an antibody, enzyme, hormone, therapeutic polypeptide, antigen, chemotoxin, radiotoxin, cytokine or variant or fragment thereof. In some embodiments, the cargo polypeptide of one monomeric protein functions in synergy with the cargo polypeptide of another monomeric protein.
  • Provided herein are heteromultimer proteins that comprise at least two monomeric proteins, wherein each monomeric protein comprises at least one cargo polypeptide, and an annexin based polypeptide, such that said annexin based polypeptides self-assemble to form the heteromultimer with a quasi-native structure of annexin or analog thereof. In some embodiments, the annexin is Annexin A1. In some embodiments, a heteromultimer described herein is a heterodimer. In some embodiments cargo polypeptide is an antibody, enzyme, hormone, therapeutic polypeptide, antigen, chemotoxin, radiotoxin, cytokine, ligand to a receptor, receptor or variant or fragment thereof. In some embodiments, the cargo polypeptide of one monomeric protein functions in synergy with the cargo polypeptide of another monomeric protein. In some embodiments the cargo polypeptide can be an agonist or antagonist to the cargo polypeptide of another monomeric protein.
  • Provided herein are heterodimer proteins that comprise at least two monomeric fusion proteins, wherein each monomeric fusion proteins comprises at least one cargo polypeptide fused to an albumin derived polypeptide, such that said albumin derived polypeptides self-assemble to form the multifunctional heterodimer. In certain embodiments are heterodimeric proteins comprising a first monomer which comprises at least one cargo polypeptide fused to an albumin derived polypeptide; and a second monomer that comprises at least one cargo polypeptide fused to an albumin derived polypeptide. In certain embodiments, the at least one cargo polypeptide of the first monomer is different from the at least one cargo polypeptide of the second monomer. In certain embodiments, the at least one cargo polypeptide of the first monomer is the same as the at least one cargo polypeptide of the second monomer.
  • In certain embodiments are heteromultimer proteins that comprise at least two monomeric fusion proteins, wherein each monomeric fusion proteins comprises at least one cargo polypeptide fused to an alloalbumin derived polypeptide, such that said alloalbumin derived polypeptides self-assemble to form the multifunctional heteromultimer. In certain embodiments are heteromultimer proteins that comprise at least two monomeric fusion proteins, wherein each monomeric fusion proteins comprises at least one cargo polypeptide fused to a transferrin derived polypeptide, such that said transferrin derived polypeptides self-assemble to form the heteromultimer. In certain embodiments are heteromultimer proteins that comprise at least two monomeric fusion proteins, wherein each monomeric fusion proteins comprises at least one cargo polypeptide fused to an annexin derived polypeptide, such that said annexin derived polypeptides self-assemble to form the heteromultimer. In certain embodiments, the annexin is Annexin A2.
  • In certain embodiments are heteromultimer proteins comprising a first monomer which comprises at least one cargo polypeptide fused to an alloalbumin derived polypeptide; and a second monomer that comprises at least one cargo polypeptide fused to an alloalbumin derived polypeptide. In certain embodiments, the at least one cargo polypeptide of the first monomer is different from the at least one cargo polypeptide of the second monomer. In certain embodiments, the at least one cargo polypeptide of the first monomer is the same as the at least one cargo polypeptide of the second monomer.
  • Provided herein is a heteromultimer that comprises: at least two monomers, each comprising a transporter polypeptide and optionally at least one cargo molecule attached to said transporter polypeptide, wherein each transporter polypeptide is obtained by segmentation of a whole protein such that said transporter polypeptides self-assemble to form quasi-native whole protein. In certain embodiments, the heteromultimer is multispecific. In certain embodiments, the transporter polypeptides are not derived from an antibody. In some embodiments, each monomer preferentially forms the heteromultimer as compared to a monomer or a homomultimer. In an embodiment of the heteromultimer, at least one cargo molecule is a therapeutic agent, or a biomolecule. In some embodiments, at least one cargo molecule is a biomolecule which is selected from a polypeptide, DNA, PNA, or RNA. In some embodiments, each transporter polypeptide is a derivate of albumin or alloalbumin. In an embodiment, each transporter polypeptide is a derivate of annexin. In certain embodiments, each transporter polypeptide is a derivate of transferrin.
  • In certain embodiments are pharmaceutical formulations that comprise an albumin-based and/or alloalbumin-based heteromultimeric protein described herein and a pharmaceutically acceptable diluent or carrier. In certain embodiments are pharmaceutical formulations that comprise a transferrin-based heteromultimeric protein described herein and a pharmaceutically acceptable diluent or carrier. In certain embodiments are pharmaceutical formulations that comprise an annexin-based heteromultimeric protein described herein and a pharmaceutically acceptable diluent or carrier. In certain embodiments are pharmaceutical formulations that comprise an Annexin-A2 based heteromultimeric protein described herein and a pharmaceutically acceptable diluent or carrier. In certain embodiments, a formulation described herein is provided as part of a kit or container. In certain embodiments, the kit or container is packaged with instructions pertaining to extended shelf life of the therapeutic protein. In some embodiments, a heteromultimer described herein is used in a method of treating (e.g., ameliorating) preventing, or diagnosing a disease or disease symptom in an individual, comprising the step of administering said formulation to the individual.
  • Provided herein is a method of obtaining fusion protein scaffolds with a known number of conjugation sites based on any transport protein of interest.
  • Also provided are transgenic organisms modified to contain nucleic acid molecules described herein to encode and express monomeric fusion proteins described herein.
  • Other aspects and features of the present invention will become apparent to those ordinarily skilled in the art upon review of the following description of specific embodiments of the invention in conjunction with the accompanying figures.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • In drawings which illustrate embodiments of the invention,
  • FIG. 1 depicts the structure of the Human Serum Albumin (HSA) molecule. The alpha helical sections of the secondary structure are shown schematically along with the bonds represented as sticks.
  • FIG. 2 is a plot of buried solvent accessible surface area at the interface of two albumin-based polypeptides.
  • FIG. 3 depicts two albumin-based polypeptides expressed separately. The two polypeptides are shown in light and dark grey respectively. Each polypeptide comprises two fusion sites for functional cargo proteins and these sites are represented as spheres. The disulphide residues in structure are shown as sticks.
  • FIG. 4 is a schematic representation of bispecific and other multifunctional therapeutics based on the multispecific heteromultimer described herein. The albumin-based, or alloalbumin-based polypeptides are denoted A1 and A2. Multifunctional heteromultimers are obtained by conjugating antigen binding motifs, cytokines and other forms of signaling molecules, chemotoxin, radiotoxins or other functionally relevant immunoconjugates to N and/or C terminal sites on A1 and A2 and this is represented by the + symbol.
  • FIG. 5 is a schematic of a bispecific antibody derived from a heterodimeric Fc domain. Albumin or alloalbumin based polypeptides are connected to the C-terminal of the Fc to selectively drive the formation of heterodimers.
  • FIGS. 6A-6C show native gel electrophoresis profiles of full-length HSA and heterodimer scaffolds Albumin-based heteromultimer-1 (ABH1) and Albumin-based heteromultimer-2 (ABH2) formed by coexpression of HSA based transporter polypeptides.
  • FIG. 7 shows stability of wild type HSA and heterodimer scaffolds ABH1 and ABH2 stuided using Differential Scanning Calorimetry
  • FIGS. 8A-8B show equilibrium binding isotherms 3000 nM FcRN 3× dilution series over 3000 RUs. FIG. 8A shows Albumin and FIG. 8B shows heteromultimer scaffold ABH1
  • FIG. 9 shows scheme for multivalent Albumin based heteromultimers comprising anti-Her2/neu and anti-CD16 scFv bioactive fusions
  • FIGS. 10A-10B contain a non-reducing SDS PAGE analysis of the heteromultimer ABH2 fusions described in table 8. The gel indicates all constructs form the correct complex with expected MW.
  • FIG. 11 shows structure of Annexin molecule based on the PDB structure 1MCX. The two monomers that will be derived by splitting the Annexin molecule are color coded as light and dark grey units. The sites of fusion for the cargo protein are represented as spheres.
  • FIG. 12 shows a plot of the buried solvent accessible surface area at the interface of Annexin based tranporter polypeptide-1, and Annexin based tranporter polypeptide-2.
  • FIG. 13 shows structure of transferrin molecule based on the PDB structure 1H76. The two monomers derived by splitting the transferrin molecule are color coded as light and dark grey units. The sites of fusion for the cargo protein are represented as spheres.
  • FIG. 14 shows a plot of the buried solvent accessible surface area at the interface of two transferrin based transporter polypeptides described herein. A split transferrin near residue position 330 as designed herein, forms a heterodimer with about 1800 Å2 of buried surface area.
  • FIGS. 15A-15SS shows sequences of multimers comprising transporter polypeptides based on human serum albumin.
  • DETAILED DESCRIPTION
  • In the realm of therapeutic proteins, bispecific molecules exhibit dual target specificities or are able to simultaneously perform multiple functional roles by providing the necessary spatiotemporal organization necessary for drug action. In one aspect, bispecific molecules are particularly interesting when the mode of therapeutic action involves retargeting of effector cells or molecules to a target such as a tumor cell [Muller D. and Kontermann R. E. (2010) Biodrugs 24, 89-98]. The development of bispecific therapeutic proteins with favorable pharmacokinetics and functional activity in stable and homogeneous condition has been a challenge. Attempts have been made to assemble bispecific units from multiple antigen binding domains using a number of approaches. These techniques have involved using heterodimeric antibody IgG molecule, using leucine zipper proteins such as the Fos/Jun pair or other scaffolds assembled from the alternate organizations of the light and heavy chains of the variable domains in an antibody. Kipriyanov and Le Gall have reviewed the design of a variety of bispecific constructs [Kipriyanov S. M. & Le Gall F. (2004) Curr Opin Drug Discov Dev 7, 233-242]. The use of a heterodimeric antibody IgG molecule wherein mutations are introduced in the CH3 domain of the antibody to achieve the heterodimer and hence introduce the two unique antigen binding sites into one molecule is very attractive because of the natural immunoglobulin like structure of this construct. Further, the Fc portion of the antibody is involved in interactions with the neonatal Fc receptor (FcRn) which mediates an endocytic salvage pathway and this is attributed to improved serum half-life of the antibody molecule [Roopenian D. & Akilesh S. (2007) Nature Rev Immunol 7, 715-725]. On the other hand, antibody based bispecific molecules have been problematic in clinical trials because of the strong cytokine responses as a result of the concurrent effector activity induced via the Fc portion of the bispecific antibody [Weiner L. M.; Alpaugh R. K. et al. (1996) Cancer Immunol Immunother 42, 141-150]. This highlights the needs for novel scaffolds that can aid in the design of bispecific and immunoconjugate molecules.
  • The human serum album (HSA) protein is the most abundant component of blood, accounting for close to 60% of the total protein in blood serum at a concentration of about 40 mg/ml. Albumin is also one of the longest-lived proteins in the circulatory system with a half-life of about 19 days. Interestingly, the same endocytic salvage pathway dependent on FcRn molecules that prevents antibody degradation is known to interact with the HSA molecule as well [Chaudhary C.; Mehnaz S. et al. (2003) J Exp Med 197, 315-322].
  • HSA (shown in FIG. 1) is a non-glycosylated 585-residue single polypeptide protein and the 3-dimensional structure of the protein was first observed using X-ray crystallography by Carter and coworkers [reviewed in Carter, D. C. & Ho, J. X. (1994) Adv Prot Chem 45, 153-203]. The HSA protein consists of three homologous domains: DI, DII, DIII, attributed to gene duplication, a feature common to the serum albumin in other species as well [Gray J. E. & Doolittle R. F. (1992) Protein Sci 1, 289-302]. Each of the three domains have been expressed and characterized separately and shown to be independently stable [Dockal M., Carter D. C. & Ruker F. (1999) J Biol Chem 274, 29303-29310]. Each domain is made up of 10 helical segments and based on the inter-helical organization each domain can be further classified into 2 sub-domains comprised of helix 1-6 and 7-10 respectively. HSA has 17 disulphide bonds in total and all these cysteine pairs forming the linkages are within the individual domains. In general, HSA is a very stable due to the large number of disulphide bonds as well as the predominantly helical fold. The sequence identities of albumin molecules across a number of species is quite large, greater than 70% among albumin cDNA derived from humans, horse, bovine, rat, etc. [Carter, D. C. & Ho, J. X. (1994) Adv Prot Chem 45, 153-203].
  • Split protein pairs have been used as sensors to understand protein-protein interactions in the area of functional proteomics. The approach involves identifying suitable segments from a protein that can reconstitute to form an active native-like protein. Generating new split proteins is technically demanding. For a protein to be split in a functionally useful manner, the segmentation site has to yield two segments that efficiently reconstitute into the quasi-native protein when associated to each other. Further, the component protein segments should be soluble enough to stay in solution and selectively associate with the partner segments such that manufacture yields and purification will be economical. Deriving split protein segments that would recombine to form the quasi-native structure is quite challenging [Tafelmeyer P., Johnsson N. & Johnsson K. Chem & Biol 11, 681-689]. Such split proteins have not been used in the design of protein therapeutics, or as cargo delivery vehicles in the past.
  • Definitions
  • It is to be understood that this invention is not limited to the particular protocols; cell lines, constructs, and reagents described herein and as such may vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only, and is not intended to limit the scope of the present invention, which will be limited only by the appended claims.
  • As used herein and in the appended claims, the singular forms “a,” “an,” and “the” include plural reference unless the context clearly indicates otherwise. Thus, for example, reference to a “HSA”, “HA”, “albumin”, “human serum albumin” and various capitalized, hyphenated and unhyphenated forms is a reference to one or more such proteins and includes variants, derivatives, fragments, equivalents thereof known to those of ordinary skill in the art, and so forth.
  • Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood to one of ordinary skill in the art to which this invention belongs. Although any methods, devices, and materials similar or equivalent to those described herein can be used in the practice or testing of the invention, the preferred methods, devices and materials are now described.
  • All publications and patents mentioned herein are incorporated herein by reference for the purpose of describing and disclosing, for example, the constructs and methodologies that are described in the publications, which might be used in connection with the presently described invention. The publications discussed herein are provided solely for their disclosure prior to the filing date of the present application. Nothing herein is to be construed as an admission that the inventors are not entitled to antedate such disclosure by virtue of prior invention or for any other reason.
  • A “heteromultimer” or “heteromultimeric polypeptide” is a molecule comprising at least a first monomer comprising a first transporter polypeptide and a second monomer comprising a second transporter polypeptide, wherein the second polypeptide differs in amino acid sequence from the first polypeptide by at least one amino acid residue. The heteromultimer can comprise a “heterodimer” formed by the first and second transporter polypeptides. In certain embodiments, the heteromultimer can form higher order tertiary structures such as, but not restricted to trimers and tetramers. In some embodiments, transporter polypeptides in addition to the first and second transporter polypeptides are present. In certain embodiments, the assembly of transporter polypeptides to form the heteromultimer is driven by surface area burial. In some embodiments, the transporter polypeptides interact with each other by means of electrostatic interactions and/or salt-bridge interactions that drive heteromultimer formation by favoring heteromultimer formation and/or disfavoring homomultimer formation. In some embodiments, the transporter polypeptides inteact with each other by means of hydrophobic interactions that drive heteromultimer formation by favoring heteromultimer formation and/or disfavoring homomultimer formation. In certain embodiments, the transporter polypeptides inteact with each other by means of covalent bond formation. In certain embodiments, the covalent bonds are formed between naturally present or introduced cysteines that drive heteromultimer formation. In certain embodiments of the heteromultimers described herein, no covalent bonds are formed between the monomers. In some embodiments, the transporter polypeptides inteact with each other by means of packing/size-complementarity/knobs-into-holes/protruberance-cavity type interactions that drive heteromultimer formation by favoring heteromultimer formation and/or disfavoring homomultimer formation. In some embodiments, the transporter polypeptides inteact with each other by means of cation-pi interactions that drive heteromultimer formation by favoring heteromultimer formation and/or disfavoring homomultimer formation. In certain embodiments the individual transporter polypeptides cannot exist as isolated monomers in solution. In certain embodiments, the heteromultimer is the preferred state of the individual transporter polypeptides as compared to the monomer.
  • The term “bispecific” is intended to include any agent, e.g., heteromultimer, monomer, protein, peptide, or protein or peptide complex, which has two different binding specificities. For example, in some embodiments, the molecule may bind to, or interact with, (a) a cell surface target molecule and (b) an Fc receptor on the surface of an effector cell. In certain embodiments of a heteromultimer described herein, at least one monomer is bispecific formed by attaching to the same transporter polypeptide, two cargo molecules with different binding specificities. In certain embodiments of a heteromultimer described herein, the heteromultimer is itself bispecific formed by attaching to the transporter polypeptides, at least two cargo molecules with different specificities. The term “multispecific molecule” or “heterospecific molecule” is intended to include any agent, e.g., a protein, peptide, or protein or peptide complex, which has more than two different binding specificities. For example, the molecule may bind to, or interact with, (a) a cell surface target molecule such as but not limited to cell surface antigens, (b) an Fc receptor on the surface of an effector cell, and (c) at least one other component. Accordingly, embodiments of the heteromultimers described herein, are inclusive of, but not limited to, bispecific, trispecific, tetraspecific, and other multispecific molecules. In certain embodiments, these molecules are directed to cell surface antigens, such as CD30, and to other targets, such as Fc receptors on effector cells.
  • Unless indicated otherwise, the expression “multivalent” is used throughout this specification to denote a heteromultimer comprising at least two sites of attachment for target molecules. The multivalent heteromultimer is designed to have multiple binding sites for desired targets. In certain embodiments, the binding sites are on at least one cargo molecules attached to a transporter polypeptide. In certain embodiments, at least one binding site is on a transporter polypeptide. The expression “bivalent” is used throughout this specification to denote a heteromultimer comprising two target binding sites. In certain embodiments of a bivalent heteromultimer, both binding sites are on the same monomer. The expression “trivalent” is used throughout this specification to denote a heteromultimer comprising three target binding sites. The expression “tetravalent” is used throughout this specification to denote a heteromultimer comprising four target binding sites.
  • “Fusion proteins” and polypeptides are created by joining two or more genes that originally code for separate polypeptides. Translation of this fusion gene results in a single polypeptide with functional properties derived from each of the original polypeptides. In embodiments of the heteromultimers described herein, at least one monomer may comprise a fusion protein formed by the fusion of at least one cargo polypeptide to the N- or C-terminus of a transporter polypeptide.
  • The term “substantially purified” refers to a heteromultimer described herein, or variant thereof that may be substantially or essentially free of components that normally accompany or interact with the protein as found in its naturally occurring environment, i.e. a native cell, or host cell in the case of recombinantly produced heteromultimer that in certain embodiments, is substantially free of cellular material includes preparations of protein having less than about 30%, less than about 25%, less than about 20%, less than about 15%, less than about 10%, less than about 5%, less than about 4%, less than about 3%, less than about 2%, or less than about 1% (by dry weight) of contaminating protein. When the heteromultimer or variant thereof is recombinantly produced by the host cells, the protein in certain embodiments is present at about 30%, about 25%, about 20%, about 15%, about 10%, about 5%, about 4%, about 3%, about 2%, or about 1% or less of the dry weight of the cells. When the heteromultimer or variant thereof is recombinantly produced by the host cells, the protein, in certain embodiments, is present in the culture medium at about 5 g/L, about 4 g/L, about 3 g/L, about 2 g/L, about 1 g/L, about 750 mg/L, about 500 mg/L, about 250 mg/L, about 100 mg/L, about 50 mg/L, about 10 mg/L, or about 1 mg/L or less of the dry weight of the cells. In certain embodiments, “substantially purified” heteromultimer produced by the methods described herein, has a purity level of at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, specifically, a purity level of at least about 75%, 80%, 85%, and more specifically, a purity level of at least about 90%, a purity level of at least about 95%, a purity level of at least about 99% or greater as determined by appropriate methods such as SDS/PAGE analysis, RP-HPLC, SEC, and capillary electrophoresis.
  • A “recombinant host cell” or “host cell” refers to a cell that includes an exogenous polynucleotide, regardless of the method used for insertion, for example, direct uptake, transduction, f-mating, or other methods known in the art to create recombinant host cells. The exogenous polynucleotide may be maintained as a nonintegrated vector, for example, a plasmid, or alternatively, may be integrated into the host genome.
  • As used herein, the term “medium” or “media” includes any culture medium, solution, solid, semi-solid, or rigid support that may support or contain any host cell, including bacterial host cells, yeast host cells, insect host cells, plant host cells, eukaryotic host cells, mammalian host cells, CHO cells, prokaryotic host cells, E. coli, or Pseudomonas host cells, and cell contents. Thus, the term may encompass medium in which the host cell has been grown, e.g., medium into which the protein has been secreted, including medium either before or after a proliferation step. The term also may encompass buffers or reagents that contain host cell lysates, such as in the case where a heteromultimer described herein is produced intracellularly and the host cells are lysed or disrupted to release the heteromultimer.
  • “Refolding,” as used herein describes any process, reaction or method which transforms disulfide bond containing polypeptides from an improperly folded or unfolded state to a native or properly folded conformation with respect to disulfide bonds.
  • “Cofolding,” as used herein, refers specifically to refolding processes, reactions, or methods which employ at least two monomeric polypeptides which interact with each other and result in the transformation of unfolded or improperly folded polypeptides to native, properly folded polypeptides.
  • As used herein, the term “modulated serum half-life” means the positive or negative change in circulating half-life of a cargo polypeptide that is comprised by a heteromultimer described herein relative to its native form. Serum half-life is measured by taking blood samples at various time points after administration of heteromultimer, and determining the concentration of that molecule in each sample. Correlation of the serum concentration with time allows calculation of the serum half-life. Increased serum half-life desirably has at least about two-fold, but a smaller increase may be useful, for example where it enables a satisfactory dosing regimen or avoids a toxic effect. In some embodiments, the increase is at least about three-fold, at least about five-fold, or at least about ten-fold.
  • The term “modulated therapeutic half-life” as used herein means the positive or negative change in the half-life of the therapeutically effective amount of a cargo polypeptide comprised by a heteromultimer described herein, relative to its non-modified form. Therapeutic half-life is measured by measuring pharmacokinetic and/or pharmacodynamic properties of the molecule at various time points after administration. Increased therapeutic half-life desirably enables a particular beneficial dosing regimen, a particular beneficial total dose, or avoids an undesired effect. In some embodiments, the increased therapeutic half-life results from increased potency, increased or decreased binding of the modified molecule to its target, increased or decreased breakdown of the molecule by enzymes such as proteases, or an increase or decrease in another parameter or mechanism of action of the non-modified molecule or an increase or decrease in receptor-mediated clearance of the molecule.
  • The term “isolated,” when applied to a nucleic acid or protein, denotes that the nucleic acid or protein is free of at least some of the cellular components with which it is associated in the natural state, or that the nucleic acid or protein has been concentrated to a level greater than the concentration of its in vivo or in vitro production. It can be in a homogeneous state. Isolated substances can be in either a dry or semi-dry state, or in solution, including but not limited to, an aqueous solution. It can be a component of a pharmaceutical composition that comprises additional pharmaceutically acceptable carriers and/or excipients. Purity and homogeneity are typically determined using analytical chemistry techniques such as polyacrylamide gel electrophoresis or high performance liquid chromatography. A protein which is the predominant species present in a preparation is substantially purified. In particular, an isolated gene is separated from open reading frames which flank the gene and encode a protein other than the gene of interest. The term “purified” denotes that a nucleic acid or protein gives rise to substantially one band in an electrophoretic gel. Particularly, it may mean that the nucleic acid or protein is at least 85% pure, at least 90% pure, at least 95% pure, at least 99% or greater pure.
  • The term “nucleic acid” refers to deoxyribonucleotides, deoxyribonucleosides, ribonucleosides, or ribonucleotides and polymers thereof in either single- or double-stranded form. Unless specifically limited, the term encompasses nucleic acids containing known analogues of natural nucleotides which have similar binding properties as the reference nucleic acid and are metabolized in a manner similar to naturally occurring nucleotides. Unless specifically limited otherwise, the term also refers to oligonucleotide analogs including PNA (peptidonucleic acid), analogs of DNA used in antisense technology (phosphorothioates, phosphoroamidates, and the like). Unless otherwise indicated, a particular nucleic acid sequence also implicitly encompasses conservatively modified variants thereof (including but not limited to, degenerate codon substitutions) and complementary sequences as well as the sequence explicitly indicated. Specifically, degenerate codon substitutions may be achieved by generating sequences in which the third position of one or more selected (or all) codons is substituted with mixed-base and/or deoxyinosine residues (Batzer et al., Nucleic Acid Res. 19:5081 (1991); Ohtsuka et al., J. Biol. Chem. 260:2605-2608 (1985); Rossolini et al., Mol. Cell. Probes 8:91-98 (1994)).
  • The terms “polypeptide,” “peptide” and “protein” are used interchangeably herein to refer to a polymer of amino acid residues. That is, a description directed to a polypeptide applies equally to a description of a peptide and a description of a protein, and vice versa. The terms apply to naturally occurring amino acid polymers as well as amino acid polymers in which one or more amino acid residues is a non-naturally encoded amino acid. As used herein, the terms encompass amino acid chains of any length, including full length proteins, wherein the amino acid residues are linked by covalent peptide bonds.
  • The term “amino acid” refers to naturally occurring and non-naturally occurring amino acids, as well as amino acid analogs and amino acid mimetics that function in a manner similar to the naturally occurring amino acids. Naturally encoded amino acids are the 20 common amino acids (alanine, arginine, asparagine, aspartic acid, cysteine, glutamine, glutamic acid, glycine, histidine, isoleucine, leucine, lysine, methionine, phenylalanine, praline, serine, threonine, tryptophan, tyrosine, and valine) and pyrrolysine and selenocysteine. Amino acid analogs refers to compounds that have the same basic chemical structure as a naturally occurring amino acid, i.e., an a carbon that is bound to a hydrogen, a carboxyl group, an amino group, and an R group, such as, homoserine, norleucine, methionine sulfoxide, methionine methyl sulfonium. Such analogs have modified R groups (such as, norleucine) or modified peptide backbones, but retain the same basic chemical structure as a naturally occurring amino acid. Reference to an amino acid includes, for example, naturally occurring proteogenic L-amino acids; D-amino acids, chemically modified amino acids such as amino acid variants and derivatives; naturally occurring non-proteogenic amino acids such as β-alanine, ornithine, etc.; and chemically synthesized compounds having properties known in the art to be characteristic of amino acids. Examples of non-naturally occurring amino acids include, but are not limited to, α-methyl amino acids (e.g. α-methyl alanine), D-amino acids, histidine-like amino acids (e.g., 2-amino-histidine, β-hydroxy-histidine, homohistidine), amino acids having an extra methylene in the side chain (“homo” amino acids), and amino acids in which a carboxylic acid functional group in the side chain is replaced with a sulfonic acid group (e.g., cysteic acid). The incorporation of non-natural amino acids, including synthetic non-native amino acids, substituted amino acids, or one or more D-amino acids into the proteins of the present invention may be advantageous in a number of different ways. D-amino acid-containing peptides, etc., exhibit increased stability in vitro or in vivo compared to L-amino acid-containing counterparts. Thus, the construction of peptides, etc., incorporating D-amino acids can be particularly useful when greater intracellular stability is desired or required. More specifically, D-peptides, etc., are resistant to endogenous peptidases and proteases, thereby providing improved bioavailability of the molecule, and prolonged lifetimes in vivo when such properties are desirable. Additionally, D-peptides, etc., cannot be processed efficiently for major histocompatibility complex class II-restricted presentation to T helper cells, and are therefore, less likely to induce humoral immune responses in the whole organism.
  • Amino acids may be referred to herein by either their commonly known three letter symbols or by the one-letter symbols recommended by the IUPAC-IUB Biochemical Nomenclature Commission. Nucleotides, likewise, may be referred to by their commonly accepted single-letter codes.
  • “Conservatively modified variants” applies to both amino acid and nucleic acid sequences. With respect to particular nucleic acid sequences, “conservatively modified variants” refers to those nucleic acids which encode identical or essentially identical amino acid sequences, or where the nucleic acid does not encode an amino acid sequence, to essentially identical sequences. Because of the degeneracy of the genetic code, a large number of functionally identical nucleic acids encode any given protein. For instance, the codons GCA, GCC, GCG and GCU all encode the amino acid alanine. Thus, at every position where an alanine is specified by a codon, the codon can be altered to any of the corresponding codons described without altering the encoded polypeptide. Such nucleic acid variations are “silent variations,” which are one species of conservatively modified variations. Every nucleic acid sequence herein which encodes a polypeptide also describes every possible silent variation of the nucleic acid. One of ordinary skill in the art will recognize that each codon in a nucleic acid (except AUG, which is ordinarily the only codon for methionine, and TGG, which is ordinarily the only codon for tryptophan) can be modified to yield a functionally identical molecule. Accordingly, each silent variation of a nucleic acid which encodes a polypeptide is implicit in each described sequence.
  • As to amino acid sequences, one of ordinary skill in the art will recognize that individual substitutions, deletions or additions to a nucleic acid, peptide, polypeptide, or protein sequence which alters, adds or deletes a single amino acid or a small percentage of amino acids in the encoded sequence is a “conservatively modified variant” where the alteration results in the deletion of an amino acid, addition of an amino acid, or substitution of an amino acid with a chemically similar amino acid. Conservative substitution tables providing functionally similar amino acids are known to those of ordinary skill in the art. Such conservatively modified variants are in addition to and do not exclude polymorphic variants, interspecies homologs, and alleles of the invention.
  • Conservative substitution tables providing functionally similar amino acids are known to those of ordinary skill in the art. The following eight groups each contain amino acids that are conservative substitutions for one another:
  • 1) Alanine (A), Glycine (G);
  • 2) Aspartic acid (D), Glutamic acid (E);
  • 3) Asparagine (N), Glutamine (Q);
  • 4) Arginine (R), Lysine (K);
  • 5) Isoleucine (I), Leucine (L), Methionine (M), Valine (V);
  • 6) Phenylalanine (F), Tyrosine (Y), Tryptophan (W);
  • 7) Serine (S), Treonine (T); and [0139] 8) Cysteine (C), Methionine (M) (see, e.g., Creighton, Proteins: Structures and Molecular Properties (W H Freeman & Co.; 2nd edition (December 1993)
  • The terms “identical” or percent “identity,” in the context of two or more nucleic acids or polypeptide sequences, refer to two or more sequences or subsequences that are the same. Sequences are “substantially identical” if they have a percentage of amino acid residues or nucleotides that are the same (i.e., about 60% identity, about 65%, about 70%, about 75%, about 80%, about 85%, about 90%, or about 95% identity over a specified region), when compared and aligned for maximum correspondence over a comparison window, or designated region as measured using one of the following sequence comparison algorithms (or other algorithms available to persons of ordinary skill in the art) or by manual alignment and visual inspection. This definition also refers to the complement of a test sequence. The identity can exist over a region that is at least about 50 amino acids or nucleotides in length, or over a region that is 75-100 amino acids or nucleotides in length, or, where not specified, across the entire sequence of a polynucleotide or polypeptide. A polynucleotide encoding a polypeptide of the present invention, including homologs from species other than human, may be obtained by a process comprising the steps of screening a library under stringent hybridization conditions with a labeled probe having a polynucleotide sequence of the invention or a fragment thereof, and isolating full-length cDNA and genomic clones containing said polynucleotide sequence. Such hybridization techniques are well known to the skilled artisan.
  • For sequence comparison, typically one sequence acts as a reference sequence, to which test sequences are compared. When using a sequence comparison algorithm, test and reference sequences are entered into a computer, subsequence coordinates are designated, if necessary, and sequence algorithm program parameters are designated. Default program parameters can be used, or alternative parameters can be designated. The sequence comparison algorithm then calculates the percent sequence identities for the test sequences relative to the reference sequence, based on the program parameters.
  • A “comparison window”, as used herein, includes reference to a segment of any one of the number of contiguous positions selected from the group consisting of from 20 to 600, usually about 50 to about 200, more usually about 100 to about 150 in which a sequence may be compared to a reference sequence of the same number of contiguous positions after the two sequences are optimally aligned. Methods of alignment of sequences for comparison are known to those of ordinary skill in the art. Optimal alignment of sequences for comparison can be conducted, including but not limited to, by the local homology algorithm of Smith and Waterman (1970) Adv. Appl. Math. 2:482c, by the homology alignment algorithm of Needleman and Wunsch (1970) J. Mol. Biol. 48:443, by the search for similarity method of Pearson and Lipman (1988) Proc. Nat'l. Acad. Sci. USA 85:2444, by computerized implementations of these algorithms (GAP, BESTFIT, FASTA, and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group, 575 Science Dr., Madison, Wis.), or by manual alignment and visual inspection (see, e.g., Ausubel et al., Current Protocols in Molecular Biology (1995 supplement)).
  • One example of an algorithm that is suitable for determining percent sequence identity and sequence similarity are the BLAST and BLAST 2.0 algorithms, which are described in Altschul et al. (1997) Nuc. Acids Res. 25:3389-3402, and Altschul et al. (1990) J. Mol. Biol. 215:403-410, respectively. Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information available at the World Wide Web at ncbi.nlm.nih.gov. The BLAST algorithm parameters W, T, and X determine the sensitivity and speed of the alignment. The BLASTN program (for nucleotide sequences) uses as defaults a wordlength (W) of 11, an expectation (E) or 10, M=5, N=−4 and a comparison of both strands. For amino acid sequences, the BLASTP program uses as defaults a wordlength of 3, and expectation (E) of 10, and the BLOSUM62 scoring matrix (see Henikoff and Henikoff (1992) Proc. Natl. Acad. Sci. USA 89:10915) alignments (B) of 50, expectation (E) of 10, M=5, N=−4, and a comparison of both strands. The BLAST algorithm is typically performed with the “low complexity” filter turned off.
  • The BLAST algorithm also performs a statistical analysis of the similarity between two sequences (see, e.g., Karlin and Altschul (1993) Proc. Natl. Acad. Sci. USA 90:5873-5787). One measure of similarity provided by the BLAST algorithm is the smallest sum probability (P(N)), which provides an indication of the probability by which a match between two nucleotide or amino acid sequences would occur by chance. For example, a nucleic acid is considered similar to a reference sequence if the smallest sum probability in a comparison of the test nucleic acid to the reference nucleic acid is less than about 0.2, or less than about 0.01, or less than about 0.001.
  • The phrase “selectively (or specifically) hybridizes to” refers to the binding, duplexing, or hybridizing of a molecule only to a particular nucleotide sequence under stringent hybridization conditions when that sequence is present in a complex mixture (including but not limited to, total cellular or library DNA or RNA).
  • The phrase “stringent hybridization conditions” refers to hybridization of sequences of DNA, RNA, or other nucleic acids, or combinations thereof under conditions of low ionic strength and high temperature as is known in the art. Typically, under stringent conditions a probe will hybridize to its target subsequence in a complex mixture of nucleic acid (including but not limited to, total cellular or library DNA or RNA) but does not hybridize to other sequences in the complex mixture. Stringent conditions are sequence-dependent and will be different in different circumstances. Longer sequences hybridize specifically at higher temperatures. An extensive guide to the hybridization of nucleic acids is found in Tijssen, Laboratory Techniques in Biochemistry and Molecular Biology—Hybridization with Nucleic Probes, “Overview of principles of hybridization and the strategy of nucleic acid assays” (1993).
  • As used herein, the term “eukaryote” refers to organisms belonging to the phylogenetic domain Eucarya such as animals (including but not limited to, mammals, insects, reptiles, birds, etc.), ciliates, plants (including but not limited to, monocots, dicots, algae, etc.), fungi, yeasts, flagellates, microsporidia, protists, etc.
  • As used herein, the term “prokaryote” refers to prokaryotic organisms. For example, a non-eukaryotic organism can belong to the Eubacteria (including but not limited to, Escherichia coli, Thermus thermophilus, Bacillus stearothermophilus, Pseudomonas fluorescens, Pseudomonas aeruginosa, Pseudomonas putida, etc.) phylogenetic domain, or the Archaea (including but not limited to, Methanococcus jannaschii, Methanobacterium thermoautotrophicum, Halobacterium such as Haloferax volcanii and Halobacterium species NRC-1, Archaeoglobus fulgidus, Pyrococcus furiosus, Pyrococcus horikoshii, Aeuropyrum pernix, etc.) phylogenetic domain.
  • The term “subject” as used herein, refers to an animal, in some embodiments a mammal, and in other embodiments a human, who is the object of treatment, observation or experiment. An animal may be a companion animal (e.g., dogs, cats, and the like), farm animal (e.g., cows, sheep, pigs, horses, and the like) or a laboratory animal (e.g., rats, mice, guinea pigs, and the like).
  • The term “effective amount” as used herein refers to that amount of heteromultimer being administered, which will relieve to some extent one or more of the symptoms of the disease, condition or disorder being treated. Compositions containing the heteromultimer described herein can be administered for prophylactic, enhancing, and/or therapeutic treatments.
  • The terms “enhance” or “enhancing” means to increase or prolong either in potency or duration a desired effect. Thus, in regard to enhancing the effect of therapeutic agents, the term “enhancing” refers to the ability to increase or prolong, either in potency or duration, the effect of other therapeutic agents on a system. An “enhancing-effective amount,” as used herein, refers to an amount adequate to enhance the effect of another therapeutic agent in a desired system. When used in a patient, amounts effective for this use will depend on the severity and course of the disease, disorder or condition, previous therapy, the patient's health status and response to the drugs, and the judgment of the treating physician.
  • The term “modified,” as used herein refers to any changes made to a given polypeptide, such as changes to the length of the polypeptide, the amino acid sequence, chemical structure, co-translational modification, or post-translational modification of a polypeptide. The form “(modified)” term means that the polypeptides being discussed are optionally modified, that is, the polypeptides under discussion can be modified or unmodified.
  • The term “post-translationally modified” refers to any modification of a natural or non-natural amino acid that occurs to such an amino acid after it has been incorporated into a polypeptide chain. The term encompasses, by way of example only, co-translational in vivo modifications, co-translational in vitro modifications (such as in a cell-free translation system), post-translational in vivo modifications, and post-translational in vitro modifications.
  • The term “segmentation” refers to a precise internal splice of the original protein sequence which results in “segments” of the protein sequence that preferentially associate as heteromultimers to form a quasi-protein.
  • Quasi-Native Structure:
