US20060177443A1 - Integrin antagonists - Google Patents

Integrin antagonists Download PDF

Info

Publication number
US20060177443A1
US20060177443A1 US11/392,436 US39243606A US2006177443A1 US 20060177443 A1 US20060177443 A1 US 20060177443A1 US 39243606 A US39243606 A US 39243606A US 2006177443 A1 US2006177443 A1 US 2006177443A1
Authority
US
United States
Prior art keywords
adam
seq
amino acids
disintegrin domain
integrin
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/392,436
Inventor
William Fanslow
Douglas Cerretti
Kurt Poindexter
Roy Black
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Immunex Corp
Original Assignee
Immunex Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Immunex Corp filed Critical Immunex Corp
Priority to US11/392,436 priority Critical patent/US20060177443A1/en
Publication of US20060177443A1 publication Critical patent/US20060177443A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/48Hydrolases (3) acting on peptide bonds (3.4)
    • C12N9/50Proteinases, e.g. Endopeptidases (3.4.21-3.4.25)
    • C12N9/64Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue
    • C12N9/6421Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue from mammals
    • C12N9/6489Metalloendopeptidases (3.4.24)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • A61P27/06Antiglaucoma agents or miotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs 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
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • 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
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • This invention relates to methods and compositions that are useful for antagonizing the interaction between integrins and their ligands.
  • the invention relates to the use of ADAM disintegrin domains for antagonizing the interaction between integrins and their ligands.
  • Integrins are a family of cell surface proteins that mediate adhesion between cells (cell-cell adhesion) and between cells and extracellular matrix proteins (cell-ECM adhesion). Integrins are heterodimeric structures composed of noncovalently bound ⁇ and ⁇ subunits. In humans, at least fifteen different ⁇ subunits and eight different ⁇ subunits combine to form integrins with diverse biological activities and ligand specificities. Integrins play important roles in biological processes including embryonic development, platelet aggregation, immune reactions, tissue repair and remodeling, bone resorption, and tumor invasion and metastasis. Integrins are, therefore, important targets for therapeutic intervention in human disease.
  • the disintegrins are a family of low molecular weight, soluble, cysteine-rich peptides which have been isolated from snake venom (reviewed in Niewiarowski et al., Seminars in Hematology 31(4):289, 1994).
  • the snake venom disintegrins typically contain an RGD (Arg-Gly-Asp, SEQ ID NO:19) motif.
  • the RGD motif is recognized by many integrins, and is present in several integrin ligands including fibronectin, vitronectin, and von Willebrand factor. Disintegrins disrupt normal adhesion processes by inhibiting the binding of cell surface integrins to their ligands.
  • Disintegrin-like domains have been identified in cellular proteins from both invertebrates and vertebrates (see, e.g., Westcamp and Blobel, Proc. Natl. Acad. Sci. USA 91:2748, 1994; Wolfsberg et al., Dev. Biol. 169:378, 1995; Alfandari et al., Dev. Biol. 182:314, 1997), including the ADAM family of transmembrane proteins.
  • the ADAMs which have also been called MDCs, are a family of type I transmembrane cysteine-rich glycoproteins (Weskamp et al., Proc. Natl. Acad. Sci. USA, 91:2748, 1994; Wolfsberg et al., Dev. Biol. 169:378, 1995).
  • the multidomain structure of the ADAMs typically includes an amino-terminal metalloprotease domain, a disintegrin domain, a cysteine-rich region (the region between the disintegrin domain and the transmembrane domain), a transmembrane region, and a cytoplasmic domain. At least 30 ADAM family members have been identified, in a variety of animal species.
  • ADAMs The structure of the ADAMs suggests that they may be involved in a variety of biological processes, including cell adhesion, cell fusion, signal transduction, and proteolysis.
  • Members of the ADAM family have, in fact, been shown to play roles in sperm-egg binding and fusion, myotube formation, neurogenesis, and proteolysis.
  • ADAM-15 also called MDC-15 or metargidin
  • MDC-15 is the only ADAM identified to date which contains an RGD motif within its disintegrin domain.
  • Zhang et al. J. Biol. Chem. 273(13):7345, 1998) have reported that the isolated disintegrin domain of ADAM-15, expressed in E. coli as a glutathione S-transferase fusion protein, specifically interacts with ⁇ v ⁇ 3 integrin and that the interaction is mediated by the RGD tripeptide sequence.
  • the recombinant fusion protein did not interact with other integrins tested, including ⁇ IIb ⁇ 3 and ⁇ 5 ⁇ 1 . Nath et al. (J.
  • Angiogenesis the generation of new blood vessels, is a spatially and temporally regulated process in which endothelial and smooth muscle cells proliferate, migrate, and assemble into tubes, in response to endogenous positive and negative regulatory molecules. Angiogenesis plays important roles in both normal and pathological physiology.
  • angiogenesis is involved in fetal and embryonic development, wound healing, organ regeneration, and female reproductive remodeling processes including formation of the endometrium, corpus luteum, and placenta.
  • Angiogenesis is stringently regulated under normal conditions, especially in adult animals, and perturbation of the regulatory controls can lead to pathological angiogenesis.
  • Pathological angiogenesis has been implicated in the manifestation and/or progression of inflammatory diseases, certain eye disorders, and cancer.
  • angiogenesis is essential for the growth and persistence of solid tumors and their metastases (see, e.g., Folkman, N. Engl. J. Med. 285:1182, 1971; Folkman et al., Nature 339:58, 1989; Kim et al., Nature 362:841, 1993; Hori et al., Cancer Res., 51:6180, 1991; Zetter, Annu. Rev. Med. 49:407, 1998).
  • angiogenesis inhibitors are presently being developed and tested for the prevention (e.g., treatment of premalignant conditions), intervention (e.g., treatment of small tumors), and regression (e.g., treatment of large tumors) of cancers (see, e.g., Bergers et al., Science 284:808, 1999) and other forms of pathological angiogenesis. Because many steps in the angiogenic process, including endothelial cell migration, proliferation, and morphogenesis require vascular cell adhesion, certain integrin antagonists have been tested as anti-angiogenic agents.
  • ⁇ v ⁇ 3 integrins are expressed on the surface of cultured endothelial and smooth muscle cells, including ⁇ v ⁇ 3 integrin.
  • the ⁇ v ⁇ 3 integrin is an endothelial cell receptor for von Willebrand factor, fibrin, fibrinogen, and fibronectin, and a marker of angiogenic vascular tissue. Brooks et al. have reported that monoclonal antibodies to ⁇ v ⁇ 3 integrin, as well as cyclic peptide inhibitors, disrupt angiogenesis and that ⁇ v ⁇ 3 antibodies promote tumor regression (Science 264:569, 1994; Cell 79:1157, 1994). These results suggest that ⁇ v ⁇ 3 integrin is a useful therapeutic target for diseases characterized by pathological angiogenesis.
  • compositions and methods of antagonizing the interaction between integrins and their ligands there is great need for additional compositions and methods of antagonizing the interaction between integrins and their ligands.
  • additional compositions and methods of inhibiting angiogenesis for the prevention, abrogation, and mitigation of disease processes that are dependent upon pathological angiogenesis.
  • the present invention is based upon the discovery that ADAM disintegrin domains are useful for inhibiting the biological activity of integrins and for inhibiting endothelial cell migration and angiogenesis, including the unexpected discovery that these inhibitory activities reside in ADAM disintegrin domains that lack an RGD motif.
  • the invention is directed to methods of antagonizing the binding of an integrin to its ligands, and thereby inhibiting the biological activity of the integrin, comprising contacting the integrin with an effective amount of an ADAM disintegrin domain polypeptide.
  • the invention is further directed to methods of inhibiting endothelial cell migration and methods of inhibiting angiogenesis comprising administering an effective amount of an ADAM disintegrin domain polypeptide.
  • the ADAM disintegrin domain polypeptide is in the form of a multimer, preferably a leucine zipper multimer or Fc polypeptide.
  • the ADAM disintegrin domain is from a human ADAM, and preferably from ADAM-8, ADAM-9, ADAM-10, ADAM-15, ADAM-17, ADAM-20, ADAM-21, ADAM-22, ADAM-23, or ADAM-29.
  • the ADAM disintegrin domain is preferably produced in a recombinant cell, and is preferably present in a composition comprising a pharmaceutically acceptable carrier.
  • the ADAM disintegrin domain polypeptide comprises an amino acid sequence selected from the group consisting of: amino acids 23-264 of SEQ ID NO:2, amino acids 23-303 of SEQ ID NO:4, amino acids 23-235 of SEQ ID NO:6, amino acids 23-292 of SEQ ID NO:8, amino acids 23-216 of SEQ ID NO:10, amino acids 23-305 of SEQ ID NO:12, amino acids 23-293 of SEQ ID NO:14, amino acids 23-312 of SEQ ID NO:16, amino acids 23-310 of SEQ ID NO:18, and amino acids 23-298 of SEQ ID NO:22.
  • the ADAM disintegrin domain polypeptide comprises an amino acid sequence selected from the group consisting of: amino acids 34-91 of SEQ ID NO:2, amino acids 34-92 of SEQ ID NO:4, amino acids 34-99 of SEQ ID NO:6, amino acids 34-92 of SEQ ID NO:8, amino acids 34-93 of SEQ ID NO:10, amino acids 34-91 of SEQ ID NO:12, amino acids 34-91 of SEQ ID NO:14, amino acids 34-92 of SEQ ID NO:16, amino acids 34-91 of SEQ ID NO:18, and amino acids 34-91 of SEQ ID NO:22.
  • the ADAM disintegrin domain polypeptide comprises an amino acid sequence selected from the group consisting of: amino acids 78-91 of SEQ ID NO:2, amino acids 79-92 of SEQ ID NO:4, amino acids 87-99 of SEQ ID NO:6, amino acids 79-92 of SEQ ID NO:8, amino acids 79-93 of SEQ ID NO:10, amino acids 78-91 of SEQ ID NO:12, amino acids 78-91 of SEQ ID NO:14, amino acids 79-92 of SEQ ID NO:16, amino acids 78-91 of SEQ ID NO:18, and amino acids 78-91 of SEQ ID NO:22.
  • a therapeutically effective amount of the ADAM disintegrin domain is administered to a mammal in need of such treatment.
  • the mammal is afflicted with a condition mediated by angiogenesis, an ocular disorder, malignant or metastatic condition, inflammatory disease, osteoporosis and other conditions mediated by accelerated bone resorption, restenosis, inappropriate platelet activation, recruitment, or aggregation, thrombosis, or a condition requiring tissue repair or wound healing.
  • the ADAM disintegrin domain is, in some embodiments, administered in combination with radiation therapy and/or in combination with one or more additional therapeutic agents.
  • the invention also encompasses methods for identifying compounds that modulate integrin biological activity, that modulate the interaction between an integrin and an ADAM disintegrin domain, that inhibit endothelial cell migration, or that inhibit angiogenesis, comprising combining a test compound with an integrin or with endothelial cells and with an ADAM disintegrin domain polypeptide that binds to the integrin or endothelial cells and determining whether the test compound alters the binding of the ADAM disintegrin domain polypeptide to the integrin or endothelial cells.
  • 4-1BB and 4-1BB ligand (4-1BB-L) are polypeptides described, inter alia, in U.S. Pat. No. 5,674,704, including soluble forms thereof.
  • ADAMs are a family of transmembrane glycoproteins having disintegrin and metalloproteinase domains, also called MDC, metalloprotease/disintegrin/cysteine-rich proteins.
  • Disintegrin domain is a disintegrin domain
  • ADAMdis is an ADAM disintegrin domain
  • CD40 ligand (CD40L) is a polypeptide described, inter alia, in U.S. Pat. No. 5,716,805, including soluble forms thereof.
  • CD148 is a protein tyrosine phosphatase, also called DEP-1, ECRTP, and PTPRJ. CD148 binding proteins are described in Daniel et al., PCT Publication No. WO 00/15258, 23 Mar. 2000.
  • DMEM Dulbecco's Modified Eagle Medium
  • FACS Fluorescence activated cell sorting
  • Flt3L is Flt3 ligand, a polypeptide described, inter alia, in U.S. Pat. No. 5,554,512, including soluble forms thereof.
  • HRMEC human renal microvascular endothelial cells.
  • HMVEC-d are human dermal microvascular endothelial cells.
  • mAb is a monoclonal antibody.
  • MDC is a family of cysteine-rich proteins having metalloprotease and disintegrin domains, also called ADAM.
  • Nectin-3 is a cell adhesion molecule in the nectin family (which is described, inter alia, in Satoh-Horikawa et al., J. Biol. Chem. 275(14):10291, 2000).
  • GenBank accession numbers of human nectin-3 nucleic acid and polypeptide sequences are AF282874 and AAF97597 respectively (Reymond et al., 2000).
  • PMA is phorbol-12-myristate-13-acetate.
  • Tek which has also been called Tie2 and ork, is an receptor tyrosine kinase (RTK) that is predominantly expressed in vascular endothelium.
  • RTK receptor tyrosine kinase
  • the molecular cloning of human Tek (ork) has been described by Ziegler, U.S. Pat. No. 5,447,860.
  • Tek antagonists are described, inter alia, in Cerretti et al., PCT Publication No. WO 00/75323, 14 Dec. 2000.
  • TNF tumor necrosis factor
  • TNFR tumor necrosis factor receptor
  • TNFR/Fc tumor necrosis factor receptor-Fc fusion polypeptide
  • TRAIL is TNF-related apoptosis-inducing ligand, a type II transmembrane polypeptide in the TNF family described, inter alia, in U.S. Pat. No. 5,763,223, including soluble forms thereof.
  • TWEAK is TNF-weak effector of apoptosis, a type II transmembrane polypeptide in the TNF family described, inter alia, in Chicheportiche et al., J. Biol. Chem., 272(51):32401, 1997, including soluble forms thereof.
  • TWEAK-R is the “TWEAK receptor,” which is described, inter alia, in U.S. Ser. Nos. 60/172,878 and 60/203,347 and Feng et al., Am. J. Pathol. 156(4):1253, 2000, including soluble forms thereof.
  • TWEAK-R/Fc is a TWEAK receptor-Fc fusion polypeptide.
  • VEGF vascular endothelial growth factor
  • VPF vascular permeability factor
  • Table 1 provides reference information for selected human ADAMs.
  • ADAM disintegrin domains show sequence homology to the snake venom disintegrins, and are characterized by a framework of cysteines.
  • a typical disintegrin sequence comprises a framework such as: (SEQ ID NO:20) CDCGX 3-5 CX 3-6 CCX 2-4 CX 7 CX 4-6 CCX 2-4 CX 8 CX 5-7 CX 3-5 C
  • SEQ ID NO:20 CDCGX 3-5 CX 3-6 CCX 2-4 CX 7 CX 4-6 CCX 2-4 CX 8 CX 5-7 CX 3-5 C
  • the sequences of several ADAM disintegrin domains are shown in Table 2 and in the Sequence Listing.
  • the present invention encompasses the use of various forms of ADAM disintegrin domains that retain at least one activity selected from the group consisting of integrin binding activity, inhibition of endothelial cell migration, and inhibition of angiogenesis.
  • ADAM disintegrin domain polypeptide is intended to encompass polypeptides containing all or part of a native ADAM disintegrin domain, with or without other ADAM domains (such as the cysteine-rich region), as well as related forms including, but not limited to: (a) fragments, (b) variants, (c) derivatives, (d) fusion polypeptides, and (e) multimeric forms (multimers).
  • variant includes polypeptides that are substantially homologous to native ADAM disintegrin domains, but which have an amino acid sequence different from that of a native ADAM disintegrin domain because of one or more deletions, insertions or substitutions.
  • Particular embodiments include, but are not limited to, ADAM disintegrin domain polypeptides that comprise from one to ten deletions, insertions or substitutions of amino acid residues, when compared to a native ADAM disintegrin domain sequence.
  • ADAM disintegrin domain polypeptides include those variants that are naturally occurring, such as allelic forms and alternatively spliced forms, as well as variants that have been constructed by modifying the amino acid sequence of a ADAM disintegrin domain polypeptide or the nucleotide sequence of a nucleic acid encoding a ADAM disintegrin domain polypeptide.
  • substitutions for one or more amino acids present in the native polypeptide should be made conservatively.
  • conservative substitutions include substitution of amino acids outside of the active domain(s), and substitution of amino acids that do not alter the secondary and/or tertiary structure of the ADAM disintegrin domain. Additional examples include substituting one aliphatic residue for another, such as Ile, Val, Leu, or Ala for one another, or substitutions of one polar residue for another, such as between Lys and Arg; Glu and Asp; or Gln and Asn, or substitutions of one aromatic residue for another, such as Phe, Trp, or Tyr for one another.
  • Other such conservative substitutions for example, substitutions of entire regions having similar hydrophobicity characteristics, are known in the art.
  • the ADAM disintegrin domain variant is at least about 70% identical in amino acid sequence to the amino acid sequence of a native ADAM disintegrin domain; in some preferred embodiments the ADAM disintegrin domain variant is at least about 80% identical in amino acid sequence to the amino acid sequence of a native ADAM disintegrin domain. In some more preferred embodiments the ADAM disintegrin domain variant is at least about 90% identical in amino acid sequence to the amino acid sequence of a native ADAM disintegrin domain; in some more preferred embodiments the ADAM disintegrin domain variant is at least about 95% identical in amino acid sequence to the amino acid sequence of a native ADAM disintegrin domain.
  • the ADAM disintegrin domain variant is at least about 98% identical in amino acid sequence to the amino acid sequence of a native ADAM disintegrin domain; in some most preferred embodiments the ADAM disintegrin domain variant is at least about 99% identical in amino acid sequence to the amino acid sequence of a native ADAM disintegrin domain.
  • Percent identity in the case of both polypeptides and nucleic acids, may be determined by visual inspection. Percent identity may be determined using the alignment method of Needleman and Wunsch (J. Mol. Biol. 48:443, 1970) as revised by Smith and Waterman (Adv. Appl. Math 2:482, 1981. Preferably, percent identity is determined by using a computer program, for example, the GAP computer program version 10.x available from the Genetics Computer Group (GCG; Madison, Wis., see also Devereux et al., Nucl. Acids Res. 12:387, 1984).
  • GAP computer program version 10.x available from the Genetics Computer Group (GCG; Madison, Wis., see also Devereux et al., Nucl. Acids Res. 12:387, 1984).
