CA2347131C - Modified peptides as therapeutic agents - Google Patents

Modified peptides as therapeutic agents Download PDF

Info

Publication number
CA2347131C
CA2347131C CA2347131A CA2347131A CA2347131C CA 2347131 C CA2347131 C CA 2347131C CA 2347131 A CA2347131 A CA 2347131A CA 2347131 A CA2347131 A CA 2347131A CA 2347131 C CA2347131 C CA 2347131C
Authority
CA
Canada
Prior art keywords
peptide
seq
peptides
tmp
protein
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.)
Expired - Lifetime
Application number
CA2347131A
Other languages
French (fr)
Other versions
CA2347131A1 (en
Inventor
Ulrich Feige
Chuan-Fa Liu
Janet Cheetham
Thomas Charles Boone
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.)
Amgen Inc
Original Assignee
Amgen Inc
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=26802505&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=CA2347131(C) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Amgen Inc filed Critical Amgen Inc
Publication of CA2347131A1 publication Critical patent/CA2347131A1/en
Application granted granted Critical
Publication of CA2347131C publication Critical patent/CA2347131C/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Classifications

    • 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
    • C12N9/6489Metalloendopeptidases (3.4.24)
    • C12N9/6491Matrix metalloproteases [MMP's], e.g. interstitial collagenase (3.4.24.7); Stromelysins (3.4.24.17; 3.2.1.22); Matrilysin (3.4.24.23)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • 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
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/04Antihaemorrhagics; Procoagulants; Haemostatic agents; Antifibrinolytic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/08Plasma substitutes; Perfusion solutions; Dialytics or haemodialytics; Drugs for electrolytic or acid-base disorders, e.g. hypovolemic shock
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • 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
    • C07K14/505Erythropoietin [EPO]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/524Thrombopoietin, i.e. C-MPL ligand
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/525Tumour necrosis factor [TNF]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/54Interleukins [IL]
    • C07K14/545IL-1
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/81Protease inhibitors
    • C07K14/8107Endopeptidase (E.C. 3.4.21-99) inhibitors
    • C07K14/8146Metalloprotease (E.C. 3.4.24) inhibitors, e.g. tissue inhibitor of metallo proteinase, TIMP
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto

Abstract

The present invention concerns fusion of Fc domains with biologically active peptides and a process for preparing pharmaceutical agents using biologically active peptides. In this invention, pharmacologically active compounds are prepared by a process comprising: a) selecting at least one peptide that modulates the activity of a protein of interest; and b) preparing a pharmacologic agent comprising an Fc domain covalently linked to at least one amino acid of the selected peptide. Linkage to the vehicle increases the half-life of the peptide, which otherwise would be quickly degraded in vivo. The preferred vehicle is an Fc domain. The peptide is preferably selected by phage display, E. coli display, ribosome display, RNA-peptide screening, or chemical-peptide screening.

Description

Modified Peptides as Therapeutic Agents Background of the Invention Recombinant proteins are an emerging class of therapeutic agents.
Such recombinant therapeutics have engendered advances in protein formulation and chemical modification. Such modifications can protect therapeutic proteins, primarily by blocking their exposure to proteolytic enzymes. Protein modifications may also increase the therapeutic protein's stability, circulation time, and biological activity. A review article describing protein modification and fusion proteins is Francis (1992), Focus on Growth Factors 3:4-10 (Mediscript, London)õ

One useful modification is combination with the "Fc" domain of an antibody. Antibodies comprise two functionally independent parts, a variable domain known as "Fab", which binds antigen, and a constant domain known as "Fc", which links to such effector functions as complement activation and attack by phagocytic cells. An Fc has a long serum half-life, whereas an Fab is short-lived. Capon et al. (1989), Nature 337:525-31. When constructed together with a therapeutic protein, an Fc domain can provide longer half-life or incorporate such functions as Fc receptor binding, protein A binding, complement fixation and perhaps even placental transfer. Id. Table 1 summarizes use of Fc fusions known in the art.

Table 1--Fc fusion with therapeutic proteins Form of Fc - Fusion Therapeutic partner implications Reference IgG1 N-terminus of Hodgkin's disease; U.S. Patent No.
CD30-L anaplastic lymphoma; T- 5,480,981 cell leukemia Murine Fcy2a IL-10 anti-inflammatory; Zheng gLW. (1995), L
transplant resection Immunol. 154: 5590-600 IgG1 TNF receptor septic shock Fisher aLaL (1996), tL
Engl. J. Med. 334:1697-1702; Van Zee, K. W, (1996), J. Immunol. 156:

IgG, IgA, TNF receptor inflammation, autoimmune U.S. Pat. No. 5,808,029, IgM, or IgE disorders issued September 15, (excluding 1998 the first domain) IgG1 CD4 receptor AIDS Capon gJAL (1989), Nature 37: 525-31 IgG1, N-terminus anti-cancer, antiviral Harvill et al. (1995).
IgG3 of IL-2 Immunotech. 1:95-105 IgG1 C-erminus of osteoarthritis; WO 97/23614, published OPG bone density July 3, 1997 IgG1 N-terminus of anti-obesity . PCT/US 97/23183, filed leptin December 11, 1997 Human Ig CTLA-4 autoimmune disorders Linsley (1991), J. Exp.
Cyl M. 174:561-9 A much different approach to development of therapeutic agents is peptide library screening. The interaction of a protein ligand with its receptor often takes place at a relatively large interface. However, as demonstrated for human growth hormone and its receptor, only a few key residues at the interface contribute to most of the binding energy.
Clackson et al. (1995), Science 267: 383-6. The bulk of the protein ligand merely displays the binding epitopes in the right topology or serves functions unrelated to binding. Thus, molecules of only "peptide" length (2 to 40 amino acids) can bind to the receptor protein of a given large protein ligand. Such peptides may mimic the bioactivity oFthe large protein ligand ("peptide agonises") or, through competitive binding, inhibit the bioactivity of the large protein ligand ("peptide antagonists").

Phage display peptide libraries have emerged as a powerful method in identifying such peptide agonists and antagonists. See, for example, Scott et al. (1990), Science 249: 386; Devlin et al. (1990), Science 249:404; U.S. Pat. No. 5,223,409, issued June 29,1993; U.S. Pat. No.
5,733,731, issued March 31, 1998; U.S. Pat. No. 5,498,530, issued March 12, 1996; U.S. Pat. No. 5,432,018, issued July 11, 1995; U.S. Pat. No. 5,338,665, issued August 16,1994; U.S. Pat. No. 5,922,545, issued July 13,1999; WO
96/40987, published December 19,1996; and WO 98/15833, published April 16, 1998 (each of which is incorporated by reference). In such libraries, random peptide sequences are displayed by fusion with coat proteins of filamentous phage. Typically, the displayed peptides are affinity-eluted against an antibody immobilized extracellular domain of a receptor. The retained phages may be enriched by successive rounds of affinity purification and repropagation. The best binding peptides may be sequenced to identify key residues within one or more structurally related families of peptides. See, e.g., Cwirla et al. (1997), Science 276: 1696-9, in which two distinct families were identified. The peptide sequences may also suggest which residues may be safely replaced by alanine scanning or by mutagenesis at the DNA level. Mutagenesis libraries may be created and screened to further optimize the sequence of the best binders.
Lowman (1997), Ann. Rev. Bioõphys. Biomol. Struct. 26:401-24.
Structural analysis of protein-protein interaction may also be used to suggest peptides that mimic the binding activity of large protein ligands. In such an analysis, the crystal structure may suggest the identity .25 and relative orientation of critical residues of the large protein ligand, from which a peptide may be designed. See, e.g., Takasaki et al. (1997), Nature Biotech. 15: 1266-70. These analytical methods may-also-be used to investigate the interaction between a receptor protein and peptides selected by phage display, which may suggest further modification of the peptides to increase binding affinity.
Other methods compete with phage display in peptide research. A
peptide library can be fused to the carboxyl terminus of the lac. repressor and expressed in E. coll. Another E. coli based method allows display on the cell's outer membrane by fusion with a peptidoglycan-associated lipoprotein (PAL). Hereinafter, these and related methods are collectively referred to as "E. coli display." In another method, translation of random RNA is halted prior to ribosome release, resulting in a library of polypeptides with their associated RNA still attached. Hereinafter, this and related methods are collectively referred to as "r bosome display."
Other methods employ chemical linkage of peptides to RNA; see, for example, Roberts & Szostak (1997), Proc. Natl. Acad. Sci. USA, 94:12297-303. Hereinafter, this and related methods are collectively referred to as "RNA peptide screening." Chemically derived peptide libraries have been developed in which peptides are immobilized on stable, non biological materials, such as polyethylene rods or solvent permeable resins. Another chemically derived peptide library uses photolithography to scan peptides immobilized on glass slides. Hereinafter, these and related methods are collectively referred to as "chemical-peptide screening." Chemical peptide screening may be advantageous in that it allows use of D-amino acids and other unnatural analogues, as well as non-peptide elements. Both biological and chemical methods are reviewed in Wells & Lowman (1992), Curr. Opin. Biotechnol. 3:355-62.
Conceptually, one may discover peptide mimetics of any protein using phage display and the other methods mentioned above. These methods have been used for epitope mapping, for identification of critical amino acids in protein-protein interactions, and as leads for the discovery of new therapeutic agents. E.g., Cortese et al. (1996), Curr. O in. Biotech.
7:
616-21. Peptide libraries are now being used most often in immunological studies, such as epitope mapping. Kreeger (1996), The Scientist 10(13):19-20.
Of particular interest here is use of peptide libraries and other techniques in the discovery of pharmacologically active peptides. A
number of such peptides identified in the art are summarized in Table 2.
The peptides are described in the listed publications, each of which is hereby incorporated by reference. The pharmacologic activity of the peptides is described, and in many instances is followed by a shorthand term therefor in parentheses. Some of these peptides have been modified (e.g., to form C-terminally cross-linked dimers). Typically, peptide libraries were screened for binding to a receptor for a pharmacologically active protein (e.g., EPO receptor). In at least one instance (CTLA4), the peptide library was screened for binding to a monclonal antibody.
Table 2-Pharmacologically active peptides Binding Form of partner/ Pharmacologic Reference peptide protein of activity interest' intrapeptide EPO receptor EPO-mimetic Wrighton et.Sl. (1996), disulfide- Science 273: 458-63;
bonded U.S. Pat. No. 5,773,569, issued June 30,1998 to Wrighton g,Ul.
C-terminally EPO receptor EPO-mimetic Uvnah gI.a(. (1996), cross-linked Science 273:464-71;
dimer Wrighton gam. (1997), Nature Biotechnoloav 15:
1261-5; International patent application WO
96/40772, published . -Dec. 19, 1996 linear EPO receptor EPO-mimetic Naranda ate. (1999), PM, Nat], Acad. Sci.
= 96:7569-74 linear c-Mpl TPO-mimetic Cwirla ".(1997) Science 276:1696-9;
U.S. Pat. No. 5,869,451, issued Feb. 9,1999; U.S.
Pat. No. 5,932,946, issued Aug. 3, 1999 C-terminally c-Mpl TPO-mimetic Cwirla at t. (1997), cross-linked Science 276:1696-9 dimer disulfide- stimulation of Paukovits ". (1984), linked dimer hematopoiesis Hoppe-Seylers Z.
("G-CSF-mimetic") Physiol. Chem. 365: 303-11; Laerum el.W. (1988), Exn. Hemat. 16:274-80 alkylene- G-CSF-mimetic Bhatnagar gam(. (1996), linked dimer J. Med. Chem. 39:3814-9; Cuthbertson ".
(1997), J. Med. Chem.
40: 2876-82; King ate.
(1991), Exo. Hematol.
19:481; King AL&I.
(1995), Ziood 86 (Suppl.
1): 309a linear IL-1 receptor inflammatory and U.S. Pat. No. 5,608,035;
autoimmune diseases U.S. Pat. No. 5,786,331;
("IL-1 antagonist" or U.S-Pat. No. 5,880,096;
"IL-1 ra-mimetic") Yanofsky aLal. (1996), ' The protein listed in this column may be bound by the associated peptide (e.g., EPO
receptor, IL-1 receptor) or mimicked by the associated peptide. The references listed for each clarity whether the molecule is bound by or mimicked by the peptides.
Proc. Nati. Acad. Sci. 93:
7381-6; Akeson g(.
(1996), J. Biol. Chem.
271: 30517-23;
Wiekzorek ". (1997), Pal. J. P-harmacol. 49:
107-17; Yanofsky (1996), PNAs, 93:7381-7386.
linear Facteur stimulation of Inagaki-Ohara gam.
thymique lymphocytes (1996), Cellular lmmunol.
serique (FTS) ("FTS-mimetic") 171: 30-40; Yoshida (1984), i,=
Immunoo armacol, 6:141-6.
intrapeptide CTLA4 MAb CTLA4-mimetic ejaL (1998), disulfide Nature Biotech. 16:267-bonded 70 exocyclic TNF-a receptor TNF-u antagonist Takasaki f<5..;~L. (1997), Nature Biotech. 15:1266-70; WO 98/53842, published December 3, linear TNF-a receptor TNF-a antagonist Chirinos-Rojas Imm., 5621-5626.
intrapeptide Cab inhibition of complement Sahu ftW. (1996),, disulfide activation; autoimmune Immunol. 157: 884-91;
bonded diseases- Morikis aU. (1998), Mb-antagonist') Protein Sci. 7:619-27 linear vinculin cell adhesion processes- Adey gta[. (1997), cell growth, differentiation, Biochem. J. 324:523-8 wound healing, tumor metastasis (`vinculin binding") linear C4 binding anti-thrombotic Linse 2-g(. (1997),,L
protein (C4BP) Blol. Chem. 272:14658-linear urokinase processes associated with Goodson 1t6(. (1994), receptor urokinase interaction with Proc. Nati Acad. Sci. 91:
its receptor (e.g., 7129-33; International angiogenesis, tumor cell application WO
invasion and metastasis); 97/35969, published ("UKR antagonist') October 2,1997 linear Mdm2, Hdm2 Inhibition of inactivation of Picksley (. (1994), p53 mediated by Mdm2 or Onnoong 9:2523-9;
hdm2; anti-tumor Bottger .a,(. (1997) ,L
("Mdm/hdm antagonist") 1. Biol. 269: 744-56;
Bottger aW. (1996), Qgj2 13: 2141-7 linear p21 WAR anti-tumor by mimicking - Bait 91jil. (1997), Quri .
the activity of p21 ""F' B!. 7: 71-8.0 linear farnesyl anti-cancer by preventing Gibbs et al. (1994), b FTS is a thymic hormone mimicked by the molecule of this Invention rather than a receptor bound by the molecule of this invention.
transferase activation of ras oncogene 77:175-178 linear Ras effector anti-cancer by inhibiting Moodie et al. (1994), domain biological function of the Trends Genet 10: 44-48 ras oncogene Rodriguez et al. (1994), 1 ture 370.527-532 linear SH2/SH3 anti-cancer by inhibiting Pawson et al (1993), domains tumor growth with rr. Biol. i 3:434-432 activated tyrosine kinases Yu et al. (1994), Ca( 76:933-945 linear p16 anti-cancer by mimicking FAhraeus gf al-. (1996), activity of p16; e.g., rr. Blot. 6:8491 inhibiting cyclin D-Cdk complex ("p16-mimetic") linear Src, Lyn inhibition of Mast cell Stauffer et i. (1997), activation, IgE-related Biochgm. 36: 9388-94 conditions, type I
hypersensitivity ("Mast cell antagonist") linear Mast cell treatment of inflammatory International application protease disorders mediated by WO 98/33812, published release of tryptase-6 August 6, 1998 ("Mast cell protease inhibitors') linear SH3 domains treatment of SH3- Rickles l3La}. (1994), mediated disease states EMBO J. 13: 5598-5604;
("SH3 antagonist") Sparks Stal. (1994), ,L
Biol. Chem. 269: 23853-6; Sparks per(. (1996), Proc. Natl. Acad. Sci. 93:
1540.4 linear HBV core treatment of HBV viral Dyson & Muray (1995), antigen (HBcAg) infections ("anti-HBV') Proc. Natl. Acad. Sd. 92:

linear selectins neutrophil adhesion; Martens gt,g(. (1995), inflammatory diseases 151o l. Chem. 270: 21129-("selectin antagonist') 36; European-patent application EP 0 714 912, published June 5, linear, calmodulin c almodulin antagonist Pierce gt.g1. (1995), cyclized MAoiec. Diversity 1: 259-65; Dedman ffi..ai.
(1993), 1jj*ol. Chem.
268:23025-30; Adey &
Kay (1996), i 169:

linear, integrins tumor-homing; treatment International applications cyclized- for conditions related to WO 95/14714, published integrin-mediated cellular Jung 1, 1995; WO
events, including platelet 97108203, published aggregation, thrombosis, March 6, 1997; WO
wound healing, 98110795, published osteoporosis, tissue March 19, 1998; WO
repair, angiogenesis (e.g., 99124462, published May for treatment of cancer), 20, 1999; Kraft e.t..3.
and tumor invasion (1999), J. Biol. Chem.
("integrin-binding") 274:1979-1985 cyclic, linear fibronectin and treatment of inflammatory WO 98/09985, published extracellular and autoimmune March 12, 1998 matrix conditions components of T
cells and macrophages linear somatostatin treatment or prevention of European patent and cortistatin hormone-producing application 0 911 393, tumors, acromegaly, published April 28, 1999 giantism, dementia, gastric ulcer, tumor growth, inhibition of hormone secretion, modulation of sleep or neural activity linear bacterial antibiotic; septic shock; U.S. Pat. No. 5,877,151, lipopolysac- disorders modulatable by issued March 2, 1999 charide CAP37 linear or pardaxin, mellitin antipathogenic WO 97/31019, published cyclic, 28 August 1997 including D-amino acids linear, cyclic VIP impotence, WO 97/40070, published neurodegenerative October 30, 1997 disorders linear CTLs cancer EP 0 770 624, published May 2, 1997 linear THF-gamma2 Bumstein (1988), Biochem.,, 27:4066-71.
linear Amylin Cooper (1987), Proc=
Natl. Acad. Sci..
84:8628-32.
linear Adrenomedullin Kitamura (1993), i $G, 192:553-60.
cyclic, linear VEGF anti-angiogenic; cancer, Fairbrother (1998), rheumatoid arthritis, Biochem., 37:17754-diabetic retinopathy, 17764.
psoriasis ("VEGF
antagonist cyclic MMP inflammation and Koivunen (1999), Nature autoimmune disorders; Mech., 17:768-774.
tumor growth ("MMP inhibitor") HGH fragment U.S. Pat. No. 5,869,452 Echistatin inhibition of platelet Gan (1988), J. Biol.
aggregation õ 263:19827-32.
linear SLE SLE WO 96/30057, published autoantibody October 3, 1996 GD1alpha suppression of tumor Ishikawa 1a1. (1998), metastasis FEBS Lett. 441 (1): 20-4 antiphospholipid endothelial cell activation, Blank pJAL (1999), P
beta-2- antiphospholipid Nati. Acad. Sci. USA 96:
glycoprotein-1 syndrome (APS), 5164-8 (R2GPt) thromboembolic antibodies phenomena, thrombocytopenia, and recurrent fetal loss linear T Cell Receptor diabetes WO 96/1.1214, published beta chain April 18, 1996 Peptides identified by peptide library screening have been regarded as "leads" in development of therapeutic agents rather than as therapeutic agents themselves. Like other proteins and peptides, they would be rapidly removed in vivo either by renal filtration, cellular clearance mechanisms in the reticuloendothelial system, or proteolytic degradation.
Francis (1992), Focus on Growth Factors 3:4-11. As a result, the art presently uses the identified peptides to validate drug targets or as scaffolds for design of organic compounds that might not have been as easily or as quickly identified through chemical library screening.
Lowman (1997), Ann. Rev. Biophys. Biomol. Struct. 26:401-24; Kay et al.
(1998), Drug Disc. Today 3: 370-8. The art would benefit from a process by which such peptides could more readily yield therapeutic agents.
Summary of the Invention The present invention concerns a process by which the in vivo half-life of one or more biologically active peptides is increased by fusion with a vehicle. In this invention, pharmacologically active compounds are prepared by a process comprising:
a) selecting at least one peptide that modulates the activity of a protein of interest; and b) preparing a pharmacologic agent comprising at least one vehicle covalently linked to at least one amino acid sequence of the selected peptide.
The preferred vehicle is an Fc domain. The peptides screened in step (a) are preferably expressed in a phage display library. The vehicle and the peptide may be linked through the N- or C-terminus of the peptide or the vehicle, as described further below. Derivatives of the above compounds (described below) are also encompassed by this invention.
The compounds of this invention may be prepared by standard synthetic methods, recombinant DNA techniques, or any other methods of preparing peptides and fusion proteins. Compounds of this invention that encompass non-peptide portions may be synthesized by standard organic *
chemistry reactions, in addition to standard peptide chemistry reactions when applicable.
The primary use contemplated is as therapeutic or prophylactic agents. The vehicle-linked peptide may have activity comparable to-or even greater than the natural ligand mimicked by the peptide. In addition, certain natural ligand-based therapeutic agents might induce antibodies against the patient's own endogenous ligand; the vehicle-linked peptide-avoids this pitfall by having little or typically no sequence identity with the natural ligand.
Although mostly contemplated as therapeutic agents, compounds of this invention may also be useful in screening for such agents. For example, one could use an Fc-peptide (e.g., Fc-SH2 domain peptide) in an assay employing anti-Fc coated plates. The vehicle, especially Fc, may make insoluble peptides soluble and thus useful in a number of assays.
The compounds of this invention may be used for therapeutic or prophylactic purposes by formulating them with appropriate pharmaceutical carrier materials and administering an effective amount to a patient, such as a human (or other mammal) in need thereof. Other related aspects are also included in the instant invention.
Numerous additional aspects and advantages of the present invention will become apparent upon consideration of the figures and detailed description of the invention.

Brief Description of the Figures Figure 1 shows a schematic representation of an exemplary process of the invention. In this preferred process, the vehicle is an Fc domain, which is linked to the peptide covalently by expression from a DNA
construct encoding both the Fc domain and the peptide. As noted in Figure 1, the Fc domains spontaneously form a dimer in this process.
Figure 2 shows exemplary Fc dimers that may be derived from an IgG1 antibody. "Fc" in the figure represents any of the Fc variants within the meaning of "Fc domain" herein. "X"' and "X2" represent peptides or linker-peptide combinations as defined hereinafter. The specific dieters are as follows: -A, D: Single disulfide-bonded dimers. IgG1 antibodies typically have two disulfide bonds at the hinge region between the constant and variable domains. The Fc domain in Figures 2A and 2 D may be formed by truncation between the two disulfide bond sites or by substitution of a cysteinyl residue with an unreactive residue (e.g., alanyl). In Figure 2A, the Fc domain is linked at the amino terminus of the peptides; in 2D, at the carboxyl terminus.
B, E: Doubly disulfide-bonded dimers. This Fc domain may be formed by truncation of the parent antibody to retain both cysteinyl residues in the Fc domain chains or by expression from a construct including a sequence encoding such an Fc domain. In Figure 2B, the Fc domain is linked at the amino terminus of the peptides; in 2E, at the carboxyl terminus.
C, F: Noncovalent dimers. This Fc domain may be formed by elimination of the cysteinyl residues by either truncation or substitution.
One may desire to eliminate the cysteinyl residues to avoid impurities formed by reaction of the cysteinyl residue with cysteinyl residues of other }

proteins present in the host cell. The noncovalent bonding of the Fc domains is sufficient to hold together the dimer.
Other dimers may be formed by using Fc domains derived from different types of antibodies (e.g., IgG2, IgM).
Figure 3 shows the structure of preferred compounds of the invention that feature tandem repeats of the pharmacologically active peptide. Figure 3A shows a single chain molecule and may also represent the DNA construct for the molecule. Figure 3B shows a dieter in which the linker-peptide portion is present on only one chain of the dimer. Figure 3C
shows a dimer having the peptide portion on both chains. The dieter of Figure 3C will form spontaneously in certain host cells upon expression of a DNA construct encoding the single chain shown in Figure 3A. In other host cells, the cells could be placed in conditions favoring formation of dimers or the dimers can be formed in vitro.
Figure 4 shows exemplary nucleic acid and amino acid sequences (SEQ ID NOS:1 and 2, respectively) of human IgGi Fc that may be used in this invention.
Figure 5 shows a synthetic scheme for the preparation of PEGylated peptide 19 (SEQ ID NO: 3).
Figure 6 shows a synthetic scheme for the preparation of PEGylated peptide 20 (SEQ ID NO: 4).
Figure 7 shows the nucleotide and amino acid sequences (SEQ ID
NOS: 5 and 6, respectively) of the molecule identified as "Fc-TMP" in Example 2 hereinafter.
Figure 8 shows the nucleotide and amino acid sequences (SEQ. ID.
NOS: 7 and 8, respectively) of the molecule identified as "Fc-TMP TMP" in Exaiilple 2 hereinafter. -Figure 9 shows the nucleotide and amino acid sequences (SEQ. ID.
NOS: 9 arid 10, respectively) of the molecule identified as "TMP-TMP-Fc"
in Example 2 hereinafter.
Figure 10 shows the nucleotide and amino acid sequences (SEQ. ID.
NOS: 11 and 12, respectively) of the molecule identified as "TMP-Fc" in Example 2 hereinafter.
Figure 11 shows the number of platelets generated in vivo in normal female BDF1 mice treated with one 100 g/kg bolus injection of various compounds, with the terms defined as follows.
PEG-MGDF: 20 kD average molecular weight PEG attached by reductive amination to the N-terminal amino group of amino acids 1-163 of native human TPO, which is expressed in E. coli (so that it is not glycosylated);
TMP: the TPO-mimetic peptide having the amino acid sequence IEGPTLRQWLAARA (SEQ ID NO: 13);
TMP-TMP: the TPO-mimetic peptide having the amino acid sequence IEGPTLRQWLAARA-GGGGGGGG-IEGPTLRQWLAARA (SEQ ID NO: 14);
PEG-TMP-TMP: the peptide of SEQ ID NO: 14, wherein the PEG
group is a 5 kD average molecular weight PEG attached as shown in Figure 6;
Fc TMP-TMP: the compound of SEQ ID NO: 8 (Figure 8) dimerized with an identical second monomer (i.e., Cys residues 7 and 10 are bound to the corresponding Cys residues in the second monomer to form a dimer, as shown in Figure 2); and TMP-TMP-Fc is the compound of SEQ ID NO: 10 (Figure 9) dimerized in the same way as TMP-TMP-Fc except that the Fc domain is attached at the C-terminal end rather than the N-terminal end of the TMP-TMP peptide.

Figure 12 shows the number of platelets generated in vivo in normal BDF1 mice treated with various compounds delivered via implanted osmotic pumps over a 7-day period. The compounds are as defined for Figure 7.
Figure 13 shows the nucleotide and amino acid sequences (SEQ. ID.
NOS: 15 and 16, respectively) of the molecule identified as "Fc-EMP" in Example 3 hereinafter.
Figure 14 shows the nucleotide and amino acid sequences (SEQ ID
NOS: 17 and 18, respectively) of the molecule identified as "EMP-Fc" in Example 3 hereinafter.
Figure 15 shows the nucleotide and amino acid sequences (SEQ ID
NOS:19 and 20, respectively) of the molecule identified as "EMP-EMP-Fc"
in Example 3 hereinafter.
Figure 16 shows the nucleotide and amino acid sequences (SEQ ID
NOS: 21 and 22, respectively) of the molecule identified as "Fc-EMP-EMP"
in Example 3 hereinafter.
Figures 17A and 17B show the DNA sequence (SEQ ID NO: 23) inserted into pCFM1656 between the unique AatU (position #4364 in pCFM1656) and SacU (position #4585 in pCFMI656) restriction sites to form expression plasmid pAMG21 (ATCC accession no. 98113).
Figure 18A shows the hemoglobin, red blood cells, and hematocrit generated in vivo in normal female BDF1 mice treated with one 100 g/kg bolus injection of various compounds. Figure 18B shows the same results with mice treated with 100 jig/kg per day delivered 9e by 7-day micro-osmotic pump with the EMPs delivered at 100 g/kg, rhEPO at 30U/mouse. (In both experiments, neutrophils, lymphocytes, and platelets were unaffected.) In these figures, the terms are defined as follows.
Fc-EMP: the compound of SEQ ID NO: 16 (Figure 13) dimerized with an identical second monomer (i.e., Cys residues 7 and 10 are bound to the corresponding Cys residues in the second monomer to form a dimer, as shown in Figure 2);
EMP-Fc: the compound of SEQ ID NO: 18 (Figure 14) dimerized in the same way as Fc-EMP except that the Fc domain is attached at the C-terminal end rather than the N-terminal end of the EMP
peptide.
"EMP-EMP-Fc" refers to a tandem repeat of the same peptide (SEQ
ID NO: 20) attached to the same Fc domain by the carboxyl terminus of the peptides. "Fc-EMP-EMP" refers to the same tandem repeat of the peptide but with the same Fc domain attached at the amino terminus of the tandem repeat. All molecules are expressed in E. coli and so are not glycosylated.
Figures 19A and 19B show the nucleotide and amino acid sequences (SEQ ID NOS: 1055 and 1056) of the Fc-TNF-a inhibitor fusion molecule described in Example 4 hereinafter.
Figures 20A and 20B show the nucleotide and amino acid sequences (SEQ ID NOS: 1057 and 1058) of the TNF-a inhibitor-Fc fusion molecule described in Example 4 hereinafter.
Figures 21A and 21B show the nucleotide and amino acid sequences (SEQ ID NOS: 1059 and 1060) of the Fc-IL-1 antagonist fusion molecule described in Example 5 hereinafter.
Figures 22A and 22B show the nucleotide and amino acid sequences (SEQ ID NOS: 1061 and 1062) of the IL-1 antagonist-Fc fusion molecule described in Example 5 hereinafter.
Figures 23A, 23B, and 23C show the nucleotide and amino acid sequences (SEQ ID NOS: 1063 and 1064) of the Fc-VEGF antagonist fusion molecule described in Example 6 hereinafter.

Figures 24A and 24B show the nucleotide and amino acid sequences (SEQ ID NOS: 1065 and 1066) of the VEGF antagonist-Fc fusion molecule described in Example 6 hereinafter.
Figures 25A and 25B show the nucleotide and amino acid sequences (SEQ ID NOS: 1067 and 1068) of the Fc-MMP inhibitor fusion molecule described in Example 7 hereinafter.
Figures 26A and 26B show the nucleotide and amino acid sequences (SEQ ID NOS: 1069 and 1070) of the MMP inhibitor-Fc fusion molecule described in Example 7 hereinafter.
Detailed Description of the Invention Definition of Terms The terms used throughout this specification are defined as follows, unless otherwise limited in specific instances.
The term "comprising" means that a compound may include additional amino acids on either or both of the N- or C- termini of the given sequence. Of course, these additional amino acids should not significantly interfere with the activity of the compound.
The term "vehicle" refers to a molecule that prevents degradation and/or increases half-life, reduces toxicity, reduces immunogenicity, or increases biological activity of a therapeutic protein. Exemplary vehicles include an Fc domain (which is preferred) as well as a linear polymer (e.g., polyethylene glycol (PEG), polylysine, dextran, etc.); a branched-chain polymer (see, for example, U.S. Patent No. 4,289,872 to Denkenwalter et al., issued September 15,1981; 5,229,490 to Tam, issued July 20,1993; WO
93/21259 by Frechet et al., published 28 October 1993); a lipid; a cholesterol group (such as a steroid); a carbohydrate or oligosaccharide; or any natural or synthetic protein, polypeptide or peptide that binds to a salvage receptor. Vehicles are further described hereinafter.

The term "native Fc" refers to molecule or sequence comprising the sequence of a non-antigen-binding fragment resulting from digestion of whole antibody, whether in monomeric or multimeric form. The original immunoglobulin source of the native Fc is preferably of human origin and may be any of the immunoglobulins, although IgGI and IgG2 are preferred. Native Fc's are made up of monomeric polypeptides that may be linked into dimeric or multimeric forms by covalent (i.e., disulfide bonds) and non-covalent association. The number of intermolecular disulfide bonds between monomeric subunits of native Fc molecules ranges from 1 to 4 depending on class (e.g., IgG, IgA, IgE) or subclass (e.g., IgGi, IgG2, IgG3, IgA1, IgGA2). One example of a native Fc is a disulfide-bonded diner resulting from papain digestion of an IgG (see Ellison et al.
(1982), Nucleic Acids Res. 10: 4071-9). The term "native Fc" as used herein is generic to the monomeric, dimeric, and multimeric forms.
The term "Fc variant" refers to a molecule or sequence that is modified from a native Fc but still comprises a binding site for the salvage receptor, FcRn. International applications WO 97/34631 (published 25 September 1997) and WO 96/32478 describe exemplary Fc variants, as well as interaction with the salvage receptor, and are hereby incorporated by reference. Thus, the term "Fc variant" comprises a molecule or sequence that is humanized from a non-human native Fc. Furthermore, a native Fc comprises sites that may be removed because they provide structural features or biological activity that are not required for the fusion molecules of the present invention. Thus, the term "Fc variant" comprises a molecule or sequence that lacks one or more native Fc sites or residues that affect or are involved in (1) disulfide bond formation, (2) incompatibility with a. selected host cell (3) N-terminal heterogeneity upon expression in a selected host cell, (4) glycosylation, (5) interaction with complement, (6) binding to an Fc receptor other than a salvage receptor, or (7) antibody-dependent cellular cytotoxicity (ADCC). Fc variants are described in further detail hereinafter.
The term "Fc domain" encompasses native Fc and Fc variant molecules and sequences as defined above. As with Fc variants. and native Fc's, the term "Fc domain" includes molecules in monomeric or multimeric form, whether digested from whole antibody or produced by other means.
The term "multimer" as applied to Fc domains or molecules comprising Fc domains refers to molecules having two or more polypeptide chains associated covalently, noncovalently, or by both covalent and non-covalent interactions. IgG molecules typically form dimers; IgM, pentamers; IgD, dieters; and IgA, monomers, dimers, trimers, or tetramers. Multimers may be formed by exploiting the sequence and resulting activity of the native Ig source of the Fc or by derivatizing (as defined below) such a native Fc, The term "diner" as applied to Fc domains or molecules.
comprising Fc domains refers to molecules having two polypeptide chains associated covalently or non-covalently. Thus, exemplary dieters within the scope of this invention are as shown in Figure 2.
The terms "derivatizing" and "derivative" or "derivatized"
comprise processes and resulting compounds respectively in which (1) the compound has a cyclic portion; for example, cross-linking between cysteinyl residues within the compound; (2) the compound is cross-linked or has a cross linking site; for example, the compound has a cysteinyl residue and thus forms cross-linked dimers in culture or in vivo; (3) one or more peptidyl linkage is replaced by a non-peptidyl linkage; (4) the N-terminus is replaced by NRR', NRC(O)R', -NRC(O)OR', -NRS(O)2R', -NHC(O)NHR, a succinimide group, or substituted or unsubstituted benzyloxycarbonyl-NH-, wherein R and R' and the ring substituents are A I vV ir- -4AN% f'^*A7 oh.o I

as defined hereinafter; (5) the C-terminus is replaced by -C(O)R2 or -NR3R`
wherein R2, R3 and R` are as defined hereinafter; and (6) compounds in which individual amino acid moieties are modified through treatment with agents capable of reacting with selected side chains or terminal residues. Derivatives are further described hereinafter.
The term "peptide" refers to molecules of 2 to 40 amino acids, with molecules of 3 to 20 amino acids preferred and those of 6 to 15 amino acids most preferred. Exemplary peptides may be randomly generated by any of the methods cited above, carried in a peptide library (e.g., a phage display library), or derived by digestion of proteins.
The term "randomized" as used to refer to peptide sequences refers to fully random sequences (e.g., selected by phage display methods) and sequences in which one or more residues of a naturally occurring molecule is replaced by an amino acid residue not appearing in that position in the naturally occurring molecule. Exemplary methods for identifying peptide sequences include phage display, E. coli display, ribosome display, RNA-peptide screening, chemical screening, and the like.
The term "pharmacologically active" means that a substance so described is determined to have activity that affects a medical parameter (e.g., blood pressure, blood cell count, cholesterol level) or disease state (e.g., cancer, autoimmune disorders). Thus, pharmacologically active peptides comprise agonistic or mimetic and antagonistic peptides as defined below.
The terms "-mimetic peptide" and "-agonist peptide" refer to a peptide having biological activity comparable to a protein (e.g., EPO, TPO, G-CSF) that interacts with a protein of interest. These terms further include peptides that indirectly mimic the activity of a protein of interest, such as by potentiating the effects of the natural ligand of the protein of interest; see, for example, the G-CSF-mimetic peptides listed in Tables 2 and 7. Thus, the term "EPO-mimetic peptide" comprises any peptides that .can be identified or derived as described in Wrighton et al. (1996), Science 273: 458-63, Naranda et al. (1999), Proc. Natl. Acad. Sci. USA 96: 7569-74, or any other reference in Table 2 identified as having EPO-mimetic subject matter. Those of ordinary skill in the art appreciate that each of these references enables one to select different peptides than actually disclosed therein by following the disclosed procedures with different peptide libraries.
The term "FPO-mimetic peptide" comprises peptides that can be identified or derived as described in Cwirla et al. (1997), Science 276:1696-9, U.S. Pat. Nos. 5,869,451 and 5,932,946 and any other reference in Table 2 identified as having TPO-mimetic subject matter, as well as WO 00/24770.