  • With reference to a native protein or its structure, quasi-native proteins and/or ‘quasi-native structures’ present the native protein like functional and structural characteristics. Proteins are naturally dynamics molecules and display an ensemble of structural configurations although we ascribe a native structure to it, such as the one obtained by X-ray crystallography. The alternate structural configurations observed in the ensemble of geometries of that protein can be deemed to be quasi-native structures relative to each other or relative to the structure observed in the crystal. On a different front, homologous proteins sequences or proteins belonging to common structural families tend to fold into similar structural geometries. The member proteins belonging to this family can be deemed to achieve a quasi-native structure relative to each other. Some of the unique sequences in the protein family could also exhibit similar functional attributes and hence can be referred to as quasi-native proteins relative to each other. In the case of heteromultimers described here comprising of two or more monomeric proteins each of which have a transporter polypeptide component, the transporter polypeptides assemble to form a quasi-native structure. The reference native protein in this case is the protein from which the transporter polypeptide is derived and the reference native structure is the structure of the protein from which the transporter polypeptide is derived. We describe a case where two or more different polypeptides self-assemble to form a heteromultimeric structural and exhibit functional characteristics like a native protein which itself is a monomeric entity. In certain embodiments, we present polypeptide segments derived from albumin that self-assemble to form a heteromultimer that exhibits native albumin like functional characteristics such as FcRn binding and structural characteristics. In certain embodiments, we present polypeptide segments derived from transferrin that self-assemble to form a heteromultimer that exhibits native transferrin like structural and functional characteristics. In certain embodiments, we present polypeptide segments derived from annexin that self-assemble to form a heteromultimer that exhibits native annexin like structural and functional characteristics. These heteromultimers are referred to as being quasi-native.
  • Transporter Polypeptide
  • As used herein, the term “transporter polypeptide” or “transporter polypeptide” or “transporter peptide” or “transporter” refers to a polypeptide, such that said transporter polypeptide is capable of forming heteromultimeric proteins with other such transporter polypeptides in solution, and wherein said heteromultimeric proteins have a quasi-native structure of a monomeric protein from which at least one transporter polypeptide is derived. In certain embodiments of the heteromultimers described herein, all transporter polypeptides are derived from the same albumin or alloalbumin protein. In certain other embodiments, the heteromultimers are formed by transporter polypeptides derived from various albumin and alloalbumin proteins. In certain embodiments of the heteromultimers described herein, the transporter polypeptides are derived from transferrin. In certain embodiments of the heteromultimers described herein, all transporter polypeptides are derived from annexin proteins. In certain embodiments, the heteromultimers are formed by transporter polypeptides derived from the same annexin protein. In some embodiments, the heteromultimers are formed by transporter polypeptides derived from different annexin proteins. In an embodiment, the heteromultimers are formed by transporter polypeptides derived from annexin A2.
  • In certain embodiments, transporter polypeptides are segments of a whole protein, wherein said segments are capable of assembling to form a heteromultimer. In certain embodiments, the transporter polypeptides are segments derived from a coiled coil protein. In certain embodiments, the transporter polypeptides are segments derived from a leucine-zipper protein. In an embodiment, the transporter polypeptides are segments from a beta-barrel protein. In an embodiment, transporter polypeptides are segments obtained from a beta-propeller protein. In some embodiments, the transporter polypeptides are segments obtained from a helical bundle protein. In embodiments, the transporter polypeptides are generated from for instance, but not restricted to proteins comprising a zinc finger motif, a helix-turn-helix motif or a beta-hairpin motif. In some embodiments, the transporter polypeptides are segments obtained from non-immunogenic proteins that are structurally stable, and have favorable biological properties.
  • Albumin
  • As used herein, “albumin” refers collectively to albumin protein or amino acid sequence, or an albumin segment or variant, having one or more functional activities (e.g., biological activities) of albumin. In particular “albumin” refers to human albumin or segments thereof (see for example, EP 201239, EP 322 094 WO 97/24445, WO95/23857) especially the mature form of human albumin as shown in FIG. 1, or albumin from other vertebrates, or segments thereof, or analogs or variants of these molecules or fragments thereof. In certain embodiments, albumin refers to a truncated version of albumin.
  • The term “quasi-albumin” refers to a heteromultimer molecule that has structure and/or function similar to the whole albumin, and wherein said heteromultimer molecule is formed by the assembly of two or more monomeric polypeptides designed based on the sequence of the whole albumin. In certain embodiments, the monomeric polypeptides are “segments” that preferentially associate as heteromultimeric pairs to form a quasi-protein. In some embodiments, the quasi-albumin has 90% of the activity of the whole albumin. In some embodiments, the quasi-albumin has 75% of the activity of whole-albumin. In an embodiment, the quasi-albumin has 50% of the activity of whole albumin. In some embodiments, the quasi-albumin has 50-75% of the activity of whole albumin. In an embodiment, quasi-albumin has 80% of the activity of whole albumin. In some embodiments, the quasi-albumin has 90% of the structure of whole albumin as determined by molecular modeling. In some embodiments, the quasi-albumin has 80% of the structure of whole albumin as determined by molecular modeling. In some embodiments, the quasi-albumin has 70% of the structure of whole albumin as determined by molecular modeling. In some embodiments, the quasi-albumin has 50% of the structure of whole albumin as determined by molecular modeling. In some embodiments, the quasi-albumin has 50%-75% of the structure of whole albumin as determined by molecular modeling.
  • The terms, human serum albumin (HSA) and human albumin (HA) are used interchangeably herein. The terms, “albumin and serum albumin” are broader, and encompass human serum albumin (and fragments and variants thereof) as well as albumin from other species (and fragments and variants thereof).
  • In certain embodiments, each albumin-based monomer of the heteromultimeric proteins described herein is based on a variant of normal HA. Each cargo polypeptide portion of the heteromultimeric proteins of the invention may also be variants of the Therapeutic proteins as described herein. The term “variants” includes insertions, deletions and substitutions, either conservative or non conservative, where such changes do not substantially alter one or more of the oncotic, useful ligand-binding and non-immunogenic properties of albumin, or the active site, or active domain which confers the therapeutic activities of the Therapeutic proteins.
  • In certain embodiments, the heteromultimeric proteins described herein include naturally occurring polymorphic variants of human albumin and fragments of human albumin, for example those fragments disclosed in EP 322 094 (namely HA (Pn), where n is 369 to 419).
  • In certain embodiments, the albumin is derived from any vertebrate, especially any mammal that includes but is not limited to human, cow, sheep, rat, mouse, rabbit, horse, dog or pig. In certain embodiments, the albumin is derived from non-mammalian albumins including, but are not limited to hen and salmon.
  • The sequence of human albumin is as shown:
  • SEQ ID NO: 1
    MKWVTFISLLFLFSSAYSRGVFRRDAHKSEVAHRFKDLGEENFKALVLIA
    FAQYLQQCPFEDHVKLVNEVTEFAKTCVADESAENCDKSLHTLFGDKLCT
    VATLRETYGEMADCCAKQEPERNECFLQHKDDNPNLPRLVRPEVDVMCTA
    FHDNEETFLKKYLYEIARRHPYFYAPELLFFAKRYKAAFTECCQAADKAA
    CLLPKLDELRDEGKASSAKQRLKCASLQKFGERAFKAWAVARLSQRFPKA
    EFAEVSKLVTDLTKVHTECCHGDLLECADDRADLAKYICENQDSISSKLK
    ECCEKPLLEKSHCIAEVENDEMPADLPSLAADFVESKDVCKNYAEAKDVF
    LGMFLYEYARRHPDYSVVLLLRLAKTYETTLEKCCAAADPHECYAKVFDE
    FKPLVEEPQNLIKQNCELFEQLGEYKFQNALLVRYTKKVPQVSTPTLVEV
    SRNLGKVGSKCCKHPEAKRMPCAEDYLSVVLNQLCVLHEKTPVSDRVTKC
    CTESLVNRRPCFSALEVDETYVPKEFNAETFTFHADICTLSEKERQIKKQ
    TALVELVKHKPKATKEQLKAVMDDFAAFVEKCCKADDKETCFAEEGKKLV
    AASQAALGL
  • Alloalbumin
  • An alloalbumin is a genetic variant of albumin. In certain embodiments the alloalbumin is human alloalbumin (HAA). Alloalbumins that differ in electrophoretic mobility from albumin have been identified through population genetics surveys in the course of clinical electrophoresis, or in blood donor surveys. As markers of mutation and migration, alloalbumins are of interest to geneticists, biochemists, and anthropologists, but most of these alloalbumin are not associated with disease (Minchioti et al. Human Mutations 29(8), 1007-1016(2008)).
  • TABLE 1
    List of substitutions comprised by various alloalbumins
    as compared to HA of SEQ ID NO: 1. Thermostability,
    half-life information and other HAAs are provided in
    Krogh-hansen et al. Biochim Biophys Acta 1747, 81-88(2005);
    and WO2011051489 incorporated by reference herein.
    Thermostability (C.) Effect on
    (positive = stabilizing, half-life
    Mutation negative = destabilizing) (% change)
    H3Y N/A N/A
    H3Q N/A N/A
    Q32Stop N/A N/A
    E60K N/A N/A
    D63N 6.07 N/A
    L66P N/A N/A
    E82K 2.03 N/A
    R114G N/A N/A
    R114Stop N/A N/A
    E119K N/A N/A
    V122E 0.57 N/A
    H128R N/A N/A
    Y140C N/A N/A
    A175Stop N/A N/A
    C177F −1.59  N/A
    R218H N/A N/A
    R218P N/A N/A
    K225Q* −4.86  N/A
    K240E N/A N/A
    E244Stop N/A N/A
    Q268R N/A N/A
    D269G 3.67 N/A
    K276N 4.87 N/A
    K313N −7.16  N/A
    D314G −0.38  N/A
    D314V N/A N/A
    N318K N/A N/A
    A320T, & -1R N/A 6.16
    E321K 1.42 N/A
    E333K −2.56  N/A
    E354K N/A N/A
    E358K N/A N/A
    K359K −6.56  N/A
    D365H 0.89 N/A
    D365V N/A N/A
    E368G N/A N/A
    K372E N/A N/A
    D375N N/A N/A
    D375H −0.09  N/A
    E376K N/A N/A
    E376Q N/A N/A
    E382K N/A N/A
    Q385Stop N/A N/A
    Y401Stop N/A N/A
    R410C N/A N/A
    E479K N/A N/A
    D494N N/A 0.84
    E501K 0.13 N/A
    E505K 1.87 N/A
    I513N N/A N/A
    V533M N/A N/A
    K536E N/A N/A
    K541E 6.12 N/A
    D550G N/A N/A
    D550A N/A N/A
    K560E 0.70 N/A
    D563N 4.17 N/A
    E565K N/A N/A
    E570K −6.53  N/A
    K573E 2.08 2.7 
    K574N N/A N/A
    L575insertion(TCCCKSSCLR −5.30  N/A
    LITSHLKASQPTMRIRERK)
    Frameshift after 567; Stop N/A −5.7% 
    at 582
    Frameshift after 572; Stop N/A −8.9% 
    at 578
  • Annexin:
  • As used herein, “annexin” refers to a group of cellular proteins found in eukaryotic organisms. Annexin is also known as lipocortin. As used herein “annexin” may refer to any annexin protein, or to specific annexin proteins such as “annexin A1,” “annexin A2,” and “annexin A5.” Annexins are characterized by their calcium dependent ability to bind negatively charged phospholipids (i.e. membrane walls). Annexins are characterized by a repeat protein scaffold limited to 30-50 kDa in size with fairly ubiquitous tissue distribution. The basic structure of an annexin is composed of two domains: a structurally conserved C terminal “core” region and a divergent N terminal domain. The core region binds the phospholipid cellular membrane in a Ca2+ dependent manner. The N terminal region binds cytoplasmic proteins. Annexins are important in various cellular and physiological processes and provide a membrane scaffold. The C terminal core is composed of four annexin repeats. Annexin is characterized by its flexible repeat-like nature that influences its intrinsic membrane-sensing abilities. For instance, the affinity towards specific biomembranes can be controlled by the number of repeats. With the characteristic phospholipid sensing, annexin can be useful to sense/target intestinal junctions for drug delivery. Another potential application for an annexin is targeting intestinal tight junctions and the Zonula Occludens region (ZO-1), which is known to be particularly difficult to traverse for larger protein therapeutics, significantly impairing drug absorption.
  • The term “quasi-annexin” refers to a heteromultimer molecule that has structure and/or function similar to the whole annexin, and wherein said heteromultimer molecule is formed by the assembly of two or more monomeric polypeptides designed based on the sequence of the whole annexin. In certain embodiments, the monomeric polypeptides are “segments” that preferentially associate as heteromultimeric pairs to form a quasi-protein. In some embodiments, the quasi-annexin has 90% of the activity of the whole annexin. In some embodiments, the quasi-annexin has 75% of the activity of whole-annexin. In an embodiment, the quasi-annexin has 50% of the activity of whole annexin. In some embodiments, the quasi-annexin has 50-75% of the activity of whole annexin. In an embodiment, quasi-annexin has 80% of the activity of whole annexin. In some embodiments, the quasi-annexin has 90% of the structure of whole annexin as determined by molecular modeling. In some embodiments, the quasi-annexin has 80% of the structure of whole annexin as determined by molecular modeling. In some embodiments, the quasi-annexin has 70% of the structure of whole annexin as determined by molecular modeling. In some embodiments, the quasi-annexin has 50% of the structure of whole annexin as determined by molecular modeling. In some embodiments, the quasi-annexin has 50%-75% of the structure of whole annexin as determined by molecular modeling.
  • The sequence of Human wild-type Annexin A2 is as shown:
  • SEQ ID NO: 14
    GSAVSPYPTFNPSSDVAALHKAIMVKGVDEATIIDILTKRNNAQRQQIKA
    AYLQETGKPLDETLKKALTGHLEEVVLALLKTPAQFDADELRAAMKGLGT
    DEDTLIEILASRTNKEIRDINRVYREELKRDLAKDITSDTSGDFRNALLS
    LAKGDRSEDFGVNEDLADSDARALYEAGERRKGTDVNVFNTILTTRSYPQ
    LRRVFQKYTKYSKHDMNKVLDLELKGDIEKCLTAIVKCATSKPAFFAEKL
    HQAMKGVGTRHKALIRIMVSRSEIDMNDIKAFYQKMYGISLCQAILDETK
    GDYEKILVALCGGN
  • Transferrin:
  • Transferrins are monomeric proteins of about 76 kDa molecular weight present in all vertebrates and function as a iron-binding and transporting protein. Recombinant human transferrin and its fusions is being considered for the management of various diseases including thalassemia, atransferrinemia, age related macular degeneration, type 2 diabetes, during stem cell transplantation and in the treatment of acute infectious disease caused by the anthrax bacteria. Transferrin is stable in the gastrointestinal environment and a number of studies have shown that intact protein-transferrin conjugates can be orally delivered and remain bioactive.
  • The term “quasi-transferrin” refers to a heteromultimer molecule that has structure and/or function similar to the whole transferrin, and wherein said heteromultimer molecule is formed by the assembly of two or more monomeric polypeptides designed based on the sequence of the whole transferrin. In certain embodiments, the monomeric polypeptides are “segments” that preferentially associate as heteromultimeric pairs to form a quasi-protein. In some embodiments, the quasi-transferrin has 90% of the activity of the whole transferrin. In some embodiments, the quasi-transferrin has 75% of the activity of whole-transferrin. In an embodiment, the quasi-transferrin has 50% of the activity of whole transferrin. In some embodiments, the quasi-transferrin has 50-75% of the activity of whole transferrin. In an embodiment, quasi-transferrin has 80% of the activity of whole transferrin. In some embodiments, the quasi-transferrin has 90% of the structure of whole transferrin as determined by molecular modeling. In some embodiments, the quasi-transferrin has 80% of the structure of whole transferrin as determined by molecular modeling. In some embodiments, the quasi-transferrin has 70% of the structure of whole transferrin as determined by molecular modeling. In some embodiments, the quasi-transferrin has 50% of the structure of whole transferrin as determined by molecular modeling. In some embodiments, the quasi-transferrin has 50%-75% of the structure of whole transferrin as determined by molecular modeling.
  • The sequence of wildtype Human Transferrin is as shown:
  • SEQ ID NO: 19
    MRLAVGALLV CAVLGLCLAV PDKTVRWCAV SEHEATKCQS
    FRDHMKSVIP SDGPSVACVK KASYLDCIRA IAANEADAVT
    LDAGLVYDAY LAPNNLKPVV AEFYGSKEDP QTFYYAVAVV
    KKDSGFQMNQ LRGKKSCHTG LGRSAGWNIP IGLLYCDLPE
    PRKPLEKAVA NFFSGSCAPC ADGTDFPQLC QLCPGCGCST
    LNQYFGYSGA FKCLKDGAGD VAFVKHSTIF ENLANKADRD
    QYELLCLDNT RKPVDEYKDC HLAQVPSHTV VARSMGGKED
    LIWELLNQAQ EHFGKDKSKE FQLFSSPHGK DLLFKDSAHG
    FLKVPPRMDA KMYLGYEYVT AIRNLREGTC PEAPTDECKP
    VKWCALSHHE RLKCDEWSVN SVGKIECVSA ETTEDCIAKI
    MNGEADAMSL DGGFVYIAGK CGLVPVLAEN YNKSDNCEDT
    PEAGYFAVAV VKKSASDLTW DNLKGKKSCH TAVGRTAGWN
    IPMGLLYNKI NHCRFDEFFS EGCAPGSKKD SSLCKLCMGS
    GLNLCEPNNK EGYYGYTGAF RCLVEKGDVA FVKHQTVPQN
    TGGKNPDPWA KNLNEKDYEL LCLDGTRKPV EEYANCHLAR
    APNHAVVTRK DKEACVHKIL RQQQHLFGSN VTDCSGNFCL
    FRSETKDLLF RDDTVCLAKL HDRNTYEKYL GEEYVKAVGN
    LRKCSTSSLL EACTFRRP
  • Cargo Molecule:
  • A heteromultimer described herein comprises monomers that comprise at least one cargo molecule, and at least one transporter polypeptide, said cargo molecule and transporter polypeptide associated with one another, by means inclusive of, but not restricted to genetic fusion or chemical conjugation. In certain embodiments, at least one cargo molecule is a therapeutic agent. In certain agents, the cargo molecule is a toxin. In certain embodiments, the cargo molecule is an antigen, or analogs thereof. In an embodiment, the cargo molecule is a natural product, analog, or prodrug thereof. In certain embodiments, the cargo molecule is a therapeutic agent such as a cytotoxin, e.g., a cytostatic or cytocidal agent, a therapeutic agent or a radioactive metal ion, e.g., alpha-emitters such as, for example, 213Bi. A cytotoxin or cytotoxic agent includes any agent that is detrimental to cells. Examples include paclitaxol, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicin, doxorubicin, daunorubicin, dihydroxy anthracin dione, mitoxantrone, mithramycin, actinomycin D, 1-dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, and puromycin and analogs or homologs thereof. Therapeutic agents include, but are not limited to, antimetabolites (e.g., methotrexate, 6mercaptopurine, 6thioguanine, cytarabine, 5-fluorouracil decarbazine), alkylating agents (e.g., mechlorethamine, thioepa chlorambucil, melphalan, carmustine (BSNU) and lomustine (CCNU), cyclothosphamide, busulfan, dibromomannitol, streptozotocin, mitomycin C, and cis-dichlorodiamine platinum (II) (DDP) cisplatin), anthracyclines (e.g., daunorubicin (formerly daunomycin) and doxorubicin), antibiotics (e.g., dactinomycin (formerly actinomycin), bleomycin, mithramycin, and anthramycin (AMC)), and anti-mitotic agents (e.g., vincristine and vinblastine).
  • In certain embodiment, the cargo molecule is a biomolecule. In an embodiment, the cargo molecule is a natural or synthetic nucleic acid. In some embodiments, at least one cargo molecule is one or more of a DNA, PNA, and/or RNA oligomer. In certain embodiments, a heteromultimer described herein comprises monomeric proteins that comprise at least one cargo polypeptide, or fragments or variants thereof, and at least one transporter polypeptide, said cargo polypeptide and transporter polypeptide associated with one another, by means inclusive of, but not restricted to genetic fusion or chemical conjugation
  • As used herein, “Cargo polypeptide” refers to proteins, polypeptides, antibodies, peptides or fragments or variants thereof, having one or more therapeutic and/or biological activities. Cargo polypeptides encompassed by the invention include but are not limited to, proteins, polypeptides, peptides, antibodies, substrates or ligands to therapeutically relevant target proteins and biologics. (The terms peptides, proteins, and polypeptides are used interchangeably herein.) Specifically the term “Cargo polypeptide” encompasses antibodies and fragments and variants thereof. Thus a heteromultimer described herein may contain at least a fragment or variant of a cargo polypeptide, and/or at least a fragment or variant of an antibody. Additionally, in certain embodiments, the term “Cargo polypeptide” refers to the endogenous or naturally occurring correlate of a cargo polypeptide.
  • As a non-limiting example, a “Cargo biomolecule” is a biomolecule such as but not restricted to a protein, DNA, or RNA that is useful to treat, prevent or ameliorate a disease, condition or disorder. As a non-limiting example, a “Cargo polypeptide” may be one that binds specifically to a particular cell type (normal (e.g., lymphocytes) or abnormal e.g., (cancer cells)) and therefore may be used to target a compound (drug, or cytotoxic agent) to that cell type specifically.
  • In another non-limiting example, a “Cargo molecule” is a molecule that has a biological, activity, and in particular, a biological activity that is useful for treating preventing or ameliorating a disease. A non-inclusive list of biological activities that may be possessed by a Cargo molecule, for instance a Cargo polypeptide includes, enhancing the immune response, promoting angiogenesis, inhibiting angiogenesis, regulating hematopoietic functions, stimulating nerve growth, enhancing an immune response, inhibiting an immune response, or any one or more of the biological activities described herein.
  • Cargo polypeptides corresponding to a cargo polypeptide portion of a heteromultimer protein described herein, such as cell surface and secretory proteins, are often modified, by the attachment of one or more oligosaccharide groups. The modification, referred to as glycosylation, can dramatically affect the physical properties of proteins and can be important in protein stability, secretion, and localization. Glycosylation occurs at specific locations along the polypeptide backbone. There are usually two major types of glycosylation: glycosylation characterized by O-linked oligosaccharides, which are attached to serine or threonine residues; and glycosylation characterized by N-linked oligosaccharides, which are attached to asparagine residues in an Asn-X-Ser/Thr sequence, where X can be any amino acid except proline. N-acetylneuramic acid (also known as sialic acid) is usually the terminal residue of both N-linked and Blinked oligosaccharides. Variables such as protein structure and cell type influence the number and nature of the carbohydrate units within the chains at different glycosylation sites. Glycosylation isomers are also common at the same site within a given cell type.
  • Table 2 provides a non-exhaustive list of Cargo polypeptides that correspond to a Cargo polypeptide portion of a heteromultimer described herein. The “Cargo Polypeptide” column discloses Cargo polypeptide molecules followed by parentheses containing scientific and brand names that comprise, or alternatively consist of, that Cargo polypeptide molecule or a fragment or variant thereof. In an embodiment the cargo molecule is a molecule that binds to a protein disclosed in the “Cargo polypeptide” column, or in Zhu et al. (Nucleic Acids Res. 38(1), D787-D791 (2009)); Wishart et al. (Nucleic Acids Res 36, D901-D906 (2008)); Ahmed et al. (Nucleic Acids Res 39, D960-D967 (2011)) incorporated by reference herein, or a protein that belongs in the class of therapeutic target molecules.
  • “Cargo polypeptide” as used herein may refer either to an individual Cargo polypeptide molecule (as defined by the amino acid sequence obtainable from the CAS and Genbank accession numbers), or to the entire group of Cargo polypeptide associated with a given Cargo polypeptide molecule disclosed in this column, or a Cargo polypeptide that binds to a polypeptide molecule disclosed in this column.
  • TABLE 2
    Non-exhaustive list of Cargo polypeptides that correspond to a Cargo polypeptide portion of a heteromultimer
    Cargo Polypeptide Biological Activity Exemplary Activity Assay Indication
    EPO (Erythropoietin; Stimulates cellular Cell proliferation assay Anemia; Anemia in Renal Disease;
    Epoetin alfa; Epoetin differentiation of bone- using a erythroleukemic Anemia in Oncology Patients;
    beta; Gene- activated marrow stem cells at an cell line TF-1. (Kitamura Bleeding Disorders; Chronic Renal
    erythropoietin; early stage of et al. 1989 J. Cell. Failure; Chronic Renal Failure in Pre-
    Darbepoetin- alpha; etythropoiesis; Physiol. 140: 323) Dialysis Patients; Renal Disease;
    NESP; Epogen; Procrit; accelerates the End-Stage Renal Disease; End-Stage
    Eprex; Erypo; Espo; proliferation and Renal Disease in Dialysis Patients;
    Epoimmun; EPOGIN; maturation of Chemotherapy; Chemotherapy in
    NEORECORMON; terminally Cancer Patients; Anemia in
    HEMOLINK; Dynepo; differentiating cells into zidovudine-treated HIV patients;
    ARANESP) erythrocytes; and Anemia in zidovudine-treated
    modulates the level of patients; Anemia in HIV patients;
    circulating erythrocytes. Anemia in premature infants;
    Surgical patients (pre and/or post
    surgery); Surgical patients (pre
    and/or post surgery) who are anemic;
    Surgical patients (pre and/or post
    surgery) who are undergoing elective
    surgery; Surgical patients (pre and/or
    post surgery) who are undergoing
    elective, non-cardiac surgery;
    Surgical patients (pre and/or post
    surgery) who are undergoing elective,
    non-cardiac, non-vascular surgery;
    Surgical patients (pre and/or post
    surgery) who are undergoing elective,
    non-vascular surgery; Surgical
    patients (pre and/or post surgery)
    who are undergoing cardiac and/or
    vascular surgery; Aplastic anemia;
    Refractory anemia; Anemia in
    Inflammatory Bowel Disease;
    Refractory anemia in Inflammatory
    Bowel Disease; Transfusion
    avoidance; Transfusion avoidance for
    surgical patients; Transfusion
    avoidance for elective surgical
    patients; Transfusion avoidance for
    elective orthopedic surgical patients;
    Patients who want to Increase Red
    Blood Cells.
    G-CSF (Granulocyte Stimulates the Proliferation of murine Chemoprotection; Adjunct to
    colony- stimulating proliferation and NFS-60 cells (Weinstein Chemotherapy; Inflammatory
    factor; Granulokine; differentiation of the et al, Proc Natl Acad Sci disorders; Cancer; Leukemia;
    KRN 8601; Filgrastim; progenitor cells for USA 1986; 83, pp5010-4) Myelocytic leukemia; Neutropenia,
    Lenograstim; granulocytes and Primary neutropenias (e.g.;
    Meograstim; monocytes- Kostmann syndrome); Secondary
    Nartograstim; macrophages. neutropenia; Prevention of
    Neupogen; NOPIA; neutropenia; Prevention and
    Gran; GRANOCYTE; treatment of neutropenia in HIV-
    Granulokine; Neutrogin; infected patients; Prevention and
    Neu-up; Neutromax) treatment of neutropenia associated
    with chemotherapy; Infections
    associated with neutropenias;
    Myelopysplasia; Autoimmune
    disorders; Psoriasis; Mobilization of
    hematopoietic progenitor cells;
    Wound Healing; Autoimmune
    Disease; Transplants; Bone marrow
    transplants; Acute myelogeneous
    leukemia; Lymphoma, Non-
    Hodgkin's lymphoma; Acute
    lymphoblastic leukemia; Hodgkin's
    disease; Accelerated myeloid
    recovery; Glycogen storage disease.
    GM-CSF (Granulocyte- Regulates Colony Stimulating Bone Marrow Disorders; Bone
    macrophage colony- hematopoietic cell Assay: Testa, N. G., et marrow transplant; Chemoprotection;
    stimulating factor; differentiation, gene al., “Assays for Hepatitis C; HIV Infections; Cancer;
    rhuGM- CSF; BI 61012; expression, growth, and hematopoietic growth Lung Cancer; Melanoma; Malignant
    Prokine; Molgramostim; function. factors.” Balkwill F R melanoma; Mycobacterium avium
    Sargramostim; GM- (edt) Cytokines, A complex; Mycoses; Leukemia;
    CSF/IL 3 fusion; practical Approach, pp Myeloid Leukemia; Infections;
    Milodistim; 229-44; IRL Press Neonatal infections; Neutropenia;
    Leucotropin; Oxford 1991. Mucositis; Oral Mucositis; Prostate
    PROKINE; Cancer; Stem Cell Mobilization;
    LEUKOMAX; Vaccine Adjuvant; Ulcers (such as
    Interberin; Leukine; Diabetic, Venous Stasis, or Pressure
    Leukine Liquid; Ulcers); Prevention of neutropenia;
    Pixykine) Acute myelogenous leukemia;
    Hematopoietic progenitor cell
    mobilization; Lymphoma; Non-
    Hodgkin's lymphoma; Acute
    Lymphoblastic Leukemia; Hodgkin's
    disease; Accelerated myeloid
    recovery; Transplant Rejection;
    Xenotransplant Rejection.
    Human growth hormone Binds to two GHR Ba/F3-hGHR Acromegaly; Growth failure; Growth
    (Pegvisamont; molecules and Induces proliferation assay, a hormone replacement; Growth
    Somatrem; Somatropin; signal transduction novel specific bioassay hormone deficiency; Pediatric
    TROVERT; through receptor for serum human growth Growth Hormone Deficiency; Adult
    PROTROPIN; BIO- dimerization hormone. J Clin Growth Hormone Deficiency;
    TROPIN; Endocrinol Metab 2000 Idiopathic Growth Hormone
    HUMATROPE; November; 85(11): Deficiency; Growth retardation;
    NUTROPIN; 4274-9 Prader- Willi Syndrome; Prader-Willi
    NUTROPIN AQ; Plasma growth hormone Syndrome in children 2 years or
    NUTROPHIN; (GH) immunoassay and older; Growth deficiencies; Growth
    NORDITROPIN; tibial bioassay, Appl failure associated with chronic renal
    GENOTROPIN; Physiol 2000 December; insufficiency; Osteoporosis;
    SAIZEN; SEROSTIM) 89(6): Postmenopausal osteoporosis;
    2174-8 Growth hormone Osteopenia, Osteoclastogenesis;
    (hGH) receptor mediated burns; Cachexia; Cancer Cachexia;
    cell mediated Dwarfism; Metabolic Disorders;
    proliferation, Growth Obesity; Renal failure; Turner's
    Horm IGF Res Syndrome; Fibromyalgia; Fracture
    2000 October; treatment; Frailty, AIDS wasting;
    10(5): 248-55 Muscle Wasting; Short Stature;
    International standard Diagnostic Agents; Female
    for growth hormone, Infertility; lipodystrophy.
    Horm Res 1999; 51
    Suppl 1: 7-12
    Insulin (Human insulin; Stimulates glucose Insulin activity may be Hyperglycemia; Diabetes; Diabetes
    Insulin aspart; Insulin uptake and promotes assayed in vitro using a Insipidus; Diabetes mellitus; Type 1
    Glargine; Insulin lispro; glycogenesis and [3-H]-glucose uptake diabetes; Type 2 diabetes; Insulin
    Lys-B28 Pro- B29; lipogenesis. assay. (J Biol Chem 1999 resistance; Insulin deficiency;
    lyspro; LY 275585; Oct. 22; 274(43): 30864- Hyperlipidemia; Hyperketonemia;
    diarginylinsulin; Des- 30873). Non- insulin dependent Diabetes
    B26- B30-insulin- B25- Mellitus (NIDDM); Insulin-
    amide; Insulin detemir; dependent Diabetes Mellitus
    LABI; NOVOLIN; (IDDM); A Condition Associated
    NOVORAPID; With Diabetes Including, But Not
    HUMULIN; Limited To Obesity, Heart Disease,
    NOVOMIX 30; Hyperglycemia, Infections,
    VELOSULIN; Retinopathy, And/Or Ulcers;
    NOVOLOG; LANTUS; Metabolic Disorders; Immune
    ILETIN; HUMALOG; Disorders; Obesity; Vascular
    MACRULIN; Disorders; Suppression of Body
    EXUBRA; INSUMAN; Weight; Suppression of Appetite;
    ORALIN; ORALGEN; Syndrome X.
    HUMAHALE;
    HUMAHALIN)
    Interferon alfa Confers a range of Anti-viral assay: Viral infections; HIV Infections;
    (Interferon alfa-2b; cellular responses Rubinstein S, Familletti Hepatitis; Chronic Hepatitis;
    recombinant; Interferon including antiviral, P C, Pestka S. (1981) Hepatitis B; Chronic Hepatitis B;
    alfa- n1; Interferon alfa- antiproliferative, Convenient assay for Hepatitis C; Chronic Hepatitis C;
    n3; Peginterferon alpha- antitumor and interferons. J. Virol. Hepatitis D; Chronic Hepatitis D;
    2b; Ribavirin and immunomodulatory 37(2): 755-8; Anti- Human Papillomavirus; Herpes
    interferon alfa- 2b; activities; stimulate proliferation assay: Gao Simplex Virus Infection; External
    Interferon alfacon-1; production of two Y, et al (1999) Sensitivity Condylomata Acuminata; HIV; HIV
    interferon consensus; enzymes: a protein of an epstein-barr virus- Infection; Oncology; Cancer; Solid
    YM 643; CIFN; kinase and an positive tumor line, Tumors; Melanoma; Malignant
    interferon- alpha oligoadenylate Daudi, to alpha interferon Melanoma; Renal Cancer (e.g., Renal
    consensus; recombinant synthetase. correlates with Cell Carcinoma); Lung Cancer (e.g,.
    methionyl consensus expression of a GC-rich Non-Small Cell Lung Cancer or
    interferon; recombinant viral transcript. Mol Cell Small Cell Lung Cancer) Colon
    consensus interferon; Biol. 19(11): 7305-13. Cancer; Breast Cancer; Liver Cancer;
    CGP 35269; RO Prostate Cancer; Bladder Cancer;
    253036; RO 258310; Gastric Cancer; Sarcoma; AIDS-
    INTRON A; PEG- Related Kaposi's Sarcoma;
    INTRON; OIF; Lymphoma; T Cell Lymphoma;
    OMNIFERON; PEG- Cutaneous T-Cell Lymphoma; Non-
    OMNIFERON; Hodgkin's Lymphoma; Brain Cancer;
    VELDONA; PEG- Glioma; Glioblastoma Multiforme;
    REBETRON; Cervical Dysplasia; Leukemia;
    ROFERON A; Preleukemia; Bone Marrow
    WELLFERON; Disorders; Bone Disorders; Hairy
    ALFERON N/LDO; Cell Leukemia; Chronic
    REBETRON; Myelogeonus Leukemia;
    ALTEMOL; Hematological Malignancies;
    VIRAFERON PEG; Hematological Disorders; Multiple
    PEGASYS; Myeloma; Bacterial Infections;
    VIRAFERON; Chemoprotection;
    VIRAFON; Thrombocytopenia; Multiple
    AMPLIGEN; Sclerosis; Pulmonary Fibrosis; Age-
    INFERGEN; Related Macular Degeneration;
    INFAREX; ORAGEN) Macular Degeneration; Crohn's
    Disease; Neurological Disorders;
    Arthritis; Rheumatoid Arthritis;
    Ulcerative Colitis; Osteoporosis,
    Osteopenia, Osteoclastogenesis;
    Fibromyalgia; Sjogren's Syndrome;
    Chronic Fatigue Syndrome; Fever;
    Hemmorhagic Fever; Viral
    Hemmorhagic Fevers;
    Hyperglycemia; Diabetes; Diabetes
    Insipidus; Diabetes mellitus; Type 1
    diabetes; Type 2 diabetes; Insulin
    resistance; Insulin deficiency;
    Hyperlipidemia; Hyperketonemia;
    Non- insulin dependent Diabetes
    Mellitus (NIDDM); Insulin-
    dependent Diabetes Mellitus
    (IDDM); A Condition Associated
    With Diabetes Including, But Not
    Limited To Obesity, Heart Disease,
    Hyperglycemia, Infections,
    Retinopathy, And/Or Ulcers;
    Metabolic Disorders; Immune
    Disorders; Obesity; Vascular
    Disorders; Suppression of Body
    Weight; Suppression of Appetite;
    Syndrome X.
    Calcitonin (Salmon Regulates levels of Hypocalcemic Rat Bone Disorders; Fracture prevention;
    Calcitonin (Salcatonin); calcium and phosphate Bioassay, bone resorbing Hypercalcemia; Malignant
    Calcitonin human- in serum; causes a assay and the pit assay, hypercalcemia; Osteoporosis; Paget's
    salmon hybrid; reduction in serum CT receptor binding disease; Osteopenia,
    Forcaltonin; Fortical; calcium--an effect assay, CAMP stimulation Osteoclastogenesis; osteolysis;
    Calcitonin; Calcitonina opposite to that of assay: J Bone Miner Res osteomyelitis; osteonecrosis;
    Almirall; Calcitonina human parathyroid 1999 August; 14(8): periodontal bone loss; osteoarthritis;
    Rubber; Calcimar; hormone. 1425-31 rheumatoid arthritis; osteopetrosis;
    Calsynar; Calogen; periodontal, lytic, or metastatic bone
    Miacalcic; Miacalcin; disease; osteoclast differentiation
    SB205614; Macritonin; inhibition; bone disorders; bone
    Cibacalcin; Cibacalcina; healing and regeneration.
    Cibacalcine;
    Salmocalcin;
    PowderJect Calcitonin)
    (CAS-21215-62-3)
    Interferon beta Modulates MHC Anti-viral assay: Multiple Sclerosis; Oncology;
    (Interferon beta-1a; antigen expression, NK Rubinstein S, Familletti Cancer; Solid Tumors; Melanoma;
    Interferon beta 1b; cell activity and IFNg P C, Pestka S. (1981) Malignant Melanoma; Renal Cancer
    Interferon- beta-serine; production and IL12 Convenient assay for (e.g., Renal Cell Carcinoma); Lung
    SH 579; ZK 157046; production in interferons. J. Virol. Cancer (e.g,. Non-Small Cell Lung
    BCDF; beta-2 IF; monocytes. 37(2): 755-8; Anti- Cancer or Small Cell Lung Cancer)
    Interferon- beta-2; rhIL- proliferation assay: Gao Colon Cancer; Breast Cancer; Liver
    6; SJ0031; DL 8234; Y, et al (1999) Sensitivity Cancer; Prostate Cancer; Bladder
    FERON; IFNbeta; of an epstein-barr virus- Cancer; Gastric Cancer; Sarcoma;
    BETASERON; positive tumor line, AIDS-Related Kaposi's Sarcoma;
    AVONEX; REBIF; Daudi, to alpha interferon Lymphoma; T Cell Lymphoma;
    BETAFERON; correlates with Cutaneous T-Cell Lymphoma; Non-
    SIGOSIX) expression of a GC-rich Hodgkin's Lymphoma; Brain Cancer;
    viral transcript. Mol Cell Glioma; Glioblastoma Multiforme;
    Biol. 19(11): 7305-13. Cervical Dysplasia; Leukemia;
    Preleukemia; Bone Marrow
    Disorders; Bone Disorders; Hairy
    Cell Leukemia; Chronic
    Myelogeonus Leukemia;
    Hematological Malignancies;
    Hematological Disorders; Multiple
    Myeloma; Bacterial Infections;
    Chemoprotection;
    Thrombocytopenia; Viral infections;
    HIV Infections; Hepatitis; Chronic
    Hepatitis; Hepatitis B; Chronic
    Hepatitis B; Hepatitis C; Chronic
    Hepatitis C; Hepatitis D; Chronic
    Hepatitis D; Human Papillomavirus;
    Herpes Simplex Virus Infection;
    External Condylomata Acuminata;
    HIV; HIV infection; Pulmonary
    Fibrosis; Age-Related Macular
    Degeneration; Macular Degeneration;
    Crohn's Disease; Neurological
    Disorders; Arthritis; Rheumatoid
    Arthritis; Ulcerative Colitis;
    Osteoporosis, Osteopenia,
    Osteoclastogenesis; Fibromyalgia;
    Sjogren's Syndrome; Chronic Fatigue
    Syndrome; Fever; Hemmorhagic
    Fever; Viral Hemmorhagic Fevers;
    Hyperglycemia; Diabetes; Diabetes
    Insipidus; Diabetes mellitus; Type 1
    diabetes; Type 2 diabetes; Insulin
    resistance; Insulin deficiency;
    Hyperlipidemia; Hyperketonemia;
    Non- insulin dependent Diabetes
    Mellitus (NIDDM); Insulin-
    dependent Diabetes Mellitus
    (IDDM); A Condition Associated
    With Diabetes Including, But Not
    Limited To Obesity, Heart Disease,
    Hyperglycemia, Infections,
    Retinopathy, And/Or Ulcers;
    Metabolic Disorders; Immune
    Disorders; Obesity; Vascular
    Disorders; Suppression of Body
    Weight; Suppression of Appetite;
    Syndrome X.
    Growth hormone Acts on the anterior Growth hormone- Acromegaly; Growth failure; Growth
    releasing factor; Growth pituitary to stimulate releasing peptides hormone replacement; Growth
    hormone releasing the production and (GHRPs) are known to hormone deficiency; Pediatric
    hormone (Sermorelin secretion of growth release growth hormone Growth Hormone Deficiency; Adult
    acetate; Pralmorelin; hormone and exert a (GH) in vivo and in vitro Growth Hormone Deficiency;
    Somatorelin; trophic effect on the by a direct action on Idiopathic Growth Hormone
    Somatoliberin; Geref; gland. receptors in anterior Deficiency; Growth retardation;
    Gerel; Groliberin) pituitary cells. Biological Prader- Willi Syndrome; Prader-Willi
    activity can be measured Syndrome in children 2 years or
    in cell lines expressing older; Growth deficiencies; Growth
    growth hormone failure associated with chronic renal
    releasing factor receptor insufficiency; Osteoporosis;
    (Mol Endocrinol 1992 Osteopenia, Osteoclastogenesis;
    October; 6(10): Postmenopausal osteoporosis; burns;
    1734-44, Cachexia; Cancer Cachexia;
    Molecular Dwarfism; Metabolic Disorders;
    Endocrinology, Vol 7, Obesity; Renal failure; Turner's
    77-84). Syndrome; Fibromyalgia; Fracture
    treatment; Frailty, AIDS wasting;
    Muscle Wasting; Short Stature;
    Diagnostic Agents; Female
    Infertility; lipodystrophy.
    IL-2 (Aldesleukin; Promotes the growth of T cell proliferation assay Cancer; Solid Tumors; Metastatic
    interleukin-2 fusion B and T cells and “Biological activity of Renal Cell Carcinoma; Metastatic
    toxin; T cell growth augments NK cell and recombinant human Melanoma; Malignant Melanoma;
    factor; PROLEUKIN; CTL cell killing interleukin-2 produced in Melanoma; Renal Cell Carcinoma;
    IMMUNACE; activity. Escherichia coli.” Renal Cancer; Lung Cancer (e.g,.
    CELEUK; ONCOLIPIN Science 223: 1412-1415, Non-Small Cell Lung Cancer or
    2; MACROLIN) 1984. natural killer (NK) Small Cell Lung Cancer); Colon
    cell and CTL cytotoxicity Cancer; Breast Cancer; Liver Cancer;
    assay “Control of Leukemia; Preleukemia;
    homeostasis of CD8+ Hematological Malignancies;
    memory T cells by Hematological Disorders; Acute
    opposing cytokines. Myeloid Leukemia; Melanoma;
    Science 288: 675-678, Malignant Melanoma; Non-
    2000; CTLL-2 Hodgkin's Lymphoma; Ovarian
    Proliferation: Gillis et al Cancer; Prostate Cancer; Brain
    (1978) J. Immunol. 120, Cancer; Glioma; Glioblastoma
    2027 Multiforme; Hepatitis; Hepatitis C;
    Lymphoma; HIV Infection (AIDS);
    Inflammatory Bowel Disorders;
    Kaposi's Sarcoma; Multiple
    Sclerosis; Arthritis; Rheumatoid
    Arthritis; Transplant Rejection;
    Diabetes; Type 1 Diabetes Mellitus;
    Type 2 Diabetes.
    Parathyroid hormone; Acts in conjuction with Adenylyl cyclase Bone Disorders; Fracture prevention;
    parathyrin (PTH; calcitonin to control stimulation in rat Hypercalcemia; Malignant
    Ostabolin; ALX1-11; calcium and phosphate osteosarcoma cells, hypercalcemia; Osteoporosis; Paget's
    hPTH 1-34; LY 333334; metabolism; elevates ovariectomized rat disease; Osteopenia,
    MN 10T; parathyroid blood calcium level; model of Osteoclastogenesis; osteolysis;
    hormone (1-31); stimulates the activity osteoporosis: IUBMB osteomyelitis; osteonecrosis;
    FORTEO; of osteocytes; enhances Life 2000 Februray; periodontal bone loss; osteoarthritis;
    PARATHAR) absorption of Ca+/Pi 49(2): 131-5 rheumatoid arthritis; osteopetrosis;
    from small intestine periodontal, lytic, or metastatic bone
    into blood; promotes disease; osteoclast differentiation
    reabsorption of Ca+ and inhibition; bone disorders; bone
    inhibits Pi by kidney healing and regeneration.
    tubules.
    Resistin Mediates insulin Ability of resistin to Hyperglycemia; Diabetes; Diabetes
    resistance in Type II influence type II diabetes Insipidus; Diabetes mellitus; Type 1
    diabetes; inhibits can be determined using diabetes; Type 2 diabetes; Insulin
    insulin-stimulated assays known in the art: resistance; Insulin deficiency;
    glucose uptake Pontoglio et al., J Clin Hyperlipidemia; Hyperketonemia;
    Invest 1998 May 15; Non- insulin dependent Diabetes
    101(10): 2215-22. Mellitus (NIDDM); Insulin-
    dependent Diabetes Mellitus
    (IDDM); A Condition Associated
    With Diabetes Including, But Not
    Limited To Obesity, Heart Disease,
    Hyperglycemia, Infections,
    Retinopathy, And/Or Ulcers;
    Metabolic Disorders; Immune
    Disorders; Obesity; Vascular
    Disorders; Suppression of Body
    Weight; Suppression of Appetite;
    Syndrome X.
    TR6 (DcR3; Decoy Inhibits Fas Ligand and Cellular apoptosis can Fas Ligand or LIGHT induced
    Receptor 3; FASTR) AIM-2 (TL5, LIGHT) be measured by annexin apoptotic disorders; hepatitis; liver
    mediated apoptosis. staining, TUNEL failure (including fulminant liver
    staining, measurement of failure); graft versus host disease;
    caspase levels. Inhibition graft rejection; myelodysplastic
    of cell growth can also be syndrome; renal failure; insulin
    directly measured, for dependent diabetes mellitus;
    example by ALOMAR rheumatoid arthritis; inflammatory
    Blue staining. Assay refs: bowel disease; autoimmune disease;
    cytotoxicity assay on toxic epidermal necrolysis; multiple
    human fibrosarcoma sclerosis.
    (Epsevik and Nissen-
    Meyer, 1986, J.
    Immunol. methods).
    DeCAF (D- SLAM; Inhibits proliferation DeCAF activity can be B cell and/or T cell mediated immune
    BCM-like membrane and differentiation of B determined using assays disorders; Immunodeficiency (e.g.,
    protein; BLAME (B cells; Antagonize BLyS known in the art, such as Common Variable
    lymphocyte activator activity for example, those Immunodeficiency, Selective IgA
    macrophage described in Examples Deficiency)
    expressed))x 32-33 of International
    Publication No.
    WO0111046.
    BLyS (B Lymphocyte Promotes proliferation, BLyS activity can be B cell and/or T cell mediated immune
    Stimulator; Neutrokine differentiation and determined using assays disorders, particularly immune
    alpha; TL7; BAFF; survival of B cells; known in the art, such as, system disorders associated with low
    TALL-1; THANK; Promotes for example, the B cell numbers or low serum
    radiolabeled BLyS) immunoglobulin costimulatory immunoglobulin; Immunodeficiency
    production by B cells. proliferation assay and (e.g., Common Variable
    other assays disclosed by Immunodeficiency, Selective IgA
    Moore et al., 1999, Deficiency). Radiolabeled forms;
    Science, 285(5425): 260-3. lymphoma, non-Hodgkins
    lymphoma, chronic lymphocytic
    leukemia, multiple myeloma.
    Anti-BLyS single chain Agonize or antagonize BLyS agonist or B cell and/or T cell mediated immune
    antibody (scFvI116A01, BlyS activity. antagonist activity can be disorders; Autoimmune disorders,
    scFvI050B11, determined using assays particularly autoimmune diseases
    scFvI006D08) and known in the art, such as, associated with the production of
    others. for example, a modified autoantibodies; Rheumatoid Arthritis,
    version the costimulatory Systemic Lupus Erythmatosus;
    proliferation assay Sjogren's Syndrome, cancers
    disclosed by Moore et al., expressing Blys as an autocrine
    1999, Science, growth factor, e.g. certain chronic
    285(5425): 260-3, in lymphocytic leukemias.
    which BlyS is mixed or
    preincubated with the
    anti-BlyS antibody prior
    to being applied to the
    responder B
    lymphocytes.
    MPIF-1 (Myeloid Inhibits myeloid MPIF-1 activity can be Chemoprotection; Adjunct to
    Progenitor Inhibitory progenitor cells; and measured using the Chemotherapy; Inflammatory
    Factor; CK beta-8; activates monocytes myeloprotection assay disorders; Cancer; Leukemia;
    Mirostipen) and chemotaxis assay Myelocytic leukemia; Neutropenia,
    described in U.S. Pat. No. Primary neutropenias (e.g.;
    6,001,606. Kostmann syndrome); Secondary
    neutropenia; Prevention of
    neutropenia; Prevention and
    treatment of neutropenia in HIV-
    infected patients; Prevention and
    treatment of neutropenia associated
    with chemotherapy; Infections
    associated with neutropenias;
    Myelopysplasia; Autoimmune
    disorders; Psoriasis; Mobilization of
    hematopoietic progenitor cells;
    Wound Healing; Autoimmune
    Disease; Transplants; Bone marrow
    transplants; Acute myelogeneous
    leukemia; Lymphoma, Non-
    Hodgkin's lymphoma; Acute
    lymphoblastic leukemia; Hodgkin's
    disease; Accelerated myeloid
    recovery; Glycogen storage disease.
    KDI (Keratinocyte Inhibits bone marrow KDI activity can be Multiple sclerosis; Hepatitis; Cancer;
    Derived Interferon; proliferation; and shows measured using the Viral infections, HIV infections,
    Interferon Kappa antiviral activity. antiviral and cell Leukemia.
    Precursor) proliferation assays
    described in Examples
    57-63 of International
    Publication No.
    WO0107608.
    TNFR2 (p75) Binds both TNFα and T-cell proliferation can Autoimmune disease; Rheumatoid
    (ENBREL) TNFβ; mediates T-cell be measured using assays Arthritis; Psoriatic arthritis; Still's
    proliferation by TNF; known in the art. For Disease; Ankylosing Spondylitis;
    reduces signs and example, “Lymphocytes: Cardiovascular Diseases; Vasulitis;
    structural damage in a practical approach” Wegener's granulomatosis;
    patients with edited by: S L Rowland, Amyloidosis; Systemic Lupus
    moderately to severely A J McMichael - chapter Erythematosus, Insulin-Dependent
    active rheumatoid 6, pages 138-160 Oxford Diabetes Mellitus; Immunodeficiency
