WO2005000892A2 - Glp-1 analog fusion plroteins - Google Patents

Glp-1 analog fusion plroteins Download PDF

Info

Publication number
WO2005000892A2
WO2005000892A2 PCT/US2004/015595 US2004015595W WO2005000892A2 WO 2005000892 A2 WO2005000892 A2 WO 2005000892A2 US 2004015595 W US2004015595 W US 2004015595W WO 2005000892 A2 WO2005000892 A2 WO 2005000892A2
Authority
WO
WIPO (PCT)
Prior art keywords
gly
glu
ser
val
lys
Prior art date
Application number
PCT/US2004/015595
Other languages
French (fr)
Other versions
WO2005000892A3 (en
Inventor
Wolfgang Glaesner
Rohn Lee. Millican, Jr.
Andrew Mark Vick
Original Assignee
Eli Lilly And Company
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=33551775&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2005000892(A2) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to CN200480015953XA priority Critical patent/CN1802386B/en
Priority to NZ543292A priority patent/NZ543292A/en
Priority to CA2528591A priority patent/CA2528591C/en
Priority to AU2004251145A priority patent/AU2004251145C1/en
Priority to EP04752589A priority patent/EP1641823B1/en
Priority to US10/558,627 priority patent/US7452966B2/en
Priority to DK04752589.4T priority patent/DK1641823T3/en
Priority to BRPI0411132A priority patent/BRPI0411132B8/en
Priority to EA200600015A priority patent/EA008831B1/en
Priority to JP2006533197A priority patent/JP4629047B2/en
Priority to AT04752589T priority patent/ATE525395T1/en
Application filed by Eli Lilly And Company filed Critical Eli Lilly And Company
Priority to PL04752589T priority patent/PL1641823T3/en
Priority to MXPA05013565A priority patent/MXPA05013565A/en
Priority to KR1020057023668A priority patent/KR100758755B1/en
Priority to SI200431776T priority patent/SI1641823T1/en
Publication of WO2005000892A2 publication Critical patent/WO2005000892A2/en
Publication of WO2005000892A3 publication Critical patent/WO2005000892A3/en
Priority to IL171926A priority patent/IL171926A/en
Priority to US12/262,832 priority patent/US8273854B2/en
Priority to HR20110714T priority patent/HRP20110714T1/en
Priority to FR15C0010C priority patent/FR15C0010I2/en
Priority to CY2015002C priority patent/CY2015002I2/en
Priority to BE2015C007C priority patent/BE2015C007I2/fr
Priority to LTPA2015007C priority patent/LTC1641823I2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/26Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against hormones ; against hormone releasing or inhibiting factors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • 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
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/48Drugs for disorders of the endocrine system of the pancreatic hormones
    • 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
    • C07K14/50Fibroblast growth factors [FGF]
    • C07K14/503Fibroblast growth factors [FGF] basic FGF [bFGF]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • C07K14/605Glucagons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto

Definitions

  • the present invention relates to glucagon-like peptide analogs fused to proteins that have the effect of extending the in vivo half-life of the peptides. These fusion proteins can be used to treat diabetes as well as a variety of other conditions or disorders.
  • Glucagon-like peptide- 1 (GLP-1) analogs and derivatives show promise in clinical trials for the treatment of type 2 diabetes. GLP-1 induces numerous biological effects such as stimulating insulin secretion, inhibiting glucagon secretion, inhibiting gastric emptying, inhibiting gastric motility or intestinal motility, and inducing weight loss.
  • GLP-1 A significant characteristic of GLP-1 is its ability to stimulate insulin secretion without the associated risk of hypoglycemia that is seen when using insulin therapy or some types of oral therapies that act by increasing insulin expression.
  • the usefulness of therapy involving GLP-1 peptides has been limited by the fact that GLP-l(l-37) is poorly active, and the two naturally occurring truncated peptides, GLP-l(7-37)OH and GLP-1(7-36)NH 2 , are rapidly cleared in vivo and have extremely short in vivo half lives.
  • DPP-IV dipeptidyl-peptidase IN
  • GLP-1 peptide or reduce clearance of the peptide from the body while maintaining biological activity involves fusing a GLP-1 peptide to the Fc portion of an immunoglobulin.
  • Immunoglobulins typically have long circulating half-lives in vivo. For example, IgG molecules can have a half-life in humans of up to 23 days.
  • the Fc portion of the immunoglobulin is responsible, in part, for this in vivo stability.
  • GLP-1 -Fc fusion proteins take advantage of the stability provided by the Fc portion of an immunoglobulin while preserving the biological activity of the GLP-1 molecule.
  • the present invention seeks to overcome the problems associated with the potential immunogenicity and effector activity associated with administration of GLP-1 - Fc fusions by identifying specific GLP-1 -Fc fusion proteins that have a reduced risk of inducing an immune response after repeated and prolonged administration and no longer have effector function.
  • Compounds of the present invention include heterologous fusion proteins comprising a GLP-1 analog comprising a sequence selected from the group consisting of a) (SEQ LD NO:l) His-Xaa 8 -Glu-Gly-Thr-Phe-Thr-Ser-Asp ⁇ Val-Ser-Ser-Tyr-Leu-Glu-Glu- Gln-Ala-Ala-Lys-Glu-Phe-Ile-Ala-Trp-Leu-Val-Lys-Gly-Gly-Gly-Gly wherein Xaa 8 is selected from Gly and Val; b) (SEQ ID ⁇ O:2) His-Xaa 8 -Glu-Gly-Thr-Phe-Thr-Ser-Asp-Val-Ser-Ser-Tyr-Leu-Glu-Glu- Gln-Ala-Ala-Lys-Glu-Phe-Ile-Ala-Trp-Leu-Lys-Asn-
  • the C- terminus of the GLP-1 analog portion and the N- terminus of the Fc portion of the heterologous fusion proteins of the present invention are preferably fused together via 1, 1.5 or 2 repeats of a G-rich peptide linker having the sequence Gly-Gly-Gly-Gly- Ser-Gly-Gly-Gly-Gly-Ser-Gly-Gly-Gly-Gly-Gly-Ser (SEQ ID NO: 8).
  • the present invention also includes polynucleotides encoding the heterologous fusion proteins of the present invention, as well as vectors and host cells comprising such polynucleotides.
  • the heterologous fusion proteins of the present invention comprise a GLP-1 analog portion and an Fc portion.
  • the GLP-1 analog portion and the Fc portion comprise substitutions to the native GLP-1 sequence and the human IgG4 sequence respectively that provide the protein with increased potency and in vivo stability compared to native GLP-1 or GLP-1 analogs not fused to an Fc sequence while decreasing the potential for inducing antibody formation after prolonged and repeated administration in humans.
  • Native GLP-1 is processed in vivo such that the first 6 amino acids are cleaved from the molecule.
  • the amino terminus of GLP-1 has been assigned the number 7 and the carboxy- terminus, number 37.
  • the other amino acids in the polypeptide are numbered consecutively as shown in SEQ ID NO:9.
  • position 8 is alanine and position 22 is glycine.
  • the processed peptide may be further modified in vivo such that the C-terminal glycine residue is removed and replaced with an amide group.
  • GLP-l(7-37)OH and GLP-l(7-36)amide represent the two native forms of the molecule.
  • GLP-1 (7-37)OH has the amino acid sequence of SEQ ID NO:9:
  • the GLP-1 analog portion of the heterologous fusion protein comprises three primary substitutions at positions 8, 22, and 36 relative to native GLP-1(7- 37).
  • the substitution at position 8 reduces the rate at which the endogenous enzyme dipeptidyl-peptidase IV (DPP-IV) inactivates the analog.
  • DPP-IV cleaves native GLP-1 between the 2 nd and 3 rd amino acids (between position 8 and 9) and the resulting molecule is less active.
  • the heterologous fusion proteins of the present invention are DPP- IV resistant.
  • the substitution at position 22 reduces the potential of the molecule to aggregate and increases the potency of the molecule.
  • T H cell activation is initiated by interaction of the T-cell receptor (TCR)-CD3 complex with a processed antigenic peptide bound to a class II major histocompatibility (MHC) molecule in the presence of an antigen-presenting cell (APC).
  • TCR T-cell receptor
  • MHC major histocompatibility
  • T H cell Interaction of a T H cell with antigen initiates a cascade of biochemical events that induces the resting T H cell to enter the cell cycle (Go to Gi transition).
  • the activated T cell progresses through the cell cycle, proliferating and differentiating into memory cells or effector cells.
  • Epitope as used herein refers to a region of a protein molecule to which an antibody can bind.
  • An immunogenic epitope is defined as the part of the protein that elicits an antibody response when the whole protein is the immunogen.
  • Epitope mapping involved the scanning of sequences using a sliding nine amino acid window coupled with advanced statistical analysis techniques to extract the information contained in these patterns.
  • a proprietary software package known as EpiMatrixTM was used to analyze the sequence and identify peptides that are highly likely to provoke an immune response when presented to T-cells. Eight highly common alleles were used in the analysis for Class II MHC receptor interaction.
  • a strong epitope was predicted to be located at the junction of the C- terminus of the GLP-1 analog portion and the beginning of the linker. The sequence of this epitope is Trp-Leu-Val-Lys-Gly-Arg-Gly-Gly-Gly (SEQ ID NO: 11) which interacts with DRB 1 *0801.
  • the present invention encompasses the discovery that this epitope can be eliminated by changing the GLP-1 analog C-terminus to one of the following sequences: Trp-Leu-Val-Lys-Gly-Gly-Gly (SEQ ID NO: 12); Trp-Leu-Lys-Asn-Gly-Gly-Gly (SEQ ID NO: 13); Trp-Leu- Val-Lys-Gly-Gly-Pro (SEQ ID NO: 14); Trp-Leu-Lys-Asn-Gly-Gly-Pro (SEQ ID NO: 15); Trp-Leu-Val-Lys-Gly-Gly (SEQ ID NO: 16); and Trp-Leu-Lys-Asn- Gly-Gly (SEQ ID NO:17).
  • the heterologous fusion proteins of the present invention contain an Fc portion which is derived from human IgG4, but comprises one or more substitutions compared to the wild-type human sequence.
  • the Fc portion of an immunoglobulin has the meaning commonly given to the term in the field of immunology. Specifically, this term refers to an antibody fragment which does not contain the two antigen binding regions (the Fab fragments) from the antibody.
  • the Fc portion consists of the constant region of an antibody from both heavy chains, which associate through non-covalent interactions and disulfide bonds.
  • the Fc portion can include the hinge regions and extend through the CH2 and CH3 domains to the c-terminus of the antibody.
  • the Fc portion can further include one or more glycosylation sites.
  • IgG human immunoglobulins with different effector functions and pharmcokinetic properties.
  • IgG is the most stable of the five types having a serum half -life in humans of about 23 days.
  • IgG subclasses Gl, G2, G3, and G4 each of which have different biological functions known as effector functions.
  • Fc ⁇ R Fc receptor
  • binding Clq and fixing complement Fc receptor
  • binding to Fc ⁇ R can lead to antibody dependent cell mediated cytolysis, whereas binding to complement factors can lead to complement mediated cell lysis.
  • Fc ⁇ R Fc receptor
  • complement factors can lead to complement mediated cell lysis.
  • heterologous Fc fusion proteins wherein the Fc portion is being utilized solely for its ability to extend half-life, it is important to minimize any effector function.
  • the heterologous fusion proteins of the present invention are derived from the human IgG4 Fc region because of its reduced ability to bind Fc ⁇ R and complement factors compared to other IgG sub-types. IgG4, however, has been shown to deplete target cells in humans [Issacs et al., (1996) Clin. Exp. Immunol. 106:427-433]. Because the heterologous fusion proteins of the present invention target beta cells in the pancreas to induce insulin expression, using an IgG4 derived region in an Fc fusion protein could initiate an immune response against the pancreateic beta cell through interaction of the fusion protein with the GLP-1 receptor present on pancreatic beta cells.
  • the IgG4 Fc region which is part of the heterologous fusion proteins of the present invention contains substitutions that eliminate effector function.
  • the IgG4 Fc portion of the fusion proteins of the present invention may contain one or more of the following substitutions: substitution of proline for glutamate at residue 233, alanine or valine for phenylalanine at residue 234 and alanine or glutamate for leucine at residue 235 (EU numbering, Kabat, E.A. et al. (1991) Sequences of Proteins of Immunological Interest, 5 Ed. U.S. Dept. of Health and Human Services, Bethesda, MD, NIH Publication no. 91-3242).
  • residues corresponds to positions 16, 17 and 18 in SEQ ID NO:7.
  • removing the N-linked glycosylation site in the IgG4 Fc region by substituting Ala for Asn at residue 297 (EU numbering) which corresponds to position 80 of SEQ ID NO:7 is another way to ensure that residual effector activity is eliminated in the context of a heterologous fusion protein.
  • the IgG4 Fc portion of the heterologous fusion proteins of the present invention contain a substitution that stabilizes heavy chain dimer formation and prevents the formation of half-IgG4 Fc chains.
  • the heterologous fusion proteins of the present invention preferably exist as dimers joined together by disulfide bonds and various non-covalent interactions.
  • Wild-type IgG4 contains a Pro-Pro-Cys-Pro-Ser-Cys (SEQ ID NO: 18) motif beginning at residue 224 (EU numbering).
  • This motif in a single GLP-1 analog-Fc chain forms disulfide bonds with the corresponding motif in another GLP-1 analog-Fc chain.
  • the presence of serine in the motif causes the formation of single chain fusion proteins.
  • the present invention encompasses heterologous Fc fusion proteins wherein the IgG4 sequence is further modified such that serine at position at 228 (EU numbering) is substituted with proline (amino acid residue ll in SEQ ID NO:7).
  • the C-terminal lysine residue present in the native molecule may be deleted in the IgG4 derivative Fc portion of the heterologous fusion proteins discussed herein (position 230 of SEQ ID NO:7; deleted lysine referred to as des- K).
  • Fusion proteins expressed in some cell types (such as NSO cells) wherein lysine is encoded by the C-terminal codon are heterogeneous in that a portion of the molecules have lysine as the C-terminal amino acid and a portion have lysine deleted.
  • the deletion is due to protease action during expression in some types of mammalian cells.
  • Fc fusion expression constructs lack a C-terminal codon for lysine.
  • the C-terminal amino acid of the GLP-1 analog portion discussed herein is fused to the N-terminus of the IgG4 Fc analog portion via a glycine-rich linker.
  • the in vivo function and stability of the heterologous fusion proteins of the present invention can be optimized by adding small peptide linkers to prevent potentially unwanted domain interactions.
  • a glycine- rich linker provides some structural flexibility such that the GLP-1 analog portion can interact productively with the GLP-1 receptor on target cells such as the beta cells of the pancreas. These linkers, however, can significantly increase the risk that the fusion protein will be immunogenic in vivo.
  • the preferred glycine-rich linker comprises the sequence: Gly-Gly-Gly-Gly-Ser-Gly-Gly-Gly-Gly- Gly-Ser-Gly-Gly-Gly-Gly-Ser (SEQ ID NO:8). Although more copies of this linker may be used in the heterologous fusion proteins of the present invention, it is preferred that a single copy of this linker be used to minimize the risk of immunogenicity associated with prolonged and repeated administration.
  • Preferred GLP-1 -Fc heterologous fusion proteins of the present invention include the following proteins: Gly 8 -Glu 22 -Gly 36 -GLP-l(7-37)-lL-IgG4 (S228P), Gly 8 -Glu 22 - Gly 36 -GLP-l(7-37)-lL-IgG4 (S228P, F234A, L235A), Gly 8 -Glu 22 -Gly 36 -GLP-l(7-37)- lL-IgG4 (S228P, N297A), Gly 8 -Glu 22 -Gly 36 -GLP-l(7-37)-lL-IgG4 (S228P, F234A, L235A, N297A), Gly 8 -Glu 22 -Gly 36 -GLP-l(7-37)-1.5L-IgG4 (S228P), Gly 8 -Glu 22 -Gly 36 -GLP
  • GLP-1 (7-37) indicates that the GLP-1 portion of the mature fusion protein begins with His at position 7 and ends with Gly at position 37.
  • L refers to a linker with the sequence Gly-Gly-Gly-Gly-Ser-Gly-Gly-Gly-Gly-Gly-Ser-Gly-Gly-Gly-Gly-Gly-Gly-Ser (SEQ ID NO:8).
  • the number immediately preceding the L refers to the number of linkers separating the GLP-1 portion from the Fc portion.
  • a linker specified as 1.5L refers to the sequence Gly-Ser- Gly-Gly-Gly-Gly-Ser-Gly-Gly-Gly-Gly-Gly-Ser-Gly-Gly-Gly-Gly-Gly-Ser-Gly-Gly-Gly-Gly-Gly-Gly-Ser (SEQ ID NO: 19)
  • IgG4 refers to an analog of the human IgG4 Fc sequence specified as SEQ ID NO:7. Substitutions in the IgG4 Fc portion of the heterologous fusion protein are indicated in parenthesis.
  • the wild-type amino acid is specified by its common abbreviation followed by the position number in the context of the entire IgG4 sequence using the EU numbering system followed by the amino acid being substituted at that position specified by its common abbreviation.
  • the heterologous fusion proteins of the present invention can be made by a variety of different methods, because of the size of the fusion protein, recombinant methods are preferred.
  • the following general molecular biology terms and abbreviations are defined below.
  • “Base pair” or "bp” as used herein refers to DNA or RNA.
  • the abbreviations A,C,G, and T correspond to the 5 '-monophosphate forms of the deoxyribonucleosides (deoxy)adenosine, (deoxy)cytidine, (deoxy)guanosine, and thymidine, respectively, when they occur in DNA molecules.
  • the abbreviations U,C,G, and A correspond to the 5'- monophosphate forms of the ribonucleosides uridine, cytidine, guanosine, and adenosine, respectively when they occur in RNA molecules.
  • base pair may refer to a partnership of A with T or C with G.
  • heteroduplex base pair may refer to a partnership of A with U or C with G.
  • “Digestion” or “Restriction” of DNA refers to the catalytic cleavage of the DNA with a restriction enzyme that acts only at certain sequences in the DNA (“sequence- specific endonucleases”).
  • the various restriction enzymes used herein are commercially available and their reaction conditions, cofactors, and other requirements were used as would be known to one of ordinary skill in the art. Appropriate buffers and substrate amounts for particular restriction enzymes are specified by the manufacturer or can be readily found in the literature.
  • Ligation refers to the process of forming phosphodiester bonds between, two double stranded nucleic acid fragments. Unless otherwise provided, ligation may be accomplished using known buffers and conditions with a DNA ligase, such as T4 DNA ligase.
  • Plasmid refers to an extrachromosomal (usually) self-replicating genetic element.
  • Recombinant DNA cloning vector refers to any autonomously replicating agent, including, but not limited to, plasmids and phages, comprising a DNA molecule to which one or more additional DNA segments can or have been added.
  • Recombinant DNA expression vector refers to any recombinant DNA cloning vector in which a promoter to control transcription of the inserted DNA has been incorporated.
  • Transcription refers to the process whereby information contained in a nucleotide sequence of DNA is transferred to a complementary RNA sequence.
  • Transfection refers to the uptake of an expression vector by a host cell whether or not any coding sequences are, in fact, expressed. Numerous methods of transfection are known to the ordinarily skilled artisan, for example, calcium phosphate co- precipitation, liposome transfection, and electroporation. Successful transfection is generally recognized when any indication of the operation of this vector occurs within the host cell.
  • Transformation refers to the introduction of DNA into an organism so that the
  • DNA is replicable, either as an extrachromosomal element or by chromosomal integration.
  • Methods of transforming bacterial and eukaryotic hosts are well known in the art, many of which methods, such as nuclear injection, protoplast fusion or by calcium treatment using calcium chloride are summarized in J. Sambrook, et al, Molecular Cloning: A Laboratory Manual, (1989).
  • transformation refers to the process whereby the genetic information of messenger RNA (mRNA) is used to specify and direct the synthesis of a polypeptide chain.
  • Vector refers to a nucleic acid compound used for the transfection and/or transformation of cells in gene manipulation bearing polynucleotide sequences corresponding to appropriate protein molecules which, when combined with appropriate control sequences, confers specific properties on the host cell to be transfected and/or transformed. Plasmids, viruses, and bacteriophage are suitable vectors. Artificial vectors are constructed by cutting and joining DNA molecules from different sources using restriction enzymes and ligases.
  • the term "vector” as used herein includes Recombinant DNA cloning vectors and Recombinant DNA expression vectors.
  • “Primer” refers to a nucleic acid fragment which functions as an initiating substrate for enzymatic or synthetic elongation.
  • “Promoter” refers to a DNA sequence which directs transcription of DNA to RNA.
  • “Probe” refers to a nucleic acid compound or a fragment, thereof, which hybridizes with another nucleic acid compound.
  • Leader sequence refers to a sequence of amino acids which can be enzymatically or chemically removed to produce the desired polypeptide of interest.
  • “Secretion signal sequence” refers to a sequence of amino acids generally present at the N-terminal region of a larger polypeptide functioning to initiate association of that polypeptide with the cell membrane compartments like endoplasmic reticulum and secretion of that polypeptide through the plasma membrane.
  • Wild-type human IgG4 proteins can be obtained from a variety of sources. For example, these proteins can be obtained from a cDNA library prepared from cells which express the mRNA of interest at a detectable level. Libraries can be screened with probes designed using the published DNA or protein sequence for the particular protein of interest.
  • immunoglobulin light or heavy chain constant regions are described in Adams, et al. (1980) Biochemistry 19:2711-2719; Goughet, et al. (1980) Biochemistry 19:2702-2710; Dolby, et al. (1980) Proc. Natl. Acad. Sci. USA 77:6027- 6031; Rice et al. (1982) Proc. Natl. Acad. Sci. USA 79:7862-7862; Falkner, et al. (1982) Nature 298:286-288; and Morrison, et al. (1984) Ann. Rev. Immunol. 2:239-256.
  • PCR primers can be designed based on published sequences.
  • the full-length wild-type sequences cloned from a particular library can serve as a template to create the IgG4 Fc analog fragments of the present invention that retain the ability to confer a longer plasma half -life on the GLP-1 analog that is part of the fusion protein.
  • the IgG4 Fc analog fragments can be generated using PCR techniques with primers designed to hybridize to sequences corresponding to the desired ends of the fragment. PCR primers can also be designed to create restriction enzyme sites to facilitate cloning into expression vectors.
  • DNA encoding the GLP-1 analogs of the present invention can be made by a variety of different methods including cloning methods like those described above as well as chemically synthesized DNA.
  • the gene encoding a fusion protein can then be constructed by ligating DNA encoding a GLP-1 analog in-frame to DNA encoding the IgG Fc proteins described herein.
  • the DNA encoding wild-type GLP-1 and IgG4 Fc fragments can be mutated either before ligation or in the context of a cDNA encoding an entire fusion protein. A variety of mutagenesis techniques are well known in the art.