Those of ordinary skill in the art appreciate that each of these references enables, one to select different peptides than actually disclosed therein by following the disclosed procedures with different peptide libraries.
The term "G-CSF-mimetic peptide" comprises any peptides that can be identified or described in Paukovits et al. (1984), Hoppe-Seylers Z.
Physiol. Chem. 365: 303-11 or any of the references in Table 2 identified as having G-CSF-mimetic subject matter. Those of ordinary skill in the art appreciate that each of these references enables one to select different peptides than actually disclosed therein by following the disclosed .
procedures with different peptide libraries.
The term "CTLA4-mimetic peptide" comprises any peptides that can be identified or derived as described in Fukumoto et al. (1998), Nature Biotech. 16: 267-70. Those of ordinary skill in the art appreciate-that each of these references enables one to select different peptides than actually disclosed therein by following the disclosed procedures with different peptide libraries.
The term "-antagonist peptide" or "inhibitor peptide" refers to a peptide that blocks or in some way interferes with the biological activity of the associated protein of interest, or has biological activity comparable to a known antagonist or inhibitor of the associated protein of interest. Thus, the term "TNF-antagonist peptide" comprises peptides that can be identified or derived as described in Takasaki et al. (1997), Nature Biotech.
15: 1266-70 or any of the references in Table 2 identified as having TNF-antagonistic subject matter. Those of ordinary skill in the art appreciate that each of these references enables one to select different peptides than actually disclosed therein by following the disclosed procedures with different peptide libraries.
The terms "II.-1 antagonist" and "IL-Ira-mimetic peptide"
comprises peptides that inhibit or down-regulate activation of the IL-1 receptor by IL-1. IL-1 receptor activation results from formation of a complex among IL-1, IL-1 receptor, and IL-1 receptor accessory protein.
IL-1 antagonist or IL-1ra-mimetic peptides bind to IL-1, IL-1 receptor, or IL-1 receptor accessory protein and obstruct complex formation among any two or three components of the complex. Exemplary IL-1 antagonist or IL-1ra-mimetic peptides can be identified or derived as described in U.S. Pat. Nos. 5,608,035, 5,786,331, 5,880,096, or any of the references in Table 2 identified as having IL-1ra-mimetic or IL-1 antagonistic subject matter. Those of ordinary skill in the art appreciate that each of these references enables one to select different peptides than actually disclosed therein by following the disclosed procedures with different peptide libraries.
The term "VEGF-antagonist peptide" comprises peptides that can be identified or derived as described in Fairbrother (1998), Biochem. 37:

WO 00/24782 PCT/US99n5044 17754-64, and in any of the references in Table 2 identified as having .VEGF-antagonistic subject matter. Those of ordinary skill in the art appreciate that each of these references enables one to select different peptides than actually disclosed therein by following the disclosed procedures with different peptide libraries.
The term "MMP inhibitor peptide" comprises peptides that can be identified or derived as described in Koivunen (1999), Nature Biotech. 17:
768-74 and in any of the references in Table 2 identified as having MMP
inhibitory subject matter. Those of ordinary skill in the art appreciate that each of these references enables one to select different peptides than actually disclosed therein by following the disclosed procedures with different peptide libraries.
Additionally, physiologically acceptable salts of the compounds of this invention are also encompassed herein. By "physiologically acceptable salts" is meant any salts that are known or later discovered to be pharmaceutically acceptable. Some specific examples are: acetate;
trifluoroacetate; hydrohalides, such as hydrochloride and hydrobromide;
sulfate; citrate; tartrate; glycolate; and oxalate.
Structure of compounds In General. In the compositions of matter prepared in accordance with this invention, the peptide may be attached to the vehicle through the peptide's N-terminus or C-terminus. Thus, the vehicle-peptide molecules of this invention may be described by the following formula I:
I

(X')a F'-(X2)4 wherein:
Fl is a vehicle (preferably an Fc domain);
X' and X2 are each independently selected from -(L')- P', -(L')'CP'-(L2)d -P2; 4L')~-P'4L2)a P2-(L), P3, and -(L1)-P1-(L2)4-P2-(L3) . -P3-(L4)f-P4 P', P2, P3, and P` are each independently sequences of pharmacologically active peptides;
L', L2, L3, and L` are each independently linkers; and a, b, c, d, e, and f are each independently 0 or 1, provided that at least one of aand bis 1.
Thus, compound I comprises preferred compounds of the formulae II
X'-F' and multimers thereof wherein F' is an Fc domain and is attached at the C-terminus of X';
III _ -F'-X2 and multimers thereof wherein F' is an Fc domain and is attached at the N-terminus of X2;

IV

and multimers thereof wherein F' is an Fc domain and is attached at the N-terminus of -(L')-P'; and V
F1-(L')c-P1-(L2)d P2 and multimers thereof wherein F' is an Fc domain and is attached at the N-terminus of -L'-P'-L2-P2.
Peptides. Any number of peptides may be used in conjunction with the present invention. Of particular interest are peptides that mimic the activity of EPO, TPO, growth hormone, G-CSF, GM-CSF, IL-1ra, leptin, CTLA4, TRAIL, TGF-a, and TGF-R. Peptide antagonists are also of interest, particularly those antagonistic to the activity of TNF, leptin, any of the interleukins (IL-1, 2,3,...), and proteins involved in complement activation (e.g., C3b). Targeting peptides are also of interest, including tumor-homing peptides, membrane-transporting peptides, and the like.
All of these-classes of peptides may be discovered by methods described in the references cited in this specification and other references.
Phage display, in particular, is useful in generating peptides for use in the present invention. It has been stated that affinity selection from libraries of random peptides can be used to identify peptide ligands for any site of any gene product. Dedman et al. (1993), J. Biol. Chem. 268:
23025-30. Phage display is particularly well suited for identifying peptides that bind to such proteins of interest as cell surface receptors or any proteins having linear epitopes. Wilson et al. (1998), Can. T. Microbiol. 44:
313-29; Kay it al. (1998), Drug Disc. Today 3: 370-8. Such proteins are extensively reviewed in Herz et al. (1997), 1. Receptor & Signal Transduction Res. 17(5): 671-776.
Such proteins of interest are preferred for use in this invention.
A particularly preferred group of peptides are those that bind to cytokine receptors. Cytokines have recently been classified according to their receptor code. See Inglot (1997), Archivum Immunologiae et Therapiae Experimentalis 45:353-7.
Among these receptors, most preferred are the CKRs (family I in Table 3). The receptor classification appears in Table 3.

e..c.vvan. .... ,sweese I

Table 3-Cytokine Receptors Classified by Receptor Code Cytokines Ii ands Receptor Type family subfamily family subfamily 1. Hematopoietic 1. IL-2, IL-4, IL-7, I. Cytokine R 1. shared yCr cytokines IL-9, IL-13, IL- (CKR) 2. IL-3, IL-5, GM- 2. shared GP 140 CSF pR
3. IL-6, ILA 1, IL- 3. 3.shared RP
12, LIF, OSM, 130 CNTF, leptin (OB) 4. G-CSF, EPO, 4. "single chain" R
TPO, PRL, GH
5. IL-17, HVS-IL- 5. other R`

It. IL-10 ligands IL-10, BCRF-1, II. IL-10 R

III. interferon 1. IFN-al, a2, a4, _ Ill. Interferon R 1. IFNAR
m, t, IFN-Q' 2. IFN 2. IFNGR
IV. IL-1 ligands 1. IL-1a, IL-1j3, IL- IV. IL-1R
1 Ra V. TNF ligands 1. TNF-a, TNF-P V. NGF/TNF R' (LT), FAS1, CD40 L, CD30L, CD27 L
VI. Chemokines 1. a chemokines: VI. Chemokine R 1. CXCR
IL-8, GRO
a, P, T, IF-10, PF-4, SDF-1 -2. p chemokines: 2. CCR
MIP1a, MIP1 MCP-1,2,3,4, RANTES, eotaxin 3. y chemokines: 3. CR
lymphotactin 4. DARC' IL-17R belongs to the CKR family but is not assigned to any of the 4 indicated subjamilies.
Other IFN type I subtypes remain unassigned. Hematopoietic cytokines, IL-10 ligands and interferons do not possess functional intrinsic protein kinases. The signaling molecules for the cytokines are JAK's, STATs and related non-receptor molecules. IL-14, IL-16 and IL-18 have been cloned but according to the receptor code they remain unassigned.
TNF F-receptors use multiple, distinct intracellular molecules for signal transduction including "death domain` of FAS R and 55 kDa TNF-aR that participates in their cytotoxic effects. NGF/TNF
R can bind both NGF and related factors as well as TNF ligands. Chemokine receptors are G
protein-coupled, seven transmembrane (7TM, serpentine) domain receptors.
The Duffy blood group antigen (DARC) is an erythrocyte receptor that can bind several different chemokines. It belongs to the immunoglobulin superfamily but characteristics of its signal transduction events remain unclear.

VII. Growth factors VII. RKF 1. TK sub-family 1.1 SCF, M-CSF, 1.1 IgTK III R
PDGF-AA, AB, BB, FLT-3L, VEGF, SSV-PDGF
1.2 FGFa, FGF(3 1.2 IgTK IV R
1.3 EGF, TGF-a, 1.3 Cysteine-rich VV-F19 (EGF- TM
like) 1.4 IGF-I, IGF-II, 1.4 Cysteine rich Insulin TK-II
1.5 NGF, BDNF, 1.5 Cysteine knot NT-3, NT-49 TK V
2. TGF- 1, 2, 3 2. STK subfamily"
Exemplary peptides for this invention appear in Tables 4 through 20 below. These peptides may be prepared by methods disclosed in the art. Single letter amino acid abbreviations are used. The X in these sequences (and throughout this specification, unless specified otherwise in a particular instance) means that any of the 20 naturally occurring amino acid residues may be present. Any of these peptides may be linked in tandem (i.e., sequentially), with or without linkers, and a few tandem-linked examples are provided in the table. Linkers are listed as "A" and may be any of the linkers described herein. Tandem repeats and linkers are shown separated by dashes for clarity. Any peptide containing a cysteinyl residue may be cross-linked with another Cys-containing peptide, either or both of which may be linked to a vehicle. A few cross-linked examples are provided in the table. Any peptide having more than one Cys residue may form an intrapeptide disulfide bond, as well; see, for example, EPO-mimetic peptides in Table 5. A few examples of intrapeptide disulfide-bonded peptides are specified in the table. Any of these peptides may be derivatized as described herein, and a few 2o derivatized examples are provided in the table. Derivatize3 peptides in The neurotrophic cytokines can associate with NGFITNF receptors also.

the tables are exemplary rather than limiting, as the associated underivatized peptides may be employed in this invention, as well. For derivatives in which the carboxyl terminus may be capped with an amino group, the capping amino group is shown as -NH2. For derivatives in which amino acid residues are substituted by moieties other than amino acid residues, the substitutions are denoted by a, which signifies any of the moieties described in Bhatnagar et al. (1996), T. Med. Chem. 39: 3814-9 and Cuthbertson et al. (1997), T. Med. Chem. 40:2876-82, The J substituent and the Z substituents (Zs, Z6, ...Z1,) are as defined in U.S. Pat. Nos. 5,608,035,5,786,331, and 5,880,096.
For the EPO-mimetic sequences (Table 5), the substituents X2 through X,, and the integer "n" are as defined in WO 96/40772. 'The substituents "T,-"G," and "+" are as defined in Sparks et al. (1996), Proc. Natl. Acad. Sci.
93:
15404. X414 X6, and X, are as defined in U.S. Pat. No. 5,773,569, except that for integrity binding peptides, X,, X2, X3, X,, Xs, X6, X7, and X1 are as defined in International applications WO 95 / 14714, published June 1,1995 and WO 97/08203, published March 6,1997;
'and for VIP-mimetic peptides, X,, X,', XI", X2, X3, X4, Xs, X6 and Z and the integers m and n are as defined in WO
97/40070, published October 30,1997, Xaa and Yaa below are as defined in WO 98/09985, published March 12,1998,. AA,, AA2, AB,, AB2, and AC are as defined in International application WO 98/53842, published December 3,1998.. X1, X2, X3, and X` in Table 17 only are as defined in European application EP 0 911 L STKS may encompass many other TGF P-related factors that remain unassigned.
The protein kinases are intrinsic part of the intracellular domain of receptor kinase family (RKF). The enzymes participate in the signals transmission via the receptors.

393, published April 28,1999. Residues appearing in boldface are D-amino acids.-All peptides are linked through peptide bonds unless otherwise noted. Abbreviations are listed at the end of this specification. In the "SEQ ID NO." column, "NR" means that no sequence listing is required for the given sequence.

Table 4-IL-1 antagonist peptide sequences Sequence/structure SEQ
ID NO:

xxoz,yzexx 907 zlz,ozxlz,zl, 909 ETPFTWEESNAYYWOPYALPL = 215 AcFEWTPGWYQJY 222 AcFEWTPGYWQJY 228 FEWTPSarWYQJY 230 FEWTPvYWQJY 234 FEWTPecGYWQJY _ 235 FEWTPAibYWOJY 236 FEWTSarGYWQJY 237 AcFEWTPGWYQJY 241 FEWTPGW-pY-QJY 242 AcFEWTPGWYJQJY 247 FEWTPSarWYQJY 249 FEWTPecGYWQJY 254 FEWTPAibYWQJY 255 FEWTSarGYWQJY 256 1 Na EWTPGYYQJY 258 .FEWTPNYWQJY 262 YYDG VYWOPYSVQ VMPA ' 787 VKQKWR WYQPYALPL _ 807 DGYDRWRQSGERY_ WQPY L 860 .TANVSSFEWTPGYWQPYALPL 861 QYAEPSALNDWG YWQPYALPL _ 865 SVGEDHNFWTSE YWQPYALPL. 875 DSTFTWEESNAY YWQPYALP.L 891 SWTDYGYWQPYALPISGL. 919 FEWTPecGYWQJY 941 FEWTPAibYWQIY 1 6.- 942 FEWTPSarWYQJY 943 FEWTSarGYWQJY 944 AcFEWTPGWYQJY 948 AcFEWTPGYWQJY 949 INa EWTPGYYQJY 950 FEWTPsYYQJY 954 FEWTPnYYQJY 955 SHLY-Nap-OPYSVQM 956 TLVY-Nap-QPYSLQT 957 RGDY-Nap-QPYSVQS 958 NMVY-Nap-QPYSIQT 959 FEWTPGYYQJ-B a 963 XaaFEWTPGYYQJ-Bpa 964 FEWTPGY-Bpa-QJY 965 AcFEWTPGY-Bpa-QJY 966 AcFEWTPG-Bpa-YQJY 968 AcFE-B -TPGYYQJY 969 AcFE-B -TPGYYQJY 970 a-EWTPGYYQJY 971 Ac6 a-EWTPGYYQJY 972 TI DNTAWYANLVQ TYPO ' 99.1 pY-INap-pY-QJYALPL 1002 AcFEWTPGYWQJY 1008 AcFEWTPGWYQJY 1009 AcFEWTPGYYQJY 1010 AcFEWTPaYWQJY 1011 AcFEWTPaWYQJY 1012 AcFEWTPaYYQJY 1013 AcFEWTPGYWQJY 1018 AcFEWTPGWYQJY 1019 AcFEWTPGYYQJY 1020 AcFEWTPAYWQJY 1021 AcFEWTPAWYQJY 1022 AcFEWTPAYYQJY 1023 Table 5-EPO-mimetic peptide sequences Sequence/structure SEQ
ID NO:

YXCXXGPXTWXCXP-A- (-tee) 86 ~K

YXCXXGPXTWXCXP-A / (a-amine) GGTYSCHFG PLTWVCKPQGG

GGTYSCHFGPLTWVCKPQGG

GGTYSCHFGPLTWVCKPQGGSSK

GGTYSCHFGPLTWVCKPQGGSSK
GGTYSCHFGPLTWVCKPOGGSS (E-tee) 97 ~K

GGTYSCHFGPLTWVCKPQGGSS (a-amine) GGTYSCHFGPLTWVCKPQGGSSK(-A-biotin) 98 Table 6-TPO-mimetic peptide sequences Sequence/structure SEQ
ID NO:

I I
IEGPTLRQWLAARA-A-K BrAc -A-IEGPTLRQWLAARA 29 I

CA 02347131 2004-02-09 ]j Table 7-G-CSF-mimetic peptide sequences Sequence/structure SEQ
ID NO:

EEDaK 100 EEDaK 100 pGIuEDcrK 101 pGIuEDaK 101 IuEDeK 101 PicSDaK 102 PicSDcK 102 PicSD,K 102 Table 8-TNF-antagonist peptide sequences Sequence/structure SEQ
ID NO:

FCRKEMGCY _ 119 AA,-AB, NR
AC

AA,-AB, Table 9-Integrin-binding peptide sequences Sequence/structure SEQ
ID NO:

)~XADDX,X,XeXA 454 NGR NR
GSL NR
RGD NR

RGD NR
NGR NR
GSL NR

Table 10-Selectin antagonist peptide sequences Sequence/structure SEQ
ID NO:

BNSDOGD: cWO_Q0247e2II,J~

Table 11-Antipathogenic peptide sequences Sequeiice/structure SEQ
ID NO:

GFFALtPKIIS 510 AllVHIRLRIIHHIRL 561 RllVK1RLRIIKKIRLIKK 568 c CKCKGFFALIPKIISSPLFKTLLSAVC 580 C IicCRIVIRIRIRLIRIRC 583 C clicCKPGHKARPHIIRYKlIC 584 C icCRFAVKIRLRIIKKIRLIKKIRKRVIKC 585 Table 12-VIP-mimetic peptide sequences Sequence/structure SEQ
ID NO:

Me HSDAVFYDNYTR LRKQMAVKKYLN SILN 591 X, X' X"X 592 I I
CH2 m Z CH2 n KKYV _ 601 SILauN 602 KKYLNIe 603 LauKKYL 607 CapKKYL 608 KYL NR
KKYNIe 609 SILauN 622 KKYLNIe 625 SILauN 632 LauKKYL 633 CapKKYL 634 KYL - NR
KYL NR
KKYNIe 635 KKYLNIe 639 clicCKKYLC 645 I

Table 13-Mdm/hdm antagonist peptide sequences Sequencelstructure SEQ
ID NO:

Table 14-Calmodulin antagonist peptide sequences Sequence/structure SEQ
ID NO:

CA 02347131 2004-02-09 {{
a) Table 15-Mast cell antagonists/Mast cell protease inhibitor peptide sequences Sequence/structure SEQ
ID NO:

RNR NR
QT NR
RQK NR
NRQ NR
ROK NR

Table 16-SH3 antagonist peptide sequences Sequence/structure SEQ
ID NO:

LXXRPLPX%PP 314 'YXXRPLPXLP 315 PPXOXPPP'VP 316 +PP'YPXKPXWL 317 RPX`YP`YR+SXP 318 'PP'PLP'PK 320 +ODXPLPXLP 321 WO 00/24782 .PCT/US99/25044 Table 17-,Somatostatin or cortistatin mimetic peptide sequences Sequencelstructure SEQ
ID NO:
X'-X -Asn-Phe-Phe-Tr -L s-Thr-Phe-X -Ser-X` 473 Asp Arg Met Pro Cys Arg Asn Phe Phe Trp Lys Thr Phe Ser Ser Cys Lys 474 Met Pro C s Arg Asn Phe Phe Trp Lys Thr Phe Ser Ser Cys Lys 475 Cys Arg Asn Phe Phe Trp Lys Thr Phe Ser Ser C s L .s 476 Asp Arg Met Pro Cys Arg Asn Phe Phe T Lys Thr Phe Ser Ser Cys 477 Met Pro Cys A Asn Phe Phe T Lys Thr Phe Ser Ser Cys 478 Cys Arg Asn Phe Phe T Lys Thr Phe Ser Ser Cy; 479 Asp Arg Met Pro Cys Lys Asn Phe Phe T Lys Thr Phe Ser Ser Cys 480 Met Pro Cys Lys Asn Phe Phe T Lys Thr Phe Ser Ser Cys Lys 481 Cys Lys Asn Phe Phe T Lys Thr Phe Ser Ser Cys Lys 482 Asp A Met Pro Cys Lys Asn Phe Phe T Lys Thr Phe Ser Ser Cys 483 Met Pro Cys Lys Asn Phe Phe T . L Thr Phe Ser Ser Cys 484 Cys Lys Asn Phe Phe T Lys Thr Phe Ser Ser Cys 485 Asp Arg Met Pro Cys Arg Asn Phe Phe T Lys, Thr. Phe Thr Ser Cys Lys 486 Met Pro Cys Arg Asn Phe Phe T Lys Thr Phe Thr Ser Cys Lys .487 Cys A Asn Phe Phe T Lys Thr Phe Thr Ser Cys L s 488 Asp Arg Met Pro Cys Arg Asn Phe Phe T Lys Thr Phe Thr Ser Cys 489 Met Pro Cys Arg Asn Phe Phe T Lys Thr Phe Thr Ser s 490 Cys Arg Asn Phe Phe T Lys Thr Phe Thr Ser Cys 491 Asp Arg Met Pro Cys Lys Asn Phe Phe T Lys Thr Phe Thr Ser Cys L 492 Met Pro Cys Lys Asn Phe Phe T Lys Thr Phe Thr Ser C Lys 493 Cys Lys Asn Phe Phe T Lys Thr Phe Thr Ser C Lys 494 Asp Arg Met Pro Cys Lys Asn Phe Phe T Lys Thr Phe Thr Ser Cys 495 Met Pro Cys Lys Asn Phe Phe T Lys Thr Phe Thr Ser Cys 496 Cys Lys Asn Phe Phe T Lys Thr Phe Thr Ser Cys 497 Table 18-UKR antagonist peptide sequences Sequence/structure SEQ
ID NO:

Table 19-Macrophage and/or T-cell inhibiting peptide sequences Sequence/structure SEQ
ID NO:
Xaa-Yaa-Arg NR
Arg-Yaa-Xaa NR
Xaa-Ar -Yaa NR
Yaa-Arg-Xaa NR
Ala-Ar NR
Ar -A NR
Asn-Ar NR
As -A NR
C s-A NR
Gin-A NR
Glu-A NR
Gly-Arg - NR
His-arg NR
fle-Arg NR
Leu-Arg NR
L s-A NR
Met-Arg NR
Phe-Arg NR
Ser-Arg NR
Thr-A NR
T Ar NR
T -A NR
Val-Arg NR
Ala-GIu-Ar NR
Arg-Glu-Arg NR
Asn-Glu-Arg NR
As GIu-A -NR
Cys-Glu-Arg NR
Gin-GIu-A NR
Glu-Glu-Arg NR
GI -Glu-Ar NR
His-GIu-A NR
Ile-GIu- NR
Leu-Glu-A NR
L s-GIu-A NR
Met-Glu- NR
Phe-Glu-Arg NR
Pro-Glu-Ar NR
Ser-Glu-Arg - NR
Thr-GIu-A NR
T Iu-A NR
T -GIu-A NR
Val-GIu-A NR

Ar -AIa NR
Arg-Asp NR
Arg-Cys NR
Ar -Gin NR
Ar -GIu NR
Arg-Gly NR
Arg-His NR
Ar -IIe NR
Arg-Leu NR
Arg-Lys NR
Arg-Met NR
Arg-Phe NR
A -Pro NR
Arg-Ser NR
Arg-Thr NR
Ar -T NR
Ar -T r NR
Ar -VaI NR
A -GIu-Ala NR
Arg-Glu-Asn NR
A -GIu-As NR
A -GIu-C NR
Ar GIu-GIn NR
A -GIu-GIu NR
Arg-Glu-Gly NR
Arg-Glu-His NR
A -GIu-IIe NR
Ar -GIu-Leu NR
A -GIu-L s NR
Ar -GIu-Met NR
Ar GIu-Phe NR
A -GIu-Pro NR
A -GIu-Ser NR
A -GIu-Thr NR
Arg-Glu-Trp NR
A -GIu-T r NR
A -GIu-VaI NR
Ala-A -Glu NR
Ar Ar -GIu NR
Asn-A -GIu NR
As Ar -GIu NR
C -A -GIu NR
Gin-A -GIu NR
GIu-A -GIu NR
GI -A -GIu NR
His-Ar -GIu - NR
Ile-A -GIu NR
Leu- -GIu NR
L s-A -GIu NR
Met-Ar -GIu NR

Phe-Ar -Glu NR
Pro-A -Gtu NR
Ser-Ar -Glu NR
Thr-Ar -GIu NR
T -A -GIu NR
T r-A -GIu NR
Val-Ar -GIu N`
GIu-Ar -Ala, NR
Glu-Arg-Arg NR
GIu-Ar -Asn NR
Glu-Arg-Asp NR
GIu-A -C s NR
Glu-Ar -Gln NR
Glu-A -Gt NR
GIu-Ar -His NR
GIu-Ar -Ile NR
GIu-Ar -Leu NR
Giu-A -L - NR
Glu-A -Met NR
GIu-A -Phe NR
GIu-A -Pro NR
GIu-A -Ser NR
GIu-A -Thr NR
Glu-A -T NR
Glu-A -T r NR
GIu-A -VaI }QTR

Table 20-Additional Exemplary Pharmacologically Active Peptides Sequence/structure SEQ Activity ID
NO:
VEPNCDIHVMWEWECFERL VEGF-antagonist GE$WCFDGPLTWVCGEES 398-1 VEGF-antagonist WVEICVADDNGMCVTEAQ 1085 VEGF-anta onist GWDECDVARMWEWECFAGV 1086 VEGF- antagonist GERWCFDGPRAWVCGWEI 501 VEGF- antagonist EELWCFDGPRAWVCGYVK 502 VEGF- a nist RGWVEICAADDYGRCLTEAQ 1031 VEGF- antagonist RGWVEICESDVWGRCL 1087 VEGF- ants nist RGWVEICESDVWGRCL 1088 VEGF- antagonist GGNECDIARMWEWECFERL 1089 VEGF- antagonist RGWVEICAADDYGRCL 1090 VEGF-antagonist CTTHWGFTLC _ 1028 MMP inhibitor CLRSGXGC 1091 MMP inhibitor CXXHWGFXXC 1092 MMP inhibitor CXPXC 1093 MMP inhibitor CRRHWGFEFC 1094 MMP inhibitor STTHWGFTLS 1095 MMP inhibitor CSLHWGFWWC 1096 CTLA4-mimetic GFVCSGIFAVGVGRC 125 CTLA4-mimetic APGVRLGCAVLGRYC 126 CTLA4-mimetic LLGRMK 105 Antiviral (HBV) ICVVQDWGHHRCTAGHM LTSHASAI 127 C3b antagonist ICWQDWGHHRCT 128 C3b antagonist CWODWGHHAC 129 C3b antagonist STGGFDDVYDWARGVSSALTTTLVATR, 185 Vinculin-binding STGGFDDVYDWARRVSSAL LVATR. 186 Vinculin-binding SRGVNFSEWLYDMSAAM SNVFPS RSR 187 "Vinculin-binds SSQNWDMEAGVEDLTAAM GLLSTIH. ,SR 188 Vinculin-binds SSPSLYTQFLVNYESAATRIQDLLIASRP$R 189 Vinculin-binding SSTGWVDLLGALQRAADATRTSIPPSLONSR 190 Vinculin-binds DVYTKKEUECARRVSEK - 191 Vinculin-binds EKGSYYPGSGIAQFHIDYNNVS 192 C41912-binding SGIAQFHIDYNNVSSAEG HVN 193 C41313-binds LVTVEKGSYYPGSGIAQFHIDYNNVSSAE WHVN 194 C4BP-bindi.
SGIAQFHIDYNNVS 195 C4BP-binding LLGRMK 279 anti-HBV
ALLGRMKG 280 anti-HBV
LDPAFR 281 anti-HBV
CXXRGDC 322 Inhibition of pi let a tion RPLPPLP 323 Src antagonist PPVPPR 324 Src antagonist XFXDXWXXLXX 325 Anti-cancer (particularly for sarcomas) KACRRLFGPVDSEQLSRDCD 326 16-mimetic RERWNFDFVTETPLEGDFAW 327 16-mimetic .KRRQTSMTDFYHSKRRLIFS 328 p16-mimetic TSMTDFYHSKRRLIFSKRKP 329 16-mimetic RRLIF 330 pl KRRQTSATDFYHSKRRLIFSRQIKIWFQNRRMKWKK 331 p16-Mimetic KRRLIFSKRQIKIWFQNRRMKWKK 332 16-mimetic Asn Gin Gly Arg His Phe Cys Gly Gly Ala Leu Ile His Ala 498 CAP37 mimetic/LPS
Arg Phe Val Met Thr Ala Ala Ser Cys Phe Gin bindin Arg His Phe Cys Gly Gly Ala Leu Ile His Ala Arg Phe Val 499 CAP37 mimetic/LPS
Met Thr Ala Ala Ser Cys binding Gly Thr Arg Cys Gin Val Ala Gly Trp Gly Ser Gin Arg Ser 500 CAP37 mimetic/LPS
Gly Gly Arg Leu Ser Arg Phe Pro Arg Phe Val Asn Val binding WHWRHRIPLQLAAGR 1097 carbohydrate (GD1 alpha) mimetic LKTPRV 1098 2GPI Ab binding NTLKTPRV 1099 2GPI Ab binding NTLKTPRVGGC 1100 2GPI Ab binding KDKATF 1101 2GPI Ab binding KDKATFGCHD 1102 2GPI Ab binding KDKATFGCHDGC 1103 2GPI Ab bindi TLRVVK 1104 2GPI Ab binding ATLRVYKGG 1105 2GPI Ab bindi CATLRVYKGG 1106 2GPI Ab binding INLKALAALAKKIL 1107 Membrane-transporting GWT NR Membrane-_____________________________________________ tnans rti GWTLNSAGYLLG 1108 Membrane-transporting GWTLNSAGYLLGKINLKALAALAKKIL 1109 Membrane-tti The present invention is also particularly useful with peptides having activity in treatment of:
= cancer, wherein the peptide is a VEGF-mimetic or a VEGF receptor antagonist, a HER2 agonist or antagonist, a CD20 antagonist and the like;
= asthma, wherein the protein of interest is a CKR3 antagonist, an IL-5 receptor antagonist, and the like;
= thrombosis, wherein the protein of interest is a GPIIb antagonist, a GPIIIa antagonist, and the like;

= autoimmune diseases and other conditions involving immune modulation, wherein the protein of interest is an IL-2 receptor antagonist, a CD40 agonist or antagonist, a CD40L agonist or antagonist, a thymopoietin mimetic and the like.

Vehicles. This invention requires the presence of at least one vehicle (F', F2) attached to a peptide through the N-terminus, C-terminus or a sidechain of one of the amino acid residues. Multiple vehicles may also be used; e.g., Fc's at each terminus or an Fc at a terminus and a PEG group at the other terminus or a sidechain.
An Fc domain is the preferred vehicle. The Fc domain may be fused to the N or C termini of the peptides or at both the N and C termini. For the TPO-mimetic peptides, molecules having the Fc domain fused to the N
terminus of the peptide portion of the molecule are more bioactive than other such fusions, so fusion to the N terminus is preferred.
As noted above, Fc variants are suitable vehicles within the scope of this invention. A native Fc may be extensively modified to form an Fc variant in accordance with this invention, provided binding to the salvage receptor is maintained; see, for example WO 97/34631 and WO 96/32478.
In such Fc variants, one may remove one or more sites of a native Fc that provide structural features or functional activity not required by the fusion molecules of this invention. One may remove these sites by, for example, substituting or deleting residues, inserting residues into the site, or truncating portions containing the site. The inserted or substituted residues may also be altered amino acids, such as peptidomimetics or D-amino acids. Fc variants may be desirable for a number of reasons, several of which are described below. Exemplary Fc variants include molecules and sequences in which: -1. Sites involved in disulfide bond formation are removed. Such removal may avoid reaction with other cysteine-containing proteins present in the host cell used to produce the molecules of the invention. For this purpose,-the cysteine-containing segment at the N-terminus may be truncated or cysteine residues may be deleted or substituted with other amino acids (e.g., alanyl, seryl). In particular, one may truncate the N-terminal 20-amino acid segment of SEQ ID NO. 2 or delete or substitute the cysteine residues at positions 7 and 10 of SEQ ID NO: 2.
Even when cysteine residues are removed, the single chain Fc domains can still form a dimeric Fc domain that is held together non-covalently.
2. A native Fc is modified to make it more compatible with a selected host cell. For example, one may remove the PA sequence near the N-terminus of a typical native Fc, which may be recognized by a digestive enzyme in E. coli such as proline iminopeptidase. One may also add an N-terminal methionine residue, especially when the molecule is expressed recombinantly in a bacterial cell such as E. coli. The Fc domain of SEQ ID NO: 2 (Figure 4) is one such Fc variant.
3. A portion of the N-terminus of a native Fc is removed to prevent N-terminal heterogeneity when expressed in a selected host cell. For this purpose, one may delete any of the first 20 amino acid residues at the N-terminus, particularly those at positions 1, 2, 3, 4 and 5.
4. One or more glycosylation sites are removed. Residues that are typically glycosylated (e.g., asparagine) may confer cytolytic response.
Such residues may be deleted or substituted with unglycosylated residues (e.g., alanine).
5. Sites involved in interaction with complement, such as the Clq binding site, are removed. For example, one may delete or substitute the EKK
sequence of human IgG1. Complement recruitment may not be advantageous for the molecules of this invention and so may be avoided with such an Fc variant.

6. Sites are removed that affect binding to Fc receptors other than a salvage receptor. A native Fc may have sites for interaction with certain white blood cells that are not required for the fusion molecules of the present invention and so may be removed.
7. The ADCC site is removed. ADCC sites are known in the art; see, for example, Molec. Immunol. 29 (5): 633-9 (1992) with regard to ADCC
sites in IgG1. These sites, as well, are not required for the fusion molecules of the present invention and so may be removed.
8. When the native Fc is derived from a non-human antibody, the native Fc may be humanized. Typically, to humanize a native Fc, one will substitute selected residues in the non-human native Fc with residues that are normally found in human native Fc. Techniques for antibody humanization are well known in the art.
Preferred Fc variants include the following. In SEQ ID NO: 2 (Figure 4) the leucine at position 15 may be substituted with glutamate; the glutamate at position 99, with alanine; and the lysines at positions 101-and 103, with alanines. In addition, one or more tyrosine residues can be replaced by phenyalanine residues.
An alternative vehicle would be a protein, polypeptide, peptide, antibody, antibody fragment, , or small molecule (e.g., a peptidomimetic compound) capable of binding to a salvage receptor. For example, one could use as a vehicle a polypeptide as described in U.S. Pat. No. 5,739,277, issued April 14,1998 to Presta et al. Peptides could also be selected by phage display for binding to the FcRn salvage receptor. Such salvage receptor-binding compounds are also included within the meaning of "vehicle" and are within the scope of this invention. Such vehicles should be selected for increased half-life (e.g., by avoiding sequences recognized by proteases) and decreased immunogenicity (e.g., by favoring non immunogenic sequences, as discovered in antibody humanization).