    arthritis (RA). University Press (2000); Disorders; Infection; Inflammation;
    and “Current Protocols Inflammatory Bowel Disease;
    on CD-ROM” section Chrohn's Disease; Psoriasis; AIDS;
    3.12 Proliferation Assays Graft Rejection; Graft Versus Host
    for T-cell Function John Disease.
    Wiley & Soncs, Inc.
    (1999).
    Keratinocyte growth Stimulates epithelial KGF-2 activity can be Stimulate Epithelial Cell
    factor 2 (Repifermin; cell growth. measured using the Proliferation; Stimulate Basal
    KGF-2; Fibroblast wound healing assays and Keratinocytes; Wound Healing;
    Growth Factor-10; FGF-10) epithelial cell Stimulate Hair Follicle Production;
    proliferation assays Healing Of Dermal Wounds. Wound
    described in U.S. Pat. No. Healing; Eye Tissue Wounds, Dental
    6,077,692. Tissue Wounds, Oral Cavity
    Wounds, Diabetic Ulcers, Dermal
    Ulcers, Cubitus Ulcers, Arterial
    Ulcers, Venous Stasis Ulcers, Burns
    Resulting From Heat Exposure Or
    Chemicals, or Other Abnormal
    Wound Healing Conditions such as
    Uremia, Malnutrition, Vitamin
    Deficiencies or Complications
    Associated With Systemic Treatment
    With Steroids, Radiation Therapy or
    Antineoplastic Drugs or
    Antimetabolites; Promote Dermal
    Reestablishment Subsequent To
    Dermal Loss; Increase the Adherence
    Of Skin Grafts To A Wound Bed;
    Stimulate Re-Epithelialization from
    The Wound Bed; To Promote Skin
    Strength; Improve The Appearance
    Of Aged Skin; Proliferate
    Hepatocytes, Lung, Breast, Pancreas,
    Stomach, Bladder, Small Intestine,
    Large Intestine; Sebocytes, Hair
    Follicles, Type II Pneumocytes,
    Mucin- Producing Goblet Cells, or
    Other Epithelial Cells, Endothelial
    Cells, Keratinocytes, or Basal
    Keratinocytes (and Their Progenitors)
    Contained Within The Skin, Lung,
    Liver, Bladder, Eye, Salivary Glands,
    or Gastrointestinal Tract; Reduce The
    Side Effects Of Gut Toxicity That
    Result From Radiation,
    Chemotherapy Treatments Or Viral
    Infections; Cytoprotector, especially
    of the Small Intestine Mucosa or
    Bladder; Mucositis (Mouth Ulcers);
    Regeneration Of Skin; Full and/or
    Partial Thickness Skin Defects,
    including Burns, (e.g., Repopulation
    Of Hair Follicles, Sweat Glands, And
    Sebaceous Glands); Psoriasis;
    Epidermolysis Bullosa; Blisters;
    Gastric and/or Doudenal Ulcers;
    Reduce Scarring; Inflamamatory
    Bowel Diseases; Crohn's Disease;
    Ulcerative Colitis; Gut Toxicity;
    Lung Damage; Repair Of Alveoli
    And/or Brochiolar Epithelium; Acute
    Or Chronic Lung Damage;
    Emphysema, ARDS; Inhalation
    Injuries; Hyaline Membrane
    Diseases; Infant Respiratory Distress
    Syndrome; Bronchopulmonary
    Displasia In Premature Infants;
    Fulminant Liver Failure; Cirrhosis,
    Liver Damage caused by Viral
    Hepatitis and/or Toxic Substances;
    Diabetes Mellitus; Inflammation.
    TR2 (and TR2sv1, Inhibits B cell Co-stimulation B-cell Herpes; immune disorders;
    TR2SV2; TNFRSF14; proliferation, and proliferation assay and Ig autoimmune disease; graft versus
    HVEM; Herpes Virus mediates and inhibits production assay (Moore host disease; graft rejection; variable
    Entry Mediator; ATAR) Herpes Simplex Virus et al., 1999, Science, immunodeficiency;
    (HSV) infection. 285(5425): 260-3.). immunodeficiency syndromes;
    HSV-1 and HSV-2 cancer.
    Infectivity Assay:
    International Publication
    No. WO 97/
    Macrophage derived Chemotactic for 04658 Chemokine Inflammatory diseases; wound
    chemokine, MDC monocyte-derived activities can be healing;
    (Ckbeta-13) dendritic cells and IL-2- determined using assays angiogenesis; AIDS infection.
    activated natural killer known in the art:
    cells. Methods in Molecular
    Biology, 2000, vol. 138:
    Chemokine Protocols.
    Edited by: A. E. I.
    Proudfoot, T. N. C.
    Wells, and C. A. Power.
     ©Humana Press Inc.,
    Totowa, NJ
    HAGDG59 Activates MIP1a Dendritic cell assays Immune disorders; cancer; viral
    (Retinal release in Dendritic are well known in the infection; inflammation; sepsis;
    short-chain Cells. art. For example, J. arthritis; asthma.
    dehydrogenase) Immunol. 158: 2919-2925
    (1997); J. Leukoc. Biol.
    65: 822-828 (1999).
    GnRH (Gonadotropin Promotes release of GnRH is known to cause Infertility; Kallmann's syndrome or
    Releasing Hormone) follicle-stimulating the release of follicle other forms of hypergonadotropic
    hormone and stimulating hormone hypergonadism (failure to go through
    luteinizing hormone (FSH) and/or luteinizing puberty naturally).
    from anterior pituitary. hormone (LH) in vivo by
    a direct action on
    receptors in anterior
    pituitary gonadotropes.
    GnRH activity can be
    determined by measuring
    FSH levels in the
    medium of cultured
    gonadotropes before and
    after GnRH
    supplementation. For
    example, Baker et al.
    Biol Reprod 2000 September;
    63(3): 865-71.
    Teprotide Inhibits angiotensin Inhibition of ACE can be Hypertension; congestive heart
    converting enzyme determined using assays failure.
    (ACE). known in the art. For
    example,
    Anzenbacherova et al.,
    J. Pharma Biomed Anal
    2001 March; 24(5-6):
    1151-6.
    Human chemokine Involved in Chemokine activities can Autoimmune disorders; Immunity;
    HCC-1 (ckBeta-1; inflammation, allergy, be determined using Vascular and Inflammatory disorders;
    HWFBD) tissue rejection, viral assays known in the art: HIV; AIDS; infectious diseases.
    infection, and tumor Methods in Molecular
    biology; enhances Biology, 2000, vol. 138:
    proliferation of CD34+ Chemokine Protocols.
    myeloid progenitor Edited by: A. E. I.
    cells. Proudfoot, T. N. C.
    Wells, and C. A. Power.
     ©Humana Press Inc.,
    Totowa, NJ
    ACE2 inhibitor Inhibits production of Inhibition of angiotensin Treatment for elevated angiotensin II
    (DX512) angiotensin II which can be determined using and/or aldosterone levels, which can
    induces aldosterone assays known in the art. lead to vasoconstriction, impaired
    production, arteriolar For example, in vitro cardiac output and/or hypertension;
    smooth muscle using a proliferation Cardiovascular Disease; Cardiac
    vasoconstriction, and assay with rat cardiac Failure; Diabetes; Type II Diabetes;
    proliferation of cardiac fibroblasts as described in Proteinuria; Renal disorders,
    fibroblasts, Induces Naunyn Schmiedebergs congestive heart failure.
    angiogenesis; an Arch Pharmacol 1999
    enzyme that converts May; 359(5): 394-9.
    angiotensin I to
    angiotensin1-9; also
    cleaves des-Arg,
    bradykinin and
    neurotensin.
    TR1 (OCIF; Inhibits Coculture Assay for Osteoporosis; Paget's disease;
    Osteoclastogenesis osteoclastogenesis and Osteoclastogenesis, Bone osteopenia; osteolysis; osteomyelitis;
    inhibitory factor; bone resorption, and resorption assay using osteonecrosis; periodontal bone loss;
    osteoprotegerin, OPG; induces fibroblast fetal long-bone organ osteoarthritis; rheumatoid arthritis;
    tumor necrosis factor proliferation. culture system, dentine osteopetrosis; periodontal, lytic, or
    receptor superfamily resorption assay, and metastatic bone disease; osteoclast
    member 11B precursor;) fibroblast proliferation differentiation inhibition; bone
    assays are each described disorders; bone healing and
    in Kwon et al., FASEB J. regeneration; organ calcification;
    12: 845-854 (1998). vascular calcification.
    Human chemokine Chemotactic for both Chemokine activities can Cancer; Wound healing;
    Ckbeta-7 activated (CD3+) T be determined using Inflammatory disorders;
    cells and nonactivated assays known in the art: Immmunoregulatory disorders;
    (CD14−) lymphocytes Methods in Molecular Atherosclerosis; Parasitic Infection;
    and (CD4+) and Biology, 2000, vol. 138: Rheumatoid Arthritis; Asthma;
    (CD8+) T lymphocytes Chemokine Protocols. Autoimmune disorders.
    and (CD45RA+) T cells Edited by: A. E. I.
    Proudfoot, T. N. C.
    Wells, and C. A. Power.
     ©Humana Press Inc.,
    Totowa, NJ
    CKbeta4 (HGBAN46; Attracts and activates Chemokine activities can Cancer; Solid Tumors; Chronic
    HE9DR66) microbicidal be determined using Infection; Autoimmune Disorders;
    leukocytes; Attracts assays known in the art: Psoriasis; Asthma; Allergy;
    CCR6-expressing Methods in Molecular Hematopoiesis; Wound Healing;
    immature dendritic cells Biology, 2000, vol. 138: Bone Marrow Failure; Silicosis;
    and memory/effector T Chemokine Protocols. Sarcoidosis; Hyper-Eosinophilic
    cells; B-cell Edited by: A. E. I. Syndrome; Lung Inflammation;
    chemotaxis; inhibits Proudfoot, T. N. C. Fibrotic Disorders; Atherosclerosis;
    proliferation of myeloid Wells, and C. A. Power. Periodontal diseases; Viral diseases;
    progenitors; chemotaxis  ©Humana Press Inc., Hepatitis.
    of PBMC's. Totowa, NJ
    Leptin Controls obesity in vivo modulation of Hyperglycemia; Diabetes; Diabetes
    through regulation of food intake, reduction in Insipidus; Diabetes mellitus; Type 1
    appetite, reduction of body weight, and diabetes; Type 2 diabetes; Insulin
    body weight, and lowering of insulin and resistance; Insulin deficiency;
    lowering of insulin and glucose levels in ob/ob Hyperlipidemia; Hyperketonemia;
    glucose level. mice, radioimmunoassay Non-insulin dependent Diabetes
    (RIA) and activation of Mellitus (NIDDM); Insulin-
    the leptin receptor in a dependent Diabetes Mellitus
    cell-based assay. Protein (IDDM); a Condition Associated
    Expr Purif 1998 December; With Diabetes Including, But Not
    14(3): 335-42 Limited To Obesity, Heart Disease,
    Hyperglycemia, Infections,
    Retinopathy, And/Or Ulcers;
    Metabolic Disorders; Immune
    Disorders; Obesity; Vascular
    Disorders; Suppression of Body
    Weight; Suppression of Appetite;
    Syndrome X; Immunological
    Disorders; Immunosuppression.
    IL-1 receptor antagonist Binds IL1 receptor 1) Competition for IL-1 Autoimmune Disease; Arthritis;
    (Anakinra; soluble without activating the binding to IL-1 receptors Rheumatoid Arthritis; Asthma;
    interleukin-1 receptor; target cells; inhibits the in YT-NCI or C3H/HeJ Diabetes; Diabetes Mellitus; GVHD;
    IRAP; KINERET; binding of IL1-alpha cells (Carter et al., Nature Inflammatory Bowel Disorders;
    ANTRIL) and IL1-beta; and 344: 633-638, 1990); 2) Chron's Disease; Ocular
    neutralizes the biologic Inhibition of IL-1- Inflammation; Psoriasis; Septic
    activity of IL1-alpha induced endothelial cell- Shock; Transplant Rejection;
    and IL1-beta. leukocyte adhesion Inflammatory Disorders; Rheumatic
    (Carter et al., Nature 344: Disorders; Osteoporosis;
    633-638, 1990); 3) Postmenopausal Osteoporosis;
    Proliferation assays on Stroke.
    A375-C6 cells, a human
    melanoma cell line highly
    susceptible to the
    antiproliferative action of
    IL-1 (Murai T et al., J.
    Biol. Chem. 276: 6797-
    6806, 2001).
    TREM-1 (Triggering Mediates activation of Secretion of cytokines, Inflammation; Sepsis; bacterial
    Receptor Expressed on neutrophil and chemokines, infection; autoimmune diseases;
    Monocytes 1) monocytes; Stimulates degranulation, and cell GVHD.
    neutrophil and surface activation
    monocyte-mediated markers can be
    inflammatory response; determined using assays
    Promotes secretion of described in Bouchon et
    TNF, IL-8, and MCP-1; al, J Immunol 2000 May
    Induces neutrophil 15; 164(10): 4991-5.
    degranulation, Ca2+
    mobilization and
    tyrosine
    phosphorylation of
    extracellular signal-
    related kinase 1
    (ERK1), ERK2 and
    phospholipase C-
    gamma.
    HCNCA73 Induces T-cell FMAT can be used to Autoimmune disorders; Inflammation
    activation- expression measure T-cell surface of the gastrointestinal tract; Cancer;
    of CD152 marker; markers (CD69, CD152, Colon Cancer; Allergy; Crohn's
    Stimulates release of CD71, HLA-DR) and T- disease.
    TNF-a and MIP- 1a cell cytokine production
    from immature, (e.g., IFNg production).
    monocyte-derived J. of Biomol. Screen. 4:
    dendritic cells; 193-204 (1999). Other T-
    Promotes maturation of cell proliferation assays:
    dendritic cells. “Lymphocytes: a
    practical approach”
    edited by: S L Rowland,
    A J McMichael - Chapter
    6, pages 138-160 Oxford
    University Press (2000);
    WO 01/21658 Examples
    11-14, 16-17 and 33.
    VEGF-2 (Vascular Promotes endothelial VEGF activity can be Coronary artery disease; Critical limb
    Endothelial Growth cell proliferation. determined using assays ischemia; Vascular disease;
    Factor-2; VEGF-C) known in the art, such as proliferation of endothelial cells, both
    those disclosed in vascular and lymphatic. Antagonists
    International Publication may be useful as anti-angiogenic
    No. WO0045835, for agents; Cancer.
    example.
    HCHNF25 (jumping Activates MIP1a Dendritic cell assays are Immune disorders; cancer.
    translocation Release in Dendritic well known in the art. For
    breakpoint) Cells. example, J. Immunol.
    158: 2919-2925 (1997);
    J. Leukoc. Biol. 65: 822-
    828 (1999).
    HLDOU18 (Bone Activates L6/GSK3 Assays for activation of Hyperglycemia; Diabetes; Diabetes
    Morphogenic Protein
    9 kinase assay. GSK3 kinase activity are Insipidus; Diabetes mellitus; Type 1
    (BMP9); Growth well known in the art. For diabetes; Type 2 diabetes; Insulin
    differentiation factor-2 example, Biol. Chem. resistance; Insulin deficiency;
    precursor (GDF-2 379(8-9): (1998) 1101- Hyperlipidemia; Hyperketonemia;
    precursor)) 1110.; Biochem J. 1993 Non- insulin dependent Diabetes
    Nov. 15; 296 (Pt 1): 15-9. Mellitus (NIDDM); Insulin-
    dependent Diabetes Mellitus
    (IDDM); A Condition Associated
    With Diabetes Including, But Not
    Limited To Obesity, Heart Disease,
    Hyperglycemia, Infections,
    Retinopathy, And/Or Ulcers;
    Metabolic Disorders; Immune
    Disorders; Obesity; Vascular
    Disorders; Suppression of Body
    Weight; Suppression of Appetite;
    Syndrome X.
    Glucagon- Like-Peptide Stimulates the synthesis GLP1 activity may be Hyperglycemia; Diabetes; Diabetes
    1 (GLP1; and release of insulin; assayed in vitro using a Insipidus; Diabetes mellitus; Type 1
    Insulinotropin) enhances the sensitivity [3-H]-glucose uptake diabetes; Type 2 diabetes; Insulin
    of adipose, muscle, and assay. (J Biol Chem 1999 resistance; Insulin deficiency;
    liver tissues towards Oct. 22; 274(43): 30864- Hyperlipidemia; Hyperketonemia;
    insulin; stimulates 30873). Non- insulin dependent Diabetes
    glucose uptake; slows Mellitus (NIDDM); Insulin-
    the digestive process; dependent Diabetes Mellitus
    suppresses appetite; (IDDM); A Condition Associated
    blocks the secretion of With Diabetes Including, But Not
    glucagon. Limited To Obesity, Heart Disease,
    Hyperglycemia, Infections,
    Retinopathy, And/Or Ulcers;
    Metabolic Disorders; Immune
    Disorders; Obesity; Vascular
    Disorders; Suppression of Body
    Weight; Suppression of Appetite;
    Syndrome X.
    Exendin-4 (AC-2993) Stimulates the synthesis Exendin-4 activity may Hyperglycemia; Diabetes; Diabetes
    and release of insulin; be assayed in vitro using Insipidus; Diabetes mellitus; Type 1
    enhances the sensitivity a [3-H]-glucose uptake diabetes; Type 2 diabetes; Insulin
    of adipose, muscle, and assay. (J Biol Chem 1999 resistance; Insulin deficiency;
    liver tissues towards Oct. 22; 274(43): Hyperlipidemia; Hyperketonemia;
    insulin; stimulates 30864-30873). Non- insulin dependent Diabetes
    glucose uptake; slows Mellitus (NIDDM); Insulin-
    the digestive process; dependent Diabetes Mellitus
    suppresses appetite; (IDDM); A Condition Associated
    blocks the secretion of With Diabetes Including, But Not
    glucagon. Limited To Obesity, Heart Disease,
    Hyperglycemia, Infections,
    Retinopathy, And/Or Ulcers;
    Metabolic Disorders; Immune
    Disorders; Obesity; Vascular
    Disorders; Suppression of Body
    Weight; Suppression of Appetite;
    Syndrome X.
    T20 (T20 HIV a peptide from residues Virus inhibition assays as HIV; AIDS; SIV (simian
    inhibitory peptide, 643-678 of the HIV described in Zhang et al., immunodeficiency virus) infection.
    DP178; DP178 HIV gp41 transmembrane Sep. 26, 2002,
    inhibitory peptide) protein ectodomain Sciencexpress
    which binds to gp41 in (www.sciencexpress.org).
    its resting state and
    prevents transformation
    to the fusogenic state
    T1249 (T1249 HIV a second generation Virus inhibition assays as HIV; AIDS; SIV (simian
    inhibitory peptide; HIV fusion inbitor described in Zhang et al., immunodeficiency virus) infection
    T1249 anti-HIV Sep. 26, 2002,
    peptide) Sciencexpress
    (www.sciencexpress.org).
    Interferon Hybrids, Confers a range of Anti-viral assay: Viral infections; HIV Infections;
    specifically preferred: cellular responses Rubinstein S, Familletti Hepatitis; Chronic Hepatitis;
    IFNalpha A/D hybrid including antiviral P C, Peslka S. (1981) Hepatitis B; Chronic Hepatitis B;
    (BgIII version) antiproliferative, Convenient assay for Hepatitis C; Chronic Hepatitis C;
    IFNalpha A/D hybrid antitumor and interferons. J. Virol. Hepatitis D; Chronic Hepatitis D;
    (PvuII version) immunomodulatory 37(2): 755-8; Anti- Human Papillomavirus; Herpes
    IFNalpha A/F hybrid activities; stimulate proliferation assay: Gao Simplex Virus Infection; External
    IFNalpha A/B hybrid production of two Y, et al (1999) Sensitivity Condylomata Acuminata; HIV; HIV
    IFNbeta
    1/alpha D enzymes: a protein of an epstein-barr virus- Infection; Oncology; Cancer; Solid
    hybrid (IFNbeta- kinase and an positive tumor line. Tumors; Melanoma; Malignant
    1/alpha-1 hybrid) oligoadenylate Daudi, to alpha interferon Melanoma; Renal Cancer (e.g., Renal
    IFNalpha/beta hybrid synthetase. Also, correlates with Cell Carcinoma); Lung Cancer (e.g.,
    modulates MHC expression of a GC-rich Non-Small Cell Lung Cancer or
    antigen expression, NK viral transcript. Mol Cell Small Cell Lung Cancer) Colon
    cell activity and IFNg Biol. 19(11): 7305-13. Cancer; Breast Cancer; Liver Cancer;
    production and IL12 Prostate Cancer; Bladder Cancer;
    production in Gastric Cancer; Sarcoma; AIDS-
    monocytes. Related Kaposi's Sarcoma;
    Lymphoma; T Cell Lymphoma;
    Cutaneous T-Cell Lymphoma; Non-
    Hodgkin's Lymphoma; Brain Cancer;
    Glioma; Glioblastoma Multiforme;
    Cervical Dysplasia; Leukemia;
    Preleukemia; Bone Marrow
    Disorders; Bone Disorders; Hairy
    Cell Leukemia; Chronic
    Myelogeonus Leukemia;
    Hematological Malignancies;
    Hematological Disorders; Multiple
    Myeloma; Bacterial Infections;
    Chemoprotection;
    Thrombocytopenia; Multiple
    Sclerosis; Pulmonary Fibrosis; Age-
    Related Macular Degeneration;
    Macular Degeneration; Crohn's
    Disease; Neurological Disorders;
    Arthritis; Rheumatoid Arthritis;
    Ulcerative Colitis; Osteoporosis,
    Osteopenia, Osteoclastogenesis;
    Fibromyalgia; Sjogren's Syndrome;
    Chronic Fatigue Syndrome; Fever;
    Hemmorhagic Fever; Viral
    Hemmorhagic Fevers;
    Hyperglycemia; Diabetes; Diabetes
    Insipidus; Diabetes mellitus; Type 1
    diabetes; Type 2 diabetes; insulin
    resistance; Insulin deficiency;
    Hyperlipidemia; Hyperketonemia;
    Non- insulin dependent Diabetes
    Mellitus (NIDDM); Insulin-
    dependent Diabetes Mellitus
    (IDDM); A Condition Associated
    With Diabetes Including, But Not
    Limited To Obesity, Heart Disease,
    Hyperglycemia, Infections,
    Retinopathy, And/Or Ulcers;
    Metabolic Disorders; Immune
    Disorders; Obesity; Vascular
    Disorders; Suppression of Body
    Weight; Suppression of Appetite;
    Syndrome X.
    B-type natriuretic stimulates smooth Inhibition of angiotensin Congestive heart failure; cardiac
    peptide (BNP, brain muscle relaxation and can be determined using volume overload; cardiac
    natriuretic peptide) vasodilation, assays known in the art, decompensation; Cardiac Failure;
    natriuresis, and for example using an in Left Ventricular Dysfunction;
    suppression of renin- vitro proliferation assay Dyspnea
    angiotensin and with rat cardiac
    endothelin. fibroblasts as described in
    Naunyn Schmiedebergs
    Arch Pharmacol 1999
    May; 359(5): 394-9.
    Vasodilation can be
    measured in animals by
    measuring the myogenic
    responses of small renal
    arteries in an isobaric
    arteriograph system (see
    Am J Physiol Regul
    Integr Comp Physiol
    2002 August; 283(2): R349-
    R355). Natriuesis is
    determined by measuring
    the amount of sodium in
    the urine.
    α-defensin, including Suppression of HIV Virus inhibition assays as HIV, AIDS; ARC.
    alpha 1 defensin, alpha 2 replication; active described in Zhang et al.,
    defensin, alpha 3 against bacteria, fungi, Sep. 26, 2002,
    defensin (myeloid- and enveloped viruses. Sciencexpress
    related defensin; (www.sciencexpress.org).
    DEFA1; neutrophil-
    specific defensin; CAF)
    Phosphatonin (matrix Regulation of Blood phosphate levels Hyperphosphatemia;
    extracellular phosphate metabolism. can be measured using Hyperphosphatemia in chronic renal
    phosphoglycoprotein; methods known in the art failure; hypophosphatemia;
    MEPE) such as the Osteomalacia; Rickets; X-linked
    Hypophosphatemic Rat dominant hypophosphatemic
    Bioassay. Zoolog Sci rickets/osteomalacia (XLH) ;
    1995 October; 12(5): autosomal dominant
    607-10, hypophosphatemic
    rickets/osteomalacia (ADHR); tumor-
    induced rickets/osteomalacia (TIO).
    P1pal-12 (pepducin, Regulation of protease- Platelet aggregation can Protection against systemic platelet
    PAR1-based pepducin) activated receptor be measured using activation, thrombus, heart attack,
    (PAR) signal methods known in the art stroke, and/or coagulation disorders.
    transduction and such as described in
    thrombin-mediated Nature Medicine 2002
    aggregation of human October; 8(10): 1161-1165.
    platelets.
    P4pal-10 (pepducin, Regulation of protease- Platelet aggregation can Protection against systemic platelet
    PAR4-based pepducin) activated receptor be measured using activation, thrombus, heart attack,
    (PAR) signal methods known in the art stroke, and/or coagulation disorders.
    transduction and such as described in
    thrombin-mediated Nature Medicine 2002
    aggregation of human October; 8(10): 1161-1165.
    platelets.
    HRDFD27 Involved in the T-cell proliferation can Chemoprotection; Adjunct to
    proliferation of T cells; be measured using assays Chemotherapy; Inflammatory
    Production of known in the art. For disorders; Cancer; Leukemia;
    TNFgamma. example, “Lymphocytes: Myelocytic leukemia; Neutropenia,
    a practical approach” Primary neutropenias (e.g.;
    edited by: S L Rowland, Kostmann syndrome); Secondary
    A J McMichael - chapter neutropenia; Prevention of
    6, pages 138-160 Oxford neutropenia; Prevention and
    University Press (2000); treatment of neutropenia in HIV-
    and “Current Protocols infected patients; Prevention and
    on CD-ROM” section treatment of neutropenia associated
    3.12 Proliferation Assays with chemotherapy; Infections
    for T-cell Function John associated with neutropenias;
    Wiley & Soncs, Inc. Myelopysplasia; Autoimmune
    (1999). disorders; Psoriasis; Mobilization of
    hematopoietic progenitor cells;
    Wound Healing; Autoimmune
    Disease; Transplants; Bone marrow
    transplants; Acute myelogeneous
    leukemia; Lymphoma, Non-
    Hodgkin's lymphoma; Acute
    lymphoblastic leukemia; Hodgkin's
    disease; Accelerated myeloid
    recovery; Glycogen storage disease
    HWHGZ51 (CD59; Stimulates an immune The ability to affect Skeletal diseases and disorders;
    Metastasis- associated response and induces chondrocyte Musculoskeletal diseases and
    GPI-adhered protein inflammation by differentiation can be disorders; Bone fractures and/or
    homolog) inducing mononuclear measured using methods breaks; Osteoporosis
    cell, eosinophil and known in the art, such as (postmenopausal, senile, or idiopathic
    PMN infiltration; described in Bone (1995) juvenile); Gout and/or pseudogout;
    Inhibits growth of September; 17(3): Paget's disease; Osteoarthritis;
    breast cancer, ovarian 279-86. Tumors and/or cancers of the bone
    cancer, leukemia, and (osteochondromas, benign
    melanoma; chondromas, chondroblastomas,
    Overexpressed in colon, chondromyxoid fibromas, osteoid
    lung, breast and rectal osteomas, giant cell tumors, multiple
    tumors; Regulates myelomas, osteosarcomas,
    glucose and/or FFA fibrosarcomas, malignant fibrous
    update by adipocytes histiocytomas, chondrosarcomas,
    and skeletal muscle; Ewing's tumors, and/or malignant
    Induces lymphomas); Bone and joint
    redifferentiation of infections (osteomyelitits and/or
    chondrocytes infectious arthritis); Charcot's joints;
    Heel spurs; Sever's disease; Sport's
    injuries; Cancer; Solid Tumors;
    Melanoma; Malignant Melanoma;
    Renal Cancer (e.g., Renal Cell
    Carcinoma); Lung Cancer (e.g.. Non-
    Small Cell Lung Cancer or Small
    Cell Lung Cancer) Colon Cancer;
    Breast Cancer; Liver Cancer; Prostate
    Cancer; Bladder Cancer; Gastric
    Cancer; Sarcoma; AIDS-Related
    Kaposi's Sarcoma; Lymphoma; T
    Cell Lymphoma; Cutaneous T-Cell
    Lymphoma; Non-Hodgkin's
    Lymphoma; Brain Cancer; Glioma;
    Glioblastoma Multiforme; Cervical
    Dysplasia; Leukemia; Preleukemia;
    Bone Marrow Disorders; Bone
    Disorders; Hairy Cell Leukemia;
    Chronic Myelogeonus Leukemia;
    Hematological Malignancies;
    Hematological Disorders; Multiple
    Myeloma; Kidney diseases and
    disorders; Shonlein-Henoch purpura,
    Berger disease, celiac disease,
    dermatitis herpetiformis, Chron
    disease; Diabetes; Diabetes Insipidus;
    Diabetes mellitus; Type 1 diabetes;
    Type 2 diabetes; Insulin resistance;
    Insulin deficiency; Hyperlipidemia;
    Hyperketonemia; Non- insulin
    dependent Diabetes Mellitus
    (NIDDM); Insulin-dependent
    Diabetes Mellitus (IDDM); A
    Condition Associated With Diabetes
    Including, But Not Limited To
    Obesity, Heart Disease,
    Hyperglycemia, Infections,
    Retinopathy, And/Or Ulcers;
    Metabolic Disorders; Immune
    Disorders; Obesity; Vascular
    Disorders; Suppression of Body
    Weight; Suppression of Appetite;
    Syndrome X; Kidney disorders;
    Hyperinsulinemia; Hypoinsulinemia;
    Immunological disorders (e.g.
    arthritis, asthma, immunodeficiency
    diseases, AIDS, rheumatoid arthritis,
    granulomatous disease, inflammatory
    bowl disease, sepsis, acne,
    neutropenia, neutrophilia, psoriasis,
    hypersensitivities, T-cell mediated
    cytotoxicity, host-versus-graft
    disease, autoimmunity disorders,
    demyelination, systemic lupus
    erythematosis, drug induced
    hemolytic anemia, rheumatoid
    arthritis, Sjorgren's disease,
    scleroderma)
    C17 (cytokine- like Inhibits glucose and/or Proliferation of kidney Kidney diseases and disorders;
    protein C17) FFA uptake by mesangial cells can be Shonlein- Henoch purpura, Berger
    adipocytes; Induces assayed using techniques disease, celiac disease, dermatitis
    proliferation of kidney described in J. Investig. herpetiformis, Chron disease;
    mesangial cells; Med. (1998) August; Diabetes; Diabetes Insipidus;
    Regulation of cytokine 46(6): 297-302, Diabetes mellitus; Type 1 diabetes;
    production and antigen Type 2 diabetes; Insulin resistance;
    presentation Insulin deficiency; Hyperlipidemia;
    Hyperketonemia; Non-insulin
    dependent Diabetes Mellitus
    (NIDDM); Insulin- dependent
    Diabetes Mellitus (IDDM); A
    Condition Associated With Diabetes
    Including, But Not Limited To
    Obesity, Heart Disease,
    Hyperglycemia, Infections,
    Retinopathy, And/Or Ulcers;
    Metabolic Disorders; Immune
    Disorders; Obesity; Vascular
    Disorders; Suppression of Body
    Weight; Suppression of Appetite;
    Syndrome X; Kidney disorders;
    Hyperinsulinemia; Hypoinsulinemia;
    Hematopoietic disorders;
    Immunological diseases and
    disorders; Developmental diseases
    and disorders; Hepatic diseases and
    disorders; Cancer (particularly
    leukemia); Immunological disorders
    (e.g. arthritis, asthma,
    immunodeficiency diseases, AIDS,
    rheumatoid arthritis, granulomatous
    disease, inflammatory bowl disease,
    sepsis, acne, neutropenia,
    neutrophilia, psoriasis,
    hypersensitivities, T-cell mediated
    cytotoxicity, host- versus-graft
    disease, autoimmunity disorders,
    demyelination, systemic lupus
    erythematosis, drug induced
    hemolytic anemia, rheumatoid
    arthritis, Sjorgren's disease,
    scleroderma)
    HDPBQ71 Regulates production Such assays that may be Blood disorders and infection (e.g.,
    and secretion of used or routinely viral infections, tuberculosis,
    IFN gamma; Activation modified to test infections associated with chronic
    of myeloid cells and/or immunomodulatory granulomatosus disease and
    hematopoietic cells activity of polypeptides malignant osteoporosis);
    of the invention Autoimmune disease (e.g.,
    (including antibodies and rheumatoid arthritis, systemic lupus
    agonists or antagonists of erythematosis, multiple sclerosis);
    the invention) include the Immunodeficiency, boosting a T cell-
    assays disclosed in mediated immune response, and
    Miraglia el al., J suppressing a T cell- mediated
    Biomolecular Screening immune response; Inflammation and
    4: 193-204 (1999); inflammatory disorders; Idiopathic
    Rowland et al., pulmonary fibrosis; Neoplastic
    ““Lymphocytes: a diseases (e.g., leukemia, lymphoma,
    practical approach”” melanoma); Neoplasms and cancers,
    Chapter 6: 138-160 such as, for example, leukemia,
    (2000); Gonzalez et al., J lymphoma, melanoma, and prostate,
    Clin Lab Anal 8(5): 225- breast, lung, colon, pancreatic,
    233 (1995); Billiau et al., esophageal, stomach, brain, liver and
    Ann NY Acad Sci 856: urinary cancer;. Benign
    22-32 (1998); Boehm et dysproliferative disorders and pre-
    al., Annu Rev Immunol neoplastic conditions, such as, for
    15: 749-795 (1997), and example, hyperplasia, metaplasia,
    Rheumatology (Oxford) and/or dysplasia; Anemia;
    38(3): 214-20 (1999) Pancytopenia; Leukopenia;
    Thrombocytopenia; Hodgkin's
    disease; Acute lymphocytic anemia
    (ALL); Plasmacytomas; Multiple
    myeloma; Burkitt's lymphoma;
    Arthritis; AIDS; Granulomatous
    disease; Inflammatory bowel disease;
    Sepsis; Neutropenia; Neutrophilia;
    Psoriasis; Suppression of immune
    reactions to transplanted organs and
    tissues; Hemophilia;
    Hypercoagulation; Diabetes mellitus;
    Endocarditis; Meningitis; Lyme
    Disease; Asthma; Allergy
    Oscar (osteoclast- Regulator of osteoclast Assay to detect osteoclast Skeletal diseases and disorders;
    associated receptor differentiation; differentiation is Musculoskeletal diseases and
    isoform-3) regulator of innate and described in J. Exp. Med. disorders; Bone fractures and/or
    adaptive immune (2002) Jan. 21; 195(2): breaks; Osteoporosis
    responses 201-9. (postmenopausal, senile, or idiopathic
    juvenile); Gout and/or pseudogout;
    Paget's disease; Osteoarthritis;
    Tumors and/or cancers of the bone
    (osteochondromas, benign
    chondromas, chondroblastomas,
    chondromyxoid fibromas, osteoid
    osteomas, giant cell tumors, multiple
    myelomas, osteosarcomas,
    fibrosarcomas, malignant fibrous
    histiocytomas, chondrosarcomas,
    Ewing's tumors, and/or malignant
    lymphomas); Bone and joint
    infections (osteomyelitits and/or
    infectious arthritis); Charcot's joints;
    Heel spurs; Sever's disease; Sport's
    injuries
    Tumstatin (T5, T7 or T8 Inhibits angiogenesis; A tumor cell proliferation Cancer; Solid Tumors; Melanoma;
    peptide; α3(IV)NC1) Inhibits tumor growth; assay is described in J. Malignant Melanoma; Renal Cancer
    Inhibits protein Biol. Chem. (1997) 272: (e.g., Renal Cell Carcinoma); Lung
    synthesis 20395-20401, Protein Cancer (e.g,. Non-Small Cell Lung
    synthesis can be Cancer or Small Cell Lung Cancer)
    measured as described in Colon Cancer; Breast Cancer; Liver
    Science (2002) Jan. 4; Cancer; Prostate Cancer; Bladder
    295(5552): 140-3. Cancer; Gastric Cancer; Sarcoma;
    AIDS- Related Kaposi's Sarcoma;
    Lymphoma; T Cell Lymphoma;
    Cutaneous T-Cell Lymphoma; Non-
    Hodgkin's Lymphoma; Brain Cancer;
    Glioma; Glioblastoma Multiforme;
    Cervical Dysplasia; Leukemia;
    Preleukemia; Bone Marrow
    Disorders; Bone Disorders; Hairy
    Cell Leukemia; Chronic
    Myelogeonus Leukemia;
    Hematological Malignancies;
    Hematological Disorders; Multiple
    Myeloma; Angiogenesis
    CNTF (Ciliary Enhances myelin Regulation of myelin Neurological and neural diseases and
    neurotrophic factor) formation; Reduces formation can be assayed disorders, particularly diseases and
    photoreceptor as described in J. disorders associated with myelin and
    degradation; Regulates Neurosci. (2002) Nov. 1; demyelination, such as, for example,
    calcium currents 22(21): 9221-7. ALS, multiple sclerosis, Huntington's
    disease; Neuronal and spinal cord
    injuries; Disorders of the eye, such
    as, for example, retinitis pigmentosa,
    blindness, color-blindness, macular
    degeneration.
    Somatostatin Inhibits growth Inhibition of growth Cancer; Metastatic carcinoid tumors;
    (Octreotide; octreotide hormone, glucagons hormone release in Vasoactive Intestinal Peptide
    acetate; Sandostating and insulin; Suppresses humans by somatostatin secreting adenomas; Diarrhea and
    LAR ®) LF response to GnRH; can be measured as Flushing; Prostatic disorders and
    Decreases splanchnic described in J. Clin. cancers; Breast cancer;
    blood flow; Inhibits Endocrinol. Metab. Gastrointestinal disorders and
    release of serotonin, (1973) October; 37(4): cancers; Cancers of the endocrine
    gastrin, vasoactive 632-4. system; Head and neck
    intestinal peptide, Inhibition of insulin paragangliomas; Liver disorders and
    secretin, motilin, and secretion by somatostatin cancers; Nasopharyngeal cancers;
    pancreatic polypeptide. can be measured as Thyroid disorders and cancers;
    described in the Lancet Acromegaly; Carcinoid Syndrome;
    (1973) Dec. 8; 2(7841): Gallbladder disorders, such as
    1299-1301. gallbladder contractility diseases and
    abnormal bile secretion; Psoriasis;
    Diabetes; Diabetes Insipidus;
    Diabetes mellitus; Type 1 diabetes;
    Type 2 diabetes; Insulin resistance;
    Insulin deficiency; Hyperlipidemia;
    Hyperketonemia; Non- insulin
    dependent Diabetes Mellitus
    (NIDDM); Insulin-dependent
    Diabetes Mellitus (IDDM); A
    Condition Associated With Diabetes
    Including, But Not Limited To
    Obesity, Heart Disease,
    Hyperglycemia, Infections,
    Retinopathy, And/Or Ulcers;
    Metabolic Disorders; Immune
    Disorders; Obesity; Vascular
    Disorders; Suppression of Body
    Weight; Suppression of Appetite;
    Syndrome X; Kidney disorders;
    Neurological disorders and diseases,
    including Alzheimers Disease.
    Parkinson's disease and dementia;
    Neuropsychotic disorders, including
    Bipolar affective disorder;
    Rheumatoid arthritis; Hypertension;
    Intracranial hypertension; Esophageal
    varices; Graves' disease; Seizures;
    Epilepsy; Gastritis; Angiogenesis;
    IL-22 (IL22, interleukin- Stimulates glucose IL-22 activity may be Hyperglycemia; Diabetes; Diabetes
    22; IL17D, 1L27) uptake in skeletal assayed in vitro using a Insipidus; Diabetes mellitus; Type 1
    muscle cells; increases [3-H]-glucose uptake diabetes; Type 2 diabetes; Insulin
    skeletal muscle insulin assay. (J Biol Chem 1999 resistance; Insulin deficiency;
    sensitivity. Oct. 22; 274(43): 30864- Hyperlipidemia; Hyperketonemia;
    30873). Non- insulin dependent Diabetes
    Mellitus (NIDDM); Insulin-
    dependent Diabetes Mellitus
    (IDDM); A Condition Associated
    With Diabetes Including, But Not
    Limited To Obesity, Heart Disease,
    Hyperglycemia, Infections,
    Retinopathy, And/Or Ulcers;
    Metabolic Disorders; Immune
    Disorders; Obesity; Vascular
    Disorders; Suppression of Body
    Weight; Suppression of Appetite;
    Syndrome X.
    HCE1P80 Stimulates glucose HCE1P80 activity may Hyperglycemia; Diabetes; Diabetes
    uptake in; increases be assayed in vitro using Insipidus; Diabetes mellitus; Type 1
    insulin sensitivity, a [3-H]-glucose uptake diabetes; Type 2 diabetes; Insulin
    assay. (J Biol Chem 1999 resistance; Insulin deficiency;
    Oct. 22; 274(43): 30864- Hyperlipidemia; Hyperketonemia;
    30873), Non- insulin dependent Diabetes
    Mellitus (NIDDM); Insulin-
    dependent Diabetes Mellitus
    (IDDM); A Condition Associated
    With Diabetes Including, But Not
    Limited To Obesity, Heart Disease,
    Hyperglycemia, Infections,
    Retinopathy, And/Or Ulcers;
    Metabolic Disorders; Immune
    Disorders; Obesity; Vascular
    Disorders; Suppression of Body
    Weight; Suppression of Appetite;
    Syndrome X.
    HDRMI82 Stimulates glucose HDRMI82 activity may Hyperglycemia; Diabetes; Diabetes
    uptake; increases be assayed in vitro using Insipidus; Diabetes mellitus; Type 1
    insulin sensitivity. a [3-H]-glucose uptake diabetes; Type 2 diabetes; Insulin
    assay. (J Biol Chem 1999 resistance; Insulin deficiency;
    Oct. 22; 274(43): 30864- Hyperlipidemia; Hyperketonemia;
    30873), Non- insulin dependent Diabetes
    Mellitus (NIDDM); Insulin-
    dependent Diabetes Mellitus
    (IDDM); A Condition Associated
    With Diabetes Including, But Not
    Limited To Obesity, Heart Disease,
    Hyperglycemia, Infections,
    Retinopathy, And/Or Ulcers;
    Metabolic Disorders; Immune
    Disorders; Obesity; Vascular
    Disorders; Suppression of Body
    Weight; Suppression of Appetite;
    Syndrome X.
    HDALV07 Modulates insulin Insulin activity may be Diabetes; Diabetes Insipidus;
    (adiponectin; gelatin- action assayed in vitro using a Diabetes mellitus; Type 1 diabetes;
    binding 28k protein [3-H]-glucose uptake Type 2 diabetes; Insulin resistance;
    precursor; adipose most assay. (J Biol Chem 1999 Insulin deficiency; Hyperlipidemia;
    abundant gene Oct. 22; 274(43): 30864- Hyperketonemia; Non-insulin
    transcript; APM-1; 30873), dependent Diabetes Mellitus
    GBP28; ACRP30; (NIDDM); Insulin- dependent
    ADIPOQ) Diabetes Mellitus (IDDM); A
    Condition Associated With Diabetes
    Including, But Not Limited To
    Obesity, Heart Disease,
    Hyperglycemia, Infections,
    Retinopathy, And/Or Ulcers;
    Metabolic Disorders; Immune
    Disorders; Obesity; Vascular
    Disorders; Suppression of Body
    Weight; Suppression of Appetite;
    Syndrome X; Hyperglycemia;
    Familial combined hyperlipidemia;
    Metabolic syndrome; Inflammatory
    disorders; Atherogenic disorders
    C Peptide An insulin precursor C-peptide concentrations Diabetes; Diabetes Insipidus;
    involved in insulin can be measured using Diabetes mellitus; Type 1 diabetes;
    regulation assays well known in the Type 2 diabetes; Insulin resistance;
    art, such as the one Insulin deficiency; Hyperlipidemia;
    described in PNAS Hyperketonemia; Non-insulin
    (1970) September; 67(1): dependent Diabetes Mellitus
    148-55 (NIDDM); Insulin- dependent
    Diabetes Mellitus (IDDM); A
    Condition Associated With Diabetes
    Including, But Not Limited To
    Obesity, Heart Disease,
    Hyperglycemia, Infections,
    Retinopathy, And/Or Ulcers;
    Metabolic Disorders; Immune
    Disorders; Obesity; Vascular
    Disorders; Suppression of Body
    Weight; Suppression of Appetite;
    Syndrome X; Hyperglycemia;
    Familial combined hyperlipidemia;
    Metabolic syndrome
    HCBOG68 (enteric Controls proliferation/ Activation of cAMP- Treatment of Obesity; treatment of
    adipokine; Fat SID; differentiation or mediated transcription in Diabetes; suppression of body weight
    proline rich acidic metabolism/ adipocytes can be gain; suppression of appetite.
    protein) physiology/pathology/ assayed using methods Hyperglycemia; Diabetes; Diabetes
    of adipocytes and known in the art (Berger Insipidus; Diabetes mellitus; Type 1
    adipose tissue in et al., Gene 66: 1-10 diabetes; Type 2 diabetes; Insulin
    response to dietary (1998); Cullen and resistance; Insulin deficiency;
    conditions. Malm, Methods in Hyperlipidemia; Hyperketonemia;
    Enzymol 216: 362-368 Non- insulin dependent Diabetes
    (1992); Henthorn et al., Mellitus (NIDDM); Insulin-
    Proc Natl Acad Sci USA dependent Diabetes Mellitus
    85: 6342-6346 (1988); (IDDM); A Condition Associated
    Reusch et al., Mol Cell With Diabetes Including, But Not
    Biol 20(3): 1008-1020 Limited To Obesity, Heart Disease,
    (2000); and Klemm et al., Hyperglycemia, Infections,
    J Biol Chem 273: 917- Retinopathy, And/Or Ulcers;
    923 (1998)). Metabolic Disorders; Immune
    Disorders; Obesity; Vascular
    Disorders; Suppression of Body
    Weight; Suppression of Appetite;
    Syndrome X. Other indications for
    antibodies and/or antagonists, include
    treatment of weight loss; treatment of
    AIDS wasting; appetite stimulant;
    treatment of cachexia.
    PYY (Peptide YY), Decreases appetite; Appetite and food intake Most Preferred: Treatment of
    including PYY3-36 increases satiety; can be can be measured Obesity; treatment of Diabetes;
    (amino acid residues 31- decreases food intake. by methods known in the suppression of body weight gain;
    64 of full length PYY, art (Batterham et al. suppression of appetite.
    amino acid residues 3- Nature 2002; 418: Hyperglycemia; Diabetes; Diabetes
    36 of mature PYY) 650654) Insipidus; Diabetes mellitus; Type 1
    diabetes; Type 2 diabetes; Insulin
    resistance; Insulin deficiency;
    Hyperlipidemia; Hyperketonemia;
    Non- insulin dependent Diabetes
    Mellitus (NIDDM); Insulin-
    dependent Diabetes Mellitus
    (IDDM); A Condition Associated
    With Diabetes Including, But Not
    Limited To Obesity, Heart Disease,
    Hyperglycemia, Infections,
    Retinopathy, And/Or Ulcers;
    Metabolic Disorders; Immune
    Disorders; Obesity; Vascular
    Disorders; Suppression of Body
    Weight; Suppression of Appetite;
    Syndrome X. Other indications for
    antibodies, antagonists; treatment of
    weight loss; treatment of AIDS
    wasting; appetite stimulant; treatment
    of cachexia.
    WNT10b Inhibits adipogenesis. WNT10b activity can be Most Preferred: Treatment of
    measured using Obesity; suppression of body weight
    adipogenesis inhibition gain; suppression of appetite. Other
    assays (Ross et al., indications; Hyperglycemia;
    Science 2000; 289(5481): Diabetes; Diabetes Insipidus;
    950-953 Diabetes mellitus; Type 1 diabetes;
    Type 2 diabetes; Insulin resistance;
    Insulin deficiency; Hyperlipidemia;
    Hyperketonemia; Non-insulin
    dependent Diabetes Mellitus
    (NIDDM); Insulin- dependent
    Diabetes Mellitus (IDDM).
    WNT11 Promotes cardiogenesis. WNT11 activity can be Treatment of Cardiovascular
    measured using assays disorders; Congestive Heart Failure;
    known in the art, Myocardial Infarction.
    including cardiogenesis
    assays (Eisenberg et al.,
    Dev Dyn 1999 September;
    216(1): 45-58).
    Herstatin Inhibits cancer Herstatin activity can be Oncology; Cancer; Solid Tumors;
    proliferation. measured using cell Melanoma; Malignant Melanoma;
    proliferation assays Renal Cancer (e.g., Renal Cell
    known in the art (Doherty Carcinoma); Lung Cancer (e.g,. Non-
    et al., PNAS 1999; Small Cell Lung Cancer or Small
    96(19): 10869-10874. Cell Lung Cancer); Colon Cancer;
    Breast Cancer; Liver Cancer; Prostate
    Cancer; Bladder Cancer; Gastric
    Cancer; Sarcoma; AIDS- Related
    Kaposi's Sarcoma; Lymphoma; T
    Cell Lymphoma; Cutaneous T-Cell
    Lymphoma; Non-Hodgkin's
    Lymphoma; Brain Cancer; Glioma;
    Glioblastoma Multiforme; Cervical
    Dysplasia; Leukemia; Preleukemia;
    Hairy Cell Leukemia; Chronic
    Myelogeonus Leukemia;
    Hematological Malignancies;
    Hematological Disorders; Multiple
    Myeloma.
    Adrenomedullin stimulates vasodilation; Vasodilation can be Treatment of Congestive Heart
    promotes bone growth. measured using assays Failure; Hypertension; Myocardial
    known in the art (Ashton Infarction; Septic Shock;
    et al. Pharmacology Osteoporosis; Postmenopausal
    2000; 61(2): 101-105. osteoporosis; Osteopenia.
    The promotion of bone
    growth can be measured
    using assays known in
    the art, such as the
    osteoblast proliferation
    assay (Cornish et al. Am
    J Physiol 1997 December;
    273(6 Pt 1): E1113- 20).
    Nogo Receptor Receptor for the axon The promotion of axon Treatment of Central Nervous
    growth inhibitor, Nogo. regeneration and growth System Damage; Spinal Cord Injury;
    can be measured using Peripheral Nerve Damage;
    assays known in the art Neurodegenerative Diseases;
    (Fournier et al. Nature Parkinson's Disease; Alzheimer's
    2001; 409(6818): 341-346). Disease; Huntington's Disease;
    Amyotrophic Lateral Sclerosis;
    Progressive Supranuclear Palsy;
    Creutzfeld-Jacob Disease; Motor
    Neuron Disease.
    CART (Cocaine- and Inhibits food intact and Appetite and food intake Most Preferred: Treatment of
    Amphetamine- fat storage; promotes can be can be measured Obesity; suppression of body weight
    Regulated Transcript) lipid oxidation. by methods known in the gain; suppression of appetite. Other
    art (Batterham et al. indications; Hyperglycemia;
    Nature 2002; 418: Diabetes; Diabetes Insipidus;
    650654) Diabetes mellitus; Type 1 diabetes;
    Type 2 diabetes; Insulin resistance;
    Insulin deficiency; Hyperlipidemia;
    Hyperketonemia; Non-insulin
    dependent Diabetes Mellitus
    (NIDDM); Insulin- dependent
    Diabetes Mellitus (IDDM).
    RegIV (Colon Specific Stimulates glucose RegIV activity may be Hyperglycemia; Diabetes; Diabetes
    Gene; Colon Specific uptake; increases assayed in vitro using a Insipidus; Diabetes mellitus; Type 1
    Protein) insulin sensitivity. [3-H]-glucose uptake diabetes; Type 2 diabetes; Insulin
    assay. (J Biol Chem 1999 resistance; Insulin deficiency;
    Oct. 22; 274(43): 30864- Hyperlipidemia; Hyperketonemia;
    30873). Non- insulin dependent Diabetes
    Mellitus (NIDDM); Insulin-
    dependent Diabetes Mellitus
    (IDDM); A Condition Associated
    With Diabetes Including, But Not
    Limited To Obesity, Heart Disease,
    Hyperglycemia, Infections,
    Retinopathy, And/Or Ulcers;
    Metabolic Disorders; Immune
    Disorders; Obesity; Vascular
    Disorders; Suppression of Body
    Weight; Suppression of Appetite;
    Syndrome X.
    Cosyntropin (Cortrosyn) Synthetic corticotropin; The activity of Endocrine; Addison's disease;
    (CAS-16960- 16-0) stimulates the release of cosyntropin can be Cushing's syndrome; pituitary
    cortisol. assessed in vivo by dysfunction; acute adrenal crisis
    measuring serum cortisol
    levels. (Frank et al. J.
    Am. Vet. Med. Assoc.
    1998 212(10): 1569-71).
    Pexiganan Acetate Disrupts bacterial Pexiganan acetate Treatment of Infectious Diseases;
    (CAS-172820-23-4) membranes. activity can be assessed Treatment of Bacterial Infections.
    using in vitro
    antibacterial assays
    known in the art. (Zasloff
    et al., Antimicrobial
    Agents and
    Chemotherapy 1999, 43:
    782-788).