  • the preferred default parameters for the GAP program include: (1) a unary comparison matrix (containing a value of 1 for identities and 0 for non-identities) for nucleotides, and the weighted comparison matrix of Gribskov and Burgess, Nucl. Acids Res. 14:6745, 1986, as described by Schwartz and Dayhoff, eds., Atlas of protein Sequence and Structure , National Biomedical Research Foundation, pp. 353-358, 1979 for amino acids; (2) a penalty of 30 (amino acids) or 50 (nucleotides) for each gap and an additional 1 (amino acids) or 3 (nucleotides) penalty for each symbol in each gap; (3) no penalty for end gaps; and (4) no maximum penalty for long gaps.
  • Other programs used by one skilled in the art of sequence comparison may also be used.
  • the percent identity is calculated based on that portion of ADAM disintegrin domain that is present in the fragment.
  • Subunits of the inventive polypeptides may be constructed by deleting terminal or internal residues or sequences. Additional guidance as to the types of mutations that can be made is provided by a comparison of the sequence of ADAM disintegrin domain polypeptides to polypeptides that have similar structures, as well as by performing structural analysis of the inventive polypeptides.
  • variant also includes ADAM disintegrin domain polypeptides that are encoded by nucleic acids capable of hybridizing under moderately stringent conditions (e.g., prewashing solution of 5 ⁇ SSC, 0.5% SDS, 1.0 mM EDTA (pH 8.0) and hybridization conditions of 50° C., 5 ⁇ SSC, overnight) or higher stringency conditions to DNA sequences encoding ADAM disintegrin domain polypeptides, and which encode polypeptides that retain at least one activity selected from the group consisting of integrin binding activity, inhibition of endothelial cell migration, and inhibition of angiogenesis.
  • moderately stringent conditions e.g., prewashing solution of 5 ⁇ SSC, 0.5% SDS, 1.0 mM EDTA (pH 8.0) and hybridization conditions of 50° C., 5 ⁇ SSC, overnight
  • higher stringency conditions include higher temperatures for hybridization and post-hybridization washes, and/or lower salt concentration.
  • Mutations can be introduced into nucleic acids by synthesizing oligonucleotides containing a mutant sequence, flanked by restriction sites enabling ligation to fragments of the native sequence. Following ligation, the resulting reconstructed sequence encodes a variant having the desired amino acid insertion, substitution, or deletion.
  • oligonucleotide-directed site-specific mutagenesis procedures can be employed to provide an altered gene having particular codons altered according to the substitution, deletion, or insertion required.
  • the well known polymerase chain reaction (PCR) procedure also may be employed to generate and amplify a DNA sequence encoding a desired polypeptide or fragment thereof.
  • Oligonucleotides that define the desired termini of the DNA fragment are employed as 5′ and 3′ primers.
  • the oligonucleotides may additionally contain recognition sites for restriction endonucleases to facilitate insertion of the amplified DNA fragment into an expression vector.
  • ADAM disintegrin domain polypeptides with or without associated native-pattern glycosylation.
  • ADAM disintegrin domain expressed in yeast or mammalian expression systems e.g., COS-1 or COS-7 cells
  • yeast or mammalian expression systems may be similar to or significantly different from a native ADAM disintegrin domain polypeptide in molecular weight and glycosylation pattern, depending upon the choice of expression system.
  • Expression of ADAM disintegrin domain polypeptides in bacterial expression systems, such as E. coli provides non-glycosylated molecules.
  • Different host cells may also process polypeptides differentially, resulting in heterogeneous mixtures of polypeptides with variable N- or C-termini.
  • ADAM disintegrin domain polypeptides may be modified to create derivatives by forming covalent or aggregative conjugates with other chemical moieties, such as glycosyl groups, lipids, phosphate, acetyl groups and the like.
  • Covalent derivatives of ADAM disintegrin domain polypeptides may be prepared by linking particular functional groups to ADAM disintegrin domain amino acid side chains or at the N-terminus or C-terminus of a ADAM disintegrin domain polypeptide.
  • Fusion polypeptides of ADAM disintegrin domains that are useful in practicing the invention include covalent or aggregative conjugates of ADAMdis or its fragments with other polypeptides, such as by synthesis in recombinant culture as N-terminal or C-terminal fusions.
  • One class of fusion polypeptides are discussed below in connection with ADAM disintegrin oligomers.
  • a fusion polypeptide may comprise a signal peptide (which is also variously referred to as a signal sequence, signal, leader peptide, leader sequence, or leader) at the N-terminal region or C-terminal region of an ADAM disintegrin domain polypeptide which co-translationally or post-translationally directs transfer of the polypeptide from its site of synthesis to a site inside or outside of the cell membrane or cell wall. It is particularly advantageous to fuse a signal peptide that promotes extracellular secretion to the N-terminus of a soluble ADAMdis polypeptide. In this case, the signal peptide is typically cleaved upon secretion of the soluble polypeptide from the cell.
  • Secreted soluble polypeptides may be identified (and distinguished from its non-soluble membrane-bound counterparts) by separating intact cells which express the desired polypeptide from the culture medium, e.g., by centrifugation, and assaying the medium (supernatant) for the presence of the desired polypeptide. The presence of the desired polypeptide in the medium indicates that the polypeptide was secreted from the cells and thus is a soluble form of the polypeptide.
  • Soluble polypeptides may be prepared by any of a number of conventional techniques. A DNA sequence encoding a desired soluble polypeptide may be subcloned into an expression vector for production of the polypeptide, or the desired encoding DNA fragment may be chemically synthesized.
  • Soluble ADAM disintegrin domain polypeptides comprise all or part of the ADAM disintegrin domain, with or without additional segments from the extracellular portion of the ADAM (such as the cysteine-rich region) but generally lack a transmembrane domain that would cause retention of the polypeptide at the cell surface. Soluble polypeptides may include part of the transmembrane domain or all or part of the cytoplasmic domain as long as the polypeptide is secreted from the cell in which it is produced. Examples of soluble ADAM disintegrin domain polypeptides are provided in the examples.
  • a multimeric form of a soluble ADAM disintegrin domain polypeptide is used to inhibit integrin binding to ligands and, hence, integrin biological activity.
  • the soluble ADAM disintegrin domain polypeptide is used to inhibit endothelial cell migration and/or inhibit angiogenesis. These inhibitory activities may include both integrin-mediated and integrin-independent mechanisms.
  • ADAM disintegrin domain multimers are covalently-linked or non-covalently-linked multimers, including dimers, trimers, and higher multimers. Oligomers may be linked by disulfide bonds formed between cysteine residues on different ADAM disintegrin domain polypeptides.
  • One embodiment of the invention is directed to multimers comprising multiple ADAM disintegrin domain polypeptides joined via covalent or non-covalent interactions between peptide moieties fused to the ADAM disintegrin domain polypeptides.
  • Such peptides may be peptide linkers (spacers), or peptides that have the property of promoting multimerization.
  • Leucine zippers and certain polypeptides derived from antibodies are among the peptides that can promote multimerization of ADAM disintegrin domain polypeptides attached thereto, as described in more detail below.
  • the multimers comprise from two to four ADAM disintegrin domain polypeptides.
  • a ADAM disintegrin domain multimer is prepared using polypeptides derived from immunoglobulins. Preparation of fusion proteins comprising certain heterologous polypeptides fused to various portions of antibody-derived polypeptides (including the Fc domain) has been described, e.g., by Ashkenazi et al. (Proc. Natl. Acad. Sci. USA 88:10535, 1991); Byrn et al. (Nature 344:677, 1990); and Hollenbaugh and Aruffo (“Construction of Immunoglobulin Fusion Proteins”, in Current Protocols in Immunology , Suppl. 4, pages 10.19.1-10.19.11, 1992).
  • a preferred embodiment of the present invention is directed to an ADAM disintegrin domain (ADAMdis) dimer comprising two fusion polypeptides created by fusing an ADAM disintegrin domain to an Fc polypeptide.
  • a gene fusion encoding the ADAMdis-Fc fusion polypeptide is inserted into an appropriate expression vector.
  • ADAMdis-Fc fusion polypeptides are expressed in host cells transformed with the recombinant expression vector, and allowed to assemble much like antibody molecules, whereupon interchain disulfide bonds form between the Fc moieties to yield divalent soluble ADAMdis polypeptides.
  • the term “Fc polypeptide” as used herein includes native and mutein forms of polypeptides derived from the Fc region of an antibody. Truncated forms of such polypeptides containing the hinge region that promotes dimerization are also included.
  • Fc polypeptide is a single chain polypeptide extending from the N-terminal hinge region to the native C-terminus of the Fc region of a human IgG1 antibody.
  • Another useful Fc polypeptide is the Fc mutein described in U.S. Pat. No. 5,457,035 and by Baum et al., EMBO J. 13:3992, 1994.
  • the amino acid sequence of this mutein is identical to that of the native Fc sequence presented in WO 93/10151, except that amino acid 19 has been changed from Leu to Ala, amino acid 20 has been changed from Leu to Glu, and amino acid 22 has been changed from Gly to Ala.
  • the mutein exhibits reduced affinity for Fc receptors.
  • Fusion polypeptides comprising Fc moieties, and multimers formed therefrom, offer an advantage of facile purification by affinity chromatography over Protein A or Protein G columns, and Fc fusion polypeptides may provide a longer in vivo half life, which is useful in therapeutic applications, than unmodified polypeptides.
  • a soluble ADAM disintegrin domain polypeptide may be substituted for the variable portion of an antibody heavy or light chain. If fusion proteins are made with both heavy and light chains of an antibody, it is possible to form an ADAM disintegrin domain multimer with as many as four soluble ADAM disintegrin domain polypeptides.
  • the ADAM disintegrin domain multimer is a fusion polypeptide comprising multiple ADAM disintegrin domain polypeptides, with or without peptide linkers (spacers), or peptides that have the property of promoting multimerization.
  • suitable peptide linkers are those described in U.S. Pat. Nos. 4,751,180 and 4,935,233.
  • a DNA sequence encoding a desired peptide linker may be inserted between, and in the same reading frame as, the DNA sequences encoding ADAMdis, using conventional techniques known in the art. For example, a chemically synthesized oligonucleotide encoding the linker may be ligated between sequences encoding ADAMdis.
  • a fusion protein comprises from two to four ADAM disintegrin domain polypeptides, separated by peptide linkers.
  • Leucine zipper domains are peptides that promote multimerization of the proteins in which they are found. Leucine zippers were originally identified in several DNA-binding proteins (Landschulz et al., Science 240:1759, 1988), and have since been found in a variety of different proteins. Among the known leucine zippers are naturally occurring peptides and derivatives thereof that dimerize or trimerize. Examples of leucine zipper domains suitable for producing soluble oligomeric proteins are described in PCT application WO 94/10308, and the leucine zipper derived from lung surfactant protein D (SPD) described in Hoppe et al. FEBS Lett.
  • SPD lung surfactant protein D
  • the ADAM disintegrin domain polypeptides used in the present invention may be prepared using a recombinant expression system. Host cells transformed with a recombinant expression vector encoding the ADAM disintegrin domain polypeptide are cultured under conditions that promote expression of ADAM disintegrin domain and the ADAM disintegrin domain is recovered. ADAM disintegrin domain polypeptides can also be produced in transgenic plants or animals.
  • Recombinant expression vectors include DNA encoding an ADAM disintegrin domain polypeptide operably linked to suitable transcriptional and translational regulatory nucleotide sequences, such as those derived from a mammalian, microbial, viral, or insect gene. Nucleotide sequences are operably linked when the regulatory sequence functionally relates to the ADAM disintegrin domain DNA sequence.
  • a promoter nucleotide sequence is operably linked to an ADAM disintegrin domain DNA sequence if the promoter nucleotide sequence controls the transcription of the ADAM disintegrin domain DNA sequence.
  • regulatory sequences include transcriptional promoters, operators, or enhancers, an mRNA ribosomal binding site, and appropriate sequences which control transcription and translation initiation and termination.
  • a sequence encoding an appropriate signal peptide can be incorporated into expression vectors.
  • a DNA sequence for a signal peptide secretory leader may be fused in frame to the ADAM disintegrin domain sequence so that the ADAM disintegrin domain polypeptide is initially translated as a fusion protein comprising the signal peptide.
  • a signal peptide that is functional in the intended host cells promotes extracellular secretion of the ADAM disintegrin domain polypeptide.
  • the signal peptide is cleaved from the ADAM disintegrin domain polypeptide upon secretion from the cell.
  • Suitable host cells for expression of ADAM disintegrin domain polypeptides include prokaryotes, yeast and higher eukaryotic cells, including insect and mammalian cells. Appropriate cloning and expression vectors for use with bacterial, fungal, yeast, insect, and mammalian cellular hosts are known in the art.
  • ADAM disintegrin domain polypeptides comprising various additions or substitutions of amino acid residues or sequences, or deletions of terminal or internal residues or sequences, including ADAM disintegrin domain fragments, variants, derivatives, multimers, and fusion polypeptides.
  • ADAM disintegrin domain polypeptides may be purified using methods known in the art, including one or more concentration, salting-out, ion exchange, hydrophobic interaction, affinity purification, HPLC, or size exclusion chromatography steps. Fusion polypeptides comprising Fc moieties (and multimers formed therefrom) offer the advantage of facile purification by affinity chromatography over Protein A or Protein G columns.
  • the disclosed methods may be used to inhibit integrin binding and integrin biological activity, and to inhibit endothelial cell migration, and/or angiogenesis in a mammal in need of such treatment.
  • the treatment is advantageously administered in order to prevent the onset or the recurrence of a disease or condition mediated by an integrin, or to treat a mammal that has a disease or condition mediated by an integrin.
  • ADAM disintegrin domain polypeptides and compositions thereof include the treatment of individuals afflicted with conditions mediated by angiogenesis such as ocular disorders, dermatological disorders, and malignant or metastatic conditions, inflammatory diseases, osteoporosis and other conditions mediated by accelerated bone resorption, restenosis, inappropriate platelet activation, recruitment, or aggregation, thrombosis, or a condition requiring tissue repair or wound healing.
  • angiogenesis such as ocular disorders, dermatological disorders, and malignant or metastatic conditions, inflammatory diseases, osteoporosis and other conditions mediated by accelerated bone resorption, restenosis, inappropriate platelet activation, recruitment, or aggregation, thrombosis, or a condition requiring tissue repair or wound healing.
  • ocular disorders that can be treated according to the present invention are eye diseases characterized by ocular neovascularization including, but not limited to, diabetic retinopathy (a major complication of diabetes), retinopathy of prematurity (this devastating eye condition, that frequently leads to chronic vision problems and carries a high risk of blindness, is a severe complication during the care of premature infants), neovascular glaucoma, retinoblastoma, retrolental fibroplasia, rubeosis, uveitis, macular degeneration, and corneal graft neovascularization.
  • Other eye inflammatory diseases, ocular tumors, and diseases associated with choroidal or iris neovascularization can also be treated according to the present invention.
  • the present invention can also be used to treat malignant and metastatic conditions such as solid tumors.
  • Solid tumors include both primary and metastatic sarcomas and carcinomas.
  • the present invention can also be used to treat inflammatory diseases including, but not limited to, arthritis, rheumatism, inflammatory bowel disease, and psoriasis.
  • thrombosis Among the conditions mediated by inappropriate platelet activation, recruitment, aggregation, or thrombosis that can be treated according to the present invention are coronary artery disease or injury, myocardial infarction or injury following myocardial infarction, stroke, unstable angina, atherosclerosis, arteriosclerosis, preeclampsia, embolism, platelet-associated ischemic disorders including lung ischemia, coronary ischemia, and cerebral ischemia, restenosis following percutaneous coronary intervention including angioplasty, atherectomy, stent placement, and bypass surgery, thrombotic disorders including coronary artery thrombosis, cerebral artery thrombosis, intracardiac thrombosis, peripheral artery thrombosis, venous thrombosis, thrombosis and coagulopathies associated with exposure to a foreign or injured tissue surface, and reocclusion following thrombosis, deep venous thrombosis (DVT), pulmonary embo
  • the methods according to the invention are used in individuals at high risk for thrombus formation or reformation, advanced coronary artery disease, or for occlusion, reocclusion, stenosis and/or restenosis of blood vessels, or stroke.
  • the methods according to the invention are used in combination with angioplasty procedures, such as balloon angioplasty, laser angioplasty, coronary atherectomy or similar techniques, carotid endarterectomy, anastomosis of vascular grafts, surgery having a high risk of thrombus formation (i.e., coronary bypass surgery, insertion of a prosthetic valve or vessel and the like), atherectomy, stent placement, placement of a chronic cardiovascular device such as an in-dwelling catheter or prosthetic valve or vessel, organ transplantation, or bypass surgery.