As noted above, polymer vehicles may also be used for F' and F2.
Various means for attaching chemical moieties useful as vehicles are currently available, see, e.g., Patent Cooperation Treaty ("PCT") International Publication No. WO 96/11953, entitled "N-Terminally Chemically Modified Protein Compositions and Methods."
This PCT publication discloses, among other things, the selective attachment of water soluble polymers to the N-terminus of proteins.
A preferred polymer vehicle is polyethylene glycol (PEG). The PEG
group may be of any convenient molecular weight and may be linear or branched. The average molecular weight of the PEG will preferably range from about 2 kiloDalton ("kD") to about 100 kDa, more preferably from about 5 kDa to about 50 kDa, most preferably from about 5 kDa to about 10 kDa. The PEG groups will generally be attached to the compounds of the invention via acylation or reductive alkylation through a reactive group on the PEG moiety (e.g., an aldehyde, amino, thiol, or ester group) to a reactive group on the inventive compound (e.g., an aldehyde, amino, or ester group).
A useful strategy for the PEGylation of synthetic peptides consists of combining, through forming a conjugate linkage in solution, a peptide and a PEG moiety, each bearing a special functionality that is mutually reactive toward the other. The peptides can be easily prepared with conventional solid phase synthesis (see, for example, Figures 5 and 6 and the accompanying text herein). The peptides are "preactivated" with an appropriate functional group at a specific site. The precursors are purified and fully characterized prior to reacting with the PEG moiety. Ligation of the peptide with PEG usually takes place in aqueous phase and can be easily monitored by reverse phase analytical HPLC. The PEGylated peptides can be easily purified by preparative HPLC and characterized by analytical HPLC, amino acid analysis and laser desorption mass spectrometry.
Polysaccharide polymers are another type of water soluble polymer which may be used for protein modification. Dextrans are polysaccharide polymers comprised of individual subunits of glucose predominantly linked by al-6 linkages. The dextran itself is available in many molecular weight ranges, and is readily available in molecular weights from about 1 kD to about 70 kD. Dextran is a suitable water soluble polymer for use in the present invention as a vehicle by itself or in combination with another vehicle (e.g., Fc). See, for example, WO 96 / 11953 and WO 96/05309. The use of dextran conjugated to therapeutic or diagnostic immunoglobulins has been reported; see, for example, European Patent Publication No. 0 315 456', Dextran of about 1 kD
to about 20 kD is preferred when dextran is used as a vehicle in accordance with the present invention.
Linkers. Any "linker" group is optional. When present, its chemical structure is not critical, since it serves primarily as a spacer. The linker is preferably made up of amino acids linked together by peptide bonds.
Thus, in preferred embodiments, the linker is made up of from 1 to 20 amino acids linked by peptide bonds, wherein the amino acids are selected from the 20 naturally occurring amino acids. Some of these amino acids may be glycosylated, as is well understood by those in the art. In a more preferred embodiment, the 1 to 20 amino acids are selected from glycine, alanine, proline, asparagine, glutamine, and lysine. Even more preferably, a linker is made up of a majority of amino acids that are sterically unhindered, such as glycine and alanine. Thus, preferred linkers are polyglycines (particularly (Gly),, (Gly)5), poly(Gly-Ala), 'anzl polyalanines.
Other specific examples of linkers are:
(Gly)3Lys(Gly)4 (SEQ ID NO: 333);

(Gly),AsnGlySer(Gly)2 (SEQ ID NO: 334);
(Gly),Cys(Gly), (SEQ ID NO: 335); and GlyProAsnGlyGly (SEQ ID NO: 336).
To explain the above nomenclature, for example, (Gly)3Lys(Gly)4 means Gly-Gly-Gly-Lys-Gly-Gly-Gly-Gly. Combinations of Gly and Ala are also preferred. The linkers shown here are exemplary; linkers within the scope of this invention may be much longer and may include other residues.
Non peptide linkers are also possible. For example, alkyl linkers such as -NH-(CH2),-C(O)-, wherein s = 2-20 could be used. These alkyl linkers may further be substituted by any non-sterically hindering group such as lower alkyl (e.g., C1-C6) lower acyl, halogen (e.g., Cl, Br), CN, NI-4, phenyl, etc. An exemplary non-peptide linker is a PEG linker, VI
O
\N ~ n wherein n is such that the linker has a molecular weight of 100 to 5000 kD, preferably 100 to 500 kD. The peptide linkers may be altered to form derivatives in the same manner as described above.
Derivatives. The inventors also contemplate derivatizing the peptide and/or vehicle portion of the compounds. Such derivatives may improve the solubility, absorption, biological half life, and the like of the compounds. The moieties may alternatively eliminate or attenuate any undesirable side-effect of the compounds and the like. Exemplary derivatives include compounds in which:
1. The compound or some portion thereof is cyclic. For example, the peptide portion may be modified to contain two or more Cys residues (e.g., in the linker), which could cyclize by disulfide bond formation.

For citations to references on preparation of cyclized derivatives, see Table 2.
2. The compound is cross-linked or is rendered capable of cross-linking between molecules. For example, the peptide portion may be modified to contain one Cys residue and thereby be able to form an intermolecular disulfide bond with a like molecule. The compound may also be cross-linked through its C-terminus, as in the molecule shown below.
VII

H O
F' -(X' )b-Co -N NH2 F' -(X' )b-Co-N NH

3. O
4. One or more peptidyl [-C(O)NR-] linkages (bonds) is replaced by a non-peptidyl linkage. Exemplary non-peptidyl linkages are -CH2-carbamate [-CH2-OC(O)NR-], phosphonate, -CH2-sulfonamide [-CH2-S(O)2NR ], urea [-NHC(O)NH-1, -CH2-secondary amine, and alkylated peptide [-C(O)NK`- wherein R6 is lower alkyll.
5. The N-terminus is derivatized. Typically, the N-terminus may be acylated or modified to a substituted amine. Exemplary N-terminal derivative groups include -NRR' (other than -NH2), -NRC(O)R1, NRC(O)OR', -NRS(O)2R', -NHC(O)NHR', succinimide, or benzyloxycarbonyl-NH- (CBZ-NH-), wherein R and R' are each independently hydrogen or lower alkyl and wherein the phenyl ring may be substituted with 1 to 3 substituents selected from the group consisting of C1-C4 alkyl, C1-C4 alkoxy, chloro, and bromo.
6. The free C-terminus is derivatized. Typically, the C-terminus is esterified or amidated. For example, one may use methods described in the art to add (NH-CHZ-CHZ NH2), to compounds of this invention WO 00/24782 PCT/US99n5044 having any of SEQ ID NOS: 504 to 508 at the C-terminus. Likewise, one may use methods described in the art to add -NH2 to compounds of this invention having any of SEQ ID NOS: 924 to 955, 963 to 972, 1005 to 1013, or 1018 to 1023 at the C-terminus. Exemplary C-terminal derivative groups include, for example, -C(O)R2 wherein R2 is lower alkoxy or -NR3R' wherein R3 and R4 are independently hydrogen or C;
CB alkyl (preferably C1-C4 alkyl).
7. A disulfide bond is replaced with another, preferably more stable, cross-linking moiety (e.g., an alkylene). See, e.g., Bhatnagar et al.
(1996), T. Med. Chem. 39:3814-9; Alberts et al. (1993) Thirteenth Am.
Pep. S mp., 357-9.
8. One or more individual amino acid residues is modified. Various derivatizing agents are known to react specifically with selected sidechains or terminal residues, as described in detail below.
Lysinyl residues and amino terminal residues may be reacted with succinic or other carboxylic acid anhydrides, which reverse the charge of the lysinyl residues. Other suitable reagents for derivatizing alpha-amino-containing residues include imidoesters such as methyl picolinimidate;
pyridoxal phosphate; pyridoxal; chloroborohydride; trinitrobenzenesulfonic acid; O-methylisourea; 2,4 pentanedione; and transaminase-catalyzed reaction with glyoxylate.
Arginyl residues may be modified by reaction with any one or combination of several conventional reagents, including phenylglyoxal, 2,3-butanedione, 1,2-cyclohexanedione, and ninhydrin. Derivatization of arginyl residues requires that the reaction be performed in alkaline conditions because of the high pKa of the guanidine functional group. Furthermore, these reagents may react with the groups of lysine as well as the argiriincrepsilon-amino group.

Specific modification of tyrosyl residues has been studied extensively, with particular interest in introducing spectral labels into tyrosyl residues by reaction with aromatic diazonium compounds or tetranitromethane. Most commonly, N-acetylimidizole and tetranitromethane are used to form O-acetyl tyrosyl species and 3-nitro derivatives, respectively.
Carboxyl sidechain groups (aspartyl or glutamyl) may be selectively modified by reaction with carbodiimides (R'-N=C=N-R') such as 1-cyclohexyl-3-(2-morpholinyl-(4-ethyl) carbodiimide or 1-ethyl-3-(4-azonia-4,4-dimethylpentyl) carbodiimide. Furthermore, aspartyl and glutamyl residues may be converted to asparaginyl and glutaminyl residues by reaction with ammonium ions.
Glutaminyl and asparaginyl residues may be deamidated to the corresponding glutamyl and aspartyl residues. Alternatively, these residues are deamidated under mildly acidic conditions. Either form of these residues falls within the scope of this invention.
Cysteinyl residues can be replaced by amino acid residues or other moieties either to eliminate disulfide bonding or, conversely, to stabilize cross-linking. See, e.g., Bhatnagar et al. (1996), T. Med. Chem. 39:3814-9.
Derivatization with bifunctional agents is useful for cross-linking the peptides or their functional derivatives to a water-insoluble support matrix or to other macromolecular vehicles. Commonly used cross-linking agents include, e.g., 1,1-bis(diazoacetyl)2 phenylethane, glutaraldehyde, N-hydroxysucciniunide esters, for example, esters with 4-azidosalicylic acid, homobifunctional imidoesters, including disuccinimidyl esters such as 3,3'-dithiobis(succinimidylpropionate), and bifunctional maleimides such as bis-N-maleimido-1,8-octane. Derivatizing agents such as methyl-3-[(p-azidophenyl)dithio]propioimidate yield photoactivatable' intermediates that are capable of forming crosslinks in the presence of light. Alternatively, reactive water-insoluble matrices such as cyanogen bromide-activated carbohydrates and the reactive substrates described in U.S. Pat. Nos. 3,969,287; 3,691,016;
4,195,128; 4,247,642; 4,229,537; and 4,330,440 are employed for protein immobilization.
Carbohydrate (oligosaccharide) groups may conveniently be attached to sites that are known to be glycosylation sites in proteins.
Generally, O-linked oligosaccharides are attached to serine (Ser) or threonine (Thr) residues while N-linked oligosaccharides are attached to asparagine (Asn) residues when they are part of the sequence Asn-X-Ser/Thr, where X can be any amino acid except proline. X is preferably one of the 19 naturally occurring amino acids other than proline. The structures of N-linked and O-linked oligosaccharides and the sugar residues found in each type are different. One type of sugar that is commonly found on both is N-acetylneuraminic acid (referred to as sialic acid). Sialic acid is usually the terminal residue of both N-linked and 0-linked oligosaccharides and, by virtue of its negative charge, may confer acidic properties to the glycosylated compound. Such site(s) may be incorporated in the linker of the compounds of this invention and are preferably glycosylated by a cell during recombinant production of the polypeptide compounds (e.g., in mammalian cells such as CHO, BHK, COS). However, such sites may further be glycosylated by synthetic or semi-synthetic procedures known in the art.
Other possible modifications include hydroxylation of proline and lysine, phosphorylation of hydroxyl groups of seryl or threonyl residues, oxidation of the sulfur atom in Cys, methylation of the alpha amino groups of lysine, arginine, and histidine side chains. Creighton, Proteins:
Structure and Molecule Properties (W. H. Freeman & Co., San Francisco), pp. 79-86 (1983). - -Compounds of the present invention may be changed at the DNA
level, as well. The DNA sequence of any portion of the compound may be changed to codons more compatible with the chosen host cell. For E. coli, which is the preferred host cell, optimized codons are known in the art.
Codons may be substituted to eliminate restriction sites or to include silent restriction sites, which may aid in processing of the DNA in the. selected host cell. The vehicle, linker and peptide DNA sequences may be modified to include any of the foregoing sequence changes.

Methods of Making The compounds of this invention largely may be made in transformed host cells using recombinant DNA techniques. To do so, a recombinant DNA molecule coding for the peptide is prepared. Methods of preparing such DNA molecules are well known in the art. For instance, sequences coding for the peptides could be excised from DNA using suitable restriction enzymes. Alternatively, the DNA molecule could be synthesized using chemical synthesis techniques, such as the phosphoramidate method. Also, a combination of these techniques could be used.
The invention also includes a vector capable of expressing the peptides in an appropriate host. The vector comprises the DNA molecule that codes for the peptides operatively linked to appropriate expression control sequences. Methods of effecting this operative linking, either before or after the DNA molecule is inserted into the vector, are well known. Expression control sequences include promoters, activators, enhancers, operators, ribosomal binding sites, start signals, stop signals, cap signals, polyadenylation signals, and other signals involved with the control of transcription or translation.
The resulting vector having the DNA molecule thereon is used to transform an appropriate host. This transformation may lye performed using methods well known in the art.

Any of a large number of available and well-known host cells may be used in the practice of this invention. The selection of a particular host is dependent upon a number of factors recognized by the art. These include, for example, compatibility with the chosen expression vector, toxicity of the peptides encoded by the DNA molecule, rate of transformation, ease of recovery of the peptides, expression characteristics, bio-safety and costs. A balance of these factors must be struck with the understanding that not all hosts may be equally effective for the expression of a particular DNA sequence. Within these general guidelines, lo useful microbial hosts include bacteria (such as E. coli sp.), yeast (such as Saccharomyces sp.) and other fungi, insects, plants, mammalian (including human) cells in culture, or other hosts known in the art.
Next, the transformed host is cultured and purified. Host cells may be cultured under conventional fermentation conditions so that the desired compounds are expressed. Such fermentation conditions are well known in the art. Finally, the peptides are purified from culture by methods well known in the art.
The compounds may also be made by synthetic methods. For example, solid phase synthesis techniques may be used. Suitable techniques are well known in the art, and include those described in Merrifield (1973), Chem. Polytides, pp. 335-61 (Katsoyannis and Panayotis eds.); Merrifield (1963), T. Am. Chem. Soc. 85: 2149; Davis et al.
(1985), Biochem. Intl. 10: 394-414; Stewart and Young (1969), Solid Phase Peptide Synthesis: U.S. Pat. No. 3,941,763; Finn et al. (1976), The Proteins (3rd ed.) 2:105-253; and Erickson et al. (1976), The Proteins (3rd ed.) 2:
257-527. Solid phase synthesis is the preferred technique of making individual peptides since it is-the most cost-effective method of making small peptides.

Compounds that contain derivatized peptides or which contain non-peptide groups may be synthesized by well-known organic chemistry techniques.

Uses of the Compounds In general. The compounds of this invention have pharmacologic activity resulting from their ability to bind to proteins of interest as agonists, mimetics or antagonists of the native ligands of such proteins of interest. The utility of specific compounds is shown in Table 2. The activity of these compounds can be measured by assays known in the art. For the TPO-mimetic and EPO-mimetic compounds, in vivo assays are further described in the Examples section herein.
In addition to therapeutic uses, the compounds of the present invention are useful in diagnosing diseases characterized by dysfunction of their associated protein of interest. In one embodiment, a method of detecting in a biological sample a protein of interest (e.g., a receptor) that is capable of being activated comprising the steps of. (a) contacting the sample with a compound of this invention; and (b) detecting activation of the protein of interest by the compound. The biological samples include tissue specimens, intact cells, or extracts thereof. The compounds of this invention may be used as part of a diagnostic kit to detect the presence of their associated proteins of interest in a biological sample. Such kits employ the compounds of the invention having an attached label to allow for detection. The compounds are useful for identifying normal or abnormal proteins of interest. For the EPO-mimetic compounds, for example, presence of abnormal protein of interest in a biological sample may be indicative of such disorders as Diamond Blackfan anemia, where it is believed that the EPO receptor is dysfunctional.
Therapeutic uses of EPO-mimetic compounds. The EPO-mimetic compounds of the invention are useful for treating disorders characterized by low red blood cell levels. Included in the invention are methods of modulating the endogenous activity of an EPO receptor in a mammal, preferably methods of increasing the activity of an EPO receptor. In general, any condition treatable by erythropoietin, such as anemia, may also be treated by the EPO-mimetic compounds of the invention. These compounds are administered by an amount and route of delivery that is appropriate for the nature and severity of the condition being treated and may be ascertained by one skilled in the art. Preferably, administration is by injection, either subcutaneous, intramuscular, or intravenous.
Therapeutic uses of TPO-mimetic compounds. For the TPO-mimetic compounds, one can utilize such standard assays as those described in W095/26746 entitled "Compositions and Methods for Stimulating Megakaryocyte Growth and Differentiation". In vivo assays also appear in the Examples hereinafter.
The conditions to be treated are generally those that involve an existing megakaryocyte/platelet deficiency or an expected megakaryocyte/platelet deficiency (e.g., because of planned surgery or platelet donation). Such conditions will usually be the result of a deficiency (temporary or permanent) of active Mpl ligand in vivo. The generic term for platelet deficiency is thrombocytopenia, and hence the methods and compositions of the present invention are generally available for treating thrombocytopenia in patients in need thereof.
Thrombocytopenia (platelet deficiencies) may be present for various reasons, including chemotherapy and other therapy with a variety of drugs, radiation therapy, surgery, accidental blood loss, and other specific disease conditions. Exemplary specific disease conditions that involve thrombocytopenia and may be treated in accordance with this invention are: aplastic anemia, idiopathic thrombocytopenia, metastatic tumors which result in thrombocytopenia, systemic lupus erythematosus, splenomegaly, Fanconi's syndrome, vitamin B12 deficiency; folic acid deficiency, May-Hegglin anomaly, Wiskott-Aldrich syndrome, and paroxysmal nocturnal hemoglobinuria. Also, certain treatments for AIDS

WO 00/24782 PCT/US99n5044 result in thrombocytopenia (e.g., AZT). Certain wound healing disorders might also benefit from an increase in platelet numbers.
With regard to anticipated platelet deficiencies, e.g., due to future surgery, a compound of the present invention could be administered several days to several hours prior to the need for platelets. With regard to acute situations, e.g., accidental and massive blood loss, a compound of this invention could be administered along with blood or purified platelets.
The TPO-mimetic compounds of this invention may also be useful in l 0 stimulating certain cell types other than megakaryocytes if such cells are found to express Mpl receptor. Conditions associated with such cells that express the. -Mpl receptor, which are responsive to stimulation by the Mpl ligand, are also within the scope of this invention.
The TPO-mimetic compounds of this invention may be used in any situation in which production of platelets or platelet precursor cells is desired, or in which stimulation of the c-Mpl receptor is desired. Thus, for example, the compounds of this invention may be used to treat any condition in a mammal wherein there is a need of platelets, megakaryocytes, and the like. Such conditions are described in detail in the following exemplary sources:
W095/26746; W095/21919; WO95/18858; W095/21920.

The TPO-mimetic compounds of this invention may also be useful in maintaining the viability or storage life of platelets and/or megakaryocytes and related cells. Accordingly, it could be useful to include an effective amount of one or more such compounds in a composition containing such cells.
The therapeutic methods, compositions and compounds of the present invention may also be employed, alone or in combination with other cytokines, soluble Mpl receptor, hematopoietic factors, interleukins, growth factors or antibodies in the treatment of disease states characterized by other symptoms as well as platelet deficiencies. It is anticipated that the inventive compound will prove useful in treating some forms of thrombocytopenia in combination with general stimulators of hematopoiesis, such as IL-3 or GM-CSF. Other megakaryocytic stimulatory factors, i.e., meg-CSF, stem cell factor (SCF), leukemia inhibitory factor (LIF), oncostatin M (OSM), or other molecules with megakaryocyte stimulating activity may also be employed with Mpl ligand. Additional exemplary cytokines or hematopoietic factors for such co-administration include IL-1 alpha, IL-1 beta, IL-2, IL-3, IL-4, IL-5, IL-6, IL-11, colony stimulating factor-1 (CSF-1), SCF, GM-CSF, granulocyte colony stimulating factor (G-CSF), EPO, interferon-alpha (IFN-alpha), consensus interferon, IFN-beta, or IFN-gamma. It may further be useful to administer, either simultaneously or sequentially, an effective amount of a soluble mammalian Mpl receptor, which appears to have an effect of causing megakaryocytes to fragment into platelets once the megakaryocytes have reached mature form. Thus, administration of an inventive compound (to enhance the number of mature megakaryocytes) followed by administration of the soluble Mpl receptor (to inactivate the ligand and allow the mature megakaryocytes to produce platelets) is expected to be a particularly effective means of stimulating platelet production. The dosage recited above would be adjusted to compensate for such additional components in the therapeutic composition. Progress of the treated patient can be monitored by conventional methods.
In cases where the inventive compounds are added to compositions of platelets and/or megakaryocytes and related cells, the amount to be included will generally be ascertained experimentally by techniques and assays known in the art. An exemplary range of amounts is-0.1 jig-i mg inventive compound per 106 cells.

Pharmaceutical Compositions In General. The present invention also provides methods of using pharmaceutical compositions of the inventive compounds. Such pharmaceutical compositions may be for administration for injection, or for oral, pulmonary, nasal, transdermal or other forms of administration. In general, the invention encompasses pharmaceutical compositions comprising effective amounts of a compound of the invention together with pharmaceutically acceptable diluents, preservatives, solubilizers, emulsifiers, adjuvants and/or carriers. Such compositions include diluents of various buffer content (e.g., Tris-HC1, acetate, phosphate), pH and ionic strength;
additives such as detergents and solubilizing agents (e.g., Tween 80, Polysorbate 80), anti-oxidants (e.g., ascorbic acid, sodium metabisulfite), preservatives (e.g., Thimersol, benzyl alcohol) and bulking substances (e.g., lactose, mannitol); incorporation of the material into particulate preparations of polymeric compounds such as polylactic acid, polyglycolic acid, etc. or into liposomes. Hyaluronic acid may also be used, and this may have the effect of promoting sustained duration in the circulation. Such compositions may influence the physical state, stability, rate of in vivo release, and rate of in vivo clearance of the present proteins and derivatives. See, e.g., Remington's Pharmaceutical Sciences, 18th Ed. (1990, Mack Publishing Co., Easton, PA
18042) pages 1435-1712.. The compositions may be prepared in liquid form, or may be in dried powder, such as lyophilized form. Implantable sustained release formulations are also contemplated, as are transdermal formulations.
Oral dosage forms. Contemplated for use herein are oral solid dosage forms, which are described generally in Chapter 89 of Remington's Pharmaceutical Sciences (1990),18th Ed., Mack PublishingCo. Easton PA
18042. Solid dosage forms include tablets, capsules, pills, troches or lozenges, cachets or pellets.
Also, * Trade-mark 77 liposomal or proteinoid encapsulation may be used to formulate the present compositions (as, for example, proteinoid microspheres reported in U.S. Patent No. 4,925,673). Liposomal encapsulation may be used and the liposomes may be derivatized with various polymers (e.g., U.S. Patent No. 5,013,556). A description of possible solid dosage forms for the therapeutic is given in Chapter 10 of Marshall, K., Modern Pharmaceutics (1979), edited by G. S. Banker and C. T. Rhodesõ
In general, the formulation will include the inventive compound, and inert ingredients which allow for protection against the stomach environment, and release of the biologically active material in the intestine. -Also specifically contemplated are oral dosage forms of the above inventive compounds. If necessary, the compounds may be chemically modified so that oral delivery is efficacious. Generally, the chemical modification contemplated is the attachment of at least one moiety to the compound molecule itself, where said moiety permits (a) inhibition of proteolysis; and (b) uptake into the blood stream from the stomach or intestine. Also desired is the increase in overall stability of the compound and increase in circulation time in the body. Moieties useful as covalently attached vehicles in this invention may also be used for this purpose.
Examples of such moieties include: PEG, copolymers of ethylene glycol and propylene glycol, carboxymethyl cellulose, dextran, polyvinyl alcohol, polyvinyl pyrrolidone and polyproline. See, for example, Abuchowski and Davis, Soluble Polymer-Enzyme Adducts, Enzymes as Drugs (1981), Hocenberg and Roberts, eds., Wiley-Interscience, New York, NY,, pp 367-83; Newmark, et al. (1982), T. Appl. Biochem. 4:185-9. Other polymers that couldbe used are poly-1,3-dioxolane and poly-1,3,6-tioxocane. Preferred for pharmaceutical usage, as indicated above, are PEG moieties.

For oral delivery dosage forms, it is also possible to use a salt of a modified aliphatic amino acid, such as sodium N-(8-[2-hydroxybenzoyl]
amino) caprylate (SNAC), as a carrier to enhance absorption of the therapeutic compounds of this invention. The clinical efficacy of a heparin formulation using SNAC has been demonstrated in a Phase II trial conducted by Emisphere Technologies. See US Patent No. 5,792,451, "Oral drug delivery composition and methods".
The compounds of this invention can be included in the formulation as fine multiparticulates in the form of granules or pellets of particle size about 1 mm. The formulation of the material for capsule administration could also be as a powder, lightly compressed plugs or even as tablets. The therapeutic could be prepared by compression.
Colorants and flavoring agents may all be included. For example, the protein (or derivative) may be formulated (such as by liposome or microsphere encapsulation) and then further contained within an edible product, such as a refrigerated beverage containing colorants and flavoring agents.
One may dilute or increase the volume of the compound of the invention with an inert material. These diluents could include carbohydrates, especially mannitol, a-lactose, anhydrous lactose, cellulose, sucrose, modified dextrans and starch. Certain inorganic salts may also be used as fillers including calcium triphosphate, magnesium carbonate and sodium chloride. Some commercially available diluents are Fast-Float Emden, STA-Rx*1500, Emcompress and Avicell.
Disintegrants may be included in the formulation of the therapeutic into a solid dosage form. Materials used as disintegrants include but are not limited to starch including the commercial disintegrantbased on starch, Explotab. Sodium starch glycolate, Amberlite;` sodium carboxymethylcellulose, ultramylopectin, sodium alginate, gelatin, orange * Trade-mark 79 peel, acid carboxymethyl cellulose, natural sponge and bentonite may all be used-. Another form of the disintegrants are the insoluble cationic exchange resins. Powdered gums may be used as disintegrants and as binders and these can include powdered gums such as agar, Karaya or tragacanth. Alginic acid and its sodium salt are also useful as disintegrants.
Binders may be used to hold the therapeutic agent together to form a hard tablet and include materials from natural products such as acacia, tragacanth, starch and gelatin. Others include methyl cellulose (MC), ethyl cellulose (EC) and carboxymethyl cellulose (CMC). Polyvinyl pyrrolidone (PVP) and hydroxypropylmethyl cellulose (HPMC) could both be used in alcoholic solutions to granulate the therapeutic.
An antifrictional agent may be included in the formulation of the therapeutic to prevent sticking during the formulation process. Lubricants may be used as a layer between the therapeutic and the die wall, and these can include but are not limited to; stearic acid including its magnesium and calcium salts, polytetrafluoroethylene (PTFE), liquid paraffin, vegetable oils and waxes. Soluble lubricants may also be used such as sodium lauryl sulfate, magnesium lauryl sulfate, polyethylene glycol of various molecular weights, Carbowax 4000 and 6000.
Glidants that might improve the flow properties of the drug during formulation and to aid rearrangement during compression might be added. The glidants may include starch, talc, pyrogenic silica and hydrated silicoaluminate.
To aid dissolution of the compound of this invention into the aqueous environment a surfactant might be added as a wetting agent.
Surfactants may include anionic detergents such as sodium tauryl sulfate, dioctyl sodium sulfosuccinate and dioctyl sodium sulfonate. Cationic detergents might be used and could include benzalkonium chloride or * Trade-mark 80 benzethonium chloride. The list of potential nonionic detergents that could-be included in the formulation as surfactants are lauromacrogol 400, polyoxyl 40 stearate, polyoxyethylene hydrogenated castor oil 10, 50 and 60, glycerol monostearate, polysorbate 40, 60, 65 and 80, sucrose fatty acid ester, methyl cellulose and carboxymethyl cellulose. These surfactants could be present in the formulation of the protein or derivative either alone or as a mixture in different ratios.
Additives may also be included in the formulation to enhance uptake of the compound. Additives potentially having this property are for instance the fatty acids oleic acid, linoleic acid and linolenic acid.
Controlled release formulation may be desirable. The compound of .
this invention could be incorporated into an inert matrix which permits release by either diffusion or leaching mechanisms e.g., gums. Slowly degenerating matrices may also be incorporated into the formulation, e.g., alginates, polysaccharides. Another form of a controlled release of the compounds of this invention is by a method based on the Oros therapeutic system (Alza Corp.), i.e., the drug is enclosed in a semipermeable membrane which allows water to enter and push drug out through a single small opening due to osmotic effects. Some enteric coatings also have a delayed release effect.
Other coatings may be used for the formulation. These include a variety of sugars which could be applied in a coating pan. The therapeutic agent could also be given in a film coated tablet and the materials used in this instance are divided into 2 groups. The first are the nonenteric materials and include methyl cellulose, ethyl cellulose, hydroxyethyl cellulose, methylhydroxy-ethyl cellulose, hydroxypropyl cellulose, hydroxypropyl-methyl cellulose, sodium carboxy-methyl cellulose, providone and the polyethylene glycols. The second group consists of the enteric materials that are commonly esters of phthalic acid.

A mix of materials might be used to provide the optimum film coating. Film coating may be carried out in a pan coater or in a fluidized bed or by compression coating.
Pulmonary delivery forms. Also contemplated herein is.pulmonary delivery of the present protein (or derivatives thereof). The protein (or derivative) is delivered to the lungs of a mammal while inhaling and traverses across the lung epithelial lining to the blood stream. (Other reports'of this include Adjei et al., Pharma. Res. (1990) 7: 565-9; Adjei et al.
(1990), Internati.1. Pharmaceutics 63:135-44 (leuprolide acetate); Braquet et al. (1989), 1. Cardiovasc. Pharmacol.13 (suppl.5): s.143-146 (endothelin-1); Hubbard et al. (1989), Annals Int. Med. 3:206-12 (al-antitrypsin); Smith et al. (1989), T. Clin. Invest. 84:1145-6 (al-proteinase); Oswein et al.
(March 1990), "Aerosolization of Proteins", Proc. Svmp. Resp. Drug Delivery II, Keystone, Colorado (recombinant human growth hormone); Debs et al.
(1988), I. Immunol. 140: 3482-8 (interferon-y and tumor necrosis factor a) and Platz et al., U.S. Patent No. 5,284,656 (granulocyte colony stimulating factor).
Contemplated for use in the practice of this invention are a wide range of mechanical devices designed for pulmonary delivery of therapeutic products, including but not limited to nebulizers, metered dose inhalers, and powder inhalers, all of which are familiar to those skilled in the art . Some specific examples of commercially available devices suitable for the practice of this invention are the Ultravent*
nebulizer, manufactured by Mallinckrodt, Inc., St. Louis, Missouri; the Acorn*II nebulizer, manufactured by Marquest Medical Products, Englewood, Colorado; the Ventolin metered dose inhaler, manufactured by Glaxo Inc., Research Triangle Park, North Carolina; and-the Spinhaler*
powder inhaler, manufactured by Fisons Corp., Bedford, Massachusetts.
* Trade-mark All such devices require the use of formulations suitable for the dispensing ofthe inventive compound. Typically, each formulation is specific to the type of device employed and may involve the use of an appropriate propellant material, in addition to diluents, adjuvants and/or carriers useful in therapy.
The inventive compound should most advantageously be prepared in particulate form with an average particle size of less than 10 m (or microns), most preferably 0.5 to 5 m, for most effective delivery to the distal lung.
Pharmaceutically acceptable carriers include carbohydrates such as trehalose, mannitol, xylitol, sucrose, lactose, and sorbitol. Other ingredients for use in formulations may include DPPC, DOPE, DSPC and DOPC. Natural or synthetic surfactants may be used. PEG may be used (even apart from its use in derivatizing the protein or analog). Dextrans, such as cyclodextran, may be used. Bile salts and other related enhancers may be used. Cellulose and cellulose derivatives may be used. Amino .
acids may be used, such as use in a buffer formulation.
Also, the use of liposomes, microcapsules or microspheres, inclusion complexes, or other types of carriers is contemplated.
Formulations suitable for use with a nebulizer, either jet or ultrasonic, will typically comprise the inventive compound dissolved in water at a concentration of about 0.1 to 25 mg of biologically active protein per mL of solution. The formulation may also include a buffer and a simple sugar (e.g., for protein stabilization and regulation of osmotic pressure). The nebulizer formulation may also contain a surfactant, to reduce or prevent surface induced aggregation of the protein caused by atomization of the solution in forming the aerosol, Formulations for use with a metered-dose inhaler device will generally comprise a finely divided powder containing the inventive CA 02347131 2004-02-09 }

compound suspended in a propellant with the aid of a surfactant. The propellant may be any conventional material employed for this purpose, such as a chiorofluorocarbon, a hydrochlorofluorocarbon, a hydrofluorocarbon, or a hydrocarbon, including trichiorofluoromethane, dichiorodifluoromethane, dichlorotetrafluoroethanol, and 1,1,1,2-tetrafluoroethane, or combinations thereof. Suitable surfactants include sorbitan trioleate and soya lecithin. Oleic acid may also be useful as a surfactant.
Formulations for dispensing from a powder inhaler device will comprise a finely divided dry powder containing the inventive compound and may also include a bulking agent, such as lactose, sorbitol, sucrose, mannitol, trehalose, or xylitol in amounts which facilitate dispersal of the powder from the device, e.g., 50 to 90% by weight of the formulation.
Nasal delivery forms. Nasal delivery of the inventive compound is also contemplated. Nasal delivery allows the passage of the protein to the blood stream directly after administering the therapeutic product to the nose, without the necessity for deposition of the product in the lung.
Formulations for nasal delivery include those with dextran or cyclodextran. Delivery via transport across other mucous membranes is also contemplated.
Dosages. The dosage regimen involved in a method for treating the above-described conditions will be determined by the attending physician, considering various factors which modify the action of drugs, e.g. the age, condition, body weight, sex and diet of the patient, the severity of any infection, time of administration and other clinical factors. Generally, the daily regimen should be in the range of 0.1-1000 micrograms of the inventive compound per kilogram of body weight, preferably 0.1-150 micrograms per kilogram.