    Pramlintide (Amylin) Slows gastric emptying; Appetite and food intake Treatment of Obesity; treatment of
    (CAS-151126-32-8) decreases food intake. can be can be measured Diabetes; suppression of body weight
    by methods known in the gain; suppression of appetite;
    art (Batterham et al. treatment of endocrine disorders;
    Nature 2002; 418: Hyperglycemia; Diabetes; Diabetes
    650654) Insipidus; Diabetes mellitus; Type 1
    diabetes; Type 2 diabetes; Insulin
    resistance; Insulin deficiency;
    Hyperlipidemia; Hyperketonemia;
    Non- insulin dependent Diabetes
    Mellitus (NIDDM); Insulin-
    dependent Diabetes Mellitus
    (IDDM); A Condition Associated
    With Diabetes Including, But Not
    Limited To Obesity, Heart Disease,
    Hyperglycemia, Infections,
    Retinopathy, And/Or Ulcers;
    Metabolic Disorders; Immune
    Disorders; Obesity; Vascular
    Disorders; Suppression of Body
    Weight; Suppression of Appetite;
    Syndrome X. Other indications for
    antibodies, antagonists; treatment of
    weight loss; treatment of AIDS
    wasting; appetite stimulant; treatment
    of cachexia.
    Teriparatide (CAS- Acts in conjuction with Adenylyl cyclase Bone Disorders; Fracture prevention;
    52232- 67-4) calcitonin to control stimulation in rat Hypercalcemia; Malignant
    calcium and phosphate osteosarcoma cells, hypercalcemia; Osteoporosis; Paget's
    metabolism; elevates ovariectomized rat model disease; Osteopenia,
    blood calcium level; of osteoporosis: IUBMB Osteoclastogenesis; osteolysis;
    stimulates the activity Life 2000 February; 49(2): osteomyelitis; osteonecrosis;
    of osteocytes; enhances 131-5 periodontal bone loss; osteoarthritis;
    absorption of Ca+/Pi rheumatoid arthritis; osteopetrosis;
    from small intestine periodontal, lytic, or metastatic bone
    into blood; promotes disease; osteoclast differentiation
    reabsorption of Ca+ and inhibition; bone disorders; bone
    inhibits Pi by kidney healing and regeneration.
    tubules.
    Terlipressin (triglycyl Analog of vasopressin; Terlipressin activity can Variceal hemorrhage; cirrhosis;
    lycine vasopressin) induces be measured using assays portal hypertension; hepatorenal
    (CAS-14636- 12-5) vasoconstriction. of vasoconstriction, such syndrome; Blood-related disorders
    as the isolated arterial
    ring preparation.
    (Landstrom et al., Hum
    Reprod 1999 January; 14(1):
    151-5),
    Ularitide (CAS-118812- Stimulates natriuresis, Ularitide activity can be Excretory disorders; Acute renal
    69-4) diuresis, and assessed by measuring failure; asthma; congestive heart
    vasodilation. cGMP accumulation in failure; hypertension; pulmonary
    rat renal cells. (Valentin hypertension; cardiovascular
    et al., Hypertension 1993 disorders
    April; 21(4): 432-8).
    Aprotinin (Trasylol) Serine protease Inhibition of thrombin- Inhibition of fibrinolysis; reduction
    (CAS-9087-70-1; CAS- inhibitor; attenuates induced platelet of blood loss during surgery;
    11061-94-2; CAS- Systemic Inflammatory aggregation can be Treatment of Inflammation and
    12407- 79-3) Response, fibrinolysis measured using methods immune Disorders.
    and thrombin-induced known in the art. (Poullis
    platelet aggregation. et al., J Thorac
    Cardiovasc Surg
    2000
    August; 120(2): 370-8).
    Aspartocin (CAS-4117- Antibacteria Aspartocin activity can Treatment of Infectious Diseases;
    65-1; CAS- 1402-89-7) be assessed using in vitro treatment of bacterial infections.
    antibacterial assays
    known in the art. (Zasloff
    et al., Antimicrobial
    Agents and
    Chemotherapy 1999, 43:
    782-788).
    Calcitonin (Calcimar) Regulates levels of Hypocalcemic Rat Musculoskeletal; Osteroporosis;
    (CAS-21215-62-3) calcium and phosphate Bioassay, bone resorbing Paget's disease; hypercalcemia; Bone
    in serum; causes a assay and the pit assay, Disorders; Fracture prevention;
    reduction in serum CT receptor binding Malignant hypercalcemia;
    calcium--an effect assay, CAMP stimulation Osteopenia, Osteoclastogenesis;
    opposite to that of assay: J Bone Miner Res osteolysis; osteomyelitis;
    human parathyroid 1999 August; 14(8): osteonecrosis; periodontal bone loss;
    hormone. 1425-31 osteoarthritis; rheumatoid arthritis;
    osteopetrosis; periodontal, lytic, or
    metastatic bone disease; osteoclast
    differentiation inhibition; bone
    disorders; bone healing and
    regeneration.
    Carperitide (HANP; Stimulates natriuresis, Carperitide activity can Treatment of Heart Failure;
    recombinant human diuresis, and be assessed in vitro by Cardiovascular disorders; Respiratory
    atrial natriuretic peptide) vasodilation. measuring cGMP disorders; Acute respiratory distress
    (CAS-89213- 87-6) accumulation in a number syndrome.
    of cell lines, including
    PC12 cells and cultured
    human glomerular cells.
    (Medvede et al., Life Sci
    2001 Aug. 31; 69(15):
    1783-90; Green et al., J
    Am Soc Nephrol 1994
    October; 5(4): 1091-8).
    Desirudin (recombinant Inhibits thrombin; Desirudin activity can be Blood-related disorder; Thrombosis;
    hirudin; Revasc) (CAS- inhibits blood clotting. assessed using blood thrombocytopenia; hemorrhages.
    120993- 53-5) clotting assays known in
    the art, such as in vitro
    platelet aggregation
    assays. (Glusa,
    Haemostasis 1991; 21
    Suppl 1: 116-20).
    Emoctakin (interleukin proinflammatory Treatment of Inflammation, Immune
    8) (CAS-142298- 00-8) cytokine disorders. RSV infection.
    Felypressin (CAS-56- Derivative of Felypressin Treatment of pain; to induce local
    59-7) Vasopressin; Stimulates vasoconstriction activity anesthesia.
    vasoconstriction; can be measured using
    Induces local assays of
    anesthesia. vasoconstriction, such as
    the isolated arterial ring
    preparation. (Landstrom
    et al., Hum Reprod 1999
    January; 14(1): 151-5).
    Glucagon (CAS-16941- Induces hyperglycemia. Glucagon activity may be Hypoglycemia; Diabetes; Diabetes
    32-5) assayed in vitro using a Insipidus; Diabetes mellitus; Type 1
    [3-H]-glucose uptake diabetes; Type 2 diabetes; Insulin
    assay. (J Biol Chem 1999 resistance; Insulin deficiency;
    Oct. 22; 274(43): 30864- Hyperlipidemia; Hyperketonemia;
    30873). Non- insulin dependent Diabetes
    Mellitus (NIDDM); Insulin-
    dependent Diabetes Mellitus
    (IDDM); A Condition Associated
    With Diabetes Including, But Not
    Limited To Obesity, Heart Disease,
    Hyperglycemia, Infections,
    Retinopathy, And/Or Ulcers;
    Metabolic Disorders; Immune
    Disorders; Obesity; Vascular
    Disorders; Suppression of Body
    Weight; Suppression of Appetite;
    Syndrome X; Endocrine disorders.
    Nagrestipen (CAS- Inflammation; Immune
    166089- 33-4)
    Pentigetide (Pentyde) Respiratory; Allergy; Immune
    (CAS-62087- 72-3)
    Proinsulin (CAS-67422- Stimulates glucose Insulin activity may be Hyperglycemia; Diabetes; Diabetes
    14-4) uptake and promotes assayed in vitro using a Insipidus; Diabetes mellitus; Type 1
    glycogenesis and [3-H]-glucose uptake diabetes; Type 2 diabetes; Insulin
    lipogenesis. assay. (J Biol Chem 1999 resistance; Insulin deficiency;
    Oct. 22; 274(43): 30864- Hyperlipidemia; Hyperketonemia;
    30873). Non- insulin dependent Diabetes
    Mellitus (NIDDM); Insulin-
    dependent Diabetes Mellitus
    (IDDM); A Condition Associated
    With Diabetes Including, But Not
    Limited To Obesity, Heart Disease,
    Hyperglycemia, Infections,
    Retinopathy, And/Or Ulcers;
    Metabolic Disorders; Immune
    Disorders; Obesity; Vascular
    Disorders; Suppression of Body
    Weight; Suppression of Appetite;
    Syndrome X.
    Becaplermin (Regranex; Promotes wound Becaplermin activity can Stimulate Epithelial Cell
    recombinant PDGF-BB) healing. be assessed using animal Proliferation; Stimulate Basal
    (CAS-165101- 51-9) wound healing models Keratinocytes; Promote Wound
    known in the art. (Saba et Healing; Stimulate Hair Follicle
    al., Ann Plast Surg 2002 Production; Healing Of Dermal
    July; 49(1): 62-6). Wounds. Wound Healing; Eye Tissue
    Wounds, Dental Tissue Wounds,
    Oral Cavity Wounds, Diabetic
    Ulcers, Dermal Ulcers, Cubitus
    Ulcers, Arterial Ulcers, Venous
    Stasis Ulcers, Burns Resulting From
    Heat Exposure Or Chemicals, or
    Other Abnormal Wound Healing
    Conditions such as Uremia,
    Malnutrition, Vitamin Deficiencies or
    Complications Associated With
    Systemic Treatment With Steroids,
    Radiation Therapy or Antineoplastic
    Drugs or Antimetabolites; Promote
    Dermal Reestablishment Subsequent
    To Dermal Loss; Increase the
    Adherence Of Skin Grafts To A
    Wound Bed; Stimulate Re-
    Epithelialization from The Wound
    Bed; To Promote Skin Strength;
    Improve The Appearance Of Aged
    Skin; Proliferate Hepatocytes, Lung,
    Breast, Pancreas, Stomach, Bladder,
    Small Intestine, Large Intestine;
    Sebocytes, Hair Follicles, Type II
    Pneumocytes, Mucin-Producing
    Goblet Cells, or Other Epithelial
    Cells, Endothelial Cells,
    Keratinocytes, or Basal Keratinocytes
    (and Their Progenitors) Contained
    Within The Skin, Lung, Liver,
    Bladder, Eye, Salivary Glands, or
    Gastrointestinal Tract; Reduce The
    Side Effects Of Gut Toxicity That
    Result From Radiation,
    Chemotherapy Treatments Or Viral
    Infections; Cytoprotector, especially
    of the Small Intestine Mucosa or
    Bladder; Mucositis (Mouth Ulcers);
    Regeneration Of Skin; Full and/or
    Partial Thickness Skin Defects,
    including Burns, (e.g., Repopulation
    Of Hair Follicles, Sweat Glands, And
    Sebaceous Glands); Psoriasis;
    Epidermolysis Bullosa; Blisters;
    Gastric and/or Doudenal Ulcers;
    Reduce Scarring; Inflamamatory
    Bowel Diseases; Crohn's Disease;
    Ulcerative Colitis; Gut Toxicity;
    Lung Damage; Repair Of Alveoli
    And/or Brochiolar Epithelium; Acute
    Or Chronic Lung Damage;
    Emphysema, ARDS; Inhalation
    Injuries; Hyaline Membrane
    Diseases; Infant Respiratory Distress
    Syndrome; Bronchopulmonary
    Displasia In Premature Infants;
    Fulminant Liver Failure; Cirrhosis,
    Liver Damage caused by Viral
    Hepatitis and/or Toxic Substances;
    Diabetes Mellitus; Inflammation;
    Cancer; Digestive disorders.
    Ghrelin Stimulates release of Appetite and food Endocrine; loss of body weight; loss of
    (Genbank growth hormone intake can be can be body weight associated with cancer or
    Accession No. from anterior measured by methods anorexia nervosa; loss of appetite;
    AB029434) pituitary. Stimulates known in the art excessive appetite; body weight gain;
    appetite and reduces (Batterham et al. Obesity; Diabetes; Acromegaly;
    fat burning. Nature 2002; Growth
    418: 650654) failure; Growth hormone deficiency;
    Growth failure and growth
    retardation
    Prader-Willi syndrome in children 2
    years or older; Growth deficiencies;
    Growth failure associated with
    chronic
    renal insufficiency; Postmenopausal
    osteoporosis; burns; cachexia; cancer
    cachexia; dwarfism; metabolic
    disorders;
    obesity; renal failure; Turner's
    Syndrome, pediatric and adult;
    fibromyalgia; fracture treatment;
    frailty, AIDS wasting
    Ghrelin - binding Inhibits growth Appetite and food intake Endocrine; Obesity; Diabetes; body
    antibody including hormone release in can be can be measured weight gain; excessive appetite; loss
    antibody fragment, or response to Ghrelin; by methods known in the of appetite; loss of body weight.
    dominant- negative form inhibits increase in art (Batterham et al.
    of Ghrelin appetite. Nature 2002; 418:
    650654)
    receptor NOGO-66 Neurodegenerative disorders; spinal
    peptide fragment cord injury; neuronal injury; brain
    (Genbank Accession trauma; stroke; multiple sclerosis;
    No. NP_008939 (amino demyelinating disorders; neural
    acids 62-101)) activity and neurological diseases;
    neural cell (e.g., neuron, glial cell,
    and schwann cell) regeneration
    and/or growth
    Gastric inhibitory Increases nutrient Nutrient uptake and Most Preferred: loss of body weight,
    polypeptide (GIP), uptake and tryglyceride tryglyceride AIDS wasting, cachexia, and loss of
    including GIP fragments accumulation in accumulation can be appetite. Other: Obesity; Diabetes;
    (Genbank Accession adipocytes, which leads measured by methods insulin resistance; body weight gain;
    No. NM_004123) to obesity and insulin described in Miyawaki et excessive appetite.
    resistance. al., Nat. Medicine, 2002,
    Vol 8(7): 738-742,
    Gastric inhibitory Increased use of fat as Fat utilization as an Obesity; Diabetes; Insulin resistance;
    polypeptide antibody, or predominant energy energy source can be body weight gain.
    antibody fragments source; decreased measured as described in
    accumulation of fat in Miyawaki et al., Nat.
    adipocytes. Medicine, 2002, Vol
    8(7): 738-742,
    Gastric inhibitory Increased use of fat as Fat utilization as an Most preferred: Obesity; Diabetes;
    peptide receptor or predominant energy energy source can be body weight gain; excessive appetite;
    receptor fragments or source; decreased measured as described in insulin resistance. Other: loss of body
    variants including accumulation of fat in Miyawaki et al., Nat. weight, AIDS wasting, loss of
    soluble fragments or adipocytes. Medicine, 2002, Vol appetite.
    variants (Genbank 8(7): 738-742.
    Accession Number
    NM_000164)
    POMC Activity of POMC- Preferred: resistance to stress; anti-
    (proopiomelanocortin), derived fragments are inflammatory activity; analgesic
    including fragments or diverse, and well- activity; increased skin pigmentation;
    variants (such as, for known in the art. See, increased protein catabolism;
    example, alpha- for example, Hadley et increased gluconeogenesis; obesity;
    melanocyte stimulating al., Ann NY Acad Sci diabetes. Other: decreased protein
    hormone, αMSH, 1999 Oct. 20; 885: 1-21; catabolism, decreased skin
    gamma melanocyte Dores, Prog Clin Biol pigmentation, Addison's disease,
    stimulating hormone, Res 1990; 342: 22-7; Cushing's syndrome
    γMSH, beta- melanocyte Blalock, Ann NY Acad
    stimulating hormone, Sci. 1999 Oct. 20; 885:
    βMSH, 161-72).
    adrenocorticotropin,
    ACTH, beta- endorphin,
    met- enkephalin)
    (Genbank Accession
    No. NM_000930)
    HP 467, HP228 (U.S. See U.S. Pat. No. See U.S. Pat. No. Resistance to stress; anti-
    Pat. No. 6,350,430) 6,350,430 6,350,430 inflammatory activity; analgesic
    activity; increased skin pigmentation;
    increased protein catabolism;
    increased gluconeogenesis.
    NDP (U.S. Pat. No. See U.S. Pat. No. See U.S. Pat. No. Resistance to stress; anti-
    6,350,430) 6,350,430 6,350,430 inflammatory activity; analgesic
    activity; increased skin pigmentation;
    increased protein catabolism;
    increased gluconeogenesis.
    Interleukin-21 (IL-21) Immunomodulator; IL-21 activity can be Autoimmune disorders;
    inhibits interferon assessed by measuring Inflammatory disorders; Treatment of
    gamma production by interferon gamma Psoriasis; Rheumatoid Arthritis;
    Th1 cells, production in Th1 cells. Inflammatory bowel disease.
    (Wurster et al.,: J Exp
    Med 2002 Oct. 7; 196(7):
    969-77)
    Interleukin-4 (IL-4) Immunomodulator; IL-4 activity can be Treatment of Psoriasis; Autoimmune
    promotes the assessed by measuring disorders; Rheumatoid Arthritis;
    differentiation of T cells Th1/Th2 cytokine Inflammatory bowel disease;
    into Th2 phenotype. responses of isolated Inflammatory disorders.
    spleen cells in vitro.
    (Waltz et al., Horm
    Metab Res 2002 October;
    34(10): 561-9).
    Osteoclast Inhibitory Inhibits osteoclast Osteoclast Inhibitory Treatment of Bone Disorders;
    Lectin (OCIL) formation. Lectin activity can be Osteoporosis; Fracture prevention;
    assessed using osteoclast Hypercalcemia; Malignant
    formation assays known hypercalcemia; Paget's disease;
    in the art. (Zhou et al., J Osteopenia, Osteoclastogenesis;
    Biol Chem 2002 Dec. 13; osteolysis; osteomyelitis;
    277(50): 48808-15) osteonecrosis; periodontal bone loss;
    osteoarthritis; rheumatoid arthritis;
    osteopetrosis; periodontal, lytic, or
    metastatic bone disease; osteoclast
    differentiation inhibition; bone
    healing and regeneration.
    PCSK9 Inhibitor Inhibits the interaction Further LDL lowering Treatment of coronary heart disease.
    of PCSK9 with LDL through targeting PCSK9
    Receptor. for coronary artery
    disease. (Cao et al.
    Endocrine, Metabolic &
    Immune Disorders-Drug
    Targets
    2008, 8, 238-243)
  • Functional Activity:
  • “A polypeptide having functional activity” refers to a polypeptide capable of displaying one or more known functional activities associated with the full-length, pro-protein, and/or mature form of a cargo polypeptide. Such functional activities include, but are not limited to, biological activity, antigenicity [ability to bind (or compete with a polypeptide for binding) to an anti-polypeptide antibody], immunogenicity (ability to generate antibody which binds to a specific polypeptide described herein), ability to form multimers with polypeptides described herein, and ability to bind to a receptor or ligand for a polypeptide. In certain embodiments, the functional activity includes the ability to improve the expression and stability of a partner protein.
  • “A polypeptide having biological activity” refers to a polypeptide exhibiting activity similar to, but not necessarily identical to, an activity of a therapeutic protein described herein, including mature forms, as measured in a particular biological assay, with or without dose dependency. In the case where dose dependency does exist, it need not be identical to that of the polypeptide, but rather substantially similar to the dose-dependence in a given activity as compared to the polypeptide described herein (i.e., the candidate polypeptide will exhibit greater activity or not more than about 25-fold less, or not more than about tenfold less activity, or not more than about three-fold less activity relative to a polypeptide described herein, or presented in Table 2).
  • In certain embodiments, a heteromultimer described herein has at least one biological and/or therapeutic activity associated with the cargo molecule when said cargo molecule is not linked to the transporter polypeptide. In certain embodiments, a heteromultimer described herein has at least one biological and/or therapeutic activity associated with the cargo polypeptide when said cargo polypeptide is not linked to the transporter polypeptide. In certain embodiments, a heteromultimeric protein described herein has at least one biological and/or therapeutic activity associated with the cargo polypeptide portion (or fragment or variant thereof) when said cargo polypeptide is not linked to the albumin or alloalbumin based polypeptide.
  • The heteromultimeric proteins described herein can be assayed for functional activity (e.g., biological activity) using or routinely modifying assays known in the art, as well as assays described herein. Additionally, one of skill in the art may routinely assay fragments of a protein corresponding to a cargo protein portion of an albumin or alloalbumin based monomeric polypeptide, for activity using assays referenced in its corresponding row of Table 2 (e.g., in column 3 of Table 2). In certain embodiments, are assay of fragments of an albumin protein corresponding to an albumin protein portion of a heteromultimer, for activity using assays known in the art and/or as described in the Examples section below.
  • For example, in one embodiment where one is assaying for the ability of a heteromultimeric protein described herein to bind or compete with a Cargo polypeptide for binding to an anti-Cargo polypeptide antibody and/or anti-albumin antibody, various immunoassays known in the art can be used, including but not limited to, competitive and non-competitive assay systems using techniques such as radioimmunoassays, ELISA (enzyme linked immunosorbent assay), “sandwich” immunoassays, immunoradiometric assays, gel diffusion precipitation reactions, immunodiffusion assays, in situ immunoassays (using colloidal gold, enzyme or radioisotope labels, for example), western blots, precipitation reactions, agglutination assays (e.g., gel agglutination assays, hemagglutination assays), complement fixation assays, immunofluorescence assays, protein A assays, and immunoelectrophoresis assays, etc. In one embodiment, antibody binding is detected by detecting a label on the primary antibody. In another embodiment, the primary antibody is detected by detecting binding of a secondary antibody or reagent to the primary antibody. In a further embodiment, the secondary antibody is labeled. Many means are known in the art for detecting binding in an immunoassay and are within the scope of the present invention.
  • In certain embodiments, where a binding partner (e.g., a receptor or a ligand) is identified for a cargo molecule comprised by a heteromultimer described herein, binding to that binding partner by a heteromultimer described herein is assayed, e.g., by means well-known in the art, such as, for example, reducing and non-reducing gel chromatography, protein affinity chromatography, and affinity blotting. See generally, Phizicky et al., Microbiol. Rev. 59:94-123 (1995). In another embodiment, the ability of physiological correlates of a heteromultimeric protein to bind to a substrate(s) of polypeptides corresponding to the cargo protein portion of the heteromultimer can be routinely assayed using techniques known in the art.
  • Biological Activities
  • In certain embodiments, heteromultimers described herein, are used in assays to test for one or more biological activities. If a heteromultimer exhibits an activity in a particular assay, it is likely that at least one cargo protein comprised by one or more monomers of the heteromultimer is implicated in the diseases associated with the biological activity. Thus, the heteromultimer is of use in a treatment of the associated disease.
  • In certain embodiments, provided is a method of treating a disease or disorder comprising administering to a patient in which such treatment, prevention or amelioration is desired, a heteromultimer described herein, in an amount effective to treat, prevent or ameliorate the disease or disorder.
  • Provided herein are monomeric albumin or alloalbumin based fusion proteins produced by a cell, wherein said proteins are encoded by polynucleotides, wherein said monomeric proteins comprise at least one cargo protein, and an albumin or alloalbumin derived polypeptide, such that said monomers form heteromultimers in solution. In certain embodiments, when the polynucleotides are used to express the encoded protein from a cell, the cell's natural secretion and processing steps produces a protein that lacks at least one signal sequence. The specific amino acid sequence of the signal sequence is well known in the art.
  • In certain embodiments, heteromultimers described herein are used in the diagnosis, prognosis, prevention and/or treatment of diseases and/or disorders of the endocrine system. In some embodiments, heteromultimers described herein are used in the diagnosis, prognosis, prevention and/or treatment of diseases and/or disorders of the nervous system.
  • In certain embodiments, heteromultimers described herein are used in the diagnosis, prognosis, prevention and/or treatment of diseases and/or disorders of the immune system. In certain embodiments, heteromultimers described herein are used in the diagnosis, prognosis, prevention and/or treatment of diseases and/or disorders of the respiratory system.
  • In certain embodiments, heteromultimers described herein are used in the diagnosis, prognosis, prevention and/or treatment of diseases and/or disorders of the cardiovascular system. In some embodiments, heteromultimers described herein are used in the diagnosis, prognosis, prevention and/or treatment of diseases and/or disorders of the reproductive system.
  • In certain embodiments, heteromultimers described herein are used in the diagnosis, prognosis, prevention and/or treatment of diseases and/or disorders of the digestive system. In certain embodiments, heteromultimer proteins described herein are used in the diagnosis, prognosis, prevention and/or treatment of diseases or disorders relating to the blood.
  • In certain embodiments, heteromultimers described herein are used in the diagnosis and/or prognosis of diseases and/or disorders associated with at least one tissue(s) in which at least one gene of interest is expressed, wherein a heteromultimer described herein comprises a cargo molecule that binds said at least one gene of interest.
  • In some embodiments, heteromultimers described herein and/or polynucleotides encoding the albumin/alloalbumin based monomers that associate to form heteromultimers described herein, are used in the diagnosis, detection and/or treatment of diseases and/or disorders associated with activities that include, but are not limited to, prohormone activation, neurotransmitter activity, cellular signaling, cellular proliferation, cellular differentiation, and cell migration.
  • Therapeutic Uses:
  • In an aspect, heteromultimers described herein are directed to antibody-based therapies which involve administering heteromultimers described comprising cargo polypeptide(s) which is an antibody, a fragment or variant of an antibody, to a patient for treating one or more of the disclosed diseases, disorders, or conditions. Therapeutic compounds described herein include, but are not limited to, heteromultimers described herein, nucleic acids encoding heteromultimers described herein.
  • In a specific embodiment, are antibody-based therapies which involve administering heteromultimers described herein comprising at least a fragment or variant of an antibody to a patient for treating one or more diseases, disorders, or conditions, including but not limited to: neural disorders, immune system disorders, muscular disorders, reproductive disorders, gastrointestinal disorders, pulmonary disorders, cardiovascular disorders, renal disorders, proliferative disorders, and/or cancerous diseases and conditions, and/or as described elsewhere herein.
  • A summary of the ways in which the heteromultimer proteins of the invention comprising at least a fragment or variant of an antibody are used therapeutically includes binding locally or systemically in the body or by direct cytotoxicity of the antibody, e.g. as mediated by complement (CDC) or by effector cells (ADCC). Some of these approaches are described in more detail below. Armed with the teachings provided herein, one of ordinary skill in the art will know how to use the heteromultimers described herein for diagnostic, monitoring or therapeutic purposes without undue experimentation.
  • The heteromultimers described herein, comprising at least a fragment or variant of an antibody may be administered alone or in combination with other types of treatments (e.g., radiation therapy, chemotherapy, hormonal therapy, immunotherapy and anti-tumor agents). Generally, administration of products of a species origin or species reactivity (in the case of antibodies) that is the same species as that of the patient is preferred. Thus, in an embodiment, human antibodies, fragments derivatives, analogs, or nucleic acids, are administered to a human patient for therapy or prophylaxis.
  • Gene Therapy:
  • In a specific embodiment, nucleic acids comprising sequences encoding heteromultimer proteins described herein are administered to treat, inhibit or prevent a disease or disorder associated with aberrant expression and/or activity of a protein, by way of gene therapy. Gene therapy refers to therapy performed by the administration to a subject of an expressed or expressible nucleic acid. In this embodiment of the invention, the nucleic acids produce their encoded protein that mediates a therapeutic effect. Any of the methods for gene therapy available in the art can be used.
  • Demonstration of Therapeutic or Prophylactic Activity:
  • The heteromultimers or pharmaceutical compositions described herein are tested in vitro, and then in vivo for the desired therapeutic or prophylactic activity, prior to use in humans. For example, in vitro assays to demonstrate the therapeutic or prophylactic utility of a compound or pharmaceutical composition include, the effect of a compound on a cell line or a patient tissue sample. The effect of the compound or composition on the cell line and/or tissue sample can be determined utilizing techniques known to those of skill in the art including, but not limited to, rosette formation assays and cell lysis assays. In accordance with the invention, in vitro assays which can be used to determine whether administration of a specific compound is indicated, include in vitro cell culture assays in which a patient tissue sample is grown in culture, and exposed to or otherwise administered a heteromultimer, and the effect of such heteromultimer upon the tissue sample is observed.
  • Therapeutic/Prophylactic Administration and Composition
  • Provided are methods of treatment, inhibition and prophylaxis by administration to a subject of an effective amount of a heteromultimer or pharmaceutical composition described herein. In an embodiment, the heteromultimer is substantially purified (e.g., substantially free from substances that limit its effect or produce undesired side-effects). In certain embodiments, the subject is an animal, including but not limited to animals such as cows, pigs, horses, chickens, cats, dogs, etc., and in certain embodiments, a mammal, and most preferably human.
  • Various delivery systems are known and can be used to administer a heteromultimer formulation described herein, e.g., encapsulation in liposomes, microparticles, microcapsules, recombinant cells capable of expressing the compound, receptor-mediated endocytosis (see, e.g., Wu and Wu, J. Biol. Chem. 262:4429-4432 (1987)), construction of a nucleic acid as part of a retroviral or other vector, etc. Methods of introduction include but are not limited to intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural, and oral routes. The compounds or compositions may be administered by any convenient route, for example by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa, etc.) and may be administered together with other biologically active agents. Administration can be systemic or local. In addition, in certain embodiments, it is desirable to introduce the heteromultimer compositions described herein into the central nervous system by any suitable route, including intraventricular and intrathecal injection; intraventricular injection may be facilitated by an intraventricular catheter, for example, attached to a reservoir, such as an Ommaya reservoir. Pulmonary administration can also be employed, e.g., by use of an inhaler or nebulizer, and formulation with an aerosolizing agent.
  • In a specific embodiment, it is desirable to administer the heteromultimers, or compositions described herein locally to the area in need of treatment; this may be achieved by, for example, and not by way of limitation, local infusion during surgery, topical application, e.g., in conjunction with a wound dressing after surgery, by injection, by means of a catheter, by means of a suppository, or by means of an implant, said implant being of a porous, non-porous, or gelatinous material, including membranes, such as sialastic membranes, or fibers. Preferably, when administering a protein, including an antibody, of the invention, care must be taken to use materials to which the protein does not absorb.
  • In another embodiment, the heteromultimers or composition can be delivered in a vesicle, in particular a liposome (see Langer, Science 249:1527-1533 (1990); Treat et al., in Liposomes in the Therapy of Infectious Disease and Cancer, Lopez-Berestein and Fidler (eds.), Liss, New York, pp. 353-365 (1989); Lopez-Berestein, ibid., pp. 317-327; see generally ibid.)
  • In yet another embodiment, the heteromultimers or composition can be delivered in a controlled release system. In one embodiment, a pump may be used (see Langer, supra; Sefton, CRC Crit. Ref. Biomed. Eng. 14:201 (1987); Buchwald et al., Surgery 88:507 (1980); Saudek et al., N. Engl. J. Med. 321:574 (1989)). In another embodiment, polymeric materials can be used (see Medical Applications of Controlled Release, Langer and Wise (eds.), CRC Pres., Boca Raton, Fla. (1974); Controlled Drug Bioavailability, Drug Product Design and Performance, Smolen and Ball (eds.), Wiley, New York (1984); Ranger and Peppas, J., Macromol. Sci. Rev. Macromol. Chem. 23:61 (1983); see also Levy et al., Science 228:190 (1985); During et al., Ann. Neurol. 25:351 (1989); Howard et al., J. Neurosurg. 71:105 (1989)). In yet another embodiment, a controlled release system can be placed in proximity of the therapeutic target, e.g., the brain, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, in Medical Applications of Controlled Release, supra, vol. 2, pp. 115-138 (1984)).
  • Other controlled release systems are discussed in the review by Langer (Science 249:1527-1533 (1990)).
  • In a specific embodiment comprising a nucleic acid encoding a heteromultimer described herein, the nucleic acid can be administered in vivo to promote expression of its encoded protein, by constructing it as part of an appropriate nucleic acid expression vector and administering it so that it becomes intracellular, e.g., by use of a retroviral vector (see U.S. Pat. No. 4,980,286), or by direct injection, or by use of microparticle bombardment (e.g., a gene gun; Biolistic, Dupont), or coating with lipids or cell-surface receptors or transfecting agents, or by administering it in linkage to a homeobox-like peptide which is known to enter the nucleus (see e.g., Joliot et al., Proc. Natl. Acad. Sci. USA 88:1864-1868 (1991)), etc. Alternatively, a nucleic acid can be introduced intracellularly and incorporated within host cell DNA for expression, by homologous recombination.
  • Also provided herein are pharmaceutical compositions. Such compositions comprise a therapeutically effective amount of a compound, and a pharmaceutically acceptable carrier. In a specific embodiment, the term “pharmaceutically acceptable” means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans. The term “carrier” refers to a diluent, adjuvant, excipient, or vehicle with which the therapeutic is administered. Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Water is a preferred carrier when the pharmaceutical composition is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions. Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like. The composition, if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents. These compositions can take the form of solutions, suspensions, emulsion, tablets, pills, capsules, powders, sustained-release formulations and the like. The composition can be formulated as a suppository, with traditional binders and carriers such as triglycerides. Oral formulation can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc. Examples of suitable pharmaceutical carriers are described in “Remington's Pharmaceutical Sciences” by E. W. Martin. Such compositions will contain a therapeutically effective amount of the compound, preferably in purified form, together with a suitable amount of carrier so as to provide the form for proper administration to the patient. The formulation should suit the mode of administration.
  • In certain embodiments, the composition comprising the heteromultimer is formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous administration to human beings. Typically, compositions for intravenous administration are solutions in sterile isotonic aqueous buffer. Where necessary, the composition may also include a solubilizing agent and a local anesthetic such as lignocaine to ease pain at the site of the injection. Generally, the ingredients are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent. Where the composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline. Where the composition is administered by injection, an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.
  • In certain embodiments, the compositions described herein are formulated as neutral or salt forms. Pharmaceutically acceptable salts include those formed with anions such as those derived from hydrochloric, phosphoric, acetic, oxalic, tartaric acids, etc., and those formed with cations such as those derived from sodium, potassium, ammonium, calcium, ferric hydroxide isopropylamine, triethylamine, 2-ethylamino ethanol, histidine, procaine, etc.
  • The amount of the composition described herein which will be effective in the treatment, inhibition and prevention of a disease or disorder associated with aberrant expression and/or activity of a Therapeutic protein can be determined by standard clinical techniques. In addition, in vitro assays may optionally be employed to help identify optimal dosage ranges. The precise dose to be employed in the formulation will also depend on the route of administration, and the seriousness of the disease or disorder, and should be decided according to the judgment of the practitioner and each patient's circumstances. Effective doses are extrapolated from dose-response curves derived from in vitro or animal model test systems.
  • Methods of Recombinant and Synthetic Production of Heteromultimer Proteins:
  • In certain embodiments are heteromultimers produced as recombinant molecules by secretion from yeast, a microorganism such as a bacterium, or a human or animal cell line. In embodiments, the polypeptides are secreted from the host cells.
  • Embodiments include a cell, such as a yeast cell transformed to express a heteromultimer protein described herein. In addition to the transformed host cells themselves, are provided culture of those cells, preferably a monoclonal (clonally homogeneous) culture, or a culture derived from a monoclonal culture, in a nutrient medium. If the polypeptide is secreted, the medium will contain the polypeptide, with the cells, or without the cells if they have been filtered or centrifuged away. Many expression systems are known and may be used, including bacteria (for example E. coli and Bacillus subtilis), yeasts (for example Saccharomyces cerevisiae, Kluyveromyces lactis and Pichia pastoris, filamentous fungi (for example Aspergillus), plant cells, animal cells and insect cells.
  • A heteromultimer described herein is produced in conventional ways, for example from a coding sequence inserted in the host chromosome or on a free plasmid. The yeasts are transformed with a coding sequence for the desired protein in any of the usual ways, for example electroporation. Methods for transformation of yeast by electroporation are disclosed in Becker & Guarente (1990) Methods Enzymol. 194, 182.
  • Successfully transformed cells, i.e., cells that contain a DNA construct of the present invention, can be identified by well known techniques. For example, cells resulting from the introduction of an expression construct can be grown to produce the desired polypeptide. Cells can be harvested and lysed and their DNA content examined for the presence of the DNA using a method such as that described by Southern (1975) J. Mol. Biol. 98, 503 or Berent et al. (1985) Biotech. 3, 208. Alternatively, the presence of the protein in the supernatant can be detected using antibodies.
  • Useful yeast plasmid vectors include pRS403-406 and pRS413-416 and are generally available from Stratagene Cloning Systems, La Jolla, Calif. 92037, USA. Plasmids pRS403, pRS404, pRS405 and pRS406 are Yeast Integrating plasmids (YIps) and incorporate the yeast selectable markers HIS3, 7RP1, LEU2 and URA3. Plasmids pRS413-416 are Yeast Centromere plasmids (Ycps).
  • A variety of methods have been developed to operably link DNA to vectors via complementary cohesive termini. For instance, complementary homopolymer tracts can be added to the DNA segment to be inserted to the vector DNA. The vector and DNA segment are then joined by hydrogen bonding between the complementary honmopolymeric tails to form recombinant DNA molecules.
  • Synthetic linkers containing one or more restriction sites provide an alternative method of joining the DNA segment to vectors. The DNA segment, generated by endonuclease restriction digestion, is treated with bacteriophage T4 DNA polymerase or E. coli DNA polymerase 1, enzymes that remove protruding, -single-stranded termini with their 3′ 5′-exonucleolytic activities, and fill in recessed 3′-ends with their polymerizing activities.
  • The combination of these activities therefore generates blunt-ended DNA segments. The blunt-ended segments are then incubated with a large molar excess of linker molecules in the presence of an enzyme that is able to catalyze the ligation of blunt-ended DNA molecules, such as bacteriophage T4 DNA ligase. Tus, the products of the reaction are DNA segments carrying polymeric linker sequences at their ends. These DNA segments are then cleaved with the appropriate restriction enzyme and ligated to an expression vector that has been cleaved with an enzyme that produces termini compatible with those of the DNA segment.
  • Synthetic linkers containing a variety of restriction endonuclease sites are commercially available from a number of sources including International Biotechnologies Inc, New Haven, Conn., USA.
  • Exemplary genera of yeast contemplated to be useful in the practice of the present invention as hosts for expressing the albumin, fusion proteins are Pichua (formerly classified as Hansenula), Saccharomyces, Kluyveromyces, Aspergillus, Candida, Torulopsis, Torulaspora, Schizosaccharomyces, Citeromyces, Pachysolen, Zygosaccharomyces, Debaromyces, Trichoderma, Cephalosporium, Humicola, Mucor, Neurospora, Yarrowia, Metschunikowia, Rhodosporidium, Leucosporidium, Botryoascus, Sporidiobolus, Endomycopsis, and the like. Preferred genera are those selected from the group consisting of Saccharomyces, Schizosaccharomyces, Kluyveromyces, Pichia and Torulaspora. Examples of Saccharomyces spp. are S. cerevisiae, S. italicus and S. rouxii.
  • Examples of Kluyveromyces spp. are K. fragilis, K. lactis and K. marxianus. A suitable Torulaspora species is T. delbrueckii. Examples of Pichia (Hansenula) spp. are P. angusta (formerly H. polymorpha), P. anomala (formerly H. anomala) and P. pastoris. Methods for the transformation of S. cerevisiae are taught generally in EP 251744, EP 258 067 and WO 90/01063, all of which are incorporated herein by reference.
  • Preferred exemplary species of Saccharomyces include S. cerevisiae, S. italicus, S. diastaticus, and Zygosaccharomyces rouxii. Preferred exemplary species of Kluyveromyces include K. fragilis and K. lactis. Preferred exemplary species of Hansenula include H. polymorpha (now Pichia angusta), H. anomala (now Pichia anomala), and Pichia capsulata. Additional preferred exemplary species of Pichia include P. pastoris. Preferred exemplary species of Aspergillus include A. niger and A. nidulans. Preferred exemplary species of Yarrowia include Y. lipolytica. Many preferred yeast species are available from the ATCC. For example, the following preferred yeast species are available from the ATCC and are useful in the expression of albumin fusion proteins: Saccharomyces cerevisiae, Hansen, teleomorph strain BY4743 yap3 mutant (ATCC Accession No. 4022731); Saccharomyces cerevisiae Hansen, teleomorph strain BY4743 hsp150 mutant (ATCC Accession No. 4021266); Saccharomyces cerevisiae Hansen, teleomorph strain BY4743 pmt1 mutant (ATCC Accession No. 4023792); Saccharomyces cerevisiae Hansen, teleomorph (ATCC Accession Nos. 20626; 44773; 44774; and 62995); Saccharomyces diastaticus Andrews et Gilliland ex van der Walt, teleomorph (ATCC Accession No. 62987); Kluyveromyces lactis (Dombrowski) van der Walt, teleomorph (ATCC Accession No. 76492); Pichia angusta (Teunisson et al.) Kurtzman, teleomorph deposited as Hansenula polymorpha de Morais et Maia, teleomorph (ATCC Accession No. 26012); Aspergillus niger van Tieghem, anamorph (ATCC Accession No. 9029); Aspergillus niger van Tieghem, anamorph (ATCC Accession No. 16404); Aspergillus nidulans (Eidam) Winter, anamorph (ATCC Accession No. 48756); and Yarrowia lipolytica (Wickerham et al.) van der Walt et von Arx, teleomorph (ATCC Accession No. 201847).
  • Suitable promoters for S. cerevisiae include those associated with the PGKI gene, GAL1 or GAL10 genes, CYCI, PH05, TRP1, ADH1, ADH2, the genes for glyceraldehyde-3-phosphate dehydrogenase, hexokinase, pyruvate decarboxylase, phosphofructokinase, triose phosphate isomerase, phosphoglucose isomerase, glucokinase, alpha-mating factor pheromone, [a mating factor pheromone], the PRBI promoter, the GUT2 promoter, the GPDI promoter, and hybrid promoters involving hybrids of parts of 5′ regulatory regions with parts of 5′ regulatory regions of other promoters or with upstream activation sites (e.g. the promoter of EP-A-258 067).
  • Convenient regulatable promoters for use in Schizosaccharomyces pombe are the thiamine-repressible promoter from the nmt gene as described by Maundrell (1990) J. Biol. Chem. 265, 10857-10864 and the glucose repressible jbpl gene promoter as described by Hoffman & Winston (1990) Genetics 124, 807-816.
  • Methods of transforming Pichia for expression of foreign genes are taught in, for example, Cregg et al. (1993), and various Phillips patents (e.g. U.S. Pat. No. 4,857,467, incorporated herein by reference), and Pichia expression kits are commercially available from Invitrogen BV, Leek, Netherlands, and Invitrogen Corp., San Diego, Calif. Suitable promoters include AOX1 and AOX2. Gleeson et al. (1986) J. Gen. Microbiol. 132, 3459-3465 include information on Hansenula vectors and transformation, suitable promoters being MOX1 and FMD1; whilst EP 361991, Fleer et al. (1991) and other publications from Rhone-Poulenc Rorer teach how to express foreign proteins in Kluyveromyces spp., a suitable promoter being PGKI.
  • The transcription termination signal is preferably the 3′ flanking sequence of a eukaryotic gene which contains proper signals for transcription termination and polyadenylation. Suitable 3′ flanking sequences may, for example, be those of the gene naturally linked to the expression control sequence used, i.e. may correspond to the promoter. Alternatively, they may be different in which case the termination signal of the S. cerevisiae ADHI gene is preferred.
  • In certain embodiments, the desired heteromultimer protein is initially expressed with a secretion leader sequence, which may be any leader effective in the yeast chosen. Leaders useful in S. cerevisiae include that from the mating factor alpha polypeptide (MFα-1) and the hybrid leaders of EP-A-387 319. Such leaders (or signals) are cleaved by the yeast before the mature albumin is released into the surrounding medium. Further such leaders include those of S. cerevisiae invertase (SUC2) disclosed in JP 62-096086 (granted as 911036516), acid phosphatase (PH05), the pre-sequence of MFα-1, 0 glucanase (BGL2) and killer toxin; S. diastaticus glucoamylase Il; S. carlsbergensis α-galactosidase (MEL1); K. lactis killer toxin; and Candida glucoamylase.