  • the gene encoding the GLP- 1 analog and the gene encoding the IgG4 Fc analog protein can also be joined in-frame via DNA encoding a G-rich linker peptide.
  • a preferred DNA sequence encoding one of the preferred heterologous fusion proteins of the present invention, Gly 8 -Glu 2 -Gly 36 - GLP-l(7-37)-lL-IgG4 (S228P, F234A, L235A, des K), is provided as SEQ ID NO:20: CACGGCGAGGGCACCTTCACCTCCGACGTGTCCTCCTATCTCGAGGAGCAGG CCGCCAAGGAATTCATCGCCTGGCTGGTGAAGGGCGGCGGCGGTGGTGGTGG CTCCGGAGGCGGCGGCTCTGGTGGCGGTGGCAGCGCTGAGTCCAAATATGGT CCCCCATGCCCACCCTGCCCAGCACCTGAGGCCGCCGGGGGACCATCAGTCTT CCTGTTCCCCCCAAAACCCAAGGACACTCTCATGATCTCCCGGACCCCTGAGG TCACGTGCGTGGTGGTGGACGTGAGCCAGGAAGACCCCGAGGTCCAGTTCAA CTGGTACGTGGATGGCGTGGAGGTGCATAATGC
  • the culture conditions can be selected by the skilled artisan without undue experimentation.
  • principles, protocols, and practical techniques for maximizing the productivity of cell cultures can be found in Mammalian Cell Biotechnology: A Practical Approach, M. Butler, ed. (IRL Press, 1991) and Sambrook, et al, supra. Methods of transfection are known to the ordinarily skilled artisan, for example, CaPO 4 and electroporation.
  • General aspects of mammalian cell host system transformations have been described in U.S. Patent No. 4,399,216. Transformations into yeast are typically carried out according to the method of van Solingen et al., J Bact.
  • Suitable host cells for cloning or expressing the nucleic acid (e.g., DNA) in the vectors herein include yeast or higher eukaryote cells.
  • Eukaryotic microbes such as filamentous fungi or yeast are suitable cloning or expression hosts for fusion protein vectors.
  • Saccharomyces cerevisiae is a commonly used lower eukaryotic host microorganism.
  • Others include Schizosaccharomyces pombe [Beach and Nurse, Nature 290: 140-3 (1981); EP 139,383 published 2 May 1995]; Muyveromyces hosts [U.S. Patent No.
  • Methylotropic yeasts are selected from the genera consisting of
  • Suitable host cells for the expression of the fusion proteins of the present invention are derived from multicellular organisms.
  • invertebrate cells include insect cells such as Drosophila S2 and Spodoptera Sp, Spodoptera high5 as well as plant cells.
  • useful mammalian host cell lines include NSO myeloma cells, Chinese hamster ovary (CHO), SP2, and COS cells.
  • monkey kidney CV1 line transformed by SV40 (COS-7, ATCC CRL 1651); human embryonic kidney line [293 or 293 cells subcloned for growth in suspension culture, Graham, et al, J. Gen Virol, 36(1): 59-74 (1977)]; Chinese hamster ovary cells/-DHFR [CHO, Urlaub and Chasin, Proc. Natl. Acad. Sci. USA, 77(7): 4216-20 (1980)]; mouse sertoli cells [TM4, Mather, Biol. Reprod. 23(l):243-52 (1980)]; human lung cells (W138.
  • COS-7 monkey kidney CV1 line transformed by SV40
  • human embryonic kidney line [293 or 293 cells subcloned for growth in suspension culture, Graham, et al, J. Gen Virol, 36(1): 59-74 (1977)]
  • Chinese hamster ovary cells/-DHFR [CHO, Urlaub and Chasin, Proc. Natl. Acad. Sci. USA
  • a preferred cell line for production of the Fc fusion proteins of the present invention is the NSO myeloma cell line available from the European Collection of Cell Cultures (ECACC, catalog #85110503) and described in Galfre, G. and Milstein, C. ((1981) Methods in Enzymology 73(13):3-46; and Preparation of Monoclonal Antibodies: Strategies and Procedures, Academic Press, ⁇ .Y.,
  • the fusion proteins of the present invention may be recombinantly produced directly, or as a protein having a signal sequence or other additional sequences which create a specific cleavage site at the N-terminus of the mature fusion protein.
  • the signal sequence may be a component of the vector, or it may be a part of the fusion protein-encoding DNA that is inserted into the vector.
  • the signal sequence may be, e.g., the yeast invertase leader, alpha factor leader (including Saccharomyces and Kluyveromyces cc-factor leaders, the latter described in U.S. Patent No. 5,010,182), or acid phosphatase leader, the C.
  • mammalian signal sequences may be used to direct secretion of the protein, such as signal sequences from secreted polypeptides of the same or related species as well as viral secretory leaders.
  • Both expression and cloning vectors contain a nucleic acid sequence that enables the vector to replicate in one or more selected host cells. Expression and cloning vectors will typically contain a selection gene, also termed a selectable marker.
  • Typical selection genes encode proteins that (a) confer resistance to antibiotics or other toxins, e.g., neomycin, methotrexate, or tetracycline, (b) complement autotrophic deficiencies, or (c) supply critical nutrients not available from complex media, e.g., the gene encoding D-alanine racemase for Bacilli.
  • An example of suitable selectable markers for mammalian cells are those that enable the identification of cells competent to take up the fusion protein-encoding nucleic acid, such as DHFR or thymidine kinase.
  • An appropriate host cell when wild-type DHFR is employed is the CHO cell line deficient in DHFR activity, prepared and propagated as described [Urlaub and Chasin, Proc.
  • a suitable selection gene for use in yeast is the trpl gene present in the yeast plasmid YRp7 [Stinchcomb, et al, Nature 282(5734): 39-43 (1979); Kingsman, et al, Gene 7(2): 141-52 (1979); Tschumper, et al, Gene 10(2): 157-66 (1980)].
  • the trpl gene provides a selection marker for a mutant strain of yeast lacking the ability to grow in tryptophan, for example, ATCC No. 44076 or PEPC1 [Jones, Genetics 85: 23-33 (1977)].
  • Expression and cloning vectors usually contain a promoter operably linked to the fusion protein-encoding nucleic acid sequence to direct mRNA synthesis. Promoters recognized by a variety of potential host cells are well known. Examples of suitable promoting sequences for use with yeast hosts include the promoters for 3-phosphoglycerate kinase [Hitzeman, et al, J. Biol Chem. 255(24): 12073-80 (1980)] or other glycolytic enzymes [Hess et al., J. Adv. Enzyme Reg.
  • yeast promoters which are inducible promoters having the additional advantage of transcription controlled by growth conditions, are the promoter regions for alcohol dehydrogenase 2, isocytochrome C, acid phosphatase, degradative enzymes associated with nitrogen metabolism, metallothionein, gl ceraldehyde-3 -phosphate dehydrogenase, and enzymes responsible for maltose and galactose utilization. Suitable vectors and promoters for use in yeast expression are further described in EP 73,657.
  • Transcription of fusion protein-encoding mRNA from vectors in mammalian host cells may be controlled, for example, by promoters obtained from the genomes of viruses such as polyoma virus, fowlpox virus, adenovirus (such as Adeno virus 2), bovine papilloma virus, avian sarcoma virus, cytomegalovirus, a retrovirus, hepatitis-B virus and Simian Virus 40 (SV40), from heterologous mammalian promoters, e.g., the actin promoter or an immunoglobulin promoter, and from heat-shock promoters, provided such promoters are compatible with the host cell systems.
  • viruses such as polyoma virus, fowlpox virus, adenovirus (such as Adeno virus 2), bovine papilloma virus, avian sarcoma virus, cytomegalovirus, a retrovirus, hepatitis-B virus and Simian
  • Enhancers are cis-acting elements of DNA, usually about from 10 to 300 bp, that act on a promoter to increase its transcription.
  • Many enhancer sequences are now known from mammalian genes (globin, elastase, albumin, a-ketoprotein, and insulin). Typically, however, one will use an enhancer from a eukaryotic cell virus.
  • Examples include the SV40 enhancer on the late side of the replication origin (bp 100-270), the cytomegalovirus early promoter enhancer, the polyoma enhancer on the late side of the replication origin, and adenovirus enhancers.
  • the enhancer may be spliced into the vector at a position 5' or 3' to the fusion protein coding sequence but is preferably located at a site 5' from the promoter.
  • Expression vectors used in eukaryotic host cells yeast, fungi, insect, plant, animal, human, or nucleated cells from other multicellular organisms
  • Such sequences are commonly available from the 5' and occasionally 3' untranslated regions of eukaryotic or viral DNAs or cDNAs. These regions contain nucleotide segments transcribed as polyadenylated fragments in the untranslated portion of the mRNA encoding the fusion protein.
  • Various forms of a fusion protein may be recovered from culture medium or from host cell lysates. If membrane-bound, it can be released from the membrane using a suitable detergent solution (e.g., Triton-X 100) or by enzymatic cleavage.
  • Cells employed in expression of a fusion protein can be disrupted by various physical or chemical means, such as freeze- thaw cycling, sonication, mechanical disruption, or cell lysing agents.
  • the analogs can be isolated and purified.
  • the following procedures are exemplary of suitable purification procedures: fractionation on carboxymethyl cellulose; gel filtration such as Sephadex G-75; anion exchange resin such as DEAE or Mono-Q; cation exchange such as CM or Mono-S; metal chelating columns to bind epitope-tagged forms of the polypeptide; reversed-phase HPLC; chromatofocusing; silica gel; ethanol precipitation; and ammonium sulfate precipitation.
  • fusion proteins comprising an Fc fragment can be effectively purified using a Protein A or Protein G affinity matrix.
  • Low or high pH buffers can be used to elute the fusion protein from the affinity matrix. Mild elution conditions will aid in preventing irreversible denaturation of the fusion protein.
  • the heterologous fusion proteins of the present invention may be formulated with one or more excipients.
  • the fusion proteins of the present invention may be combined with a pharmaceutically acceptable buffer, and the pH adjusted to provide acceptable stability, and a pH acceptable for administration such as parenteral administration.
  • a pharmaceutically acceptable buffer such as a pharmaceutically acceptable buffer
  • one or more pharmaceutically-acceptable anti-microbial agents may be added. Meta-cresol and phenol are preferred pharmaceutically-acceptable microbial agents.
  • One or more pharmaceutically- acceptable salts may be added to adjust the ionic strength or tonicity.
  • One or more excipients may be added to further adjust the isotonicity of the formulation. Glycerin is an example of an isotonicity-adjusting excipient.
  • Pharmaceutically acceptable means suitable for administration to a human or other animal and thus, does not contain toxic elements or undesirable contaminants and does not interfere with the activity of the active compounds therein.
  • the heterologous fusion proteins of the present invention may be formulated as a solution formulation or as a lyophilized powder that can be reconstituted with an appropriate diluent.
  • a lyophilized dosage form is one in which the fusion protein is stable, with or without buffering capacity to maintain the pH of the solution over the intended in-use shelf- life of the reconstituted product. It is preferable that the solution comprising the heterologous fusion proteins discussed herein before lyphilization be substantially isotonic to enable formation of isotonic solutions after reconstitution.
  • a pharmaceutically-acceptable salt form of the heterologous fusion proteins of the present invention are within the scope of the invention.
  • Acids commonly employed to form acid addition salts are inorganic acids such as hydrochloric acid, hydrobromic acid, hydriodic acid, sulfuric acid, phosphoric acid, and the like, and organic acids such as -toluenesulfonic acid, methanesulfonic acid, oxalic acid, -bromophenyl-sulfonic acid, carbonic acid, succinic acid, citric acid, benzoic acid, acetic acid, and the like.
  • Preferred acid addition salts are those formed with mineral acids such as hydrochloric acid and hydrobromic acid.
  • Base addition salts include those derived from inorganic bases, such as ammonium or alkali or alkaline earth metal hydroxides, carbonates, bicarbonates, and the like. Such bases useful in preparing the salts of this invention thus include sodium hydroxide, potassium hydroxide, ammonium hydroxide, potassium carbonate, and the like.
  • the heterologous fusion proteins of the present invention have biological activity. Biological activity refers to the ability of the fusion protein to bind to and activate the GLP-1 receptor in vivo and elicit a response.
  • GLP-1 fusion proteins were tested for in vitro as well as in vivo activity. Examples 1 and 2 provide in vitro activity based on the ability of the fusion protein to interact with and activate the human GLP-1 receptor. In both sets of experiments, HEK293 cells over-expressing the human GLP-1 receptor were used.
  • Example 1 (table 1) provides data wherein the reporter gene is beta lactamase, and example 2 (table 2) provides data wherein the reporter gene is luciferase.
  • Example 3 provides data generated after administration of one of the heterologous fusion proteins of the present invention to rats. Together the data show that the fusion proteins are able to bind to and activate the GLP-1 receptor and appear more potent in vitro than Val 8 -GLP-l(7-37)OH.
  • the data generated in rats indicate the fusion proteins are active in vivo and have a longer half-life than native GLP-1.
  • Administration of the heterogeneous fusion proteins may be via any route known to be effective by the physician of ordinary skill.
  • Peripheral parenteral is one such method. Parenteral administration is commonly understood in the medical literature as the injection of a dosage form into the body by a sterile syringe or some other mechanical device such as an infusion pump. Peripheral parenteral routes can include intravenous, intramuscular, subcutaneous, and intraperitoneal routes of administration.
  • the heterologous fusion proteins of the present invention may also be amenable to administration by oral, rectal, nasal, or lower respiratory routes, which are non-parenteral routes.
  • the fusion proteins of the present invention can be used to treat a wide variety of diseases and conditions.
  • the fusion proteins of the present invention primarily exert their biological effects by acting at a receptor referred to as the "GLP-1 receptor.”
  • Subjects with diseases and/or conditions that respond favorably to GLP-1 receptor stimulation or to the administration of GLP-1 compounds can therefore be treated with the GLP-1 fusion proteins of the present invention. These subjects are said to "be in need of treatment with GLP-1 compounds" or "in need of GLP-1 receptor stimulation".
  • Subjects with impaired glucose tolerance or impaired fasting glucose subjects whose body weight is about 25% above normal body weight for the subject's height and body build, subjects with a partial pancreatectomy, subjects having one or more parents with non-insulin dependent diabetes, subjects who have had gestational diabetes and subjects who have had acute or chronic pancreatitis are at risk for developing non-insulin dependent diabetes.
  • An effective amount of the GLP-1 -Fc fusion proteins described herein is the quantity which results in a desired therapeutic and/or prophylactic effect without causing unacceptable side-effects when administered to a subject in need of GLP-1 receptor stimulation.
  • a “desired therapeutic effect” includes one or more of the following: 1) an amelioration of the symptom(s) associated with the disease or condition; 2) a delay in the onset of symptoms associated with the disease or condition; 3) increased longevity compared with the absence of the treatment; and 4) greater quality of life compared with the absence of the treatment.
  • an "effective amount" of a GLP-l-Fc fusion protein for the treatment of diabetes is the quantity that would result in greater control of blood glucose concentration than in the absence of treatment, thereby resulting in a delay in the onset of diabetic complications such as retinopathy, neuropathy or kidney disease.
  • an "effective amount" of a GLP-l-Fc fusion protein for the prevention of diabetes is the quantity that would delay, compared with the absence of treatment, the onset of elevated blood glucose levels that require treatment with anti-hypoglycaemic drugs such as sulfonyl ureas, thiazolidinediones, insulin and/or bisguanidines.
  • the dose of fusion protein effective to normalize a patient's blood glucose will depend on a number of factors, among which are included, without limitation, the subject's sex, weight and age, the severity of inability to regulate blood glucose, the route of administration and bioavailability, the pharmacokinetic profile of the fusion protein, the potency, and the formulation.
  • Doses may be in the range of 0.01 to 1 mg/kg body weight, preferably in the range of 0.05 to 0.5 mg/kg body weight. It is preferable that the fusion proteins of the present invention be administered either once every two weeks or once a week. Depending on the disease being treated, it may be necessary to administer the fusion protein more frequently such as two to three time per week.
  • the present invention will now be described only by way of non-limiting example with reference to the following Examples.
  • Example 1 In vitro GLP-1 receptor activation assay HEK-293 cells expressing the human GLP-1 receptor, using a CRE-BLAM system, are seeded at 20,000 to 40,000 cells/well/ 100 ⁇ l DMEM medium with 10%FBS into a poly-d-lysine coated 96 well black, clear-bottom plate. The day after seeding, the medium is flicked off and 80 ⁇ l plasma-free DMEM medium is added. On the third day after seeding, 20 ⁇ l of plasma-free DMEM medium with 0.5% BSA containing different concentrations of various GLP-l-Fc heterologous fusion protein is added to each well to generate a dose response curve.
  • Gly 8 -Glu 22 -GLP-l(7-37)-1.5L-IgG4 S228P, F234A, L235A: 314% 45
  • Gly 8 -Glu 22 -GLP-l(7-37)-lL-IgG4 S228P, F234A, L235A: 468% 120
  • Example 2 In vitro GLP-1 receptor activation assay HEK-293 cells stably expressing the human GLP-1 receptor, using a CRE- Luciferase system, are seeded at 30,000 cells/well/80 ⁇ l low serum DMEM F12 medium into 96 well plates. The day after seeding, 20 ⁇ l aliquots of test protein dissolved in 0.5% BSA are mixed and incubated with the cells for 5 hours. Generally 12 dilutions containing from 3 pM to 3 nM are prepared at a 5X concentration for each test protein before addition to the cells to generate a dose response curve from which EC 50 values are determined.
  • Gly 8 -Glu 22 -Gly 36 -GLP-l(7-37)-2L-IgG4 (S228P, F234A, L235A): 398% 62
  • Gly 8 -Glu 22 -Gly 36 -GLP-l(7-37)-lL-IgG4 (S228P, F234A, L235A): 417% 140
  • Group I receives vehicle (table 3)
  • Group II receives 1.79 mg/kg of Gly 8 -Glu 22 -Gly 36 -GLP-l(7-37)-L-IgG4 (S228P,F234A,L235A) as a single subcutaneous injection (table 4)
  • Group III receives 0.179 mg/kg of Gly 8 -Glu 22 -Gly 36 -GLP-l(7-37)-L-IgG4 (S228P,F234A,L235A) as a single subcutaneous injection (table 5).
  • Rats are subcutaneously injected the morning of Day 1. Twenty-four hours following the first injection, 1 ⁇ L of glucose (D50) per gram rat body weight is infused as a bolus. Blood samples are taken at 2, 4, 6, 10, 20, and 30 minutes following the bolus infusion of glucose.
  • D50 glucose
  • Insulin AUC (ng*min/mL) Average SEM 0-2 11 9.4 7 11 9.6 2-4 18.1 9.7 5.6 10.6 8.8 4-6 13.4 7 3.4 9.6 5.9 6-10 7.9 3.5 2.5 6 2.9 10-20 3.7 3 2.4 3 2.4 20-30 2 0 0 0 2.4 sum 56.1 32.6 20.9 40.2 32 36.4 5.8
  • Example 4 Pharmacokinetic Study Following a Single Subcutaneous Injection to Cynomolgus Monkeys.
  • a study is performed to characterize the pharmacokinetics (PK) of the Fc fusion protein, Gly 8 -Glu 22 -Gly 36 -GLP-l(7-37)-L-IgG4 (S228P,F234A,L235A), when administered as a 0.1 mg/kg by subcutaneous (SC) injection to male cynomolgus monkeys.
  • RIA antibody is specific for the middle portion of GLP.
  • ELISA uses an N- terminus specific capture antibody and an Fc specific detection antibody. Resulting plasma concentrations from both the ELISA and the RIA are used to determine the represented pharmacokinetic parameter values.
  • a representation of the resulting PK parameter values is summarized in table 6.
  • Single-dose SC PK from the RIA is associated with a mean C max of 446.7 ng/mL with a corresponding T max of 17.3 hours.
  • the mean elimination half-life is approximately 79.3 hours (3.3 days).
  • the PK from the ELISA is associated with a mean C max of 292.2 ng/mL with a corresponding T max of 16J hours.
  • the mean elimination half-life is approximately 51.6 hours (2.2 days).
  • Microtiter plates are coated with Gly 8 -Glu 22 -Gly 36 -GLP- l(7-37)-L-IgG4 (S228P,F234A,L235A) at a 0.1 ⁇ g/mL concentration.
  • Monkey serum samples are diluted 50, 500,1000 and 5000 fold into blocking solution, and 0.05 mL sample/well are incubated approximately one hour.
  • Secondary antibody, Goat ⁇ Human Fab'2>-Peroxidase (with 75% cross reactivity to human), is diluted 10,000 fold into block and added at 0.05 mL/well and incubated approximately one hour.
  • TMB tetramethylbenzidine
  • a GLP-1 antibody was used as a positive control and goat ⁇ rabbit>(H+L)-Peroxidase conjugate is the secondary used for detection.
  • Point serum samples are collected prior to dosing, at 24 hours following the second dose, and 168 hours following the first and second SC dose for an evaluation of potential immunogenicity.
  • the presence of antibody titers to G8E22-CEX-L-hIgG4 is interpreted by comparison to predose serum samples and positive control. A representation of the results is presented in table 7.
  • Example 6 Pharmacodynamic Study Following a Single Subcutaneously Injection to Cynomolgus Monkevs.in the Fasting State and During a Graded Intravenous Glucose Infusion.
  • Phase 1 a subcutaneous injection of vehicle is administered.
  • a graded intravenous glucose (20% dextrose) infusion of 5, 10, and 25 mg/kg/min is then administered immediately after the vehicle injection.
  • Phase 2 a subcutaneous injection of a GLP-1 fusion protein (0.1 mg/kg) is administered.
  • Phase 3 a graded intravenous glucose infusion is performed approximately 96 hours following the GLP-1 fusion injection.
  • Graded intravenous glucose infusion procedures are conducted in sedated monkeys after a 16-hr overnight fast.
  • baseline samples will be drawn every 10 min for 20 min to define baseline.
  • a stepped-up glucose infusion is initiated at +20 min at a rate of 5 mg kg/min, followed by infusions of 10 mg/kg/min, and 25 mg/kg/min. Each infusion rate is administered for a period of 20 minutes.
  • Blood samples are taken at 10 minute intervals for measurement of glucose, insulin, and glucagon. Approximately 1.0 mL of blood is collected at -20, -10 min, 0 pre- glucose infusions, and at 10, 20, 30, 40, 50, and 60 minutes following glucose infusion for Phases 1 and 3. A representation of the data are shown in table 8.
  • Glucagon levels were not statistically different between the vehicle and the GLP-1 fusion protein dosed monkeys.
  • Example 7 Pharmacodynamic Study Following Single Subcutaneously Injections of Three Different Doses to Rats in the Fasting State and During a Graded Intravenous Glucose Infusion.
  • Chronically cannulated rats are assigned to either vehicle control (saline) or one of 3 treatment groups (GLP-1 fusion protein; 0.0179 mg/kg, 0.179 mg/kg, or 1.79 mg/kg).