  • angioplasty procedures such as balloon angioplasty, laser angioplasty, coronary atherectomy or similar techniques, carotid endarterectomy, anastomosis of vascular grafts, surgery having a high risk of thrombus formation (i.e., coronary bypass surgery, insertion of a prosthetic valve or vessel and the like), atherectomy, stent placement, placement of a chronic cardiovascular device such as an in-dwelling
  • diseases and conditions that can be treated according to the present invention include benign tumors and preneoplastic conditions, myocardial angiogenesis, hemophilic joints, scleroderma, vascular adhesions, asthma and allergy, eczema and dermatitis, graft versus host disease, sepsis, adult respirator distress syndrome, telangiectasia, and wound granulation.
  • the methods according to the present invention can be tested in in vivo animal models for the desired prophylactic or therapeutic activity, as well as to determine the optimal therapeutic dosage, prior to administration to humans.
  • the amount of a particular ADAM disintegrin domain polypeptide that will be effective in a particular method of treatment depends upon age, type and severity of the condition to be treated, body weight, desired duration of treatment, method of administration, and other parameters. Effective dosages are determined by a physician or other qualified medical professional. Typical effective dosages are about 0.01 mg/kg to about 100 mg/kg body weight. In some preferred embodiments the dosage is about 0.1-50 mg/kg; in some preferred embodiments the dosage is about 0.5-10 mg/kg.
  • the dosage for local administration is typically lower than for systemic administration. In some embodiments a single administration is sufficient; in some embodiments the ADAM disintegrin domain is administered as multiple doses over one or more days.
  • the ADAM disintegrin domain polypeptides are typically administered in the form of a pharmaceutical composition comprising one or more pharmacologically acceptable carriers.
  • Pharmaceutically acceptable carriers include diluents, fillers, adjuvants, excipients, and vehicles which are pharmaceutically acceptable for the route of administration, and may be aqueous or oleaginous suspensions formulated using suitable dispersing, wetting, and suspending agents.
  • Pharmaceutically acceptable carriers are generally sterile and free of pyrogenic agents, and may include water, oils, solvents, salts, sugars and other carbohydrates, emulsifying agents, buffering agents, antimicrobial agents, and chelating agents.
  • the particular pharmaceutically acceptable carrier and the ratio of active compound to carrier are determined by the solubility and chemical properties of the composition, the mode of administration, and standard pharmaceutical practice.
  • ADAM disintegrin domain polypeptides are administered to the patient in a manner appropriate to the indication.
  • ADAM disintegrin domain polypeptides, or pharmaceutical compositions thereof may be administered by intravenous, transdermal, intradermal, intraperitoneal, intramuscular, intranasal, epidural, oral, topical, subcutaneous, intracavity, sustained release from implants, peristaltic routes, or by any other suitable technique. Parenteral administration is preferred.
  • the treatment further comprises treating the mammal with one or more additional therapeutic agents.
  • the additional therapeutic agent(s) may be administered prior to, concurrently with, or following the administration of the ADAM disintegrin domain polypeptide.
  • the use of more than one therapeutic agent is particularly advantageous when the mammal that is being treated has a solid tumor.
  • the treatment further comprises treating the mammal with radiation. Radiation, including brachytherapy and teletherapy, may be administered prior to, concurrently with, or following the administration of the ADAM disintegrin domain polypeptide and/or additional therapeutic agent(s).
  • the method includes the administration of, in addition to an ADAM disintegrin domain polypeptide, one or more therapeutics selected from the group consisting of alkylating agents, antimetabolites, vinca alkaloids and other plant-derived chemotherapeutics, antitumor antibiotics, antitumor enzymes, topoisomerase inhibitors, platinum analogs, adrenocortical suppressants, hormones and antihormones, antibodies, immunotherapeutics, radiotherapeutics, and biological response modifiers.
  • one or more therapeutics selected from the group consisting of alkylating agents, antimetabolites, vinca alkaloids and other plant-derived chemotherapeutics, antitumor antibiotics, antitumor enzymes, topoisomerase inhibitors, platinum analogs, adrenocortical suppressants, hormones and antihormones, antibodies, immunotherapeutics, radiotherapeutics, and biological response modifiers.
  • the method includes administration of, in addition to an ADAM disintegrin domain polypeptide, one or more therapeutics selected from the group consisting of cisplatin, cyclophosphamide, mechloretamine, melphalan, bleomycin, carboplatin, fluorouracil, 5-fluorodeoxyuridine, methotrexate, taxol, asparaginase, vincristine, and vinblastine, lymphokines and cytokines such as interleukins, interferons (alpha., beta.
  • one or more therapeutics selected from the group consisting of cisplatin, cyclophosphamide, mechloretamine, melphalan, bleomycin, carboplatin, fluorouracil, 5-fluorodeoxyuridine, methotrexate, taxol, asparaginase, vincristine, and vinblastine, lymphokines and cytokines such as interleukins, interferons (alpha., beta.
  • TNF chlorambucil, busulfan, carmustine, lomustine, semustine, streptozocin, dacarbazine, cytarabine, mercaptopurine, thioguanine, vindesine, etoposide, teniposide, dactinomycin, daunorubicin, doxorubicin, bleomycin, plicamycin, mitomycin, L-asparaginase, hydroxyurea, methylhydrazine, mitotane, tamoxifen, fluoxymesterone, IL-8 inhibitors, angiostatin, endostatin, kringle 5, angiopoietin-2 or other antagonists of angiopoietin-1, antagonists of platelet-activating factor, antagonists of basic fibroblast growth factor, and COX-2 inhibitors.
  • the method includes administration of, in addition to an ADAM disintegrin domain polypeptide, one or more therapeutic polypeptides, including soluble forms thereof, selected from the group consisting of Flt3 ligand, CD40 ligand, interleukin-2, interleukin-12, 4-1BB ligand, anti-4-1BB antibodies, TRAIL, TNF antagonists and TNF receptor antagonists including TNFR/Fc, Tek antagonists, TWEAK antagonists and TWEAK-R antagonists including TWEAK-R/Fc, VEGF antagonists including anti-VEGF antibodies, VEGF receptor (including VEGF-R1 and VEGF-R2, also known as Flt1 and Flk1 or KDR) antagonists, CD148 (also referred to as DEP-1, ECRTP, and PTPRJ, see Takahashi et al., J. Am. Soc. Nephrol. 10:2135-45, 1999; and PCT Publication No. WO 00/15258, 23 Mar. 2000
  • ADAM disintegrin domain polypeptides of the invention are used as a component of, or in combination with, “metronomic therapy,” such as that described by Browder et al. and Klement et al. (Cancer Research 60:1878, 2000; J. Clin. Invest. 105(8):R15, 2000; see also Barinaga, Science 288:245, 2000).
  • the terms “therapy,” “therapeutic,” “treat,” and “treatment” generally include prophylaxis, i.e. prevention, in addition to therapy or treatment for an extant disease or condition.
  • the methods of the present invention may be used as a first line treatment, for the treatment of residual disease following primary therapy, or as an adjunct to other therapies. Methods of measuring biological effectiveness are known in the art and are illustrated in the Examples below.
  • This example describes one method for the recombinant production of ADAM disintegrin domain polypeptides.
  • Expression cassettes encoding an IgKappa leader sequence, ADAM disintegrin domain, and C-terminal Fc region were constructed in bacterial plasmids then transferred into eukaryotic expression vectors (pDC409, EMBO J. 10:2821, 1991, or another mammalian expression vector).
  • the coding regions of the various constructs are summarized in Table 2.
  • these constructs encode additional portions of the extracellular portion of the ADAM (e.g., cysteine-rich region and EGF-like domain).
  • the expression vectors were transfected into COS-1, CV-1/EBNA, or 293/EBNA cells. Two days after transfection the cells were 35 S labeled for four hours. Supernatants and total cell lysates were prepared and aliquots were immunoprecipitated using protein A-sepharose beads to capture the Fc tagged polypeptides. 35 S labeled ADAM disintegrin-Fc polypeptides were run on 8-16% reducing gels and detected via autoradiography.
  • ADAM disintegrin-Fc polypeptides were purified from the supernatants using affinity chromatography (protein A column). The polypeptides were characterized by determining the N-terminal amino acid sequence, amino acid composition, and protein integrity (SDS-PAGE under reducing and non-reducing conditions) before the polypeptides were used in FACS, immunoprecipitations, and biological assays such as those described below.
  • ADAM Disintegrin Domain Polypeptide Constructs SEQ ID Nos: ADAM DNA/ IgK disintegrin 1,3 Fc Construct polypeptide Leader 1,2 (dis Framework) 1,4 Region 1 ADAM- 1/2 1-20 23-264 267-494 8dis-Fc (34-91) ADAM- 3/4 1-20 23-303 306-533 9dis-Fc (34-92) ADAM- 5/6 1-20 23-235 238-465 10dis-Fc (34-99) ADAM- 7/8 1-20 23-292 295-522 15dis-Fc (34-92) ADAM- 9/10 1-20 23-216 219-446 17dis-Fc (34-93) ADAM- 11/12 1-20 23-305 308-535 20dis-Fc (34-91) ADAM- 13/14 1-20 23-293 296-523 21dis-Fc (34-91) ADAM- 15/16 1-20 23-312 315-542 22dis-Fc (34-92) ADAM- 17/18 1-20 23
  • This example describes a flow cytometric integrin mAb based binding inhibition assay, which is used to show binding of ADAM disintegrin-Fc polypeptides to integrins expressed on the surface of endothelial cells.
  • Human endothelial cells express ⁇ v ⁇ 3 , ⁇ v ⁇ 5 , ⁇ 1 , ⁇ 4 , ⁇ 1 , ⁇ 2 , ⁇ 3 , ⁇ 4 , ⁇ 5 , and ⁇ 6 integrins.
  • HMVEC-d Primary human dermal microvascular endothelial cells
  • endothelial growth medium Clonetics Corporation, Walkersville, Md.
  • the ADAM disintegrin-Fc polypeptides produced in Example 1 were shown to bind specifically to HMVEC-d.
  • Monoclonal antibodies specific for human integrins ⁇ v ⁇ 3 (LM609, anti CD51/61, Chemicon, Temecula, C A Brooks et al., Science 264:569, 1994), ⁇ 2 ⁇ 1 (BHA2.1 anti CD49b, Chemicon, Wang et al., Mol. Biol.
  • integrin mAbs to inhibit the binding of ADAM disintegrin-Fc polypeptides reveals which integrins the disintegrin domains bind and, indirectly, which integrin binding activities the disintegrin domains are able to antagonize.
  • the ability of the antibodies to inhibit binding of the ADAM disintegrin-Fc polypeptides to endothelial cells was tested as described below.
  • HMVEC-d Prior to performing binding studies, HMVEC-d were removed from culture vessels using trypsin-EDTA. The cells were washed in media containing serum and resuspended in binding medium which consisted of PBS containing 1 mM Ca2+, 1 mM Mg2+ and 0.5 mM Mn2+, 0.1% sodium azide, 10% Normal goat serum, 2% rabbit serum and 2% fetal bovine serum. Under these binding conditions, ADAM-8, -9, -10, -15, -17, -20, -21, -22, -23, and -29dis-Fc all bind to human endothelial cells.
  • Unbound Fc polypeptides were washed away by centrifugation of cells in 2 mls of binding media. The washed cell pellets were resuspended in binding medium and then incubated at 30° C. for 30 minutes with goat anti-human Fc-specific biotinylated antibody at a concentration of 2.5 ⁇ g/ml for 30 minutes. After centrifugation and washing of the cell pellets, the cells were resuspended in binding medium and bound anti-human Fc-biotin was detected by adding streptavidin-phycoerythrin conjugate to the cell suspension at a 1:1000 dilution (1 ⁇ g/ml) and incubating at 30° C. for 30 minutes.
  • the unbound streptavidin-phycoerythrin was washed away and the cells were resuspended in binding medium containing propidum iodide.
  • the level of fluorescent binding was determined by flow cytometry.
  • ADAM-10, -22, and -23 disintegrin polypeptides did significantly affect the binding of ADAM-10, -22, and -23 disintegrin polypeptides to endothelial cells, suggesting that these ADAMdis polypeptides interact with integrin sites other than or in addition to the ligand (e.g., fibronectin, vitronectin) binding site.
  • ligand e.g., fibronectin, vitronectin
  • ADAM disintegrin-Fc polypeptides that bind to selected integrins are further tested for the ability to disrupt integrin-ligand interactions and to modulate endothelial cell function, angiogenesis, and other biological activities in vitro and in vivo.
  • ADAMdis-Fc polypeptides were purified from whole blood. These cells were used in FACS experiments to assess cell surface binding of purified ADAMdis-Fc polypeptides. ADAMdis-Fc binding was assessed with and without Con A (5 ⁇ g/ml) or immobilized OTK3 antibody (1 mg/ml, immobilized for 1 hour, 37° C.) stimulation. ADAMdis-Fc polypeptides (20 ⁇ g/ml) were bound at either 4° C. or 30° C. in the presence of cations (Ca++, Mg++, Mn++, 0.5 mM each). Cell surface integrin expression was assessed using a panel of murine and rat anti-human integrin antibodies.
  • ADAMdis-Fc polypeptides did not bind to primary human T-cells at 4° C.
  • ADAM-8-, ADAM-9-, ADAM-15-, ADAM-20-, ADAM-21-, ADAM-22-, and ADAM-23-dis-Fc polypeptides did bind primary T-cells at 30° C. with Con A stimulation.
  • ADAMdis-Fc binding was not inhibited by a three-fold molar excess of antibodies to the integrins listed above.
  • ADAMdis-Fc polypeptides to citrated washed resting platelets was performed at 4° C. or 30° C. Binding was analyzed by flow cytometry using a biotinylated-anti-human Fc specific antibody and streptavidin-PE. Resting platelets express the integrins CD41/CD61 and CD49e. ADAM-9dis-Fc and ADAM-8dis-Fc bound resting platelets at 30° C. but not at 4° C. ADAM-9dis-Fc binding to resting platelets at 30° C. was not inhibited by a ten-fold excess of CD41a mAb.
  • ADAM Disintegrin Domain Polypeptides in a Wound Closure Assay
  • a planar endothelial cell migration (wound closure) assay was used to quantitate the inhibition of angiogenesis by ADAM disintegrin-Fc polypeptides in vitro.
  • endothelial cell migration is measured as the rate of closure of a circular wound in a cultured cell monolayer.
  • the rate of wound closure is linear, and is dynamically regulated by agents that stimulate and inhibit angiogenesis in vivo.
  • HRMEC Primary human renal microvascular endothelial cells, HRMEC, were isolated, cultured, and used at the third passage after thawing, as described in Martin et al., In Vitro Cell Dev Biol 33:261, 1997. Replicate circular lesions, “wounds,” (600-800 micron diameter) were generated in confluent HRMEC monolayers using a silicon-tipped drill press. At the time of wounding the medium (DMEM+1% BSA) was supplemented with 20 ng/ml PMA (phorbol-12-myristate-13-acetate), a range of concentrations of ADAM disintegrin-Fc polypeptide, or combinations of PMA and ADAM disintegrin-Fc polypeptide.
  • PMA phorbol-12-myristate-13-acetate
  • the residual wound area was measured as a function of time (0-12 hours) using a microscope and image analysis software (Bioquant, Arlington, Tenn.). The relative migration rate was calculated for each agent and combination of agents by linear regression of residual wound area plotted over time. The inhibition of PMA-induced endothelial migration by ADAM disintegrin-Fc polypeptides is shown in Table 4.
  • ADAM-20 and -23dis-Fc polypeptides showed the greatest inhibition of both EGF- and PMA-induced endothelial migration at 15 ⁇ g/ml.
  • ADAM-15 and -17dis-Fc polypeptides were less effective at inhibiting endothelial cell migration at 15 ⁇ g/ml.
  • Hu IgG did not inhibit EGF- or PMA-induced endothelial cell migration in any of the experiments performed where it was included as a control Fc protein.
  • ADAM Disintegrin Domain Polypeptides in a Corneal Pocket Assay
  • a mouse corneal pocket assay is used to quantitate the inhibition of angiogenesis by ADAM disintegrin-Fc polypeptides in vivo.
  • agents to be tested for angiogenic or anti-angiogenic activity are immobilized in a slow release form in a hydron pellet, which is implanted into micropockets created in the corneal epithelium of anesthetized mice.
  • Vascularization is measured as the appearance, density, and extent of vessel ingrowth from the vascularized corneal limbus into the normally avascular cornea.
  • Hydron pellets as described in Kenyon et al., Invest Opthamol. & Visual Science 37:1625, 1996, incorporate sucralfate with bFGF (90 ng/pellet), bFGF and IgG (11 ⁇ g/pellet, control), or bFGF and a range of concentrations of ADAM disintegrin-Fc polypeptide.
  • the pellets are surgically implanted into corneal stromal micropockets created by micro-dissection 1 mm medial to the lateral corneal limbus of 6-8 week old male C57BL mice.
  • the corneas are photographed, using a Zeiss slit lamp, at an incipient angle of 35-50° from the polar axis in the meridian containing the pellet. Images are digitized and processed by subtractive color filters (Adobe Photoshop 4.0) to delineate established microvessels by hemoglobin content. Image analysis software (Bioquant, Nashville, Tenn.) is used to calculate the fraction of the corneal image that is vascularized, the vessel density within the vascularized area, and the vessel density within the total cornea. The inhibition of bFGF-induced corneal angiogenesis, as a function of the dose of ADAM disintegrin-Fc polypeptide, is determined.
  • a murine heterotopic cardiac isograft model is used to demonstrate the antagonistic effects of ADAM disintegrin-Fc polypeptides on neovascularization.
  • mice Female BALB/c (12 weeks of age) recipients are given neonatal heart grafts from donor mice of the same strain.
  • the donor heart tissue is grafted into the left ear pinnae of the recipient on day 0 and the mice are divided into two groups.
  • the control group receives human IgG (Hu IgG) while the other group receives ADAM disintegrin-Fc polypeptide, both intraperitoneally.
  • the treatments are continued for five consecutive days.
  • the functionality of the grafts is determined by monitoring visible pulsatile activity on days 7 and 14 post-engraftment.
  • the inhibition of functional engraftment, as a function of the dose of ADAM disintegrin-Fc polypeptide, is determined.
  • the histology of the transplanted hearts is examined is order to visualize the effects of ADAM disintegrin-Fc polypeptides on edema at the site of transplant and host and donor tissue vasculature (using, e.g., Factor VIII staining).
  • ADAM disintegrin-Fc polypeptides are tested in animal models of solid tumors. The effect of the ADAM disintegrin-Fc polypeptides is determined by measuring tumor frequency and tumor growth.
  • ADAM disintegrin-Fc polypeptides The biological activity of ADAM disintegrin-Fc polypeptides is also demonstrated in other in vitro, ex vivo, and in vivo assays known to the skilled artisan, such as calcium mobilization assays and assays to measure platelet activation, recruitment, or aggregation.