  • Provided are vectors containing a polynucleotide encoding a heteromultimer protein described herein, host cells, and the production of the heteromultimer proteins by synthetic and recombinant techniques. The vector may be, for example, a phage, plasmid, viral, or retroviral vector. Retroviral vectors may be replication competent or replication defective. In the latter case, viral propagation generally will occur only in complementing host cells.
  • In certain embodiments, the polynucleotides encoding heteromultimer proteins described herein are joined to a vector containing a selectable marker for propagation in a host. Generally, a plasmid vector is introduced in a precipitate, such as a calcium phosphate precipitate, or in a complex with a charged lipid. If the vector is a virus, it may be packaged in vitro using an appropriate packaging cell line and then transduced into host cells.
  • In certain embodiments, the polynucleotide insert is operatively linked to an appropriate promoter, such as the phage lambda PL promoter, the E. coli lac, trp, phoA and rac promoters, the SV40 early and late promoters and promoters of retroviral LTRs, to name a few. Other suitable promoters will be known to the skilled artisan. The expression constructs will further contain sites for transcription initiation, termination, and, in the transcribed region, a ribosome binding site for translation. The coding portion of the transcripts expressed by the constructs will preferably include a translation initiating codon at the beginning and a termination codon (UAA, UGA or UAG) appropriately positioned at the end of the polypeptide to be translated.
  • As indicated, the expression vectors will preferably include at least one selectable marker. Such markers include dihydrofolate reductase, G418, glutamine synthase, or neomycin resistance for eukaryotic cell culture, and tetracycline, kanamycin or ampicillin resistance genes for culturing in E. coli and other bacteria. Representative examples of appropriate hosts include, but are not limited to, bacterial cells, such as E. coli, Streptomyces and Salmonella typhimurium cells; fungal cells, such as yeast cells (e.g., Saccharomyces cerevisiae or Pichia pastoris (ATCC Accession No. 201178)); insect cells such as Drosophila S2 and Spodoptera Sf cells; animal cells such as CHO, COS, NSO, 293, and Bowes melanoma cells; and plant cells. Appropriate culture mediums and conditions for the above-described host cells are known in the art.
  • Among vectors preferred for use in bacteria include pQE70, pQE60 and pQE-9, available from QIAGEN, Inc.; pBluescript vectors, Phagescript vectors, pNH8A, pNH16a, pNH18A; pNH46A, available from Stratagene Cloning Systems, Inc.; and ptrc99a, pKK223-3, pKK233-3, pDR540, pRIT5 available from Pharmacia Biotech, Inc. Among preferred eukaryotic vectors are pWLNEO, pSV2CAT, pOG44, pXTI and pSG available from Stratagene; and pSVK3, pBPV, pMSG and pSVL available from Pharmacia. Preferred expression vectors for use in yeast systems include, but are not limited to pYES2, pYDI, pTEFI/Zeo, pYES2/GS, pPICZ, pGAPZ, pGAPZalph, pPIC9, pPIC3.5, pHIL-D2, pHIL-S1, pPIC3.5K, pPIC9K, and PA0815 (all available from Invitrogen, Carlbad, Calif.). Other suitable vectors will be readily apparent to the skilled artisan.
  • In one embodiment, polynucleotides encoding a heteromultimer protein described herein are fused to signal sequences that will direct the localization of a protein of the invention to particular compartments of a prokaryotic or eukaryotic cell and/or direct the secretion of a protein of the invention from a prokaryotic or eukaryotic cell. For example, in E. coli, one may wish to direct the expression of the protein to the periplasmic space. Examples of signal sequences or proteins (or fragments thereof) to which the heteromultimeric proteins are fused in order to direct the expression of the polypeptide to the periplasmic space of bacteria include, but are not limited to, the pelB signal sequence, the maltose binding protein (MBP) signal sequence, MBP, the ompA signal sequence, the signal sequence of the periplasmic E. coli heat-labile enterotoxin B-subunit, and the signal sequence of alkaline phosphatase. Several vectors are commercially available for the construction of fusion proteins which will direct the localization of a protein, such as the pMAL series of vectors (particularly the pMAL-.rho. series) available from New England Biolabs. In a specific embodiment, polynucleotides albumin fusion proteins of the invention may be fused to the pelB pectate lyase signal sequence to increase the efficiency of expression and purification of such polypeptides in Gram-negative bacteria. See, U.S. Pat. Nos. 5,576,195 and 5,846,818, the contents of which are herein incorporated by reference in their entireties.
  • Examples of signal peptides that are fused to a heteromultimeric protein in order to direct its secretion in mammalian cells include, but are not limited to, the MPIF-1 signal sequence (e.g., amino acids 1-21 of GenBank Accession number AAB51134), the stanniocalcin signal sequence (MLQNSAVLLLLVISASA), and a consensus signal sequence (MPTWAWWLFLVLLLALWAPARG). A suitable signal sequence that may be used in conjunction with baculoviral expression systems is the gp67 signal sequence (e.g., amino acids 1-19 of GenBank Accession Number AAA72759).
  • Vectors which use glutamine synthase (GS) or DHFR as the selectable markers can be amplified in the presence of the drugs methionine sulphoximine or methotrexate, respectively. An advantage of glutamine synthase based vectors are the availability of cell lines (e.g., the murine myeloma cell line, NSO) which are glutamine synthase negative. Glutamine synthase expression systems can also function in glutamine synthase expressing cells (e.g., Chinese Hamster Ovary (CHO) cells) by providing additional inhibitor to prevent the functioning of the endogenous gene. A glutamine synthase expression system and components thereof are detailed in PCT publications: WO87/04462; WO86/05807; WO89/10036; WO89/10404; and WO91/06657, which are hereby incorporated in their entireties by reference herein. Additionally, glutamine synthase expression vectors can be obtained from Lonza Biologics, Inc. (Portsmouth, N.H.). Expression and production of monoclonal antibodies using a GS expression system in murine myeloma cells is described in Bebbington et al., Bio/technology 10:169(1992) and in Biblia and Robinson Biotechnol. Prog. 11:1(1995) which are herein incorporated by reference.
  • Also provided are host cells containing vector constructs described herein, and additionally host cells containing nucleotide sequences that are operably associated with one or more heterologous control regions (e.g., promoter and/or enhancer) using techniques known of in the art. The host cell can be a higher eukaryotic cell, such as a mammalian cell (e.g., a human derived cell), or a lower eukaryotic cell, such as a yeast cell, or the host cell can be a prokaryotic cell, such as a bacterial cell. A host strain may be chosen which modulates the expression of the inserted gene sequences, or modifies and processes the gene product in the specific fashion desired. Expression from certain promoters can be elevated in the presence of certain inducers; thus expression of the genetically engineered polypeptide may be controlled. Furthermore, different host cells have characteristics and specific mechanisms for the translational and post-translational processing and modification (e.g., phosphorylation, cleavage) of proteins. Appropriate cell lines can be chosen to ensure the desired modifications and processing of the foreign protein expressed.
  • Introduction of the nucleic acids and nucleic acid constructs of the invention into the host cell can be effected by calcium phosphate transfection, DEAE-dextran mediated transfection, cationic lipid-mediated transfection, electroporation, transduction, infection, or other methods. Such methods are described in many standard laboratory manuals, such as Davis et al., Basic Methods In Molecular Biology (1986). It is specifically contemplated that the polypeptides of the present invention may in fact be expressed by a host cell lacking a recombinant vector.
  • In addition to encompassing host cells containing the vector constructs discussed herein, the invention also encompasses primary, secondary, and immortalized host cells of vertebrate origin, particularly mammalian origin, that have been engineered to delete or replace endogenous genetic material (e.g., the coding sequence corresponding to a Cargo polypeptide is replaced with a heteromultimer protein corresponding to the Cargo polypeptide), and/or to include genetic material. The genetic material operably associated with the endogenous polynucleotide may activate, alter, and/or amplify endogenous polynucleotides.
  • In addition, techniques known in the art may be used to operably associate heterologous polynucleotides (e.g., polynucleotides encoding an albumin protein, or a fragment or variant thereof) and/or heterologous control regions (e.g., promoter and/or enhancer) with endogenous polynucleotide sequences encoding a Therapeutic protein via homologous recombination (see, e.g., U.S. Pat. No. 5,641,670, issued Jun. 24, 1997; International Publication Number WO 96/29411; International Publication Number WO 94/12650; Koller et al., Proc. Natl. Acad. Sci. USA 86:8932-8935 (1989); and Zijlstra et al., Nature 342:435-438 (1989), the disclosures of each of which are incorporated by reference in their entireties).
  • Heteromultimer proteins described herein can be recovered and purified from recombinant cell cultures by well-known methods including ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography, hydrophobic charge interaction chromatography and lectin chromatography. Most preferably, high performance liquid chromatography (“HPLC”) is employed for purification.
  • In certain embodiments the heteromultimer proteins of the invention are purified using Anion Exchange Chromatography including, but not limited to, chromatography on Q-sepharose, DEAE sepharose, poros HQ, poros DEAF, Toyopearl Q, Toyopearl QAE, Toyopearl DEAE, Resource/Source Q and DEAE, Fractogel Q and DEAE columns.
  • In specific embodiments the proteins described herein are purified using Cation Exchange Chromatography including, but not limited to, SP-sepharose, CM sepharose, poros HS, poros CM, Toyopearl SP, Toyopearl CM, Resource/Source S and CM, Fractogel S and CM columns and their equivalents and comparables.
  • In addition, heteromultimer proteins described herein can be chemically synthesized using techniques known in the art (e.g., see Creighton, 1983, Proteins: Structures and Molecular Principles, W. H. Freeman & Co., N.Y and Hunkapiller et al., Nature, 310:105-111 (1984)). For example, a polypeptide corresponding to a fragment of a polypeptide can be synthesized by use of a peptide synthesizer. Furthermore, if desired, nonclassical amino acids or chemical amino acid analogs can be introduced as a substitution or addition into the polypeptide sequence. Non-classical amino acids include, but are not limited to, to the D-isomers of the common amino acids, 2,4diaminobutyric acid, alpha-amino isobutyric acid, 4aminobutyric acid, Abu, 2-amino butyric acid, g-Abu, e-Ahx, 6amino hexanoic acid, Aib, 2-amino isobutyric acid, 3-amino propionic acid, omithine, norleucine, norvaline, hydroxyproline, sarcosine, citrulline, homocitrulline, cysteic acid, t-butylglycine, t-butylalanine, phenylglycine, cyclohexylalanine, β-alanine, fluoro-amino acids, designer amino acids such as β-methyl amino acids, Cα-methyl amino acids, Nα-methyl amino acids, and amino acid analogs in general. Furthermore, the amino acid can be D (dextrorotary) or L (levorotary).
  • Provided are heteromultimers which are differentially modified during or after translation, e.g., by glycosylation, acetylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, linkage to an antibody molecule or other cellular ligand, etc. Any of numerous chemical modifications may be carried out by known techniques, including but not limited, to specific chemical cleavage by cyanogen bromide, trypsin, chymotrypsin, papain, V8 protease, NaBH4; acetylation, formylation, oxidation, reduction; metabolic synthesis in the presence of tunicamycin; etc.
  • Additional post-translational modifications encompassed herein include, for example, e.g., N-linked or O-linked carbohydrate chains, processing of N-terminal or C-terminal ends), attachment of chemical moieties to the amino acid backbone, chemical modifications of N-linked or O-linked carbohydrate chains, and addition or deletion of an N-terminal methionine residue as a result of procaryotic host cell expression. The heteromultimer proteins are modified with a detectable label, such as an enzymatic, fluorescent, isotopic or affinity label to allow for detection and isolation of the protein.
  • Examples of suitable enzymes include horseradish peroxidase, alkaline phosphatase, beta-galactosidase, or acetylcholinesterase; examples of suitable prosthetic group complexes include streptavidin biotin and avidin/biotin; examples of suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; an example of a luminescent material includes luminol; examples of bioluminescent materials include luciferase, luciferin, and aequorin; and examples of suitable radioactive material include iodine, carbon, sulfur, tritium, indium, technetium, thallium, gallium, palladium, molybdenum, xenon, fluorine.
  • In specific embodiments, heteromultimer proteins or fragments or variants thereof are attached to macrocyclic chelators that associate with radiometal ions.
  • As mentioned, the heteromultimer described herein is modified by either natural processes, such as post-translational processing, or by chemical modification techniques which are well known in the art. It will be appreciated that the same type of modification may be present in the same or varying degrees at several sites in a given polypeptide. Polypeptides of the invention may be branched, for example, as a result of ubiquitination, and they may be cyclic, with or without branching. Cyclic, branched, and branched cyclic polypeptides may result from posttranslation natural processes or may be made by synthetic methods. Modifications include acetylation, acylation, ADP-ribosylation, amidation, covalent attachment of flavin, covalent attachment of a heme moiety, covalent attachment of a nucleotide or nucleotide derivative, covalent attachment of a lipid or lipid derivative, covalent attachment of phosphotidylinositol, cross-linking, cyclization, disulfide bond formation, demethylation, formation of covalent cross-links, formation of cysteine, formation of pyroglutamate, formylation, gamma-carboxylation, glycosylation, GPI anchor formation, hydroxylation, iodination, methylation, myristylation, oxidation, pegylation, proteolytic processing, phosphorylation, prenylation, racemization, selenoylation, sulfation, transfer-RNA mediated addition of amino acids to proteins such as arginylation, and ubiquitination. (See, for instance, PROTEINS—STRUCTURE AND MOLECULAR PROPERTIES, 2nd Ed., T. E. Creighton, W. H. Freeman and Company, New York (1993); POST-TRANSLATIONAL COVALENT MODIFICATION OF PROTEINS, B. C. Johnson, Ed., Academic Press, New York, pgs. 1-12 (1983); Seifter et al., Meth. Enzymol. 182:626-646 (1990); Rattan et al., Ann. N.Y. Acad. Sci. 663:48-62 (1992)).
  • In certain embodiments, heteromultimeric proteins may also be attached to solid supports, which are particularly useful for immunoassays or purification of polypeptides that are bound by, that bind to, or associate with albumin fusion proteins of the invention. Such solid supports include, but are not limited to, glass, cellulose, polyacrylamide, nylon, polystyrene, polyvinyl chloride or polypropylene.
  • In embodiments where the heteromultimeric protein comprises only the VH domain of an antibody, it may be necessary and/or desirable to coexpress the protein with the VL domain of the same antibody, such that the VH-albumin fusion protein and VL protein will associate (either covalently or non-covalently) post-translationally.
  • In embodiments where the heteromultimeric protein comprises only the VL domain of an antibody, it may be necessary and/or desirable to coexpress the fusion protein with the VH domain of the same antibody, such that the VL-albumin fusion protein and VH protein will associate (either covalently or non-covalently) post-translationally.
  • Also provided herein are chemically modified derivatives of the heteromultimeric proteins which may provide additional advantages such as increased solubility, stability and circulating time of the polypeptide, or decreased immunogenicity (see U.S. Pat. No. 4,179,337). The chemical moieties for derivitization may be selected from water soluble polymers such as polyethylene glycol, ethylene glycol/propylene glycol copolymers, carboxymethylcellulose, dextran, polyvinyl alcohol and the like. The proteins may be modified at random positions within the molecule, or at predetermined positions within the molecule and may include one, two, three or more attached chemical moieties.
  • The polymer may be of any molecular weight, and may be branched or unbranched. For polyethylene glycol, the preferred molecular weight is between about 1 kDa and about 100 kDa (the term “about” indicating that in preparations of polyethylene glycol, some molecules will weigh more, some less, than the stated molecular weight) for ease in handling and manufacturing. Other sizes may be used, depending on the desired therapeutic profile (e.g., the duration of sustained release desired, the effects, if any on biological activity, the ease in handling, the degree or lack of antigenicity and other known effects of the polyethylene glycol to a Therapeutic protein or analog). For example, the polyethylene glycol may have an average molecular weight of about 200, 500, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 5500, 6000, 6500, 7000, 7500, 8000, 8500, 9000, 9500, 10,000, 10,500, 11,000, 11,500, 12,000, 12,500, 13,000, 13,500, 14,000, 14,500, 15,000, 105,500, 16,000, 16,500, 17,000, 17,500, 18,000, 18,500, 19,000, 19,500, 20,000, 25,000, 30,000, 35,000, 40,000, 45,000, 50,000, 55,000, 60,000, 65,000, 70,000, 75,000, 80,000, 85,000, 90,000, 95,000, or 100,000 kDa.
  • The presence and quantity of heteromultimer proteins described herein may be determined using ELISA, a well known immunoassay known in the art. In one ELISA protocol that would be useful for detecting/quantifying heteromultimers described herein, comprises the steps of coating an ELISA plate with an anti-human serum albumin antibody, blocking the plate to prevent non-specific binding, washing the ELISA plate, adding a solution containing the protein described herein (at one or more different concentrations), adding a secondary anti-cargo polypeptide specific antibody coupled to a detectable label (as described herein or otherwise known in the art), and detecting the presence of the secondary antibody. In an alternate version of this protocol, the ELISA plate might be coated with the anti-cargo polypeptide specific antibody and the labeled secondary reagent might be the anti-human albumin specific antibody.
  • Provided herein are multifunctional heteromultimers that comprise: at least two monomers, wherein at least one monomer comprises at least one cargo molecule attached to a transporter polypeptide, such that said monomers associate to form the heteromultimer; wherein at least one transporter polypeptide is derived from a monomeric protein and wherein said transporter polypeptides self-assemble to form a quasi-native structure of said monomeric protein or analog thereof. In certain embodiments, the cargo molecule is a biomolecule. In specific embodiments is a heteromultimer that comprises: at least two monomeric proteins, wherein each monomeric protein comprises at least one cargo polypeptide, attached to a transporter polypeptide, such that said monomeric proteins self-assemble to form the heteromultimer. In certain embodiments, the heteromultimer is a heterodimer. In an embodiment, the heteromultimer is bispecific. In an embodiment, the heteromultimer is multispecific. In certain embodiments, at least one transporter polypeptide is not derived from an antibody. In certain embodiments, the transporter polypeptides are not derived from an antibody. In an embodiment, the heteromultimer is multifunctional. In certain embodiments, the transporter polypeptides are derivatives of albumin. In certain embodiments of the heteromultimer described herein, the transporter polypeptides are derived from human serum albumin of SEQ ID No. 1. In certain embodiments of the heteromultimer described herein, the transporter polypeptides are derived from alloalbumins. In certain embodiments, the cargo polypeptides are therapeutic proteins described herein, or fragments or variants thereof. In some embodiments, at least one cargo polypeptide is fused to the transporter polypeptide. In certain embodiments, at least one cargo polypeptide is attached to the N-terminus of the transporter polypeptide. In some embodiments, at least one cargo polypeptide is attached to the C-terminus of the transporter polypeptide.
  • Provided herein are heteromultimers, each heteromultimer comprising: at least a first monomer that comprises at least one cargo molecule, and a first transporter polypeptide; and at least a second monomer that comprises at least one cargo molecule and a second transporter polypeptide wherein at least one transporter polypeptide is derived from a monomeric protein and wherein said transporter polypeptides self-assemble to form a quasi-native structure of said monomeric protein or analog thereof. In certain embodiments, at least one cargo molecule is a therapeutic agent described herein. In certain embodiments, at least one cargo molecule is a biomolecule described herein. Provided herein are heteromultimers, each heteromultimer comprising: at least a first monomeric protein that comprises at least one cargo polypeptide and a first transporter polypeptide; and at least a second monomeric protein that comprises at least one cargo polypeptide and a second transporter polypeptide. In certain embodiments, the heteromultimer is a heterodimer. In certain embodiments, the heteromultimer is multivalent. In an embodiment, the heteromultimer is bivalent. In some embodiments, the heteromultimer is multispecific. In an embodiment, the heteromultimer is bispecific. In certain embodiments, the transporter polypeptides are derivatives of albumin. In certain embodiments of the heteromultimer described herein, the transporter polypeptides are derived from human serum albumin of SEQ ID No. 1.
  • In certain embodiments, are heteromultimers, each heteromultimer comprising: at least a first monomeric protein that comprises at least one cargo polypeptide and a first transporter polypeptide comprising a sequence of SEQ ID NO:2; and at least a second monomeric protein that comprises at least one cargo polypeptide and a second transporter polypeptide comprising a sequence of SEQ ID NO: 3. In certain embodiments of the heteromultimer described herein, at least one transporter polypeptide is derived from alloalbumins. In certain embodiments, both transporter polypeptides are derived from alloalbumins. In certain embodiments, all transporter polypeptides are derivatives of the same alloalbumin. In some other embodiments, the transporter polypeptides are derivatives of different alloalbumins. In some embodiments, each transporter polypeptide is an alloalbumin derivative based on an alloalbumin selected from Table 2. In certain embodiments, the first monomeric protein comprises two cargo polypeptides. In some embodiments, the second monomeric protein comprises two cargo polypeptides.
  • In some embodiments of the heteromultimer described herein, the transporter polypeptides are derivatives of an annexin protein. In an embodiment, the transporter polypeptides are derived from different annexin proteins. In certain embodiments, the transporter polypeptides are derived from the same annexin protein. In an embodiment, at least one transporter polypeptide is derived from Annexin A1 or lipocortin I. In certain embodiments of the heteromultimer, all transporter polypeptides are derived from Annexin A1 of SEQ ID NO: 14. In certain embodiments of the heteromultimer, at least one transporter polypeptides is derived from a sequence homologous to SEQ ID NO: 14. In an embodiment, at least one transporter polypeptide is derived from Annexin A2 or annexin II. In certain embodiments of the heteromultimer, all transporter polypeptides are derived from Annexin A2 or lipocortin II. In an embodiment, at least one transporter polypeptide is derived from Annexin like protein. In certain embodiments of the heteromultimer, all transporter polypeptides are derived from Annexin like protein. In an embodiment, at least one transporter polypeptide is derived from the group comprising Annexin A1-Annexin A7. In an embodiment of the heteromultimer described herein, all transporter polypeptides are derived from the group comprising Annexin A1-Annexin A7. 14. In certain embodiments, the first annexin based transporter polypeptide has a sequence comprising SEQ ID NO:15, and the second annexin based transporter polypeptide has a sequence comprising SEQ ID NO: 16.
  • In some embodiments of the heteromultimer described herein, the transporter polypeptides are derivatives of transferrin. In an embodiment, at least one transporter polypeptide is derived from transferrin. In certain embodiments of the heteromultimer, at least one transporter polypeptides are derived from transferrin of SEQ ID NO: 19 or analog thereof. In certain embodiments of the heteromultimer, at least one transporter polypeptide is derived from a polypeptide sequence homologous to the transferrin. In certain embodiments of the heteromultimer described herein, at least one transporter polypeptide is derived from apo-transferrin. In certain embodiments, the first transferrin based transporter polypeptide has a sequence comprising SEQ ID NO:15 and the second transferrin based transporter polypeptide has a sequence comprising SEQ ID NO: 16. Provided herein are heteromultimers, each heteromultimer comprising: at least a first monomeric protein that comprises at least one cargo polypeptide and a first transporter polypeptide; and at least a second monomeric protein that comprises at least one cargo polypeptide and a second transporter polypeptide, wherein said cargo polypeptides are selected from the proteins listed in Table 2, and wherein at least one transporter polypeptide is derived from a monomeric protein and wherein said transporter polypeptides self-assemble to form a quasi-native structure of said monomeric protein or analog thereof. In certain embodiments, are heteromultimers, each heteromultimer comprising: at least a first monomeric protein that comprises at least one cargo polypeptide and a first transporter polypeptide; and at least a second monomeric protein that comprises at least one cargo polypeptide and a second transporter polypeptide, wherein at least one at least one cargo polypeptide is an antibody, or fragment or variant thereof. In certain embodiments, all cargo polypeptides are antibodies or fragments or variants thereof. In certain embodiments, at least one cargo molecule attached to the first transporter polypeptide is the same as at least one cargo molecule attached to the second transporter polypeptide. In certain embodiments, the cargo molecules attached to the first transporter polypeptide are different from the cargo molecule on the second transporter polypeptide. In certain embodiments, there are at least two cargo molecules attached to the first transporter polypeptide and at least two cargo molecule attached to the second transporter polypeptide. In certain embodiments the cargo molecules attached to the first transporter polypeptide are the same. In certain embodiments at least two cargo molecules attached to the first transporter polypeptide are different from each other. In certain embodiments at least two cargo molecules attached to the second transporter polypeptide are the same. In certain embodiments at least two cargo molecules attached to the second transporter polypeptide are different. In some embodiments, the antibody fragment comprises antibody Fc region. In some embodiments, the antibody is an immunoglobulin selected from the group consisting of IgG, IgA, IgD, IgE, and IgM. In certain embodiments, the IgG is of subtype selected from IgG1, IgG2a, IgG2b, IgG3 and IgG4. In certain embodiments, the antibody is a multispecific antibody. In some embodiments, the multispecific antibody is a bispecific antibody.
  • Provided herein are heteromultimers, each heteromultimer comprising: at least a first monomeric protein that comprises at least one cargo polypeptide and a first transporter polypeptide; and at least a second monomeric protein that comprises at least one cargo polypeptide and a second transporter polypeptide, wherein at least one cargo polypeptide is a therapeutic antibody. In some embodiments of the heteromultimers described herein, at least one cargo polypeptide is a therapeutic antibody or fragment or variant thereof, wherein the antibody is selected from antibody is selected from abagovomab, adalimumab, alemtuzumab, aurograb, bapineuzumab, basiliximab, belimumab, bevacizumab, briakinumab, canakinumab, catumaxomab, certolizumab pegol, certuximab, daclizumab, denosumab, efalizumab, galiximab, gemtuzumab ozagamicin, golimumab, ibritumomab tiuxetan, infliximab, ipilimumab, lumiliximab, mepolizumab, motavizumab, muromonab, mycograb, natalizumab, nimotuzumab, ocrelizumab, ofatumumab, omalizumab, palivizumab, panitumumab, pertuzumab, ranizumab, reslizumab, rituximab, teplizumab, toclizumab, tositumomab, trastuzumab, Proxinium, Rencarex, ustekinumab, and zalutumumab. In certain embodiments, the therapeutic antibody binds a cancer antigen.
  • Provided herein are heteromultimers, each heteromultimer comprising: at least a first monomeric protein that comprises at least one cargo polypeptide and a first transporter polypeptide; and at least a second monomeric protein that comprises at least one cargo polypeptide and a second transporter polypeptide, wherein at least one cargo polypeptide is an enzyme, hormone, therapeutic polypeptide, antigen, chemotoxin, radiotoxin, cytokine or variant or fragment thereof.
  • Provided herein are heteromultimers, each heteromultimer comprising: at least a first monomeric protein that comprises at least one cargo polypeptide and a first transporter polypeptide; and at least a second monomeric protein that comprises at least one cargo polypeptide and a second transporter polypeptide, wherein the cargo polypeptide is attached to the transporter polypeptide by chemical conjugation, native ligation, chemical ligation, a disulfide bond or fusion.
  • Provided herein are host cells comprising nucleic acid encoding a heteromultimer described herein. In certain embodiments, the nucleic acid encoding the first monomeric protein and the nucleic acid encoding the second monomeric protein are present in a single vector. In certain embodiments, the nucleic acid encoding the first monomeric protein and the nucleic acid encoding the second monomeric protein are present in separate vectors.
  • Provided herein is a method of making a heteromultimer, wherein said method comprises: culturing a host cell described herein such that the nucleic acid encoding a heteromultimer described herein is expressed; and recovering the heteromultimer from the cell culture. In some embodiments, the host cell is a prokaryotic cell or a eukaryotic cell. In certain embodiments, the host cell is yeast cell. In some embodiments, the yeast is S. cerevisiae. In some embodiments, the yeast is glycosylation deficient, and/or protease deficient. In some embodiments, the host cell is a bacterial cell. In some embodiments, the host cell expressing a heteromultimer descried herein is a mammalian cell. In certain embodiments, the mammalian cell is a CHO cell, a BHK cell, NSO cell, COS cell or a human cell.
  • Provided is a pharmaceutical composition that comprises a heteromultimer described herein and a pharmaceutically acceptable adjuvant. Also provided are methods of treating an individual suffering from a disease or disorder, said method comprising administering to the individual an effective amount of a formulation or pharmaceutical composition described herein. In certain embodiments is a method of treating cancer in a patient, said method comprising administering to the patient a therapeutically effective amount of a heteromultimer described herein. In some embodiments is a method of treating an immune disorder in a patient, said method comprising administering to the patient a therapeutically effective amount of a heteromultimer described herein. Also provided is a method of treating an infectious disease in a patient, said method comprising administering to the patient a therapeutically effective amount of a heteromultimer described herein. In certain embodiments is a method of treating a cardiovascular disorder in a patient, said method comprising administering to the patient a therapeutically effective amount of a heteromultimer described herein. In certain embodiments is a method of treating a respiratory disorder in a patient, said method comprising administering to the patient a therapeutically effective amount of a heteromultimer described herein.
  • Provided is a kit for detecting the presence of a biomarker of interest in an individual, said kit comprising (a) an amount of a heteromultimer described herein, wherein said heteromultimer comprises at least one cargo polypeptide such that said cargo polypeptide is capable of binding to the biomarker of interest; and (b) instructions for use.
  • Provided herein are heteromultimer proteins that comprise at least two monomeric proteins, wherein each monomeric protein comprises at least one cargo polypeptide, and an albumin based polypeptide, such that said monomeric proteins self-assemble to form the heteromultimer.
  • In certain embodiments, the cargo polypeptide is fused to the albumin or alloalbumin based polypeptide. In some embodiments, the cargo polypeptide is chemically conjugated to the albumin or alloalbumin based polypeptide. In certain embodiments, the cargo polypeptide is attached to the albumin or alloalbumin based polypeptide by means of chemical ligation or a disulfide bond.
  • Provided herein are heteromultimer proteins that comprise at least two monomeric proteins, wherein each monomeric protein comprises at least one cargo polypeptide, and an alloalbumin based polypeptide, such that said alloalbumin based polypeptides self-assemble to form the heteromultimer with a quasi-native structure of said alloalbumin or analog thereof. In some embodiments, a heteromultimer described herein is a heterodimer. In some embodiments cargo polypeptide is an antibody, enzyme, hormone, therapeutic polypeptide, antigen, chemotoxin, radiotoxin, cytokine or variant or fragment thereof. In some embodiments, the cargo polypeptide of one monomeric protein functions in synergy with the cargo polypeptide of another monomeric protein.
  • In an aspect described herein is a method to derive protein segments from a protein of interest that can efficiently fold and selectively associate together to form an active quasi-native protein like structure.
  • Provided herein is a strategy for creating polypeptides based on a monomeric protein such as but not restricted to human serum albumin (HSA) that yield a quasi-native monomeric protein like structure and function when associated with each other. In embodiments described herein, this strategy is also used to design heteromultimers comprising monomeric polypeptides that comprise transporter polypeptides that are derivatives of HSA variants, alloalbumins other homologous albumin molecules from other species and also Annexin and Transferrin. The monomers described herein can be engineered using a variety of strategies to improve biophysical characteristics such as the stability of the individual transporter polypeptides or their associated complex.
  • In an embodiment is a scaffold for the development of bispecific or other multispecific or multifunctional protein molecules based on fragments derived from HSA.
  • Provided is a transporter polypeptide which is a HAS, HAA, Annexin or Transferrin derived scaffold that can be conjugated or fused with cargo polypeptides such as other functional domains such as antigen binding protein units, target substrates or inhibitors or payloads such as chemotoxins, radiotoxins, cytokines, etc. to achieve a multispecific or multifunctional therapeutic protein.
  • Described herein are fusions of heterodimeric Fc with transporter polypeptides based on HSA to yield bispecific antibody based therapeutics with sufficient purity and stability for pharmaceutical applications.
  • In an aspect, described herein is a method of deriving a multispecific or multifunctional protein comprising self-assembling monomers that comprise transporter polypeptides based on HSA, such that, the protein has a number of favorable pharmacokinetic properties including improved half-life, improved stability, low immunogenicity, etc.
  • Provided herein are heterodimer proteins that comprise at least two monomeric fusion proteins, wherein each monomeric fusion proteins comprises at least one cargo polypeptide fused to an albumin derived polypeptide, such that said albumin derived polypeptides self-assemble to form the multifunctional heterodimer with a quasi-native structure of albumin or an analog thereof.
  • In certain embodiments are heterodimer proteins that comprise at least two monomeric fusion proteins, wherein each monomeric fusion proteins comprises at least one cargo polypeptide fused to an alloalbumin derived polypeptide, such that said alloalbumin derived polypeptides self-assemble to form the multifunctional heterodimer.
  • In certain embodiments described herein are heteromultimer proteins that comprise at least two monomeric fusion proteins, wherein each monomeric fusion proteins comprises at least one cargo polypeptide fused to an alloalbumin derived polypeptide, such that said alloalbumin derived polypeptides self-assemble to form the multifunctional heterodimer. In certain embodiments are heterodimeric proteins comprising a first monomer which comprises at least one cargo polypeptide fused to an alloalbumin derived polypeptide; and a second monomer that comprises at least one cargo polypeptide fused to an alloalbumin derived polypeptide. In certain embodiments, the at least one cargo polypeptide of the first monomer is different from the at least one cargo polypeptide of the second monomer.
  • Provided herein is a heteromultimer that comprises: at least two monomers, each comprising a transporter polypeptide and optionally at least one cargo molecule attached to said transporter polypeptide, wherein each transporter polypeptide is obtained by segmentation of a whole protein such that said transporter polypeptides self-assemble to form quasi-native whole protein. In certain embodiments, the heteromultimer is multispecific. In certain embodiments, the transporter polypeptides are not derived from an antibody. In some embodiments, each monomer preferentially forms the heteromultimer as compared to a monomer or a homomultimer. In an embodiment of the heteromultimer, at least one cargo molecule is a therapeutic agent, or a biomolecule. In some embodiments, at least one cargo molecule is a biomolecule which is selected from a polypeptide, DNA, PNA, or RNA. In some embodiments, each transporter polypeptide is a derivate of albumin or alloalbumin. In an embodiment, each transporter polypeptide is a derivate of annexin. In certain embodiments, each transporter polypeptide is a derivate of transferrin.
  • In certain embodiments are pharmaceutical formulations that comprise an albumin-based and/or alloalbumin-based heteromultimeric protein described herein and a pharmaceutically acceptable diluent or carrier. In certain embodiments, a formulation described herein is provided as part of a kit or container. In certain embodiments, the kit or container is packaged with instructions pertaining to extended shelf life of the therapeutic protein. In some embodiments, a heteromultimer described herein is used in a method of treating (e.g., ameliorating) preventing, or diagnosing a disease or disease symptom in an individual, comprising the step of administering said formulation to the individual.
  • Also provided are transgenic organisms modified to contain nucleic acid molecules described herein to encode and express monomeric fusion proteins described herein.
  • Various publications are cited herein, the disclosures of which are incorporated by reference in their entireties.
  • EXAMPLES Example 1: The Protein Splitting Method
  • Specific protein-protein association is driven by strong surface complementarity between interacting partners and the accompanying structural and thermodynamic changes. The surface complementarity provides an opportunity to form contacts that support the creation of favorable electrostatic and hydrophobic interactions. Electrostatic interactions involve the formation of salt bridges, hydrogen bonds and the pervasive dispersion interactions. Solvent exclusion and reorganization around non-polar atomic groups at the interface and its associated entropic effects play a role in the hydrophobic component of the binding thermodynamics. Residues with geometries that are optimized for hydrophobic interaction with one another will form contacts (i.e. stacking, pi-pi, cation-pi contacts favorable for stabilizing a protein-protein interface. Similar thermodynamic effects control multi-step protein folding processes that involve the pre-organization of secondary structural units and tertiary domains, which is followed by their association to form the folded quaternary state of the protein. An alternate mechanism to protein folding and binding involves a coupled protein folding and binding process that ultimately results in the quaternary state of the protein. In the context of protein association, the individual protein components need to be co-expressed or be present in the same medium and each of the components or monomers will stably fold into its final structural state only on association with its obligate partner. (FIG. 6)
  • Generation of a split protein involves recognizing a segmentation site in the native protein, using information from sequence, secondary structure and fold that will yield at least two transporter polypeptides that efficiently form the quasi-native protein structure by self-assembling to form a heteromultimer together. For example, these split protein transporter polypeptides selectively self-assemble and form the quasi-native state when co-expressed. While generating a split protein complementary pair of transporter polypeptides, in a way, the attempt is to emulate a number of naturally occurring obligate protein-protein complexes that exhibit their functionality as a complex while being non-functional in their uncomplexed state. A successful implementation of the strategy results in polypeptides that selectively self-assemble to form heteromultimers with each other, are soluble as individual entities and for functional relevance, do not impair the folding, binding and activity of other components in the environment. The intrinsic nature of the polypeptides to reconstitute with each other has applications in area of creating heteromultimeric fusion entities out of cargo molecules that are not efficient at forming multimers by themselves. The functional role of the split protein segments is to act as transporter polypeptides that drive heteromultimerization.
  • Example 2:Preparation of HA/Alloalbumin Based Heteromultimer Proteins
  • Shown is a method to determine the segmentation site along the HSA sequence and structure that will yield monomeric polypeptide chains that stably fold and fuse to form a quasi-native quaternary structure of the original protein. One of the critical requirements for such stable association is the formation of a large buried area of surface complementarity at the interface between the polypeptide chains. The native fold of the original protein provides indication of the natural complementarity of regions within the protein.
  • FIG. 2 shows the solvent accessible surface area buried at the interface of two albumin-based polypeptides that would ideally fold into the quasi-native structure of HSA, when the segmentation point is moved along the protein sequence. The analysis indicates that a large surface area, of the order of about 2000 Å2 is buried when the split segmentation is introduced anywhere between residues 30 and 520 with a few exceptions. Albumin has an exceptionally large number of disulphides bridges that contributes to the stability of the native protein structure. Section of the protein near residues 110, 190, 300, 390 and 500 provide sites for segmentation that do not split the residues involved in a disulphide link across the two transporter polypeptides. Segmentation in other regions would result in heterodimers with a cross linking disulphide bond between the two transporter polypeptide pairs. FIG. 3 presents a model representation of one such quasi-native albumin structure derived by removal of loop from residues 294 to 303 in the HSA sequence. The total buried surface area for the two albumin based polypeptides of SEQ ID No. 2, and SEQ ID No: 3 shown herein is approximately 2500 Å2. This is within the average range of 1910-3880 Å2 observed in a number of protein-protein heterodimeric and homodimeric co-complex structures [Bahadhur R P. & Zacharias M. (2008) Cell Mol Life Sci 65, 1059-1072]. his suggests that there is a strong likelihood for the two polypeptides to selectively associate with each other if the folding pathway of the two polypeptides is fairly independent of each other.
  • In an aspect of this invention, selective formation of a stable quasi-native structure with the two polypeptides (the pair formed by SEQ ID No. 2 and SEQ ID No. 3 or the transporter pair formed by SEQ ID No. 8 and SEQ ID No. 10) gives us the opportunity to employ these polypeptides to drive the formation of bispecific or other multifunctional molecules after fusing the appropriate cargo proteins of interest to the N or C terminus of the albumin based polypeptides employed as transporter polypeptides. A number of other alternate segmentation patterns resulting in transportation polypeptide pair heterodimer can be designed. The fused cargo proteins can be antigen binding domains or other payloads such as chemotoxins, radiotoxins or cytokines (as represented in FIG. 4). The resulting heterodimers have many of the favorable properties intrinsic to HSA including properties like improved half-life, stability and low immunogenicity. Traditional linkers such as (Gly4Ser)x can be used for the association of the cargo protein with the transporter polypeptide.
  • In another aspect of this invention, each of the HSA based transporter polypeptides is fused independently to the C-terminus of two heavy chains in a bispecific Fc molecule (as represented in FIG. 5). The strong and selective pairing of the two transporter polypeptides (such as SEQ ID No. 2, and SEQ ID No. 3) drives the selectively heterodimerization of the Fc and also contribute to its stability and other valuable pharmacokinetic properties.
  • Serum albumin preprotein NP_000468.1 GI 4502027 mRNA sequence from NM_000477.5, Consensus CDS (CCDS) ID 3555.1
  • SEQ ID No. 4:
    Residue 1-29 (EFATMAVMAPRTLVLLLSGALALTQTWAG)