  • the GLP-1 fusion protein and vehicle are administered via subcutaneous injection.
  • Twenty-four hours after treatment, overnight fasted (16h) rats are subjected to a graded intravenous glucose infusion test.
  • the graded glucose infusion test consists of a baseline saline infusion period (20 min), followed by two 30 min glucose infusion phases at 5 and 15 mg/kg/min, respectively.
  • Plasma samples are collected at -20, -10 min, 0 pre-glucose infusions (baseline), and at 10, 20, 30, 40, 50, and 60 minutes.
  • a representation of the data are shown in table 9.

Abstract

The invention provides specific GLP-1 analogs fused to specific IgG4-Fc derivatives. These fusion proteins have an increased half-life, decreased immunogenicity, and reduce effector activity. The fusion proteins are useful in treating diabetes, obesity, irritable bowel syndrome and other conditions that would be benefited by lowering plasma glucose, inhibiting gastric and/or intestinal motility and inhibiting gastric and/or intestinal emptying, or inhibiting food intake.

Description

GLP-1 ANALOG FUSION PROTEINS
FIELD OF THE INVENTION The present invention relates to glucagon-like peptide analogs fused to proteins that have the effect of extending the in vivo half-life of the peptides. These fusion proteins can be used to treat diabetes as well as a variety of other conditions or disorders. Glucagon-like peptide- 1 (GLP-1) analogs and derivatives show promise in clinical trials for the treatment of type 2 diabetes. GLP-1 induces numerous biological effects such as stimulating insulin secretion, inhibiting glucagon secretion, inhibiting gastric emptying, inhibiting gastric motility or intestinal motility, and inducing weight loss. A significant characteristic of GLP-1 is its ability to stimulate insulin secretion without the associated risk of hypoglycemia that is seen when using insulin therapy or some types of oral therapies that act by increasing insulin expression. The usefulness of therapy involving GLP-1 peptides has been limited by the fact that GLP-l(l-37) is poorly active, and the two naturally occurring truncated peptides, GLP-l(7-37)OH and GLP-1(7-36)NH2, are rapidly cleared in vivo and have extremely short in vivo half lives. It is known that endogenously produced dipeptidyl-peptidase IN (DPP-IV) inactivates circulating GLP-1 peptides by removing the Ν- terminal histidine and alanine residues and is a major reason for the short in vivo half-life. Various approaches have been undertaken to extend the elimination half-life of a
GLP-1 peptide or reduce clearance of the peptide from the body while maintaining biological activity. One approach involves fusing a GLP-1 peptide to the Fc portion of an immunoglobulin. Immunoglobulins typically have long circulating half-lives in vivo. For example, IgG molecules can have a half-life in humans of up to 23 days. The Fc portion of the immunoglobulin is responsible, in part, for this in vivo stability. GLP-1 -Fc fusion proteins take advantage of the stability provided by the Fc portion of an immunoglobulin while preserving the biological activity of the GLP-1 molecule. Although this approach is feasible for QLP-1 therapeutics (See WO 02/46227), there is a general concern regarding the antigenicity of various fusion proteins when administered repeatedly over prolonged periods of time. This is especially a concern for GLP-1 -Fc fusion therapeutics as a patient with diabetes must be treated for her entire life once diagnosed with the disease. In addition, Fc fusion protein therapeutics can be a concern if the Fc portion retains unwanted effector functions. The present invention seeks to overcome the problems associated with the potential immunogenicity and effector activity associated with administration of GLP-1 - Fc fusions by identifying specific GLP-1 -Fc fusion proteins that have a reduced risk of inducing an immune response after repeated and prolonged administration and no longer have effector function. These specific fusion proteins have substitutions at various positions in the GLP-1 portion as well as the Fc portion of the molecule. The substitutions described herein provide increased potency, increased in vivo stability, elimination of effector function and decreased likelihood the molecule will be recognized by the adaptive elements of the immune system. Compounds of the present invention include heterologous fusion proteins comprising a GLP-1 analog comprising a sequence selected from the group consisting of a) (SEQ LD NO:l) His-Xaa8-Glu-Gly-Thr-Phe-Thr-Ser-Asp~Val-Ser-Ser-Tyr-Leu-Glu-Glu- Gln-Ala-Ala-Lys-Glu-Phe-Ile-Ala-Trp-Leu-Val-Lys-Gly-Gly-Gly wherein Xaa8 is selected from Gly and Val; b) (SEQ ID ΝO:2) His-Xaa8-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Val-Ser-Ser-Tyr-Leu-Glu-Glu- Gln-Ala-Ala-Lys-Glu-Phe-Ile-Ala-Trp-Leu-Lys-Asn-Gly-Gly-Gly wherein Xaa8 is selected from Gly and Val; c) (SEQ ID NO:3) His-Xaa8-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Val-Ser-Ser-Tyr-Leu-Glu-Glu- Gln-Ala-Ala-Lys-Glu-Phe-Ile-Ala-Trp-Leu-Val-Lys-Gly-Gly-Pro wherein Xaa8 is selected from Gly and Val; d) (SEQ ID NO:4) His-Xaa8-Glu-Gly-Thr-Phe-Thr-Ser-Asρ-Val-Ser-Ser-Tyr-Leu-Glu-Glu- Gln- Ala-Ala-Lys-Glu-Phe-Ile-Ala-Trp-Leu-Lys-Asn-Gly-Gly-Pro wherein Xaa8 is selected from Gly and Val; e) (SEQ ID NO:5) His-Xaa8-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Val-Ser-Ser-Tyr-Leu-Glu-Glu- Gln-Ala-Ala-Lys-Glu-Phe-Ile-Ala-Trp-Leu-Val-Lys-Gly-Gly wherein Xaa8 is selected from Gly and Val; f) (SEQ ID NO:6) His-Xaa8-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Val-Ser-Ser-Tyr-Leu-Glu-Glu- Gln-Ala-Ala-Lys-Glu-Phe-Ile-Ala-Trp-Leu-Lys-Asn-Gly-Gly wherein Xaa8 is selected from Gly and Val; fused to the Fc portion of an immunoglobulin comprising the sequence of SEQ ID NO:7 Ala-Glu-Ser-Lys-Tyr-Gly-Pro-Pro-Cys-Pro-Pro-Cys-Pro-Ala-Pro- Xaa16-Xaaι7-Xaaι8-Gly-Gly-PiO-Ser-Val-Phe-Leu-Phe-Pro-Pro-Lys-Pro- Lys-Asp-Thι--Leu-Met-Ile-Ser-Arg-Thr-Pro-Glu-Val-Thr-Cys-Val- Val-Val-Asp-Val-Ser-Gln-Glu-Asp-Pro-GIu-Val-Gln-Phe-Asn-Trp- Tyr-Val-Asp-Gly-Val-Glu-Val-His-Asn-Ala-Lys-Thr-Lys-Pro-Arg- Glu-Glu-Gln-Phe-Xaa80-Ser-Thr-Tyr-Arg-Val-Val-Ser-Val-Leu-Thr- Val-Leu-His-Gln-Asp-Trp-Leu-Asn-Gly-Lys-Glu-Tyr-Lys-Cys-Lys- Val-Ser-Asn-Lys-Gly-Leu-Pro-Ser-Ser-Ile-Glu-Lys-Thr-Ile-Ser- Lys-Ala-Lys-Gly-Gln-Pro-Arg-Glu-Pro-Gln-Val-Tyr-Thr-Leu-Pro- Pro-Ser-Gln-Glu-Glu-Met-Thr-Lys-Asn-Gln-Val-Ser-Leu-Thr-Cys- Leu-Val-Lys-Gly-Phe-Tyr-Pro-Ser-Asp-Ile-Ala-Val-Glu-Trp-Glu- Ser-Asn-Gly-Gln-Pro-Glu-Asn-Asn-Tyr-Lys-Thr-Thr-Pro-Pro-Val- Leu-Asp-Ser-Asp-Gly-Ser-Phe-Phe-Leu-Tyr-Ser-Arg-Leu-Thr-Val- Asp-Lys-Ser-Arg-Trp-Gln-Glu-Gly-Asn-Val-Phe-Ser-Cys-Ser-Val- Met-His-Glu-Ala-L i-His-Asn-His-Tyr-Thr-Gln-Lys-Ser-Leu-Ser- Leu-Ser-Leu-Gly-Xaa23o (SEQ ID NO:7) wherein: Xaa at position 16 is Pro or Glu; Xaa at position 17 is Phe, Val, or Ala; Xaa at position 18 is Leu, Glu, or Ala; Xaa at position 80 is Asn or Ala; and Xaa at position 230 is Lys or is absent. The C- terminus of the GLP-1 analog portion and the N- terminus of the Fc portion of the heterologous fusion proteins of the present invention are preferably fused together via 1, 1.5 or 2 repeats of a G-rich peptide linker having the sequence Gly-Gly-Gly-Gly- Ser-Gly-Gly-Gly-Gly-Ser-Gly-Gly-Gly-Gly-Ser (SEQ ID NO: 8). The present invention also includes polynucleotides encoding the heterologous fusion proteins of the present invention, as well as vectors and host cells comprising such polynucleotides. Methods of treating patients suffering from non-insulin dependent as well as insulin dependent diabetes mellitus, obesity, and various other disorders and conditions comprising administering the heterologous fusion proteins discussed herein are also encompassed by the present invention. The heterologous fusion proteins of the present invention comprise a GLP-1 analog portion and an Fc portion. The GLP-1 analog portion and the Fc portion comprise substitutions to the native GLP-1 sequence and the human IgG4 sequence respectively that provide the protein with increased potency and in vivo stability compared to native GLP-1 or GLP-1 analogs not fused to an Fc sequence while decreasing the potential for inducing antibody formation after prolonged and repeated administration in humans. Native GLP-1 is processed in vivo such that the first 6 amino acids are cleaved from the molecule. Thus, by custom in the art, the amino terminus of GLP-1 has been assigned the number 7 and the carboxy- terminus, number 37. The other amino acids in the polypeptide are numbered consecutively as shown in SEQ ID NO:9. For example, position 8 is alanine and position 22 is glycine. The processed peptide may be further modified in vivo such that the C-terminal glycine residue is removed and replaced with an amide group. Thus, GLP-l(7-37)OH and GLP-l(7-36)amide represent the two native forms of the molecule. GLP-1 (7-37)OH has the amino acid sequence of SEQ ID NO:9:
1His-Ala-Glu-10Gly-Thr-Phe-Thr-Ser-15Asp-Val-Ser-Ser-Tyr-20Leu-Glu-Gly- GGlinn--AAllaa--2255AAllaa--LLys-Glu-Phe-Ile-30Ala-Trp-Leu-Val-Lys-35GIy-Arg-37Gly (SEQ ID NO:9)
The GLP-1 analog portion of the heterologous fusion protein comprises three primary substitutions at positions 8, 22, and 36 relative to native GLP-1(7- 37). The substitution at position 8 reduces the rate at which the endogenous enzyme dipeptidyl-peptidase IV (DPP-IV) inactivates the analog. DPP- IV cleaves native GLP-1 between the 2nd and 3rd amino acids (between position 8 and 9) and the resulting molecule is less active. Thus, the heterologous fusion proteins of the present invention are DPP- IV resistant. The substitution at position 22 reduces the potential of the molecule to aggregate and increases the potency of the molecule. The substitution at position 36 in the context of the analog with changes at 8 and 22 as well as in the context of the entire fusion protein reduces the risk that the fusion protein will induce a neutralizing immune response after repeated and prolonged administration in humans. The central event in the generation of both humoral and cell-mediated immune responses is the activation and clonal expansion of T-helper (TH) cells. TH cell activation is initiated by interaction of the T-cell receptor (TCR)-CD3 complex with a processed antigenic peptide bound to a class II major histocompatibility (MHC) molecule in the presence of an antigen-presenting cell (APC). Interaction of a TH cell with antigen initiates a cascade of biochemical events that induces the resting TH cell to enter the cell cycle (Go to Gi transition). The activated T cell progresses through the cell cycle, proliferating and differentiating into memory cells or effector cells. The following sequence was analyzed to identify potential epitopes: His-Gly-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Val-Ser-Ser-Tyr-Leu-Glu-Glu-Gln-Ala- Ala-Lys-Glu-Phe-Ile-Ala-Trp-Leu-Val-Lys-Gly-Arg-Gly-Gly-Gly-Gly-Gly-Ser-Gly-Gly- Gly-Gly-Ser-Gly-Gly-Gly-Gly-Ser-Gly-Gly-Gly-Gly-Ser- Gly-Gly-Gly-Gly-Ser-Gly- Gly-Gly-Gly-Ser-Ala-Glu-Ser-Lys-Tyr-Gly-Pro-Pro-Cys-Pro (SEQ ID NO: 10) This sequence is a GLP-1 analog sequence with changes at positions 8 and 22 relative to the native sequence followed by 2 copies of a G-rich linker sequence followed by the first 10 amino acids of an Fc region derived from human IgG4. Epitope as used herein refers to a region of a protein molecule to which an antibody can bind. An immunogenic epitope is defined as the part of the protein that elicits an antibody response when the whole protein is the immunogen. Epitope mapping involved the scanning of sequences using a sliding nine amino acid window coupled with advanced statistical analysis techniques to extract the information contained in these patterns. A proprietary software package known as EpiMatrix™ was used to analyze the sequence and identify peptides that are highly likely to provoke an immune response when presented to T-cells. Eight highly common alleles were used in the analysis for Class II MHC receptor interaction. These alleles included DRB1*0101, DRB1*0301, DRB1*0401, DRB1*0701, DRB1*0801, DRB 1101, DRBP1301, and DRB1*1501. A strong epitope was predicted to be located at the junction of the C- terminus of the GLP-1 analog portion and the beginning of the linker. The sequence of this epitope is Trp-Leu-Val-Lys-Gly-Arg-Gly-Gly-Gly (SEQ ID NO: 11) which interacts with DRB 1 *0801. The present invention encompasses the discovery that this epitope can be eliminated by changing the GLP-1 analog C-terminus to one of the following sequences: Trp-Leu-Val-Lys-Gly-Gly-Gly (SEQ ID NO: 12); Trp-Leu-Lys-Asn-Gly-Gly-Gly (SEQ ID NO: 13); Trp-Leu- Val-Lys-Gly-Gly-Pro (SEQ ID NO: 14); Trp-Leu-Lys-Asn-Gly-Gly-Pro (SEQ ID NO: 15); Trp-Leu-Val-Lys-Gly-Gly (SEQ ID NO: 16); and Trp-Leu-Lys-Asn- Gly-Gly (SEQ ID NO:17). The heterologous fusion proteins of the present invention contain an Fc portion which is derived from human IgG4, but comprises one or more substitutions compared to the wild-type human sequence. As used herein, the Fc portion of an immunoglobulin has the meaning commonly given to the term in the field of immunology. Specifically, this term refers to an antibody fragment which does not contain the two antigen binding regions (the Fab fragments) from the antibody. The Fc portion consists of the constant region of an antibody from both heavy chains, which associate through non-covalent interactions and disulfide bonds. The Fc portion can include the hinge regions and extend through the CH2 and CH3 domains to the c-terminus of the antibody. The Fc portion can further include one or more glycosylation sites. There are five types of human immunoglobulins with different effector functions and pharmcokinetic properties. IgG is the most stable of the five types having a serum half -life in humans of about 23 days. There are four IgG subclasses (Gl, G2, G3, and G4) each of which have different biological functions known as effector functions. These effector functions are generally mediated through interaction with the Fc receptor (FcγR) or by binding Clq and fixing complement. Binding to FcγR can lead to antibody dependent cell mediated cytolysis, whereas binding to complement factors can lead to complement mediated cell lysis. In designing heterologous Fc fusion proteins wherein the Fc portion is being utilized solely for its ability to extend half-life, it is important to minimize any effector function. Thus, the heterologous fusion proteins of the present invention are derived from the human IgG4 Fc region because of its reduced ability to bind FcγR and complement factors compared to other IgG sub-types. IgG4, however, has been shown to deplete target cells in humans [Issacs et al., (1996) Clin. Exp. Immunol. 106:427-433]. Because the heterologous fusion proteins of the present invention target beta cells in the pancreas to induce insulin expression, using an IgG4 derived region in an Fc fusion protein could initiate an immune response against the pancreateic beta cell through interaction of the fusion protein with the GLP-1 receptor present on pancreatic beta cells. Thus, the IgG4 Fc region which is part of the heterologous fusion proteins of the present invention contains substitutions that eliminate effector function. The IgG4 Fc portion of the fusion proteins of the present invention may contain one or more of the following substitutions: substitution of proline for glutamate at residue 233, alanine or valine for phenylalanine at residue 234 and alanine or glutamate for leucine at residue 235 (EU numbering, Kabat, E.A. et al. (1991) Sequences of Proteins of Immunological Interest, 5 Ed. U.S. Dept. of Health and Human Services, Bethesda, MD, NIH Publication no. 91-3242). These residues corresponds to positions 16, 17 and 18 in SEQ ID NO:7. Further, removing the N-linked glycosylation site in the IgG4 Fc region by substituting Ala for Asn at residue 297 (EU numbering) which corresponds to position 80 of SEQ ID NO:7 is another way to ensure that residual effector activity is eliminated in the context of a heterologous fusion protein. In addition, the IgG4 Fc portion of the heterologous fusion proteins of the present invention contain a substitution that stabilizes heavy chain dimer formation and prevents the formation of half-IgG4 Fc chains. The heterologous fusion proteins of the present invention preferably exist as dimers joined together by disulfide bonds and various non-covalent interactions. Wild-type IgG4 contains a Pro-Pro-Cys-Pro-Ser-Cys (SEQ ID NO: 18) motif beginning at residue 224 (EU numbering). This motif in a single GLP-1 analog-Fc chain forms disulfide bonds with the corresponding motif in another GLP-1 analog-Fc chain. However, the presence of serine in the motif causes the formation of single chain fusion proteins. The present invention encompasses heterologous Fc fusion proteins wherein the IgG4 sequence is further modified such that serine at position at 228 (EU numbering) is substituted with proline (amino acid residue ll in SEQ ID NO:7). The C-terminal lysine residue present in the native molecule may be deleted in the IgG4 derivative Fc portion of the heterologous fusion proteins discussed herein (position 230 of SEQ ID NO:7; deleted lysine referred to as des- K). Fusion proteins expressed in some cell types (such as NSO cells) wherein lysine is encoded by the C-terminal codon are heterogeneous in that a portion of the molecules have lysine as the C-terminal amino acid and a portion have lysine deleted. The deletion is due to protease action during expression in some types of mammalian cells. Thus, to avoid this heterogeneity, it is preferred that Fc fusion expression constructs lack a C-terminal codon for lysine. It is preferred that the C-terminal amino acid of the GLP-1 analog portion discussed herein is fused to the N-terminus of the IgG4 Fc analog portion via a glycine-rich linker. The in vivo function and stability of the heterologous fusion proteins of the present invention can be optimized by adding small peptide linkers to prevent potentially unwanted domain interactions. Further, a glycine- rich linker provides some structural flexibility such that the GLP-1 analog portion can interact productively with the GLP-1 receptor on target cells such as the beta cells of the pancreas. These linkers, however, can significantly increase the risk that the fusion protein will be immunogenic in vivo. Thus, it is preferred that the length be no longer than necessary to prevent unwanted domain interactions and/or optimize biological activity and/or stability. The preferred glycine-rich linker comprises the sequence: Gly-Gly-Gly-Gly-Ser-Gly-Gly-Gly- Gly-Ser-Gly-Gly-Gly-Gly-Ser (SEQ ID NO:8). Although more copies of this linker may be used in the heterologous fusion proteins of the present invention, it is preferred that a single copy of this linker be used to minimize the risk of immunogenicity associated with prolonged and repeated administration. Preferred GLP-1 -Fc heterologous fusion proteins of the present invention include the following proteins: Gly8-Glu22-Gly36-GLP-l(7-37)-lL-IgG4 (S228P), Gly8-Glu22- Gly36-GLP-l(7-37)-lL-IgG4 (S228P, F234A, L235A), Gly8-Glu22-Gly36-GLP-l(7-37)- lL-IgG4 (S228P, N297A), Gly8-Glu22-Gly36-GLP-l(7-37)-lL-IgG4 (S228P, F234A, L235A, N297A), Gly8-Glu22-Gly36-GLP-l(7-37)-1.5L-IgG4 (S228P), Gly8-Glu22-Gly36- GLP-l(7-37)-1.5L-IgG4 (S228P, F234A, L235A), Gly8-Glu22-Gly36-GLP-l(7-37)-1.5L- IgG4 (S228P, N297A), Glys-Glu22-Gly36-GLP-l(7-37)-1.5L-IgG4 (S228P, F234A, L235A, N297A), Gly8-Glu22-Gly36-GLP-l(7-37)-2L-IgG4 (S228P), Gly8-Glu22-Gly36- GLP-l(7-37)-2L-IgG4 (S228P, F234A, L235A), Gly8-Glu22-Gly36-GLP-l(7-37)-2L-IgG4 (S228P, N297A), and Gly8-Glu22-Gly36-GLP-l(7-37)-2L-IgG4 (S228P, F234A, L235A, N297A), and the Val8 and des-K forms of all of the above. The nomenclature used herein to refer to specific heterologous fusion proteins is defined as follows: Specific substitutions to the GLP-1 portion of the fusion protein are indicated using the specific amino acid being substituted followed by the residue number. GLP-1 (7-37) indicates that the GLP-1 portion of the mature fusion protein begins with His at position 7 and ends with Gly at position 37. L refers to a linker with the sequence Gly-Gly-Gly-Gly-Ser-Gly-Gly-Gly-Gly-Ser-Gly-Gly-Gly-Gly-Ser (SEQ ID NO:8). The number immediately preceding the L refers to the number of linkers separating the GLP-1 portion from the Fc portion. A linker specified as 1.5L refers to the sequence Gly-Ser- Gly-Gly-Gly-Gly-Ser-Gly-Gly-Gly-Gly-Ser-Gly-Gly-Gly-Gly-Ser-Gly-Gly-Gly-Gly-Ser (SEQ ID NO: 19) IgG4 refers to an analog of the human IgG4 Fc sequence specified as SEQ ID NO:7. Substitutions in the IgG4 Fc portion of the heterologous fusion protein are indicated in parenthesis. The wild-type amino acid is specified by its common abbreviation followed by the position number in the context of the entire IgG4 sequence using the EU numbering system followed by the amino acid being substituted at that position specified by its common abbreviation. Although the heterologous fusion proteins of the present invention can be made by a variety of different methods, because of the size of the fusion protein, recombinant methods are preferred. For purposes of the present invention, as disclosed and claimed herein, the following general molecular biology terms and abbreviations are defined below. "Base pair" or "bp" as used herein refers to DNA or RNA. The abbreviations A,C,G, and T correspond to the 5 '-monophosphate forms of the deoxyribonucleosides (deoxy)adenosine, (deoxy)cytidine, (deoxy)guanosine, and thymidine, respectively, when they occur in DNA molecules. The abbreviations U,C,G, and A correspond to the 5'- monophosphate forms of the ribonucleosides uridine, cytidine, guanosine, and adenosine, respectively when they occur in RNA molecules. In double stranded DNA, base pair may refer to a partnership of A with T or C with G. In a DNA/RNA, heteroduplex base pair may refer to a partnership of A with U or C with G. (See the definition of "complementary", infra.) "Digestion" or "Restriction" of DNA refers to the catalytic cleavage of the DNA with a restriction enzyme that acts only at certain sequences in the DNA ("sequence- specific endonucleases"). The various restriction enzymes used herein are commercially available and their reaction conditions, cofactors, and other requirements were used as would be known to one of ordinary skill in the art. Appropriate buffers and substrate amounts for particular restriction enzymes are specified by the manufacturer or can be readily found in the literature. "Ligation" refers to the process of forming phosphodiester bonds between, two double stranded nucleic acid fragments. Unless otherwise provided, ligation may be accomplished using known buffers and conditions with a DNA ligase, such as T4 DNA ligase. "Plasmid" refers to an extrachromosomal (usually) self-replicating genetic element. "Recombinant DNA cloning vector" as used herein refers to any autonomously replicating agent, including, but not limited to, plasmids and phages, comprising a DNA molecule to which one or more additional DNA segments can or have been added. "Recombinant DNA expression vector" as used herein refers to any recombinant DNA cloning vector in which a promoter to control transcription of the inserted DNA has been incorporated. "Transcription" refers to the process whereby information contained in a nucleotide sequence of DNA is transferred to a complementary RNA sequence. "Transfection" refers to the uptake of an expression vector by a host cell whether or not any coding sequences are, in fact, expressed. Numerous methods of transfection are known to the ordinarily skilled artisan, for example, calcium phosphate co- precipitation, liposome transfection, and electroporation. Successful transfection is generally recognized when any indication of the operation of this vector occurs within the host cell. "Transformation" refers to the introduction of DNA into an organism so that the