Abstract

The present invention provides methods and compositions for inhibiting the biological activity of integrins, for inhibiting endothelial cell migration, and for inhibiting angiogenesis. In particular, the invention provides compositions comprising ADAM disintegrin domains and methods for using said compositions. In preferred embodiments the methods and compositions of the invention are used to inhibit angiogenesis and to treat diseases or conditions mediated by angiogenesis.

Description

    CROSS REFERENCE TO RELATED APPLICATIONS
  • This application is a divisional of application U.S. Ser. No. 09/792,200, filed Feb. 23, 2001, which claims the benefit of U.S. provisional application Ser. No. 60/184,865, filed Feb. 25, 2000, all of which are incorporated in their entirety by reference herein.
  • FIELD OF THE INVENTION
  • This invention relates to methods and compositions that are useful for antagonizing the interaction between integrins and their ligands. In particular, the invention relates to the use of ADAM disintegrin domains for antagonizing the interaction between integrins and their ligands.
  • BACKGROUND OF THE INVENTION
  • A. Integrins and Disintegrins
  • Integrins are a family of cell surface proteins that mediate adhesion between cells (cell-cell adhesion) and between cells and extracellular matrix proteins (cell-ECM adhesion). Integrins are heterodimeric structures composed of noncovalently bound α and β subunits. In humans, at least fifteen different α subunits and eight different β subunits combine to form integrins with diverse biological activities and ligand specificities. Integrins play important roles in biological processes including embryonic development, platelet aggregation, immune reactions, tissue repair and remodeling, bone resorption, and tumor invasion and metastasis. Integrins are, therefore, important targets for therapeutic intervention in human disease.
  • The disintegrins are a family of low molecular weight, soluble, cysteine-rich peptides which have been isolated from snake venom (reviewed in Niewiarowski et al., Seminars in Hematology 31(4):289, 1994). The snake venom disintegrins typically contain an RGD (Arg-Gly-Asp, SEQ ID NO:19) motif. The RGD motif is recognized by many integrins, and is present in several integrin ligands including fibronectin, vitronectin, and von Willebrand factor. Disintegrins disrupt normal adhesion processes by inhibiting the binding of cell surface integrins to their ligands.
  • Disintegrin-like domains have been identified in cellular proteins from both invertebrates and vertebrates (see, e.g., Westcamp and Blobel, Proc. Natl. Acad. Sci. USA 91:2748, 1994; Wolfsberg et al., Dev. Biol. 169:378, 1995; Alfandari et al., Dev. Biol. 182:314, 1997), including the ADAM family of transmembrane proteins.
  • B. ADAMs
  • The ADAMs, which have also been called MDCs, are a family of type I transmembrane cysteine-rich glycoproteins (Weskamp et al., Proc. Natl. Acad. Sci. USA, 91:2748, 1994; Wolfsberg et al., Dev. Biol. 169:378, 1995). The multidomain structure of the ADAMs typically includes an amino-terminal metalloprotease domain, a disintegrin domain, a cysteine-rich region (the region between the disintegrin domain and the transmembrane domain), a transmembrane region, and a cytoplasmic domain. At least 30 ADAM family members have been identified, in a variety of animal species. The structure of the ADAMs suggests that they may be involved in a variety of biological processes, including cell adhesion, cell fusion, signal transduction, and proteolysis. Members of the ADAM family have, in fact, been shown to play roles in sperm-egg binding and fusion, myotube formation, neurogenesis, and proteolysis.
  • ADAM-15, also called MDC-15 or metargidin, is the only ADAM identified to date which contains an RGD motif within its disintegrin domain. Zhang et al. (J. Biol. Chem. 273(13):7345, 1998) have reported that the isolated disintegrin domain of ADAM-15, expressed in E. coli as a glutathione S-transferase fusion protein, specifically interacts with αvβ3 integrin and that the interaction is mediated by the RGD tripeptide sequence. The recombinant fusion protein did not interact with other integrins tested, including αIIbβ3 and α5β1. Nath et al. (J. Cell Science 112:579, 1999) have reported that the entire ADAM-15 extracellular domain, expressed as an Fc fusion protein in COS cells, interacts with αvβ3 and α5β1 integrins on hematopoietic cells and that the interaction is mediated by the RGD tripeptide sequence. Zhang et al. and Nath et al. commented that the RGD-dependent interaction between ADAM-15 and αvβ3 integrin suggests a role in processes such as malignancy and angiogenesis.
  • C. Angiogenesis
  • Angiogenesis, the generation of new blood vessels, is a spatially and temporally regulated process in which endothelial and smooth muscle cells proliferate, migrate, and assemble into tubes, in response to endogenous positive and negative regulatory molecules. Angiogenesis plays important roles in both normal and pathological physiology.
  • Under normal physiological conditions, angiogenesis is involved in fetal and embryonic development, wound healing, organ regeneration, and female reproductive remodeling processes including formation of the endometrium, corpus luteum, and placenta. Angiogenesis is stringently regulated under normal conditions, especially in adult animals, and perturbation of the regulatory controls can lead to pathological angiogenesis.
  • Pathological angiogenesis has been implicated in the manifestation and/or progression of inflammatory diseases, certain eye disorders, and cancer. In particular, several lines of evidence support the concept that angiogenesis is essential for the growth and persistence of solid tumors and their metastases (see, e.g., Folkman, N. Engl. J. Med. 285:1182, 1971; Folkman et al., Nature 339:58, 1989; Kim et al., Nature 362:841, 1993; Hori et al., Cancer Res., 51:6180, 1991; Zetter, Annu. Rev. Med. 49:407, 1998). The formation of new blood vessels provides a growing tumor with oxygen, nutrients, waste removal, and a conduit by which invasive cells can enter the circulatory system and establish distant metastases. Various classes of angiogenesis inhibitors are presently being developed and tested for the prevention (e.g., treatment of premalignant conditions), intervention (e.g., treatment of small tumors), and regression (e.g., treatment of large tumors) of cancers (see, e.g., Bergers et al., Science 284:808, 1999) and other forms of pathological angiogenesis. Because many steps in the angiogenic process, including endothelial cell migration, proliferation, and morphogenesis require vascular cell adhesion, certain integrin antagonists have been tested as anti-angiogenic agents.
  • Several integrins are expressed on the surface of cultured endothelial and smooth muscle cells, including αvβ3 integrin. The αvβ3 integrin is an endothelial cell receptor for von Willebrand factor, fibrin, fibrinogen, and fibronectin, and a marker of angiogenic vascular tissue. Brooks et al. have reported that monoclonal antibodies to αvβ3 integrin, as well as cyclic peptide inhibitors, disrupt angiogenesis and that αvβ3 antibodies promote tumor regression (Science 264:569, 1994; Cell 79:1157, 1994). These results suggest that αvβ3 integrin is a useful therapeutic target for diseases characterized by pathological angiogenesis.
  • There is great need for additional compositions and methods of antagonizing the interaction between integrins and their ligands. In particular, there is great need for additional compositions and methods of inhibiting angiogenesis for the prevention, abrogation, and mitigation of disease processes that are dependent upon pathological angiogenesis.
  • SUMMARY OF THE INVENTION
  • The present invention is based upon the discovery that ADAM disintegrin domains are useful for inhibiting the biological activity of integrins and for inhibiting endothelial cell migration and angiogenesis, including the unexpected discovery that these inhibitory activities reside in ADAM disintegrin domains that lack an RGD motif.
  • The invention is directed to methods of antagonizing the binding of an integrin to its ligands, and thereby inhibiting the biological activity of the integrin, comprising contacting the integrin with an effective amount of an ADAM disintegrin domain polypeptide. The invention is further directed to methods of inhibiting endothelial cell migration and methods of inhibiting angiogenesis comprising administering an effective amount of an ADAM disintegrin domain polypeptide. In some embodiments the ADAM disintegrin domain polypeptide is in the form of a multimer, preferably a leucine zipper multimer or Fc polypeptide. In some embodiments the ADAM disintegrin domain is from a human ADAM, and preferably from ADAM-8, ADAM-9, ADAM-10, ADAM-15, ADAM-17, ADAM-20, ADAM-21, ADAM-22, ADAM-23, or ADAM-29. The ADAM disintegrin domain is preferably produced in a recombinant cell, and is preferably present in a composition comprising a pharmaceutically acceptable carrier.
  • In some preferred embodiments the ADAM disintegrin domain polypeptide comprises an amino acid sequence selected from the group consisting of: amino acids 23-264 of SEQ ID NO:2, amino acids 23-303 of SEQ ID NO:4, amino acids 23-235 of SEQ ID NO:6, amino acids 23-292 of SEQ ID NO:8, amino acids 23-216 of SEQ ID NO:10, amino acids 23-305 of SEQ ID NO:12, amino acids 23-293 of SEQ ID NO:14, amino acids 23-312 of SEQ ID NO:16, amino acids 23-310 of SEQ ID NO:18, and amino acids 23-298 of SEQ ID NO:22. In some more preferred embodiments the ADAM disintegrin domain polypeptide comprises an amino acid sequence selected from the group consisting of: amino acids 34-91 of SEQ ID NO:2, amino acids 34-92 of SEQ ID NO:4, amino acids 34-99 of SEQ ID NO:6, amino acids 34-92 of SEQ ID NO:8, amino acids 34-93 of SEQ ID NO:10, amino acids 34-91 of SEQ ID NO:12, amino acids 34-91 of SEQ ID NO:14, amino acids 34-92 of SEQ ID NO:16, amino acids 34-91 of SEQ ID NO:18, and amino acids 34-91 of SEQ ID NO:22. In some most preferred embodiments the ADAM disintegrin domain polypeptide comprises an amino acid sequence selected from the group consisting of: amino acids 78-91 of SEQ ID NO:2, amino acids 79-92 of SEQ ID NO:4, amino acids 87-99 of SEQ ID NO:6, amino acids 79-92 of SEQ ID NO:8, amino acids 79-93 of SEQ ID NO:10, amino acids 78-91 of SEQ ID NO:12, amino acids 78-91 of SEQ ID NO:14, amino acids 79-92 of SEQ ID NO:16, amino acids 78-91 of SEQ ID NO:18, and amino acids 78-91 of SEQ ID NO:22.
  • In some embodiments a therapeutically effective amount of the ADAM disintegrin domain is administered to a mammal in need of such treatment. In preferred embodiments the mammal is afflicted with a condition mediated by angiogenesis, an ocular disorder, malignant or metastatic condition, inflammatory disease, osteoporosis and other conditions mediated by accelerated bone resorption, restenosis, inappropriate platelet activation, recruitment, or aggregation, thrombosis, or a condition requiring tissue repair or wound healing. The ADAM disintegrin domain is, in some embodiments, administered in combination with radiation therapy and/or in combination with one or more additional therapeutic agents.
  • The invention also encompasses methods for identifying compounds that modulate integrin biological activity, that modulate the interaction between an integrin and an ADAM disintegrin domain, that inhibit endothelial cell migration, or that inhibit angiogenesis, comprising combining a test compound with an integrin or with endothelial cells and with an ADAM disintegrin domain polypeptide that binds to the integrin or endothelial cells and determining whether the test compound alters the binding of the ADAM disintegrin domain polypeptide to the integrin or endothelial cells.
  • These and other aspects of the present invention will become evident upon reference to the following detailed description, examples, and claims.
  • DETAILED DESCRIPTION OF THE INVENTION
  • A. Abbreviations and Terminology Used in the Specification
  • “4-1BB” and “4-1BB ligand” (4-1BB-L) are polypeptides described, inter alia, in U.S. Pat. No. 5,674,704, including soluble forms thereof.
  • “ADAMs” are a family of transmembrane glycoproteins having disintegrin and metalloproteinase domains, also called MDC, metalloprotease/disintegrin/cysteine-rich proteins.
  • “Dis” is a disintegrin domain; “ADAMdis” is an ADAM disintegrin domain.
  • “CD40 ligand” (CD40L) is a polypeptide described, inter alia, in U.S. Pat. No. 5,716,805, including soluble forms thereof.
  • “CD148” is a protein tyrosine phosphatase, also called DEP-1, ECRTP, and PTPRJ. CD148 binding proteins are described in Daniel et al., PCT Publication No. WO 00/15258, 23 Mar. 2000.
  • “DMEM” is Dulbecco's Modified Eagle Medium.
  • “FACS” is fluorescence activated cell sorting.
  • “Flt3L” is Flt3 ligand, a polypeptide described, inter alia, in U.S. Pat. No. 5,554,512, including soluble forms thereof.
  • “HRMEC” are human renal microvascular endothelial cells.
  • “HMVEC-d” are human dermal microvascular endothelial cells.
  • “mAb” is a monoclonal antibody.
  • “MDC” is a family of cysteine-rich proteins having metalloprotease and disintegrin domains, also called ADAM.
  • “Nectin-3” is a cell adhesion molecule in the nectin family (which is described, inter alia, in Satoh-Horikawa et al., J. Biol. Chem. 275(14):10291, 2000). The GenBank accession numbers of human nectin-3 nucleic acid and polypeptide sequences are AF282874 and AAF97597 respectively (Reymond et al., 2000).
  • “PMA” is phorbol-12-myristate-13-acetate.
  • “Tek,” which has also been called Tie2 and ork, is an receptor tyrosine kinase (RTK) that is predominantly expressed in vascular endothelium. The molecular cloning of human Tek (ork) has been described by Ziegler, U.S. Pat. No. 5,447,860. “Tek antagonists” are described, inter alia, in Cerretti et al., PCT Publication No. WO 00/75323, 14 Dec. 2000.
  • “TNF” is tumor necrosis factor. “TNFR” is a tumor necrosis factor receptor, including soluble forms thereof. “TNFR/Fc” is a tumor necrosis factor receptor-Fc fusion polypeptide.
  • “TRAIL” is TNF-related apoptosis-inducing ligand, a type II transmembrane polypeptide in the TNF family described, inter alia, in U.S. Pat. No. 5,763,223, including soluble forms thereof.
  • “TWEAK” is TNF-weak effector of apoptosis, a type II transmembrane polypeptide in the TNF family described, inter alia, in Chicheportiche et al., J. Biol. Chem., 272(51):32401, 1997, including soluble forms thereof. “TWEAK-R” is the “TWEAK receptor,” which is described, inter alia, in U.S. Ser. Nos. 60/172,878 and 60/203,347 and Feng et al., Am. J. Pathol. 156(4):1253, 2000, including soluble forms thereof. TWEAK-R/Fc is a TWEAK receptor-Fc fusion polypeptide.
  • “VEGF” is vascular endothelial growth factor, also known as VPF or vascular permeability factor.
  • B. ADAM Polypeptides and ADAM Disintegrin Domain Polypeptides
  • At least thirty ADAMs have been described. Table 1 provides reference information for selected human ADAMs.
  • ADAM disintegrin domains show sequence homology to the snake venom disintegrins, and are characterized by a framework of cysteines. For example, a typical disintegrin sequence comprises a framework such as:
    (SEQ ID NO:20)
    CDCGX3-5CX3-6CCX2-4CX7CX4-6CCX2-4CX8CX5-7CX3-5C