Specific preferred embodiments The inventors have determined preferred peptide sequences for molecules having many different kinds of activity. The inventors have further determined preferred structures of these preferred peptides combined with preferred linkers and vehicles. Preferred structures for these preferred peptides listed in Table 21 below.
Table 21-Preferred embodiments Sequence/structure SEQ Activity ID
NO:
F'- G -IEGPTLRQWLAARA- G -IEGPTLRQWLAARA 337 TPO-mimetic IEGPTLRQWLAARA- G -IEGPTLROWLAARA- G - F' 338 TPO-mimetic F'-(G)s IEGPTLRQWLAARA - TPO-mimetic IEGPTLRQWLAARA -(G),- F' TPO-mimetic F'-(G)S GGTYSCHFGPLTWVCKPQGG-(G); 339 EPO-mimetic GGTYSCHFGPLTWVCKPOGG
GGTYSCHFGPLTWVCKPQGG-(G)4 EPO-mimetic GGTYSCHFGPLTWVCKPOGG-(G) -F' 340 GGTYSCHFGPLTWVCKPQGG-(G),F' EPO-mimetic F'-(G)5 DFLPHYKNTSLGHRP TNF-a inhibitor DFLPHYKNTSLGHRP-(G)s F' TNF-a, inhibitor F'-(G),FEWTPGYWQPYALPL 1047 IL-1 R antagonist FEWTPGYWQPYALPL-(G)s F' IL-1 R antagonist F'-(G)S VEPNCDIHVMWEWECFERL VEGF-antagonist VEPNCDIHVMWEWECFERL-(G)s F' VEGF-antagonist F'-(G)S CTTHWGFTLC MMP inhibitor CTTHWGFTLC-(G),F' MMP inhibitor "F'" is an Fc domain as defined previously herein.
...Working examples The compounds described above may be prepared as described below. ' These examples comprise preferred embodiments of the invention and are illustrative rather than limiting.
Eam TPO-Mimeti ccs The following example uses peptides identified by the numbers appearing in Table A hereinafter.
Preparation of peptide 19. Peptide 17b (12 mg) and MeO-PEG-SH
5000 (30 mg, 2 equiv.) were dissolved in 1 ml aqueous buffer (pH 8). The mixture was incubated at RT for about 30 minutes and the reaction was checked by analytical HPLC, which showed a > 80% completion of the reaction. The pegylated material was isolated by preparative HPLC.
Preparation of peptide 20. Peptide 18 (14 mg) and MeO-PEG-maleimide (25 mg) were dissolved in about 1.5 ml aqueous buffer (pH 8).
The mixture was incubated at RT for about 30 minutes, at which time about 70% transformation was complete as monitored with analytical HPLC by applying an aliquot of sample to the HPLC column. The pegylated material was purified by preparative HPLC.
Bioactivity assay. The TPO in vitro bioassay is a mitogenic assay utilizing an IL-3 dependent clone of murine 32D cells that have been transfected .with human mpl receptor. This assay is described in greater detail in WO 95/26746. Cells are maintained in MEM medium containing 10% Fetal Clone II and 1 ng/ml mIL-3. Prior to sample addition, cells are prepared by rinsing twice with growth medium lacking mIL-3. An extended twelve point TPO standard curve is prepared, ranging from 33 to 39 pg/ml. Four dilutions, estimated to fall within the linear portion of the standard curve, (100 to 125 pg/ml), are prepared for each sample and run in triplicate. A volume of 100 pl of each dilution of sample or standard is added to appropriate wells of a 96 well microliter plate containing 10,000 cells/well. After forty-four hours at 37 C and 10% CO, .MTS (a tetrazolium compound which is bioreduced by cells to a formazan) is added to each well. Approximately six hours later, the optical density is read on a plate reader at 490 run. A dose response curve (log TPO
concentration vs. O.D.- Background) is generated and linear regression analysis of points which fall in the linear portion of the standard curve is performed. Concentrations of unknown test samples are determined using the resulting linear equation and a correction for the dilution factor.
TMP tandem repeats with polyglycine linkers. Our design of sequentially linked TMP repeats was based on the assumption that a dimeric form of TMP was required for its effective interaction with c-Mpl (the TPO receptor) and that depending on how they were wound up against, each other in the receptor context, the two TMP molecules could be tethered together in the C- to N-terminus configuration in a way that would not perturb the global dimeric conformation. Clearly, the success of the design of tandem linked repeats depends on proper selection of the length and composition of the linker that joins the C- and N-termini of the two sequentially aligned TMP monomers. Since no structural information of the TMP bound to c-MpI was available, a series of repeated peptides with linkers composed of 0 to 10 and 14 glycine residues (Table A) were synthesized. Glycine was chosen because of its simplicity and flexibility, based on the rationale that a flexible polyglycine peptide chain might allow for the free folding of the two tethered TMP repeats into the required conformation, while other amino acid sequences may adopt undesired secondary structures whose rigidity might disrupt the correct packing of the repeated peptide in the receptor context.
The resulting peptides are readily accessible by conventional solid phase peptide synthesis methods (Merrifield (1963), 1. Amer. Chem. Soc.
85: 2149) with either Fmoc or t-Boc chemistry. Unlike the synthesis of the C-terminally linked parallel dimer which required the use of an orthogonally protected lysine residue as the initial branch point to build the two peptide chains in a pseudosymmetrical way (Cwirla et al. (1997), Science 276: 1696-9), the synthesis of these tandem repeats was a straightforward, stepwise assembly of the continuous peptide chains from the C- to N-terminus. Since dimerization of TMP had a more dramatic effect on the proliferative activity than binding affinity as shown for the C-terminal dieter (Cwirla et al. (1997)), the synthetic peptides were tested directly for biological activity in a TPO-dependent cell-proliferation assay using an IL-3 dependent clone of murine 32D cells transfected with the full-length c-Mpl (Palacios et aL,. Ce1141:727 (1985)). As the test results showed, all the polyglycine linked tandem repeats demonstrated >1000 fold increases in potency as compared to the monomer, and were even more potent than the C-terminal dimer in this cell proliferation assay. The absolute activity of the C-terminal diner in our assay was lower than that of the native TPO protein, which is different from the previously reported findings in which the C-terminal dimer was found to be as active as the natural ligand (Cwirla et al. (1997)). This might be due to differences in the conditions used in the two assays. Nevertheless, the difference in activity between tandem (C terminal of first monomer linked to N
terminal of second monomer) and C-terminal (C terminal of first monomer linked to C terminal of second monomer; also referred to as parallel) dimers in the same assay dearly demonstrated the superiority of tandem repeat strategy over parallel peptide dimerization. It is interesting to note that a wide range of length is tolerated by the linker. The optimal linker between tandem peptides with the selected TMP monomers apparently is composed of 8 glycines. V -Other tandem repeats. Subsequent to this first series of TMP
tandem repeats, several other molecules were designed either with different linkers or containing modifications within the monomer itself.
The first of these molecules, peptide 13, has a linker composed of GPNG, a sequence known to have a high propensity to form a R-turn-type secondary structure. Although still about 100-fold more potent than the monomer, this peptide was found to be >10-fold less active than the equivalent GGGG-linked analog. Thus, introduction of a relatively rigid R-turn at the linker region seemed to have caused a slight distortion of the optimal agonist conformation in this short linker form.
The Trp9 in the TMP sequence is a highly conserved residue among the active peptides isolated from random peptide libraries. There is also a highly conserved Trp in the consensus sequences of EPO mimetic peptides and this Trp residue was found to be involved in the formation of a hydrophobic core between the two EMPs and contributed to hydrophobic interactions with the EPO receptor. Livnah et al. (1996), Science 273: 464-71). By analogy, the Trp9 residue in TMP might-have a similar function in dimerization of the peptide ligand, and as an attempt to modulate and estimate the effects of noncovalent hydrophobic forces exerted by the two indole rings, several analogs were made resulting from mutations at the Trp. So in peptide 14, the Trp residue was replaced in each of the two TMP monomers with a Cys, and an intramolecular disulfide bond was formed between the two cysteines by oxidation which was envisioned to mimic the hydrophobic interactions between the two Trp residues in peptide dimerization. Peptide 15 is the reduced form of peptide 14. In peptide 16, the two Trp residues were replaced by Ala. As the assay data show, all three analogs were inactive. These data further demonstrated that Trp is critical for the activity of the TPO mimetic peptide, not just for dieter formation. -The next two peptides (peptide 17a, and 18) each contain in their 8-amino acid linker a Lys or Cys residue. These two compounds are precursors to the two PEGylated peptides (peptide 19 and 20) in which the side chain of the Lys or Cys is modified by a PEG moiety. A PEG moiety was introduced at the middle of a relatively long linker, so that the large PEG component (5 kDa) is far enough away from the critical binding sites in the peptide molecule. PEG is a known biocompatible polymer which is increasingly used as a covalent modifier to improve the pharmacokinetic profiles of peptide- and protein-based therapeutics.
A modular, solution based method was devised for convenient PEGylation of synthetic or recombinant peptides. The method is based on the now well established chemoselective ligation strategy which utilizes the specific reaction between a pair of mutually reactive functionalities.
So, for pegylated peptide 19, the lysine side chain was preactivated with a bromoacetyl group to give peptide 17b to accommodate reaction with a thiol-derivatized PEG. To do that, an orthogonal. protecting group, Dde, was employed for the protection of the lysine E-amine. Once the whole peptide chain was assembled, the N-terminal amine was reprotected with t-Boc. Dde was then removed to allow for the bromoacetylation. This strategy gave a-high quality crude peptide which was easily purified using conventional reverse phase HPLC. Ligation of the peptide with the thiol-modified PEG took place in aqueous buffer at pH 8 and the reaction completed within 30 minutes. MALDI-MS analysis of the purified, pegylated material revealed a characteristic, bell-shaped spectrum with an increment of 44 Da between the adjacent peaks. For PEG-peptide 20, a cysteine residue was placed in the linker region and its side chain thiol group would serve as an attachment site for a maleimide-containing PEG.
Similar conditions were used for the pegylation of this peptide. As the assay data revealed, these two pegylated peptides had even higher in vitro bioactivity as compared to their unpegylated counterparts.

Peptide 21 has in its 8-amino acid linker a potential glycosylation motif,-NGS. Since our exemplary tandem repeats are made up of natural amino acids linked by peptide bonds, expression of such a molecule in an appropriate eukaryotic cell system should produce a glycopeptide with the carbohydrate moiety added on the side chain carboxyamide of Asn.
Glycosylation is a common post-translational modification process which can have many positive impacts on the biological activity of a given protein by increasing its aqueous solubility and in vivo stability. As the assay data show, incorporation of this glycosylation motif into the linker maintained high bioactivity. The synthetic precursor of the potential glycopeptide had in effect an activity comparable to that of the -(G);
linked analog. Once glycosylated, this peptide is expected to have the same order of activity as the pegylated peptides, because of the similar chemophysical properties exhibited by a PEG and a carbohydrate moiety.
The last peptide is a dimer of a tandem repeat. It was prepared by oxidizing peptide 18, which formed an intermolecular disulfide bond between the two cysteine residues located at the linker. This peptide was designed to address the possibility that TMP was active as a tetramer. The assay data showed that this peptide was not more active than an average tandem repeat on an adjusted molar basis, which indirectly supports the idea that the active form of TMP is indeed a dimer, otherwise dimerization of a tandem repeat would have a further impact on the bioactivity.
In order to confirm the in vitro data in animals, one pegylated TMP
tandem repeat (compound 20 in Table A) was delivered subcutaneously to normal mice via osmotic pumps. Time and dose-dependent increases were seen in platelet numbers for the duration of treatment.. Peak platelet levels-over 4-fold baseline were seen on day 8. A dose of 1(T g/kg/day of the pegylated TMP repeat produced a similar response to rHuMGDF
(non-pegylated) at 100 g/kg/day delivered by the same route.

Table A-TPO-mimetic Peptides Peptide Compound SEQ ID Relative No. NO: Potency TPO ++++
TMP monomer 13 +
TMP C-C dimer +++-TMP-(G)~ TMP:
1 n = 0 341 ++++-2 n=1 342 ++++
3 n=2 343 ++
4 n=3 344 ++++
n=4 345 +++
6 n=5 _ 346 ++++
7 n=6 347 +++
8 n=7 348 +++
9 n=8 349 ++++-n=9 350 ++++
11 n=10 351 ++++
12 n =14 352 ++++
13 TMP-GPNG-TMP 353 +++
I
(cyclic) IEGPTLRQ.QLAARA-GGGGGGGG- 355 -IEGPTLRQQLAARA (linear) IEGPTLRQALAARA
17a TMP-GGGKGGGG-TMP 357 ++++
17b TMP-GGGK(BrAc)GGGG-TMP 358 ND
18 TMP-GGGCGGGG-TMP 359 ++++
19 TMP-GGGK(PEG)GGGG-TMP 360 +++++
TMP-GGGC(PEG)GGGG-TMP 361 +++++
21 TMP-GGGN*GSGG-TMP 362 ++++
22 TMP-GGGCGGGG-TMP 363- _ Discussion. It is well accepted that MGDF acts in a way similar to hGH, i.e., one molecule of the protein ligand binds two molecules of the receptor for its activation. Wells et al.(1996), Ann. Rev. Biochem. 65: 609-34. Now, this interaction is mimicked by the action of a much smaller peptide, TMP. However, the present studies suggest that this mimicry requires the concerted action of two TMP molecules, as covalent dimerization of TMP in either a C-C parallel or C-N sequential fashion increased the in vitro biological potency of the original monomer by a factor of greater than 103. The relatively low biopotency of the monomer is probably due to inefficient formation of the noncovalent dimer. A
preformed covalent repeat has the ability to eliminate the entropy barrier for the formation of a noncovalent dimer which is exclusively driven by weak, noncovalent interactions between two molecules of the small, 14-residue peptide.
It is intriguing that this tandem repeat approach had a similar effect on enhancing bioactivity as the reported C-C dimerization is intriguing.
These two strategies brought about two very different molecular configurations. The C-C dimer is a quasi-symmetrical molecule, while the tandem repeats have no such symmetry in their linear structures. Despite this difference in their primary structures, these two types of molecules appeared able to fold effectively into a similar biologically active conformation and cause the dimerization and activation of c-Mpl. These experimental observations provide a number of insights into how the two TMP molecules may interact with one another in binding to c-Mpl. First, the two C-termini of the two bound TMP molecules must be in relatively close proximity with each other, as suggested by data on the C-terminal dimer. - Second, the respective N- and C-termini of the two TMP molecules in the receptor complex must also be very closely aligned with each other, such that they can be directly tethered together with a single peptide bond to realize the near maximum activity-enhancing effect brought about by the tandem repeat strategy. Insertion of one or more (up to 14) glycine residues at the junction did not increase (or decrease) significantly the activity any further. This may be due to the fact that a flexible polyglycine peptide chain is able to loop out easily from the junction without causing any significant changes in the overall conformation. This flexibility seems to provide the freedom of orientation for the TMP peptide chains to fold into the required conformation in interacting with the receptor and validate it as a site of modification. Indirect evidence supporting this came from the study on peptide 13, in which a much more rigid b-turn-forming sequence as the linker apparently forced a deviation of the backbone alignment around the linker which might have resulted in a slight distortion of the optimal conformation, thus resulting in a moderate (10-fold) decrease in activity as compared with the analogous compound with a 4-Gly linker. Third, Trp9 in TMP plays a similar role as Trp13 in EMP, which is involved not only in peptide--peptide interaction for the formation of dimers but also is important for contributing hydrophobic forces in peptide:receptor interaction. Results obtained with the W to C
mutant analog, peptide 14, suggest that a covalent disulfide linkage is not sufficient to approximate the hydrophobic interactions provided by the Trp pair and that, being a short linkage, it might bring the two TMP
monomers too dose, therefore perturbing the overall conformation of the optimal dimeric structure.
An analysis of the possible secondary structure of the TMP peptide can provide further. understanding on the interaction between TMP and c-Mpl. This can be facilitated by making reference to the reported structure of the EPO mimetic peptide. Livnah et al. (1996), Science 273:464-75 The receptor-bound EMP has a b-hairpin structure with a b-turn formed by the highly consensus Gly-Pro-Leu Thr at the center of its sequence. Instead of GPLT, TMP has a highly selected GPTL sequence which is likely to form a similar-turn. However, this turn-like motif is located near the N-terminal part in TMP. Secondary structure prediction using Chau-Fasman method suggests that the C-terminal half of the peptide has a tendency to adopt a helical conformation. Together with the highly conserved Trp at position 9, this C-terminal helix may contribute to the stabilization of the dimeric structure. It is interesting to note that most of our tandem repeats are more potent than the C-terminal parallel dimer. Tandem repeats seem to give the molecule a better fit conformation than does the C-C parallel dimerization. The seemingly asymmetric feature of a tandem repeat might have brought it closer to the natural ligand which, as an asymmetric molecule, uses two different sites to bind two identical receptor molecules.
Introduction of a PEG moiety was envisaged to enhance the in vivo activity of the modified peptide by providing it a protection against proteolytic degradation and by slowing down its clearance through renal filtration. It was unexpected that pegylation could further increase the in vitro bioactivity of a tandem repeated TMP peptide in the cell-based proliferation assay.
Example, 2 Fc-TMP fusions TMPs (and EMPs as described in Example 3) were expressed in either monomeric or dimeric form as either N -terminal or C-terminal fusions to the Fc region of human IgG1. In all cases, the expression construct utilized the luxPR promoter promoter in the plasmid expression vector pAMG21.
Fc-TMI'. A DNA sequence coding for the Fc region of human IgG1 fused in-frame to a monomer of the TPO-mimetic peptide was constructed using standard PCR technology. Templates for PCR reactions were the pFc-A3 vector and a synthetic TMP gene. The synthetic gene was ~ j constructed from the 3 overlapping oligonucleotides (SEQ ID NOS: 364, 365, and 366; respectively) shown below:

CTG ACG CAG AGT CGG ACC

TTT
These oligonucleotides were annealed to form the duplex encoding an amino acid sequence (SEQ ID NOS: 367 and 368, respectively) shown below:

AAAGGTGGAGGTGGTGGTATCGAAGGTCCGACTCTGCGTCAGTGGCTGGCTGCTCGTGCT
1 ---------+---------+---------+---------+---------+---------+ 60 CCAGGCTGAGACGCAGTCACCGACCGACGAGCACGA
a K G G G G G I E G P T L R Q W L A A R A
TAATCTCGAGGATCCTTTTTT
61 ---------+---------+- 81 ATTAGAGCTCCTAGGAAAAAA
a This duplex was amplified in a PCR reaction using 1842-98 and 1842-97 as the sense and antisense primers.
The Fc portion of the molecule was generated in a PCR reaction with pFc-A3 using the primers shown below (SEQ ID NOS: 369 and 370):

The oligonucleotides 1830-51 and 1842-98 contain an overlap of 24 nucleotides, allowing the two genes to be fused together in the correct reading frame by combining the above PCR products in a third reaction using the outside primers, 1216-52 and 1842-97.
The final PCR gene product (the full length fusion gene) was digested with restriction endonucleases XbaI and BamHI, and then ligated into the vector pAMG21 and transformed into competent E. coil strain 2596 cells as described for EMP-Fc herein. Clones were screened for the ability to produce the recombinant protein product and to possess the gene fusion having the correct nucleotide sequence. A single such clone was selected-and designated Amgen strain #3728.
The nucleotide and amino acid sequences (SEQ ID NOS: 5 and 6) of the fusion protein are shown in Figure 7.
Fc-TMP-TMP. A DNA sequence coding for the Fc region of human IgG1 fused in-frame to a dimer of the TPO-mimetic peptide was constructed using standard PCR technology. Templates for PCR reactions were the pFc-A3 vector and a synthetic TMP-TMP gene. The synthetic gene was constructed from the 4 overlapping oligonudeotides (SEQ ID
NOS: 371 to 374, respectively) shown below:

ACT CTG CGT CAG TGG CTG GCT GCT CGT GCT

CCA CTG ACG CAG AGT CGG ACC

CTT CGC CAA TGG CTT GCA GCA CGC GCA.

ATT GGC GAA GGG TTG GGC CCT CAA TAC CTC CGC CGC C
The 4 oligonudeotides were annealed to form the duplex encoding an amino acid sequence (SEQ ID NOS: 375 and 376, respectively) shown below:

AAAGGTGGAGGTGGTGGTATCGAAGGTCCGACTCTGCGTCAGTGGCTGGCTGCTCGTGCT
1 ---------+---------+---------+---------+---------+---------+ 60 CCAGGCTGAGACGCAGTCACCGACCGACGAGCACGA
a K G G G G G I E G P T L R Q W L A A R A
GGTGGTGGAGGTGGCGGCGGAGGTATTGAGGGCCCAACCCTTCGCCAATGGCTTGCAGCA
61 ---------+---------+---------+---------+---------+---------+ 120 CCACCACCTCCACCGCCGCCTCCATAACTCCCGGGTTGGGAAGCGGTTACCGAACGTCGT
a G G G G G G G. G I E G P T L R Q W L A A
CGCGCA

GCGCGTATTAGAGCTCCTAGGAAAAAAA

a R A This duplex was amplified in a PCR reaction using 1830-52 and 1830-55 as the sense and antisense primers.
The Fc portion of the molecule was generated in a PCR reaction with pFc-A3 using the primers 1216-52 and 1830-51 as described above for Fc-TMP. The full length fusion gene was obtained from a third PCR
reaction using the outside primers 1216-52 and 1830-55.
The final PCR gene product (the full length fusion gene) was digested with restriction endonucleases XbaI and BamFH, and then ligated into the vector pAMG21 and transformed into competent E. coli strain 2596 cells as described in example 1. Clones were screened for the ability to produce the recombinant protein product and to possess the gene fusion having the correct nucleotide sequence. A single such done was selected and designated Amgen strain #3727.
The nucleotide and amino acid sequences (SEQ ID NOS: 7 and 8) of the fusion protein are shown in Figure 8.
TMP-TMP-Fc. A DNA sequence coding for a tandem repeat of the TPO-mimetic peptide fused in-frame to the Fc region of human IgG1 was constructed using standard PCR technology. Templates for PCR reactions were the EMP-Fc plasmid from strain #3688 (see Example 3) and a synthetic gene encoding the TMP dieter. The synthetic gene for the tandem repeat was constructed from the 7 overlapping oligonucleotides shown below (SEQ ID NOS: 377 to 383, respectively):

GAT CAT ATG

CAC ACA

TCC GCC ACC ACC GCC

GGT GGG GAC AAA ACT

These oligonudeotides were annealed to form the duplex shown encoding 4 0 an amino acid sequence shown below (SEQ ID NOS 384 and 385):

TTTTTTCATATGATCGAAGGTCCGACTCTGCGTCAGTGGCTGGCTGCTCGTGCTGGCGGT
1 ---------+---------+---------+---------+---------+-...-----+ 60 GTATACTAGCTTCCAGGCTGAGACGCAGTCACCGACCGACGAGCACGACCGCCA
a M I E G P T L R Q W L A A R A G G -GGTGGCGGAGGGGGTGGCATTGAGGGCCCAACCCTTCGCCAATGGCTGGCTGCTCGTGCT
61 ---------+---------+---------+---------+---------+---------+ 120 CCACCGCCTCCCCCACCGTAACTCCCGGGTTGGGAAGCGGTTACCGAACGTCGTGCGCGT
a G G G G G G I E G P T L R Q W L A A R A
GGTGGAGGCGGTGGGGACAAAACTCTGGCTGCTCGTGCTGGTGGAGGCGGTGGGGACAAA
121 ---------+---------+---------+------- +---------+---------+ 180 CCCCCTCCGCCACCC
a G G G G G D K T L A A R A G G G G G D K -ACTCACACA

a T H T -This duplex was amplified in a PCR reaction using 1885-52 and 1885-58 as the sense and antisense primers.
The Fc portion of the molecule-was generated in a PCR reaction with DNA from the EMP-Fc fusion strain #3688 (see Example 3) using the primers 1885-54 and 1200-54. The full length fusion gene was obtained from a third PCR reaction using the outside primers 1885-52 and 1200-54.
The final PCR gene product (the full length fusion gene) was digested with restriction endonucleases XbaI and BamHI, and then ligated into the vector pAMG21 and transformed into competent E. coli strain 2596 cells as described for Fc-EMP herein. Clones were screened for the ability to produce the recombinant protein product and to possess the gene fusion having the correct nucleotide sequence. A single such done was selected and designated Amgen strain #3798.
The nucelotide and amino acid sequences (SEQ ID NOS: 9 and 10) of the fusion protein are shown in Figure 9.
TMP-Fc. A DNA sequence coding for a monomer of the TPO-mimetic peptide fused in-frame to the Fc region of human IgGI was obtained fortuitously in the ligation in TMP TMP Fc, presumably due to the ability of primer 1885-54 to anneal to 1885-53 as well as to 1885-58. A
single clone having the correct nucleotide sequence for the TMP-Fc construct was selected and designated Amgen strain #3788.

The nucleotide and amino acid sequences (SEQ ID NOS: 11 and 12) of the fusion protein are shown in Figure 10.
Expression in E. coli. Cultures of each of the pAMG21-Fc-fusion constructs in E. coli GM221 were grown at 37 C in Luria Broth medium containing 50 mg/ml kanamycin. Induction of gene product expression from the luxPR promoter was achieved following the addition of the synthetic autoinducer N-(3-oxohexanoyl)-DL-homoserine lactone to the culture media to a final concentration of 20 ng/ml. Cultures were incubated at 37 C for a further 3 hours. After 3 hours, the bacterial cultures were examined by microscopy for the presence of inclusion bodies and were then collected by centrifugation. Refractile inclusion bodies were observed in induced cultures indicating that the Fc-fusions were most likely produced in the insoluble fraction in E. coli. Cell pellets were lysed directly by resuspension in Laemmli sample buffer containing 10% b-mercaptoethanol and were analyzed by SDS-PAGE. In each case, an intense coomassie-stained band of the appropriate molecular weight was observed on an SDS-PAGE gel.
pAMG21. The expression plasmid pAMG21 can be derived from the Amgen expression vector pCFM1656 (ATCC #69576) which in turn be derived from the Amgen expression vector system described in US Patent No. 4,710,473. The pCFM1656 plasmid can be derived from the described pCFM836 plasmid (Patent No. 4,710,473) by:
(a) destroying the two endogenous NdeI restriction sites by end filling with T4 polymerase enzyme followed by blunt end ligation;
(b) replacing the DNA sequence between the unique AatII and Clal restriction sites containing the synthetic PL promoter with a similar fragment obtained from pCFM636 (patent No. 4,710,473) containing the PL promoter (see SEQ ID NO: 386 below); and (c) substituting the small DNA sequence between the unique C1aI
and KpnI restriction sites with the oligonucleotide having the sequence of SEQ ID NO: 388.
SEQ ID NO: 386:
III
5' CTAATTCCGCTCTCACCTACCAAACAATGCCCCCCTGCAAAAAATAAATTCATAT-3' TGCAGATTAAGGCGAGAGTGGATGGTTTGTTACGGGGGGACGTTTTTTATTTAAGTATA--AAAAAACATACAGATAACCATCTGCGGTGATAAATTATCTCTGGCGGTGTTGACATAAA--TTTTTTGTATGTCTATTGGTAGACGCCACTATTTAATAGAGACCGCCACAACTGTATTT-- TACCACTGGCGGTGATACTGAGCACAT 3' -ATGGTGACCGCCACTATGACTCGTGTAGC 5' Clai SEQ ID NO: 387:
5' CGATTTGATTCTAGAAGGAGGAATAACATATGGTTAACGCGTTGGAATTCGGTAC 3' 3' TAAACTAAGATCTTCCTCCTTATTGTATACCAATTGCGCAACCTTAAGC 5' CUd ,K nI
The expression plasmid pAMG21 can then be derived from pCFM1656 by making a series of site-directed base changes by PCR overlapping oligo mutagenesis and DNA sequence substitutions. Starting with the Bg1II site (plasmid bp # 180) immediately 5' to the plasmid replication promoter PcopB and proceeding toward the plasmid replication genes, the base pair changes are as shown in Table B below.

Table B-Base pair changes resulting in pAMG21 pAMG21 by # by in pCFM1656 bo changed to in A 1 # 204 T/A C/G
# 428 A/T G/C
# 509 G/C Apr # 617 - - insert two G/C bp # 679 G/C T/A
# 980 T/A C/G
# 994 G/C A/T
# 1004 A/T C/G
# 1007 C/G T/A
# 1028 A/T T/A
# 1047 C/G T/A
# 1178 G/C T/A
# 1466 G/C T/A
# 2028 G/C bp deletion # 2187 C/G T/A
# 2480 A/T T/A

# 2499-2502 AGTG GTCA
TCAC CAGT
# 2642 TCCGAGC 7 bp deletion AGGCTCG
# 3435 G/C A/T
# 3446 G/C A/T
# 3643 A/T T/A

^1 {

The DNA sequence between the unique Aatl (position #4364 in pCFM1656) and SacII (position #4585 in pCFM1656) restriction sites is substituted with the DNA sequence (SEQ ID NO: 23) shown in Figures 17A and 17B. During the ligation of the sticky ends of this substitution DNA sequence, the outside AatII and Sacl sites are destroyed. There are unique AatII and SacII sites in the substituted DNA.
GM221 (Amgen #2596). The Amgen host strain #2596 is an E.coli K-12 strain derived from Amgen strain #393. It has been modified to contain both the temperature sensitive lambda repressor d857s7 in the early ft region and the lacI repressor in the late ft region (68 minutes). The presence of these two repressor genes allows the use of this host with a variety of expression systems, however both of these repressors are irrelevant to the expression from IuxPR. The untransformed host has no antibiotic resistances.
The ribosome binding site of the c1857s7 gene has been modified to include an enhanced RBS. It has been inserted into the ft operon between nucleotide position 1170 and 1411 as numbered in Genbank accession number M64441Gb_Ba with deletion of the intervening ft sequence. The sequence of the insert is shown below with lower case letters representing the ft sequences flanking the insert shown below (SEQ ID NO: 388):
ttattttcgtGCGGCCGCACCATTATCACCGCCAGAGGTAAACTAGTCAACACGCACGGTGTTAGATATTTAT
CCCTTGCGGTGATAGATTGAGCACATCGATTTGATTCTAGAAGGAGGGATAATATATGAGCACAAAAAAGAAA
CCATTAACACAAGAGCAGCTTGAGGACGCACGTCGCCTTAAAGCAATTTATG TGAACTTG
GCTTATCCCAGGAATCTGTCGCAGACAAGATGGGGATGGGGCAGTCAGGCGTTGGTGCTTTATTTAATGGCAT
CAATGCATTAAATGCTTATAACGCCGCATTGCTTACAAAAATTCTCAAAGTTAGCGTTGAAGAATTTAGCCCT
TCAATCGCCAGAGAATCTACGAGATGTATGAAGCGGTTAGTATGCAGCCGTCACTTAGAAGTGAGTATGAGTA
CCCTGTTTTTTCTCATGTTCAGGCAGGGATGTTCTCACCTAAGCTTAGAACCTTTACCAAAGGTGATGCGGAG
AGATGGGTAAGCACAACCAAAAAAGCCAGTGATTCTGCATTCTGGCTTGAGGTTGAAGGTAATTCCATGACCG
CACCAACAGGCTCCAAGCCAAGCTTTCCTGACGGAATGTTAATTCTCGTTGACCCTGAGCAGGCTGTTGAGCC
AGGTGATTTCTGCATAGCCAGACTTGGGGGTGATGAGTTTACCTTCAAGAAACTGATCAGGGATAGCGGTCAG
GTGTTTTTACAACCACTAAACCCACAGTACCCAATGATCCCATGCAATGAGAGTTGTTCCGTTGTGGGGAAAG
TTATCGCTAGTCAGTGGCCTGAAGAGACGTTTGGCTGATAGACTAGTGGATCCACTAGTgtttctgccc The construct was delivered to the chromosome using a recombinant phage called MMebg-cI857s7enhanced RBS #4 into F'tet / 393.
After recombination and resolution only the chromosomal insert described ,J r` v "n- -Wr1 meaxeee I

above remains in the cell. It was renamed F'tet/GM101. F'tet/GM101 was then modified by the delivery of a lack construct into the e_g operon between nucleotide position 2493 and 2937 as numbered in the Genbank accession number M64441Gb_Ba with the deletion of the intervening ft sequence. The sequence of the insert is shown below with the lower case letters representing the ft sequences flanking the insert (SEQ ID NO:
389) shown below:
ggcggaaaccGACGTCCATCGAATGGTGCAAAACCTTTCGCGGTATGGCATGATAGCGCCCGGAAGAGAGTCA
ATTCAGGGTGGTGAATGTGAAACCAGTAACGTTATACGATGTCGCAGAGTATGCCGGTGTCTCTTATCAGACC
GTTTCCCGCGTGGTGAACCAGGCCAGCCACGTTTCTGCGAAAACGCGGGAAAAAGTCGAAGCGGCGATGGCGG
AGCTGAATTACATTCCCAACCGCGTGGCACAACAACTGGCGGGCAAACAGTCGCTCCTGATTGGCGTTGCCAC
CTCCAGTCTGGCCCTGCACGCGCCGTCGCAAATTGTCGCGGCGATTAAATCTCGCGCCGATCAACTGGGTGCC
AGCGTGGTGGTGTCGATGGTAGAACGAAGCGGCGTCGAAGCCTGTAAAGCGGCGGTGCACAATCTTCTCGCGC
AACGCGTCAGTGGGCTGATCATTAACTATCCGCTGGATGACCAGGATGCCATTGCTGTGGAAGCTGCCTGCAC
TAATGTTCCGGCGTTATTTCTTGATGTCTCTGACCAGACACCCATCAACAGTATTATTTTCTCCCATGAAGAC
GGTACGCGACTGGGCGTGGAGCATCTGGTCGCATTGGGTCACCAGCAAATCGCGCTGTTAGCGGGCCCATTAA
GTTCTGTCTCGGCGCGTCTGCGTCTGGCTGGCTGGCATAAATATCTCACTCGCAATCAAATTCAGCCGATAGC
GGAACGGGAAGGCGACTGGAGTGCCATGTCCGGTTTTCAACAAACCATGCAAATGCTGAATGAGGGCATCGTT
CCCACTGCGATGCTGGTTGCCAACGATCAGATGGCGCTGGGCGCAATGCGCGCCATTACCGAGTCCGGGCTGC
GCGTTGGTGCGGATATCTCGGTAGTGGGATACGACGATACCGAAGACAGCTCATGTTATATCCCGCCGTTAAC
CACCATCAAACAGGATTTTCGCCTGCTGGGGCAAACCAGCGTGGACCGCTTGCTGCAACTCTCTCAGGGCCAG
GCGGTGAAGGGCAATCAGCTGTTGCCCGTCTCACTGGTGAAAAGAAAAACCACCCTGGCGCCCAATACGCAAA
CCGCCTCTCCCCGCGCGTTGGCCGATTCATTAATGCAGCTGGCACGACAGGTTTCCCGACTGGAAAGCGGACA
GTAAGGTACCATAGGATCCaggcacagga The construct was delivered to the chromosome using a recombinant phage called AGebg-LacIQ#5 into F'tet/GM101. After recombination and resolution only the chromosomal insert described above remains in the cell. It was renamed F'tet/GM221. The F'tet episome was cured from the strain using acridine orange at a concentration of 25 g/ml in LB. The cured strain was identified as tetracyline sensitive and was stored as GM221.

faTression. Cultures of pAMG21-Fc-TMP-TMP in E. coli GM221 in Luria Broth medium containing 50 g/ml kanamycin were incubated at 37 C prior to induction. Induction of Fc-TMP-TMP gene product expression from the luxPR promoter was achieved followinj the addition of the synthetic autoinducer N-(3-oxohexanoyl)-DL-homoserine lactone to the culture media to a final concentration of 20 ng/ml and cultures were incubated at 37 C for a further 3 hours. After 3 hours, the bacterial WO 00/24782 PC'rIUS99n5044 cultures were examined by microscopy for the presence of inclusion bodies and were then collected by centrifugation. Refractile inclusion bodies were observed in induced cultures indicating that the Fc-TMP-TMP
was most likely produced in the insoluble fraction in E. coli. Cell pellets were lysed directly by resuspension in Laemmli sample buffer containing 10% =-mercaptoethanol and were analyzed by SDS-PAGE. An intense Coomassie stained band of approximately 30kDa was observed on an SDS-PAGE gel. The expected gene product would be 269 amino acids in length and have an expected molecular weight of about 29.5 kDa.
Fermentation was also carried out under standard batch conditions at the 10 L scale, resulting in similar expression levels of the Fc-TMP-TMP to those obtained at bench scale.
Purification of Fc-TMP-TMP. Cells are broken in water (1/10) by high pressure homogenization (2 passes at 14,000 PSI) and inclusion bodies are harvested by centrifugation (4200 RPM in J-6B for 1 hour).
Inclusion bodies are solubilized in 6M guanidine, 50mM Tris, 8mM DTT, pH 8.7 for 1 hour at a 1 /10 ratio. The solubilized mixture is diluted 20 times into 2M urea, 50 mM tris, 160mM arginine, 3mM cysteine, pH 8.5.
The mixture is stirred overnight in the cold and then concentrated about 10 fold by ultafiltration. It is then diluted 3 fold with 10mM Tris, 1.5M
urea, pH 9. The pH of this mixture is then adjusted to pH 5 with acetic acid. The precipitate is removed by centrifugation and the supernatant is loaded onto a SP-Sepharose Fast Flow column equilibrated in 20mM
NaAc, 100 mM NaCl, pH 5(10mg/ml protein load, room temperature).
The protein is eluted off using a 20 column volume gradient in the same buffer ranging from 100mM NaCl to 500mM NaCl. The pool from the column is diluted 3 fold and loaded onto a SP-Sepharose HP column in 20 mM NaAc, 150 mM NaCl, pH 5(10 mg/ml protein load, room temperature). The protein is eluted off using a 20 column volume gradient * Trade-mark 105 in the same buffer ranging from 150 mM NaCI to 400 mM NaCl. The peak is pooled and filtered.
Characterization of Fc-TMP activity. The following is a summary of in vivo data in mice with various compounds of this invention..
Mice: Normal female BDF1 approximately 10-12 weeks of age.
Bleed schedule: Ten mice per group treated on day 0, two groups started 4 days apart for a total of 20 mice per group. Five mice bled at each time point, mice were bled a minimum of three times a week. Mice were anesthetized with isoflurane and a total volume of 140-160 pl of blood was obtained by puncture of the orbital sinus. Blood was counted on a Technicon ME blood analyzer running software for murine blood.
Parameters measured were white blood cells, red blood cells, hematocrit, hemoglobin, platelets, neutrophils.
Treatments: Mice were either injected subcutaneously for a bolus treatment or implanted with 7-day micro-osmotic pumps for continuous delivery. Subcutaneous injections were delivered in a volume of 0.2 ml.
Osmotic pumps were inserted into a subcutaneous incision made in the skin between the scapulae of anesthetized mice. Compounds were diluted in PBS with 0.1% BSA. All experiments included one control group, labeled "carrier" that were treated with this diluent only. The concentration of the test articles in the pumps was adjusted so that the calibrated flow rate from the pumps gave the treatment levels indicated in the graphs.
Compounds: A dose titration of the compound was delivered to mice in 7 day micro-osmotic pumps. Mice were treated with various compounds at a single dose of 100 g/kg in 7 day osmotic pumps. Some of the same compounds were*then given to mice as a single-bolus injection.
Activity test results: The results of the activity experiments are shown in Figures 11 and 12. In dose response assays using 7-day micro-osmotic pumps, the maximum effect was seen with the compound of SEQ
ID NO: 18 was at 100 g/kg/day; the 10 g/kg/day dose was about 50%G
maximally active and 1 g/kg/day was the lowest dose at which activity could be seen in this assay system. The compound at 10 g/kg/day dose was about equally active as 100 g/kg/day unpegylated rHu-MGDF in the same experiment.