DNA is replicable, either as an extrachromosomal element or by chromosomal integration. Methods of transforming bacterial and eukaryotic hosts are well known in the art, many of which methods, such as nuclear injection, protoplast fusion or by calcium treatment using calcium chloride are summarized in J. Sambrook, et al, Molecular Cloning: A Laboratory Manual, (1989). Generally, when introducing DNA into Yeast the term transformation is used as opposed to the term transfection. "Translation" as used herein refers to the process whereby the genetic information of messenger RNA (mRNA) is used to specify and direct the synthesis of a polypeptide chain. "Vector" refers to a nucleic acid compound used for the transfection and/or transformation of cells in gene manipulation bearing polynucleotide sequences corresponding to appropriate protein molecules which, when combined with appropriate control sequences, confers specific properties on the host cell to be transfected and/or transformed. Plasmids, viruses, and bacteriophage are suitable vectors. Artificial vectors are constructed by cutting and joining DNA molecules from different sources using restriction enzymes and ligases. The term "vector" as used herein includes Recombinant DNA cloning vectors and Recombinant DNA expression vectors. "Complementary" or "Complementarity", as used herein, refers to pairs of bases (purines and pyrimidines) that associate through hydrogen bonding in a double stranded nucleic acid. The following base pairs are complementary: guanine and cytosine; adenine and thymine; and adenine and uracil. "Primer" refers to a nucleic acid fragment which functions as an initiating substrate for enzymatic or synthetic elongation. "Promoter" refers to a DNA sequence which directs transcription of DNA to RNA. "Probe" refers to a nucleic acid compound or a fragment, thereof, which hybridizes with another nucleic acid compound. "Leader sequence" refers to a sequence of amino acids which can be enzymatically or chemically removed to produce the desired polypeptide of interest. "Secretion signal sequence" refers to a sequence of amino acids generally present at the N-terminal region of a larger polypeptide functioning to initiate association of that polypeptide with the cell membrane compartments like endoplasmic reticulum and secretion of that polypeptide through the plasma membrane. Wild-type human IgG4 proteins can be obtained from a variety of sources. For example, these proteins can be obtained from a cDNA library prepared from cells which express the mRNA of interest at a detectable level. Libraries can be screened with probes designed using the published DNA or protein sequence for the particular protein of interest. For example, immunoglobulin light or heavy chain constant regions are described in Adams, et al. (1980) Biochemistry 19:2711-2719; Goughet, et al. (1980) Biochemistry 19:2702-2710; Dolby, et al. (1980) Proc. Natl. Acad. Sci. USA 77:6027- 6031; Rice et al. (1982) Proc. Natl. Acad. Sci. USA 79:7862-7862; Falkner, et al. (1982) Nature 298:286-288; and Morrison, et al. (1984) Ann. Rev. Immunol. 2:239-256. Screening a cDNA or genomic library with the selected probe may be conducted using standard procedures, such as described in Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory Press, NY (1989). An alternative means to isolate a gene encoding an immunoglobulin protein is to use PCR methodology [Sambrook et al., supra; Dieffenbach et al., PCR Primer: A Laboratory Manual, Cold Spring Harbor Laboratory Press, NY (1995)]. PCR primers can be designed based on published sequences. Generally the full-length wild-type sequences cloned from a particular library can serve as a template to create the IgG4 Fc analog fragments of the present invention that retain the ability to confer a longer plasma half -life on the GLP-1 analog that is part of the fusion protein. The IgG4 Fc analog fragments can be generated using PCR techniques with primers designed to hybridize to sequences corresponding to the desired ends of the fragment. PCR primers can also be designed to create restriction enzyme sites to facilitate cloning into expression vectors. DNA encoding the GLP-1 analogs of the present invention can be made by a variety of different methods including cloning methods like those described above as well as chemically synthesized DNA. Chemical synthesis may be attractive given the short length of the encoded peptide. The amino acid sequence for GLP-1 has been published as well as the sequence of the preproglucagon gene. [Lopez, et al. (1983) Proc. Natl. Acad. Sci., USA 80:5485-5489; Bell, et al. (1983) Nature, 302:716-718; Heinrich, G., et al. (1984) Endocrinol, 115:2176-2181; Ghiglione, M., et al. 91984) Diabetologia 27:599- 600]. Thus, primers can be designed based on the native sequence to generate DNA encoding the GLP-1 analogs described herein. The gene encoding a fusion protein can then be constructed by ligating DNA encoding a GLP-1 analog in-frame to DNA encoding the IgG Fc proteins described herein. The DNA encoding wild-type GLP-1 and IgG4 Fc fragments can be mutated either before ligation or in the context of a cDNA encoding an entire fusion protein. A variety of mutagenesis techniques are well known in the art. The gene encoding the GLP- 1 analog and the gene encoding the IgG4 Fc analog protein can also be joined in-frame via DNA encoding a G-rich linker peptide. A preferred DNA sequence encoding one of the preferred heterologous fusion proteins of the present invention, Gly8-Glu 2-Gly36- GLP-l(7-37)-lL-IgG4 (S228P, F234A, L235A, des K), is provided as SEQ ID NO:20: CACGGCGAGGGCACCTTCACCTCCGACGTGTCCTCCTATCTCGAGGAGCAGG CCGCCAAGGAATTCATCGCCTGGCTGGTGAAGGGCGGCGGCGGTGGTGGTGG CTCCGGAGGCGGCGGCTCTGGTGGCGGTGGCAGCGCTGAGTCCAAATATGGT CCCCCATGCCCACCCTGCCCAGCACCTGAGGCCGCCGGGGGACCATCAGTCTT CCTGTTCCCCCCAAAACCCAAGGACACTCTCATGATCTCCCGGACCCCTGAGG TCACGTGCGTGGTGGTGGACGTGAGCCAGGAAGACCCCGAGGTCCAGTTCAA CTGGTACGTGGATGGCGTGGAGGTGCATAATGCCAAGACAAAGCCGCGGGAG GAGCAGTTCAACAGCACGTACCGTGTGGTCAGCGTCCTCACCGTCCTGCACCA GGACTGGCTGAACGGCAAGGAGTACAAGTGCAAGGTCTCCAACAAAGGCCTC CCGTCCTCCATCGAGAAAACCATCTCCAAAGCCAAAGGGCAGCCCCGAGAGC CACAGGTGTACACCCTGCCCCCATCCCAGGAGGAGATGACCAAGAACCAGGT CAGCCTGACCTGCCTGGTCAAAGGCTTCTACCCCAGCGACATCGCCGTGGAGT GGGAAAGCAATGGGCAGCCGGAGAACAACTACAAGACCACGCCTCCCGTGCT GGACTCCGACGGCTCCTTCTTCCTCTACAGCAGGCTAACCGTGGACAAGAGC AGGTGGCAGGAGGGGAATGTCTTCTCATGCTCCGTGATGCATGAGGCTCTGC ACAACCACTACACACAGAAGAGCCTCTCCCTGTCTCTGGGT (SEQ ID NO:20) Host cells are transfected or transformed with expression or cloning vectors described herein for heterologous fusion protein production and cultured in conventional nutrient media modified as appropriate for inducing promoters, selecting transformants, or amplifying the genes encoding the desired sequences. The culture conditions, such as media, temperature, pH and the like, can be selected by the skilled artisan without undue experimentation. In general, principles, protocols, and practical techniques for maximizing the productivity of cell cultures can be found in Mammalian Cell Biotechnology: A Practical Approach, M. Butler, ed. (IRL Press, 1991) and Sambrook, et al, supra. Methods of transfection are known to the ordinarily skilled artisan, for example, CaPO4 and electroporation. General aspects of mammalian cell host system transformations have been described in U.S. Patent No. 4,399,216. Transformations into yeast are typically carried out according to the method of van Solingen et al., J Bact. 130(2): 946-7 (1977) and Hsiao et al, Proc. Natl. Acad. Sci. USA 76(8): 3829-33 (1979). However, other methods for introducing DNA into cells, such as by nuclear microinjection, electroporation, bacterial protoplast fusion with intact cells, or polycations, e.g., polybrene or polyomithine, may also be used. For various techniques for transforming mammalian cells, see Keown, et al., Methods in Enzymology 185: 527-37 (1990) and Mansour, et al., Nature 336(6197): 348-52 (1988). Suitable host cells for cloning or expressing the nucleic acid (e.g., DNA) in the vectors herein include yeast or higher eukaryote cells. Eukaryotic microbes such as filamentous fungi or yeast are suitable cloning or expression hosts for fusion protein vectors. Saccharomyces cerevisiae is a commonly used lower eukaryotic host microorganism. Others include Schizosaccharomyces pombe [Beach and Nurse, Nature 290: 140-3 (1981); EP 139,383 published 2 May 1995]; Muyveromyces hosts [U.S. Patent No. 4,943,529; Fleer, et al, Bio Technology 9(10): 968-75 (1991)] such as, e.g., K lactis (MW98-8C, CBS683, CBS4574) [de Louvencourt et al, /. Bacteriol. 154(2): 737-42 (1983)]; K. fiagilis (ATCC 12,424), K. bulgaricus (ATCC 16,045), K wickeramii (ATCC 24,178), K waltii (ATCC 56,500), K. drosophilarum (ATCC 36.906) [Van den Berg et al., Bio/Technology 8(2): 135-9 (1990)]; K. thermotoierans, and K. marxianus; yarrowia (EP 402,226); Pichia pastoris (EP 183,070) [Sreekrishna et al, J. Basic Microbiol. 28(4): 265-78 (1988)]; Candid; Trichoderma reesia (EP 244,234); Neurospora crassa [Case, et al., Proc. Natl. Acad Sci. USA 76(10): 5259-63 (1979)]; Schwanniomyces such as Schwanniomyces occidentulis (EP 394,538 published 31 October 1990); and filamentous fungi such as, e.g., Neurospora, Penicillium, Tolypocladium (WO 91/00357 published 10 January 1991), and Aspergillus hosts such as A. nidulans [Ballance et al., Biochem. Biophys. Res. Comm. 112(1): 284-9 (1983)]; Tilburn, et al, Gene 26(2-3): 205-21 (1983); Yelton, et al, Proc. Natl. Acad. Sci. USA 81(5): 1470-4 (1984)] and A. niger [Kelly and Hynes, EMBO J. 4(2): 475-9 (1985)]. Methylotropic yeasts are selected from the genera consisting of
Hansenula, Candida, Kloeckera, Pichia, Saccharomyces, Torulopsis, and Rhodotoruia. A list of specific species that are exemplary of this class of yeast may be found in C. Antony, The Biochemist^ of Methylotrophs 269 (1982). Suitable host cells for the expression of the fusion proteins of the present invention are derived from multicellular organisms. Examples of invertebrate cells include insect cells such as Drosophila S2 and Spodoptera Sp, Spodoptera high5 as well as plant cells. Examples of useful mammalian host cell lines include NSO myeloma cells, Chinese hamster ovary (CHO), SP2, and COS cells. More specific examples include monkey kidney CV1 line transformed by SV40 (COS-7, ATCC CRL 1651); human embryonic kidney line [293 or 293 cells subcloned for growth in suspension culture, Graham, et al, J. Gen Virol, 36(1): 59-74 (1977)]; Chinese hamster ovary cells/-DHFR [CHO, Urlaub and Chasin, Proc. Natl. Acad. Sci. USA, 77(7): 4216-20 (1980)]; mouse sertoli cells [TM4, Mather, Biol. Reprod. 23(l):243-52 (1980)]; human lung cells (W138. ATCC CCL 75); human liver cells (Hep G2, HB 8065); and mouse mammary tumor (MMT 060562, ATCC CCL51). A preferred cell line for production of the Fc fusion proteins of the present invention is the NSO myeloma cell line available from the European Collection of Cell Cultures (ECACC, catalog #85110503) and described in Galfre, G. and Milstein, C. ((1981) Methods in Enzymology 73(13):3-46; and Preparation of Monoclonal Antibodies: Strategies and Procedures, Academic Press, Ν.Y.,
N.Y.). The fusion proteins of the present invention may be recombinantly produced directly, or as a protein having a signal sequence or other additional sequences which create a specific cleavage site at the N-terminus of the mature fusion protein. In general, the signal sequence may be a component of the vector, or it may be a part of the fusion protein-encoding DNA that is inserted into the vector. For yeast secretion the signal sequence may be, e.g., the yeast invertase leader, alpha factor leader (including Saccharomyces and Kluyveromyces cc-factor leaders, the latter described in U.S. Patent No. 5,010,182), or acid phosphatase leader, the C. albicans glucoamylase leader (EP 362,179), or the signal described in WO 90/13646. In mammalian cell expression, mammalian signal sequences may be used to direct secretion of the protein, such as signal sequences from secreted polypeptides of the same or related species as well as viral secretory leaders. Both expression and cloning vectors contain a nucleic acid sequence that enables the vector to replicate in one or more selected host cells. Expression and cloning vectors will typically contain a selection gene, also termed a selectable marker. Typical selection genes encode proteins that (a) confer resistance to antibiotics or other toxins, e.g., neomycin, methotrexate, or tetracycline, (b) complement autotrophic deficiencies, or (c) supply critical nutrients not available from complex media, e.g., the gene encoding D-alanine racemase for Bacilli. An example of suitable selectable markers for mammalian cells are those that enable the identification of cells competent to take up the fusion protein-encoding nucleic acid, such as DHFR or thymidine kinase. An appropriate host cell when wild-type DHFR is employed is the CHO cell line deficient in DHFR activity, prepared and propagated as described [Urlaub and Chasin, Proc. Natl. Acad. Sci. USA, 77(7): 4216-20 (1980)]. A suitable selection gene for use in yeast is the trpl gene present in the yeast plasmid YRp7 [Stinchcomb, et al, Nature 282(5734): 39-43 (1979); Kingsman, et al, Gene 7(2): 141-52 (1979); Tschumper, et al, Gene 10(2): 157-66 (1980)]. The trpl gene provides a selection marker for a mutant strain of yeast lacking the ability to grow in tryptophan, for example, ATCC No. 44076 or PEPC1 [Jones, Genetics 85: 23-33 (1977)]. Expression and cloning vectors usually contain a promoter operably linked to the fusion protein-encoding nucleic acid sequence to direct mRNA synthesis. Promoters recognized by a variety of potential host cells are well known. Examples of suitable promoting sequences for use with yeast hosts include the promoters for 3-phosphoglycerate kinase [Hitzeman, et al, J. Biol Chem. 255(24): 12073-80 (1980)] or other glycolytic enzymes [Hess et al., J. Adv. Enzyme Reg. 7: 149 (1968); Holland, Biochemistry 17(23): 4900-7 (1978)], such as enolase, glyceraldehyde-3 -phosphate dehydrogenase, hexokinase, pyruvate decarboxylase, phosphofructokinase, glucose-6-ρhosρhate isomerase, 3- phosphoglycerate mutase, pyruvate kinase, triosephosphate isomerase, phosphoglucose isomerase, and glucokinase. Other yeast promoters, which are inducible promoters having the additional advantage of transcription controlled by growth conditions, are the promoter regions for alcohol dehydrogenase 2, isocytochrome C, acid phosphatase, degradative enzymes associated with nitrogen metabolism, metallothionein, gl ceraldehyde-3 -phosphate dehydrogenase, and enzymes responsible for maltose and galactose utilization. Suitable vectors and promoters for use in yeast expression are further described in EP 73,657. Transcription of fusion protein-encoding mRNA from vectors in mammalian host cells may be controlled, for example, by promoters obtained from the genomes of viruses such as polyoma virus, fowlpox virus, adenovirus (such as Adeno virus 2), bovine papilloma virus, avian sarcoma virus, cytomegalovirus, a retrovirus, hepatitis-B virus and Simian Virus 40 (SV40), from heterologous mammalian promoters, e.g., the actin promoter or an immunoglobulin promoter, and from heat-shock promoters, provided such promoters are compatible with the host cell systems. Transcription of a polynucleotide encoding a fusion protein by higher eukaryotes may be increased by inserting an enhancer sequence into the vector. Enhancers are cis-acting elements of DNA, usually about from 10 to 300 bp, that act on a promoter to increase its transcription. Many enhancer sequences are now known from mammalian genes (globin, elastase, albumin, a-ketoprotein, and insulin). Typically, however, one will use an enhancer from a eukaryotic cell virus. Examples include the SV40 enhancer on the late side of the replication origin (bp 100-270), the cytomegalovirus early promoter enhancer, the polyoma enhancer on the late side of the replication origin, and adenovirus enhancers. The enhancer may be spliced into the vector at a position 5' or 3' to the fusion protein coding sequence but is preferably located at a site 5' from the promoter. Expression vectors used in eukaryotic host cells (yeast, fungi, insect, plant, animal, human, or nucleated cells from other multicellular organisms) will also contain sequences necessary for the termination of transcription and for stabilizing the mRNA. Such sequences are commonly available from the 5' and occasionally 3' untranslated regions of eukaryotic or viral DNAs or cDNAs. These regions contain nucleotide segments transcribed as polyadenylated fragments in the untranslated portion of the mRNA encoding the fusion protein. Various forms of a fusion protein may be recovered from culture medium or from host cell lysates. If membrane-bound, it can be released from the membrane using a suitable detergent solution (e.g., Triton-X 100) or by enzymatic cleavage. Cells employed in expression of a fusion protein can be disrupted by various physical or chemical means, such as freeze- thaw cycling, sonication, mechanical disruption, or cell lysing agents. Once the heterologous fusion proteins of the present invention are expressed in the appropriate host cell, the analogs can be isolated and purified. The following procedures are exemplary of suitable purification procedures: fractionation on carboxymethyl cellulose; gel filtration such as Sephadex G-75; anion exchange resin such as DEAE or Mono-Q; cation exchange such as CM or Mono-S; metal chelating columns to bind epitope-tagged forms of the polypeptide; reversed-phase HPLC; chromatofocusing; silica gel; ethanol precipitation; and ammonium sulfate precipitation. Various methods of protein purification may be employed and such methods are known in the art and described, for example, in Deutscher, Methods in Enzymology 182: 83-9 (1990) and Scopes, Protein Purification: Principles and Practice, Springer- Verlag, NY (1982). The purification step(s) selected will depend on the nature of the production process used and the particular fusion protein produced. For example, fusion proteins comprising an Fc fragment can be effectively purified using a Protein A or Protein G affinity matrix. Low or high pH buffers can be used to elute the fusion protein from the affinity matrix. Mild elution conditions will aid in preventing irreversible denaturation of the fusion protein. The heterologous fusion proteins of the present invention may be formulated with one or more excipients. The fusion proteins of the present invention may be combined with a pharmaceutically acceptable buffer, and the pH adjusted to provide acceptable stability, and a pH acceptable for administration such as parenteral administration. Optionally, one or more pharmaceutically-acceptable anti-microbial agents may be added. Meta-cresol and phenol are preferred pharmaceutically-acceptable microbial agents. One or more pharmaceutically- acceptable salts may be added to adjust the ionic strength or tonicity. One or more excipients may be added to further adjust the isotonicity of the formulation. Glycerin is an example of an isotonicity-adjusting excipient. Pharmaceutically acceptable means suitable for administration to a human or other animal and thus, does not contain toxic elements or undesirable contaminants and does not interfere with the activity of the active compounds therein. The heterologous fusion proteins of the present invention may be formulated as a solution formulation or as a lyophilized powder that can be reconstituted with an appropriate diluent. A lyophilized dosage form is one in which the fusion protein is stable, with or without buffering capacity to maintain the pH of the solution over the intended in-use shelf- life of the reconstituted product. It is preferable that the solution comprising the heterologous fusion proteins discussed herein before lyphilization be substantially isotonic to enable formation of isotonic solutions after reconstitution. A pharmaceutically-acceptable salt form of the heterologous fusion proteins of the present invention are within the scope of the invention. Acids commonly employed to form acid addition salts are inorganic acids such as hydrochloric acid, hydrobromic acid, hydriodic acid, sulfuric acid, phosphoric acid, and the like, and organic acids such as -toluenesulfonic acid, methanesulfonic acid, oxalic acid, -bromophenyl-sulfonic acid, carbonic acid, succinic acid, citric acid, benzoic acid, acetic acid, and the like. Preferred acid addition salts are those formed with mineral acids such as hydrochloric acid and hydrobromic acid. Base addition salts include those derived from inorganic bases, such as ammonium or alkali or alkaline earth metal hydroxides, carbonates, bicarbonates, and the like. Such bases useful in preparing the salts of this invention thus include sodium hydroxide, potassium hydroxide, ammonium hydroxide, potassium carbonate, and the like. The heterologous fusion proteins of the present invention have biological activity. Biological activity refers to the ability of the fusion protein to bind to and activate the GLP-1 receptor in vivo and elicit a response. Responses include, but are not limited to, secretion of insulin, suppression of glucagon, inhibition of appetite, weight loss, induction of satiety, inhibition of apoptosis, induction of pancreatic beta cell proliferation, and differentiation of pancreatic beta cells. A representative number of GLP-1 fusion proteins were tested for in vitro as well as in vivo activity. Examples 1 and 2 provide in vitro activity based on the ability of the fusion protein to interact with and activate the human GLP-1 receptor. In both sets of experiments, HEK293 cells over-expressing the human GLP-1 receptor were used. Activation of the GLP-1 receptor in these cells causes adenylyl cyclase activation which in turn induces expression of a reporter gene driven by a cyclic AMP response element (CRE). Example 1 (table 1) provides data wherein the reporter gene is beta lactamase, and example 2 (table 2) provides data wherein the reporter gene is luciferase. Example 3 provides data generated after administration of one of the heterologous fusion proteins of the present invention to rats. Together the data show that the fusion proteins are able to bind to and activate the GLP-1 receptor and appear more potent in vitro than Val8-GLP-l(7-37)OH. In addition, the data generated in rats indicate the fusion proteins are active in vivo and have a longer half-life than native GLP-1. Administration of the heterogeneous fusion proteins may be via any route known to be effective by the physician of ordinary skill. Peripheral parenteral is one such method. Parenteral administration is commonly understood in the medical literature as the injection of a dosage form into the body by a sterile syringe or some other mechanical device such as an infusion pump. Peripheral parenteral routes can include intravenous, intramuscular, subcutaneous, and intraperitoneal routes of administration. The heterologous fusion proteins of the present invention may also be amenable to administration by oral, rectal, nasal, or lower respiratory routes, which are non-parenteral routes. Of these non-parenteral routes, the lower respiratory route and the oral route are preferred. The fusion proteins of the present invention can be used to treat a wide variety of diseases and conditions. The fusion proteins of the present invention primarily exert their biological effects by acting at a receptor referred to as the "GLP-1 receptor." Subjects with diseases and/or conditions that respond favorably to GLP-1 receptor stimulation or to the administration of GLP-1 compounds can therefore be treated with the GLP-1 fusion proteins of the present invention. These subjects are said to "be in need of treatment with GLP-1 compounds" or "in need of GLP-1 receptor stimulation". Included are subjects with non- insulin dependent diabetes, insulin dependent diabetes, stroke (see WO 00/16797), myocardial infarction (see WO 98/08531), obesity (see WO 98/19698), catabolic changes after surgery (see U.S. Patent No. 6,006,753), functional dyspepsia and irritable bowel syndrome (see WO 99/64060). Also included are subjects requiring prophylactic treatment with a GLP-1 compound, e.g., subjects at risk for developing non-insulin dependent diabetes (see WO 00/07617). Subjects with impaired glucose tolerance or impaired fasting glucose, subjects whose body weight is about 25% above normal body weight for the subject's height and body build, subjects with a partial pancreatectomy, subjects having one or more parents with non-insulin dependent diabetes, subjects who have had gestational diabetes and subjects who have had acute or chronic pancreatitis are at risk for developing non-insulin dependent diabetes. An effective amount of the GLP-1 -Fc fusion proteins described herein is the quantity which results in a desired therapeutic and/or prophylactic effect without causing unacceptable side-effects when administered to a subject in need of GLP-1 receptor stimulation. A "desired therapeutic effect" includes one or more of the following: 1) an amelioration of the symptom(s) associated with the disease or condition; 2) a delay in the onset of symptoms associated with the disease or condition; 3) increased longevity compared with the absence of the treatment; and 4) greater quality of life compared with the absence of the treatment. For example, an "effective amount" of a GLP-l-Fc fusion protein for the treatment of diabetes is the quantity that would result in greater control of blood glucose concentration than in the absence of treatment, thereby resulting in a delay in the onset of diabetic complications such as retinopathy, neuropathy or kidney disease. An "effective amount" of a GLP-l-Fc fusion protein for the prevention of diabetes is the quantity that would delay, compared with the absence of treatment, the onset of elevated blood glucose levels that require treatment with anti-hypoglycaemic drugs such as sulfonyl ureas, thiazolidinediones, insulin and/or bisguanidines. The dose of fusion protein effective to normalize a patient's blood glucose will depend on a number of factors, among which are included, without limitation, the subject's sex, weight and age, the severity of inability to regulate blood glucose, the route of administration and bioavailability, the pharmacokinetic profile of the fusion protein, the potency, and the formulation. Doses may be in the range of 0.01 to 1 mg/kg body weight, preferably in the range of 0.05 to 0.5 mg/kg body weight. It is preferable that the fusion proteins of the present invention be administered either once every two weeks or once a week. Depending on the disease being treated, it may be necessary to administer the fusion protein more frequently such as two to three time per week. The present invention will now be described only by way of non-limiting example with reference to the following Examples.