    The sequences of several ADAM disintegrin domains are shown in Table 2 and in the Sequence Listing.
  • The present invention encompasses the use of various forms of ADAM disintegrin domains that retain at least one activity selected from the group consisting of integrin binding activity, inhibition of endothelial cell migration, and inhibition of angiogenesis. The term “ADAM disintegrin domain polypeptide” is intended to encompass polypeptides containing all or part of a native ADAM disintegrin domain, with or without other ADAM domains (such as the cysteine-rich region), as well as related forms including, but not limited to: (a) fragments, (b) variants, (c) derivatives, (d) fusion polypeptides, and (e) multimeric forms (multimers). The ability of these related forms to inhibit integrin binding, endothelial cell migration, and/or inhibition of angiogenesis may be determined in vitro or in vivo by using methods such as those exemplified below or by using other assays known in the art.
    TABLE 1
    Selected Members of the ADAM Family
    GenBank
    Accession Published
    ADAM Other Names Number (Human) Description
    ADAM-8 MS2, CD156 D26579 Genomics 41(1):
    56, 1997
    ADAM-9 MDC9, meltrin U41766 J. Cell. Biol.
    gamma 132(4): 717, 1996
    ADAM-10 MADM, AF009615 J. Biol. Chem.
    kuzbanian, 272(39): 24588,
    reprolysin 1997
    ADAM-15 Metargidin, U46005 J. Biol. Chem.
    MDC15 271(9): 4593, 1996
    ADAM-17 TACE, cSVP U86755 WO 96/41624
    ADAM-20 SVPH1-26 AF029899 WO 99/23228
    ADAM-21 SVPH1-8 AF029900 WO 99/36549
    ADAM-22 SVPH3-13, MDC2 AB009671 WO 99/41388
    ADAM-23 SVPH3-17, MDC3 AB009672 WO 99/41388
    ADAM-29 SVPH1 AF171929 Biochem. Biophys.
    Res. Commun.
    263: 810, 1999
  • The term “variant” includes polypeptides that are substantially homologous to native ADAM disintegrin domains, but which have an amino acid sequence different from that of a native ADAM disintegrin domain because of one or more deletions, insertions or substitutions. Particular embodiments include, but are not limited to, ADAM disintegrin domain polypeptides that comprise from one to ten deletions, insertions or substitutions of amino acid residues, when compared to a native ADAM disintegrin domain sequence. Included as variants of ADAM disintegrin domain polypeptides are those variants that are naturally occurring, such as allelic forms and alternatively spliced forms, as well as variants that have been constructed by modifying the amino acid sequence of a ADAM disintegrin domain polypeptide or the nucleotide sequence of a nucleic acid encoding a ADAM disintegrin domain polypeptide.
  • Generally, substitutions for one or more amino acids present in the native polypeptide should be made conservatively. Examples of conservative substitutions include substitution of amino acids outside of the active domain(s), and substitution of amino acids that do not alter the secondary and/or tertiary structure of the ADAM disintegrin domain. Additional examples include substituting one aliphatic residue for another, such as Ile, Val, Leu, or Ala for one another, or substitutions of one polar residue for another, such as between Lys and Arg; Glu and Asp; or Gln and Asn, or substitutions of one aromatic residue for another, such as Phe, Trp, or Tyr for one another. Other such conservative substitutions, for example, substitutions of entire regions having similar hydrophobicity characteristics, are known in the art.
  • In some preferred embodiments the ADAM disintegrin domain variant is at least about 70% identical in amino acid sequence to the amino acid sequence of a native ADAM disintegrin domain; in some preferred embodiments the ADAM disintegrin domain variant is at least about 80% identical in amino acid sequence to the amino acid sequence of a native ADAM disintegrin domain. In some more preferred embodiments the ADAM disintegrin domain variant is at least about 90% identical in amino acid sequence to the amino acid sequence of a native ADAM disintegrin domain; in some more preferred embodiments the ADAM disintegrin domain variant is at least about 95% identical in amino acid sequence to the amino acid sequence of a native ADAM disintegrin domain. In some most preferred embodiments the ADAM disintegrin domain variant is at least about 98% identical in amino acid sequence to the amino acid sequence of a native ADAM disintegrin domain; in some most preferred embodiments the ADAM disintegrin domain variant is at least about 99% identical in amino acid sequence to the amino acid sequence of a native ADAM disintegrin domain.
  • Percent identity, in the case of both polypeptides and nucleic acids, may be determined by visual inspection. Percent identity may be determined using the alignment method of Needleman and Wunsch (J. Mol. Biol. 48:443, 1970) as revised by Smith and Waterman (Adv. Appl. Math 2:482, 1981. Preferably, percent identity is determined by using a computer program, for example, the GAP computer program version 10.x available from the Genetics Computer Group (GCG; Madison, Wis., see also Devereux et al., Nucl. Acids Res. 12:387, 1984). The preferred default parameters for the GAP program include: (1) a unary comparison matrix (containing a value of 1 for identities and 0 for non-identities) for nucleotides, and the weighted comparison matrix of Gribskov and Burgess, Nucl. Acids Res. 14:6745, 1986, as described by Schwartz and Dayhoff, eds., Atlas of protein Sequence and Structure, National Biomedical Research Foundation, pp. 353-358, 1979 for amino acids; (2) a penalty of 30 (amino acids) or 50 (nucleotides) for each gap and an additional 1 (amino acids) or 3 (nucleotides) penalty for each symbol in each gap; (3) no penalty for end gaps; and (4) no maximum penalty for long gaps. Other programs used by one skilled in the art of sequence comparison may also be used. For fragments of ADAM disintegrin domains, the percent identity is calculated based on that portion of ADAM disintegrin domain that is present in the fragment.
  • When a deletion or insertion strategy is adopted, the potential effect of the deletion or insertion on biological activity (such as integrin binding activity, inhibition of endothelial cell migration, or inhibition of angiogenesis) must be considered. Subunits of the inventive polypeptides may be constructed by deleting terminal or internal residues or sequences. Additional guidance as to the types of mutations that can be made is provided by a comparison of the sequence of ADAM disintegrin domain polypeptides to polypeptides that have similar structures, as well as by performing structural analysis of the inventive polypeptides.
  • The term “variant” also includes ADAM disintegrin domain polypeptides that are encoded by nucleic acids capable of hybridizing under moderately stringent conditions (e.g., prewashing solution of 5×SSC, 0.5% SDS, 1.0 mM EDTA (pH 8.0) and hybridization conditions of 50° C., 5×SSC, overnight) or higher stringency conditions to DNA sequences encoding ADAM disintegrin domain polypeptides, and which encode polypeptides that retain at least one activity selected from the group consisting of integrin binding activity, inhibition of endothelial cell migration, and inhibition of angiogenesis. The skilled artisan can determine additional combinations of salt and temperature that constitute moderate hybridization stringency. Conditions of higher stringency include higher temperatures for hybridization and post-hybridization washes, and/or lower salt concentration.
  • Mutations can be introduced into nucleic acids by synthesizing oligonucleotides containing a mutant sequence, flanked by restriction sites enabling ligation to fragments of the native sequence. Following ligation, the resulting reconstructed sequence encodes a variant having the desired amino acid insertion, substitution, or deletion. Alternatively, oligonucleotide-directed site-specific mutagenesis procedures can be employed to provide an altered gene having particular codons altered according to the substitution, deletion, or insertion required. The well known polymerase chain reaction (PCR) procedure also may be employed to generate and amplify a DNA sequence encoding a desired polypeptide or fragment thereof. Oligonucleotides that define the desired termini of the DNA fragment are employed as 5′ and 3′ primers. The oligonucleotides may additionally contain recognition sites for restriction endonucleases to facilitate insertion of the amplified DNA fragment into an expression vector.
  • The present invention further encompasses the use of ADAM disintegrin domain polypeptides with or without associated native-pattern glycosylation. ADAM disintegrin domain expressed in yeast or mammalian expression systems (e.g., COS-1 or COS-7 cells) may be similar to or significantly different from a native ADAM disintegrin domain polypeptide in molecular weight and glycosylation pattern, depending upon the choice of expression system. Expression of ADAM disintegrin domain polypeptides in bacterial expression systems, such as E. coli, provides non-glycosylated molecules. Different host cells may also process polypeptides differentially, resulting in heterogeneous mixtures of polypeptides with variable N- or C-termini.
  • The primary amino acid structure of ADAM disintegrin domain polypeptides may be modified to create derivatives by forming covalent or aggregative conjugates with other chemical moieties, such as glycosyl groups, lipids, phosphate, acetyl groups and the like. Covalent derivatives of ADAM disintegrin domain polypeptides may be prepared by linking particular functional groups to ADAM disintegrin domain amino acid side chains or at the N-terminus or C-terminus of a ADAM disintegrin domain polypeptide.
  • Fusion polypeptides of ADAM disintegrin domains that are useful in practicing the invention include covalent or aggregative conjugates of ADAMdis or its fragments with other polypeptides, such as by synthesis in recombinant culture as N-terminal or C-terminal fusions. One class of fusion polypeptides are discussed below in connection with ADAM disintegrin oligomers. As another example, a fusion polypeptide may comprise a signal peptide (which is also variously referred to as a signal sequence, signal, leader peptide, leader sequence, or leader) at the N-terminal region or C-terminal region of an ADAM disintegrin domain polypeptide which co-translationally or post-translationally directs transfer of the polypeptide from its site of synthesis to a site inside or outside of the cell membrane or cell wall. It is particularly advantageous to fuse a signal peptide that promotes extracellular secretion to the N-terminus of a soluble ADAMdis polypeptide. In this case, the signal peptide is typically cleaved upon secretion of the soluble polypeptide from the cell.
  • Secreted soluble polypeptides may be identified (and distinguished from its non-soluble membrane-bound counterparts) by separating intact cells which express the desired polypeptide from the culture medium, e.g., by centrifugation, and assaying the medium (supernatant) for the presence of the desired polypeptide. The presence of the desired polypeptide in the medium indicates that the polypeptide was secreted from the cells and thus is a soluble form of the polypeptide. Soluble polypeptides may be prepared by any of a number of conventional techniques. A DNA sequence encoding a desired soluble polypeptide may be subcloned into an expression vector for production of the polypeptide, or the desired encoding DNA fragment may be chemically synthesized.
  • Soluble ADAM disintegrin domain polypeptides comprise all or part of the ADAM disintegrin domain, with or without additional segments from the extracellular portion of the ADAM (such as the cysteine-rich region) but generally lack a transmembrane domain that would cause retention of the polypeptide at the cell surface. Soluble polypeptides may include part of the transmembrane domain or all or part of the cytoplasmic domain as long as the polypeptide is secreted from the cell in which it is produced. Examples of soluble ADAM disintegrin domain polypeptides are provided in the examples. In some preferred embodiments of the present invention, a multimeric form of a soluble ADAM disintegrin domain polypeptide is used to inhibit integrin binding to ligands and, hence, integrin biological activity. In some most preferred embodiments the soluble ADAM disintegrin domain polypeptide is used to inhibit endothelial cell migration and/or inhibit angiogenesis. These inhibitory activities may include both integrin-mediated and integrin-independent mechanisms.
  • ADAM disintegrin domain multimers are covalently-linked or non-covalently-linked multimers, including dimers, trimers, and higher multimers. Oligomers may be linked by disulfide bonds formed between cysteine residues on different ADAM disintegrin domain polypeptides. One embodiment of the invention is directed to multimers comprising multiple ADAM disintegrin domain polypeptides joined via covalent or non-covalent interactions between peptide moieties fused to the ADAM disintegrin domain polypeptides. Such peptides may be peptide linkers (spacers), or peptides that have the property of promoting multimerization. Leucine zippers and certain polypeptides derived from antibodies are among the peptides that can promote multimerization of ADAM disintegrin domain polypeptides attached thereto, as described in more detail below. In particular embodiments, the multimers comprise from two to four ADAM disintegrin domain polypeptides.
  • In some embodiments, a ADAM disintegrin domain multimer is prepared using polypeptides derived from immunoglobulins. Preparation of fusion proteins comprising certain heterologous polypeptides fused to various portions of antibody-derived polypeptides (including the Fc domain) has been described, e.g., by Ashkenazi et al. (Proc. Natl. Acad. Sci. USA 88:10535, 1991); Byrn et al. (Nature 344:677, 1990); and Hollenbaugh and Aruffo (“Construction of Immunoglobulin Fusion Proteins”, in Current Protocols in Immunology, Suppl. 4, pages 10.19.1-10.19.11, 1992).
  • A preferred embodiment of the present invention is directed to an ADAM disintegrin domain (ADAMdis) dimer comprising two fusion polypeptides created by fusing an ADAM disintegrin domain to an Fc polypeptide. A gene fusion encoding the ADAMdis-Fc fusion polypeptide is inserted into an appropriate expression vector. ADAMdis-Fc fusion polypeptides are expressed in host cells transformed with the recombinant expression vector, and allowed to assemble much like antibody molecules, whereupon interchain disulfide bonds form between the Fc moieties to yield divalent soluble ADAMdis polypeptides. The term “Fc polypeptide” as used herein includes native and mutein forms of polypeptides derived from the Fc region of an antibody. Truncated forms of such polypeptides containing the hinge region that promotes dimerization are also included.
  • One suitable Fc polypeptide, described in PCT application WO 93/10151, is a single chain polypeptide extending from the N-terminal hinge region to the native C-terminus of the Fc region of a human IgG1 antibody. Another useful Fc polypeptide is the Fc mutein described in U.S. Pat. No. 5,457,035 and by Baum et al., EMBO J. 13:3992, 1994. The amino acid sequence of this mutein is identical to that of the native Fc sequence presented in WO 93/10151, except that amino acid 19 has been changed from Leu to Ala, amino acid 20 has been changed from Leu to Glu, and amino acid 22 has been changed from Gly to Ala. The mutein exhibits reduced affinity for Fc receptors. Fusion polypeptides comprising Fc moieties, and multimers formed therefrom, offer an advantage of facile purification by affinity chromatography over Protein A or Protein G columns, and Fc fusion polypeptides may provide a longer in vivo half life, which is useful in therapeutic applications, than unmodified polypeptides.
  • In other embodiments, a soluble ADAM disintegrin domain polypeptide may be substituted for the variable portion of an antibody heavy or light chain. If fusion proteins are made with both heavy and light chains of an antibody, it is possible to form an ADAM disintegrin domain multimer with as many as four soluble ADAM disintegrin domain polypeptides.
  • Alternatively, the ADAM disintegrin domain multimer is a fusion polypeptide comprising multiple ADAM disintegrin domain polypeptides, with or without peptide linkers (spacers), or peptides that have the property of promoting multimerization. Among the suitable peptide linkers are those described in U.S. Pat. Nos. 4,751,180 and 4,935,233. A DNA sequence encoding a desired peptide linker may be inserted between, and in the same reading frame as, the DNA sequences encoding ADAMdis, using conventional techniques known in the art. For example, a chemically synthesized oligonucleotide encoding the linker may be ligated between sequences encoding ADAMdis. In particular embodiments, a fusion protein comprises from two to four ADAM disintegrin domain polypeptides, separated by peptide linkers.