M&JCIrV'W4f%- -AWN

Example 3 Fc-EMP fusions Fc-EMP. A DNA sequence coding for the Fc region of human IgG1 fused in-frame to a monomer of the EPO-mimetic peptide was constructed using standard PCR technology. Templates for PCR reactions were a vector containing the Fc sequence (pFc-A3, described in International application WO 97/23614, published July 3,1997) and a synthetic gene encoding EPO monomer. The synthetic gene for the monomer was constructed from the 4 overlapping oligonucleotides (SEQ ID NOS: 390 to 393, respectively) shown below:

CCA CTT CGG CCC GCT GAC TTG G

GTA AGT ACC TCC ACC ACC ACC TCC ACC TTT CAT

ACC TAT TCC TGT CAT TTT

GCC GCC GCC GCC GCC ACC CTG
The 4 oligonudeotides were annealed to form the duplex encoding an amino acid sequence (SEQ ID NOS: 394 and 395, respectively) shown below:

TATGAAA GGTACTTACTCTTGCCACTTCGGCCCGCTGAC? G
1 ----==---+-........+........................................ 60 TACTTTCCACCTCCACCACCACCTCCATGAATGAGAACGGTGAAGCCGGGCGACTGAAC
b M K G G G G G G G T Y S C H F G P L T W-GGTTTGCAAACCGCAGGGTGGCGGCGGCGGCGGCGGTGGTACCTATTCCTGfCATTTT
61 --.......+.........+.........+---------+--.........---......+..........+.-CCAAACGTTTGGCGTCCCACCGCCGCCGCCGCCGCCACCATGGATAAGGACAGTAAAACCGGGCGACTGGACC
b V C K P Q G G G G G G G G T Y S C H F
This duplex was amplified in a PCR reaction using AGG TGG AGG TGG TGG TGG AGG TAC TTA
CTC T
and CTG CGG TTT GCA A

as the sense and antisense primers (SEQ ID NOS: 396 and 397, respectively).
The Fc portion of the molecule was generated in a PCR reaction with pFc-A3 using the primers AGA CAG GGA GAG GCT CTT CTG C
which are SEQ ID NOS: 3 69 and 399, respectively. The oligonucleotides 1798-17 and 1798-18 contain an overlap of 61 nucleotides, allowing the two genes to be fused together in the correct reading frame by combining the above PCR products in a third reaction using the outside primers, 1216-52 and 1798-19. -The final PCR gene product (the full length fusion gene) was digested with restriction endonucleases XbaI and BaMHI, and then ligated into the vector pAMG21(described below), also digested with XbaI and BamHI. Ligated DNA was transformed into competent host cells of E. coli strain 25% (GM221, described herein). Clones were screened for the ability to produce the recombinant protein product and to possess the gene fusion having the correct nucleotide sequence. A single such clone was selected and designated Amgen strain #3718.
The nucleotide and amino acid sequence of the resulting fusion protein (SEQ ID NOS: 15 and 16) are shown in Figure 13.
EMP-Fc. A DNA sequence coding for a monomer of the EPO-mimetic peptide fused in-frame to the Fc region of human IgG1 was constructed using standard PCR technology. Templates for PCR reactions were the pFC-A3a vector and a synthetic gene encoding EPO monomer.
The synthetic gene for the monomer was constructed from the 4 overlapping oligonucleotides 1798-4 and 1798-5 (above) and 1798-6 and 1798-7 (SEQ ID NOS: 400 and 401, respectively) shown below:

GGA GGC GGG GGG TAA TCT CGA G

GGC TTA CAT AC
The 4 oligonucleotides were annealed to form the duplex encoding an amino acid sequence (SEQ ID NOS: 402 and 403, respectively) shown below:

GTTTGCAAACCGCAGGGTGGCGGCGGCGGCGGCGGTGGTACCTATTCCTGTCATTTTGGC
1 --....---+---------+ +---------+---------+---------+ 60 GTCCCACCGCCGCCGCCGCCGCCACCATGGATAAGGACAGTAAAACCG
A V C K P Q G G G G G G G G T Y S C H F G
CCGCTGACCTGGGTATGTAAGCCACAAGGGGGTGGGGGAGGCGGGGGGTAATCTCGAG
61 ---------+---------+---------+---------+---------+---------+- 122 GGCGACTGGACCCATACATTCGGTGTTCCCCCACCCCCTCCGCCCCCCATTAGAGCTCCTAG
A P L T W V C K P Q G G G G G G G
This duplex was amplified in-a PCR reaction using and CCC T
as the sense and antisense primers (SEQ ID NOS: 404 and 405, respectively).
The Fc portion of the molecule was generated in a PCR reaction with pFc-A3 using the primers TCC A
and which are SEQ ID NOS: 406 and 407, respectively. The oligonucleotides 1798-22 and 1798-23 contain an overlap of 43 nucleotides, allowing the two genes to be fused together in the correct reading frame by combining the above PCR products in a third reaction using the outside primers, 1787-21 and 1200-54.
The final PCR gene product (the full length fusion gene) was digested with restriction endonucleases XbaI and BamHI, and then ligated into the vector pAMG21 and transformed into competent E. coli strain 2596. cells-as described above. Clones were screened for the ability to produce the recombinant protein product and to possess the gene fusion having the correct nucleotide sequence. A single such clone was selected and designated Amgen strain #3688.
The nucleotide and amino acid sequences (SEQ ID NOS: 17 and 18) of the resulting fusion protein are shown in Figure 14.
EMP-EMP-Fc. A DNA sequence coding for a dimer of the EPO-mimetic peptide fused in-frame to the Fc region of human IgG1 was constructed using standard PCR technology. Templates for PCR reactions were the EMP-Fc plasmid from strain #3688 above and a synthetic gene . -encoding the EPO dieter. The synthetic gene for the dieter was constructed from the 8 overlapping oligonudeotides (SEQ ID NOS:408 to 415, respectively) shown below:

TAG AAG GAG GAA TAA AAT ATG

AA

GTT TGC AAA CCG

ATT TTA TTC CTC CTT C

CAT TTT GGC CCG CTG ACC TGG

ACC CTG CGG TTT GCA AAC CCA

AAA ACT CAC ACA TGT CCA

ACA TAC CCA GGT CAG CGG GCC
The 8 oligonudeotides were annealed to form the duplex encoding an amino acid sequence (SEQ ID NOS: 416 and 417, respectively) shown below: -TTTTTTATCGATTTGATTCTAGATTTGAGTTTTAACTTTTAGAAGGAGGAATAAAATATG
1 ---------+---------+---------+---------+---------+---------+ 60 AAAAAATAGCTAAACTAAGATCTAAACTCAAAATTGAAAATCTTCCTCCTTATTTTATAC
a M

GGAGGTACTTACTCTTGCCACTTCGGCCCGCTGACTTGGGTTTGCAAACCGCAGGGTGGC
61 ----------+---------+---------+---------+---------+---------+ 120 CCTCCATGAATGAGAACGGTGAAGCCGGGCGACTGAACCCAAACGTTTGGCGTCCCACCG
a G G T Y S C H P G P L T W V C K P Q G G -GGCGGCGGCGGCGGTGGTACCTATTCCTGTCATTTTGGCCCGCTGACCTGGGTATGTAAG
121 ---------+---------+--- -----+---------+---------+---------+ 180 CCGCCGCCGCCGCCACCATGGATAAGGACAGTAAAACCGGGCGACTGGACCCATACATTC
a G G G G G G T Y S C H F G P L T W V C K -CCACAAGGGGGTGGGGGAGGC TCACACATGTCCA
181 ---------+---------+---------+---------+-------- 228 GGTGTTCCCCCACCCCCTCCGCCCCCCCTGTTTTGA
a P Q G G G G G G G D K T H T C P -This duplex was amplified in a PCR reaction using 1869-23 and 1871-79 (shown above) as the sense and antisense primers.
The Fc portion of the molecule was generated in a PCR reaction with strain 3688 DNA using the primers 1798-23 and 1200-54 (shown above).
The oligonucleotides 1871-79 and 1798-23 contain an overlap of 31 nucleotides, allowing the two genes to be fused together in the correct reading frame by combining the above PCR products in a third reaction using the outside primers, 1869-23 and 1200-54.
The final PCR gene product (the full length fusion gene) was digested with restriction endonucleases XbaI and BamHI, and then ligated into the vector pAMG21 and transformed into competent E. coli strain 2596 cells as described for Fc-EMP. Clones were screened for ability to produce the recombinant protein product and possession of the gene fusion having the correct nucleotide sequence. A single such clone was selected and designated Amgen strain #3813.
The nucleotide and amino acid sequences (SEQ ID NOS: 19 and 20, respectively) of the resulting fusion protein are shown in Figure 15. There is a silent mutation at position 145 (A to G, shown in boldface) such that the.final construct has a different nucleotide sequence than the oligonudeotide 1871-72 from which it was derived.
Fc-ENTEMP. A DNA sequence coding for the Fc region of human IgG1 fused in-frame to a dimer of the EPO-mimetic peptide was constructed using standard PCR technology. Templates for PCR reactions were the plasmids from strains 3688 and 3813 above.
The Fc portion of the molecule was generated in a PCR reaction with strain 3688 DNA using the primers 1216-52 and 1798-17 (shown above). The EMP dieter portion of the molecule was the product of a second PCR reaction with strain 3813 DNA using the primers 1798-18 (also shown above) and SEQ ID NO: 418, shown below:

The oligonucleotides 1798-17 and 1798-18 contain an overlap of 61 nucleotides, allowing the two genes to be fused together in the correct reading frame by combining the above PCR products in a third reaction using the outside primers, 1216-52 and 1798-20.
The final PCR gene product (the full length fusion gene) was digested with restriction endonudeases XbaI and BamHI, and then ligated into the vector pAMG21 and transformed into competent E. coil strain 2596 cells as described for Fc-EMP. Clones were screened for the ability to produce the recombinant protein product and to possess the gene fusion having the correct nucleotide sequence. A single such done was selected and designated Amgen strain #3822.
The nucleotide and amino acid sequences (SEQ ID NOS: .21 and 22,, respectively) of the fusion protein are shown in Figure 16.
Characterization of Fc-EMP of Fc-EMP activity. Characterization was carried out in vivo as follows.
Mice: Normal female BDF1 approximately 10-12 weeks of age.
Bleed schedule: Ten mice per group treated on day 0, two groups started 4 days apart for a total of 20 mice per group.' Five mice bled at each time point, mice were bled a maximum of three times a week. Mice were anesthetized with isoflurane and a total volume of 140-160 ml of blood was obtained by puncture of the orbital sinus. Blood was counted on a Technicon H1E blood analyzer running software for murine blood.
Parameters measured were WBC, RBC, HCT, HGB, PLT, NEUT, LYMPH.
Treatments: Mice were either injected subcutaneously for a bolus treatment or implanted with 7 day micro-osmotic pumps for continuous delivery. Subcutaneous injections were delivered in a volume of 0.2 ml.
Osmotic pumps were inserted into a subcutaneous incision made in the skin between the scapulae of anesthetized mice. Compounds were diluted in PBS with 0.1% BSA. All experiments included one control group, labeled "carrier" that were treated with this diluent only. The concentration of the test articles in the pumps was adjusted so that the calibrated flow rate from the pumps gave the treatment levels indicated in the graphs.
Experiments: Various Fc-conjugated EPO mimetic peptides (EMPs) were delivered to mice as a single bolus injection at a dose of 100 g/kg.
Fc-EMPs were delivered to mice in 7-day micro-osmotic pumps. The pumps were not replaced at the end of 7 days. Mice were bled until day 51 when HGB and HCT returned to baseline levels.
Example 4 TNF-a inhibitors Fc-TNF-a inhibitors. A DNA sequence coding for the Fc region of human IgGi fused in frame to a monomer of the TNF-a inhibitory peptide was constructed using standard PCR technology. The Fc and 5 glycine linker portion of the molecule was generated in a PCR reaction with DNA
from the Fc-EMP fusion strain #3718 (see Example 3) using the sense primer 1216-52 and the antisense primer 2295-89 (SEQ ID NOS: 369 and 1112, respectively). The nucleotides encoding the TNF-a inhibitory peptide were provided by the PCR primer 2295-89 shown 'below:

CA 02347131 2004-02-09 j TGC GGC AGG AAG TCA CCA CCA CCT CCA CCT TTA CCC
The oligonucleotide 2295-89 overlaps the glycine linker and Fc portion of the template by 22 nucleotides, with the PCR resulting in the two genes being fused together in the correct reading frame.
The PCR gene product (the full length fusion gene) was digested with restriction endonudeases NdeI and BamHI, and then ligated into the vector pAMG21 and transformed into competent E. coli strain 2596 cells as described for EMP-Fc herein. Clones were screened for the ability to produce the recombinant protein product and to possess the gene fusion having the correct nucleotide sequence. A single such clone was selected and designated Amgen strain #4544.
The nucleotide and amino acid sequences (SEQ ID NOS: 1055 and 1056) of the fusion protein are shown in Figures 19A and 19B.
TNF-a inhibitor-Fc. A DNA sequence coding for a TNF-a inhibitory peptide fused in-frame to the Fc region of human IgG1 was constructed using standard PCR technology. The template for the PCR reaction was a plasmid containing an unrelated peptide fused via a five glycine linker to Fc. The nucleotides encoding the TNF-a inhibitory peptide were provided by the sense PCR primer 2295-88, with primer 1200-54 serving as the antisense primer (SEQ ID NOS: 1117 and 407, respectively). The primer sequences are shown below:

CAC CGT CCG GGT GGA GGC GGT GGG GAC AAA ACT

The oligonucleotide 2295-88 overlaps the glycine linker and Fc portion of the template by 24 nudeotides, with the PCR resulting in the two genes being fused together in the correct reading frame.

The PCR gene product (the full length fusion gene) was digested with restriction endonucleases NdeI and BamHI, and then ligated into the vector pAMG21 and transformed into competent E. coli strain 2596 cells as described for EMP-Fc herein. Clones were screened for the ability to produce the recombinant protein product and to possess the gene fusion having the correct nucleotide sequence. A single such clone was selected and designated Amgen strain #4543.
The nucleotide and amino acid sequences (SEQ ID NOS: 1057 and 1058) of the fusion protein are shown in Figures 20A and 20B.
Expression in E. coli. Cultures of each of the pAMG21-Fc-fusion constructs in E. coli GM221 were grown at 37 C in Luria Broth medium 0 containing 50 mg/ml kanamycin. Induction of gene product expression from the luxPR promoter was achieved following the addition of the synthetic autoinducer N-(3-oxohexanoyl)-DL-homoserine lactone to the is culture media to a final concentration of 20 ng/ml. Cultures were incubated at 37 C for a further 3 hours. After 3 hours, the bacterial cultures were examined by microscopy for the presence of inclusion bodies and were then collected by centrifugation. Refractile inclusion bodies were observed in induced cultures indicating that the Fc-fusions were most likely produced in the insoluble fraction in E. coli. Cell pellets were lysed directly by resuspension in Laemm i sample buffer containing 10% ti-mercaptoethanol and were analyzed by SDS-PAGE. In each case, an intense coomassie-stained band of the appropriate molecular weight was observed on an SDS-PAGE gel.
Purification of Fc-peptide fusion proteins. Cells are broken in water (1/10) by high pressure homogenization (2 passes at 14,000 PSI) and inclusion bodies are harvested by centrifugation (4200 RPM in J-6B for 1 hour). Inclusion bodies are solubilized in 6M guanidine, 50mM Tris, 8mM
DTT, pH 8.7 for 1 hour at a 1 /10 ratio. The solubilized mixture is diluted l 20 times into 2M urea, 50 mM tris, 160mM arginine, 3mM cysteine, pH 8.5.
The. mixture is stirred overnight in the cold and then concentrated about fold by ultafiltration. It is then diluted 3 fold with 10mM Tris, 1.5M
urea, pH 9. The pH of this mixture is then adjusted to pH 5 with acetic 5 acid. The precipitate is removed by centrifugation and the supernatant is loaded onto a SP-Sepharose Fast Flow column equilibrated in 20mM
NaAc, 100 mM NaG, pH 5 (10mg/ml protein load, room temperature).
The protein is eluted from the column using a 20 column volume gradient in the same buffer ranging from 100mM NaCl to 500mM NaCl. The pool 10 from the column is diluted 3 fold and loaded onto a SP-Sepharose HP
column in 20mM NaAc, 150mM NaCI, pH 5(10mg/ml protein load, room temperature). The protein is eluted using a 20 column volume gradient in the same buffer ranging from 150mM NaCl to 400mM NaCl. The peak is pooled and filtered.
Characterization of activity of Fc-TNF a inhibitor and.TNF-a inhibitor -Fc. Binding of these peptide fusion proteins to TNF- a can be characterized by BlAcore by methods available to one of ordinary skill in the art who is armed with the teachings of the present specification.
Example 5 IL -1 Antagonists Fc-IL-1 antagonist. A DNA sequence coding for the Fc region of human IgGi fused in frame to a monomer of an IL-1 antagonist peptide was constructed using standard PCR technology. The Fc and 5 glycine linker portion of the molecule was generated in a PCR reaction with DNA
from the Fc-EMP fusion strain #3718 (see Example 3) using the sense primer 1216-52 and the antisense primer 2269-70 (SEQ ID NOS: 369 and 1118, respectively). The nucleotides encoding the IL-1 antagonist peptide were provided by the PCR primer 2269-70 shown below:

GGG GTC CAT TCG AAA CCA CCA CCT CCA CCT TTA CCC

The oligonucleotide 2269-70 overlaps the glycine linker and Fc.portion of the template by 22 nucleotides, with the PCR resulting in the two genes being fused together in the correct reading frame.
The PCR gene product (the full length fusion gene) was digested with restriction endonucleases Ndel and BamHI, and then ligated into the vector pAMG21 and transformed into competent E. coli strain 2596 cells as described for EMP-Fc herein. Clones were screened for the ability to produce the recombinant protein product and to possess the gene fusion having the correct nucleotide sequence. A single such clone was selected and designated Amgen strain #4506.
The nucleotide and amino acid sequences (SEQ ID NOS: 1059 and 1060) of the fusion protein are shown in Figures 21A and 21B.
IL-i antagonist-Fc. A DNA sequence coding for an IL-1 antagonist peptide fused in-frame to the Fc region of human IgG1 was constructed using standard PCR technology. The template for the PCR reaction was a plasmid containing an unrelated peptide fused via a five glycine linker to Fc. The nucleotides encoding the IL-1 antagonist peptide were provided by the sense PCR primer 2269-69, with primer 1200-54 serving as the antisense primer (SEQ ID NOS: 1119 and 407, respectively). The primer sequences are shown below:

CTG CCG CTG GGT GGA GGC GGT GGG GAC AAA ACT

WO 00/24782 PCT/US99n5044 The.oligonucleotide 2269-69 overlaps the glycine linker and Fc portion of the template by 24 nucleotides, with the PCR resulting in the two genes being fused together in the correct reading frame.
The PCR gene product (the full length fusion gene) was digested with restriction endonucleases NdeI and BamH, and then ligated into the vector pAMG21 and transformed into competent E. coil strain 2596 cells as described for EMP-Fc herein. Clones were screened for the ability to produce the recombinant protein product and to possess the gene fusion having the correct nucleotide sequence. A single such clone was selected and designated Amgen strain #4505. . -The nucleotide and amino acid sequences (SEQ ID NOS: 1061 and 1062) of the fusion protein are shown in Figures 22A and 22B. Expression and purification were carried out as in previous examples.
Characterization of Fc-IL-1 antagonist peptide and IL-1 antagonist peptide-Fc activity. IL-1 Receptor Binding competition between IL-113, IL-1RA and Fc-conjugated IL-1 peptide sequences was carried out using the IGEN system. Reactions contained 0.4 nM biotin-IL-1R + 15 nM IL-i-TAG
+ 3 uM competitor + 20 ug/ml streptavidin-conjugate. beads, where competitors were IL-1RA, Fc-IL-1 antagonist, IL-i antagonist-FO.
Competition was assayed over a range of competitor concentrations from 3 uM to 1.5 pM. The results are shown in Table C below:

Table C-Results from IL-1 Receptor Binding Competition Assay IL-1pep-Fc Fc-IL-Ipop IL-Irv KI 281.5 59.58 1.405 EC50 530.0 112.2 2.645 95% Confidence Intervals EC50 280.2 to 1002 54.75 to 229.8 1.149 to 6.086 KI 148.9 to 532.5 29.08 to 122.1 0.6106 to 3.233 Goodness of Fit R2 0.9790 0.9687 0.9602 Example 6 VEGF-Antagonists Fc-VEGF Antagonist. A DNA sequence coding for the Fc region of human IgG1 fused in-frame to a monomer of the VEGF mimetic peptide was constructed using standard PCR technology. The templates for the PCR reaction were the pFc-A3 plasmid and a synthetic VEGF mimetic peptide gene. The synthetic gene was assembled by annealing the following two oligonucleotides primer (SEQ ID NOS: 1110 and 1111, respectively):

TGT TTT GAA CGT CTG

ACA GTT CGG TTC AAC

The two oligonucleotides anneal to form the following duplex encoding an amino acid sequence shown below (SEQ ID NOS 1113 and 1114)-GTTGAACCGAACTGTGACATCCATGTTATGTGGGAATGGGAATGTTTTGAACGTCTG
1 ---------+---------+---------+---------+---------+------- 57 CAACTTGGCTTGACACTGTAGGTACAATACACCCTTACCCTTACAAAACTTGCAGAC

a V E P N C D I H V M W E W E C F E R L

This duplex was amplified in a PCR reaction using 2293-05 and 2293-06 as the sense and antisense primers (SEQ ID Nom. 1 122 and 1123).
The Fc portion of the molecule was generated in a PCR reaction with the pFc-A3 plasmid using the primers 2293-03 and 2293-04 as-the sense and antisense primers (SEQ ID NOS. 1120 and 1121, respectively).
The full length fusion gene was obtained from a third PCR reaction using the outside primers 2293-03 and 2293-06. These primers are shown below:

ACA TGT

AGA CAG GGA

AAC TGT GAC ATC

The PCR gene product (the full length fusion gene) was digested with restriction endonudeases NdeI and BamHI, and then ligated into the vector pAMG21 and transformed into competent E. coli strain 25% cells as described for EMP-Fc herein. Clones were screened for the ability to produce the recombinant protein product and to possess the gene fusion having the correct nucleotide sequence. A single such done was selected and designated Amgen strain #4523.
The nucleotide and amino acid sequences (SEQ ID NOS: 1063 and 1064) of the fusion protein are shown in Figures 23A and 23B.
VEGF antagonist -Fc. A DNA sequence coding for a VEGF mimetic peptide fused in frame to the Fc region of human IgG1 was constructed using standard PCR technology. The templates for the PCR reaction were the pFc-A3 plasmid and the synthetic VEGF mimetic peptide gene described above. The synthetic duplex was amplified in a PCR reaction using 2293-07 and 2293-08 as the sense and antisense primers (SEQ ID
NOS. 1124 and 1125, respectively).
The Fc portion of the molecule was generated in a PCR reaction with the pFc-A3 plasmid using the primers 2293-09 and 2293-10 as the sense and antisense primers (SEQ ID NOS. 1126 and 1127, respectively).

The full length fusion gene was obtained from a third PCR reaction using the outside primers 2293-07 and 2293-10. These primers are shown below:

TGT GAC

AAA ACA TTC

10' 2293-09 GAA TGT TTT GAA CGT CTG GGT GGT GGT GGT GGT GAC AAA ACT
CAC ACA TGT

The PCR gene product (the full length fusion gene) was digested with restriction endonudeases NdeI and BamHI, and then ligated into the vector pAMG21 and transformed into competent E. coli strain 2596 cells as described for EMP-Fc herein. Clones were screened for the ability to produce the recombinant protein product and to possess the gene fusion having the correct nucleotide sequence. A single such done was selected and designated Amgen strain #4524.
The nucleotide and amino acid sequences (SEQ ID NOS: 1065 and 1066) of the fusion protein are shown in Figures 24A and 24B. Expression and purification were carried out as in previous examples.

Emile 7 MMP Inhibitors Fc-MMP inhibitor. A DNA sequence coding for the Fc region of human IgG1 fused in-frame to a monomer of an MMP inhibitory peptide was constructed using standard PCR technology. The Fc and 5 glycine liinker portion of the molecule was generated in a PCR reaction with DNA
from the Fc-TNF-a inhibitor fusion strain #4544 (see Example 4) using the sense primer 1216-52 and the antisense primer 2308-67 (SEQ ID NOS: 369 and 1115, respectively). The nucleotides encoding the MMP inhibitor peptide were provided by the PCR primer 2308-67 shown below:

CAA CCA CCA CCT CCA CCT TTA CCC
The oligonucleotide 2308-67 overlaps the glycine linker and Fc portion of the template by 22 nucleotides, with the PCR resulting in the two genes being fused together in the correct reading frame.
The PCR gene product (the full length fusion gene) was digested with restriction endonucleases NdeI and BamHI, and then ligated into the vector.pAMG21 and transformed into competent E. coli strain 2596 cells as described for EMP-Fc herein. Clones were screened for the ability to produce the recombinant protein product and to possess the gene fusion having the correct nucleotide sequence. A single such clone was selected and designated Amgen strain #4597.
The nucleotide and amino acid sequences (SEQ ID NOS: 1067 and 1068) of the fusion protein are shown in Figures 25A and 25B. Expression and purification were carried out as in previous examples.
MMP Inhibitor-Fc. A DNA sequence coding for an MMP inhibitory peptide fused in-frame to the Fc region of human IgG1 was constructed using standard PCR technology. The Fc and 5 glycine linker portion of the molecule was generated in a PCR reaction with DNA from the Fc-TNF-a inhibitor fusion strain #4543 (see Example 4). The nucleotides encoding the MMP inhibitory peptide were provided by the sense PCR primer 2308-66, with primer 1200-54 serving as the antisense primer (SEQ ID NOS:
1116 and 407, respectively). The primer sequences are shown below:

GGT GGA GGC GGT GGG GAC AAA

.The oligonucleotide 2269-69 overlaps the glycine linker and Fc portion of the template by 24 nudeotides, with the PCR resulting in the two genes being fused together in the correct reading frame.
The PCR gene product (the full length fusion gene) was digested with restriction endonucleases NdeI and BamHI, and then ligated into the vector pAMG21 and transformed into competent E. coli strain 2596 cells as described for EMP-Fc herein. Clones were screened for the ability to produce the recombinant protein product and to possess the gene fusion having the correct nucleotide sequence. A single such done was selected and designated Amgen strain #4598.-The nucleotide and amino acid sequences (SEQ ID NOS: 1069 and 1070) of the fusion protein are shown in Figures 26A and 26B.
* *

The invention now being fully described; it will be apparent to one of ordinary skill in the art that many changes and modifications can be made thereto, without departing from the spirit and scope of the invention as set forth herein.

Abbreviations Abbreviations used throughout this specification are as defined below, unless otherwise defined in specific circumstances.
Ac acetyl (used to refer to acetylated residues) AcBpa acetylated p-benzoyl-L-phenylalanine ADCC antibody-dependent cellular cytotoxicity Aib aminoisobutyric acid bA beta-alandne Bpa p-benzoyl-L-phenylalanine BrAc bromoacetyl BrCH2C(O) BSA Bovine serum albumin Bzl - Benzyl Cap Caproic acid CTL Cytotoxic T lymphocytes CTLA4 Cytotoxic T lymphocyte antigen 4 DARC Duffy blood group antigen receptor DCC Dicylcohexylcarbodiimide Dde 1-(4,4-dimethyl-2,6-dioxo-cyclohexylidene)ethyl EMP Erythropoietin-mimetic peptide ESI-MS Electron spray ionization mass spectrometry EPO Erythropoietin Fmoc fluorenylmethoxycarbonyl G-CSF Granulocyte colony stimulating factor -GH Growth hormone HCT hematocrit HGB hemoglobin hGH Human growth hormone HOBt 1-Hydroxybenzotriazole HPLC high performance liquid chromatography IL interleuldn IL-R interleukin receptor ILAR interleukin-1 receptor IL-Ira interleukin-1 receptor antagonist Lau Lauric acid LPS lipopolysaccharide LYMPH lymphocytes -. MALDI-MS Matrix-assisted laser desorption ionization-mass spectrometry Me methyl MeO methoxy MHC major histocompatibility complex MMP matrix metalloproteinase MMPI matrix metalloproteinase inhibitor 1-Nap 1-napthylalanine NEUT neutrophils NGF nerve growth factor Me norleucine NMP N-methyl-2-pyrrolidinone PAGE polyacrylamide gel electrophoresis PBS Phosphate-buffered saline Pbf 2,2,4,6,7 pendamethyldihydrobenzofuran-5-sulfonyl PCR polymerase chain reaction Pec pipecolic acid PEG Poly(ethylene glycol) pGlu pyroglutamic acid Pic picolinic acid PLT platelets pY phosphotyrosine RBC red blood cells RBS ribosome binding site RT room temperature (25 C) Sar sarcosine SDS sodium dodecyl sulfate STK serine-threonine kinases t-Boc tert-Butoxycarbonyl tBu tert-Butyl TGF tissue growth factor THE thymic humoral factor TK tyrosine kinase TMP Thrombopoietin-mimetic peptide TNF Tissue necrosis factor TPO Thrombopoietin TRAIL TNF-related apoptosis-inducing ligand Trt trityl UK urokinase UKR urokinase receptor VEGF vascular endothelial cell growth factor VII' vasoactive intestinal peptide WBC white blood cells

Claims (9)

Claims:
1. A composition of matter of the formula (X1)a F1-(X2)b and multimers thereof, wherein:
F1 is an F c domain;
X1 and X2 are each independently selected from -(L1),-P1, -(L1)c-P1-(L2)d-P2, -(L1)c-P1-(L-2)d-P1-(L3)e-P3, and -(L1)c-P1-(L2)d-P2 -(L3)e-P3-(L4)f-P4;
P1, P2, P3 and P4 are each independently randomly generated TPO-mimetic peptide sequences;

L1, L2, L3 and L4 are each independently linkers; and a, b, c, d, e, and f are each independently 0 or 1, provided that at least one of a and b is 1, and wherein the term peptide refers to molecules having 3 to 20 amino acids, and wherein neither X1 nor X2 is a native protein.
2. The composition of matter of Claim 1 of the formulae or F1-X2.
3. The composition of matter of Claim 1 of the formula F1-(L1)c-P1.
4. The composition of matter of Claim 1 of the formula F1-(L1)c-P1-(L2)d-P2.
5. The composition of matter of any one of Claims 1 to 4 wherein F1 is an IgG Fc domain.
6. The composition of matter of any one of Claims 1 to 4 wherein F1 is an IgG1 Fc domain.
7. The composition of matter of any one of Claims 1 to 4 wherein F1 comprises the sequence of SEQ ID NO: 2.
8. The composition of matter of any one of Claims 1 to 7 wherein P1 is a TPO-mimetic peptide sequence selected from SEQ ID NOS: 24, 25, 27, 28, 31, 33 to 82.
9. The composition of matter of Claim 1 having the sequence of SEQ ID NO:
6.
CA2347131A 1998-10-23 1999-10-25 Modified peptides as therapeutic agents Expired - Lifetime CA2347131C (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US10537198P 1998-10-23 1998-10-23
US60/105,371 1998-10-23
US09/428,082 US6660843B1 (en) 1998-10-23 1999-10-22 Modified peptides as therapeutic agents
US09/428,082 1999-10-22
PCT/US1999/025044 WO2000024782A2 (en) 1998-10-23 1999-10-25 Modified peptides, comprising an fc domain, as therapeutic agents

Publications (2)

Publication Number Publication Date
CA2347131A1 CA2347131A1 (en) 2000-05-04
CA2347131C true CA2347131C (en) 2012-11-27

Family

ID=26802505

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2347131A Expired - Lifetime CA2347131C (en) 1998-10-23 1999-10-25 Modified peptides as therapeutic agents

Country Status (29)

Country Link
US (7) US6660843B1 (en)
EP (1) EP1144454B2 (en)
JP (9) JP2003512011A (en)
KR (3) KR100753305B1 (en)
CN (8) CN100384879C (en)
AT (1) ATE380828T1 (en)
AU (3) AU767725B2 (en)
BG (1) BG65721B1 (en)
BR (1) BR9914708A (en)
CA (1) CA2347131C (en)
CY (1) CY1107881T1 (en)
CZ (1) CZ304242B6 (en)
DE (1) DE69937752T3 (en)
DK (1) DK1144454T4 (en)
EA (1) EA005404B1 (en)
ES (1) ES2299278T5 (en)
HK (2) HK1042097B (en)
HU (1) HU229485B1 (en)
IL (2) IL142365A0 (en)
MX (1) MXPA01003873A (en)
NO (1) NO331733B1 (en)
NZ (2) NZ528882A (en)
PL (1) PL211164B1 (en)
PT (1) PT1144454E (en)
RS (1) RS51852B (en)
SI (1) SI1144454T2 (en)
SK (1) SK287037B6 (en)
WO (1) WO2000024782A2 (en)
ZA (1) ZA200102753B (en)