EXAMPLES
Example 1 - In vitro GLP-1 receptor activation assay HEK-293 cells expressing the human GLP-1 receptor, using a CRE-BLAM system, are seeded at 20,000 to 40,000 cells/well/ 100 μl DMEM medium with 10%FBS into a poly-d-lysine coated 96 well black, clear-bottom plate. The day after seeding, the medium is flicked off and 80 μl plasma-free DMEM medium is added. On the third day after seeding, 20 μl of plasma-free DMEM medium with 0.5% BSA containing different concentrations of various GLP-l-Fc heterologous fusion protein is added to each well to generate a dose response curve. Generally, fourteen dilutions containing from 3 nanomolar to 30 nanomolar or heterologous GLP-1 Fc fusion protein are used to generate a dose response curve from which EC50 values can be determined. After 5 hours of incubation with the fusion protein, 20 μl of β-lactamase substrate (CCF2/AM, PanVera LLC) is added and incubation continued for 1 hour at which time fluorescence is determined on a cytofluor. The assay is further described in Zlokarnik, et al. (1998), Science, 278:84-88. Various GLP-l-Fc fusion proteins are tested and EC50 values are represented in Table 1. The values are relative to values determined for Val -GLP-1 (7- 37)OH which is run as an internal control with every experiment.
Table 1 Compound Activity Std. Dev.
Val8-GLP-1: 100%
Gly8-Glu22-GLP-l(7-37)-2L-IgG4 (S228P, F234A, L235A): 301 % 99
Gly8-Glu22-GLP-l(7-37)-1.5L-IgG4 (S228P, F234A, L235A): 314% 45 Gly8-Glu22-GLP-l(7-37)-lL-IgG4 (S228P, F234A, L235A): 468% 120 Gly8-Glu22-Gly36-GLP-l(7-37)-2L-IgG4 (S228P, F234A, L235A): 441% 35
Example 2 - In vitro GLP-1 receptor activation assay HEK-293 cells stably expressing the human GLP-1 receptor, using a CRE- Luciferase system, are seeded at 30,000 cells/well/80 μl low serum DMEM F12 medium into 96 well plates. The day after seeding, 20 μl aliquots of test protein dissolved in 0.5% BSA are mixed and incubated with the cells for 5 hours. Generally 12 dilutions containing from 3 pM to 3 nM are prepared at a 5X concentration for each test protein before addition to the cells to generate a dose response curve from which EC50 values are determined. After incubation, 100 μl of Lucif erase reagent is added directly to each plate and mixed gently for 2 minutes. Plates are placed in a Tri-lux luminometer and light output resulting from luciferase expression is calculated. Various GLP-l-Fc fusion proteins are tested and EC50 values are represented in Table 2. The values are relative to values determined for Val -GLP-l(7-37)OH which is run as an internal control with every experiment. Because the fusion proteins tested below are dimers, values are corrected taking into account a 2-fold difference in molarity.
Table 2
Compound Activity Std. Dev.
Val8-GLP-1: 100%
Gly8-Glu22-GLP-l(7-37)-2L-IgG4 (S228P, F234A, L235A): 535% 240
Gly8-Glu22-GLP-l(7-37)-1.5L-IgG4 (S228P, F234A, L235A): 595% 43 Gly8-Glu22-GLP-l(7~37)-lL-IgG4 (S228P, F234A, L235A): 1119% 128
Gly8-Glu22-Gly36-GLP-l(7-37)-2L-IgG4 (S228P, F234A, L235A): 398% 62 Gly8-Glu22-Gly36-GLP-l(7-37)-lL-IgG4 (S228P, F234A, L235A): 417% 140
Example 3 Intravenous Glucose Tolerance Test in Rats The Fc fusion protein, Gly8-Glu2 -Gly36-GLP-l(7-37)-L-IgG4
(S228P,F234A,L235A), is evaluated in an intravenous glucose tolerance test (IVGTT) in rats. At least four rats are included into each of three groups. Group I receives vehicle (table 3), Group II receives 1.79 mg/kg of Gly8-Glu22-Gly36-GLP-l(7-37)-L-IgG4 (S228P,F234A,L235A) as a single subcutaneous injection (table 4), and Group III receives 0.179 mg/kg of Gly8-Glu22-Gly36-GLP-l(7-37)-L-IgG4 (S228P,F234A,L235A) as a single subcutaneous injection (table 5). Rats are subcutaneously injected the morning of Day 1. Twenty-four hours following the first injection, 1 μL of glucose (D50) per gram rat body weight is infused as a bolus. Blood samples are taken at 2, 4, 6, 10, 20, and 30 minutes following the bolus infusion of glucose.
Table 3
Vehicle: Rat 1 Rat 2 Rat 3 Rat 4 Rat 5
Insulin AUC (ng*min/mL) Average SEM 0-2 11 9.4 7 11 9.6 2-4 18.1 9.7 5.6 10.6 8.8 4-6 13.4 7 3.4 9.6 5.9 6-10 7.9 3.5 2.5 6 2.9 10-20 3.7 3 2.4 3 2.4 20-30 2 0 0 0 2.4 sum 56.1 32.6 20.9 40.2 32 36.4 5.8
Table 4
GLP-1 -Fc
(1.79mg/kg) Rat 1 Rat 2 Rat 3 Rat 4 Rat 5
In
Figure imgf000025_0001
Table 5
GLP-1 -Fc
(0.179mg/kg) Rat 1 Rat 2 Rat 3 Rat 4
Ins SEM
Figure imgf000025_0002
8.7 Example 4 Pharmacokinetic Study Following a Single Subcutaneous Injection to Cynomolgus Monkeys. A study is performed to characterize the pharmacokinetics (PK) of the Fc fusion protein, Gly8-Glu22-Gly36-GLP-l(7-37)-L-IgG4 (S228P,F234A,L235A), when administered as a 0.1 mg/kg by subcutaneous (SC) injection to male cynomolgus monkeys. RIA antibody is specific for the middle portion of GLP. ELISA uses an N- terminus specific capture antibody and an Fc specific detection antibody. Resulting plasma concentrations from both the ELISA and the RIA are used to determine the represented pharmacokinetic parameter values. A representation of the resulting PK parameter values is summarized in table 6. Single-dose SC PK from the RIA is associated with a mean Cmax of 446.7 ng/mL with a corresponding Tmax of 17.3 hours. The mean elimination half-life is approximately 79.3 hours (3.3 days). The PK from the ELISA is associated with a mean Cmax of 292.2 ng/mL with a corresponding Tmax of 16J hours. The mean elimination half-life is approximately 51.6 hours (2.2 days).
Table 6
Figure imgf000026_0001
d Elimination half-life. e Total body clearance as a function of bioavailability. f Volume of distribution as a function of bioavailability. SD = Standard deviation. Example 5 Assessment of the potential formation of antibodies following repeat subcutanesous injections. Designated serum samples from cynomolgus monkeys are tested for the formation of antibodies against Gly8-Glu22-Gly36-GLP-l(7-37)-L-IgG4 (S228P,F234A,L235A) using a direct ELISA format. . Microtiter plates are coated with Gly8-Glu22-Gly36-GLP- l(7-37)-L-IgG4 (S228P,F234A,L235A) at a 0.1 μg/mL concentration. Monkey serum samples are diluted 50, 500,1000 and 5000 fold into blocking solution, and 0.05 mL sample/well are incubated approximately one hour. Secondary antibody, Goat <Human Fab'2>-Peroxidase (with 75% cross reactivity to human), is diluted 10,000 fold into block and added at 0.05 mL/well and incubated approximately one hour. Color development using tetramethylbenzidine (TMB) substrate is read at an optical density of 450nm - 630nm. Duplicate readings are averaged. A GLP-1 antibody was used as a positive control and goat<rabbit>(H+L)-Peroxidase conjugate is the secondary used for detection. Point serum samples are collected prior to dosing, at 24 hours following the second dose, and 168 hours following the first and second SC dose for an evaluation of potential immunogenicity. The presence of antibody titers to G8E22-CEX-L-hIgG4 is interpreted by comparison to predose serum samples and positive control. A representation of the results is presented in table 7.
Figure imgf000027_0001
Example 6 Pharmacodynamic Study Following a Single Subcutaneously Injection to Cynomolgus Monkevs.in the Fasting State and During a Graded Intravenous Glucose Infusion. In Phase 1 (Study Day 1) a subcutaneous injection of vehicle is administered. A graded intravenous glucose (20% dextrose) infusion of 5, 10, and 25 mg/kg/min is then administered immediately after the vehicle injection. In Phase 2 (Study Day 3), a subcutaneous injection of a GLP-1 fusion protein (0.1 mg/kg) is administered. In Phase 3, a graded intravenous glucose infusion is performed approximately 96 hours following the GLP-1 fusion injection. Graded intravenous glucose infusion procedures are conducted in sedated monkeys after a 16-hr overnight fast. For both intravenous glucose infusions, baseline samples will be drawn every 10 min for 20 min to define baseline. A stepped-up glucose infusion is initiated at +20 min at a rate of 5 mg kg/min, followed by infusions of 10 mg/kg/min, and 25 mg/kg/min. Each infusion rate is administered for a period of 20 minutes. Blood samples are taken at 10 minute intervals for measurement of glucose, insulin, and glucagon. Approximately 1.0 mL of blood is collected at -20, -10 min, 0 pre- glucose infusions, and at 10, 20, 30, 40, 50, and 60 minutes following glucose infusion for Phases 1 and 3. A representation of the data are shown in table 8.
Table 8 Glucose AUC AUC AUC Group Animal (min*mg/dl_) Group Animal (min*mg/dL) GLP-Fc # 9423 7447 vehicle 9423 8077 9424 7470 9424 15006 9510 5153 9510 7116 9513 6303 9513 7459 9516 5413 9516 8728 9530 5240 9530 7863 N 6 Mean 6171 Mean 9041 SD 1078 SD 2973 SE 440 SE 1214
Insulin AUC AUC AUC Group Animal (min*ng/ml_) Group Animal (min*ng/mL) GLP-Fc 9423 129 vehicle 9423 38 9424 138 9424 29 9510 357 9510 69 9513 161 9513 64 9516 376 9516 38 9530 215 9530 68 Mean 229 Mean 51 SD 111 SD 18 SE 45 SE 7
Glucagon levels were not statistically different between the vehicle and the GLP-1 fusion protein dosed monkeys.
Example 7 Pharmacodynamic Study Following Single Subcutaneously Injections of Three Different Doses to Rats in the Fasting State and During a Graded Intravenous Glucose Infusion. Chronically cannulated rats are assigned to either vehicle control (saline) or one of 3 treatment groups (GLP-1 fusion protein; 0.0179 mg/kg, 0.179 mg/kg, or 1.79 mg/kg). The GLP-1 fusion protein and vehicle are administered via subcutaneous injection. Twenty-four hours after treatment, overnight fasted (16h) rats are subjected to a graded intravenous glucose infusion test. The graded glucose infusion test consists of a baseline saline infusion period (20 min), followed by two 30 min glucose infusion phases at 5 and 15 mg/kg/min, respectively. Plasma samples are collected at -20, -10 min, 0 pre-glucose infusions (baseline), and at 10, 20, 30, 40, 50, and 60 minutes. A representation of the data are shown in table 9.
Table 9
~~~~~~~~ 5mg/Kg/min 15mg/Kg/min Vehicle 4.3 ± 0.2 (n=18) 12.7 ± 0.9 (n=18) 0.0179 mg/Kg 5.6 ± 0.4 (n=4) 15.9 + 1.8 (n=4) 0.179 mg/Kg 9.0 + 1.1* (n=6) 28.0 + 3.8* (n=6) 1.79 mg/Kg 20.5 + 3.0 * (n=4) 52.7 + 7.2* (n=4) *P < 0.05 versus vehicle

Claims

We Claim: 1. A heterologous fusion protein comprising a GLP-1 analog comprising a sequence selected from the group consisting of: a) (SEQ ID NO: 1) His-Xaa8-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Val-Ser-Ser-Tyr-Leu-Glu-Glu- Gln-Ala-Ala-Lys-Glu-Phe-Ile-Ala-Trp-Leu-Val-Lys-Gly-Gly-Gly wherein Xaa8 is selected from Gly and Val; b) (SEQ ID NO:2) His-Xaa8-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Val-Ser-Ser-Tyr-Leu-Glu-Glu- Gln-Ala-Ala-Lys-Glu-Phe-Ile-Ala-Trp-Leu-Lys-Asn-Gly-Gly-Gly wherein Xaa8 is selected from Gly and Val; c) (SEQ ID NO:3) His-Xaa8-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Val-Ser-Ser-Tyr-Leu-Glu-Glu- Gln-Ala-Ala-Lys-Glu-Phe-Ile-Ala-Trp-Leu-Val-Lys-Gly-Gly-Pro wherein Xaa8 is selected from Gly and Val; d) (SEQ ID NO:4) His-Xaa8-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Val-Ser-Ser-Tyr-Leu-Glu-Glu- Gln- Ala-Ala-Lys-Glu-Phe-Ile-Ala-Trp-Leu-Lys-Asn-Gly-Gly-Pro wherein Xaag is selected from Gly and Val; e) (SEQ ID NO:5) His-Xaa8-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Val-Ser-Ser-Tyr-Leu-Glu-Glu- Gln-Ala-Ala-Lys-Glu-Phe-Ile-Ala-Trp-Leu-Val-Lys-Gly-Gly wherein Xaa8 is selected from Gly and Val; f) (SEQ ID NO:6) His-Xaa8-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Val-Ser-Ser-Tyr-Leu-Glu-Glu- Gln-Ala-Ala-Lys-Glu-Phe-Ile-Ala-Trp-Leu-Lys-Asn-Gly-Gly wherein Xaa8 is selected from Gly and Val; fused to the Fc portion of an immunoglobulin comprising the sequence of SEQ ID NO:7 Ala-Glu-Ser-Lys-Tyr-Gly-Pro-Pro-Cys-Pro-Pro-Cys-Pro-Ala-Pro- Xaaiδ-Xaaπ-Xaais-Gly-Gly-Pro-Ser-Val-Phe-Leu-Phe-Pro-Pro-Lys-Pro- Lys-Asp-Thr-Leu-Met-Ile-Ser-Arg-Thr-Pro-Glu-Val-Thr-Cys-Val- Val-Val-Asp-Val-Ser-Gln-Glu-Asp-Pro-Glu-Val-Gln-Phe-Asn-Trp- Tyr-Val-Asp-Gly-Val-Glu-Val-His-Asn-Ala-Lys-Thr-Lys-Pro-Arg- Glu-Glu-Gln-Phe-Xaaso-Ser-Thr-Tyr-Arg-Val-Val-Ser-Val-Leu-Thr- Val-Leu-His-Gln-Asp-Trp-Leu-Asn-Gly-Lys-Glu-Tyr-Lys-Cys-Lys- Val-Ser-Asn-Lys-Gly-Leu-Pro-Ser-Ser-Ile-Glu-Lys-Thr-Ile-Ser- Lys-Ala-Lys-Gly-Gln-Pro-Arg-Glu-Pro-Gln-Val-Tyr-Thr-Leu-Pro- Pro-Ser-Gln-Glu-Glu-Met-Thr-Lys-Asn-Gln-Val-Ser-Leu-Thr-Cys- Leu-Val-Lys-Gly-Phe-Tyr-Pro-Ser-Asp-Ile-Ala-Val-Glu-Trp-Glu- Ser-Asn-Gly-Gln-Pro-Glu-Asn-Asn-Tyr-Lys-Thr-Thr-Pro-Pro-Val- Leu-Asp-Ser-Asp-Gly-Ser-Phe-Phe-Leu-Tyr-Ser-Arg-Leu-Thr-Val- Asp-Lys-Ser-Arg-Trp-Gln-Glu-Gly-Asn-Val-Phe-Ser-Cys-Ser-Val- Met-His-Glu-Ala-Leu-His-Asn-His-Tyr-Thr-Gln-Lys-Ser-Leu-Ser- Leu-Ser-Leu-Gly-Xaa230 (SEQ ID NO:7) wherein: Xaa at position 16 is Pro or Glu; Xaa at position 17 is Phe, Val, or Ala; Xaa at position 18 is Leu, Glu, or Ala; Xaa at position 80 is Asn or Ala; and Xaa at position 230 is Lys or is absent.
2. The heterologous fusion protein of Claim 1 wherein the C-terminal glycine residue of the GLP-1 analog is fused to the N-terminal alanine residue of the Fc portion via a peptide linker comprising a sequence selected from the group consisting of: a) Gly-Gly-GIy-Gly-Ser-Gly-Gly-Gly-Gly-Ser-Gly-Gly-Gly-Gly-Ser (SEQ ID NO:8); b) Gly-Ser-Gly-Gly-Gly-Gly-Ser-Gly-Gly-Gly-Gly-Ser-Gly-Gly-Gly-Gly-Ser- Gly-Gly-Gly-Gly-Ser (SEQ ID NO: 19); and c) Gly-Gly-Gly-Gly-Ser-Gly-Gly-Gly-Gly-Ser-Gly-Gly-Gly-Gly-Ser-Gly-Gly- Gly-Gly-Ser-Gly-Gly-Gly-Gly-Ser-Gly-Gly-Gly-Gly-Ser (SEQ ID NO:21).
3. The heterologous fusion protein of Claim 2 wherein the linker comprises the sequence of SEQ ID NO:8.
4. The heterologous fusion protein of any one of Claims 1 to 3 wherein Xaa at position 8 of the GLP-1 analog is Gly.
5. The heterologous fusion protein of any one of Claims 1 to 3 wherein Xaa at position 8 of the GLP-1 analog is Val. 6. The heterologous fusion protein of any one of Claims 1 to 3 wherein the GLP-1 analog comprises the sequence of SEQ ID NO:l.