  • Another method for preparing ADAM disintegrin domain multimers involves use of a leucine zipper domain. Leucine zipper domains are peptides that promote multimerization of the proteins in which they are found. Leucine zippers were originally identified in several DNA-binding proteins (Landschulz et al., Science 240:1759, 1988), and have since been found in a variety of different proteins. Among the known leucine zippers are naturally occurring peptides and derivatives thereof that dimerize or trimerize. Examples of leucine zipper domains suitable for producing soluble oligomeric proteins are described in PCT application WO 94/10308, and the leucine zipper derived from lung surfactant protein D (SPD) described in Hoppe et al. FEBS Lett. 344:191, 1994. The use of a modified leucine zipper that allows for stable trimerization of a heterologous protein fused thereto is described in Fanslow et al., Semin. Immunol. 6:267, 1994. Recombinant fusion polypeptides comprising an ADAM disintegrin domain polypeptide fused to a leucine zipper peptide are expressed in suitable host cells, and the ADAM disintegrin domain multimer that forms is recovered from the culture supernatant.
  • C. Recombinant Production of ADAM Disintegrin Domain Polypeptides
  • The ADAM disintegrin domain polypeptides used in the present invention may be prepared using a recombinant expression system. Host cells transformed with a recombinant expression vector encoding the ADAM disintegrin domain polypeptide are cultured under conditions that promote expression of ADAM disintegrin domain and the ADAM disintegrin domain is recovered. ADAM disintegrin domain polypeptides can also be produced in transgenic plants or animals.
  • Any suitable expression system may be employed. Recombinant expression vectors include DNA encoding an ADAM disintegrin domain polypeptide operably linked to suitable transcriptional and translational regulatory nucleotide sequences, such as those derived from a mammalian, microbial, viral, or insect gene. Nucleotide sequences are operably linked when the regulatory sequence functionally relates to the ADAM disintegrin domain DNA sequence. Thus, a promoter nucleotide sequence is operably linked to an ADAM disintegrin domain DNA sequence if the promoter nucleotide sequence controls the transcription of the ADAM disintegrin domain DNA sequence. Examples of regulatory sequences include transcriptional promoters, operators, or enhancers, an mRNA ribosomal binding site, and appropriate sequences which control transcription and translation initiation and termination. A sequence encoding an appropriate signal peptide (native or heterologous) can be incorporated into expression vectors. A DNA sequence for a signal peptide (secretory leader) may be fused in frame to the ADAM disintegrin domain sequence so that the ADAM disintegrin domain polypeptide is initially translated as a fusion protein comprising the signal peptide. A signal peptide that is functional in the intended host cells promotes extracellular secretion of the ADAM disintegrin domain polypeptide. The signal peptide is cleaved from the ADAM disintegrin domain polypeptide upon secretion from the cell. Suitable host cells for expression of ADAM disintegrin domain polypeptides include prokaryotes, yeast and higher eukaryotic cells, including insect and mammalian cells. Appropriate cloning and expression vectors for use with bacterial, fungal, yeast, insect, and mammalian cellular hosts are known in the art.
  • Using the techniques of recombinant DNA including mutagenesis and the polymerase chain reaction (PCR), the skilled artisan can produce DNA sequences that encode ADAM disintegrin domain polypeptides comprising various additions or substitutions of amino acid residues or sequences, or deletions of terminal or internal residues or sequences, including ADAM disintegrin domain fragments, variants, derivatives, multimers, and fusion polypeptides.
  • The procedures for purifying expressed ADAM disintegrin domain polypeptides will vary according to the host system employed, and whether or not the recombinant polypeptide is secreted. ADAM disintegrin domain polypeptides may be purified using methods known in the art, including one or more concentration, salting-out, ion exchange, hydrophobic interaction, affinity purification, HPLC, or size exclusion chromatography steps. Fusion polypeptides comprising Fc moieties (and multimers formed therefrom) offer the advantage of facile purification by affinity chromatography over Protein A or Protein G columns.
  • D. Therapeutic Methods
  • The disclosed methods may be used to inhibit integrin binding and integrin biological activity, and to inhibit endothelial cell migration, and/or angiogenesis in a mammal in need of such treatment. The treatment is advantageously administered in order to prevent the onset or the recurrence of a disease or condition mediated by an integrin, or to treat a mammal that has a disease or condition mediated by an integrin.
  • Examples of the therapeutic uses of ADAM disintegrin domain polypeptides and compositions thereof include the treatment of individuals afflicted with conditions mediated by angiogenesis such as ocular disorders, dermatological disorders, and malignant or metastatic conditions, inflammatory diseases, osteoporosis and other conditions mediated by accelerated bone resorption, restenosis, inappropriate platelet activation, recruitment, or aggregation, thrombosis, or a condition requiring tissue repair or wound healing.
  • Among the ocular disorders that can be treated according to the present invention are eye diseases characterized by ocular neovascularization including, but not limited to, diabetic retinopathy (a major complication of diabetes), retinopathy of prematurity (this devastating eye condition, that frequently leads to chronic vision problems and carries a high risk of blindness, is a severe complication during the care of premature infants), neovascular glaucoma, retinoblastoma, retrolental fibroplasia, rubeosis, uveitis, macular degeneration, and corneal graft neovascularization. Other eye inflammatory diseases, ocular tumors, and diseases associated with choroidal or iris neovascularization can also be treated according to the present invention.
  • The present invention can also be used to treat malignant and metastatic conditions such as solid tumors. Solid tumors include both primary and metastatic sarcomas and carcinomas.
  • The present invention can also be used to treat inflammatory diseases including, but not limited to, arthritis, rheumatism, inflammatory bowel disease, and psoriasis.
  • Among the conditions mediated by inappropriate platelet activation, recruitment, aggregation, or thrombosis that can be treated according to the present invention are coronary artery disease or injury, myocardial infarction or injury following myocardial infarction, stroke, unstable angina, atherosclerosis, arteriosclerosis, preeclampsia, embolism, platelet-associated ischemic disorders including lung ischemia, coronary ischemia, and cerebral ischemia, restenosis following percutaneous coronary intervention including angioplasty, atherectomy, stent placement, and bypass surgery, thrombotic disorders including coronary artery thrombosis, cerebral artery thrombosis, intracardiac thrombosis, peripheral artery thrombosis, venous thrombosis, thrombosis and coagulopathies associated with exposure to a foreign or injured tissue surface, and reocclusion following thrombosis, deep venous thrombosis (DVT), pulmonary embolism (PE), transient ischemic attacks (TIAs), and another conditions where vascular occlusion is a common underlying feature. In some embodiments the methods according to the invention are used in individuals at high risk for thrombus formation or reformation, advanced coronary artery disease, or for occlusion, reocclusion, stenosis and/or restenosis of blood vessels, or stroke. In some embodiments the methods according to the invention are used in combination with angioplasty procedures, such as balloon angioplasty, laser angioplasty, coronary atherectomy or similar techniques, carotid endarterectomy, anastomosis of vascular grafts, surgery having a high risk of thrombus formation (i.e., coronary bypass surgery, insertion of a prosthetic valve or vessel and the like), atherectomy, stent placement, placement of a chronic cardiovascular device such as an in-dwelling catheter or prosthetic valve or vessel, organ transplantation, or bypass surgery.
  • Other diseases and conditions that can be treated according to the present invention include benign tumors and preneoplastic conditions, myocardial angiogenesis, hemophilic joints, scleroderma, vascular adhesions, asthma and allergy, eczema and dermatitis, graft versus host disease, sepsis, adult respirator distress syndrome, telangiectasia, and wound granulation.
  • The methods according to the present invention can be tested in in vivo animal models for the desired prophylactic or therapeutic activity, as well as to determine the optimal therapeutic dosage, prior to administration to humans.
  • The amount of a particular ADAM disintegrin domain polypeptide that will be effective in a particular method of treatment depends upon age, type and severity of the condition to be treated, body weight, desired duration of treatment, method of administration, and other parameters. Effective dosages are determined by a physician or other qualified medical professional. Typical effective dosages are about 0.01 mg/kg to about 100 mg/kg body weight. In some preferred embodiments the dosage is about 0.1-50 mg/kg; in some preferred embodiments the dosage is about 0.5-10 mg/kg. The dosage for local administration is typically lower than for systemic administration. In some embodiments a single administration is sufficient; in some embodiments the ADAM disintegrin domain is administered as multiple doses over one or more days.
  • The ADAM disintegrin domain polypeptides are typically administered in the form of a pharmaceutical composition comprising one or more pharmacologically acceptable carriers. Pharmaceutically acceptable carriers include diluents, fillers, adjuvants, excipients, and vehicles which are pharmaceutically acceptable for the route of administration, and may be aqueous or oleaginous suspensions formulated using suitable dispersing, wetting, and suspending agents.
  • Pharmaceutically acceptable carriers are generally sterile and free of pyrogenic agents, and may include water, oils, solvents, salts, sugars and other carbohydrates, emulsifying agents, buffering agents, antimicrobial agents, and chelating agents. The particular pharmaceutically acceptable carrier and the ratio of active compound to carrier are determined by the solubility and chemical properties of the composition, the mode of administration, and standard pharmaceutical practice.
  • The ADAM disintegrin domain polypeptides are administered to the patient in a manner appropriate to the indication. Thus, for example, ADAM disintegrin domain polypeptides, or pharmaceutical compositions thereof, may be administered by intravenous, transdermal, intradermal, intraperitoneal, intramuscular, intranasal, epidural, oral, topical, subcutaneous, intracavity, sustained release from implants, peristaltic routes, or by any other suitable technique. Parenteral administration is preferred.
  • In certain embodiments of the claimed invention, the treatment further comprises treating the mammal with one or more additional therapeutic agents. The additional therapeutic agent(s) may be administered prior to, concurrently with, or following the administration of the ADAM disintegrin domain polypeptide. The use of more than one therapeutic agent is particularly advantageous when the mammal that is being treated has a solid tumor. In some embodiments of the claimed invention, the treatment further comprises treating the mammal with radiation. Radiation, including brachytherapy and teletherapy, may be administered prior to, concurrently with, or following the administration of the ADAM disintegrin domain polypeptide and/or additional therapeutic agent(s).
  • In some preferred embodiments the method includes the administration of, in addition to an ADAM disintegrin domain polypeptide, one or more therapeutics selected from the group consisting of alkylating agents, antimetabolites, vinca alkaloids and other plant-derived chemotherapeutics, antitumor antibiotics, antitumor enzymes, topoisomerase inhibitors, platinum analogs, adrenocortical suppressants, hormones and antihormones, antibodies, immunotherapeutics, radiotherapeutics, and biological response modifiers.
  • In some preferred embodiments the method includes administration of, in addition to an ADAM disintegrin domain polypeptide, one or more therapeutics selected from the group consisting of cisplatin, cyclophosphamide, mechloretamine, melphalan, bleomycin, carboplatin, fluorouracil, 5-fluorodeoxyuridine, methotrexate, taxol, asparaginase, vincristine, and vinblastine, lymphokines and cytokines such as interleukins, interferons (alpha., beta. or delta.) and TNF, chlorambucil, busulfan, carmustine, lomustine, semustine, streptozocin, dacarbazine, cytarabine, mercaptopurine, thioguanine, vindesine, etoposide, teniposide, dactinomycin, daunorubicin, doxorubicin, bleomycin, plicamycin, mitomycin, L-asparaginase, hydroxyurea, methylhydrazine, mitotane, tamoxifen, fluoxymesterone, IL-8 inhibitors, angiostatin, endostatin, kringle 5, angiopoietin-2 or other antagonists of angiopoietin-1, antagonists of platelet-activating factor, antagonists of basic fibroblast growth factor, and COX-2 inhibitors.
  • In some preferred embodiments the method includes administration of, in addition to an ADAM disintegrin domain polypeptide, one or more therapeutic polypeptides, including soluble forms thereof, selected from the group consisting of Flt3 ligand, CD40 ligand, interleukin-2, interleukin-12, 4-1BB ligand, anti-4-1BB antibodies, TRAIL, TNF antagonists and TNF receptor antagonists including TNFR/Fc, Tek antagonists, TWEAK antagonists and TWEAK-R antagonists including TWEAK-R/Fc, VEGF antagonists including anti-VEGF antibodies, VEGF receptor (including VEGF-R1 and VEGF-R2, also known as Flt1 and Flk1 or KDR) antagonists, CD148 (also referred to as DEP-1, ECRTP, and PTPRJ, see Takahashi et al., J. Am. Soc. Nephrol. 10:2135-45, 1999; and PCT Publication No. WO 00/15258, 23 Mar. 2000) binding proteins, and nectin-3 antagonists.
  • In some preferred embodiments the ADAM disintegrin domain polypeptides of the invention are used as a component of, or in combination with, “metronomic therapy,” such as that described by Browder et al. and Klement et al. (Cancer Research 60:1878, 2000; J. Clin. Invest. 105(8):R15, 2000; see also Barinaga, Science 288:245, 2000).
  • As used herein, the terms “therapy,” “therapeutic,” “treat,” and “treatment” generally include prophylaxis, i.e. prevention, in addition to therapy or treatment for an extant disease or condition. The methods of the present invention may be used as a first line treatment, for the treatment of residual disease following primary therapy, or as an adjunct to other therapies. Methods of measuring biological effectiveness are known in the art and are illustrated in the Examples below.
  • EXAMPLES
  • The following examples are intended to illustrate particular embodiments and not to limit the scope of the invention.
  • Example 1 ADAM Disintegrin Domain Polypeptides
  • This example describes one method for the recombinant production of ADAM disintegrin domain polypeptides.
  • Expression cassettes encoding an IgKappa leader sequence, ADAM disintegrin domain, and C-terminal Fc region were constructed in bacterial plasmids then transferred into eukaryotic expression vectors (pDC409, EMBO J. 10:2821, 1991, or another mammalian expression vector). The coding regions of the various constructs are summarized in Table 2. In addition to the disintegrin domain, these constructs encode additional portions of the extracellular portion of the ADAM (e.g., cysteine-rich region and EGF-like domain).
  • The expression vectors were transfected into COS-1, CV-1/EBNA, or 293/EBNA cells. Two days after transfection the cells were 35S labeled for four hours. Supernatants and total cell lysates were prepared and aliquots were immunoprecipitated using protein A-sepharose beads to capture the Fc tagged polypeptides. 35S labeled ADAM disintegrin-Fc polypeptides were run on 8-16% reducing gels and detected via autoradiography.
  • The cell type that produced the most soluble protein in the supernatant was used in a large scale (T-175 format, 20 flasks) transient transfection, and approximately one liter of supernatant was harvested after one week. ADAM disintegrin-Fc polypeptides were purified from the supernatants using affinity chromatography (protein A column). The polypeptides were characterized by determining the N-terminal amino acid sequence, amino acid composition, and protein integrity (SDS-PAGE under reducing and non-reducing conditions) before the polypeptides were used in FACS, immunoprecipitations, and biological assays such as those described below.
    TABLE 2
    ADAM Disintegrin Domain Polypeptide Constructs
    SEQ ID NOs: ADAM
    DNA/ IgK disintegrin1,3 Fc
    Construct polypeptide Leader1,2 (dis Framework)1,4 Region1
    ADAM- 1/2 1-20  23-264 267-494
    8dis-Fc (34-91)
    ADAM- 3/4 1-20  23-303 306-533
    9dis-Fc (34-92)
    ADAM- 5/6 1-20  23-235 238-465
    10dis-Fc (34-99)
    ADAM- 7/8 1-20  23-292 295-522
    15dis-Fc (34-92)
    ADAM-  9/10 1-20  23-216 219-446
    17dis-Fc (34-93)
    ADAM- 11/12 1-20  23-305 308-535
    20dis-Fc (34-91)
    ADAM- 13/14 1-20  23-293 296-523
    21dis-Fc (34-91)
    ADAM- 15/16 1-20  23-312 315-542
    22dis-Fc (34-92)
    ADAM- 17/18 1-20  23-310 313-540
    23dis-Fc (34-91)
    ADAM- 21/22 1-20  23-298 301-528
    29dis-Fc (34-91)