    is the N-terminal export signal sequence region that gets cleaved. his sequence fulfills the same role as the natural signal sequence but it's optimized for mammalian and CHO cell lines.
  • SEQ ID No. 1:
    gi|4502027|ref|NP_000468.1| serum albumin
    preproprotein [Homo sapiens]
    EFATMAVMAPRTLVLLLSGALALTQTWAGDAHKSEVAHRFKDLG
    EENFKALVLIAFAQYLQQCPFEDHVKLVNEVTEFAKTCVADESA
    ENCDKSLHTLFGDKLCTVATLRETYGEMADCCAKQEPERNECFL
    QHKDDNPNLPRLVRPEVDVMCTAFHDNEETFLKKYLYEIARRHP
    YFYAPELLFFAKRYKAAFTECCQAADKAACLLPKLDELRDEGKA
    SSAKQRLKCASLQKFGERAFKAWAVARLSQRFPKAEFAEVSKLV
    TDLTKVHTECCHGDLLECADDRADLAKYICENQDSISSKLKECC
    EKPLLEKSHCIAEVENDEMPADLPSLAADFVESKDVCKNYAEAK
    DVFLGMFLYEYARRHPDYSVVLLLRLAKTYETTLEKCCAAADPH
    ECYAKVFDEFKPLVEEPQNLIKQNCELFEQLGEYKFQNALLVRY
    TKKVPQVSTPTLVEVSRNLGKVGSKCCKHPEAKRMPCAEDYLSV
    VLNQLCVLHEKTPVSDRVTKCCTESLVNRRPCFSALEVDETYVP
    KEFNAETFTFHADICTLSEKERQIKKQTALVELVKHKPKATKEQ
    LKAVMDDFAAFVEKCCKADDKETCFAEEGKKLVAASQAALGL
    SEQ ID No. 5:
    Human serum albumin nucleotide CCDS Sequence 
    (1852 nt)
    GAATTCGCCACTATGGCTGTGATGGCCCCTAGGACCCTGGTGCT
    GCTGCTGTCCGGAGCTCTGGCTCTGACTCAGACCTGGGCTGGAG
    ATGCACACAAGAGTGAGGTTGCTCATCGGTTTAAAGATTTGGGA
    GAAGAAAATTTCAAAGCCTTGGTGTTGATTGCCTTTGCTCAGTA
    TCTTCAGCAGTGTCCATTTGAAGATCATGTAAAATTAGTGAATG
    AAGTAACTGAATTTGCAAAAACATGTGTTGCTGATGAGTCAGCT
    GAAAATTGTGACAAATCACTTCATACCCTTTTTGGAGACAAATT
    ATGCACAGTTGCAACTCTTCGTGAAACCTATGGTGAAATGGCTG
    ACTGCTGTGCAAAACAAGAACCTGAGAGAAATGAATGCTTCTTG
    CAACACAAAGATGACAACCCAAACCTCCCCCGATTGGTGAGACC
    AGAGGTTGATGTGATGTGCACTGCTTTTCATGACAATGAAGAGA
    CATTTTTGAAAAAATACTTATATGAAATTGCCAGAAGACATCCT
    TACTTTTATGCCCCGGAACTCCTTTTCTTTGCTAAAAGGTATAA
    AGCTGCTTTTACAGAATGTTGCCAAGCTGCTGATAAAGCTGCCT
    GCCTGTTGCCAAAGCTCGATGAACTTCGGGATGAAGGGAAGGCT
    TCGTCTGCCAAACAGAGACTCAAGTGTGCCAGTCTCCAAAAATT
    TGGAGAAAGAGCTTTCAAAGCATGGGCAGTAGCTCGCCTGAGCC
    AGAGATTTCCCAAAGCTGAGTTTGCAGAAGTTTCCAAGTTAGTG
    ACAGATCTTACCAAAGTCCACACGGAATGCTGCCATGGAGATCT
    GCTTGAATGTGCTGATGACAGGGCGGACCTTGCCAAGTATATCT
    GTGAAAATCAAGATTCGATCTCCAGTAAACTGAAGGAATGCTGT
    GAAAAACCTCTGTTGGAAAAATCCCACTGCATTGCCGAAGTGGA
    AAATGATGAGATGCCTGCTGACTTGCCTTCATTAGCTGCTGATT
    TTGTTGAAAGTAAGGATGTTTGCAAAAACTATGCTGAGGCAAAG
    GATGTCTTCCTGGGCATGTTTTTGTATGAATATGCAAGAAGGCA
    TCCTGATTACTCTGTCGTGCTGCTGCTGAGACTTGCCAAGACAT
    ATGAAACCACTCTAGAGAAGTGCTGTGCCGCTGCAGATCCTCAT
    GAATGCTATGCCAAAGTGTTCGATGAATTTAAACCTCTTGTGGA
    AGAGCCTCAGAATTTAATCAAACAAAATTGTGAGCTTTTTGAGC
    AGCTTGGAGAGTACAAATTCCAGAATGCGCTATTAGTTCGTTAC
    ACCAAGAAAGTACCCCAAGTGTCAACTCCAACTCTTGTAGAGGT
    CTCAAGAAACCTAGGAAAAGTGGGCAGCAAATGTTGTAAACATC
    CTGAAGCAAAAAGAATGCCCTGTGCAGAAGACTATCTATCCGTG
    GTCCTGAACCAGTTATGTGTGTTGCATGAGAAAACGCCAGTAAG
    TGACAGAGTCACCAAATGCTGCACAGAATCCTTGGTGAACAGGC
    GACCATGCTTTTCAGCTCTGGAAGTCGATGAAACATACGTTCCC
    AAAGAGTTTAATGCTGAAACATTCACCTTCCATGCAGATATATG
    CACACTTTCTGAGAAGGAGAGACAAATCAAGAAACAAACTGCAC
    TTGTTGAGCTCGTGAAACACAAGCCCAAGGCAACAAAAGAGCAA
    CTGAAAGCTGTTATGGATGATTTCGCAGCTTTTGTAGAGAAGTG
    CTGCAAGGCTGACGATAAGGAGACCTGCTTTGCCGAGGAGGGTA
    AAAAACTTGTTGCTGCAAGTCAAGCTGCCTTAGGCTTATGA
  • The protein and nucleotide sequence of albumin based polypeptides useful as transporter polypeptides are as follows:
  • Albumin Based Heteromultimer 1:
  • Albumin based Transporter polypeptide 1-Ver 1:
    SEQ ID No. 2:
    DAHKSEVAHRFKDLGEENFKALVLIAFAQYLQQCPFEDHVKLVNE
    VTEFAKTCVADESAENCDKSLHTLFGDKLCTVATLRETYGEMADC
    CAKQEPERNECFLQHKDDNPNLPRLVRPEVDVMCTAFHDNEETFL
    KKYLYEIARRHPYFYAPELLFFAKRYKAAFTECCQAADKAACLLP
    KLDELRDEGKASSAKQRLKCASLQKFGERAFKAWAVARLSQRFPK
    AEFAEVSKLVTDLTKVHTECCHGDLLECADDRADLAKYICENQDS
    ISSKLKECCEKPLLEKSHCIAEV
    Nucleotide sequence encoding Albumin based
    Transporter polypeptide 1-Ver 1:
    SEQ ID No. 6:
    GATGCACACAAGAGTGAGGTTGCTCATCGGTTTAAAGATTTGGGA
    GAAGAAAATTTCAAAGCCTTGGTGTTGATTGCCTTTGCTCAGTAT
    CTTCAGCAGTGTCCATTTGAAGATCATGTAAAATTAGTGAATGAA
    GTAACTGAATTTGCAAAAACATGTGTTGCTGATGAGTCAGCTGAA
    AATTGTGACAAATCACTTCATACCCTTTTTGGAGACAAATTATGC
    ACAGTTGCAACTCTTCGTGAAACCTATGGTGAAATGGCTGACTGC
    TGTGCAAAACAAGAACCTGAGAGAAATGAATGCTTCTTGCAACAC
    AAAGATGACAACCCAAACCTCCCCCGATTGGTGAGACCAGAGGTT
    GATGTGATGTGCACTGCTTTTCATGACAATGAAGAGACATTTTTG
    AAAAAATACTTATATGAAATTGCCAGAAGACATCCTTACTTTTAT
    GCCCCGGAACTCCTTTTCTTTGCTAAAAGGTATAAAGCTGCTTTT
    ACAGAATGTTGCCAAGCTGCTGATAAAGCTGCCTGCCTGTTGCCA
    AAGCTCGATGAACTTCGGGATGAAGGGAAGGCTTCGTCTGCCAAA
    CAGAGACTCAAGTGTGCCAGTCTCCAAAAATTTGGAGAAAGAGCT
    TTCAAAGCATGGGCAGTAGCTCGCCTGAGCCAGAGATTTCCCAAA
    GCTGAGTTTGCAGAAGTTTCCAAGTTAGTGACAGATCTTACCAAA
    GTCCACACGGAATGCTGCCATGGAGATCTGCTTGAATGTGCTGAT
    GACAGGGCGGACCTTGCCAAGTATATCTGTGAAAATCAAGATTCG
    ATCTCCAGTAAACTGAAGGAATGCTGTGAAAAACCTCTGTTGGAA
    AAATCCCACTGCATTGCCGAAGTGTGA
    Albumin based Transporter polypeptide 2-Ver1:
    SEQ ID No. 3:
    SLAADFVESKDVCKNYAEAKDVFLGMFLYEYARRHPDYSVVLLLR
    LAKTYETTLEKCCAAADPHECYAKVFDEFKPLVEEPQNLIKQNCE
    LFEQLGEYKFQNALLVRYTKKVPQVSTPTLVEVSRNLGKVGSKCC
    KHPEAKRMPCAEDYLSVVLNQLCVLHEKTPVSDRVTKCCTESLVN
    RRPCFSALEVDETYVPKEFNAETFTFHADICTLSEKERQIKKQTA
    LVELVKHKPKATKEQLKAVMDDFAAFVEKCCKADDKETCFAEEGK
    KLVAASQAALGL
    Nucleotide sequence encoding Albumin based
    Transporter polypeptide 2-Ver1:
    SEQ ID No. 7:
    TCATTAGCTGCTGATTTTGTTGAAAGTAATGGATGTTTGCAAAAA
    CTATGCTGAGGCAAAGGATGTCTTCCTGGGCATGTTTTTGTATGA
    ATATGCAAGAAGGCATCCTGATTACTCTGTCGTGCTGCTGCTGAG
    ACTTGCCAAGACATATGAAACCACTCTAGAGAAGTGCTGTGCCGC
    TGCAGATCCTCATGAATGCTATGCCAAAGTGTTCGATGAATTTAA
    ACCTCTTGTGGAAGAGCCTCAGAATTTAATCAAACAAAATTGTGA
    GCTTTTTGAGCAGCTTGGAGAGTACAAATTCCAGAATGCGCTATT
    AGTTCGTTACACCAAGAAAGTACCCCAAGTGTCAACTCCAACTCT
    TGTAGAGGTCTCAAGAAACCTAGGAAAAGTGGGCAGCAAATGTTG
    TAAACATCCTGAAGCAAAAAGAATGCCCTGTGCAGAAGACTATCT
    ATCCGTGGTCCTGAACCAGTTATGTGTGTTGCATGAGAAAACGCC
    AGTAAGTGACAGAGTCACCAAATGCTGCACAGAATCCTTGGTGAA
    CAGGCGACCATGCTTTTCAGCTCTGGAAGTCGATGAAACATACGT
    TCCCAAAGAGTTTAATGCTGAAACATTCACCTTCCATGCAGATAT
    ATGCACACTTTCTGAGAAGGAGAGACAAATCAAGAAACAAACTGC
    ACTTGTTGAGCTCGTGAAACACAAGCCCAAGGCAACAAAAGAGCA
    ACTGAAAGCTGTTATGGATGATTTCGCAGCTTTTGTAGAGAAGTG
    CTGCAAGGCTGACGATAAGGAGACCTGCTTTGCCGAGGAGGGTAA
    AAAACTTGTTGCTGCAAGTCAAGCTGCCTTAGGCTTATGA
  • Albumin Based Heteromultimer 2:
  • Albumin based Transporter polypeptide 1-Ver
    2:
    SEQ ID No. 8:
    DAHKSEVAHRFKDLGEENFKALVLIAFAQYLQQCPFEDHVKLVNEV
    TEFAKTCVADESAENCDKSLHTLFGDKLCTVATLRETYGEMADCCA
    KQEPERNECFLQHKDDNPNLPRLVRPEVDVMCTAFHDNEETFLKKY
    LYEIARRHPYFYAPELLFFAKRYKAAFTECCQAADKAACLLPKLDE
    LRDEGKASSAKQRLKCALLQKFGERAFKAWAVARLSQRFPKAEFAE
    VSKLVTDLTKVHTECCHGDLLECADDRADLAKYICENQDSISSKLK
    ECCEKPLLEKSHCIAEVENDEMPADLPSLAADFVESKDVCKNYAEA
    KDVFLGMFLYEYARA
    Nucleotide sequence encoding Albumin based
    Transporter polypeptide 1-Ver 2:
    SEQ ID No. 9:
    GATGCACACAAGAGTGAGGTTGCTCATCGGTTTAAAGATTTGGGAG
    AAGAAAATTTCAAAGCCTTGGTGTTGATTGCCTTTGCTCAGTATCT
    TCAGCAGTGTCCATTTGAAGATCATGTAAAATTAGTGAATGAAGTA
    ACTGAATTTGCAAAAACATGTGTTGCTGATGAGTCAGCTGAAAATT
    GTGACAAATCACTTCATACCCTTTTTGGAGACAAATTATGCACAGT
    TGCAACTCTTCGTGAAACCTATGGTGAAATGGCTGACTGCTGTGCA
    AAACAAGAACCTGAGAGAAATGAATGCTTCTTCGAACACAAAGATG
    ACAACCCAAACCTCCCCCGATTGGTGAGACCAGAGGTTGATGTGAT
    GTGCACTGCTTTTCATGACAATGAAGAGACATTTTTGAAAAAATAC
    TTATATGAAATTGCCAGAAGACATCCTTACTTTTATGCCCCGGAAC
    TCCTTTTCTTTGCTAAAAGGTATAAAGCTGCTTTTACAGAATGTTG
    CCAAGCTGCTGATAAAGCTGCCTGCCTGTTGCCAAAGCTCGATGAA
    CTTCGGGATGAAGGGAAGGCTTCGTCTGCCAAACAGAGACTCAAGT
    GTGCCAGTCTCCAAAAATTTGGAGAAAGAGCTTTCAAAGCATGGGC
    AGTAGCTCGCCTGAGCCAGAGATTTCCCAAAGCTGAGTTTGCAGAA
    GTTTCCAAGTTAGTGACAGATCTTACCAAAGTCCACACGGAATGCT
    GCCATGGAGATCTGCTTGAATGTGCTGATGACAGGGCGGACCTTGC
    CAAGTATATCTGTGAAAATCAAGATTCGATCTCCAGTAAACTGAAG
    GAATGCTGTGAAAAACCTCTGTTGGAAAAATCCCACTGCATTGCCG
    AAGTGGAAAATGATGAGATGCCTGCTGACTTGCCTTCATTAGCTGC
    TGATTTTGTTGAAAGTAAGGATGTTTGCAAAAACTATGCTGAGGCA
    AAGGATGTCTTCCTGGGCATGTTTTTGTATGAATATGCAAGAGCAT
    GA
    Albumin based Transporter polypeptide 2-Ver 2:
    SEQ ID No. 10:
    SVVLLLRLAKTYETTLEKCCAAADPHECYAKVFDEFKPLVEEPQNL
    IKQNCELFEQLGEYKFQNALLVRYTKKVPQVSTPTLVEVSRNLGKV
    GSKCCKHPEAKRMPCAEDYLSVVLNQLCVLHEKTPVSDRVTKCCTE
    SLVNRRPCFSALEVDETYVPKEFNAETFTFHADICTLSEKERQIKK
    QTALVELVKHKPKATKEQLKAVMDDFAAFVEKCCKADDKETCFAEE
    GKKLVAASQAALGL
    Nucleotide sequence encoding Albumin based
    Transporter polypeptide 2-Ver 2:
    SEQ ID No. 11:
    TCTGTCGTGCTGCTGCTGAGACTTGCCAAGACATATGAAACCACTC
    TAGAGAAGTGCTGTGCCGCTGCAGATCCTCATGAATGCTATGCCAA
    AGTGTTCGATGAATTTAAACCTCTTGTGGAAGAGCCTCAGAATTTA
    ATCAAACAAAATTGTGAGCTTTTTGAGCAGCTTGGAGAGTACAAAT
    TCCAGAATGCGCTATTAGTTCGTTACACCAAGAAAGTACCCCAAGT
    GTCAACTCCAACTCTTGTAGAGGTCTCAAGAAACCTAGGAAAAGTG
    GGCAGCAAATGTTGTAAACATCCTGAAGCAAAAAGAATGCCCTGTG
    CAGAAGACTATCTATCCGTGGTCCTGAACCAGTTATGTGTGTTGCA
    TGAGAAAACGCCAGTAAGTGACAGAGTCACCAAATGCTGCACAGAA
    TCCTTGGTGAACAGGCGACCATGCTTTTCAGCTCTGGAAGTCGATG
    AAACATACGTTCCCAAAGAGTTTAATGCTGAAACATTCACCTTCCA
    TGCAGATATATGCACACTTTCTGAGAAGGAGAGACAAATCAAGAAA
    CAAACTGCACTTGTTGAGCTCGTGAAACACAAGCCCAAGGCAACAA
    AAGAGCAACTGAAAGCTGTTATGGATGATTTCGCAGCTTTTGTAGA
    GAAGTGCTGCAAGGCTGACGATAAGGAGACCTGCTTTGCCGAGGAG
    GGTAAAAAACTTGTTGCTGCAAGTCAAGCTGCCTTAGGCTTATGA
  • Generation and Expression of HA or HAA Based Heteromultimers
  • The genes encoding the full length WT HA and the HA based transporter polypeptide monomers were constructed via gene synthesis using codons optimized for human/mammalian expression. The constructs were designed from known full-length Human Serum Albumin Preprotein (GENEBANK: NP_000468.1), after exclusion of the signal sequence EFATMAVMAPRTLVLLLSGALALTQTWAG. The final gene products were subcloned into the mammalian expression vector pTT5 (NRC-BRI, Canada) (Durocher et al). High level and high-throughput recombinant protein production by transient transfection of suspension-growing human CHO-3E7 was performed. See Table 3 for construct boundaries of the two scaffolds described here: Albumin based heteromultimer 1 (ABH1) and Albumin based heteromultimer 2 (ABH2). Albumin based heteromultimer 2 comprises one disulfide bond between the two transporter polypeptides, while Albumin based heteromultimer 1 is formed entirely by non-covalent interactions. FIG. 6A provides SDS-PAGE (non-reducing) gel analysis of the two heteromultimer constructs (ABH1 and ABH2), after co-expression (different DNA transfection ratios are shown). WT full-length HSA is shown as control. As expected, ABH2 retains the disulfide linkage in non-reducing SDS-PAGE, with a MW roughly double the non-disulfide linked ABH1I. FIG. 6B provides Native gel analysis of the two Albumin based heteromultimer constructs (ABH1 and ABH2), after co-expression (1:1 DNA level). WT full-length HSA is shown as control. ABH1 and ABH2 both form a complex of expected mass, comparable to the full-length WT HSA. Furthermore, upon expression, neither the transporter polypeptides forming ABH1 nor the ones forming ABH2 homodimerize; rather they preferably form a stable hetercomplex. See Table 3 below for details.
  • TABLE 3
    Albumin based heteromultimer constructs
    MW
    Construct Segment Boundaries* (KDa)
    Wild Type HA 1:585 (SEQ ID NO: 1) 64.3
    ABH1 1:293 (SEQ ID NO: 2) 32.2
    304:585 (SEQ ID NO: 3)  30.9
    ABH2 1:337 (SEQ ID NO: 8) 37
    342:585 (SEQ ID NO: 10)  26.7
  • WT-HSA and the two Albumin based heteromultimers (ABH1 and ABH2) were expressed in CHO-3E7 cell line grown in suspension in FreeStyle F17 medium (Invitrogen) supplemented with 0.1% w/v pluronic and 4 mM glutamine. The day of transfection cell density should be around 1.5-2 million cells/ml and viability must be greater than 97%. Transfection is done according to patent application WO 2009/137911 using a mixture of plasmid DNA made of 5% pTro-GFP plasmid (green fluorescent protein to determine transfection efficiency, Table 4), 15% pTT22-AKT plasmid, 21% HSA plasmids (10.63% of each), 68.37% of Salmon Sperm DNA. Following transfection, the shake flask containing cells is then placed on an orbital shaker set to 120 rpm in a humidified incubator with 5% CO2 at 37° C. Twenty-four hours post-transfection, 1% w/v TN1 and 0.5 mM VPA (Valproic acid) are added to the cultures. The cultures are then transferred on an orbital shaker (120 rpm) placed in a humidified incubator with 5% CO2 set at 32° C. At 24-48 hours, GFP positive cells should be between 30-60% as determined by flow cytometry. Cells were harvested 7 days post-transfection and spun at 4,000 rpm for 20 minutes. The supernatant was filter-sterlized (clarified) using a 0.45 μm filter (Millipore). Keep the supernatant at 4° C. for short period storage and at −80° C. for long period storage. Prior to purification, the frozen supernatant was thawed at 37° C., re-filtered and degassed through a 0.45 μm membrane filter under vacuum for 5-10 minutes.
  • TABLE 4
    Cell viability at different stages of
    expression for WT and ABH1 construct.
    HSA % GFP 48 hrs % viability 48 hrs % viability 48 hrs
    scaffold post-transfection post-transfection post-transfection
    Wild Type 67 94.6 72.3
    HSA
    ABH2 66.3 93.6 77.1
  • Purification of HSA and Heteromultimers ABH1 and ABH2
  • Purification was performed by gravity flow using a bench-top QIAGEN-tip 500 column packed with a Blue Sepharose matrix (GE Healthcare). The Blue Sepharose matrix was equilibrated with 20 ml of PBS pH 7.2. The sample was loaded at a flow rate of 5 ml/min and subsequently washed with with 20 ml of PBS. The protein was eluted with 0.1 M Na2HPO4 pH 7.2 supplemented with 1 M NaCl and collected in 1 ml fractions (20 ml total). Fractions containing HSA (as per Bradford protein assay) were pooled, and applied on a HiLoad 16/60 Superdex 200 prep grade gel filtration column coupled to an AKTA Express system (GE Healthcare) using a flow rate of 1 mlml. Protein with a purity of >85% was collected; fractions containing pure sample were pooled and concentrated by centrifugation using an Amicon Ultra membrane with a cutoff weight of 10 000 MWCO. FIG. 6C shows SDS-PAGE (non-reducing) analysis of the ABH2 heteromultimer and WT HSA, both after the final stage of purification. Both constructs show the expected MW.
  • Stability Determination of Albumin Based Heteromultimers Using Differential Scanning Calorimetry (DSC)
  • All DSC experiments were carried out using a GE or MicroCal VP-Capillary instrument. The proteins were buffer-exchanged into PBS (pH 7.4) and diluted to 0.3 to 0.7 mg/mL with 0.137 mL loaded into the sample cell and measured with a scan rate of 1° C./min from 20 to 100° C. Data was analyzed using the Origin software (GE Healthcare) with the PBS buffer background subtracted. See Table 5 and FIG. 7 for resulting melting temperature determined.
  • TABLE 5
    Melting temperature for Albumin based heteromultimers
    Measured Mass Theoretical MW Tm
    Molecule (Da) (Da) ° C.
    HSA Wild 66620 66470 75
    Type
    ABH2 66100 65880 63
  • Evaluation of FcRn Binding of HSA and ABH2 Using Surface Plasmon Resonance
  • As seen in FIGS. 8A-B, when HSA and a HSA-based heteromultimer are immobilized on the SPR surface, affinity towards FcRn appears to be comparable between the full length WT HSA and ABH2, indicating FcRn binding functionality of albumin is retained by the heteromultimer formed by the self-assembly of albumin based transporter polypeptides. The following Table 6 illustrates FcRn binding data. Values in parenthesis refer to standard deviation.
  • TABLE 6
    Kinetic and Equilibrium fit of FcRn Binding of
    HSA and ABH2 using Surface Plasmon Resonance
    Ka (1/Ms) Kd (1/s) KD (M)
    Grouped Fitted Grouped Fitted Grouped Fitted
    HAS 5.3E+04 (7E+03) 7.0E−02 (2.0E−02) 1.4E−06 (6.0E−07) Kinetic fit
    ABH2 5.0E+04 (4E+03) 4.2E−02 (8.0E−03) 8.0E−07 (2.0E−07) Kinetic fit
    HAS 9.0E−07 (1.0E−07) Equilibrium Fit
    ABH2 9.0E−07 (1.0E−07) Equilibrium Fit
  • Example 3 Generation and Expression of Albumin Based Heteromultimers with Mono- and Tetravalency Compnsing Anti-Her2/Neu and Anti-CD16 scFv Bioactive Fusions
  • Multivalent heteromultimer ABH2 was generated by expressing its single monomeric transporter polypeptides, SEQ ID NO: 8 and SEQ ID NO: 10, fused at one or both termini to cargo polypeptides that are either antiHer2scFv (4D5) and/or anti-CD16 scFv (NM3E). These form a set of 8 base construct monomers based off transporter polypeptide 1 and 8 base construct monomers based off transporter polypeptide 2. Different combinations of these base constructs were combined upon co-expression to form heteromultimers displaying all combination of the two cargo polypeptides at any of the four terminal positions of the two transporter polypeptides, ranging from monovalent to tetravalent.
  • As shown in FIG. 9, the bioactive cargo polypeptides were fused to the heteromultimer transporter polypeptides via a GGSG linker, for the N terminus of one monomer and a longer (GGS)4GG linker for all other termini in the other monomer.
  • Single fusions
    # Fusion
    1 Fusion 2
    1 NM3E2 F1
    2 F1 NM3E2
    3 NM3E2 F2
    4 F2 NM3E2
    5 4D5 F1
    6 F1 4D5
    7 4D5 F2
    8 F2 4D5|
    Double fusions
    # Fusion
    1 Fusion 2 Fusion 3
    9 NM3E2 F1 NM3E2
    10 NM3E2 F2 NM3E2
    11 4D5 F1 4D5
    12 4D5 F2 4D5
    13 NM3E2 F1 4D5
    14 4D5 F1 NM3E2
    15 NM3E2 F2 4D5
    16 4D5 F2 NM3E2
  • Multivalent constructs were generated as outlined in Example 2 using heteromultimer ABH2. The final gene products were subcloned into the mammalian expression vector pTT5 (NRC-BRI, Canada) (Durocher et al). High level and high-throughput recombinant protein production by transient transfection of suspension-growing human CHO-3E7 was performed.
  • Purification was performed by application of the cellular supernatant with expressed protein to a QIAGEN-tip 500 column packed with Blue Sepharose matrix (GE Healthcare) coupled to an AKTA Express system (GE Healthcare) using a flow rate of 1 ml/ml. The column was equilibrated with equilibrated with sample buffer composed of 20 ml of PBS pH 7.2, 300 mM NaCl. The sample was loaded at a flow rate of 5 ml/min and subsequently washed with sample buffer. The protein was eluted by application of NaCl gradient ranging from 300 mM to 2000 mM. Fractions eluting in higher salt concentration were the purest and were pooled, concentrated and subsequently applied to a HiLoad 16/60 Superdex 200 prep grade gel filtration column coupled to an AKTA Express system (GE Healthcare) using a flow rate of 1 ml/ml. Protein with a purity of >85% was collected; fractions containing pure sample were pooled and concentrated by centrifugation using an Amicon Ultra membrane with a cutoff weight of 10 000 MWCO. FIGS. 10A-10B shows SDS-PAGE (non-reducing) analysis of the ABH2 heteromultimer fused to different cargo polypeptides. The position of those polypeptides in the heteromultimer relative to the transporter polypeptides is outlined in table 8below. All constructs showed the expected molecular weight.
  • TABLE 8
    Monovalent, multivalent, and multispecific constructs that were generated
    by fusing the 4D5 and NM3 cargo polypeptides to either N or C terminus
    of transporter polypeptide 1 or transporter polypeptide 2 of ABH2.
    N terminus- C terminus- N terminus- C terminus-
    transporter transporter transporter transporter
    polypeptide
    1 polypeptide 1 polypeptide 2 polypeptide 2
    Variant (SEQ ID No: 8) (SEQ ID No: 8) (SEQ ID No: 10) (SEQ ID No: 10) Valency
    513 NM3E monovalent
    514 NM3E monovalent
    515 NM3E monovalent
    516 NM3E monovalent
    517 4D5 monovalent
    518 4D5 monovalent
    519 4D5 monovalent
    520 4D5 monovalent
    521 NM3E NM3E bivalent
    522 NM3E NM3E bivalent
    523 NM3E NM3E bivalent
    524 NM3E NM3E bivalent
    525 4D5 4D5 bivalent
    526 4D5 4D5 bivalent
    527 4D5 4D5 bivalent
    528 4D5 4D5 bivalent
    529 NM3E NM3E bivalent
    530 NM3E NM3E bivalent
    531 4D5 4D5 bivalent
    532 4D5 4D5 bivalent
    543 NM3E 4D5 bispecific
    544 NM3E 4D5 bispecific
    545 NM3E 4D5 bispecific
    546 NM3E 4D5 bispecific
    547 4D5 NM3E bispecific
    548 4D5 NM3E bispecific
    549 4D5 NM3E bispecific
    550 4D5 NM3E bispecific
    551 NM3E 4D5 bispecific
    552 4D5 NM3E bispecific
    553 NM3E 4D5 bispecific
    554 4D5 NM3E bispecific
    593 4D5 NM3E bispecific
    594 NM3E 4D5 bispecific
  • SPR Binding of Monovalent ABH2 Fused to a Single antiCD16scFv
  • Purified heteromultimer ABH2 fused to a single antiCD16scFv to the N terminus of transporter polypeptide SEQ ID 2 (construct v515) was used in a binding experiment using Surface Plasmon Resonance (SPR). Soluble CD16 was covalently immobilized onto a CM5 surface and ABH2 fused to antiCD16scFv was captured and binding kinetics were determined.
  • SPR supplies. GLM sensorchips, the Biorad ProteOn amine coupling kit (EDC, sNHS and ethanolamine), and 10 mM sodium acetate buffers were purchased from Bio-Rad Laboratories (Canada) Ltd. (Mississauga, ON). Recombinant Her-2 protein was purchased from eBioscience (San Diego, Calif.). HEPES buffer, EDTA, and NaCl were purchased from from Sigma-Aldrich (Oakville, ON). 10% Tween 20 solution was purchased from Teknova (Hollister, Calif.).
  • SPR biosensor assays. All surface plasmon resonance assays were carried out using a BioRad ProteOn XPR36 instrument (Bio-Rad Laboratories (Canada) Ltd. (Mississauga, ON)) with HBST running buffer (10 mM HEPES, 150 mM NaCl, 3.4 mM EDTA, and 0.05% Tween 20 pH 7.4) at a temperature of 25° C. The CD16 capture surface was generated using a GLM sensorchip activated by a 1:5 dilution of the standard BioRad sNHS/EDC solutions injected for 300 s at 30 μL/min in the analyte (horizontal) direction. Immediately after the activation, a 4.0 μg/mL solution of CD16 in 10 mM NaOAc pH 4.5 was injected in the ligand (vertical) direction at a flow rate of 25 μL/min until approximately 3000 resonance units (RUs) were immobilized. Remaining active groups were quenched by a 300 s injection of 1M ethanolamine at 30 μL/min in the analyte direction, and this also ensures mock-activated interspots are created for blank referencing.
  • A 500 nM 3-fold dilution series of V515 was injected over 3000 RUs CD16aWT (L6) compared to blank (L5). Flow rate 50 uL/min for 120s, with a 240s disassociation phase. Injections were repeated in standard running buffer (DPBS/3.4 mM EDTA/0.05% Tween20) and running buffer with an additional 350 mM NaCl. Sensorgrams were aligned and double-referenced using the buffer blank injection and interspots, and the resulting sensorgrams were analyzed using ProteOn Manager software v3.0. Typically, KD values were determined from binding isotherms using the Equilibrium Fit model. For high affinity interactions with slow off-rates, kinetic and affinity values were additionally determined by fitting the referenced sensorgrams to the 1:1 Langmuir binding model using local Rmax, and affinity constants (KD M) were derived from the resulting rate constants (kd s−1/ka M−1s−1). All KD values are reported as the mean and standard deviation from three independent runs.
  • As shown in Table 9, ABH2 heteromultimer fused to a single antiCD16scFv has full activity and binds its target with good reproducibility and KD similar to the free anti CD16 scFv (NM3E).
  • TABLE 9
    SPR data for monovalent ABH2 fused to a single antiCD16scFv.
    Injection #1 Injection #2
    ka kd KD ka kd KD KD (M)
    1/Ms 1/s M 1/Ms 1/s M Ave KD SD
    NM3E 5.37E+04 5.76E−03 1.07E−07 5.89E+04 6.03E−03 1.02E−07 1.05E−07 4.E−09
    V515 December 6.11E+04 6.71E−03 1.10E−07
    V515 January 5.56E+04 7.30E−03 1.31E−07
  • Example 4 Preparation of HA or HAA Based Heteromultimer Proteins Wherein Cargo Protein(s) Comprise One or More EGF-A Like Domain
  • The peptide sequence of the EGF-A domain in PCSK9 protein or another polypeptide sequence homologous to the EGF-A domain, capable of specifically binding the low density lipoprotein receptor (LDL-R) is derived by sequencing or from a database such as GenBank. The cDNA for the cargo polypeptide comprising EGF-A like domain is isolated by a variety of means including but not exclusively, from cDNA libraries, by RT-PCR and by PCR using a series of overlapping synthetic oligonucleotide primers, all using standard methods. In certain examples, the cargo protein is engineered to improve stability, target binding features or other biophysical or therapeutically relevant properties. The polypeptide is employed as the cargo protein in the creation of a heteromultimer with application in the treatment of hypercholesterolemia. The first and second monomeric fusion polypeptide sequence is derived by fusing the cargo protein sequence directly or with an intermediate linker peptide to the N-terminus and/or C-terminus of HA or HAA based transporter polypeptide such as SEQ ID No: 2, SEQ ID NO: 3, SEQ ID NO: 8 or SEQ ID NO: 10. This monomeric fusion protein sequence is reverse translated to its corresponding DNA sequence to be introduced in an expression vector, sequence optimized for expression in a particular cell line of interest. The first and second monomeric fusion proteins are transfected and coexpressed in the cell line of interest. In certain cases, the transfection is in 1:1 ratio for the two vectors. In some examples, the ratio is selected from 1.5:1, 2:1, 1:1.5, 1:2 etc.
  • Example 5 Preparation of HA or HAA Based Heteromultimeric Proteins Wherein Cargo Protein(s) are the GLP-1 and/or Glucagon
  • The peptide sequence of GLP-1 or another polypeptide sequence homologous to this peptide, capable of specifically binding the GLP-1 receptor or acting as a GLP-1 agonist is derived by sequencing or from a database such as GenBank. Alternately, the peptide sequence of Glucagon or another polypeptide sequence homologous to this peptide, capable of specifically binding the Glucagon receptor or acting as a Glucagon receptor agonist is derived by sequencing or from a database such as GenBank. The cDNA for each cargo polypeptide comprising GLP-1 or Glucagon is isolated by a variety of means including but not exclusively, from cDNA libraries, by RT-PCR and by PCR using a series of overlapping synthetic oligonucleotide primers, all using standard methods. In certain examples, these GLP-1 or Glucagon based cargo polypeptides are engineered to improve stability, target binding features or other biophysical or therapeutically relevant properties. These GLP-1 and Glucagon based polypeptides are employed as one or more cargo molecules in the creation of a heteromultimer with application in the treatment of type-2 diabetes or another disease related to glucose metabolism. The first and second monomeric fusion polypeptide sequence is derived by fusing the cargo protein sequence directly or with an intermediate linker peptide to the N-terminus and/or C-terminus of HA or HAA based transporter polypeptide such as SEQ ID No: 2, SEQ ID NO: 3, SEQ ID NO: 8 or SEQ ID NO: 10. The fusion proteins can be monospecific with either GLP-1 or Glucagon like polypeptides or be bispecific (coagonist) with both the GLP-1 and Glucagon like polypeptides. Each monomeric fusion protein sequence is reverse translated to its corresponding DNA sequence to be introduced in an expression vector, sequence optimized for expression in a particular cell line of interest. The first and second monomeric fusion proteins are transfected and coexpressed in the cell line of interest. In certain cases, the transfection is in 1:1 ratio for the two vectors. In some examples, the ratio is selected from 1.5:1, 2:1, 1:1.5, 1:2 etc
  • Sequence of Cargo molecule GLP-1
    SEQ ID No: 12:
    HAEGTFTSDVSSYLEGQAAKEFIAWLVKGRG
    Sequence of Cargo molecule Glucagon
    SEQ ID NO: 13:
    HSQGTFTSDYSKYLDSRRAQDFVQWLMNT
  • Example 6: Annexin Protein Repeat as Membrane-Sensing Multivalent Scaffold
  • Annexin is split with an extensive interface to generate a multivalent heteromultimer scaffold comprising two transporter polypeptides. Annexin is a 346 residue protein (PDB ID 1MCX). Heteromultimer comprising two transporter polypeptides based on annexin split in the region between residue 186 and 194 is shown in FIG. 11. When co-expressed in solution, the large interfacial area between the two transporter polypeptides leads to self-assembly of the heterodimer. The self-assembly of the two units allows for the design of multivalent construct with transporter polypeptides based on the annexin core. Two structures are available, Pig and Human. The two structures are superimposable with an rmsd of 0.6 A. The following stretch of sequence can be removed from the human Annexin sequence DRSEDF (residues 160 through 165). The truncation does not break any secondary structure element and does not involve introducing or removing any Proline residue.
  • Human annexin WT Sequence
    SEQ ID NO: 14:
    GSAVSPYPTFNPSSDVAALHKAIMVKGVDEATIIDILTKRNNAQRQQIKA
    AYLQETGKPLDETLKKALTGHLEEVVLALLKTPAQFDADELRAAMKGLGT
    DEDTLIEILASRTNKEIRDINRVYREELKRDLAKDITSDTSGDFRNALLS
    LAKGDRSEDFGVNEDLADSDARALYEAGERRKGTDVNVFNTILTTRSYPQ
    LRRVFQKYTKYSKHDMNKVLDLELKGDIEKCLTAIVKCATSKPAFFAEKL
    HQAMKGVGTRHKALIRIMVSRSEIDMNDIKAFYQKMYGISLCQAILDETK
    GDYEKILVALCGGN
    Sequence of Annexin based transporter
    polypeptide-1:
    SEQ ID NO: 15:
    SAVSPYPTFNPSSDVAALHKAIMVKGVDEATIIDILTKRNNAQRQQIKAA
    YLQETGKPLDETLKKALTGHLEEVVLALLKTPAQFDADELRAAMKGLGTD
    EDTLIEILASRTNKEIRDINRVYREELKRDLAKDITSDTSGDFRNALLSL
    AKG
    Sequence of Annexin based transporter
    polypeptide-2:
    SEQ ID NO: 16:
    GVNEDLADSDARALYEAGERRKGTDVNVFNTILTTRSYPQLRRVFQKYTK
    YSKHDMNKVLDLELKGDIEKCLTAIVKCATSKPAFFAEKLHQAMKGVGTR
    HKALIRIMVSRSEIDMNDIKAFYQKMYGISLCQAILDETKGDYEKILVAL
    CGGN
  • FIG. 12 shows a plot of the buried solvent accessible surface area at the interface of Annexin based transporter polypeptide-1 (ABT-1), and Annexin based transporter polypeptide-2 (ABT-2). A split annexin near residue position 186 forms a heterodimer with about 3200 Å2 of buried surface area. The transporter polypeptides such as ABT-1 and ABT-2 based on Annexin can be used to attach cargo biomolecules using the same methods as described above for albumin based transporter polypeptides.
  • Example 7: Transferrin as a Multivalent Scaffold
  • Based on the large number of therapeutically relevant properties of transferrin, this protein presents itself as an interesting scaffold molecule for the design of multivalent protein fusion drugs following the creation of a self-assembling protein and its split component parts. The structure of transferrin is shown in FIG. 13 based on the crystal structure (1H76) available in the protein data bank [Hall D R et al. Acta Crystallogr D 2002, 58, 70-80]. The transferrin molecule is composed of two structurally similar lobes, the N and C terminal lobes, connected by a short peptide linker between residues 333 and 342.
  • A heterodimer is designed based on transferrin protein, said heterodimer comprising a first transporter polypeptide involving residues 1-333 of transferrin and a second transporter polypeptide composed of residues from 342 to the C-terminus of the original transferrin sequence. When coexpressed, the two transporter polypeptides fold independently and pair to form a quasi-transferrin scaffold capable of maintaining its therapeutically relevant properties. Furthermore, such a Transferrin scaffold allows for the production of multivalent fusion molecules, e.g. a multivalent GLP-1 fusion with transporter polypeptides based on transferring. These fusions can be similar to the GLP-1-fusion polypeptides with Albumin based transporter polypeptides.
  • FIG. 13 provides structure of transferrin molecule based on the PDB structure 1H76. The two monomeric transporter polypeptides derived by splitting the transferrin molecule are color coded as light and dark grey units. The sites of fusion for the cargo molecules are represented as spheres. FIG. 14 shows a plot of the buried solvent accessible surface area at the interface of two transferrin based polypeptides. A split transferrin near residue position 330 such as the two transporter polypeptides shown below, forms a heterodimer with about 1800 A2 of buried surface area.
  • Sequence of Transferrin based transporter
    polypeptide-1:
    SEQ ID NO: 17:
    MRLAVGALLV CAVLGLCLAV PDKTVRWCAV SEHEATKCQS
    FRDHMKSVIP SDGPSVACVK KASYLDCIRA IAANEADAVT
    LDAGLVYDAY LAPNNLKPVV AEFYGSKEDP QTFYYAVAVV
    KKDSGFQMNQ LRGKKSCHTG LGRSAGWNIP IGLLYCDLPE
    PRKPLEKAVA NFFSGSCAPC ADGTDFPQLC QLCPGCGCST
    LNQYFGYSGA FKCLKDGAGD VAFVKHSTIF ENLANKADRD
    QYELLCLDNT RKPVDEYKDC HLAQVPSHTV VARSMGGKED
    LIWELLNQAQ EHFGKDKSKE FQLFSSPHGK DLLFKDSAHG
    FLKVPPRMDA KMYLGYEYVT AIRNLREG.
    Sequence of Transferrin based transporter
    polypeptide-2:
    SEQ ID NO: 18:
    ECKPVKWCALSHHE RLKCDEWSVN SVGKIECVSA ETTEDCIAKI
    MNGEADAMSL DGGFVYIAGK CGLVPVLAEN YNKSDNCEDT
    PEAGYFAVAV VKKSASDLTW DNLKGKKSCH TAVGRTAGWN
    IPMGLLYNKI NHCRFDEFFS EGCAPGSKKD SSLCKLCMGS
    GLNLCEPNNK EGYYGYTGAF RCLVEKGDVA FVKHQTVPQN
    TGGKNPDPWA KNLNEKDYEL LCLDGTRKPV EEYANCHLAR
    APNHAVVTRK DKEACVHKIL RQQQHLFGSN VTDCSGNFCL
    FRSETKDLLF RDDTVCLAKL HDRNTYEKYL GEEYVKAVGN
    LRKCSTSSLL EACTFRRP.
  • Example 9: Multiple Cargo Proteins
  • The heteromultimer proteins described herein (e.g, containing a cargo polypeptide (or fragment or variant thereof) fused to transporter albumin segment or variant thereof) may additionally be fused to other proteins to generate “multifusion proteins”. These multifusion proteins can be used for a variety of applications. For example, fusion of the proteins described herein to His-tag IgG domains, and maltose binding protein facilitates purification. (See e.g EP A 394,827; Traunecker et al., Nature 331:84-86 (1988)). Nuclear localization signals fused to the polypeptides can target the protein to a specific subcellular localization. Furthermore, the fusion of additional protein sequences to proteins described herein may further increase the solubility and/or stability of the heteromultimer. The heteromultimer proteins described above can be made using or routinely modifying techniques known in the art and/or by modifying the following protocol, which outlines the fusion of a polypeptide to an IgG molecule.
  • Briefly, the human Fc portion of the IgG molecule can be PCR amplified, using primers that span the 5′ and 3′ ends of the sequence described below. These primers also should have convenient restriction enzyme sites that will facilitate cloning into an expression vector, preferably a mammalian or yeast expression vector.
  • For example, if pC4 (ATCC Accession No. 209646) is used, the human Fc portion can be ligated into the BamHI cloning site. Note that the 3′ BamHI site should be destroyed. Next, the vector containing the human Fc portion is re-restricted with BamHI, linearizing the vector, and a polynucleotide encoding a heteromultimeric protein described herein (generated and isolated using techniques known in the art), is ligated into this BamHI site. Note that the polynucleotide encoding the fusion protein of the invention is cloned without a stop codon; otherwise an Fc containing fusion protein will not be produced.
  • If the naturally occurring signal sequence is used to produce the heteromultimeric protein described herein, pC4 does not need a second signal peptide. Alternatively, if the naturally occurring signal sequence is not used, the vector can be modified to include a heterologous signal sequence. (See, e.g., International Publication No. WO 96/34891.)