7. A heterologous fusion protein selected from the group consisting of: a) Gly8- Glu22-Gly36-GLP-l(7-37)-lL-IgG4 (S228P); b) Gly8-Glu2 -Gly36-GLP-l(7-37)- lL-IgG4 (S228P, F234A, L235A); c) Gly8-Glu22-Gly36-GLP-l(7-37)-lL-IgG4 (S228P, N297A); d) Gly8-Glu22-Gly36-GLP-l(7-37)-lL-IgG4 (S228P, F234A, L235A, N297A); e) Gly°-Glu22-Gly36-GLP-l(7-37)-1.5L-IgG4 (S228P); f) Gly8- Glu22-GIy36-GLP-l(7-37)-1.5L-IgG4 (S228P, F234A, L235A); g) Gly8-Glu22- Gly36-GLP-l(7-37)-1.5L-IgG4 (S228P, N297A); h) Gly8-Glu22-Gly36-GLP-1(7- 37)-1.5L-IgG4 (S228P, F234A, L235A, N297A); i) Gly8-Glu22-Gly36-GLP-1(7- 37)-2L-IgG4 (S228P); j) Gly8-Glu22-Gly36-GLP-l(7-37)-2L-IgG4 (S228P, F234A, L235A); k) Gly8-Glu22-Gly36-GLP-l(7-37)-2L-IgG4 (S228P, N297A); 1) Gly8- Glu22-Gly36-GLP-l(7-37)-2L-IgG4 (S228P, F234A, L235A, N297A); and the des- K forms thereof.
8. A heterologous fusion protein selected from the group consisting of: a) Val8- Glu22-Gly36-GLP-l(7-37)-lL-IgG4 (S228P); b) Val 8-Glu22-Gly36-GLP- 1(7-37)- lL-IgG4 (S228P, F234A, L235A); c) Val 8-Glu22-Gly36-GLP-l(7-37)-lL-IgG4 (S228P, N297A); d) Val 8-Glu22-Gly36-GLP-l(7-37)-lL-IgG4 (S228P, F234A, L235A, N297A); e) Val 8-Glu22-Gly36-GLP-l (7-37)- 1.5L-IgG4 (S228P); f) Val 8- Glu22-Gly36-GLP-l(7-37)-1.5L-IgG4 (S228P, F234A, L235A); g) Val 8-Glu22- Gly36-GLP-l(7-37)-1.5L-IgG4 (S228P, N297A); h) Val 8-Glu22-Gly36-GLP-l(7- 37)-1.5L-IgG4 (S228P, F234A, L235A, N297A); i) Val 8-Glu22-Gly36-GLP-l(7- 37)-2L-IgG4 (S228P); j) Val 8-Glu22-Gly36-GLP-l(7-37)-2L-IgG4 (S228P, F234A, L235A); k) Val 8-Glu22-Gly36-GLP-l(7-37)-2L-IgG4 (S228P, N297A); 1) Val 8-Glu22-Gly36-GLP-l(7-37)-2L-IgG4 (S228P, F234A, L235A, N297A); and the des-K forms thereof.
9. A polynucleotide encoding the heterologous fusion protein of any one of Claims 1 to 8.
10. A vector comprising the polynucleotide of Claim 9.
11. A host cell comprising the vector of Claim 10.
12. A host cell expressing at least one heterologous fusion protein of any one of Claims 1 to 8. 13. The host cell of Claim 12 wherein the host cell is a CHO cell.
14. The host cell of Claim 12 wherein the host cell is a NSO cell.
15. A process for producing a heterologous fusion protein comprising the steps of transcribing and translating a polynucleotide of Claim 9 under conditions wherein the heterologous fusion protein is expressed in detectable amounts. 16. A method of treating a patient with non- insulin dependent diabetes mellitus comprising the administration of a therapeutically effective amount of the heterologous fusion protein of any one of Claims 1 to 8.
17. A method of inducing weight loss in an overweight patient comprising the administrations of a therapeutically effective amount of the heterologous fusion protein of any one of Claims 1 to 8.
18. The method of Claim 16 or 17 wherein the heterologous fusion protein is administered at a dose between about 0.05 mg kg to 0.5 mg kg body weight.
19. The method of Claim 16 or 17 wherein the heterologous fusion protein is administered once a week. 20. Use of the heterologous fusion protein of any one of Claims 1 to 8 for use as a medicament.
21. Use of the heterologous fusion protein of any one of Claims 1 to 8 for the manufacture of a medicament to treat non- insulin dependent diabetes mellitus.
22. Use of the heterologous fusion protein of any one of Claims 1 to 8 for the manufacture of a medicament to treat obesity or induce weight loss in an overweight subject.
23. Use of the heterolgous fusion protein of any one of Claims 1 to 8 for the treatment of a human or animal body by therapy.
4. A heterologous fusion protein comprising a GLP-1 analog comprising a sequence selected from the group consisting of: a) (SEQ ID NO: 1) His-Xaa8-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Val-Ser-Ser-Tyr-Leu-Glu-Glu- Gln-Ala-Ala-Lys-Glu-Phe-Ile-Ala-Trp-Leu-Val-Lys-Gly-Gly-Gly wherein Xaa8 is selected from Gly and Val; b) (SEQ ID NO:2) His-Xaa8-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Val-Ser-Ser-Tyr-Leu-Glu-Glu- Gln-Ala-Ala-Lys-Glu-Phe-Ile-Ala-Trp-Leu-Lys-Asn-Gly-Gly-Gly wherein Xaa8 is selected from Gly and Val; c) (SEQ ID NO:3) His-Xaa8-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Val-Ser-Ser-Tyr-Leu-GIu-Glu- Gln-Ala-Ala-Lys-Glu-Phe-Ile-Ala-Trp-Leu-Val-Lys-Gly-Gly-Pro wherein Xaa8 is selected from Gly and Val; d) (SEQ ID NO:4) His-Xaa8-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Val-Ser-Ser-Tyr-Leu-Glu-Glu- Gln- Ala-Ala-Lys-Glu-Phe-Ile-Ala-Trp-Leu-Lys-Asn-Gly-Gly-Pro wherein Xaa8 is selected from Gly and Val; e) (SEQ ID NO:5) His-Xaa8-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Val-Ser-Ser-Tyr-Leu-Glu-Glu- Gln-Ala-Ala-Lys-Glu-Phe-Ile-Ala-Trp-Leu-Val-Lys-Gly-Gly wherein Xaa8 is selected from Gly and Val; f) (SEQ ID NO:6) His-Xaa8-Glu-Gly-Thr-Phe-Thr-Ser-Asp-Val-Ser-Ser-Tyr-Leu-Glu-Glu- GIn-Ala-Ala-Lys-Glu-Phe-Ile-Ala-Trp-Leu-Lys-Asn-Gly-Gly wherein Xaa8 is selected from Gly and Val; fused to the Fc portion of an immunoglobulin comprising the sequence of SEQ ID NO:7 Alai-Glu-Ser-Lys-Tyr-Gly-Pro-Pro-Cys-Pro-Pro-Cys-Pro-Ala-Pro- Xaai6-Xaaι7-Xaai8-Gly-Gly-Pro-Ser-Val-Phe-Leu-Phe-Pro-Pro-Lys-Pro- Lys-Asp-Thr-Leu-Met-Ile-Ser-Arg-Thr-Pro-Glu-Val-Thr-Cys-Val- Val-Val-Asp-Val-Ser-Gln-Glu-Asp-Pro-Glu-Val-Gln-Phe-Asn-Trp- Tyr-Val-Asp-Gly-Val-Glu-Val-His-Asn-Ala-Lys-Thr-Lys-Pro-Arg- Glu-Glu-Gln-Phe-Xaaso-Ser-Thr-Tyr-Arg-Val-Val-Ser-Val-Leu-Thr- Val-Leu-His-Gln-Asp-Trp-Leu-Asn-Gly-Lys-Glu-Tyr-Lys-Cys-Lys- Val-Ser-Asn-Lys-Gly-Leu-Pro-Ser-Ser-Ile-Glu-Lys-Thr-Ile-Ser- Lys-Ala-Lys-Gly-Gln-Pro-Arg-Glu-Pro-Gln-Val-Tyr-Thr-Leu-Pro-
Pro-Ser-Gln-Glu-Glu-Met-Thr-Lys-Asn-Gln-Val-Ser-Leu-Thr-Cys- Leu-Val-Lys-Gly-Phe-Tyr-Pro-Ser-Asp-Ile-Ala-Val-Glu-Trp-Glu- Ser-Asn-Gly-Gln-Pro-Glu-Asn-Asn-Tyr-Lys-Thr-Thr-Pro-Pro-Val- Leu-Asp-Ser-Asp-Gly-Ser-Phe-Phe-Leu-Tyr-Ser-Arg-Leu-Thr-Val- Asp-Lys-Ser-Arg-Trp-Gln-Glu-Gly-Asn-Val-Phe-Ser-Cys-Ser-Val-
Met-His-Glu-Ala-Leu-His-Asn-His-Tyr-Thr-Gln-Lys-Ser-Leu-Ser- Leu-Ser-Leu-Gly-Xaa230 (SEQ ID NO:7) wherein: Ala at position 1 is absent; Xaa at position 16 is Pro or Glu;
Xaa at position 17 is Phe, Val, or Ala;
Xaa at position 18 is Leu, Glu, or Ala;
Xaa at position 80 is Asn or Ala; and
Xaa at position 230 is Lys or is absent.
PCT/US2004/015595 2003-06-12 2004-06-10 Glp-1 analog fusion plroteins WO2005000892A2 (en)

Priority Applications (22)

Application Number Priority Date Filing Date Title
SI200431776T SI1641823T1 (en) 2003-06-12 2004-06-10 Glp-1 analog fusion proteins
MXPA05013565A MXPA05013565A (en) 2003-06-12 2004-06-10 Glp-1 analog fusion proteins
PL04752589T PL1641823T3 (en) 2003-06-12 2004-06-10 Glp-1 analog fusion plroteins
AU2004251145A AU2004251145C1 (en) 2003-06-12 2004-06-10 GLP-1 analog fusion proteins
NZ543292A NZ543292A (en) 2003-06-12 2004-06-10 GLP-1 analog fusion proteins
US10/558,627 US7452966B2 (en) 2003-06-12 2004-06-10 GLP-1 analog fusion proteins
DK04752589.4T DK1641823T3 (en) 2003-06-12 2004-06-10 GLP-1 analog fusion proteins
KR1020057023668A KR100758755B1 (en) 2003-06-12 2004-06-10 -1 glp-1 analog fusion proteins
EA200600015A EA008831B1 (en) 2003-06-12 2004-06-10 Glp-1 analog fusion proteins
JP2006533197A JP4629047B2 (en) 2003-06-12 2004-06-10 GLP-1 analog complex protein
AT04752589T ATE525395T1 (en) 2003-06-12 2004-06-10 FUSION PROTEINS OF GLP-1 ANALOGS
CN200480015953XA CN1802386B (en) 2003-06-12 2004-06-10 GLP-1 analog fusion proteins
CA2528591A CA2528591C (en) 2003-06-12 2004-06-10 Glp-1 analog fusion proteins
EP04752589A EP1641823B1 (en) 2003-06-12 2004-06-10 Glp-1 analog fusion plroteins
BRPI0411132A BRPI0411132B8 (en) 2003-06-12 2004-06-10 heterologous fusion protein and its uses
IL171926A IL171926A (en) 2003-06-12 2005-11-14 Glp-1 analog fusion proteins
US12/262,832 US8273854B2 (en) 2003-06-12 2008-10-31 GLP-1 analog fusion proteins
HR20110714T HRP20110714T1 (en) 2003-06-12 2011-10-05 Glp-1 analog fusion proteins
FR15C0010C FR15C0010I2 (en) 2003-06-12 2015-01-30 GLP-1 ANALOGUE HYBRID PROTEINS
CY2015002C CY2015002I2 (en) 2003-06-12 2015-01-30 GLP-1 FUSION PROTEIN ANALOGS
BE2015C007C BE2015C007I2 (en) 2003-06-12 2015-02-02
LTPA2015007C LTC1641823I2 (en) 2003-06-12 2015-02-03 GLP-1 analog fusion proteins

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US47788003P 2003-06-12 2003-06-12
US60/477,880 2003-06-12

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US10/558,627 A-371-Of-International US7452966B2 (en) 2003-06-12 2004-06-10 GLP-1 analog fusion proteins
US12/262,832 Division US8273854B2 (en) 2003-06-12 2008-10-31 GLP-1 analog fusion proteins

Publications (2)

Publication Number Publication Date
WO2005000892A2 true WO2005000892A2 (en) 2005-01-06
WO2005000892A3 WO2005000892A3 (en) 2005-03-03

Family

ID=33551775

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2004/015595 WO2005000892A2 (en) 2003-06-12 2004-06-10 Glp-1 analog fusion plroteins

Country Status (29)

Country Link
US (2) US7452966B2 (en)
EP (2) EP2368909A1 (en)
JP (1) JP4629047B2 (en)
KR (1) KR100758755B1 (en)
CN (2) CN101974090B (en)
AR (1) AR044776A1 (en)
AT (1) ATE525395T1 (en)
AU (1) AU2004251145C1 (en)
BE (1) BE2015C007I2 (en)
BR (1) BRPI0411132B8 (en)
CA (1) CA2528591C (en)
CY (2) CY1111991T1 (en)
DK (1) DK1641823T3 (en)
EA (1) EA008831B1 (en)
ES (1) ES2371072T3 (en)
FR (1) FR15C0010I2 (en)
HK (1) HK1149566A1 (en)
HR (1) HRP20110714T1 (en)
HU (1) HUS1500024I1 (en)
IL (1) IL171926A (en)
LT (1) LTC1641823I2 (en)
MX (1) MXPA05013565A (en)
NZ (1) NZ543292A (en)
PL (1) PL1641823T3 (en)
PT (1) PT1641823E (en)
SI (1) SI1641823T1 (en)
TW (1) TW200507870A (en)
UA (1) UA87458C2 (en)
WO (1) WO2005000892A2 (en)

Cited By (79)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006068910A1 (en) * 2004-12-22 2006-06-29 Eli Lilly And Company Glp-1 analog fusion protein formulations
WO2007012188A1 (en) * 2005-07-27 2007-02-01 Qinghua Wang GLP/1/EXENDM 4 IgG Fc FUSION CONSTRUCTS FOR TREATMENT OF DIABETES
WO2007016764A1 (en) 2005-08-06 2007-02-15 Qinghua Wang Composition and method for prevention and treatment of type i diabetes
US7189690B2 (en) 2001-12-21 2007-03-13 Human Genome Sciences, Inc. Albumin fusion proteins
WO2008077616A1 (en) 2006-12-22 2008-07-03 Csl Behring Gmbh Modified coagulation factors with prolonged in vivo half-life
WO2008081418A1 (en) * 2007-01-05 2008-07-10 Covx Technologies Ireland Limited Glucagon-like protein-1 receptor (glp-1r) agonist compounds
WO2008082274A1 (en) * 2007-01-05 2008-07-10 Hanmi Pharmaceutical Co., Ltd. An insulinotropic complex using an immunoglobulin fragment
JP2008540565A (en) * 2005-05-13 2008-11-20 イーライ リリー アンド カンパニー GLP-1 PEGylated compound
JP2009508505A (en) * 2005-09-22 2009-03-05 バイオコンパティブルズ ユーケー リミテッド GLP-1 (glucagon-like peptide-1) fusion peptide with improved resistance to peptidase
US7569384B2 (en) 2004-02-09 2009-08-04 Human Genome Sciences, Inc. Albumin fusion proteins
JP2010502734A (en) * 2006-09-06 2010-01-28 フェーズバイオ ファーマシューティカルズ,インコーポレイテッド Fusion peptide therapeutic composition
US7799759B2 (en) 2001-12-21 2010-09-21 Human Genome Sciences, Inc. Albumin fusion proteins
JP2010536396A (en) * 2007-08-29 2010-12-02 ベーリンガー インゲルハイム ファルマ ゲゼルシャフト ミット ベシュレンクテル ハフツング ウント コンパニー コマンディトゲゼルシャフト Method to increase protein titer
EP2371857A1 (en) 2010-04-01 2011-10-05 CSL Behring GmbH Factor XII inhibitors for treating interstitial lung disease
WO2011136361A1 (en) 2010-04-30 2011-11-03 株式会社 三和化学研究所 Peptide for improving in vivo stability of physiologically active substance or the like and physiologically active substance with improved in vivo stability
WO2011153965A1 (en) 2010-06-11 2011-12-15 北京精益泰翔技术发展有限公司 Fusion protein of exendin-4 and its analog, preparation method and use thereof
AU2011254001B2 (en) * 2007-01-05 2012-08-02 Covx Technologies Ireland Limited Glucagon-like protein-1 receptor (GLP-1R) agonist compounds
EP2497489A1 (en) 2011-03-09 2012-09-12 CSL Behring GmbH Factor XII inhibitors for the treatment of silent brain ischemia and ischemia of other organs
WO2012120128A1 (en) 2011-03-09 2012-09-13 Csl Behring Gmbh Factor xii inhibitors for the administration with medical procedures comprising contact with artificial surfaces
US8283319B2 (en) 2007-02-12 2012-10-09 Csl Behring Gmbh Therapeutic application of Kazal-type serine protease inhibitors
US8288339B2 (en) 2006-04-20 2012-10-16 Amgen Inc. GLP-1 compounds
WO2013113774A1 (en) 2012-01-31 2013-08-08 Csl Behring Gmbh Factor xii inhibitors for the treatment of neurological inflammatory disorders
WO2013120939A1 (en) 2012-02-15 2013-08-22 Csl Behring Gmbh Von willebrand factor variants having improved factor viii binding affinity
US8557769B2 (en) 2007-08-03 2013-10-15 Eli Lilly And Company Co-administration of FGF-21 and GLP-1 to treat diabetes and lower blood glucose
US8575104B2 (en) 2008-06-24 2013-11-05 Csl Behring Gmbh Factor VIII, von willebrand factor or complexes thereof with prolonged in vivo half-life
US8765915B2 (en) 2006-02-06 2014-07-01 Csl Behring Gmbh Modified coagulation factor VIIa with extended half-life
WO2014207199A1 (en) 2013-06-28 2014-12-31 Csl Behring Gmbh Combination therapy using a factor xii inhibitor and a c1-inhibitor
WO2015193457A1 (en) 2014-06-18 2015-12-23 Csl Behring Gmbh Therapy using a factor xii inhibitor in a neurotraumatic disorder
WO2016000039A1 (en) 2014-07-02 2016-01-07 Csl Limited Modified von willebrand factor
US9328154B2 (en) 2005-12-20 2016-05-03 Duke University Therapeutic agents comprising fusions of growth hormone and elastic peptides
EP1965823B1 (en) 2005-11-04 2016-05-18 Glaxosmithkline LLC Methods for administering hypoglycemic agents
WO2016178905A1 (en) * 2015-05-07 2016-11-10 Eli Lilly And Company Fusion proteins
WO2016188907A1 (en) 2015-05-22 2016-12-01 Csl Behring Recombinant Facility Ag Truncated von willebrand factor polypeptides for treating hemophilia
WO2016188905A1 (en) 2015-05-22 2016-12-01 Csl Behring Recombinant Facility Ag Methods for preparing modified von willebrand factor
US9518127B2 (en) 2011-07-22 2016-12-13 Csl Behring Gmbh Inhibitory anti-factor XII/XIIA monoclonal antibodies and their uses
WO2017074714A1 (en) 2015-10-26 2017-05-04 Eli Lilly And Company Glucagon receptor agonists
EP3184149A1 (en) 2015-12-23 2017-06-28 Julius-Maximilians-Universität Würzburg Soluble glycoprotein v for treating thrombotic diseases
WO2017117631A1 (en) 2016-01-07 2017-07-13 Csl Limited Mutated truncated von willebrand factor
US9821036B2 (en) 2008-06-27 2017-11-21 Duke University Therapeutic agents comprising a GLP-2 peptide and elastin-like peptides
US9879249B2 (en) 2009-02-17 2018-01-30 Redwood Bioscience, Inc. Aldehyde-tagged protein-based drug carriers and methods of use
US9878017B2 (en) 2013-04-22 2018-01-30 Csl Ltd. Covalent complex of von Willebrand Factor and factor VIII, compositions, and uses relating thereto
WO2018087271A1 (en) 2016-11-11 2018-05-17 Csl Behring Recombinant Facility Ag Truncated von willebrand factor polypeptides for extravascular administration in the treatment or prophylaxis of a blood coagulation disorder
WO2018088838A1 (en) 2016-11-10 2018-05-17 Yuhan Corporation Pharmaceutical composition for preventing or treating hepatitis, hepatic fibrosis, and hepatic cirrhosis comprising fusion proteins
WO2018087267A1 (en) 2016-11-11 2018-05-17 Csl Behring Recombinant Facility Ag Truncated von willebrand factor polypeptides for treating hemophilia
EP3351262A1 (en) 2016-12-30 2018-07-25 Istanbul Universitesi Rektorlugu Curaglutide for in treatment of prediabetes, diabetes, obesity and metabolic diseases associated thereto
WO2018234518A1 (en) 2017-06-22 2018-12-27 CSL Behring Lengnau AG Modulation of fviii immunogenicity by truncated vwf
US10183998B2 (en) 2011-01-14 2019-01-22 Redwood Bioscience, Inc. Aldehyde-tagged immunoglobulin polypeptides and methods of use thereof
US10258700B2 (en) 2005-12-20 2019-04-16 Duke University Methods and compositions for delivering active agents with enhanced pharmacological properties
US10286047B2 (en) 2013-03-08 2019-05-14 Csl Behring Gmbh Treatment and prevention of remote ischemia-reperfusion injury
US10294303B2 (en) 2015-12-23 2019-05-21 Amgen Inc. Method of treating or ameliorating metabolic disorders using binding proteins for gastric inhibitory peptide receptor (GIPR) in combination with GLP-1 agonists
CN111269312A (en) * 2018-12-04 2020-06-12 鲁南制药集团股份有限公司 Heterologous fusion protein
US10808023B2 (en) 2016-01-07 2020-10-20 CSL Behring Lengnau AG Mutated von Willebrand factor
US10829535B2 (en) 2013-03-14 2020-11-10 Amgen Inc. Interleukin-2 muteins for the expansion of T-regulatory cells
US10836813B2 (en) 2012-09-12 2020-11-17 Genzyme Corporation Fc containing polypeptides with altered glycosylation and reduced effector function
WO2020234195A1 (en) 2019-05-17 2020-11-26 Universitaet Zuerich Haptoglobin for use in treating an adverse secondary neurological outcome following a haemorrhagic stroke
US10851144B2 (en) 2015-04-10 2020-12-01 Amgen Inc. Interleukin-2 muteins for the expansion of T-regulatory cells
WO2021001522A1 (en) 2019-07-04 2021-01-07 CSL Behring Lengnau AG A truncated von willebrand factor (vwf) for increasing the in vitro stability of coagulation factor viii
US10905772B2 (en) 2017-01-17 2021-02-02 Amgen Inc. Method of treating or ameliorating metabolic disorders using GLP-1 receptor agonists conjugated to antagonists for gastric inhibitory peptide receptor (GIPR)
WO2021076620A1 (en) 2019-10-15 2021-04-22 Eli Lilly And Company Recombinantly engineered, lipase/esterase-deficient mammalian cell lines
WO2021094344A1 (en) 2019-11-11 2021-05-20 CSL Behring Lengnau AG Polypeptides for inducing tolerance to factor viii
US11123438B2 (en) 2016-08-19 2021-09-21 Ampsource Biopharma Shanghai Inc. Linker peptide for constructing fusion protein
US11136364B2 (en) 2015-10-28 2021-10-05 Yuhan Corporation Dual function proteins comprising FGF21 mutant protein and pharmaceutical composition comprising same
US11142557B2 (en) 2015-10-28 2021-10-12 Yuhan Corporation Long-acting FGF21 fusion proteins and pharmaceutical composition comprising same
US11174321B2 (en) 2016-04-06 2021-11-16 Csl Limited Method of treating atherosclerosis
US11186638B2 (en) 2011-09-12 2021-11-30 Genzyme Corporation Anti-αβTCR antibody
US11208632B2 (en) 2016-04-26 2021-12-28 R.P. Scherer Technologies, Llc Antibody conjugates and methods of making and using the same
EP3889185A3 (en) * 2014-03-31 2022-01-26 Hanmi Pharm. Co., Ltd. Composition for improving the solubility of a protein or peptide by using immunoglobulin fc fragment linkage
WO2022046815A1 (en) 2020-08-24 2022-03-03 The Trustees Of The University Of Pennsylvania Viral vectors encoding glp-1 receptor agonist fusions and uses thereof in treating metabolic diseases
WO2022109124A1 (en) 2020-11-20 2022-05-27 Csl Behring Gmbh Method for treating antibody-mediated rejection
WO2022162218A1 (en) 2021-02-01 2022-08-04 Csl Behring Ag Method of treating or preventing an adverse secondary neurological outcome following a haemorrhagic stroke
US11434271B2 (en) 2011-11-04 2022-09-06 Hanmi Science Co., Ltd. Method for preparing physiologically active polypeptide complex
US11472863B2 (en) 2016-08-19 2022-10-18 Ampsource Biopharma Shanghai Inc. Human coagulation factor IX (FIX) fusion protein, preparation method therefor, and use thereof
US11471513B2 (en) 2016-08-19 2022-10-18 Ampsource Biopharma Shanghai Inc. Highly glycosylated human blood-clotting factor VIII fusion protein, and manufacturing method and application of same
WO2022234070A1 (en) 2021-05-07 2022-11-10 Csl Behring Ag Expression system for producing a recombinant haptoglobin (hp) beta chain
US11560416B2 (en) 2017-04-21 2023-01-24 Yuhan Corporation Method for producing dual function proteins and its derivatives
US11697690B2 (en) 2014-03-19 2023-07-11 Genzyme Corporation Site-specific glycoengineering of targeting moieties
US11807690B2 (en) 2013-03-11 2023-11-07 Genzyme Corporation Hyperglycosylated binding polypeptides
WO2024047219A1 (en) 2022-09-02 2024-03-07 Csl Behring Ag Haptoglobin for use in treating or preventing exaggerated erectile response or erectile dysfunction
WO2024068848A1 (en) 2022-09-28 2024-04-04 Zealand Pharma A/S Methods for treating obesity

Families Citing this family (85)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7183387B1 (en) 1999-01-15 2007-02-27 Genentech, Inc. Polypeptide variants with altered effector function
US7459540B1 (en) 1999-09-07 2008-12-02 Amgen Inc. Fibroblast growth factor-like polypeptides
KR100988949B1 (en) * 2001-10-25 2010-10-20 제넨테크, 인크. Glycoprotein compositions
US20070161087A1 (en) * 2003-05-29 2007-07-12 Wolfgang Glaesner Glp-1 fusion proteins
PL1641823T3 (en) * 2003-06-12 2012-02-29 Lilly Co Eli Glp-1 analog fusion plroteins
CA2627444A1 (en) * 2005-10-24 2007-06-14 Centocor, Inc. Glp-2 mimetibodies, polypeptides, compositions, methods and uses
ES2507098T3 (en) 2005-11-07 2014-10-14 Indiana University Research And Technology Corporation Glucagon analogs showing physiological solubility and stability
US7812121B2 (en) * 2006-08-31 2010-10-12 Baker Audrey E GLP-2 mimetibodies, polypeptides, compositions, methods and uses
US8338376B2 (en) * 2006-10-20 2012-12-25 Biogen Idec Ma Inc. Compositions comprising variant LT-B-R-IG fusion proteins
AU2008205229B2 (en) * 2007-01-05 2014-03-27 Indiana University Research And Technology Corporation Glucagon analogs exhibiting enhanced solubility in physiological pH buffers
CA2677932A1 (en) 2007-02-15 2008-08-21 Indiana University Research And Technology Corporation Glucagon/glp-1 receptor co-agonists
JP5431171B2 (en) * 2007-03-15 2014-03-05 バイオジェン・アイデック・エムエイ・インコーポレイテッド Treatment of autoimmune disorders
CA2696615A1 (en) * 2007-06-19 2008-12-24 Otsuka Chemical Co., Ltd. Oligosaccharide chain added glp-1 peptide
US7960336B2 (en) * 2007-08-03 2011-06-14 Pharmain Corporation Composition for long-acting peptide analogs
US8563527B2 (en) * 2007-08-20 2013-10-22 Pharmain Corporation Oligonucleotide core carrier compositions for delivery of nucleic acid-containing therapeutic agents, methods of making and using the same
JP5771005B2 (en) * 2007-10-30 2015-08-26 インディアナ ユニバーシティー リサーチ アンド テクノロジー コーポレーションIndiana University Research And Technology Corporation Glucagon antagonist and compound showing GLP-1 agonist activity
CA2707861A1 (en) 2007-10-30 2009-05-07 Indiana University Research And Technology Corporation Glucagon antagonists
US20100317057A1 (en) * 2007-12-28 2010-12-16 Novo Nordisk A/S Semi-recombinant preparation of glp-1 analogues
JP2011511778A (en) * 2008-01-30 2011-04-14 インディアナ ユニバーシティー リサーチ アンド テクノロジー コーポレーション Ester-based peptide prodrugs
JOP20190083A1 (en) 2008-06-04 2017-06-16 Amgen Inc Fgf21 mutant fusion polypeptides and uses thereof
MX2010012695A (en) * 2008-06-17 2011-03-15 Univ Indiana Res & Tech Corp Glucagon analogs exhibiting enhanced solubility and stability in physiological ph buffers.