    1residues in the polypeptide sequence

    2the predicted cleavage site is after residue 20

    3segment of the construct that includes ADAMdis, but may also contain additional ADAM sequences

    4disintegrin framework, e.g., SEQ ID NO: 20
  • Example 2 Binding of ADAM Disintegrin Domain Polypeptides to Cells
  • A. Binding to Endothelial cells
  • This example describes a flow cytometric integrin mAb based binding inhibition assay, which is used to show binding of ADAM disintegrin-Fc polypeptides to integrins expressed on the surface of endothelial cells. Human endothelial cells express αvβ3, αvβ5, β1, β4, α1, α2, α3, α4, α5, and α6 integrins.
  • Primary human dermal microvascular endothelial cells (HMVEC-d) were maintained in supplemented endothelial growth medium (Clonetics Corporation, Walkersville, Md.). The ADAM disintegrin-Fc polypeptides produced in Example 1 were shown to bind specifically to HMVEC-d. Monoclonal antibodies specific for human integrins αvβ3 (LM609, anti CD51/61, Chemicon, Temecula, C A Brooks et al., Science 264:569, 1994), α 1 (BHA2.1 anti CD49b, Chemicon, Wang et al., Mol. Biol. of the Cell 9:865, 1998), α5β1 (SAM-1 anti CD49e, Biodesign, A. te Velde et al., J. Immunol. 140:1548, 1988), α3β1 (ASC-6 anti-CD49c, Chemicon, Pattaramalai et al., Exp. Cell. Res. 222: 281, 1996), α4β1 (HP2/1 anti CD49d, Immunotech, Marseilles, France. Workshop of the 4th International Conference on Human Leukocyte Differentiation Antigens, Vienna Austria, 1989, workshop number p091), α6β1 (GoH3 anti CD49f, Immunotech, Workshop 4th International Conference on Human Leukocyte Differentiation Antigens, workshop number p055), α6β4 (439-9B anti CD104, Pharmingen, San Diego, Calif., Schlossman et al., 1995 Leukocyte Typing V: White Cell Differentiation Antigens. Oxford University Press, New York), and αvβ5 (MAB 1961, Chemicon International, monoclonal anti-human integrin αvβ5 mAb, IgG1 isotype, inhibits αvβ5 mediated binding/adhesion to vitronectin/fibronectin; Weinaker, et al., J. Biol. Chem. 269:6940, 1994) were also shown to bind specifically to HMVEC-d. Each of these antibodies is known to specifically block binding of the indicated integrin to its ligands (e.g., fibronectin, vitronectin, fibrinogen). The ability of integrin mAbs to inhibit the binding of ADAM disintegrin-Fc polypeptides reveals which integrins the disintegrin domains bind and, indirectly, which integrin binding activities the disintegrin domains are able to antagonize. The ability of the antibodies to inhibit binding of the ADAM disintegrin-Fc polypeptides to endothelial cells was tested as described below.
  • Prior to performing binding studies, HMVEC-d were removed from culture vessels using trypsin-EDTA. The cells were washed in media containing serum and resuspended in binding medium which consisted of PBS containing 1 mM Ca2+, 1 mM Mg2+ and 0.5 mM Mn2+, 0.1% sodium azide, 10% Normal goat serum, 2% rabbit serum and 2% fetal bovine serum. Under these binding conditions, ADAM-8, -9, -10, -15, -17, -20, -21, -22, -23, and -29dis-Fc all bind to human endothelial cells.
  • One hundred microliters of cell suspension, containing 200,000 to 500,000 HMVEC-d, were added to 12×75 mm plastic test tubes. Monoclonal antibodies specific for one of the integrins, or a control monoclonal antibody (CD29 or M15), were added to the cell suspensions at a concentration of 100 μg/ml (5-8 fold mass excess) 15 minutes prior to addition of disintegrin-Fc fusion proteins. ADAM disintegrin-Fc polypeptides and control Fc fusion polypeptides (P7.5II.Fc) were added, at various concentrations from 12.5 to 20 μg/ml, to the cell suspensions and incubated for 1 hour at 30′ C. Unbound Fc polypeptides were washed away by centrifugation of cells in 2 mls of binding media. The washed cell pellets were resuspended in binding medium and then incubated at 30° C. for 30 minutes with goat anti-human Fc-specific biotinylated antibody at a concentration of 2.5 μg/ml for 30 minutes. After centrifugation and washing of the cell pellets, the cells were resuspended in binding medium and bound anti-human Fc-biotin was detected by adding streptavidin-phycoerythrin conjugate to the cell suspension at a 1:1000 dilution (1 μg/ml) and incubating at 30° C. for 30 minutes. The unbound streptavidin-phycoerythrin was washed away and the cells were resuspended in binding medium containing propidum iodide. The level of fluorescent binding (disintegrin-Fc binding) was determined by flow cytometry.
  • The level of binding of each ADAM disintegrin-Fc polypeptide was determined in the presence of anti-integrin specific mAb and in the presence of control mAb. Both the intensity of binding (MFI) and the percentage of cells binding were determined. Percent inhibition was calculated using the formula [1−(MFI control−MFI integrin mAb)/MFI control. The results of these studies are summarized in Table 3.
  • ADAM-15, -17, -20 and -22 disintegrin domain polypeptides bound to αvβ3; ADAM 23 disintegrin domain polypeptide bound to α2β1; ADAM-15, -21, -22 and -23 disintegrin domain polypeptides bound to α5β1; ADAM-10, -17, -22 and -23 disintegrin domain polypeptides bound to the α6 integrins; ADAM-10 and -15 disintegrin domain polypeptides bound to αvβ5. An excess of a non blocking αvβ5 antibody did significantly affect the binding of ADAM-10, -22, and -23 disintegrin polypeptides to endothelial cells, suggesting that these ADAMdis polypeptides interact with integrin sites other than or in addition to the ligand (e.g., fibronectin, vitronectin) binding site. Based upon results from a different type of assay, Cal et al. have reported that the ADAM-23 disintegrin domain interacts with the αvβ3 integrin through an RGD-independent mechanism (Molec. Biol. of the Cell 11:1457, 2000).
  • Binding experiments are repeated using other ADAM disintegrin domains and other monoclonal antibodies. ADAM disintegrin-Fc polypeptides that bind to selected integrins are further tested for the ability to disrupt integrin-ligand interactions and to modulate endothelial cell function, angiogenesis, and other biological activities in vitro and in vivo.
    TABLE 3
    Binding of ADAM Disintegrin-Fc Polypeptides to
    Integrins Expressed on Human Endothelial Cells
    Integrin
    Binding1 (+ or − or ND, not done) and Percent (%) Binding2
    ADAM αvβ3 α2β1 α3β1 α4β1 α5β1 α6β1, α6β4 αvβ5
    ADAM-8 ND ND − (<10) − (<10) ND ND − (<20)
    ADAM-9 − (<10) − (<10) − (<10) − (<20) − (<10) − (<10) − (<10)
    ADAM-10 − (<10) − (<10) − (<10) − (<20) − (<10) + (48) + (25)
    ADAM-15 + (60) − (<10) − (<10) − (<20) + (30) − (<10) + (25)
    ADAM-17 + (50) − (<10) − (<10) − (<10) − (<10) + (69) − (<10)
    ADAM-20 + (58) − (<10) − (<10) − (<10) − (<20) − (<10) − (<10)
    ADAM-21 − (<10) − (<10) − (<10) − (<10) + (54) − (<10) − (<10)
    ADAM-22 + (42) − (<10) − (<10) − (<10) + (36) + (32) − (<10)
    ADAM-23 − (<10) + (22) − (<10) − (<10) + (49) + (31) − (<10)

    1positive binding defined as >20% binding inhibition; normal background variation 5-10%, baseline positive approx. 2× over background

    2percent inhibition of binding by ADAM-dis-Fc in the presence of 5-8 fold excess integrin mAb as compared to control mAb