Families Citing this family (548)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6743777B1 (en) * 1992-03-19 2004-06-01 Eli Lilly And Company Cyclic peptide antifungal agents and process for preparation thereof
US7091311B2 (en) * 1996-06-07 2006-08-15 Smithkline Beecham Corporation Peptides and compounds that bind to a receptor
US7488590B2 (en) 1998-10-23 2009-02-10 Amgen Inc. Modified peptides as therapeutic agents
US6660843B1 (en) * 1998-10-23 2003-12-09 Amgen Inc. Modified peptides as therapeutic agents
MY126795A (en) 1998-10-23 2006-10-31 Amgen K A Inc Dimeric thrombopoietic peptide mimetics binding to mp1 receptor and having thrombopoietic activity.
WO2000047740A2 (en) * 1999-02-12 2000-08-17 Amgen Inc. Tnf-related proteins
WO2000051564A1 (en) * 1999-03-03 2000-09-08 Eli Lilly And Company Echinocandin pharmaceutical formulations containing micelle-forming surfactants
EP1157030B1 (en) 1999-03-03 2003-08-27 Eli Lilly And Company Echinocandin/carbohydrate complexes
JP2003503426A (en) 1999-07-02 2003-01-28 ジェネンテック・インコーポレーテッド FVIIa antagonist
EP1189931B1 (en) 1999-07-02 2006-06-21 Genentech, Inc. Peptide compounds that bind her2
CA2379388C (en) * 1999-07-13 2012-05-29 George N. Cox, Iii Immunoglobulin fusion proteins
SK782002A3 (en) 1999-07-21 2003-08-05 Lexigen Pharm Corp FC fusion proteins for enhancing the immunogenicity of protein and peptide antigens
EP1274723A4 (en) * 1999-07-29 2003-10-01 Univ Johns Hopkins ACTIVATION OF PEPTIDE PRODRUGS BY hK2
US7459540B1 (en) 1999-09-07 2008-12-02 Amgen Inc. Fibroblast growth factor-like polypeptides
US6808902B1 (en) * 1999-11-12 2004-10-26 Amgen Inc. Process for correction of a disulfide misfold in IL-1Ra Fc fusion molecules
MXPA02010324A (en) * 2000-04-21 2003-04-25 Amgen Inc Apo ai aii peptide derivatives.
AU2001259432B2 (en) * 2000-05-03 2005-04-21 Amgen Inc. Modified peptides, comprising an FC domain, as therapeutic agents
US20020090646A1 (en) * 2000-05-03 2002-07-11 Amgen Inc. Calcitonin-related molecules
US6774106B2 (en) 2000-05-12 2004-08-10 Amgen Inc. Methods and compositions of matter concerning APRIL/G70, BCMA, BLYS/AGP-3 and TACI
US7259146B2 (en) 2000-05-26 2007-08-21 Ortho-Mcneil Pharmaceutical, Inc. Neuroprotective peptides
JP2003534384A (en) * 2000-05-26 2003-11-18 オーソ−マクニール・フアーマシユーチカル・インコーポレーテツド Neuroprotective peptide
CA2410526C (en) * 2000-06-02 2012-04-17 Eidgenossische Technische Hochschule Zurich Conjugate addition reactions for the controlled delivery of pharmaceutically active compounds
US7355019B2 (en) * 2000-06-06 2008-04-08 Sibtech, Inc. Cysteine-containing peptide tag for site-specific conjugation of proteins
JP2004508043A (en) * 2000-09-05 2004-03-18 アムジェン インコーポレイテッド TNF receptor-like molecules and uses thereof
AU2003295623B2 (en) * 2000-12-05 2008-06-05 Alexion Pharmaceuticals, Inc. Rationally designed antibodies
US7396917B2 (en) 2000-12-05 2008-07-08 Alexion Pharmaceuticals, Inc. Rationally designed antibodies
EP1498429A3 (en) * 2000-12-05 2005-08-03 Alexion Pharmaceuticals, Inc. Rationally designed antibodies
US20040253242A1 (en) * 2000-12-05 2004-12-16 Bowdish Katherine S. Rationally designed antibodies
US7754208B2 (en) 2001-01-17 2010-07-13 Trubion Pharmaceuticals, Inc. Binding domain-immunoglobulin fusion proteins
US7829084B2 (en) * 2001-01-17 2010-11-09 Trubion Pharmaceuticals, Inc. Binding constructs and methods for use thereof
US20030133939A1 (en) 2001-01-17 2003-07-17 Genecraft, Inc. Binding domain-immunoglobulin fusion proteins
US7622446B2 (en) * 2001-04-18 2009-11-24 The Open University Polypeptides, derivatives and uses thereof
US7491702B2 (en) * 2001-04-18 2009-02-17 The Open University Polypeptides related to amyloid precursor protein, pharmaceutical compositions thereof, and methods of treatment using the same
ATE549354T1 (en) 2001-05-11 2012-03-15 Amgen Inc PEPTIDES AND ASSOCIATED TALL-1 BINDING MOLECULES
IL159512A0 (en) * 2001-06-26 2004-06-01 Abgenix Inc Antibodies to opgl
DE60232742D1 (en) 2001-10-04 2009-08-06 Immunex Corp UL16 BINDING PROTEIN 4
US7205275B2 (en) 2001-10-11 2007-04-17 Amgen Inc. Methods of treatment using specific binding agents of human angiopoietin-2
MX339524B (en) * 2001-10-11 2016-05-30 Wyeth Corp Novel immunogenic compositions for the prevention and treatment of meningococcal disease.
US7138370B2 (en) 2001-10-11 2006-11-21 Amgen Inc. Specific binding agents of human angiopoietin-2
US7521053B2 (en) 2001-10-11 2009-04-21 Amgen Inc. Angiopoietin-2 specific binding agents
US7332474B2 (en) 2001-10-11 2008-02-19 Amgen Inc. Peptides and related compounds having thrombopoietic activity
US20030113270A1 (en) * 2001-12-14 2003-06-19 Clark Abbot F. Vasoactive intestinal peptides for glaucomatous retinopathy
US20030138975A1 (en) * 2001-12-20 2003-07-24 Kimberly-Clark Worldwide, Inc. Diagnostic signal amplification with proteinoid microspheres
US7056535B2 (en) * 2001-12-20 2006-06-06 Kimberly-Clark Worldwide, Inc. Triggered release from proteinoid microspheres
US8188231B2 (en) 2002-09-27 2012-05-29 Xencor, Inc. Optimized FC variants
US8093357B2 (en) 2002-03-01 2012-01-10 Xencor, Inc. Optimized Fc variants and methods for their generation
US20040132101A1 (en) 2002-09-27 2004-07-08 Xencor Optimized Fc variants and methods for their generation
US7317091B2 (en) 2002-03-01 2008-01-08 Xencor, Inc. Optimized Fc variants
DE10209822A1 (en) 2002-03-06 2003-09-25 Biotechnologie Ges Mittelhesse Coupling of low molecular weight substances to a modified polysaccharide
DE10209821A1 (en) 2002-03-06 2003-09-25 Biotechnologie Ges Mittelhesse Coupling of proteins to a modified polysaccharide
JP2005521401A (en) 2002-03-27 2005-07-21 イミュネックス・コーポレーション Methods for increasing polypeptide production
US20030191056A1 (en) 2002-04-04 2003-10-09 Kenneth Walker Use of transthyretin peptide/protein fusions to increase the serum half-life of pharmacologically active peptides/proteins
US7718776B2 (en) 2002-04-05 2010-05-18 Amgen Inc. Human anti-OPGL neutralizing antibodies as selective OPGL pathway inhibitors
US6991800B2 (en) * 2002-06-13 2006-01-31 Vicuron Pharmaceuticals Inc. Antifungal parenteral products
AU2003280130B2 (en) * 2002-06-28 2009-06-11 Centocor, Inc. Mammalian CH1 deleted mimetibodies, compositions, methods and uses
EP1394180B1 (en) * 2002-08-06 2008-07-16 AplaGen GmbH Synthetic mimetic molecules of physiological binding molecules
JP4667868B2 (en) * 2002-08-06 2011-04-13 アプラーゲン ゲゼルシャフト ミット ベシュレンクテル ハフツング Binding molecule
PL376536A1 (en) 2002-08-28 2006-01-09 Immunex Corporation Compositions and methods for treating cardiovascular disease
NZ593428A (en) 2002-09-06 2013-01-25 Amgen Inc Therapeutic human anti-il-1r1 monoclonal antibody
BR0314227A (en) * 2002-09-11 2005-10-25 Fresenius Kabi De Gmbh Hydroxyalkyl Starch Derivatives
EA010759B1 (en) 2002-09-18 2008-10-30 Орто-Макнейл Фармасьютикал, Инк. Methods of increasing platelet and hematopoietic stem cell production
US6919426B2 (en) 2002-09-19 2005-07-19 Amgen Inc. Peptides and related molecules that modulate nerve growth factor activity
WO2004029207A2 (en) 2002-09-27 2004-04-08 Xencor Inc. Optimized fc variants and methods for their generation
US20040149235A1 (en) * 2002-10-04 2004-08-05 Pogue Albert S. Apparatus and method for removal of waste from animal production facilities
DE60327795D1 (en) 2002-11-09 2009-07-09 Immunocore Ltd T CELL RECEPTOR "DISPLAY"
CA2510893C (en) 2002-12-20 2012-07-10 Amgen, Inc. Binding agents which inhibit myostatin
US7125849B2 (en) * 2003-01-14 2006-10-24 The Scripps Research Institute Peptide-based angiogenesis inhibitors and methods of use thereof
DE10303974A1 (en) 2003-01-31 2004-08-05 Abbott Gmbh & Co. Kg Amyloid β (1-42) oligomers, process for their preparation and their use
KR20050102634A (en) * 2003-02-06 2005-10-26 메르크 파텐트 게엠베하 Peptidic sulfonamides
US8388955B2 (en) 2003-03-03 2013-03-05 Xencor, Inc. Fc variants
US20090010920A1 (en) 2003-03-03 2009-01-08 Xencor, Inc. Fc Variants Having Decreased Affinity for FcyRIIb
US8084582B2 (en) 2003-03-03 2011-12-27 Xencor, Inc. Optimized anti-CD20 monoclonal antibodies having Fc variants
US9051373B2 (en) 2003-05-02 2015-06-09 Xencor, Inc. Optimized Fc variants
PT2298347E (en) * 2003-05-06 2016-01-29 Biogen Hemophilia Inc Clotting factor chimeric proteins for treatment of a hemostatic disorder
TWI353991B (en) * 2003-05-06 2011-12-11 Syntonix Pharmaceuticals Inc Immunoglobulin chimeric monomer-dimer hybrids
US7348004B2 (en) * 2003-05-06 2008-03-25 Syntonix Pharmaceuticals, Inc. Immunoglobulin chimeric monomer-dimer hybrids
AU2004238263A1 (en) * 2003-05-06 2004-11-25 Syntonix Pharmaceuticals, Inc. Inhibition of drug binding to serum albumin
WO2004108885A2 (en) * 2003-05-06 2004-12-16 Syntonix Pharmaceuticals, Inc. Fc chimeric proteins with anti-hiv drugs
WO2004101600A2 (en) * 2003-05-12 2004-11-25 Affymax, Inc. Novel poly(ethylene glycol) modified compounds and uses thereof
KR101163683B1 (en) * 2003-05-12 2012-07-10 아피맥스, 인크. Novel peptides that bind to the erythropoietin receptor
EA010099B1 (en) * 2003-05-12 2008-06-30 Афимакс, Инк. Novel peptides that bind to the erythropoietin receptor and methods for use thereof
MXPA05012314A (en) * 2003-05-12 2006-04-18 Affymax Inc Spacer moiety for poly(ethylene glycol) -modified peptides.
US7485312B2 (en) * 2003-06-20 2009-02-03 Dade Behring Marburg Gmbh Surface protein (HBsAg) variant of the hepatitis B virus
EA015363B1 (en) 2003-07-18 2011-06-30 Эмджен Инк. Specific binding agents to hepatocyte growth factor and use thereof
AU2004261941B2 (en) 2003-07-25 2008-04-10 Amgen Inc. Antagonists and agonists of LDCAM and methods of use
WO2005014655A2 (en) * 2003-08-08 2005-02-17 Fresenius Kabi Deutschland Gmbh Conjugates of hydroxyalkyl starch and a protein
ATE447611T1 (en) * 2003-08-18 2009-11-15 Univ California POLYPEPTIDE DISPLAY LIBRARIES AND METHOD FOR THE PRODUCTION AND USE THEREOF
ES2304069B1 (en) * 2003-08-22 2009-08-12 Proyecto De Biomedicina Cima, S.L. PEPTIDES ABLE TO JOIN THE TRANSFORMING FACTOR OF BETA 1 GROWTH (TGF-B1).
US8158589B2 (en) 2003-08-22 2012-04-17 Proyecto Biomedicine Cima, S.L. Peptides with the capacity to bind to transforming growth factor β1 (TGF-β1)
US8101720B2 (en) 2004-10-21 2012-01-24 Xencor, Inc. Immunoglobulin insertions, deletions and substitutions
US9714282B2 (en) 2003-09-26 2017-07-25 Xencor, Inc. Optimized Fc variants and methods for their generation
US20060127404A1 (en) * 2003-09-30 2006-06-15 Chichi Huang Hinge core mimetibodies, compositions, methods and uses
UA89481C2 (en) * 2003-09-30 2010-02-10 Центокор, Инк. Human epo mimetic hinge core mimetibodies, compositions, methods and uses
EP1684791A4 (en) 2003-10-27 2009-07-01 Amgen Inc Compositions and methods to modulate an immune response to an immunogenic therapeutic agent
US8110665B2 (en) 2003-11-13 2012-02-07 Hanmi Holdings Co., Ltd. Pharmaceutical composition comprising an immunoglobulin FC region as a carrier
PL2256134T3 (en) * 2003-11-13 2014-06-30 Hanmi Science Co Ltd IgG Fc fragment for a drug carrier and method for the preparation thereof
US20110160440A1 (en) 2003-12-12 2011-06-30 Genencor International, Inc. Cab Molecules
AU2004315198A1 (en) * 2004-01-29 2005-08-18 Genentech, Inc. Variants of the extracellular domain of BCMA and uses thereof
WO2005092391A2 (en) * 2004-03-11 2005-10-06 Fresenius Kabi Deutschland Gmbh Conjugates of hydroxyalkyl starch and a protein
JP5191729B2 (en) 2004-03-11 2013-05-08 フレゼニウス・カビ・ドイチュラント・ゲゼルシャフト・ミット・ベシュレンクテル・ハフツング Conjugates of hydroxyalkyl starch and protein produced by reductive amination
ES2694252T3 (en) * 2004-04-02 2018-12-19 Swedish Orphan Biovitrum Ab (Publ) Methods to reduce the aggregation of IL-1ra
US20050222040A1 (en) * 2004-04-05 2005-10-06 Blm Group, Inc. Vertebrate peptide modulators of lipid metabolism
US8088609B2 (en) 2004-04-15 2012-01-03 Danisco Us Inc. CAB molecules
JP2005309295A (en) * 2004-04-26 2005-11-04 Nec Corp Element, device, and system for optical amplification
US20050281739A1 (en) * 2004-06-16 2005-12-22 Glen Gong Imaging damaged lung tissue using compositions
US20050281799A1 (en) * 2004-06-16 2005-12-22 Glen Gong Targeting damaged lung tissue using compositions
US20050281798A1 (en) * 2004-06-16 2005-12-22 Glen Gong Targeting sites of damaged lung tissue using composition
US7608579B2 (en) * 2004-06-16 2009-10-27 Pneumrx, Inc. Lung volume reduction using glue compositions
US7553810B2 (en) * 2004-06-16 2009-06-30 Pneumrx, Inc. Lung volume reduction using glue composition
US20050281740A1 (en) * 2004-06-16 2005-12-22 Glen Gong Imaging damaged lung tissue
US7678767B2 (en) 2004-06-16 2010-03-16 Pneumrx, Inc. Glue compositions for lung volume reduction
KR101100059B1 (en) * 2004-06-30 2011-12-29 넥타르 테라퓨틱스 Polymer-factor ix moiety conjugates
CA2572765C (en) * 2004-07-08 2013-05-21 Amgen Inc. Compound having improved bioefficiency when administered in a multidose regimen
JP5113519B2 (en) 2004-07-08 2013-01-09 ヌームアールエックス・インコーポレーテッド Treatment device, treatment method and material for pleural effusion
US20150010550A1 (en) 2004-07-15 2015-01-08 Xencor, Inc. OPTIMIZED Fc VARIANTS
ME00226B (en) 2004-07-15 2011-02-10 Medarex Llc Human anti-ngf neutralizing antibodies as selective ngf pathway inhibitors
US8080245B2 (en) * 2004-08-04 2011-12-20 University Of Massachusetts Anti-pathogen immunoadhesins
US20060210542A1 (en) * 2004-08-16 2006-09-21 Yurkow Edward J Use of TPO mimetic compounds and pharmaceutical compositions in the treatment of anemia
US7393662B2 (en) * 2004-09-03 2008-07-01 Centocor, Inc. Human EPO mimetic hinge core mimetibodies, compositions, methods and uses
EP1797127B1 (en) * 2004-09-24 2017-06-14 Amgen Inc. Modified fc molecules
EP1799700A4 (en) * 2004-09-27 2009-02-11 Centocor Inc Srage mimetibody, compositions, methods and uses
WO2006052493A1 (en) * 2004-11-04 2006-05-18 Genentech, Inc. Polypeptides that bind baff and/or april
WO2006062685A2 (en) * 2004-11-11 2006-06-15 Affymax, Inc. Novel peptides that bind to the erythropoietin receptor
EP1814910A4 (en) * 2004-11-11 2009-04-29 Affymax Inc Novel peptides that bind to the erythropoietin receptor
US8367805B2 (en) 2004-11-12 2013-02-05 Xencor, Inc. Fc variants with altered binding to FcRn
US8802820B2 (en) 2004-11-12 2014-08-12 Xencor, Inc. Fc variants with altered binding to FcRn
US8546543B2 (en) 2004-11-12 2013-10-01 Xencor, Inc. Fc variants that extend antibody half-life
KR101027427B1 (en) 2004-11-12 2011-04-11 젠코어 인코포레이티드 Fc VARIANTS WITH INCREASED BINDING TO FcRn
US20070110760A1 (en) * 2005-01-14 2007-05-17 Monroe John G Methods and compositions targeting viral and cellular ITAM motifs, and use of same in identifying compounds with therapeutic activity
US8053415B2 (en) * 2005-01-21 2011-11-08 Washington University In St. Louis Compounds having RD targeting motifs
WO2006088245A1 (en) * 2005-02-18 2006-08-24 The University Of Tokushima Polyoxyalkylene chain-containing lipid derivative and lipid film structure containing such derivative
US7723472B2 (en) * 2005-02-28 2010-05-25 The Regents Of The University Of California Extracellular matrix binding chimeric proteins and methods of use thereof
US20060241040A1 (en) * 2005-04-06 2006-10-26 Alberto Visintin Methods of treating disorders associated with toll-like receptor 4 (TLR4) signalling
US7833979B2 (en) * 2005-04-22 2010-11-16 Amgen Inc. Toxin peptide therapeutic agents
DK1877102T3 (en) 2005-04-28 2014-07-21 Danisco Us Inc TAB MOLECULES
US8324159B2 (en) * 2005-06-03 2012-12-04 Affymax, Inc. Erythropoietin receptor peptide formulations and uses
US7550433B2 (en) * 2005-06-03 2009-06-23 Affymax, Inc. Erythropoietin receptor peptide formulations and uses
US7919461B2 (en) 2005-06-03 2011-04-05 Affymax, Inc. Erythropoietin receptor peptide formulations and uses
CA2610839C (en) 2005-06-14 2019-06-25 Amgen Inc. Self-buffering protein formulations
EP1907417A2 (en) * 2005-06-23 2008-04-09 AplaGen GmbH Supravalent compounds
US7566456B2 (en) * 2005-06-23 2009-07-28 Haiming Chen Allergen vaccine proteins for the treatment and prevention of allergic diseases
KR20130080058A (en) * 2005-06-30 2013-07-11 아보트 러보러터리즈 Il-12/p40 binding proteins
EP2388277A3 (en) 2005-07-18 2013-03-20 Amgen, Inc Human anti-B7RP1 neutralizing antibodies
HUE026303T2 (en) 2005-07-25 2016-06-28 Emergent Product Dev Seattle B-cell reduction using CD37-specific and CD20-specific binding molecules
US7790679B2 (en) 2005-08-05 2010-09-07 Amgen Inc. Pharmaceutical formulations
US8008453B2 (en) 2005-08-12 2011-08-30 Amgen Inc. Modified Fc molecules
MY149128A (en) * 2005-08-16 2013-07-15 Hanmi Science Co Ltd A method for the mass production of immunoglobulin fc region deleted initial methionine residues
US7612181B2 (en) * 2005-08-19 2009-11-03 Abbott Laboratories Dual variable domain immunoglobulin and uses thereof
US20090215992A1 (en) * 2005-08-19 2009-08-27 Chengbin Wu Dual variable domain immunoglobulin and uses thereof
WO2007027935A2 (en) 2005-08-31 2007-03-08 The Regents Of The University Of California Cellular libraries of peptide sequences (clips) and methods of using the same
KR100780405B1 (en) * 2005-08-31 2007-11-28 재단법인서울대학교산학협력재단 A Process for screening of a binding peptides specific for specific RNA and RNA binding peptides therefrom
EP1762250A1 (en) * 2005-09-12 2007-03-14 Fresenius Kabi Deutschland GmbH Conjugates of hydroxyalkyl starch and an active substance, prepared by chemical ligation via thiazolidine
AU2006297173A1 (en) * 2005-09-29 2007-04-12 Viral Logic Systems Technology Corp. Immunomodulatory compositions and uses therefor
CN101277974A (en) 2005-09-30 2008-10-01 阿伯特有限及两合公司 Binding domains of proteins of the repulsive guidance molecule (RGM) protein family and functional fragments thereof, and their use
DK1931709T3 (en) 2005-10-03 2017-03-13 Xencor Inc FC VARIETIES WITH OPTIMIZED FC RECEPTOR BINDING PROPERTIES
JP4860703B2 (en) 2005-10-06 2012-01-25 ゼンコー・インコーポレイテッド Optimized anti-CD30 antibody
US8445642B1 (en) * 2005-10-13 2013-05-21 The United States Of America As Represented By The Secretary Of Agriculture Methods to differentiate protein conformers
NZ597082A (en) 2005-10-13 2013-11-29 Human Genome Sciences Inc Methods and Compositions for Use in Treatment of Patients with Autoantibody Positive Diseases
US8168592B2 (en) 2005-10-21 2012-05-01 Amgen Inc. CGRP peptide antagonists and conjugates
BRPI0617515A2 (en) * 2005-10-24 2011-07-26 Centocor Inc mimetibodies, polypeptides, compositions, methods and uses of glp-2
PL1978993T3 (en) * 2005-10-31 2017-07-31 Oncomed Pharmaceuticals, Inc. Compositions and methods for treating cancer based on human fzd receptors
US7723477B2 (en) 2005-10-31 2010-05-25 Oncomed Pharmaceuticals, Inc. Compositions and methods for inhibiting Wnt-dependent solid tumor cell growth
US8067562B2 (en) 2005-11-01 2011-11-29 Amgen Inc. Isolated nucleic acid molecule comprising the amino acid sequence of SEQ ID NO:1
SG10201706600VA (en) 2005-11-30 2017-09-28 Abbvie Inc Monoclonal antibodies and uses thereof
PT1954718E (en) 2005-11-30 2014-12-16 Abbvie Inc Anti-a globulomer antibodies, antigen-binding moieties thereof, corresponding hybridomas, nucleic acids, vectors, host cells, methods of producing said antibodies, compositions comprising said antibodies, uses of said antibodies and methods of using said antibodies
US20070149458A1 (en) * 2005-12-06 2007-06-28 Amgen Inc. Uses of myostatin antagonists
AU2006322028B2 (en) 2005-12-08 2013-06-06 Amgen Inc. Improved host cells and culture methods
TWI382990B (en) 2005-12-12 2013-01-21 Hoffmann La Roche Antibody glycosylation in the variable region
US20090054763A1 (en) * 2006-01-19 2009-02-26 The Regents Of The University Of Michigan System and method for spectroscopic photoacoustic tomography
US9012605B2 (en) * 2006-01-23 2015-04-21 Amgen Inc. Crystalline polypeptides
US7625564B2 (en) 2006-01-27 2009-12-01 Novagen Holding Corporation Recombinant human EPO-Fc fusion proteins with prolonged half-life and enhanced erythropoietic activity in vivo
AR059066A1 (en) 2006-01-27 2008-03-12 Amgen Inc COMBINATIONS OF THE ANGIOPOYETINE INHIBITOR -2 (ANG2) AND THE VASCULAR ENDOTELIAL GROWTH FACTOR INHIBITOR (VEGF)
TW200738752A (en) 2006-01-31 2007-10-16 Bayer Schering Pharma Ag Modulation of MDL-1 activity for treatment of inflammatory disease
EP1816201A1 (en) 2006-02-06 2007-08-08 CSL Behring GmbH Modified coagulation factor VIIa with extended half-life
TW200745163A (en) 2006-02-17 2007-12-16 Syntonix Pharmaceuticals Inc Peptides that block the binding of IgG to FcRn
PL2005185T3 (en) 2006-03-22 2011-05-31 Viral Logic Systems Tech Corp Methods for identifying polypeptide targets
US8129334B2 (en) 2006-03-31 2012-03-06 The Regents Of The University Of California Methods and compositions for treating neurodegenerative disorders and Alzheimer'S disease and improving normal memory
WO2007115148A2 (en) * 2006-03-31 2007-10-11 Centocor, Inc. Human mimetic epo hinge core mimetibodies
JP6084761B2 (en) 2006-04-05 2017-02-22 ザ ロックフェラー ユニバーシティー Polypeptides with enhanced anti-inflammatory properties and reduced cytotoxic properties and related methods
EP2007885B1 (en) * 2006-04-11 2010-07-21 CSL Behring GmbH Method of increasing the in vivo recovery of therapeutic polypeptides
US9283260B2 (en) * 2006-04-21 2016-03-15 Amgen Inc. Lyophilized therapeutic peptibody formulations
TWI395754B (en) 2006-04-24 2013-05-11 Amgen Inc Humanized c-kit antibody
JP2009537145A (en) * 2006-05-15 2009-10-29 バイラル ロジック システムズ テクノロジー コーポレーション Compositions and methods associated with CD47 for treating immunological diseases and disorders
US8377448B2 (en) * 2006-05-15 2013-02-19 The Board Of Trustees Of The Leland Standford Junior University CD47 related compositions and methods for treating immunological diseases and disorders
NZ612319A (en) * 2006-06-12 2015-04-24 Emergent Product Dev Seattle Single-chain multivalent binding proteins with effector function
US7981425B2 (en) 2006-06-19 2011-07-19 Amgen Inc. Thrombopoietic compounds
EP2037928B1 (en) 2006-06-26 2012-01-11 Amgen Inc. Methods for treating atherosclerosis
AU2007285976B2 (en) 2006-08-14 2011-08-18 Xencor, Inc Optimized antibodies that target CD19
CA2662549C (en) 2006-09-08 2014-10-28 Amgen Inc. Il-1 family variants
WO2008127271A2 (en) 2006-09-08 2008-10-23 Abbott Laboratories Interleukin -13 binding proteins
KR20090060334A (en) * 2006-09-08 2009-06-11 제넨테크, 인크. Wnt antagonists and their use in the diagnosis and treatment of wnt-mediated disorders
US20140147441A1 (en) * 2006-09-12 2014-05-29 The General Hospital Corporation Compositions containing alpha-1-antitrypsin and methods for use
AU2007347182A1 (en) * 2006-09-15 2008-08-21 The Burnham Institute High affinity EphB receptor binding compounds and methods of use thereof
WO2008036688A2 (en) 2006-09-18 2008-03-27 Xencor, Inc. Optimized antibodies that target hm1.24
CN101594878A (en) 2006-09-18 2009-12-02 雷普特药品公司 By giving receptor associated protein(RAP) (RAP)-conjugate to the liver treatment of conditions
US20100297218A1 (en) * 2006-09-20 2010-11-25 Pneumrx, Inc. Tissue adhesive compositions and methods thereof
US20100034194A1 (en) * 2006-10-11 2010-02-11 Siemens Communications Inc. Eliminating unreachable subscribers in voice-over-ip networks
WO2008051383A2 (en) * 2006-10-19 2008-05-02 Amgen Inc. Use of alcohol co-solvents to improve pegylation reaction yields
US7820623B2 (en) 2006-10-25 2010-10-26 Amgen Inc. Conjugated toxin peptide therapeutic agents
WO2008067438A2 (en) * 2006-11-29 2008-06-05 The Regents Of University Of Michigan System and method for photoacoustic guided diffuse optical imaging
US8455626B2 (en) 2006-11-30 2013-06-04 Abbott Laboratories Aβ conformer selective anti-aβ globulomer monoclonal antibodies
WO2008070117A1 (en) * 2006-12-04 2008-06-12 Promedior, Inc. Conjoint therapy for treating fibrotic diseases
US20100166734A1 (en) * 2006-12-20 2010-07-01 Edward Dolk Oral delivery of polypeptides
KR100888022B1 (en) 2006-12-21 2009-03-09 재단법인 목암생명공학연구소 Fusion Proteion of Imunoglobulin Fc and Human Apolipoproteina Kringle Fragment
WO2008077616A1 (en) 2006-12-22 2008-07-03 Csl Behring Gmbh Modified coagulation factors with prolonged in vivo half-life
CA2676820A1 (en) 2007-01-31 2008-08-07 Affymax, Inc. Nitrogen-based linkers for attaching modifying groups to polypeptides and other macromolecules
ES2566385T3 (en) * 2007-02-12 2016-04-12 Csl Behring Gmbh Therapeutic application of serine protease inhibitors of the Kazal type
US20100311767A1 (en) 2007-02-27 2010-12-09 Abbott Gmbh & Co. Kg Method for the treatment of amyloidoses
TWI573802B (en) 2007-03-06 2017-03-11 安美基公司 Variant activin receptor polypeptides and uses thereof
US8501678B2 (en) 2007-03-06 2013-08-06 Atara Biotherapeutics, Inc. Variant activin receptor polypeptides and uses thereof
JP2010526528A (en) 2007-05-03 2010-08-05 メディツィニーチェ ウニベルシタット インスブルック Complement factor H-derived short consensus repeat-antibody construct
US20090252729A1 (en) * 2007-05-14 2009-10-08 Farrington Graham K Single-chain Fc (scFc) regions, binding polypeptides comprising same, and methods related thereto
EP2162540A2 (en) 2007-05-22 2010-03-17 Amgen Inc. Compositions and methods for producing bioactive fusion proteins
US20090175847A1 (en) * 2007-05-30 2009-07-09 Abbott Laboratories Humanized antibodies to ab (20-42) globulomer and uses thereof
US20090232801A1 (en) * 2007-05-30 2009-09-17 Abbot Laboratories Humanized Antibodies Which Bind To AB (1-42) Globulomer And Uses Thereof
CA2689518A1 (en) * 2007-06-05 2008-12-11 Oriental Yeast Co., Ltd. A novel bone mass increasing agent
US20090054332A1 (en) * 2007-06-21 2009-02-26 Conjuchem Biotechnologies, Inc. Thombopoietin peptide conjugates
US8497243B2 (en) * 2007-07-06 2013-07-30 Promedior, Inc. Methods and compositions useful in the treatment of mucositis
US9884899B2 (en) * 2007-07-06 2018-02-06 Promedior, Inc. Methods for treating fibrosis using CRP antagonists
US8293685B2 (en) 2007-07-26 2012-10-23 The Regents Of The University Of California Methods for enhancing bacterial cell display of proteins and peptides
EP2489730B1 (en) 2007-07-26 2015-12-30 Amgen, Inc Modified lecithin-cholesterol acyltransferase enzymes
CA2720628A1 (en) 2007-07-26 2009-01-29 Novagen Holding Corporation Fusion proteins having mutated immunoglobulin hinge region
EA201070231A1 (en) * 2007-08-09 2010-10-29 Синтоникс Фармасьютикалз, Инк. IMMUNOMODULATING PEPTIDES
JP2010536786A (en) * 2007-08-17 2010-12-02 アムジエン・インコーポレーテツド Preparation of antibody and FC fusion molecule using polycation
EP2615113A3 (en) 2007-08-23 2013-11-13 Amgen Inc. Antigen binding proteins to proprotein convertase subtilisin kexin type 9 (PCSK9)
JOP20080381B1 (en) 2007-08-23 2023-03-28 Amgen Inc Antigen Binding Proteins to Proprotein Convertase subtillisin Kexin type 9 (pcsk9)
US20100291086A1 (en) * 2007-09-11 2010-11-18 Christopher Hovens Use of estrogen and androgen binding proteins in methods and compositions for treating gynaecological cancers
AU2008304111B2 (en) 2007-09-27 2014-04-24 Amgen Inc. Pharmaceutical formulations
RU2505311C2 (en) * 2007-10-02 2014-01-27 Потентия Фармасьютикалз, Инк. Prolonged delivery of compstatin analogues from gels
US7829735B2 (en) 2007-10-26 2010-11-09 Northwestern University Universal phosphoramidite for preparation of modified biomolecules and surfaces
EP3381445B1 (en) 2007-11-15 2023-10-25 Amgen Inc. Aqueous formulation of antibody stablised by antioxidants for parenteral administration
WO2009085200A2 (en) 2007-12-21 2009-07-09 Amgen Inc. Anti-amyloid antibodies and uses thereof
EP2808343B8 (en) 2007-12-26 2020-01-15 Xencor, Inc. Fc variants with altered binding to FcRn
US20140127200A1 (en) * 2008-01-03 2014-05-08 The Scripps Research Institute Multispecific Antibody Targeting and Multivalency Through Modular Recognition Domains
EP2574628B1 (en) 2008-01-25 2015-05-20 Amgen Inc. Ferroportin antibodies and methods of use
JO2913B1 (en) 2008-02-20 2015-09-15 امجين إنك, Antibodies directed to angiopoietin-1 and angiopoietin-2 and uses thereof
CN101518644B (en) * 2008-02-26 2011-07-13 上海交通大学医学院附属第九人民医院 Application of Ang-2 and genes thereof in pharmacy
AU2009219232B2 (en) * 2008-02-27 2014-02-27 Novo Nordisk A/S Conjugated Factor VIII molecules
US8962803B2 (en) 2008-02-29 2015-02-24 AbbVie Deutschland GmbH & Co. KG Antibodies against the RGM A protein and uses thereof
JP6013733B2 (en) 2008-04-11 2016-10-25 エマージェント プロダクト デベロップメント シアトル, エルエルシー CD37 immunotherapeutic and its combination with bifunctional chemotherapeutics
MX2010011955A (en) 2008-04-29 2011-01-21 Abbott Lab Dual variable domain immunoglobulins and uses thereof.
EP2294087B1 (en) 2008-05-01 2014-05-14 Amgen, Inc. Anti-hepcidin antibodies and methods of use
US8293714B2 (en) * 2008-05-05 2012-10-23 Covx Technology Ireland, Ltd. Anti-angiogenic compounds
CN104558178A (en) 2008-05-09 2015-04-29 Abbvie公司 Antibodies to receptor of advanced glycation end products (rage) and uses thereof
WO2009142460A2 (en) * 2008-05-23 2009-11-26 Samsung Electronics Co., Ltd. Antibody-peptide fused synergibody
US9035027B2 (en) 2008-06-03 2015-05-19 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
NZ589436A (en) 2008-06-03 2012-12-21 Abbott Lab Dual variable domain immunoglobulins and uses thereof
JOP20190083A1 (en) 2008-06-04 2017-06-16 Amgen Inc Fgf21 mutant fusion polypeptides and uses thereof
DK2865760T3 (en) 2008-06-24 2018-01-15 Csl Behring Gmbh Factor VIII, Von Willebrand Factor or complexes thereof with extended in vivo half-life
WO2009158432A2 (en) 2008-06-27 2009-12-30 Amgen Inc. Ang-2 inhibition to treat multiple sclerosis
NZ590074A (en) 2008-07-08 2012-12-21 Abbott Lab Prostaglandin e2 dual variable domain immunoglobulins and uses thereof
AR072596A1 (en) 2008-07-23 2010-09-08 Hanmi Pharmaceutical Co Ltd A PROTEIN COMPLEX THAT INCLUDES A DIMERICAL PROTEIN AND A NON-PEPTIDIC POLYMER
WO2010014909A1 (en) * 2008-08-01 2010-02-04 Syntonix Pharmaceuticals, Inc. Immunomodulatory peptides
EP2168590A1 (en) * 2008-09-24 2010-03-31 Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH) Antimicrobial peptides
MX2011003183A (en) 2008-09-26 2011-04-21 Oncomed Pharm Inc Frizzled-binding agents and uses thereof.
EA201791940A3 (en) 2008-11-26 2018-04-30 Амген Инк. OPTIONS OF POLYPEPTIDES OF ACTIVIN IIB RECEPTOR AND THEIR USE
WO2010081095A2 (en) * 2009-01-12 2010-07-15 The Regents Of The University Of California Methods and compositions for inhibiting hepatitis c virus replication
AU2010208637A1 (en) * 2009-01-29 2011-08-04 Abbvie Inc. IL-1 binding proteins
US20110165063A1 (en) * 2009-01-29 2011-07-07 Abbott Laboratories Il-1 binding proteins
WO2010096394A2 (en) 2009-02-17 2010-08-26 Redwood Biosciences, Inc. Aldehyde-tagged protein-based drug carriers and methods of use
SG173705A1 (en) 2009-03-05 2011-09-29 Abbott Lab Il-17 binding proteins
US8283162B2 (en) * 2009-03-10 2012-10-09 Abbott Laboratories Antibodies relating to PIVKAII and uses thereof
CA2754961C (en) * 2009-03-11 2018-04-10 Promedior, Inc. Treatment and diagnostic methods for hypersensitive disorders
PT2405929T (en) * 2009-03-11 2018-07-23 Promedior Inc A sap polypeptide for use in the treatment of autoimmune disorders and graft vs host disease
US8734796B2 (en) 2009-03-20 2014-05-27 Amgen Inc. Carrier immunoglobulins
EP2413915A2 (en) 2009-03-30 2012-02-08 F. Hoffmann-La Roche AG A method for avoiding glass fogging
BRPI1014089A2 (en) 2009-04-02 2016-04-19 Roche Glycart Ag multispecific antibodies comprising full length antibodies and single chain fab fragments
FI3248610T3 (en) 2009-05-05 2024-01-18 Amgen Inc Fgf21 mutants and uses thereof
MX2011011815A (en) 2009-05-05 2012-01-27 Amgen Inc Fgf21 mutants and uses thereof.
UA110323C2 (en) * 2009-06-04 2015-12-25 Promedior Inc Derivative of serum amyloid p and their receipt and application
EP2443137B1 (en) 2009-06-15 2015-05-20 Biokine Therapeutics Ltd. Novel chemokine binding polypeptides capable of inhibiting the course of autoimmunity, inflammation and cancer
HUE041034T2 (en) * 2009-06-17 2019-05-28 Promedior Inc Sap variants and their use
EP2443145A1 (en) 2009-06-17 2012-04-25 Amgen, Inc Chimeric fgf19 polypeptides and uses thereof
EA020621B1 (en) 2009-06-22 2014-12-30 Амген Инк. Refolding proteins using a chemically controlled redox state
JP2012531428A (en) 2009-06-25 2012-12-10 アムジエン・インコーポレーテツド Capture and purification process for proteins expressed in non-mammalian systems
IN2012DN00248A (en) 2009-07-02 2015-05-01 Angiochem Inc
IT1395137B1 (en) 2009-08-05 2012-09-05 Spider Biotech S R L NEW ANTIPATOGENIC PEPTIDES
CA2771575A1 (en) 2009-08-29 2011-03-03 Abbott Laboratories Therapeutic dll4 binding proteins
CA2772628A1 (en) 2009-09-01 2011-03-10 Abbott Laboratories Dual variable domain immunoglobulins and uses thereof
US9493578B2 (en) 2009-09-02 2016-11-15 Xencor, Inc. Compositions and methods for simultaneous bivalent and monovalent co-engagement of antigens
SG10201408401RA (en) 2009-09-16 2015-01-29 Genentech Inc Coiled coil and/or tether containing protein complexes and uses thereof
WO2011038139A1 (en) 2009-09-23 2011-03-31 Amgen Inc. Treatment of ovarian cancer using a specific binding agent of human angiopoietin-2 in combination with a taxane
US8926976B2 (en) 2009-09-25 2015-01-06 Xoma Technology Ltd. Modulators
CA2772945A1 (en) 2009-09-25 2011-03-31 Xoma Technology Ltd. Screening methods
AR078651A1 (en) 2009-10-15 2011-11-23 Abbott Lab IMMUNOGLOBULINS WITH DUAL VARIABLE DOMAIN AND USES OF THE SAME
UY32979A (en) 2009-10-28 2011-02-28 Abbott Lab IMMUNOGLOBULINS WITH DUAL VARIABLE DOMAIN AND USES OF THE SAME
TW201121568A (en) * 2009-10-31 2011-07-01 Abbott Lab Antibodies to receptor for advanced glycation end products (RAGE) and uses thereof
PT2496691T (en) 2009-11-02 2017-05-31 Univ Washington Therapeutic nuclease compositions and methods
EP2499165B1 (en) * 2009-11-13 2016-09-14 Grifols Therapeutics Inc. Von willebrand factor (vwf)-containing preparations, and methods, kits, and uses related thereto
EP2498803A4 (en) 2009-11-13 2013-05-15 Puget Sound Blood Ct Factor viii t cell epitope variants having reduced immunogenicity
EP2501407A1 (en) 2009-11-20 2012-09-26 Amgen Inc. Anti-orai1 antigen binding proteins and uses thereof
UA109888C2 (en) 2009-12-07 2015-10-26 ANTIBODY OR ANTIBODILITY ANTIBODY OR ITS BINDING TO THE β-CLOTE, FGF RECEPTORS AND THEIR COMPLEXES
CN102656190A (en) 2009-12-08 2012-09-05 雅培股份有限两合公司 Monoclonal antibodies against the RGM A protein for use in the treatment of retinal nerve fiber layer degeneration
EA201492253A1 (en) 2009-12-29 2015-06-30 Эмерджент Продакт Дивелопмент Сиэтл, Ллс CONSTRUCTORS CONNECTING WITH RON AND METHODS OF THEIR USE
TWI535445B (en) 2010-01-12 2016-06-01 安可美德藥物股份有限公司 Wnt antagonists and methods of treatment and screening
US8362210B2 (en) 2010-01-19 2013-01-29 Xencor, Inc. Antibody variants with enhanced complement activity
CA2787376A1 (en) * 2010-01-19 2011-07-28 President And Fellows Of Harvard College Engineered opsonin for pathogen detection and treatment
TWI558397B (en) 2010-01-28 2016-11-21 雷普特製藥有限公司 Method for treating liver tumors with receptor associated protein (rap) peptide-fucosidase inhibitor conjugates
US9493543B2 (en) 2010-02-16 2016-11-15 Novo Nordisk A/S Factor VIII fusion protein
PE20130580A1 (en) 2010-03-02 2013-06-02 Abbvie Inc THERAPEUTIC BINDING PROTEINS TO DLL4
US20130171128A1 (en) 2010-03-02 2013-07-04 Amgen Inc. Reducing viscosity of pharmaceutical formulations
NZ602548A (en) 2010-03-03 2014-12-24 Univ British Columbia Oligomer-specific amyloid beta epitope and antibodies
TW201138821A (en) 2010-03-26 2011-11-16 Roche Glycart Ag Bispecific antibodies
CA2794745A1 (en) 2010-03-29 2011-10-06 Zymeworks, Inc. Antibodies with enhanced or suppressed effector function
EP2371857A1 (en) 2010-04-01 2011-10-05 CSL Behring GmbH Factor XII inhibitors for treating interstitial lung disease
NZ602700A (en) 2010-04-01 2014-10-31 Oncomed Pharm Inc Frizzled-binding agents and uses thereof
AR081755A1 (en) * 2010-04-02 2012-10-17 Hanmi Holdings Co Ltd FORMULATION OF PROLONGED ACTION OF THE FOLICULES STIMULATING HORMONE WHERE AN IMMUNOGLOBULIN FRAGMENT, PREPARATION METHOD AND METHOD TO TREAT A SUBJECT SUFFERING A REPRODUCTIVE DISORDER ARE USED
ES2537814T3 (en) * 2010-04-09 2015-06-12 Amgen Inc. Proteins, nucleic acids, and BTNL9 antibodies and uses thereof
MX336196B (en) 2010-04-15 2016-01-11 Abbvie Inc Amyloid-beta binding proteins.
JP2013523184A (en) 2010-04-15 2013-06-17 アムジエン・インコーポレーテツド Human FGF receptor and β-KLOTHO binding protein
MX346731B (en) 2010-04-23 2017-03-30 Genentech Inc * Production of heteromultimeric proteins.
MX2012013144A (en) 2010-05-14 2012-11-30 Amgen Inc Enhanced death receptor agonists.
AU2011252883B2 (en) 2010-05-14 2015-09-10 Abbvie Inc. IL-1 binding proteins
CN102260343A (en) 2010-05-25 2011-11-30 健能隆医药技术(上海)有限公司 Application of recombinant human G-CSF dimer for treating nerve damage disease
US20110305670A1 (en) * 2010-06-10 2011-12-15 President And Fellows Of Harvard College Nucleic acid encoding fusion polypeptides that prevent or inhibit hiv infection
WO2012012141A1 (en) 2010-06-30 2012-01-26 Amgen Inc. Scnn1a/tnfrsf1a fusion proteins in cancer
US20120100166A1 (en) 2010-07-15 2012-04-26 Zyngenia, Inc. Ang-2 Binding Complexes and Uses Thereof
US9120862B2 (en) 2010-07-26 2015-09-01 Abbott Laboratories Antibodies relating to PIVKA-II and uses thereof
AU2011285852B2 (en) 2010-08-03 2014-12-11 Abbvie Inc. Dual variable domain immunoglobulins and uses thereof
CN103154032A (en) 2010-08-13 2013-06-12 弗·哈夫曼-拉罗切有限公司 Antibodies to IL-1beta and IL-18, for treatment of disease
WO2012024187A1 (en) 2010-08-14 2012-02-23 Abbott Laboratories Amyloid-beta binding proteins
WO2012025530A1 (en) 2010-08-24 2012-03-01 F. Hoffmann-La Roche Ag Bispecific antibodies comprising a disulfide stabilized - fv fragment
KR20130139884A (en) 2010-08-26 2013-12-23 애브비 인코포레이티드 Dual variable domain immunoglobulins and uses thereof
MY166172A (en) 2010-09-10 2018-06-07 Wyeth Llc Non-lipidated variants of neisseria meningitidis orf2086 antigens
WO2012040518A2 (en) 2010-09-22 2012-03-29 Amgen Inc. Carrier immunoglobulins and uses thereof
BR112013007385B1 (en) 2010-09-28 2022-07-26 Amylin Pharmaceuticals, Llc CHIMERIC POLYPEPTIDE, ITS USE AND COMPOSITION THAT COMPRISES IT
US9023791B2 (en) 2010-11-19 2015-05-05 Novartis Ag Fibroblast growth factor 21 mutations
TW201307388A (en) 2010-12-21 2013-02-16 Abbott Lab IL-1 binding proteins
RU2627171C2 (en) 2010-12-21 2017-08-03 Эббви Инк. Il-1 alpha and beta bispecific immunoglobulins with double variable domains and their application
WO2012085111A1 (en) 2010-12-23 2012-06-28 F. Hoffmann-La Roche Ag Polypeptide-polynucleotide-complex and its use in targeted effector moiety delivery
CN103415621A (en) 2011-01-14 2013-11-27 雷德伍德生物科技股份有限公司 Aldehyde-tagged immunoglobulin polypeptides and method of use thereof
SG192047A1 (en) 2011-01-24 2013-08-30 Univ Singapore Pathogenic mycobacteria-derived mannose-capped lipoarabinomannan antigen binding proteins
AR085138A1 (en) 2011-02-04 2013-09-11 Genentech Inc Fc VARIATIONS AND METHODS FOR PRODUCTION
US10689447B2 (en) 2011-02-04 2020-06-23 Genentech, Inc. Fc variants and methods for their production
WO2012109624A2 (en) 2011-02-11 2012-08-16 Zyngenia, Inc. Monovalent and multivalent multispecific complexes and uses thereof
CA2828405A1 (en) 2011-02-28 2012-09-07 Istituto Di Ricovero E Cura A Carattere Scientifico Materno-Infantile Bu Rlo Garofolo - Ospedale Di Alta Specializzazione E Di Rilievo Nazionale Apoptosis-inducing molecules and uses therefor
EP2497489A1 (en) 2011-03-09 2012-09-12 CSL Behring GmbH Factor XII inhibitors for the treatment of silent brain ischemia and ischemia of other organs
WO2012120128A1 (en) 2011-03-09 2012-09-13 Csl Behring Gmbh Factor xii inhibitors for the administration with medical procedures comprising contact with artificial surfaces
JP2014509859A (en) 2011-03-16 2014-04-24 アムジエン・インコーポレーテツド Potent and selective inhibitors of 17NAV1.3 and NAV1.7
WO2012138997A1 (en) 2011-04-07 2012-10-11 Amgen Inc. Novel egfr binding proteins
JP5991966B2 (en) * 2011-04-25 2016-09-14 大鵬薬品工業株式会社 Nanoparticles containing pH-responsive peptides
LT2704737T (en) 2011-04-29 2018-06-11 University Of Washington Therapeutic nuclease compositions and methods
JOP20200043A1 (en) 2011-05-10 2017-06-16 Amgen Inc Methods of treating or preventing cholesterol related disorders
EP2714738B1 (en) 2011-05-24 2018-10-10 Zyngenia, Inc. Multivalent and monovalent multispecific complexes and their uses
EP3878859B1 (en) 2011-06-10 2023-11-01 Hanmi Science Co., Ltd. Novel oxyntomodulin derivatives and pharmaceutical composition for treating obesity comprising the same
BR112013032375B1 (en) * 2011-06-17 2022-09-20 Hanmi Science Co., Ltd CONJUGATE, PHARMACEUTICAL COMPOSITION AND USE THEREOF
MX358726B (en) 2011-06-29 2018-09-03 Amgen Inc Predictive biomarker of survival in the treatment of renal cell carcinoma.
WO2013012733A1 (en) 2011-07-15 2013-01-24 Biogen Idec Ma Inc. Heterodimeric fc regions, binding molecules comprising same, and methods relating thereto
US9353172B2 (en) 2011-07-18 2016-05-31 Arts Biologics A/S Long acting biologically active luteinizing hormone (LH) compound
WO2013012855A1 (en) 2011-07-18 2013-01-24 Amgen Inc. Apelin antigen-binding proteins and uses thereof
EP2734843A2 (en) 2011-07-18 2014-05-28 President and Fellows of Harvard College Engineered microbe-targeting molecules and uses thereof
WO2013014092A1 (en) 2011-07-22 2013-01-31 Csl Behring Gmbh Inhibitory anti -factor xii/xiia monoclonal antibodies and their uses
CA2842969C (en) 2011-07-25 2018-03-27 Generon (Shanghai) Corporation Ltd. Use of g-csf dimer in preparation of medicament for treatment of neurodegenerative diseases
US9605083B2 (en) 2011-08-16 2017-03-28 Emory University JAML specific binding agents, antibodies, and uses related thereto
DE102012016127A1 (en) 2011-08-31 2013-02-28 Daniel Elias Error correction encoding apparatus for data stream, supplies one of m' mutually different synchronous words to each block in error correction information
EP2751135A4 (en) * 2011-08-31 2015-06-03 Indi Molecular Inc Vegf-specific capture agents, compositions and methods of using and making
BR112014004591A2 (en) 2011-09-02 2017-03-28 Amgen Inc light exposure analysis pharmaceutical product of a pharmaceutical product
UY34346A (en) 2011-09-26 2013-04-30 Novartis Ag FUSION PROTEINS TO TREAT METABOLIC DISORDERS
WO2013055958A1 (en) 2011-10-11 2013-04-18 Genentech, Inc. Improved assembly of bispecific antibodies
RS20140202A1 (en) 2011-10-24 2014-10-31 Abbvie Inc. Biospecific immunibinders directed against tnf and il-17
CN104093739A (en) 2011-10-24 2014-10-08 艾伯维公司 Immunobinders directed against TNF
CA2853371A1 (en) 2011-10-26 2013-05-02 Amgen Inc. Methods of reducing or eliminating protein modification and degradation arising from exposure to uv light
CN102516393B (en) * 2011-11-30 2017-03-15 北京康明百奥新药研发有限公司 Insulin-simulated peptide fusion protein and mutant and its application
CN104144947B (en) 2011-12-14 2017-10-27 艾伯维德国有限责任两合公司 Composition and method for diagnosing and treating iron phase related disorders
CA2859504A1 (en) 2011-12-19 2013-07-18 Amgen Inc. Variant activin receptor polypeptides, alone or in combination with chemotherapy, and uses thereof
PL2794626T6 (en) 2011-12-22 2020-11-02 Glycomimetics, Inc. E-selectin antagonist compounds
CN102558358A (en) * 2011-12-30 2012-07-11 张海涛 Preparation and application of human fibroblast growth factor 21 fusion protein and mutant of human fibroblast growth factor 21 fusion protein
TW201333035A (en) 2011-12-30 2013-08-16 Abbvie Inc Dual specific binding proteins directed against IL-13 and/or IL-17
EP3338617B1 (en) 2012-01-23 2020-08-19 Washington University Goggle imaging systems and devices
SG10202006762PA (en) 2012-01-27 2020-08-28 Abbvie Deutschland Composition and method for diagnosis and treatment of diseases associated with neurite degeneration
EP2623110A1 (en) 2012-01-31 2013-08-07 CSL Behring GmbH Factor XII inhibitors for the treatment of neurological inflammatory disorders
CA2861124A1 (en) 2012-02-10 2013-08-15 Genentech, Inc. Single-chain antibodies and other heteromultimers
DK2814502T3 (en) 2012-02-15 2017-12-18 Csl Behring Gmbh Von Willebrand Factor variants with improved Factor VIII binding affinity
CA2864702A1 (en) 2012-02-22 2013-08-29 Amgen Inc. Autologous mammalian models derived from induced pluripotent stem cells and related methods
SA115360586B1 (en) 2012-03-09 2017-04-12 فايزر انك Neisseria meningitidis compositions and methods thereof
BR112014021658A2 (en) 2012-03-09 2017-07-11 Pfizer neisseria meningitidis compositions and methods thereof
EP2831116A1 (en) 2012-03-28 2015-02-04 Amgen Inc. Dr5 receptor agonist combinations
WO2013162050A1 (en) * 2012-04-23 2013-10-31 株式会社Nrlファーマ Lactoferrin fusion protein and method for producing same
EA039663B1 (en) 2012-05-03 2022-02-24 Амген Инк. Use of an anti-pcsk9 antibody for lowering serum cholesterol ldl and treating cholesterol related disorders
WO2013167750A2 (en) 2012-05-11 2013-11-14 Prorec Bio Ab Method for diagnosis and treatment of prolactin associated disorders
DK2849567T3 (en) 2012-05-17 2022-06-20 Extend Biosciences Inc CARRIERS FOR IMPROVED MEDICINE ADMINISTRATION
JP2015525230A (en) 2012-06-11 2015-09-03 アムジエン・インコーポレーテツド Dual receptor antagonistic antigen binding protein and uses thereof
RU2015100656A (en) 2012-06-27 2016-08-20 Ф. Хоффманн-Ля Рош Аг METHOD FOR PRODUCING ANTIBODY FC-FRAGMENT CONNECTING, INCLUDING AT LEAST ONE CONNECTING GROUP, WHICH SPECIALLY RELATED TO THE TARGET, AND THEIR APPLICATION
CN104395339A (en) 2012-06-27 2015-03-04 弗·哈夫曼-拉罗切有限公司 Method for selection and production of tailor-made highly selective and multi-specific targeting entities containing at least two different binding entities and uses thereof
AR091755A1 (en) 2012-07-12 2015-02-25 Abbvie Inc PROTEINS OF UNION TO IL-1
CA2878400A1 (en) 2012-07-19 2014-01-23 Amgen Inc. Btnl3 proteins, nucleic acids, and antibodies and uses thereof
KR101968344B1 (en) 2012-07-25 2019-04-12 한미약품 주식회사 A composition for treating hyperlipidemia comprising oxyntomodulin analog
RU2015118063A (en) * 2012-10-17 2016-12-10 Ливерпуль Скул Оф Тропикал Медисин IMMUNOMODULATING PROTEINS
CA2887711A1 (en) 2012-10-23 2014-05-01 Oncomed Pharmaceuticals, Inc. Methods of treating neuroendocrine tumors using wnt pathway-binding agents
KR20180008921A (en) 2012-11-01 2018-01-24 애브비 인코포레이티드 Anti-vegf/dll4 dual variable domain immunoglobulins and uses thereof
MY168536A (en) 2012-11-06 2018-11-12 Hanmi Pharm Ind Co Ltd Liquid formulation of protein conjugate comprising an oxyntomodulin derivative covalently linked to a non-peptidyl polymer to an immunoglobulin fc region
KR101993393B1 (en) 2012-11-06 2019-10-01 한미약품 주식회사 A composition for treating diabetes or diabesity comprising oxyntomodulin analog
HUE038423T2 (en) 2012-12-07 2018-10-29 Glycomimetics Inc Compounds, compositions and methods using e-selectin antagonists for mobilization of hematopoietic cells
ES2875957T3 (en) * 2012-12-20 2021-11-11 Amgen Inc APJ receptor agonists and their uses
US10632179B2 (en) * 2013-01-11 2020-04-28 Case Western Reserve University Methods and compositions for treating cancer
KR102073748B1 (en) 2013-01-31 2020-02-05 한미약품 주식회사 Recombinant yeast transformant and process for preparing immunoglobulin Fc fragment employing the same
EP2951205B1 (en) 2013-02-01 2021-08-11 Santa Maria Biotherapeutics, Inc. Anti-activin-a compounds for the treatment of ovarian cancer
AU2014212081A1 (en) 2013-02-04 2015-08-13 Oncomed Pharmaceuticals, Inc. Methods and monitoring of treatment with a Wnt pathway inhibitor
SG11201506095TA (en) * 2013-02-26 2015-09-29 Hanmi Pharm Ind Co Ltd Novel insulin analog and use thereof
MX2015011257A (en) * 2013-03-08 2015-11-16 Taiho Pharmaceutical Co Ltd Novel peptide having 5 linked ctl epitopes.
JP6636334B2 (en) 2013-03-08 2020-01-29 ツェー・エス・エル・ベーリング・ゲー・エム・ベー・ハー Treatment and prevention of remote ischemia-reperfusion injury
EP2970408B1 (en) 2013-03-12 2018-01-10 Amgen Inc. Potent and selective inhibitors of nav1.7
JP6561042B2 (en) 2013-03-13 2019-08-14 ジェンザイム・コーポレーション Fusion proteins comprising PDGF and VEGF binding moieties and methods of use thereof
JOP20140087B1 (en) 2013-03-13 2021-08-17 Amgen Inc Proteins specific for baff and b7rp1 and uses thereof
US9458246B2 (en) 2013-03-13 2016-10-04 Amgen Inc. Proteins specific for BAFF and B7RP1
US9168300B2 (en) 2013-03-14 2015-10-27 Oncomed Pharmaceuticals, Inc. MET-binding agents and uses thereof
JP6387392B2 (en) 2013-03-15 2018-09-05 バイオベラティブ セラピューティクス インコーポレイテッド Factor IX polypeptide preparation
WO2014144325A1 (en) 2013-03-15 2014-09-18 President And Fellows Of Harvard College Methods and compositions for improving detection and/or capture of a target entity
CA2904448A1 (en) 2013-03-15 2014-09-18 Tariq Ghayur Dual specific binding proteins directed against il-1.beta. and/or il-17
EP2796145B1 (en) 2013-04-22 2017-11-01 CSL Ltd. A covalent complex of von willebrand factor and faktor viii linked by a disulphide bridge
WO2014190040A1 (en) * 2013-05-21 2014-11-27 President And Fellows Of Harvard College Engineered heme-binding compositions and uses thereof
EP3003372B1 (en) 2013-06-07 2019-10-09 Duke University Inhibitors of complement factor h
KR20160026905A (en) 2013-06-28 2016-03-09 체에스엘 베링 게엠베하 Combination therapy using a factor xii inhibitor and a c1-inhibitor
EP3013422A1 (en) 2013-06-28 2016-05-04 Amgen Inc. Methods for treating homozygous familial hypercholesterolemia
WO2015013168A1 (en) 2013-07-25 2015-01-29 Novartis Ag Cyclic polypeptides for the treatment of heart failure
AU2014293387B2 (en) 2013-07-25 2017-04-20 Novartis Ag Bioconjugates of synthetic apelin polypeptides
KR101905278B1 (en) 2013-09-08 2018-10-08 화이자 인코포레이티드 Neisseria meningitidis compositions and methods thereof
JP6989260B2 (en) * 2013-09-18 2022-01-05 ビーシーエヌ ペプタイズ エセ.ア.Bcn Peptides S.A. Cortistatin analogs for the treatment of inflammatory and / or immune disorders
WO2015057820A2 (en) * 2013-10-15 2015-04-23 Roberts S Kenny Peptide constructs and well-defined aggregates thereof
WO2015057908A1 (en) 2013-10-18 2015-04-23 Novartis Ag Methods of treating diabetes and related disorders
DK3061771T3 (en) 2013-10-21 2020-07-13 Taiho Pharmaceutical Co Ltd New Fire CTL epitope-linked peptide
WO2015066557A1 (en) 2013-10-31 2015-05-07 Resolve Therapeutics, Llc Therapeutic nuclease molecules with altered glycosylation and methods
WO2015095604A2 (en) 2013-12-18 2015-06-25 President And Fellows Of Harvard College Methods and assays relating to circulating tumor cells
US10344073B2 (en) 2014-01-09 2019-07-09 Hadasit Medical Research Services And Development Ltd. Cell compositions and methods for cancer therapy
US20150291689A1 (en) 2014-03-09 2015-10-15 Abbvie, Inc. Compositions and Methods for Treating Rheumatoid Arthritis
DK3129406T3 (en) * 2014-04-11 2019-04-01 Medimmune Llc CONJUGATED COMPOUNDS INCLUDING CYSTEIN MODIFIED ANTIBODIES
EP3140392B1 (en) 2014-05-06 2023-07-26 F. Hoffmann-La Roche AG Production of heteromultimeric proteins using mammalian cells
WO2015191760A2 (en) 2014-06-10 2015-12-17 Abbvie, Inc. Compositions and methods for treating rheumatoid arthritis
CA2951391C (en) 2014-06-10 2021-11-02 Amgen Inc. Apelin polypeptides
PL3157548T3 (en) 2014-06-18 2022-01-17 Csl Behring Gmbh Therapy using a factor xii inhibitor in a neurotraumatic disorder
BR112016030950A2 (en) 2014-07-02 2018-03-27 Csl Ltd modified factor viii binding polypeptide, complex, pharmaceutical composition, methods for treating a coagulopathy, to produce a polypeptide comprising a modified vwf, and to increase vwf factor viii binding affinity and factor viii half-life, use of a modified polypeptide or complex, polynucleotide, plasmid or vector, and host cell.
EP3172325B1 (en) 2014-07-22 2023-06-28 The University of Notre Dame du Lac Molecular constructs and uses thereof
US10934362B2 (en) 2014-09-15 2021-03-02 Amgen Inc. Bi-specific anti-CGRP receptor/PAC1 receptor antigen binding proteins and uses thereof
TWI772252B (en) 2014-09-16 2022-08-01 南韓商韓美藥品股份有限公司 Use of a long acting glp-1/glucagon receptor dual agonist for the treatment of non-alcoholic fatty liver disease
ES2962385T3 (en) 2014-10-15 2024-03-18 Amgen Inc Promoter and regulatory elements to improve the expression of heterologous genes in host cells
EP3220961B1 (en) 2014-10-22 2023-07-05 Extend Biosciences, Inc. Therapeutic vitamin d conjugates
US9789197B2 (en) 2014-10-22 2017-10-17 Extend Biosciences, Inc. RNAi vitamin D conjugates
WO2016065052A1 (en) 2014-10-22 2016-04-28 Extend Biosciences, Inc. Insulin vitamin d conjugates
JP2017531682A (en) 2014-10-23 2017-10-26 アムジエン・インコーポレーテツド Reduced viscosity of pharmaceutical preparations
WO2016069889A1 (en) 2014-10-31 2016-05-06 Resolve Therapeutics, Llc Therapeutic nuclease-transferrin fusions and methods
CN107001482B (en) 2014-12-03 2021-06-15 豪夫迈·罗氏有限公司 Multispecific antibodies
US10093733B2 (en) 2014-12-11 2018-10-09 Abbvie Inc. LRP-8 binding dual variable domain immunoglobulin proteins
WO2016102434A1 (en) * 2014-12-22 2016-06-30 Morphosys Ag Novel methods for displaying cyclic peptides on bacteriophage particles
KR102418477B1 (en) 2014-12-30 2022-07-08 한미약품 주식회사 Gluagon Derivatives
US20160244520A1 (en) 2015-01-24 2016-08-25 Abbvie Inc. Compositions and methods for treating psoriatic arthritis
EP3250240A4 (en) * 2015-01-30 2018-10-10 University of Utah Research Foundation Dimeric collagen hybridizing peptides and methods of using
BR112017017460A2 (en) 2015-02-19 2018-04-10 Pfizer Inc. neisseria meningitidis compositions and methods thereof
US10711067B2 (en) 2015-03-03 2020-07-14 Xoma (Us) Llc Treatment of post-prandial hyperinsulinemia and hypoglycemia after bariatric surgery
US10472408B2 (en) * 2015-03-05 2019-11-12 Peter Und Traudl Engelhorn-Stiftung Zur Förderung Der Lebenswissenschaften Fusion proteins comprising partial tetraspanin sequences and a system thereof for presenting peptides on the cell surface
WO2016164799A1 (en) 2015-04-10 2016-10-13 The Regents Of The University Of California Methods of determining patient populations amenable to immunomodulatory treatment of cancer
WO2016176427A1 (en) 2015-04-30 2016-11-03 Amgen Inc. Treatment of ovarian cancer in patients with ascites using a specific binding agent of human angiopoietin-2 in combination with a taxane
US10806804B2 (en) 2015-05-06 2020-10-20 Washington University Compounds having RD targeting motifs and methods of use thereof
EP4089109A3 (en) 2015-05-22 2023-03-01 CSL Behring Lengnau AG Methods for preparing modified von willebrand factor
MX2017014503A (en) 2015-05-22 2018-08-01 CSL Behring Lengnau AG Truncated von willebrand factor polypeptides for treating hemophilia.
TW201710286A (en) 2015-06-15 2017-03-16 艾伯維有限公司 Binding proteins against VEGF, PDGF, and/or their receptors
US10980892B2 (en) 2015-06-15 2021-04-20 Angiochem Inc. Methods for the treatment of leptomeningeal carcinomatosis
US20190004048A1 (en) 2015-06-26 2019-01-03 Amgen Inc. Biomarker of Survival in the Treatment of Renal Cell Carcinoma with a VEGFR Inhibitor and an Ang2 Inhibitor
WO2017024114A1 (en) 2015-08-06 2017-02-09 President And Fellows Of Harvard College Improved microbe-binding molecules and uses thereof
US20180237542A1 (en) 2015-09-15 2018-08-23 Amgen Inc. Tetravalent bispecific and tetraspecific antigen binding proteins and uses thereof
JP7002446B2 (en) 2015-09-21 2022-03-04 アプティーボ リサーチ アンド デベロップメント エルエルシー CD3 binding polypeptide
CA3000697A1 (en) 2015-10-01 2017-04-06 Amgen Inc. Treatment of bile acid disorders
KR102431342B1 (en) 2015-10-02 2022-08-10 에프. 호프만-라 로슈 아게 Bispecific antibodies specific for pd1 and tim3
US10975131B2 (en) 2015-10-27 2021-04-13 University Of Massachusetts Factor H-Fc immunotheraphy
KR101826792B1 (en) * 2015-10-29 2018-02-09 주식회사 와이바이오로직스 Composition for preventing or treating fatty liver or insulin resistance syndrome including extracellular domain of delta-like 1 homolog
ES2901772T3 (en) * 2015-12-15 2022-03-23 Sarepta Therapeutics Inc Peptide oligonucleotide conjugates
MA43567A (en) 2015-12-15 2018-11-14 Amgen Inc PACAP ANTIBODIES AND THEIR USES
ES2818531T3 (en) 2015-12-17 2021-04-13 Biokine Therapeutics Ltd Small molecules to inhibit chemokine activity and / or cancer cell growth
WO2017103931A1 (en) 2015-12-17 2017-06-22 Biokine Therapeutics Ltd. Small molecules against cancer
EP3184149A1 (en) 2015-12-23 2017-06-28 Julius-Maximilians-Universität Würzburg Soluble glycoprotein v for treating thrombotic diseases
CA3010720A1 (en) 2016-01-07 2017-07-13 Csl Behring Recombinant Facility Ag Mutated truncated von willebrand factor
US10808023B2 (en) 2016-01-07 2020-10-20 CSL Behring Lengnau AG Mutated von Willebrand factor
CA3019851A1 (en) 2016-04-06 2017-10-12 Csl Limited Method of treating atherosclerosis
KR101975743B1 (en) * 2016-04-07 2019-05-09 한양대학교 에리카산학협력단 Vascular Endothelial Growth Factor Receptor Targeting Peptide-elastin Fusion Polypeptides and Their Self-assembled Nanostructures
WO2017179647A1 (en) * 2016-04-14 2017-10-19 Taoヘルスライフファーマ株式会社 Peptide for inhibiting binding of amylospheroids (aspd), and evaluation and screening method
US11208632B2 (en) 2016-04-26 2021-12-28 R.P. Scherer Technologies, Llc Antibody conjugates and methods of making and using the same
TW202304996A (en) 2016-06-08 2023-02-01 美商艾伯維有限公司 Anti-b7-h3 antibodies and antibody drug conjugates
EP3478830B1 (en) 2016-07-01 2024-04-10 Resolve Therapeutics, LLC Optimized binuclease fusions and methods
MX2019000903A (en) 2016-07-22 2019-05-15 Amgen Inc Methods of purifying fc-containing proteins.
EP3502143A4 (en) 2016-08-19 2020-07-15 Ampsource Biopharma Shanghai Inc. Linker peptide for constructing fusion protein
CN106279437B (en) 2016-08-19 2017-10-31 安源医药科技(上海)有限公司 Hyperglycosylated human coagulation factor VIII fusion proteins and preparation method thereof and purposes
CN107759696A (en) 2016-08-19 2018-03-06 安源医药科技(上海)有限公司 Fusion protein of human interleukin 7 and preparation method thereof
US11459401B2 (en) 2016-10-06 2022-10-04 Amgen Inc. Reduced viscosity protein pharmaceutical formulations
JP7142915B2 (en) 2016-10-28 2022-09-28 株式会社S&Kバイオファーマ Lactoferrin/albumin fusion protein and method for producing same
SG11201903950UA (en) 2016-11-11 2019-05-30 CSL Behring Lengnau AG Truncated von willebrand factor polypeptides for treating hemophilia
ES2908008T3 (en) 2016-11-11 2022-04-27 CSL Behring Lengnau AG Truncated von Willebrand Factor polypeptides for extravascular administration in the treatment or prophylaxis of a blood coagulation disorder
JP7274417B2 (en) 2016-11-23 2023-05-16 イミュノア・セラピューティクス・インコーポレイテッド 4-1BB binding protein and uses thereof
WO2018102777A1 (en) * 2016-12-01 2018-06-07 University Of South Florida Peptibodies, compositions thereof, and methods of treating atrial fibrillation
US20190382462A1 (en) 2017-01-13 2019-12-19 Pietro P. Sanna Methods and compositions for treating hpa hyperactivity
IL303108B1 (en) 2017-01-31 2024-03-01 Pfizer Neisseria meningitidis compositions and methods thereof
TWI798209B (en) 2017-03-23 2023-04-11 南韓商韓美藥品股份有限公司 A conjugate of insulin analog with reduced affinity to insulin receptor and use thereof
BR112019017329A2 (en) 2017-04-03 2020-04-14 Hoffmann La Roche immunoconjugates, one or more polynucleotides and vectors, methods for the production of an immunoconjugate, treatment of a disease and for the stimulation of the immune system, composition, use of the immunoconjugate, invention and uses of the composition
KR102408873B1 (en) 2017-04-05 2022-06-15 에프. 호프만-라 로슈 아게 Bispecific antibodies specifically binding to pd1 and lag3
US10865238B1 (en) 2017-05-05 2020-12-15 Duke University Complement factor H antibodies
EP3641800B1 (en) 2017-06-22 2023-10-04 CSL Behring Lengnau AG Modulation of fviii immunogenicity by truncated vwf
US11725060B2 (en) 2017-07-20 2023-08-15 Aptevo Reserch and Development LLC Antigen binding proteins binding to 5T4 and 4-1BB and related compositions and methods
KR20200059213A (en) 2017-07-26 2020-05-28 잔센파마슈티카엔.브이. Methods to protect vascular integrity induced by targeted radiation therapy
WO2019028012A2 (en) 2017-07-31 2019-02-07 Dana-Farber Cancer Institute, Inc. Methods of using pembrolizumab and trebananib
MX2020001327A (en) 2017-08-04 2020-03-20 Amgen Inc Method of conjugation of cys-mabs.
JP2020530453A (en) 2017-08-09 2020-10-22 マサチューセッツ インスティテュート オブ テクノロジー Albumin-binding peptide bond and its method
EP3684811A2 (en) 2017-08-17 2020-07-29 Massachusetts Institute of Technology Multiple specificity binders of cxc chemokines and uses thereof
ES2887046T3 (en) 2017-08-17 2021-12-21 Just Evotec Biologics Inc Method of purification of glycosylated protein from host cell galectins and other contaminants
MX2020002070A (en) 2017-08-22 2020-03-24 Sanabio Llc Soluble interferon receptors and uses thereof.
EP3727447A1 (en) 2017-12-19 2020-10-28 Massachusetts Institute of Technology Antigen-adjuvant coupling reagents and methods of use
US20210046189A1 (en) * 2018-03-30 2021-02-18 HANMl PHARM. CO., LTD. Long-acting protein conjugates for brain targeting, a preparation method thereof, and a composition comprising the same
US20220128474A1 (en) 2018-10-23 2022-04-28 Amgen Inc. Automatic calibration and automatic maintenance of raman spectroscopic models for real-time predictions
EP3873488A4 (en) 2018-10-31 2022-08-03 The University of Sydney Compositions and methods for treating viral infections
AU2020204992A1 (en) 2019-01-04 2021-07-15 Resolve Therapeutics, Llc Treatment of sjogren's disease with nuclease fusion proteins
KR20220004618A (en) * 2019-01-07 2022-01-11 세나 바이오사이언스 아이엔씨. Novel peptides and uses thereof
MX2021008943A (en) 2019-01-25 2021-11-04 Janssen Pharmaceutica Nv Methods for mitigating liver injury and promoting liver hypertrophy, regeneration and cell engraftment in conjunction with radiation and/or radiomimetic treatments.
US20200237872A1 (en) 2019-01-25 2020-07-30 Janssen Pharmaceutica Nv Methods of enhancing protection against organ and vascular injury, hematopoietic recovery and survival in response to total body radiation/chemical exposure
EP3914286A2 (en) 2019-01-25 2021-12-01 Janssen Pharmaceutica NV Methods of mitigating toxic effects of vesicants and caustic gas
CN116063520A (en) 2019-01-30 2023-05-05 真和制药有限公司 anti-GAL 3 antibodies and uses thereof
KR20210127947A (en) 2019-02-15 2021-10-25 저스트-에보텍 바이오로직스, 아이엔씨. Automated biomanufacturing systems, equipment and processes
KR20220029545A (en) 2019-03-22 2022-03-08 리플렉시온 파마슈티컬스, 인크. D-peptidic compounds for VEGF
CN114144433A (en) 2019-03-22 2022-03-04 反射制药有限公司 Multivalent D-peptide compounds for proteins of interest
AU2020276793A1 (en) 2019-05-15 2022-01-20 Alonbio Ltd. Small molecules for treating cancer, inhibiting chemokine activity and/or inducing cell death
KR20220009984A (en) 2019-05-17 2022-01-25 유니베르시태트 취리히 Haptoglobin for use in the treatment of secondary neurological adverse events after hemorrhagic stroke
KR20220016139A (en) 2019-05-30 2022-02-08 암젠 인크 Hinge region engineering to induce antibody dimerization
JP2022538974A (en) 2019-06-26 2022-09-07 マサチューセッツ インスチテュート オブ テクノロジー Immunomodulatory fusion protein-metal hydroxide complexes and methods thereof
WO2020264384A1 (en) 2019-06-28 2020-12-30 Amgen Inc. Anti-cgrp receptor/anti-pac1 receptor bispecific antigen binding proteins
KR20220029733A (en) 2019-07-04 2022-03-08 체에스엘 베링 렝나우 아게 truncated von Willebrand factor (VWF) to increase the in vitro stability of coagulation factor VIII
US20220242962A1 (en) 2019-08-12 2022-08-04 Aptevo Research And Development Llc 4-1bb and ox40 binding proteins and related compositions and methods, antibodies against 4-1bb, antibodies against ox40
WO2021092355A1 (en) 2019-11-08 2021-05-14 Amgen Inc. Engineering charge pair mutations for pairing of hetero-igg molecules
JP2023500953A (en) 2019-11-11 2023-01-11 ツェー・エス・エル・ベーリング・レングナウ・アクチエンゲゼルシャフト Polypeptides for inducing tolerance to factor VIII
EP4072598A4 (en) 2019-12-13 2024-02-21 Washington University St Louis Near infrared fluorescent dyes, formulations and related methods
WO2021145946A1 (en) * 2020-01-13 2021-07-22 Invenra Inc. Multispecific treg binding molecules
WO2021146328A1 (en) 2020-01-13 2021-07-22 Aptevo Research And Development Llc Formulations for protein therapeutics
WO2021158469A1 (en) 2020-02-03 2021-08-12 Amgen Inc. Multivariate bracketing approach for sterile filter validation
WO2022006153A1 (en) 2020-06-29 2022-01-06 Resolve Therapeutics, Llc Treatment of sjogren's syndrome with nuclease fusion proteins
JP2023538897A (en) 2020-08-20 2023-09-12 アムジエン・インコーポレーテツド Antigen binding proteins with non-canonical disulfides in the Fab region
US20230374162A1 (en) 2020-10-07 2023-11-23 Amgen Inc. Rational selection of building blocks for the assembly of multispecific antibodies
CA3200603A1 (en) 2020-11-10 2022-05-19 Amgen Inc. Novel linkers of multispecific antigen binding domains
AU2021381360A1 (en) 2020-11-20 2023-06-22 Csl Behring Gmbh Method for treating antibody-mediated rejection
WO2022119976A1 (en) 2020-12-01 2022-06-09 Aptevo Research And Development Llc Heterodimeric psma and cd3-binding bispecific antibodies
US20240101598A1 (en) 2020-12-18 2024-03-28 Richter Gedeon Nyrt. Methods for the purification of refolded fc-peptide fusion protein
AR124681A1 (en) 2021-01-20 2023-04-26 Abbvie Inc ANTI-EGFR ANTIBODY-DRUG CONJUGATES
CA3206884A1 (en) 2021-02-01 2022-08-04 Kevin AKERET Method of treating or preventing an adverse secondary neurological outcome following a haemorrhagic stroke
CA3216559A1 (en) 2021-04-20 2022-10-27 Amgen Inc. Balanced charge distribution in electrostatic steering of chain pairing in multi-specific and monovalent igg molecule assembly
CN117321074A (en) 2021-05-07 2023-12-29 瑞士杰特贝林生物制品有限公司 Expression system for producing recombinant haptoglobin (Hp) beta chain
EP4341291A1 (en) 2021-05-21 2024-03-27 Aptevo Research and Development LLC Dosing regimens for protein therapeutics
WO2022266467A2 (en) 2021-06-17 2022-12-22 Dana-Farber Cancer Institute, Inc. Recombinant histone polypeptide and uses thereof
WO2023056444A1 (en) 2021-10-01 2023-04-06 Janssen Pharmaceutica N.V. Methods of increasing progenitor cell production
US20230192843A1 (en) 2021-10-14 2023-06-22 Teneobio, Inc. Mesothelin binding proteins and uses thereof
AR127458A1 (en) 2021-10-27 2024-01-24 Amgen Inc PREDICTION BASED ON DEEP LEARNING USING SPECTROSCOPY
WO2023173084A1 (en) 2022-03-11 2023-09-14 University Of Rochester Cyclopeptibodies and uses thereof
WO2023180525A1 (en) 2022-03-24 2023-09-28 Richter Gedeon Nyrt. Method for the manufacture of biopharmaceuticals
WO2024026358A1 (en) 2022-07-27 2024-02-01 Teneobio, Inc. Mesothelin binding proteins and uses thereof
WO2024047219A1 (en) 2022-09-02 2024-03-07 Csl Behring Ag Haptoglobin for use in treating or preventing exaggerated erectile response or erectile dysfunction