PL2300035T3 (en) * 2008-06-17 2016-04-29 Univ Indiana Res & Tech Corp Gip-based mixed agonists for treatment of metabolic disorders and obesity
EP2300037B1 (en) 2008-06-17 2016-03-30 Indiana University Research and Technology Corporation Glucagon/glp-1 receptor co-agonists
UA105016C2 (en) 2008-10-10 2014-04-10 Амген Інк. Fgf21 mutants and uses thereof
SG172291A1 (en) 2008-12-19 2011-07-28 Univ Indiana Res & Tech Corp Amide based glucagon superfamily peptide prodrugs
BRPI1011404B1 (en) 2009-05-05 2022-05-03 Amgen Inc Mutant fgf21 polypeptides, fusion polypeptide, multimer, pharmaceutical composition, isolated nucleic acid, vector and host cell
AU2010246038A1 (en) 2009-05-05 2011-12-01 Amgen Inc. FGF21 mutants and uses thereof
KR20120087875A (en) 2009-06-16 2012-08-07 인디애나 유니버시티 리서치 앤드 테크놀로지 코퍼레이션 Gip receptor-active glucagon compounds
WO2010148142A1 (en) * 2009-06-17 2010-12-23 Amgen Inc. Chimeric fgf19 polypeptides and uses thereof
CN101993485B (en) 2009-08-20 2013-04-17 重庆富进生物医药有限公司 Peptide analog homologous dimer capable of accelerating insulin secretion and application thereof
WO2011043530A1 (en) * 2009-10-09 2011-04-14 (주)알테오젠 Glp-1 analog fusions, and composition for preventing or treating diabetes containing the fusions as an active ingredient
EP2506861A1 (en) * 2009-12-02 2012-10-10 Amgen Inc. Binding proteins that bind to human fgfr1c, human b-klotho and both human fgfr1c and human b-klotho
UA109888C2 (en) 2009-12-07 2015-10-26 ANTIBODY OR ANTIBODILITY ANTIBODY OR ITS BINDING TO THE β-CLOTE, FGF RECEPTORS AND THEIR COMPLEXES
US8703701B2 (en) 2009-12-18 2014-04-22 Indiana University Research And Technology Corporation Glucagon/GLP-1 receptor co-agonists
IN2012DN06437A (en) 2010-01-27 2015-10-09 Univ Indiana Res & Tech Corp
US9517264B2 (en) 2010-04-15 2016-12-13 Amgen Inc. Human FGF receptor and β-Klotho binding proteins
RU2604067C2 (en) 2010-05-13 2016-12-10 Индиана Юниверсити Рисерч Энд Текнолоджи Корпорейшн Glucagon superfamily peptides exhibiting nuclear hormone receptor activity
CN103179976A (en) 2010-05-13 2013-06-26 印第安纳大学研究及科技有限公司 Glucagon superfamily peptides exhibiting g protein-coupled receptor activity
KR20130102470A (en) 2010-06-24 2013-09-17 인디애나 유니버시티 리서치 앤드 테크놀로지 코퍼레이션 Amide based glucagon superfamily peptide prodrugs
US9655974B2 (en) 2010-07-20 2017-05-23 Novo Nordisk A/S N-terminal modified FGF21 compounds
US8507428B2 (en) 2010-12-22 2013-08-13 Indiana University Research And Technology Corporation Glucagon analogs exhibiting GIP receptor activity
RS57895B1 (en) 2011-03-29 2019-01-31 Roche Glycart Ag Antibody fc variants
US8729017B2 (en) 2011-06-22 2014-05-20 Indiana University Research And Technology Corporation Glucagon/GLP-1 receptor co-agonists
MX347703B (en) 2011-06-22 2017-05-09 Univ Indiana Res & Tech Corp Glucagon/glp-1 receptor co-agonists.
JP6324315B2 (en) 2011-11-17 2018-05-16 インディアナ ユニバーシティー リサーチ アンド テクノロジー コーポレーションIndiana University Research And Technology Corporation Glucagon superfamily of peptides exhibiting glucocorticoid receptor activity
CA2877358A1 (en) 2012-06-21 2013-12-27 Indiana University Research And Technology Corporation Glucagon analogs exhibiting gip receptor activity
CN104428315B (en) 2012-07-13 2017-09-29 罗氏格黎卡特股份公司 Bispecific anti-vegf/antibody of anti-ANG 2 and its application in treatment Ocular Vessels disease
DK3366705T3 (en) * 2012-09-12 2023-07-31 Genzyme Corp FC-CONTAINING POLYPEPTIDES WITH ALTERED GLYCOSYRATION AND REDUCED EFFECTOR FUNCTION
JP6026002B2 (en) 2012-11-27 2016-11-16 アルテオジェン インコーポレイテッド Composition for stabilizing a fusion protein in which a protein and an Fc domain are fused
RU2680581C2 (en) 2012-11-27 2019-02-22 Байомарин Фармасьютикал Инк. Targeted therapeutic lysosomal enzyme fusion proteins and uses thereof
EP2970511B1 (en) 2013-03-14 2020-09-30 Indiana University Research and Technology Corporation Insulin-incretin conjugates
CN103408669B (en) * 2013-08-01 2016-01-20 江苏泰康生物医药有限公司 GLP-1 analog fusion, and its production and use
CN104592381A (en) * 2013-10-31 2015-05-06 江苏万邦生化医药股份有限公司 Preparation method of liraglutide intermediate polypeptide
PT3172227T (en) 2014-07-21 2019-12-06 Delinia Inc Molecules that selectively activate regulatory t cells for the treatment of autoimmune diseases
HUE051414T2 (en) 2014-08-11 2021-03-01 Delinia Inc Modified il-2 variants that selectively activate regulatory t cells for the treatment of autoimmune diseases
ES2822994T3 (en) 2014-09-24 2021-05-05 Univ Indiana Res & Tech Corp Incretin-insulin conjugates
CN104293834B (en) * 2014-10-11 2018-03-23 上海兴迪金生物技术有限公司 GLP 1 or its analog and antibody Fc fragment fusion protein preparation method
JP6727210B2 (en) 2014-12-23 2020-07-22 ノヴォ ノルディスク アー/エス FGF21 derivative and use thereof
KR101825048B1 (en) * 2014-12-31 2018-02-05 주식회사 제넥신 Fusion Polypeptide Comprising GLP and Immunoglobulin Hybrid Fc and use thereof
US20170204154A1 (en) 2016-01-20 2017-07-20 Delinia, Inc. Molecules that selectively activate regulatory t cells for the treatment of autoimmune diseases
US10336812B2 (en) * 2016-05-10 2019-07-02 Janssen Biotech, Inc. GDF15 fusion proteins and uses thereof
AU2017359172A1 (en) 2016-11-08 2019-05-16 Delinia, Inc. IL-2 variants for the treatment of autoimmune diseases
FI3630164T3 (en) 2017-06-01 2023-11-06 Lilly Co Eli Dulaglutide for the treatment of chronic kidney disease
MX2020002977A (en) 2017-09-22 2020-11-06 Regeneron Pharma Glucagon-like peptide 1 receptor agonists and uses thereof.
BR112020007817A2 (en) 2017-11-21 2020-10-06 Eli Lilly And Company method of use and compositions containing dulaglutide
CN109929806B (en) 2017-12-19 2020-05-08 北京吉源生物科技有限公司 Stem cell expressing GLP1 and FGF21 and application thereof
WO2019125003A1 (en) * 2017-12-22 2019-06-27 케이비바이오메드 주식회사 Oral gene carrier and use thereof
WO2019140021A1 (en) 2018-01-12 2019-07-18 Eli Lilly And Company Combination therapy
US11679143B2 (en) 2018-02-08 2023-06-20 Sunshine Lake Pharma Co., Ltd. FGF21 variant, fusion protein and application thereof
KR20200135618A (en) 2019-05-23 2020-12-03 ㈜ 디앤디파마텍 Pharmaceutical composition for preventing or treating nonalcoholic fatty liver disease comprising a polypeptide
CN112912095A (en) 2018-07-19 2021-06-04 D&D制药技术股份有限公司 Pharmaceutical compositions comprising polypeptides
CN110878127B (en) 2018-09-06 2022-06-28 浙江柏拉阿图医药科技有限公司 Long-acting recombinant GLP1-Fc-CD47 protein and preparation and application thereof
CN111234000B (en) * 2018-11-28 2023-05-26 鲁南制药集团股份有限公司 Ai Saina peptide analogues
CN109836486B (en) * 2019-01-30 2020-09-08 北京双因生物科技有限公司 Fibroblast growth factor 21 variants, fusion proteins thereof, and uses thereof
US20220064244A1 (en) * 2019-03-05 2022-03-03 Sunshine Lake Pharma Co., Ltd. A polypeptide molecule and application thereof
JP2022523972A (en) 2019-03-08 2022-04-27 アムジエン・インコーポレーテツド Proliferative differentiation factor 15 combination therapy
CN113613629A (en) 2019-03-15 2021-11-05 伊莱利利公司 Preserved formulations
CA3177693A1 (en) 2019-04-05 2020-10-05 Eli Lilly And Company Therapeutic uses of dulaglutide
CN113728013B (en) 2020-01-11 2022-06-14 北京质肽生物医药科技有限公司 Conjugates of fusion proteins of GLP-1 and FGF21
CN114106194B (en) * 2020-08-31 2024-01-16 中国科学院天津工业生物技术研究所 Fusion protein for treating diabetes and/or obesity
CN114685644A (en) 2020-12-29 2022-07-01 苏州康宁杰瑞生物科技有限公司 Human GLP-1 polypeptide variant and application thereof
AU2022308120A1 (en) 2021-07-06 2024-02-08 Suzhou Alphamab Co., Ltd. Fusion protein and application thereof
WO2023225534A1 (en) 2022-05-18 2023-11-23 Protomer Technologies Inc. Aromatic boron-containing compounds and related insulin analogs
CN114774496B (en) * 2022-06-21 2022-10-04 北京惠之衡生物科技有限公司 Method for preparing GLP-1 analogue through high-density fermentation
WO2024064842A1 (en) 2022-09-21 2024-03-28 Regeneron Pharmaceuticals, Inc. Methods of treating obesity, diabetes, and liver dysfunction

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996004388A1 (en) 1994-07-29 1996-02-15 Smithkline Beecham Plc Novel compounds
WO2002046227A2 (en) 2000-12-07 2002-06-13 Eli Lilly And Company Glp-1 fusion proteins

Family Cites Families (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4399216A (en) 1980-02-25 1983-08-16 The Trustees Of Columbia University Processes for inserting DNA into eucaryotic cells and for producing proteinaceous materials
NZ201705A (en) 1981-08-31 1986-03-14 Genentech Inc Recombinant dna method for production of hepatitis b surface antigen in yeast
US4943529A (en) 1982-05-19 1990-07-24 Gist-Brocades Nv Kluyveromyces as a host strain
AU3145184A (en) 1983-08-16 1985-02-21 Zymogenetics Inc. High expression of foreign genes in schizosaccharomyces pombe
US4879231A (en) 1984-10-30 1989-11-07 Phillips Petroleum Company Transformation of yeasts of the genus pichia
GB8610600D0 (en) 1986-04-30 1986-06-04 Novo Industri As Transformation of trichoderma
DE3883899T3 (en) 1987-03-18 1999-04-22 Sb2 Inc CHANGED ANTIBODIES.
US5010182A (en) 1987-07-28 1991-04-23 Chiron Corporation DNA constructs containing a Kluyveromyces alpha factor leader sequence for directing secretion of heterologous polypeptides
AU4005289A (en) 1988-08-25 1990-03-01 Smithkline Beecham Corporation Recombinant saccharomyces
FR2646437B1 (en) 1989-04-28 1991-08-30 Transgene Sa NOVEL DNA SEQUENCES, THEIR APPLICATION AS A SEQUENCE ENCODING A SIGNAL PEPTIDE FOR THE SECRETION OF MATURE PROTEINS BY RECOMBINANT YEASTS, EXPRESSION CASSETTES, PROCESSED YEASTS AND PROCESS FOR PREPARING THE SAME
DE394538T1 (en) 1989-04-28 1991-04-11 Rhein Biotech Ges. Fuer Biotechnologische Prozesse Und Produkte Mbh, 4000 Duesseldorf, De YEAR CELLS OF THE SCHWANNIOMYCES GENERATION.