Claims (19)

1.-54. (canceled)
55. A heteromultimer comprising:
(i) a first monomeric protein that comprises a first transporter polypeptide comprising a first segment of albumin, and at least one cargo polypeptide, and
(ii) a second monomeric protein that comprises a second transporter polypeptide comprising a second segment of albumin;
wherein the first segment of albumin and the second segment of albumin are derived from an albumin by segmentation of the albumin, the first transporter polypeptide is different from the second transporter polypeptide, and the transporter polypeptides self-assemble to form a quasi-native albumin structure.
56. The heteromultimer according to claim 55, wherein the first segment of albumin and the second segment of albumin form a complementary pair of transporter polypeptides.
57. The heteromultimer according to claim 56, wherein first transporter polypeptide and the second transporter polypeptide are derived from a mammalian albumin.
58. The heteromultimer according to claim 57, wherein the first transporter polypeptide and the second transporter polypeptide are derived from the same type of albumin.
59. The heteromultimer according to claim 58, wherein the mammalian albumin is human serum albumin or variant thereof or an alloalbumin or variant thereof.
60. The heteromultimer according to claim 57, wherein the transporter polypeptides are derived from different albumins.
61. The heteromultimer according to claim 57, wherein:
a. at least one transporter polypeptide is derived from an alloalbumin;
b. at least one transporter polypeptide is derived from human serum albumin;
c. one of the first transporter polypeptide and the second transporter polypeptide is derived from an alloalbumin and the other is derived from a different alloalbumin, or
d. one of the first transporter polypeptide and the second transporter polypeptide is derived from human serum albumin and the other is derived from an alloalbumin.
62. The heteromultimer according to claim 57, wherein the at least one cargo polypeptide is an antibody, or a fragment or variant thereof.
63. The heteromultimer according to claim 62, wherein the antibody is a bispecific antibody, a multispecific antibody, or a therapeutic antibody.
64. The heteromultimer of claim 63, wherein the therapeutic antibody binds a cancer antigen.
65. A pharmaceutical composition comprising the heteromultimer according to claim 55, and a pharmaceutically acceptable carrier.
66. One or a combination of two or more nucleic acids encoding the heteromultimer according to claim 55.
67. One or more vectors comprising the nucleic acids according to claim 66.
68. A host cell comprising nucleic acid encoding the heteromultimer according to claim 55.
69. A method of expressing a heteromultimer in cells, the method comprising:
a) transfecting at least one cell with the nucleic acids according to claim 66, to produce at least one transfected cell; and
b) culturing the at least one transfected cell under conditions suitable for expressing the heteromultimer.
70. A method of treating a disease in a subject, comprising administration of an effective amount of the heteromultimer according to claim 55 to the subject wherein the disease is selected from an immune disorder, an infectious disease, a cardiovascular disorder, a respiratory disorder, or a metabolic disorder.
71. A method of making the heteromultimer according to claim 55, comprising segmenting the albumin to obtain polypeptides such that the polypeptides self-assemble to form the heteromultimer.
72. A method of making the heteromultimer according to claim 55, comprising culturing the host cell of claim 68 such that the nucleic acid encoding the heteromultimer is expressed, and recovering the heteromultimer from the cell culture.
US16/897,583 2011-03-03 2020-06-10 Multivalent heteromultimer scaffold design and constructs Abandoned US20210009659A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US16/897,583 US20210009659A1 (en) 2011-03-03 2020-06-10 Multivalent heteromultimer scaffold design and constructs