Family Cites Families (180)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3691016A (en) 1970-04-17 1972-09-12 Monsanto Co Process for the preparation of insoluble enzymes
CA1023287A (en) 1972-12-08 1977-12-27 Boehringer Mannheim G.M.B.H. Process for the preparation of carrier-bound proteins
US4179337A (en) * 1973-07-20 1979-12-18 Davis Frank F Non-immunogenic polypeptides
US3941763A (en) 1975-03-28 1976-03-02 American Home Products Corporation PGlu-D-Met-Trp-Ser-Tyr-D-Ala-Leu-Arg-Pro-Gly-NH2 and intermediates
US4195128A (en) 1976-05-03 1980-03-25 Bayer Aktiengesellschaft Polymeric carrier bound ligands
US4330440A (en) 1977-02-08 1982-05-18 Development Finance Corporation Of New Zealand Activated matrix and method of activation
CA1093991A (en) 1977-02-17 1981-01-20 Hideo Hirohara Enzyme immobilization with pullulan gel
US4229537A (en) 1978-02-09 1980-10-21 New York University Preparation of trichloro-s-triazine activated supports for coupling ligands
IN150740B (en) * 1978-11-24 1982-12-04 Hoffmann La Roche
US4289872A (en) 1979-04-06 1981-09-15 Allied Corporation Macromolecular highly branched homogeneous compound based on lysine units
US4760067A (en) * 1979-08-15 1988-07-26 Merck & Co., Inc. Allylsulfoxide enzyme inhibitors
DE3175151D1 (en) 1980-05-21 1986-09-25 Teijin Ltd Reactive polymer and process for the preparation thereof
US4560649A (en) * 1981-10-15 1985-12-24 Cornell Research Foundation Assaying for hLH or hCG with immobilized hormone receptors
JPH0751511B2 (en) * 1982-03-15 1995-06-05 味の素株式会社 Cancer therapeutic agent containing interleukin-2
DE3378250D1 (en) 1982-04-22 1988-11-24 Ici Plc Continuous release formulations
US4587046A (en) 1982-05-18 1986-05-06 The Regents Of The University Of California Drug-carrier conjugates
EP0098110B1 (en) * 1982-06-24 1989-10-18 NIHON CHEMICAL RESEARCH KABUSHIKI KAISHA also known as JAPAN CHEMICAL RESEARCH CO., LTD Long-acting composition
US4966888A (en) * 1985-07-08 1990-10-30 Cornell Research Foundation, Inc. hCG-hLH receptor and hCG-hLH receptor-hCG complex as antigens, antibodies thereto and contraceptive vaccine
KR850004274A (en) * 1983-12-13 1985-07-11 원본미기재 Method for preparing erythropoietin
NZ210501A (en) * 1983-12-13 1991-08-27 Kirin Amgen Inc Erythropoietin produced by procaryotic or eucaryotic expression of an exogenous dna sequence
US4522750A (en) * 1984-02-21 1985-06-11 Eli Lilly And Company Cytotoxic compositions of transferrin coupled to vinca alkaloids
DE3572982D1 (en) 1984-03-06 1989-10-19 Takeda Chemical Industries Ltd Chemically modified lymphokine and production thereof
SE470099B (en) * 1984-05-17 1993-11-08 Jerker Porath Sulfone activated thioether adsorbents for separation of eg protein
US4578335A (en) * 1984-05-21 1986-03-25 Immunex Corporation Interleukin 2 receptor
US4670563A (en) * 1984-06-20 1987-06-02 Sanofi Imidazolides as intermediates for the synthesis of cytotoxic conjugates
EP0173494A3 (en) 1984-08-27 1987-11-25 The Board Of Trustees Of The Leland Stanford Junior University Chimeric receptors by dna splicing and expression
US4675285A (en) * 1984-09-19 1987-06-23 Genetics Institute, Inc. Method for identification and isolation of DNA encoding a desired protein
SE454885B (en) * 1984-10-19 1988-06-06 Exploaterings Ab Tbf POLYMER COATED PARTICLES WITH IMMOBILIZED METAL ZONES IN ITS SURFACE PROCEDURES FOR PRODUCING THEREOF
US4959314A (en) * 1984-11-09 1990-09-25 Cetus Corporation Cysteine-depleted muteins of biologically active proteins
DE3675588D1 (en) * 1985-06-19 1990-12-20 Ajinomoto Kk HAEMOGLOBIN TIED TO A POLY (ALKENYLENE OXIDE).
US4917888A (en) * 1985-06-26 1990-04-17 Cetus Corporation Solubilization of immunotoxins for pharmaceutical compositions using polymer conjugation
US4766106A (en) * 1985-06-26 1988-08-23 Cetus Corporation Solubilization of proteins for pharmaceutical compositions using polymer conjugation
US4935233A (en) 1985-12-02 1990-06-19 G. D. Searle And Company Covalently linked polypeptide cell modulators
US5985599A (en) 1986-05-29 1999-11-16 The Austin Research Institute FC receptor for immunoglobulin
CA1283046C (en) 1986-05-29 1991-04-16 Nandini Katre Tumor necrosis factor formulation
US4791192A (en) * 1986-06-26 1988-12-13 Takeda Chemical Industries, Ltd. Chemically modified protein with polyethyleneglycol
JP2876058B2 (en) 1986-08-18 1999-03-31 エミスフィア・テクノロジーズ・インコーポレイテッド Drug delivery system
US4931544A (en) * 1986-09-04 1990-06-05 Cetus Corporation Succinylated interleukin-2 for pharmaceutical compositions
IL80005A (en) * 1986-09-10 1992-11-15 Yeda Res & Dev Compositions for modulating the effect of tnf and il-1
US5229490A (en) * 1987-05-06 1993-07-20 The Rockefeller University Multiple antigen peptide system
US5359032A (en) * 1987-08-26 1994-10-25 Biogen Inc. Interkeukin-1 inhibitor
US5512544A (en) * 1987-09-13 1996-04-30 Yeda Research And Development Co. Ltd. Pharmaceutical compositions comprising an anticytokine
IL83878A (en) * 1987-09-13 1995-07-31 Yeda Res & Dev Soluble protein corresponding to tnf inhibitory protein its preparation and pharmaceutical compositions containing it
US5336603A (en) 1987-10-02 1994-08-09 Genentech, Inc. CD4 adheson variants
US4904584A (en) * 1987-12-23 1990-02-27 Genetics Institute, Inc. Site-specific homogeneous modification of polypeptides
US4847325A (en) * 1988-01-20 1989-07-11 Cetus Corporation Conjugation of polymer to colony stimulating factor-1
US5153265A (en) * 1988-01-20 1992-10-06 Cetus Corporation Conjugation of polymer to colony stimulating factor-1
US6018026A (en) * 1988-01-22 2000-01-25 Zymogenetics, Inc. Biologically active dimerized and multimerized polypeptide fusions
EP1762623A1 (en) 1988-01-22 2007-03-14 ZymoGenetics, Inc. Methods for producing biologically active peptide dimers
GB8807803D0 (en) 1988-03-31 1988-05-05 Glaxo Group Ltd Biochemical product
US5214131A (en) * 1988-05-06 1993-05-25 Sumitomo Pharmaceuticals Company, Limited Polyethylene glycol derivatives, modified peptides and production thereof
US5075222A (en) * 1988-05-27 1991-12-24 Synergen, Inc. Interleukin-1 inhibitors
US5223409A (en) 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
US5811261A (en) * 1988-09-12 1998-09-22 Yeda Research And Development Co. Ltd. Expression of the recombinant tumor necrosis factor binding protein I (TBP-I)
US5359037A (en) * 1988-09-12 1994-10-25 Yeda Research And Development Co. Ltd. Antibodies to TNF binding protein I
US5681566A (en) * 1988-10-24 1997-10-28 3I Research Exploitation Limited Antibody conjugates with two or more covalently linked FC regions
US5162430A (en) * 1988-11-21 1992-11-10 Collagen Corporation Collagen-polymer conjugates
WO1990005534A1 (en) 1988-11-23 1990-05-31 Genentech, Inc. Polypeptide derivatives
US5093475A (en) * 1988-12-22 1992-03-03 Xoma Corporation Hindered linking agents derived from 2-iminothiolanes and methods
AU638964B2 (en) 1988-12-22 1993-07-15 Genentech Inc. Method for preparing water soluble polypeptides
US5089261A (en) * 1989-01-23 1992-02-18 Cetus Corporation Preparation of a polymer/interleukin-2 conjugate
US4902502A (en) * 1989-01-23 1990-02-20 Cetus Corporation Preparation of a polymer/interleukin-2 conjugate
US5116964A (en) 1989-02-23 1992-05-26 Genentech, Inc. Hybrid immunoglobulins
US5098833A (en) 1989-02-23 1992-03-24 Genentech, Inc. DNA sequence encoding a functional domain of a lymphocyte homing receptor
US5225538A (en) 1989-02-23 1993-07-06 Genentech, Inc. Lymphocyte homing receptor/immunoglobulin fusion proteins
US5216131A (en) 1989-02-23 1993-06-01 Genentech, Inc. Lymphocyte homing receptors
US5122614A (en) * 1989-04-19 1992-06-16 Enzon, Inc. Active carbonates of polyalkylene oxides for modification of polypeptides
US5166322A (en) * 1989-04-21 1992-11-24 Genetics Institute Cysteine added variants of interleukin-3 and chemical modifications thereof
DE3922089A1 (en) * 1989-05-09 1990-12-13 Basf Ag NEW PROTEINS AND THEIR PRODUCTION
US5627262A (en) 1989-07-05 1997-05-06 The Board Of Regents Of The University Of Oklahoma Method and composition for the treatment of septic shock
US5605690A (en) * 1989-09-05 1997-02-25 Immunex Corporation Methods of lowering active TNF-α levels in mammals using tumor necrosis factor receptor
NZ235148A (en) * 1989-09-05 1991-12-23 Immunex Corp Tumour necrosis factor receptor protein and dna sequences
US5395760A (en) * 1989-09-05 1995-03-07 Immunex Corporation DNA encoding tumor necrosis factor-α and -β receptors
ATE194384T1 (en) * 1989-09-12 2000-07-15 Hoffmann La Roche TNF-BINDING PROTEINS
US5350836A (en) * 1989-10-12 1994-09-27 Ohio University Growth hormone antagonists
US5013556A (en) 1989-10-20 1991-05-07 Liposome Technology, Inc. Liposomes with enhanced circulation time
EP0502956B1 (en) * 1989-11-29 1997-04-23 Amgen Boulder Inc. Production of recombinant human interleukin-1 inhibitor
DE59105962D1 (en) 1990-01-23 1995-08-17 Merck Patent Gmbh LIQUID CRYSTAL MEDIUM.
US5136021A (en) * 1990-02-27 1992-08-04 Health Research, Inc. TNF-inhibitory protein and a method of production
US5171264A (en) * 1990-02-28 1992-12-15 Massachusetts Institute Of Technology Immobilized polyethylene oxide star molecules for bioapplications
US5349053A (en) 1990-06-01 1994-09-20 Protein Design Labs, Inc. Chimeric ligand/immunoglobulin molecules and their uses
US5723286A (en) 1990-06-20 1998-03-03 Affymax Technologies N.V. Peptide library and screening systems
AU665190B2 (en) 1990-07-10 1995-12-21 Cambridge Antibody Technology Limited Methods for producing members of specific binding pairs
CA2086323C (en) 1990-07-17 2002-05-14 Rodger P. Mcever Functionally active selectin-derived peptides and ligand for gmp-140
GB9022648D0 (en) * 1990-10-18 1990-11-28 Charing Cross Sunley Research Polypeptide and its use
US5252714A (en) * 1990-11-28 1993-10-12 The University Of Alabama In Huntsville Preparation and use of polyethylene glycol propionaldehyde
NZ241954A (en) 1991-03-15 1994-01-26 Amgen Inc Compositions of g-csf for pulmonary administration.
US6139843A (en) 1991-04-02 2000-10-31 Albert Einstein College Of Medicine Of Yeshiva University Peptide compositions for the treatment of HIV
IL99120A0 (en) * 1991-08-07 1992-07-15 Yeda Res & Dev Multimers of the soluble forms of tnf receptors,their preparation and pharmaceutical compositions containing them
US5270170A (en) 1991-10-16 1993-12-14 Affymax Technologies N.V. Peptide library and screening method
US5733731A (en) 1991-10-16 1998-03-31 Affymax Technologies N.V. Peptide library and screening method
US5376367A (en) * 1991-11-22 1994-12-27 Immunex Corporation Fusion proteins comprising MGF and IL-3
US5569779A (en) * 1991-12-23 1996-10-29 Albemarle Corporation Polyfunctional michael addition products
CA2118130A1 (en) 1992-04-14 1993-10-28 Jean M. J. Frechet Dendritic based macromolecules and method of production
EP0640094A1 (en) 1992-04-24 1995-03-01 The Board Of Regents, The University Of Texas System Recombinant production of immunoglobulin-like domains in prokaryotic cells
WO1993024135A1 (en) 1992-05-26 1993-12-09 Immunex Corporation Novel cytokine that binds cd30
US5792451A (en) * 1994-03-02 1998-08-11 Emisphere Technologies, Inc. Oral drug delivery compositions and methods
AU5098393A (en) * 1992-08-14 1994-03-15 Government Of The United States Of America, As Represented By The Secretary Of The Department Of Health And Human Services, The Recombinant toxin with increased half-life
US5382657A (en) * 1992-08-26 1995-01-17 Hoffmann-La Roche Inc. Peg-interferon conjugates
KR100232688B1 (en) * 1992-09-15 1999-12-01 스코트 쥐. 홀퀴스트 Pharmaceutical compositions for treating tnf-dependent inflammation using tumor necrosis factor antagonists
EP1550729B1 (en) 1992-09-25 2009-05-27 Avipep Pty Limited Target binding polypeptide comprising an IG-like VL domain linked to an IG-like VH domain
NZ247231A (en) * 1993-03-23 1994-10-26 Holyoake Ind Ltd Diffuser for air conditioning system; outlet air direction thermostatically controlled
EP0710121B1 (en) * 1993-07-30 2000-10-11 Kennedy Institute Of Rheumatology Method for treating multiple sclerosis
WO1995009917A1 (en) 1993-10-07 1995-04-13 The Regents Of The University Of California Genetically engineered bispecific tetravalent antibodies
US5922545A (en) 1993-10-29 1999-07-13 Affymax Technologies N.V. In vitro peptide and antibody display libraries
US5998172A (en) 1993-11-02 1999-12-07 Duke University Anti-CD6 ligand antibodies
US5446090A (en) * 1993-11-12 1995-08-29 Shearwater Polymers, Inc. Isolatable, water soluble, and hydrolytically stable active sulfones of poly(ethylene glycol) and related polymers for modification of surfaces and molecules
US5773569A (en) 1993-11-19 1998-06-30 Affymax Technologies N.V. Compounds and peptides that bind to the erythropoietin receptor
US5981478A (en) 1993-11-24 1999-11-09 La Jolla Cancer Research Foundation Integrin-binding peptides
GB2285446B (en) 1994-01-03 1999-07-28 Genentech Inc Thrombopoietin
US5786331A (en) 1994-02-02 1998-07-28 Affymax Technologies N.V. Peptides and compounds that bind to the IL-1 receptor
US5608035A (en) 1994-02-02 1997-03-04 Affymax Technologies N.V. Peptides and compounds that bind to the IL-1 receptor
US5880096A (en) 1994-02-02 1999-03-09 Affymax Technologies N.V. Peptides and compounds that bind to the IL-1 receptor
WO1995021919A2 (en) 1994-02-14 1995-08-17 Kirin Brewery Company, Limited Protein having tpo activity
CZ221496A3 (en) 1994-02-14 1997-07-16 Zymogenetics Inc Haematopoetic protein, materials and processes for preparing thereof
EP0690127B1 (en) 1994-03-31 1998-08-05 Amgen Inc. Compositions and methods for stimulating megakaryocyte growth and differentiation
BR9507618A (en) 1994-05-06 1997-08-19 Roussy Inst Gustave Soluble polypeptide fractions of the lag-3 protein production process therapeutic composition anti-idiotype antibodies
US6184205B1 (en) 1994-07-22 2001-02-06 University Of North Carolina At Chapel Hill GRB2 SH3 binding peptides and methods of isolating and using same
US6309853B1 (en) 1994-08-17 2001-10-30 The Rockfeller University Modulators of body weight, corresponding nucleic acids and proteins, and diagnostic and therapeutic uses thereof
IL111196A0 (en) 1994-10-07 1994-12-29 Yeda Res & Dev Peptides and pharmaceutical compositions comprising them
US5824784A (en) 1994-10-12 1998-10-20 Amgen Inc. N-terminally chemically modified protein compositions and methods
AU693478B2 (en) 1994-11-10 1998-07-02 Metabolic Pharmaceuticals Limited Treatment of obesity
JPH10510155A (en) 1994-12-07 1998-10-06 バイオネブラスカ・インコーポレイテッド Method for producing peptide using recombinant fusion protein construct
EP1621206A1 (en) 1994-12-12 2006-02-01 Beth Israel Deaconess Medical Center, Inc. Chimeric cytokines and uses thereof
AU3204895A (en) 1995-02-01 1996-08-21 University Of Massachusetts Medical Center Methods of selecting a random peptide that binds to a target protein
IL113159A0 (en) 1995-03-28 1995-06-29 Yeda Res & Dev Synthetic peptides and pharmaceutical compositions comprising them
US5739277A (en) 1995-04-14 1998-04-14 Genentech Inc. Altered polypeptides with increased half-life
US6096871A (en) * 1995-04-14 2000-08-01 Genentech, Inc. Polypeptides altered to contain an epitope from the Fc region of an IgG molecule for increased half-life
YU34196A (en) 1995-06-07 1999-03-04 Glaxo Group Limited Peptides and compounds that bind to a receptor
US5767078A (en) 1995-06-07 1998-06-16 Johnson; Dana L. Agonist peptide dimers
ZA964814B (en) 1995-06-07 1998-02-09 Glaxo Group Ltd Peptides and compounds that bind to a receptor.
US5869451A (en) 1995-06-07 1999-02-09 Glaxo Group Limited Peptides and compounds that bind to a receptor
IL118524A (en) 1995-06-19 2004-02-19 Akzo Nobel Nv Peptides and pharmaceutical compositions containing them useful in a peptide tolerance therapy
US5955574A (en) 1995-07-13 1999-09-21 Societe De Conseils De Recherches Et D'applications Scientifiques, S.A. Analogs of parathyroid hormone
WO1997008553A1 (en) 1995-08-22 1997-03-06 The Regents Of The University Of California Targeting of proteins to the cell wall of gram-positive bacteria
US5817750A (en) 1995-08-28 1998-10-06 La Jolla Cancer Research Foundation Structural mimics of RGD-binding sites
US6369027B1 (en) 1995-12-22 2002-04-09 Amgen Inc. Osteoprotegerin
US5723125A (en) * 1995-12-28 1998-03-03 Tanox Biosystems, Inc. Hybrid with interferon-alpha and an immunoglobulin Fc linked through a non-immunogenic peptide
EP0904112B1 (en) 1996-02-09 2009-11-18 Amgen Inc., Composition comprising il-1ra as interleukin-1 inhibitor and hyaluronan as controlled release polymer
IL117223A0 (en) 1996-02-22 1996-06-18 Yeda Res & Dev Antipathogenic polypeptides and compositions comprising them
US5747639A (en) * 1996-03-06 1998-05-05 Amgen Boulder Inc. Use of hydrophobic interaction chromatography to purify polyethylene glycols
ATE279947T1 (en) 1996-03-18 2004-11-15 Univ Texas IMMUNOGLOBULIN-LIKE DOMAIN WITH INCREASED HALF-LIFE TIMES
US5798246A (en) 1996-03-25 1998-08-25 Incyte Pharmaceuticals, Inc. Cyclic nucleotide phosphodiesterase
EP0906419A2 (en) 1996-03-28 1999-04-07 Chiron Corporation Peptide ligands of the urokinase receptor
IL118003A0 (en) 1996-04-23 1996-08-04 Yeda Res & Dev Novel vip fragments and pharmaceutical compositions comprising them
US6100071A (en) 1996-05-07 2000-08-08 Genentech, Inc. Receptors as novel inhibitors of vascular endothelial growth factor activity and processes for their production
WO1997043316A1 (en) 1996-05-10 1997-11-20 Beth Israel Deaconess Medical Center, Inc. Physiologically active molecules with extended half-lives and methods of using same
CA2257108A1 (en) 1996-06-07 1997-12-11 Takeda Chemical Industries, Ltd. Novel peptides and production and use thereof
US6126939A (en) 1996-09-03 2000-10-03 Yeda Research And Development Co. Ltd. Anti-inflammatory dipeptide and pharmaceutical composition thereof
JP2001501600A (en) 1996-09-10 2001-02-06 ザ バーナム インスティテュート Tumor homing molecules, conjugates derived therefrom, and methods of using the same
US5932546A (en) 1996-10-04 1999-08-03 Glaxo Wellcome Inc. Peptides and compounds that bind to the thrombopoietin receptor
AU4474497A (en) 1996-10-08 1998-05-05 U-Bisys B.V. Methods and means for selecting peptides and proteins having specific affinity for a target
US5958703A (en) * 1996-12-03 1999-09-28 Glaxo Group Limited Use of modified tethers in screening compound libraries
EP2002846B1 (en) 1996-12-06 2017-01-25 Amgen Inc. Combination therapy using an IL-1 inhibitor for treating IL-1 mediated diseases
DK0954588T3 (en) 1996-12-20 2007-05-14 Amgen Inc OB fusion protein compositions and methods
KR19980066046A (en) 1997-01-18 1998-10-15 정용훈 High-CTLA4-Ig fusion protein
US5955431A (en) 1997-02-05 1999-09-21 Brigham And Women's Hospital, Inc. Mast cell protease peptide inhibitors
AU7132798A (en) 1997-04-17 1998-11-11 Amgen, Inc. Compositions comprising conjugates of stable, active, human ob protein with antibody fc chain and methods
US6265535B1 (en) 1997-05-30 2001-07-24 The Trustees Of The University Of Pennsylvania Peptides and peptide analogues designed from binding sites of tumor necrosis factor receptor superfamily and their uses
IL133315A0 (en) 1997-06-06 2001-04-30 Regeneron Pharma Ntn-2 member of tnf ligand family
EP0998577B1 (en) 1997-07-24 2004-10-27 Perseptive Biosystems, Inc. Conjugates of transporter peptides and nucleic acid analogs, and their use
US6238667B1 (en) 1997-09-19 2001-05-29 Heinz Kohler Method of affinity cross-linking biologically active immunogenic peptides to antibodies
AU9790798A (en) 1997-10-06 1999-04-27 Millennium Pharmaceuticals, Inc. Signal peptide containing proteins and uses therefor
JP4439111B2 (en) 1997-10-10 2010-03-24 サイトビア インコーポレイテッド Dipeptide apoptosis inhibitors and uses thereof
JP2001522863A (en) 1997-11-07 2001-11-20 コンジュケム,インコーポレーテッド Novel conjugate of RGD-containing peptide and endogenous carrier
US20020142374A1 (en) 1998-08-17 2002-10-03 Michael Gallo Generation of modified molecules with increased serum half-lives
US6660843B1 (en) * 1998-10-23 2003-12-09 Amgen Inc. Modified peptides as therapeutic agents
PT1135153E (en) * 1998-11-20 2005-09-30 Genentech Inc USES FOR ANTAGONISTS AND AGNISTS OF EPH RECEPTORS FOR TREATING VASCULAR DISORDERS
WO2000047740A2 (en) * 1999-02-12 2000-08-17 Amgen Inc. Tnf-related proteins
CA2377731A1 (en) 1999-07-02 2001-01-11 Genentech, Inc. Fusion peptides comprising a peptide ligand domain and a multimerization domain
EP1189931B1 (en) 1999-07-02 2006-06-21 Genentech, Inc. Peptide compounds that bind her2
MXPA02007574A (en) * 2000-02-11 2002-12-13 Amgen Inc Receptor from tnf family.
US7521053B2 (en) * 2001-10-11 2009-04-21 Amgen Inc. Angiopoietin-2 specific binding agents
US7138370B2 (en) * 2001-10-11 2006-11-21 Amgen Inc. Specific binding agents of human angiopoietin-2
US7658924B2 (en) * 2001-10-11 2010-02-09 Amgen Inc. Angiopoietin-2 specific binding agents
AU2003280130B2 (en) 2002-06-28 2009-06-11 Centocor, Inc. Mammalian CH1 deleted mimetibodies, compositions, methods and uses
WO2004002424A2 (en) 2002-06-28 2004-01-08 Centocor, Inc. Mammalian epo mimetic ch1 deleted mimetibodies, compositions, methods and uses
US6919426B2 (en) * 2002-09-19 2005-07-19 Amgen Inc. Peptides and related molecules that modulate nerve growth factor activity
JP2008504248A (en) * 2004-06-25 2008-02-14 ライセンティア・リミテッド Tie receptor and Tie ligand substance and method for controlling female fertility
WO2006068953A2 (en) * 2004-12-21 2006-06-29 Astrazeneca Ab Antibodies directed to angiopoietin-2 and uses thereof