EP0402226A1 (en) 1989-06-06 1990-12-12 Institut National De La Recherche Agronomique Transformation vectors for yeast yarrowia
FR2649120B1 (en) 1989-06-30 1994-01-28 Cayla NOVEL STRAIN AND ITS MUTANTS OF FILAMENTOUS MUSHROOMS, PROCESS FOR PRODUCING RECOMBINANT PROTEINS USING SAID STRAIN, AND STRAINS AND PROTEINS OBTAINED BY SAID METHOD
JP3560609B2 (en) 1992-11-13 2004-09-02 イミュネックス・コーポレーション A novel cytokine called ELK ligand
GB9511935D0 (en) 1995-06-13 1995-08-09 Smithkline Beecham Plc Novel compound
US5723125A (en) 1995-12-28 1998-03-03 Tanox Biosystems, Inc. Hybrid with interferon-alpha and an immunoglobulin Fc linked through a non-immunogenic peptide
US6750334B1 (en) 1996-02-02 2004-06-15 Repligen Corporation CTLA4-immunoglobulin fusion proteins having modified effector functions and uses therefor
US6277819B1 (en) 1996-08-30 2001-08-21 Eli Lilly And Company Use of GLP-1 or analogs in treatment of myocardial infarction
US6006753A (en) 1996-08-30 1999-12-28 Eli Lilly And Company Use of GLP-1 or analogs to abolish catabolic changes after surgery
UA65549C2 (en) 1996-11-05 2004-04-15 Елі Ліллі Енд Компані Use of glucagon-like peptides such as glp-1, glp-1 analog, or glp-1 derivative in methods and compositions for reducing body weight
US6190909B1 (en) 1997-04-17 2001-02-20 Millennium Pharmaceuticals, Inc. TH2-specific gene
SE9802080D0 (en) 1998-06-11 1998-06-11 Hellstroem Pharmaceutical composition for the treatment of functional dyspepsia and / or irritable bowel syndrome and new use of substances therein
EP1932535A3 (en) 1998-07-31 2008-10-29 Novo Nordisk A/S Stimulation of beta cell profileration
MY155270A (en) 1998-09-24 2015-09-30 Lilly Co Eli Use of glp-1 or analogs in treatment of stroke
US6376653B1 (en) 1998-09-28 2002-04-23 Smithkline Beecham Plc Tie2 antagonist antibodies
WO2000069911A1 (en) 1999-05-17 2000-11-23 Conjuchem, Inc. Long lasting insulinotropic peptides
KR100847615B1 (en) 2000-06-16 2008-07-21 일라이 릴리 앤드 캄파니 Glucagon-like Peptide-1 Analogs
US6992174B2 (en) 2001-03-30 2006-01-31 Emd Lexigen Research Center Corp. Reducing the immunogenicity of fusion proteins
US6900292B2 (en) 2001-08-17 2005-05-31 Lee-Hwei K. Sun Fc fusion proteins of human erythropoietin with increased biological activities
EP2261250B1 (en) 2001-12-21 2015-07-01 Human Genome Sciences, Inc. GCSF-Albumin fusion proteins
PL1641823T3 (en) * 2003-06-12 2012-02-29 Lilly Co Eli Glp-1 analog fusion plroteins

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1996004388A1 (en) 1994-07-29 1996-02-15 Smithkline Beecham Plc Novel compounds
WO2002046227A2 (en) 2000-12-07 2002-06-13 Eli Lilly And Company Glp-1 fusion proteins

Cited By (130)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7238660B2 (en) 2001-12-21 2007-07-03 Human Genome Sciences, Inc. Albumin fusion proteins
US7799759B2 (en) 2001-12-21 2010-09-21 Human Genome Sciences, Inc. Albumin fusion proteins
US7977306B2 (en) 2001-12-21 2011-07-12 Human Genome Sciences, Inc. Albumin fusion proteins
US7189690B2 (en) 2001-12-21 2007-03-13 Human Genome Sciences, Inc. Albumin fusion proteins
US7569384B2 (en) 2004-02-09 2009-08-04 Human Genome Sciences, Inc. Albumin fusion proteins
KR100879662B1 (en) * 2004-12-22 2009-01-20 일라이 릴리 앤드 캄파니 Glp-1 analog fusion protein formulations
EP2168982A1 (en) * 2004-12-22 2010-03-31 Eli Lilly &amp; Company GLP-1 analog fusion protein formulations
EA011166B1 (en) * 2004-12-22 2009-02-27 Эли Лилли Энд Компани Glp-1 analog fusion protein formulations
WO2006068910A1 (en) * 2004-12-22 2006-06-29 Eli Lilly And Company Glp-1 analog fusion protein formulations
JP2008540565A (en) * 2005-05-13 2008-11-20 イーライ リリー アンド カンパニー GLP-1 PEGylated compound
US8658174B2 (en) 2005-07-27 2014-02-25 Qinghua Wang GLP/1/exendin 4 IgG Fc fusion constructs for treatment of diabetes
WO2007012188A1 (en) * 2005-07-27 2007-02-01 Qinghua Wang GLP/1/EXENDM 4 IgG Fc FUSION CONSTRUCTS FOR TREATMENT OF DIABETES
CN101273134B (en) * 2005-07-27 2012-01-04 王庆华 Glp/1/exendin 4 igg fc fusion constructs for treatment of diabetes and method
WO2007016764A1 (en) 2005-08-06 2007-02-15 Qinghua Wang Composition and method for prevention and treatment of type i diabetes
JP2009508505A (en) * 2005-09-22 2009-03-05 バイオコンパティブルズ ユーケー リミテッド GLP-1 (glucagon-like peptide-1) fusion peptide with improved resistance to peptidase
EP1965823B1 (en) 2005-11-04 2016-05-18 Glaxosmithkline LLC Methods for administering hypoglycemic agents
US9328154B2 (en) 2005-12-20 2016-05-03 Duke University Therapeutic agents comprising fusions of growth hormone and elastic peptides
US10258700B2 (en) 2005-12-20 2019-04-16 Duke University Methods and compositions for delivering active agents with enhanced pharmacological properties
US8765915B2 (en) 2006-02-06 2014-07-01 Csl Behring Gmbh Modified coagulation factor VIIa with extended half-life
US8288339B2 (en) 2006-04-20 2012-10-16 Amgen Inc. GLP-1 compounds
JP2010502734A (en) * 2006-09-06 2010-01-28 フェーズバイオ ファーマシューティカルズ,インコーポレイテッド Fusion peptide therapeutic composition
US8754194B2 (en) 2006-12-22 2014-06-17 Csl Behring Gmbh Modified coagulation factors with prolonged in vivo half-life
EP3231440A1 (en) 2006-12-22 2017-10-18 CSL Behring GmbH Modified coagulation factors with prolonged in vivo half-life
WO2008077616A1 (en) 2006-12-22 2008-07-03 Csl Behring Gmbh Modified coagulation factors with prolonged in vivo half-life
AU2011254001B2 (en) * 2007-01-05 2012-08-02 Covx Technologies Ireland Limited Glucagon-like protein-1 receptor (GLP-1R) agonist compounds
JP2010514835A (en) * 2007-01-05 2010-05-06 コヴェックス・テクノロジーズ・アイルランド・リミテッド Glucagon-like protein 1 receptor (GLP-1R) agonist compound
WO2008081418A1 (en) * 2007-01-05 2008-07-10 Covx Technologies Ireland Limited Glucagon-like protein-1 receptor (glp-1r) agonist compounds
WO2008082274A1 (en) * 2007-01-05 2008-07-10 Hanmi Pharmaceutical Co., Ltd. An insulinotropic complex using an immunoglobulin fragment
AU2008203641B2 (en) * 2007-01-05 2011-10-06 Covx Technologies Ireland Limited Glucagon-like protein-1 receptor (GLP-1R) agonist compounds
KR101224335B1 (en) * 2007-01-05 2013-01-25 씨오브이엑스 테크놀로지스 아일랜드 리미티드 Glucagon-like protein-1 receptor (glp-1r) agonist compounds
US8476230B2 (en) 2007-01-05 2013-07-02 Hanmi Science Co., Ltd Insulinotropic complex using an immunoglobulin fragment
US8283319B2 (en) 2007-02-12 2012-10-09 Csl Behring Gmbh Therapeutic application of Kazal-type serine protease inhibitors
EP2526962A1 (en) 2007-02-12 2012-11-28 CSL Behring GmbH Therapeutic application of Kazal-type serine protease inhibitors
US8557769B2 (en) 2007-08-03 2013-10-15 Eli Lilly And Company Co-administration of FGF-21 and GLP-1 to treat diabetes and lower blood glucose
JP2010536396A (en) * 2007-08-29 2010-12-02 ベーリンガー インゲルハイム ファルマ ゲゼルシャフト ミット ベシュレンクテル ハフツング ウント コンパニー コマンディトゲゼルシャフト Method to increase protein titer
US8575104B2 (en) 2008-06-24 2013-11-05 Csl Behring Gmbh Factor VIII, von willebrand factor or complexes thereof with prolonged in vivo half-life
EP2865760A1 (en) 2008-06-24 2015-04-29 CSL Behring GmbH Factor VIII, von Willebrand factor or complexes thereof with prolonged in vivo half-life
US9290561B2 (en) 2008-06-24 2016-03-22 Csl Behring Gmbh Factor VIII, von Willebrand factor or complexes thereof with prolonged in vivo half-life
US10596230B2 (en) 2008-06-27 2020-03-24 Duke University Methods of increasing nutrient absorption in the intestine using therapeutic agents comprising GLP-2 and elastin-like peptides
US11103558B2 (en) 2008-06-27 2021-08-31 Duke University Therapeutic agents comprising a BMP-9 peptide and eleastin-like peptides
US9821036B2 (en) 2008-06-27 2017-11-21 Duke University Therapeutic agents comprising a GLP-2 peptide and elastin-like peptides
US9879249B2 (en) 2009-02-17 2018-01-30 Redwood Bioscience, Inc. Aldehyde-tagged protein-based drug carriers and methods of use
US9150638B2 (en) 2010-04-01 2015-10-06 Justus-Liebig-Universität Giessen Factor XII inhibitors for treating interstitial lung disease
EP2371857A1 (en) 2010-04-01 2011-10-05 CSL Behring GmbH Factor XII inhibitors for treating interstitial lung disease
WO2011121123A1 (en) 2010-04-01 2011-10-06 Csl Behring Gmbh Factor xii inhibitors for treating interstitial lung disease
WO2011136361A1 (en) 2010-04-30 2011-11-03 株式会社 三和化学研究所 Peptide for improving in vivo stability of physiologically active substance or the like and physiologically active substance with improved in vivo stability
WO2011153965A1 (en) 2010-06-11 2011-12-15 北京精益泰翔技术发展有限公司 Fusion protein of exendin-4 and its analog, preparation method and use thereof
US10183998B2 (en) 2011-01-14 2019-01-22 Redwood Bioscience, Inc. Aldehyde-tagged immunoglobulin polypeptides and methods of use thereof
US9987328B2 (en) 2011-03-09 2018-06-05 Csl Behring Gmbh Factor XII inhibitors for the administration with medical procedures comprising contact with artificial surfaces
US9624307B2 (en) 2011-03-09 2017-04-18 The General Hospital Corporation Factor XII inhibitors for the treatment of silent brain ischemia and ischemia of other organs
WO2012120124A1 (en) 2011-03-09 2012-09-13 Csl Behring Gmbh Factor xii inhibitors for the treatment of silent brain ischemia and ischemia of other organs
WO2012120128A1 (en) 2011-03-09 2012-09-13 Csl Behring Gmbh Factor xii inhibitors for the administration with medical procedures comprising contact with artificial surfaces
US9352016B2 (en) 2011-03-09 2016-05-31 Csl Behring Gmbh Factor XII inhibitors for the administration with medical procedures comprising contact with artificial surfaces
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
US9856325B2 (en) 2011-07-22 2018-01-02 Csl Behring Gmbh Nucleic acids encoding inhibitory anti-factor XII/XIIa monoclonal antibodies
US9518127B2 (en) 2011-07-22 2016-12-13 Csl Behring Gmbh Inhibitory anti-factor XII/XIIA monoclonal antibodies and their uses
US10513560B2 (en) 2011-07-22 2019-12-24 Csl Behring Gmbh Methods of administering inhibitory anti-factor XII/XIIA monoclonal antibodies
US9856326B2 (en) 2011-07-22 2018-01-02 Csl Behring Gmbh Methods of administering inhibitory anti-factor XII/XIIA monoclonal antibodies
US11345759B2 (en) 2011-07-22 2022-05-31 Csl Behring Gmbh Methods of administering inhibitory anti-factor XII/XIIa monoclonal antibodies
US11186638B2 (en) 2011-09-12 2021-11-30 Genzyme Corporation Anti-αβTCR antibody
US11434271B2 (en) 2011-11-04 2022-09-06 Hanmi Science Co., Ltd. Method for preparing physiologically active polypeptide complex
US9957329B2 (en) 2012-01-31 2018-05-01 Csl Behring Gmbh Factor XII inhibitors for the treatment of neurological inflammatory disorders
WO2013113774A1 (en) 2012-01-31 2013-08-08 Csl Behring Gmbh Factor xii inhibitors for the treatment of neurological inflammatory disorders
US9458223B2 (en) 2012-02-15 2016-10-04 Csl Behring Gmbh Von willebrand factor variants having improved factor VIII binding affinity
WO2013120939A1 (en) 2012-02-15 2013-08-22 Csl Behring Gmbh Von willebrand factor variants having improved factor viii binding affinity
US10836813B2 (en) 2012-09-12 2020-11-17 Genzyme Corporation Fc containing polypeptides with altered glycosylation and reduced effector function
US10286047B2 (en) 2013-03-08 2019-05-14 Csl Behring Gmbh Treatment and prevention of remote ischemia-reperfusion injury
US10973891B2 (en) 2013-03-08 2021-04-13 Csl Behring Gmbh Treatment and prevention of remote ischemia-reperfusion injury
US11807690B2 (en) 2013-03-11 2023-11-07 Genzyme Corporation Hyperglycosylated binding polypeptides
US10829535B2 (en) 2013-03-14 2020-11-10 Amgen Inc. Interleukin-2 muteins for the expansion of T-regulatory cells
US9878017B2 (en) 2013-04-22 2018-01-30 Csl Ltd. Covalent complex of von Willebrand Factor and factor VIII, compositions, and uses relating thereto
WO2014207199A1 (en) 2013-06-28 2014-12-31 Csl Behring Gmbh Combination therapy using a factor xii inhibitor and a c1-inhibitor
US11697690B2 (en) 2014-03-19 2023-07-11 Genzyme Corporation Site-specific glycoengineering of targeting moieties
EP3889185A3 (en) * 2014-03-31 2022-01-26 Hanmi Pharm. Co., Ltd. Composition for improving the solubility of a protein or peptide by using immunoglobulin fc fragment linkage
WO2015193457A1 (en) 2014-06-18 2015-12-23 Csl Behring Gmbh Therapy using a factor xii inhibitor in a neurotraumatic disorder
US10253088B2 (en) 2014-07-02 2019-04-09 CSL Behring Lengnau AG Modified von Willebrand Factor
WO2016000039A1 (en) 2014-07-02 2016-01-07 Csl Limited Modified von willebrand factor
US10851144B2 (en) 2015-04-10 2020-12-01 Amgen Inc. Interleukin-2 muteins for the expansion of T-regulatory cells
EA039770B1 (en) * 2015-05-07 2022-03-11 Эли Лилли Энд Компани Fusion protein for treating diabetes mellitus
WO2016178905A1 (en) * 2015-05-07 2016-11-10 Eli Lilly And Company Fusion proteins
US10688157B2 (en) 2015-05-22 2020-06-23 CSL Behring Lengnau AG Truncated von Willebrand factor polypeptides for treating hemophilia
EP4089109A2 (en) 2015-05-22 2022-11-16 CSL Behring Lengnau AG Methods for preparing modified von willebrand factor
US10772936B2 (en) 2015-05-22 2020-09-15 CSL Behring Lengnau AG Methods for preparing modified von Willebrand factor
US10905747B2 (en) 2015-05-22 2021-02-02 CSL Behring Lengnau AG Methods for preparing modified von Willebrand factor
US11564976B2 (en) 2015-05-22 2023-01-31 CSL Behring Lengnau AG Methods for preparing modified von Willebrand Factor
WO2016188907A1 (en) 2015-05-22 2016-12-01 Csl Behring Recombinant Facility Ag Truncated von willebrand factor polypeptides for treating hemophilia
WO2016188905A1 (en) 2015-05-22 2016-12-01 Csl Behring Recombinant Facility Ag Methods for preparing modified von willebrand factor
WO2017074714A1 (en) 2015-10-26 2017-05-04 Eli Lilly And Company Glucagon receptor agonists
US9884093B2 (en) 2015-10-26 2018-02-06 Eli Lilly And Company Glucagon receptor agonists
WO2017074715A1 (en) 2015-10-26 2017-05-04 Eli Lilly And Company Glucagon receptor agonists
US9764004B2 (en) 2015-10-26 2017-09-19 Eli Lilly And Company Glucagon receptor agonists
US11136364B2 (en) 2015-10-28 2021-10-05 Yuhan Corporation Dual function proteins comprising FGF21 mutant protein and pharmaceutical composition comprising same
US11142557B2 (en) 2015-10-28 2021-10-12 Yuhan Corporation Long-acting FGF21 fusion proteins and pharmaceutical composition comprising same
US11028144B2 (en) 2015-12-23 2021-06-08 Julius-Maximilians-Universität Würzburg Soluble glycoprotein V for treating thrombotic diseases
US11046774B2 (en) 2015-12-23 2021-06-29 Amgen Inc. Method of treating or ameliorating metabolic disorders using binding proteins for gastric inhibitory peptide receptor (GIPR) in combination with GLP-1 agonists
EP3184149A1 (en) 2015-12-23 2017-06-28 Julius-Maximilians-Universität Würzburg Soluble glycoprotein v for treating thrombotic diseases
US10294303B2 (en) 2015-12-23 2019-05-21 Amgen Inc. Method of treating or ameliorating metabolic disorders using binding proteins for gastric inhibitory peptide receptor (GIPR) in combination with GLP-1 agonists
WO2017117631A1 (en) 2016-01-07 2017-07-13 Csl Limited Mutated truncated von willebrand factor
US10808023B2 (en) 2016-01-07 2020-10-20 CSL Behring Lengnau AG Mutated von Willebrand factor
US10806774B2 (en) 2016-01-07 2020-10-20 CSL Behring Lengnau AG Mutated truncated von Willebrand Factor
US11174321B2 (en) 2016-04-06 2021-11-16 Csl Limited Method of treating atherosclerosis
US11208632B2 (en) 2016-04-26 2021-12-28 R.P. Scherer Technologies, Llc Antibody conjugates and methods of making and using the same
US11788066B2 (en) 2016-04-26 2023-10-17 R.P. Scherer Technologies, Llc Antibody conjugates and methods of making and using the same
US11833212B2 (en) 2016-08-19 2023-12-05 Ampsource Biopharma Shanghai Inc. Linker peptide for constructing fusion protein
US11123438B2 (en) 2016-08-19 2021-09-21 Ampsource Biopharma Shanghai Inc. Linker peptide for constructing fusion protein
US11472863B2 (en) 2016-08-19 2022-10-18 Ampsource Biopharma Shanghai Inc. Human coagulation factor IX (FIX) fusion protein, preparation method therefor, and use thereof
US11471513B2 (en) 2016-08-19 2022-10-18 Ampsource Biopharma Shanghai Inc. Highly glycosylated human blood-clotting factor VIII fusion protein, and manufacturing method and application of same
WO2018088838A1 (en) 2016-11-10 2018-05-17 Yuhan Corporation Pharmaceutical composition for preventing or treating hepatitis, hepatic fibrosis, and hepatic cirrhosis comprising fusion proteins
US11179440B2 (en) 2016-11-10 2021-11-23 Yuhan Corporation Pharmaceutical composition containing FGF21 mutant fusion protein and method for treating hepatitis, hepatic fibrosis, and hepatic cirrhosis
WO2018087271A1 (en) 2016-11-11 2018-05-17 Csl Behring Recombinant Facility Ag Truncated von willebrand factor polypeptides for extravascular administration in the treatment or prophylaxis of a blood coagulation disorder
WO2018087267A1 (en) 2016-11-11 2018-05-17 Csl Behring Recombinant Facility Ag Truncated von willebrand factor polypeptides for treating hemophilia
US11890327B2 (en) 2016-11-11 2024-02-06 CSL Behring Lengnau AG Truncated von Willebrand factor polypeptides for extravascular administration in the treatment or prophylaxis of a blood coagulation disorder
US11814421B2 (en) 2016-11-11 2023-11-14 CSL Behring Lengnau AG Truncated von Willebrand Factor polypeptides for treating hemophilia
EP3351262A1 (en) 2016-12-30 2018-07-25 Istanbul Universitesi Rektorlugu Curaglutide for in treatment of prediabetes, diabetes, obesity and metabolic diseases associated thereto
US10905772B2 (en) 2017-01-17 2021-02-02 Amgen Inc. Method of treating or ameliorating metabolic disorders using GLP-1 receptor agonists conjugated to antagonists for gastric inhibitory peptide receptor (GIPR)
US11560416B2 (en) 2017-04-21 2023-01-24 Yuhan Corporation Method for producing dual function proteins and its derivatives
US11141466B2 (en) 2017-06-22 2021-10-12 CSL Behring Lengnau AG Modulation of FVIII immunogenicity by truncated VWF
WO2018234518A1 (en) 2017-06-22 2018-12-27 CSL Behring Lengnau AG Modulation of fviii immunogenicity by truncated vwf
CN111269312A (en) * 2018-12-04 2020-06-12 鲁南制药集团股份有限公司 Heterologous fusion protein
CN111269312B (en) * 2018-12-04 2023-05-09 鲁南制药集团股份有限公司 Heterologous fusion protein
WO2020234195A1 (en) 2019-05-17 2020-11-26 Universitaet Zuerich Haptoglobin for use in treating an adverse secondary neurological outcome following a haemorrhagic stroke
WO2021001522A1 (en) 2019-07-04 2021-01-07 CSL Behring Lengnau AG A truncated von willebrand factor (vwf) for increasing the in vitro stability of coagulation factor viii
WO2021076620A1 (en) 2019-10-15 2021-04-22 Eli Lilly And Company Recombinantly engineered, lipase/esterase-deficient mammalian cell lines
WO2021094344A1 (en) 2019-11-11 2021-05-20 CSL Behring Lengnau AG Polypeptides for inducing tolerance to factor viii
WO2022046815A1 (en) 2020-08-24 2022-03-03 The Trustees Of The University Of Pennsylvania Viral vectors encoding glp-1 receptor agonist fusions and uses thereof in treating metabolic diseases
WO2022109124A1 (en) 2020-11-20 2022-05-27 Csl Behring Gmbh Method for treating antibody-mediated rejection
WO2022162218A1 (en) 2021-02-01 2022-08-04 Csl Behring Ag Method of treating or preventing an adverse secondary neurological outcome following a haemorrhagic stroke
WO2022234070A1 (en) 2021-05-07 2022-11-10 Csl Behring Ag Expression system for producing a recombinant haptoglobin (hp) beta chain
WO2024047219A1 (en) 2022-09-02 2024-03-07 Csl Behring Ag Haptoglobin for use in treating or preventing exaggerated erectile response or erectile dysfunction
WO2024068848A1 (en) 2022-09-28 2024-04-04 Zealand Pharma A/S Methods for treating obesity

Also Published As

Publication number Publication date
HUS1500024I1 (en) 2017-04-28
EP1641823B1 (en) 2011-09-21
KR20060022262A (en) 2006-03-09
WO2005000892A3 (en) 2005-03-03
KR100758755B1 (en) 2007-09-14
LTC1641823I2 (en) 2016-12-12
HK1149566A1 (en) 2011-10-07
CY2015002I1 (en) 2015-11-04
BRPI0411132B1 (en) 2016-12-13
CY2015002I2 (en) 2017-04-05
AU2004251145C1 (en) 2011-04-14
EP1641823A2 (en) 2006-04-05
ES2371072T3 (en) 2011-12-27
AU2004251145A1 (en) 2005-01-06
IL171926A (en) 2010-12-30
BE2015C007I2 (en) 2023-08-09
HRP20110714T1 (en) 2011-11-30
CY1111991T1 (en) 2015-11-04
CA2528591A1 (en) 2005-01-06
FR15C0010I2 (en) 2015-07-24
AR044776A1 (en) 2005-10-05
EA008831B1 (en) 2007-08-31
US8273854B2 (en) 2012-09-25
US7452966B2 (en) 2008-11-18
CN1802386A (en) 2006-07-12
US20090074769A1 (en) 2009-03-19
FR15C0010I1 (en) 2015-03-13
MXPA05013565A (en) 2006-03-09
JP4629047B2 (en) 2011-02-09
UA87458C2 (en) 2009-07-27
ATE525395T1 (en) 2011-10-15
DK1641823T3 (en) 2011-12-12
SI1641823T1 (en) 2011-12-30
IL171926A0 (en) 2006-04-10
BRPI0411132A (en) 2006-07-18
PT1641823E (en) 2011-11-08
PL1641823T3 (en) 2012-02-29
TW200507870A (en) 2005-03-01
EP2368909A1 (en) 2011-09-28
CA2528591C (en) 2013-01-08
BRPI0411132B8 (en) 2021-05-25
JP2007536902A (en) 2007-12-20
NZ543292A (en) 2008-04-30
AU2004251145B2 (en) 2010-09-09
CN1802386B (en) 2010-12-15
CN101974090B (en) 2015-06-17
CN101974090A (en) 2011-02-16
US20070036806A1 (en) 2007-02-15
EA200600015A1 (en) 2006-06-30

Similar Documents

Publication Publication Date Title
US7452966B2 (en) GLP-1 analog fusion proteins
EP1641483B1 (en) Fusion proteins
AU2002226897B2 (en) GLP-1 fusion proteins
US20070161087A1 (en) Glp-1 fusion proteins
CN106496331B (en) FSH-Fc fusion protein and preparation method and application thereof

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

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

AL Designated countries for regional patents

Kind code of ref document: A2

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

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

Ref document number: 543292

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 2004251145

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 171926

Country of ref document: IL

ENP Entry into the national phase

Ref document number: 2004251145

Country of ref document: AU

Date of ref document: 20040610

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2004251145

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2007036806

Country of ref document: US

Ref document number: 10558627

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 2528591

Country of ref document: CA

Ref document number: 02522/KOLNP/2005

Country of ref document: IN

Ref document number: 2522/KOLNP/2005

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2004815953X

Country of ref document: CN

WWE Wipo information: entry into national phase

Ref document number: 1020057023668

Country of ref document: KR

Ref document number: 2006533197

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: PA/a/2005/013565

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 2004752589

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 200600015

Country of ref document: EA

WWP Wipo information: published in national office

Ref document number: 1020057023668

Country of ref document: KR

WWP Wipo information: published in national office

Ref document number: 2004752589

Country of ref document: EP

ENP Entry into the national phase

Ref document number: PI0411132

Country of ref document: BR

WWP Wipo information: published in national office

Ref document number: 10558627

Country of ref document: US