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US201161449016P 2011-03-03 2011-03-03
US13/411,353 US9499605B2 (en) 2011-03-03 2012-03-02 Multivalent heteromultimer scaffold design and constructs
US15/355,007 US10155803B2 (en) 2011-03-03 2016-11-17 Multivalent heteromultimer scaffold design and constructs
US16/180,456 US10711051B2 (en) 2011-03-03 2018-11-05 Multivalent heteromultimer scaffold design and constructs
US16/897,583 US20210009659A1 (en) 2011-03-03 2020-06-10 Multivalent heteromultimer scaffold design and constructs

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US16/180,456 Continuation US10711051B2 (en) 2011-03-03 2018-11-05 Multivalent heteromultimer scaffold design and constructs

Publications (1)

Publication Number Publication Date
US20210009659A1 true US20210009659A1 (en) 2021-01-14

Family

ID=46757329

Family Applications (4)

Application Number Title Priority Date Filing Date
US13/411,353 Active US9499605B2 (en) 2011-03-03 2012-03-02 Multivalent heteromultimer scaffold design and constructs
US15/355,007 Active 2032-03-28 US10155803B2 (en) 2011-03-03 2016-11-17 Multivalent heteromultimer scaffold design and constructs
US16/180,456 Active US10711051B2 (en) 2011-03-03 2018-11-05 Multivalent heteromultimer scaffold design and constructs
US16/897,583 Abandoned US20210009659A1 (en) 2011-03-03 2020-06-10 Multivalent heteromultimer scaffold design and constructs

Family Applications Before (3)

Application Number Title Priority Date Filing Date
US13/411,353 Active US9499605B2 (en) 2011-03-03 2012-03-02 Multivalent heteromultimer scaffold design and constructs
US15/355,007 Active 2032-03-28 US10155803B2 (en) 2011-03-03 2016-11-17 Multivalent heteromultimer scaffold design and constructs
US16/180,456 Active US10711051B2 (en) 2011-03-03 2018-11-05 Multivalent heteromultimer scaffold design and constructs

Country Status (9)

Country Link
US (4) US9499605B2 (en)
EP (1) EP2681245B1 (en)
JP (1) JP6101638B2 (en)
CN (1) CN103732628B (en)
AU (1) AU2012222833B2 (en)
CA (1) CA2828811C (en)
DK (1) DK2681245T3 (en)
ES (1) ES2676878T3 (en)
WO (1) WO2012116453A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11248037B2 (en) * 2012-07-13 2022-02-15 Zymeworks Inc. Multivalent heteromultimer scaffold design and constructs

Families Citing this family (86)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2008234248C1 (en) 2007-03-29 2015-01-22 Genmab A/S Bispecific antibodies and methods for production thereof
WO2011028952A1 (en) 2009-09-02 2011-03-10 Xencor, Inc. Compositions and methods for simultaneous bivalent and monovalent co-engagement of antigens
MX353144B (en) 2010-04-20 2017-12-20 Genmab As Heterodimeric antibody fc-containing proteins and methods for production thereof.
CN105585630B (en) 2010-07-29 2020-09-15 Xencor公司 Antibodies with modified isoelectric points
PL2635607T3 (en) 2010-11-05 2020-05-18 Zymeworks Inc. Stable heterodimeric antibody design with mutations in the fc domain
WO2012116453A1 (en) 2011-03-03 2012-09-07 Zymeworks Inc. Multivalent heteromultimer scaffold design and constructs
WO2013025846A2 (en) 2011-08-17 2013-02-21 The Regents Of The University Of Colorado, A Body Corporate Transferrin-tumstatin fusion protein and methods for producing and using the same
US10851178B2 (en) 2011-10-10 2020-12-01 Xencor, Inc. Heterodimeric human IgG1 polypeptides with isoelectric point modifications
EP2771364B1 (en) 2011-10-27 2019-05-22 Genmab A/S Production of heterodimeric proteins
WO2013063702A1 (en) 2011-11-04 2013-05-10 Zymeworks Inc. Stable heterodimeric antibody design with mutations in the fc domain
US9499634B2 (en) 2012-06-25 2016-11-22 Zymeworks Inc. Process and methods for efficient manufacturing of highly pure asymmetric antibodies in mammalian cells
ES2805361T3 (en) * 2012-07-13 2021-02-11 Zymeworks Inc Design and constructions of multivalent heteromultimer scaffolds
US9914785B2 (en) 2012-11-28 2018-03-13 Zymeworks Inc. Engineered immunoglobulin heavy chain-light chain pairs and uses thereof
US10487155B2 (en) 2013-01-14 2019-11-26 Xencor, Inc. Heterodimeric proteins
US10131710B2 (en) 2013-01-14 2018-11-20 Xencor, Inc. Optimized antibody variable regions
US10968276B2 (en) 2013-03-12 2021-04-06 Xencor, Inc. Optimized anti-CD3 variable regions
US9701759B2 (en) 2013-01-14 2017-07-11 Xencor, Inc. Heterodimeric proteins
KR102211837B1 (en) 2013-01-14 2021-02-03 젠코어 인코포레이티드 Novel heterodimeric proteins
US9605084B2 (en) 2013-03-15 2017-03-28 Xencor, Inc. Heterodimeric proteins
US11053316B2 (en) 2013-01-14 2021-07-06 Xencor, Inc. Optimized antibody variable regions
CA2897987A1 (en) 2013-01-15 2014-07-24 Xencor, Inc. Rapid clearance of antigen complexes using novel antibodies
US10519242B2 (en) 2013-03-15 2019-12-31 Xencor, Inc. Targeting regulatory T cells with heterodimeric proteins
AU2014232416B2 (en) 2013-03-15 2017-09-28 Xencor, Inc. Modulation of T Cells with Bispecific Antibodies and FC Fusions
US10858417B2 (en) 2013-03-15 2020-12-08 Xencor, Inc. Heterodimeric proteins
US10106624B2 (en) 2013-03-15 2018-10-23 Xencor, Inc. Heterodimeric proteins
RU2747857C2 (en) 2013-03-15 2021-05-17 Ин3Био Лтд. Self-assembling synthetic proteins
CN110143999B (en) 2013-03-15 2023-12-05 酵活英属哥伦比亚省公司 Cytotoxic and antimitotic compounds and methods of use thereof
PL3083689T3 (en) 2013-12-17 2020-10-19 Genentech, Inc. Anti-cd3 antibodies and methods of use
AU2014373574B2 (en) 2013-12-27 2020-07-16 Zymeworks Bc Inc. Sulfonamide-containing linkage systems for drug conjugates
RS59907B1 (en) 2014-03-28 2020-03-31 Xencor Inc Bispecific antibodies that bind to cd38 and cd3
GB201409558D0 (en) 2014-05-29 2014-07-16 Ucb Biopharma Sprl Method
GB201412658D0 (en) 2014-07-16 2014-08-27 Ucb Biopharma Sprl Molecules
GB201412659D0 (en) 2014-07-16 2014-08-27 Ucb Biopharma Sprl Molecules
KR102494557B1 (en) 2014-09-17 2023-02-02 자임워크스 비씨 인코포레이티드 Cytotoxic and anti-mitotic compounds, and methods of using the same
IL252480B2 (en) 2014-11-26 2023-12-01 Xencor Inc Heterodimeric antibodies that bind cd3 and tumor antigens
AU2015353416C1 (en) 2014-11-26 2022-01-27 Xencor, Inc. Heterodimeric antibodies that bind CD3 and CD38
US10259887B2 (en) 2014-11-26 2019-04-16 Xencor, Inc. Heterodimeric antibodies that bind CD3 and tumor antigens
US10865253B2 (en) 2014-12-19 2020-12-15 Genmab A/S Rodent bispecific heterodimeric proteins
US10428155B2 (en) 2014-12-22 2019-10-01 Xencor, Inc. Trispecific antibodies
US10227411B2 (en) 2015-03-05 2019-03-12 Xencor, Inc. Modulation of T cells with bispecific antibodies and FC fusions
JP2018526972A (en) 2015-06-16 2018-09-20 ジェネンテック, インコーポレイテッド Anti-CD3 antibody and method of use
EP4074730A1 (en) 2015-06-24 2022-10-19 F. Hoffmann-La Roche AG Anti-transferrin receptor antibodies with tailored affinity
EP3322735A4 (en) 2015-07-15 2019-03-13 Zymeworks Inc. Drug-conjugated bi-specific antigen-binding constructs
GB201601075D0 (en) 2016-01-20 2016-03-02 Ucb Biopharma Sprl Antibodies molecules
GB201601073D0 (en) 2016-01-20 2016-03-02 Ucb Biopharma Sprl Antibodies
GB201601077D0 (en) 2016-01-20 2016-03-02 Ucb Biopharma Sprl Antibody molecule
NZ741067A (en) 2015-10-02 2023-07-28 Hoffmann La Roche Bispecific anti-human cd20/human transferrin receptor antibodies and methods of use
AR106189A1 (en) 2015-10-02 2017-12-20 Hoffmann La Roche BIESPECTIFIC ANTIBODIES AGAINST HUMAN A-b AND THE HUMAN TRANSFERRINE RECEIVER AND METHODS OF USE
GB201521383D0 (en) 2015-12-03 2016-01-20 Ucb Biopharma Sprl And Ucb Celltech Method
GB201521393D0 (en) * 2015-12-03 2016-01-20 Ucb Biopharma Sprl Antibodies
GB201521382D0 (en) 2015-12-03 2016-01-20 Ucb Biopharma Sprl Antibodies
GB201521391D0 (en) * 2015-12-03 2016-01-20 Ucb Biopharma Sprl Antibodies
GB201521389D0 (en) 2015-12-03 2016-01-20 Ucb Biopharma Sprl Method
CN108699136B (en) 2015-12-07 2022-03-18 Xencor股份有限公司 Heterodimeric antibodies that bind CD3 and PSMA
RU2767357C2 (en) 2016-06-14 2022-03-17 Ксенкор, Инк. Bispecific checkpoint inhibitors antibodies
MX2018016265A (en) 2016-06-28 2019-07-04 Xencor Inc Heterodimeric antibodies that bind somatostatin receptor 2.
EP3481413A4 (en) * 2016-07-08 2020-01-08 Askgene Pharma, Inc. Fusion protein comprising leptin and methods for producing and using the same
US10793632B2 (en) 2016-08-30 2020-10-06 Xencor, Inc. Bispecific immunomodulatory antibodies that bind costimulatory and checkpoint receptors
EP3526240A1 (en) 2016-10-14 2019-08-21 Xencor, Inc. Bispecific heterodimeric fusion proteins containing il-15/il-15ralpha fc-fusion proteins and pd-1 antibody fragments
TWI791471B (en) 2016-11-15 2023-02-11 美商建南德克公司 Dosing for treatment with anti-cd20/anti-cd3 bispecific antibodies
CN108341853B (en) * 2017-01-22 2022-06-21 中国科学院化学研究所 Human serum albumin specificity recognition polypeptide and application thereof
EP3596108A4 (en) 2017-03-15 2020-12-23 Pandion Operations, Inc. Targeted immunotolerance
JOP20190222A1 (en) 2017-04-11 2019-09-24 Zymeworks Inc Anti-pd-l1-anti-tim-3 bispecific antibodies
CA3064435A1 (en) 2017-05-24 2018-11-29 Pandion Therapeutics, Inc. Targeted immunotolerance
WO2019003180A1 (en) * 2017-06-29 2019-01-03 Savitribai Phule Pune University Enhanced production, and usages, of a self-assembling protein secreted by a native yarrowia lipolytica strain
CN111132733A (en) 2017-06-30 2020-05-08 Xencor股份有限公司 Targeted heterodimeric Fc fusion proteins containing IL-15/IL-15R α and an antigen binding domain
AR112603A1 (en) 2017-07-10 2019-11-20 Lilly Co Eli BIS SPECIFIC ANTIBODIES CONTROL POINT INHIBITORS
EP3706793A1 (en) 2017-11-08 2020-09-16 Xencor, Inc. Bispecific and monospecific antibodies using novel anti-pd-1 sequences
US10981992B2 (en) 2017-11-08 2021-04-20 Xencor, Inc. Bispecific immunomodulatory antibodies that bind costimulatory and checkpoint receptors
US10946068B2 (en) 2017-12-06 2021-03-16 Pandion Operations, Inc. IL-2 muteins and uses thereof
US10174092B1 (en) 2017-12-06 2019-01-08 Pandion Therapeutics, Inc. IL-2 muteins
SG11202005732XA (en) 2017-12-19 2020-07-29 Xencor Inc Engineered il-2 fc fusion proteins
AU2019218959A1 (en) 2018-02-08 2020-09-03 Genentech, Inc. Bispecific antigen-binding molecules and methods of use
MX2020009469A (en) 2018-03-13 2021-01-29 Zymeworks Inc Anti-her2 biparatopic antibody-drug conjugates and methods of use.
WO2019195623A2 (en) 2018-04-04 2019-10-10 Xencor, Inc. Heterodimeric antibodies that bind fibroblast activation protein
MX2020010910A (en) 2018-04-18 2021-02-09 Xencor Inc Pd-1 targeted heterodimeric fusion proteins containing il-15/il-15ra fc-fusion proteins and pd-1 antigen binding domains and uses thereof.
CA3097741A1 (en) 2018-04-18 2019-10-24 Xencor, Inc. Tim-3 targeted heterodimeric fusion proteins containing il-15/il-15ra fc-fusion proteins and tim-3 antigen binding domains
EP3861016A2 (en) 2018-10-03 2021-08-11 Xencor, Inc. Il-12 heterodimeric fc-fusion proteins
WO2020132368A1 (en) * 2018-12-19 2020-06-25 Cue Biopharma, Inc. T-cell modulatory multimeric polypeptides with conjugation sites and methods of use thereof
AU2020232605A1 (en) 2019-03-01 2021-10-21 Xencor, Inc. Heterodimeric antibodies that bind ENPP3 and CD3
CN114679909A (en) 2019-05-20 2022-06-28 潘迪恩运营公司 MAdCAM-targeted immune tolerance
PE20221511A1 (en) 2019-12-13 2022-10-04 Genentech Inc ANTI-LY6G6D ANTIBODIES AND METHODS OF USE
US11919956B2 (en) 2020-05-14 2024-03-05 Xencor, Inc. Heterodimeric antibodies that bind prostate specific membrane antigen (PSMA) and CD3
WO2022040482A1 (en) 2020-08-19 2022-02-24 Xencor, Inc. Anti-cd28 and/or anti-b7h3 compositions
US11739144B2 (en) 2021-03-09 2023-08-29 Xencor, Inc. Heterodimeric antibodies that bind CD3 and CLDN6
KR20230154311A (en) 2021-03-10 2023-11-07 젠코어 인코포레이티드 Heterodimeric antibodies binding to CD3 and GPC3

Family Cites Families (106)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4179337A (en) 1973-07-20 1979-12-18 Davis Frank F Non-immunogenic polypeptides
DE3668186D1 (en) 1985-04-01 1990-02-15 Celltech Ltd TRANSFORMED MYELOMA CELL LINE AND METHOD USING THE SAME FOR EXPRESSING A GENE ENCODING AN EUKARYONTIC POLYPEPTIDE.
GB8510219D0 (en) 1985-04-22 1985-05-30 Bass Plc Isolation of fermentation products
US4980286A (en) 1985-07-05 1990-12-25 Whitehead Institute For Biomedical Research In vivo introduction and expression of foreign genetic material in epithelial cells
JPS6296086A (en) 1985-10-21 1987-05-02 Agency Of Ind Science & Technol Composite plasmid
US5576195A (en) 1985-11-01 1996-11-19 Xoma Corporation Vectors with pectate lyase signal sequence
GB8601597D0 (en) 1986-01-23 1986-02-26 Wilson R H Nucleotide sequences
GB8615701D0 (en) 1986-06-27 1986-08-06 Delta Biotechnology Ltd Stable gene integration vector
US4857467A (en) 1986-07-23 1989-08-15 Phillips Petroleum Company Carbon and energy source markers for transformation of strains of the genes Pichia
GB8620926D0 (en) 1986-08-29 1986-10-08 Delta Biotechnology Ltd Yeast promoter
US4946778A (en) 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US5869620A (en) 1986-09-02 1999-02-09 Enzon, Inc. Multivalent antigen-binding proteins
US5258498A (en) 1987-05-21 1993-11-02 Creative Biomolecules, Inc. Polypeptide linkers for production of biosynthetic proteins
GB8725529D0 (en) 1987-10-30 1987-12-02 Delta Biotechnology Ltd Polypeptides
DK159088C (en) 1988-04-06 1991-01-28 Oce Helioprint As SCANNING TO DETECT AN ORIGINAL
GB8809129D0 (en) 1988-04-18 1988-05-18 Celltech Ltd Recombinant dna methods vectors and host cells
EP0387319B1 (en) 1988-07-23 1996-03-06 Delta Biotechnology Limited Secretory leader sequences
EP0394827A1 (en) 1989-04-26 1990-10-31 F. Hoffmann-La Roche Ag Chimaeric CD4-immunoglobulin polypeptides
US5108910A (en) 1989-08-22 1992-04-28 Immunex Corporation DNA sequences encoding fusion proteins comprising GM-CSF and IL-3
US5073627A (en) 1989-08-22 1991-12-17 Immunex Corporation Fusion proteins comprising GM-CSF and IL-3
GB8924021D0 (en) 1989-10-25 1989-12-13 Celltech Ltd Recombinant dna method and vectors for the use therein
US6270989B1 (en) 1991-11-05 2001-08-07 Transkaryotic Therapies, Inc. Protein production and delivery
US5641670A (en) 1991-11-05 1997-06-24 Transkaryotic Therapies, Inc. Protein production and protein delivery
US5932448A (en) * 1991-11-29 1999-08-03 Protein Design Labs., Inc. Bispecific antibody heterodimers
FR2686899B1 (en) 1992-01-31 1995-09-01 Rhone Poulenc Rorer Sa NOVEL BIOLOGICALLY ACTIVE POLYPEPTIDES, THEIR PREPARATION AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM.
ATE419355T1 (en) 1992-02-06 2009-01-15 Novartis Vaccines & Diagnostic MARKER FOR CANCER AND BIOSYNTHETIC BINDING PROTEIN FOR IT
US6096289A (en) * 1992-05-06 2000-08-01 Immunomedics, Inc. Intraoperative, intravascular, and endoscopic tumor and lesion detection, biopsy and therapy
AU5670194A (en) 1992-11-20 1994-06-22 Enzon, Inc. Linker for linked fusion polypeptides
TW402639B (en) 1992-12-03 2000-08-21 Transkaryotic Therapies Inc Protein production and protein delivery
GB9225453D0 (en) 1992-12-04 1993-01-27 Medical Res Council Binding proteins
US5780594A (en) 1993-03-01 1998-07-14 The United States Of America As Represented By The Administrator Of The National Aeronautics And Space Administration Biologically active protein fragments containing specific binding regions of serum albumin or related proteins
AU693821B2 (en) 1993-11-18 1998-07-09 Sirtex Medical Limited Controlled release preparation
US6001606A (en) 1994-03-08 1999-12-14 Human Genome Sciences, Inc. Polynucleotides encoding myeloid progenitor inhibitory factor-1 (MPIF-1) and polypeptides encoded thereby
GB9404270D0 (en) 1994-03-05 1994-04-20 Delta Biotechnology Ltd Yeast strains and modified albumins
US6077692A (en) 1995-02-14 2000-06-20 Human Genome Sciences, Inc. Keratinocyte growth factor-2
US5731168A (en) 1995-03-01 1998-03-24 Genentech, Inc. Method for making heteromultimeric polypeptides
WO1996034891A1 (en) 1995-05-05 1996-11-07 Human Genome Sciences, Inc. Human chemokine beta-8, chemokine beta-1 and macrophage inflammatory protein-4
US6291207B1 (en) 1995-07-28 2001-09-18 Northwestern University Herpes virus entry receptor protein
GB9526733D0 (en) 1995-12-30 1996-02-28 Delta Biotechnology Ltd Fusion proteins
US7357927B2 (en) 1996-03-12 2008-04-15 Human Genome Sciences, Inc. Death domain containing receptors
AU751659B2 (en) 1997-05-02 2002-08-22 Genentech Inc. A method for making multispecific antibodies having heteromultimeric and common components
US7951917B1 (en) 1997-05-02 2011-05-31 Genentech, Inc. Method for making multispecific antibodies having heteromultimeric and common components
US20020062010A1 (en) 1997-05-02 2002-05-23 Genentech, Inc. Method for making multispecific antibodies having heteromultimeric and common components
US6350430B1 (en) 1997-10-27 2002-02-26 Lion Bioscience Science Ag Melanocortin receptor ligands and methods of using same
AU751823B2 (en) 1998-05-14 2002-08-29 Merck Patent Gmbh Fused protein
WO2001007608A1 (en) 1999-07-21 2001-02-01 Human Genome Sciences, Inc. Keratinocyte derived interferon
US7183387B1 (en) 1999-01-15 2007-02-27 Genentech, Inc. Polypeptide variants with altered effector function
CA2361615A1 (en) 1999-02-08 2000-08-10 Human Genome Sciences, Inc. The use of vascular endothelial growth factor 2 antagonist in the treatment of an injury to or disorder of an eye
EP1206541A4 (en) 1999-08-05 2002-09-18 Human Genome Sciences Inc Dendritic enriched secreted lymphocyte activation molecule
EP1218408A4 (en) 1999-09-24 2003-02-26 Human Genome Sciences Inc 32 human secreted proteins
DE19963859A1 (en) 1999-12-30 2001-07-12 Apotech Res & Dev Ltd Bi- or oligomer of a di-, tri-, quattro- or pentamer of recombinant fusion proteins
US6946134B1 (en) 2000-04-12 2005-09-20 Human Genome Sciences, Inc. Albumin fusion proteins
US20050100991A1 (en) 2001-04-12 2005-05-12 Human Genome Sciences, Inc. Albumin fusion proteins
JP2003530839A (en) 2000-04-12 2003-10-21 プリンシピア ファーマスーティカル コーポレイション Albumin fusion protein
MXPA03005036A (en) 2000-12-07 2003-09-05 Lilly Co Eli Glp-1 fusion proteins.
US7507413B2 (en) 2001-04-12 2009-03-24 Human Genome Sciences, Inc. Albumin fusion proteins
US6833441B2 (en) * 2001-08-01 2004-12-21 Abmaxis, Inc. Compositions and methods for generating chimeric heteromultimers
PT2279755E (en) 2001-10-10 2014-06-04 Ratiopharm Gmbh Remodelling and glycoconjugation of fibroblast growth factor (fgf)
WO2005003296A2 (en) 2003-01-22 2005-01-13 Human Genome Sciences, Inc. Albumin fusion proteins
EP1463752A4 (en) 2001-12-21 2005-07-13 Human Genome Sciences Inc Albumin fusion proteins
EP2261250B1 (en) 2001-12-21 2015-07-01 Human Genome Sciences, Inc. GCSF-Albumin fusion proteins
US7332580B2 (en) 2002-04-05 2008-02-19 The Regents Of The University Of California Bispecific single chain Fv antibody molecules and methods of use thereof
DK1553975T3 (en) 2002-09-27 2012-05-07 Xencor Inc Optimized Fc variants and methods for their generation.
US20070041987A1 (en) * 2003-03-19 2007-02-22 Daniel Carter Fragments or polymers of albumin with tunable vascular residence time for use in therapeutic delivery and vaccine development
SI1729795T1 (en) 2004-02-09 2016-04-29 Human Genome Sciences, Inc. Albumin fusion proteins
WO2006106905A1 (en) 2005-03-31 2006-10-12 Chugai Seiyaku Kabushiki Kaisha Process for production of polypeptide by regulation of assembly
EP1945674A2 (en) 2005-11-03 2008-07-23 Genentech, Inc. Therapeutic anti-her2 antibody fusion polypeptides
US8441210B2 (en) 2006-01-20 2013-05-14 Point Somee Limited Liability Company Adaptive current regulation for solid state lighting
JP5225266B2 (en) 2006-05-19 2013-07-03 テバ ファーマシューティカル インダストリーズ リミティド Fusion protein, use thereof, and method for producing the same
ES2469676T3 (en) * 2006-05-25 2014-06-18 Bayer Intellectual Property Gmbh Dimeric molecular complexes
PL3896090T3 (en) * 2006-06-14 2022-05-02 Csl Behring Gmbh Proteolytically cleavable fusion protein comprising a blood coagulation factor
CA2670315A1 (en) 2006-11-21 2008-11-20 The Regents Of The University Of California Anti-egfr family antibodies, bispecific anti-egfr family antibodies and methods of use thereof
WO2008131242A1 (en) 2007-04-18 2008-10-30 Zymogenetics, Inc. Single chain fc, methods of making and methods of treatment
WO2009012784A2 (en) 2007-07-20 2009-01-29 Nya Hamlet Pharma Ab Methods for preparing cytotoxic complexes of emulsifier and fatty acid
US8197196B2 (en) 2007-08-31 2012-06-12 General Electric Company Bushing and clock spring assembly for moveable turbine vane
CA2709847C (en) 2008-01-07 2018-07-10 Amgen Inc. Method for making antibody fc-heterodimeric molecules using electrostatic steering effects
EP2247618B1 (en) 2008-01-25 2014-06-11 Amgen, Inc Ferroportin antibodies and methods of use
WO2009109635A2 (en) 2008-03-05 2009-09-11 Ablynx Nv Novel antigen binding dimer-complexes, methods of making and uses thereof
US20120100558A1 (en) 2008-09-08 2012-04-26 Hanash Samir M Lung cancer diagnosis
KR20110112301A (en) 2008-11-18 2011-10-12 메리맥 파마슈티컬즈, 인크. Human serum albumin linkers and conjugates thereof
US9493545B2 (en) 2009-02-11 2016-11-15 Albumedix A/S Albumin variants and conjugates
US20120076728A1 (en) 2009-04-08 2012-03-29 The Regents Of The University Of California Human protein scaffold with controlled serum pharmacokinetics
WO2011028952A1 (en) 2009-09-02 2011-03-10 Xencor, Inc. Compositions and methods for simultaneous bivalent and monovalent co-engagement of antigens
AU2010306774A1 (en) 2009-10-14 2012-05-03 Merrimack Pharmaceuticals, Inc. Bispecific binding agents targeting IGF-1R and ErbB3 signalling and uses thereof
MX2012004793A (en) 2009-10-30 2012-07-20 Novozymes Biopharma Dk As Albumin variants.
EP2496598B1 (en) 2009-11-04 2017-08-02 Affibody AB Her3 binding polypeptides
WO2011069090A1 (en) 2009-12-04 2011-06-09 Alavita Pharmaceuticals, Inc. Inhibition of the activation of coagulation factor xii by ligands of phosphatidylserine
MX341925B (en) 2010-03-29 2016-09-07 Zymeworks Inc Antibodies with enhanced or suppressed effector function.
CA2796633C (en) 2010-04-23 2020-10-27 Genentech, Inc. Production of heteromultimeric proteins
WO2011143545A1 (en) 2010-05-14 2011-11-17 Rinat Neuroscience Corporation Heterodimeric proteins and methods for producing and purifying them
CA2800769C (en) 2010-05-27 2021-11-02 Genmab A/S Monoclonal antibodies against her2 epitope
PT2591006T (en) 2010-07-09 2019-07-29 Bioverativ Therapeutics Inc Processable single chain molecules and polypeptides made using same
PL2635607T3 (en) 2010-11-05 2020-05-18 Zymeworks Inc. Stable heterodimeric antibody design with mutations in the fc domain
WO2012116453A1 (en) 2011-03-03 2012-09-07 Zymeworks Inc. Multivalent heteromultimer scaffold design and constructs
PL2748201T3 (en) 2011-08-23 2018-04-30 Roche Glycart Ag Bispecific t cell activating antigen binding molecules
WO2013063702A1 (en) 2011-11-04 2013-05-10 Zymeworks Inc. Stable heterodimeric antibody design with mutations in the fc domain
US20130336973A1 (en) 2012-05-10 2013-12-19 Zymeworks Inc. Heteromultimer Constructs of Immunoglobulin Heavy Chains with Mutations in the Fc Domain
WO2013166604A1 (en) 2012-05-10 2013-11-14 Zymeworks Inc. Single-arm monovalent antibody constructs and uses thereof
US9499634B2 (en) 2012-06-25 2016-11-22 Zymeworks Inc. Process and methods for efficient manufacturing of highly pure asymmetric antibodies in mammalian cells
JP6498601B2 (en) 2012-07-13 2019-04-10 ザイムワークス,インコーポレイテッド Multivalent heteromultimeric scaffold designs and constructs
EP2872170A4 (en) 2012-07-13 2016-06-22 Zymeworks Inc Bispecific asymmetric heterodimers comprising anti-cd3 constructs
US20140072581A1 (en) 2012-07-23 2014-03-13 Zymeworks Inc. Immunoglobulin Constructs Comprising Selective Pairing of the Light and Heavy Chains
US9914785B2 (en) 2012-11-28 2018-03-13 Zymeworks Inc. Engineered immunoglobulin heavy chain-light chain pairs and uses thereof
CA3206122A1 (en) 2012-11-28 2014-06-05 Zymeworks Bc Inc. Engineered immunoglobulin heavy chain-light chain pairs and uses thereof
US10239951B2 (en) 2013-05-08 2019-03-26 Zymeworks Inc. Bispecific HER2 and HER3 antigen binding constructs
JP2016531100A (en) 2013-07-12 2016-10-06 ザイムワークス,インコーポレイテッド Bispecific CD3 and CD19 antigen binding constructs

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11248037B2 (en) * 2012-07-13 2022-02-15 Zymeworks Inc. Multivalent heteromultimer scaffold design and constructs

Also Published As

Publication number Publication date
EP2681245B1 (en) 2018-05-09
CN103732628B (en) 2017-06-23
US20120244577A1 (en) 2012-09-27
AU2012222833A1 (en) 2013-09-26
CA2828811C (en) 2021-09-21
US20190127443A1 (en) 2019-05-02
CA2828811A1 (en) 2012-09-07
EP2681245A4 (en) 2015-02-18
EP2681245A1 (en) 2014-01-08
JP2014512343A (en) 2014-05-22
US9499605B2 (en) 2016-11-22
DK2681245T3 (en) 2018-08-13
WO2012116453A1 (en) 2012-09-07
US10155803B2 (en) 2018-12-18
JP6101638B2 (en) 2017-03-22
US10711051B2 (en) 2020-07-14
US20170174745A1 (en) 2017-06-22
CN103732628A (en) 2014-04-16
AU2012222833B2 (en) 2017-03-16
ES2676878T3 (en) 2018-07-25

Similar Documents

Publication Publication Date Title
US20210009659A1 (en) Multivalent heteromultimer scaffold design and constructs
US11248037B2 (en) Multivalent heteromultimer scaffold design and constructs
CA2471363C (en) Albumin fusion proteins
JP5973427B2 (en) Insulin analogues containing additional disulfide bonds
JP2007536902A (en) GLP-1 analog complex protein
JP2016511275A (en) Growth hormone compounds
WO2022117044A1 (en) Glp-1/gcg dual receptor agonist polypeptide and fusion protein thereof
EP2906237A2 (en) Multivalent heteromultimer scaffold design an constructs
US10017555B2 (en) Long-acting blood sugar decreasing fusion protein
KR20180003677A (en) Pharmaceutical compositions comprising mutant proteins of human growth hormone or transferrin fusion proteins thereof
KR20220157910A (en) Composition For Combi-Therapy Comprising Growth Differentiation Factor-15 Variant and Glucagon-Like Peptide-1 Receptor Agonist
KR101504824B1 (en) Hyperglycosylated long-acting human growth hormone and method for preparing the same
EA046279B1 (en) ANALOGUES OF RELAXIN AND METHODS OF THEIR USE

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: APPLICATION DISPATCHED FROM PREEXAM, NOT YET DOCKETED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STCB Information on status: application discontinuation

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