Also Published As

Publication number Publication date
JP2007084556A (en) 2007-04-05
SK5252001A3 (en) 2002-12-03
AU2004200690A1 (en) 2004-03-18
AU2004200690C1 (en) 2009-04-09
WO2000024782A3 (en) 2002-06-06
CZ304242B6 (en) 2014-01-22
CN1908014A (en) 2007-02-07
HUP0203506A3 (en) 2012-09-28
HK1042097B (en) 2008-04-03
JP2007091747A (en) 2007-04-12
US20040057953A1 (en) 2004-03-25
ES2299278T3 (en) 2008-05-16
EP1144454B2 (en) 2012-12-12
NZ528882A (en) 2005-06-24
IL142365A0 (en) 2002-03-10
JP2003512011A (en) 2003-04-02
CN1721447A (en) 2006-01-18
CY1107881T1 (en) 2013-06-19
NO331733B1 (en) 2012-03-12
CN100384880C (en) 2008-04-30
US7189827B2 (en) 2007-03-13
RS51852B (en) 2012-02-29
IL142365A (en) 2013-02-28
US20040044188A1 (en) 2004-03-04
KR20070050977A (en) 2007-05-16
AU2004200687C1 (en) 2009-03-26
CN1781947A (en) 2006-06-07
EA200100464A1 (en) 2002-10-31
AU767725C (en) 2000-05-15
JP2007084558A (en) 2007-04-05
ES2299278T5 (en) 2013-04-03
DE69937752T2 (en) 2008-12-24
SK287037B6 (en) 2009-10-07
DE69937752T3 (en) 2013-02-28
CN1781945A (en) 2006-06-07
SI1144454T2 (en) 2013-04-30
CN1310948C (en) 2007-04-18
PT1144454E (en) 2008-02-08
WO2000024782A2 (en) 2000-05-04
CN1781946A (en) 2006-06-07
JP2007089586A (en) 2007-04-12
HK1090932A1 (en) 2007-01-05
NZ510888A (en) 2004-01-30
CN1746189A (en) 2006-03-15
AU1232200A (en) 2000-05-15
ZA200102753B (en) 2002-06-11
EP1144454B1 (en) 2007-12-12
JP2007084557A (en) 2007-04-05
US7186810B2 (en) 2007-03-06
EA005404B1 (en) 2005-02-24
MXPA01003873A (en) 2003-10-14
JP2007084555A (en) 2007-04-05
DK1144454T3 (en) 2008-05-05
AU2004200690B2 (en) 2007-05-10
KR20070050978A (en) 2007-05-16
JP2007105044A (en) 2007-04-26
SI1144454T1 (en) 2008-04-30
PL211164B1 (en) 2012-04-30
US20040071712A1 (en) 2004-04-15
KR100753304B1 (en) 2007-08-29
AU2004200687B2 (en) 2007-05-10
DE69937752D1 (en) 2008-01-24
US20040053845A1 (en) 2004-03-18
CN100384879C (en) 2008-04-30
KR100753303B1 (en) 2007-08-29
CA2347131A1 (en) 2000-05-04
JP2007091746A (en) 2007-04-12
US6660843B1 (en) 2003-12-09
YU25901A (en) 2005-07-19
EP1144454A2 (en) 2001-10-17
US20070049532A1 (en) 2007-03-01
AU2004200687A1 (en) 2004-03-18
BG65721B1 (en) 2009-08-31
NO20011963L (en) 2001-06-21
US7166707B2 (en) 2007-01-23
KR100753305B1 (en) 2007-08-29
DK1144454T4 (en) 2013-01-14
HU229485B1 (en) 2014-01-28
PL366080A1 (en) 2005-01-24
US7169905B2 (en) 2007-01-30
ATE380828T1 (en) 2007-12-15
AU767725B2 (en) 2003-11-20
BR9914708A (en) 2002-07-16
NO20011963D0 (en) 2001-04-20
KR20010099706A (en) 2001-11-09
HUP0203506A2 (en) 2003-02-28
US20040087778A1 (en) 2004-05-06
CN1331701A (en) 2002-01-16
BG105461A (en) 2003-04-30
CN1746190A (en) 2006-03-15
HK1042097A1 (en) 2002-08-02

Similar Documents

Publication Publication Date Title
CA2347131C (en) Modified peptides as therapeutic agents
AU2001259432B2 (en) Modified peptides, comprising an FC domain, as therapeutic agents
US7488590B2 (en) Modified peptides as therapeutic agents
AU2001259432A1 (en) Modified peptides, comprising an Fc domain, as therapeutic agents
AU2004200691C9 (en) Modified peptides as therapeutic agents
AU2004231208A1 (en) Modified peptides as therapeutic agents

Legal Events

Date Code Title Description
EEER Examination request
MKEX Expiry

Effective date: 20191025