    B. Binding to Primary Human T-Cells
  • Primary human T-cells were purified from whole blood. These cells were used in FACS experiments to assess cell surface binding of purified ADAMdis-Fc polypeptides. ADAMdis-Fc binding was assessed with and without Con A (5 μg/ml) or immobilized OTK3 antibody (1 mg/ml, immobilized for 1 hour, 37° C.) stimulation. ADAMdis-Fc polypeptides (20 μg/ml) were bound at either 4° C. or 30° C. in the presence of cations (Ca++, Mg++, Mn++, 0.5 mM each). Cell surface integrin expression was assessed using a panel of murine and rat anti-human integrin antibodies. αvβ5, α1, α3, α4, α6, β1, and β7 integrins were detected on the surface of these cells. ADAMdis-Fc polypeptides did not bind to primary human T-cells at 4° C. ADAM-8-, ADAM-9-, ADAM-15-, ADAM-20-, ADAM-21-, ADAM-22-, and ADAM-23-dis-Fc polypeptides did bind primary T-cells at 30° C. with Con A stimulation. ADAMdis-Fc binding was not inhibited by a three-fold molar excess of antibodies to the integrins listed above.
  • C. Binding to Resting Platelets
  • Binding of ADAMdis-Fc polypeptides to citrated washed resting platelets was performed at 4° C. or 30° C. Binding was analyzed by flow cytometry using a biotinylated-anti-human Fc specific antibody and streptavidin-PE. Resting platelets express the integrins CD41/CD61 and CD49e. ADAM-9dis-Fc and ADAM-8dis-Fc bound resting platelets at 30° C. but not at 4° C. ADAM-9dis-Fc binding to resting platelets at 30° C. was not inhibited by a ten-fold excess of CD41a mAb.
  • Example 3 Activity of ADAM Disintegrin Domain Polypeptides in a Wound Closure Assay
  • A planar endothelial cell migration (wound closure) assay was used to quantitate the inhibition of angiogenesis by ADAM disintegrin-Fc polypeptides in vitro. In this assay, endothelial cell migration is measured as the rate of closure of a circular wound in a cultured cell monolayer. The rate of wound closure is linear, and is dynamically regulated by agents that stimulate and inhibit angiogenesis in vivo.
  • Primary human renal microvascular endothelial cells, HRMEC, were isolated, cultured, and used at the third passage after thawing, as described in Martin et al., In Vitro Cell Dev Biol 33:261, 1997. Replicate circular lesions, “wounds,” (600-800 micron diameter) were generated in confluent HRMEC monolayers using a silicon-tipped drill press. At the time of wounding the medium (DMEM+1% BSA) was supplemented with 20 ng/ml PMA (phorbol-12-myristate-13-acetate), a range of concentrations of ADAM disintegrin-Fc polypeptide, or combinations of PMA and ADAM disintegrin-Fc polypeptide. The residual wound area was measured as a function of time (0-12 hours) using a microscope and image analysis software (Bioquant, Nashville, Tenn.). The relative migration rate was calculated for each agent and combination of agents by linear regression of residual wound area plotted over time. The inhibition of PMA-induced endothelial migration by ADAM disintegrin-Fc polypeptides is shown in Table 4.
  • The effect of ADAM-dis-Fc polypeptides on EGF-induced migration was also determined. For these experiments EGF (epidermal growth factor, 40 ng/ml) was added to the medium, instead of PMA, at the time of wounding. The results are shown in Table 5.
    TABLE 4
    Effect of ADAM-15, -17, -20, and -23dis-Fc Polypeptides in
    PMA-Induced Endothelial Cell Wound Closure Migration Assay
    PMA + PMA + PMA + PMA +
    No PMA PMA + ADAM- ADAM- ADAM- ADAM-
    Expt. ID Addition 20 ng/ml IgG 15dis-Fc 17dis-Fc 20dis-Fc 23dis-Fc
    HL-H-142 0.04361 0.0655 0.0499
    15 μg/ml (0.0016)2 (0.0004) (0.0009)
    dis-Fc 72%3
    HL-H-147 0.0244 0.0424 0.0449 0.0357 0.0225
    15 μg/ml (0.0023) (0.0002) (0.0012) (0.0007) (0.0022)
    dis-Fc 0% 37% 100%
    HL-H-153 0.0253 0.0460 0.0491 0.0392 0.0388 0.0317
    15 μg/ml  0.00013 (0.0022) (0.006)  (0.0016) (0.005)  (0.005) 
    dis-Fc 0% 33% 36%  70%
    HL-H-154 0.0119 0.0312 0.0283 0.0160
    15 μg/ml (0.0012) (0.0016) (0.0008) (0.0017)
    dis-Fc 15% 79%

    1Slopes to average triplicate Y values and treat as a single data point in order to test whether the slopes are significantly different

    2Data in parentheses is the +/− standard error of slopes

    3Percent inhibition compared to migration rate observed in the presence of PMA
  • TABLE 5
    Effect of ADAM-17, -20, and -23dis-Fc Polypeptides in EGF-Induced
    Endothelial Cell Wound Closure Migration Assay
    EGF + EGF + EGF +
    No EGF EGF + ADAM- ADAM- ADAM-
    Expt. ID Addition 40 ng/ml IgG 17dis-Fc 20dis-Fc 23dis-Fc
    HL-H-154 0.0119 0.0378 0.0242 0.0172 0.0310
    15 μg/ml (0.0012) (0.0061) (0.0029) (0.0031) (0.0036)
    dis-Fc 53% 80% 26%
    HL-H-155 0.0164 0.0468 0.0454 0.0412 0.0227 0.0207
    9 μg/ml (0.0010) (0.0059) (0.0052) (0.0107) (0.0035) (0.0016)
    dis-Fc 5% 18% 79% 86%

    1Slopes to average triplicate Y values and treat as a single data point in order to test whether the slopes are significantly different

    2Data in parentheses is the +/− standard error of slopes

    3Percent inhibition compared to migration rate observed in the presence of EGF alone
  • ADAM-20 and -23dis-Fc polypeptides showed the greatest inhibition of both EGF- and PMA-induced endothelial migration at 15 μg/ml. ADAM-15 and -17dis-Fc polypeptides were less effective at inhibiting endothelial cell migration at 15 μg/ml. Hu IgG did not inhibit EGF- or PMA-induced endothelial cell migration in any of the experiments performed where it was included as a control Fc protein.
  • Example 4 Activity of ADAM Disintegrin Domain Polypeptides in a Corneal Pocket Assay
  • A mouse corneal pocket assay is used to quantitate the inhibition of angiogenesis by ADAM disintegrin-Fc polypeptides in vivo. In this assay, agents to be tested for angiogenic or anti-angiogenic activity are immobilized in a slow release form in a hydron pellet, which is implanted into micropockets created in the corneal epithelium of anesthetized mice. Vascularization is measured as the appearance, density, and extent of vessel ingrowth from the vascularized corneal limbus into the normally avascular cornea.
  • Hydron pellets, as described in Kenyon et al., Invest Opthamol. & Visual Science 37:1625, 1996, incorporate sucralfate with bFGF (90 ng/pellet), bFGF and IgG (11 μg/pellet, control), or bFGF and a range of concentrations of ADAM disintegrin-Fc polypeptide. The pellets are surgically implanted into corneal stromal micropockets created by micro-dissection 1 mm medial to the lateral corneal limbus of 6-8 week old male C57BL mice. After five days, at the peak of neovascular response to bFGF, the corneas are photographed, using a Zeiss slit lamp, at an incipient angle of 35-50° from the polar axis in the meridian containing the pellet. Images are digitized and processed by subtractive color filters (Adobe Photoshop 4.0) to delineate established microvessels by hemoglobin content. Image analysis software (Bioquant, Nashville, Tenn.) is used to calculate the fraction of the corneal image that is vascularized, the vessel density within the vascularized area, and the vessel density within the total cornea. The inhibition of bFGF-induced corneal angiogenesis, as a function of the dose of ADAM disintegrin-Fc polypeptide, is determined.
  • Example 5 Inhibition of Neovascularization by ADAM Disintegrin Domain Polypeptides in a Murine Transplant Model
  • Survival of heterotopically transplanted cardiac tissue from one mouse donor to the ear skin of another genetically similar mouse requires adequate neovascularization by the transplanted heart and the surrounding tissue, to promote survival and energy for cardiac muscle function. Inadequate vasculature at the site of transplant causes excessive ischemia to the heart, tissue damage, and failure of the tissue to engraft. Agents that antagonize factors involved in endothelial cell migration and vessel formation can decrease angiogenesis at the site of transplant, thereby limiting graft tissue function and ultimately engraftment itself. A murine heterotopic cardiac isograft model is used to demonstrate the antagonistic effects of ADAM disintegrin-Fc polypeptides on neovascularization. Female BALB/c (12 weeks of age) recipients are given neonatal heart grafts from donor mice of the same strain. The donor heart tissue is grafted into the left ear pinnae of the recipient on day 0 and the mice are divided into two groups. The control group receives human IgG (Hu IgG) while the other group receives ADAM disintegrin-Fc polypeptide, both intraperitoneally. The treatments are continued for five consecutive days. The functionality of the grafts is determined by monitoring visible pulsatile activity on days 7 and 14 post-engraftment. The inhibition of functional engraftment, as a function of the dose of ADAM disintegrin-Fc polypeptide, is determined. The histology of the transplanted hearts is examined is order to visualize the effects of ADAM disintegrin-Fc polypeptides on edema at the site of transplant and host and donor tissue vasculature (using, e.g., Factor VIII staining).
  • Example 6 Treatment of Tumors with ADAM Disintegrin Domain Polypeptides
  • ADAM disintegrin-Fc polypeptides are tested in animal models of solid tumors. The effect of the ADAM disintegrin-Fc polypeptides is determined by measuring tumor frequency and tumor growth.
  • The biological activity of ADAM disintegrin-Fc polypeptides is also demonstrated in other in vitro, ex vivo, and in vivo assays known to the skilled artisan, such as calcium mobilization assays and assays to measure platelet activation, recruitment, or aggregation.
  • The relevant disclosures of publications cited herein are specifically incorporated by reference. The examples presented above are not intended to be exhaustive or to limit the scope of the invention. The skilled artisan will understand that variations and modifications and variations are possible in light of the above teachings, and such modifications and variations are intended to be within the scope of the invention.

Claims (35)

1. A method of antagonizing the binding of an integrin to its ligands comprising contacting a cell that expresses the integrin with an effective amount of an ADAM disintegrin domain polypeptide.
2. A method according to claim 1 wherein the ADAM disintegrin domain polypeptide is administered to a mammal in need of such treatment.
3. The method of claim 2 wherein the mammal is afflicted with a condition selected from the group consisting of ocular disorders, malignant and metastatic conditions, inflammatory diseases, osteoporosis and other conditions mediated by accelerated bone resorption, restenosis, inappropriate platelet activation, recruitment, or aggregation, thrombosis, or a condition requiring tissue repair or wound healing.
4. A method according to claim 2 wherein the method is a method of inhibiting angiogenesis in a mammal in need of such treatment, comprising administering to the mammal an inhibition-effective amount of an ADAM disintegrin domain polypeptide, wherein the disintegrin domain does not contain an RGD sequence.
5. The method of claim 2 wherein the ADAM disintegrin domain is in the form of a multimer.
6. The method of claim 5 wherein the multimer is a dimer or trimer.
7. The method of claim 5 wherein the multimer comprises an Fc polypeptide or a leucine zipper.
8. The method of claim 2 wherein the ADAM disintegrin domain is from a human ADAM.
9. The method of claim 8 wherein the ADAM disintegrin domain is from an ADAM selected from the group consisting of ADAM-8, ADAM-9, ADAM-10, ADAM-15, ADAM-17, ADAM-20, ADAM-21, ADAM-22, ADAM-23, and ADAM-29.
10. The method of claim 9 wherein the ADAM disintegrin domain is from ADAM-17, ADAM-20, or ADAM-23.
11. The method of claim 2 wherein the ADAM disintegrin domain polypeptide comprises an amino acid sequence selected from the group consisting of:
(a) amino acids 1-494 of SEQ ID NO:2, amino acids 23-264 of SEQ ID NO:2, amino acids 1-533 of SEQ ID NO:4, amino acids 23-303 of SEQ ID NO:4, amino acids 1-465 of SEQ ID NO:6, amino acids 23-235 of SEQ ID NO:6, amino acids 1-522 of SEQ ID NO:8, amino acids 23-292 of SEQ ID NO:8, amino acids 1-446 of SEQ ID NO:10, amino acids 23-216 of SEQ ID NO:10, amino acids 1-535 of SEQ ID NO:12, amino acids 23-305 of SEQ ID NO:12, amino acids 1-523 of SEQ ID NO:14, amino acids 23-293 of SEQ ID NO:14, amino acids 1-542 of SEQ ID NO:16, amino acids 23-312 of SEQ ID NO:16, amino acids 1-540 of SEQ ID NO:18, amino acids 23-310 of SEQ ID NO:18, amino acids 1-528 of SEQ ID NO:22, amino acids 23-298 of SEQ ID NO:22;
(b) fragments of the polypeptides of (a) wherein said fragments retain at least one ADAMdis activity; and
(c) fusion polypeptides comprising the polypeptides of (a) or (b), wherein said fusion polypeptides retain at least one ADAMdis activity,
wherein the ADAMdis activity is selected from the group consisting of integrin binding activity, inhibition of endothelial cell migration, and inhibition of angiogenesis.
12. The method of claim 11 wherein the ADAM disintegrin domain comprises an amino acid sequence selected from the group consisting of amino acids 34-91 of SEQ ID NO:2, 34-92 of SEQ ID NO:4, 34-99 of SEQ ID NO:6, 34-92 of SEQ ID NO:8, 34-93 of SEQ ID NO:10, 34-91 of SEQ ID NO:12, 34-91 of SEQ ID NO:14, 34-92 of SEQ ID NO:16, 34-91 of SEQ ID NO:18, or 34-91 of SEQ ID NO:22.
13. The method of claim 2 wherein the ADAM disintegrin domain polypeptide is a variant that is at least 70%, 80%, 90%, 95%, 98%, or 99% identical in amino acid sequence to a polypeptide selected from the group consisting of:
(a) amino acids 1-494 of SEQ ID NO:2, amino acids 23-264 of SEQ ID NO:2, amino acids 1-533 of SEQ ID NO:4, amino acids 23-303 of SEQ ID NO:4, amino acids 1-465 of SEQ ID NO:6, amino acids 23-235 of SEQ ID NO:6, amino acids 1-522 of SEQ ID NO:8, amino acids 23-292 of SEQ ID NO:8, amino acids 1-446 of SEQ ID NO:10, amino acids 23-216 of SEQ ID NO:10, amino acids 1-535 of SEQ ID NO:12, amino acids 23-305 of SEQ ID NO:12, amino acids 1-523 of SEQ ID NO:14, amino acids 23-293 of SEQ ID NO:14, amino acids 1-542 of SEQ ID NO:16, amino acids 23-312 of SEQ ID NO:16, amino acids 1-540 of SEQ ID NO:18, amino acids 23-310 of SEQ ID NO:18, amino acids 1-528 of SEQ ID NO:22, amino acids 23-298 of SEQ ID NO:22; and
(b) fragments of the polypeptides of (a),
wherein said variant polypeptide retains at least one ADAMdis activity,
wherein the ADAMdis activity is selected from the group consisting of integrin binding activity, inhibition of endothelial cell migration, and inhibition of angiogenesis.
14. The method of claim 2 wherein the ADAM disintegrin domain polypeptide is encoded by a nucleic acid comprising a sequence selected from the group consisting of:
(a) nucleotides 118-1599 of SEQ ID NO:1, nucleotides 184-909 of SEQ ID NO:1, nucleotides 46-1644 of SEQ ID NO:3, nucleotides 112-954 of SEQ ID NO:3, nucleotides 25-1419 of SEQ ID NO:5, nucleotides 91-729 of SEQ ID NO:5, nucleotides 41-1606 of SEQ ID NO:7, nucleotides 107-916 of SEQ ID NO:7, nucleotides 25-1362 of SEQ ID NO:9, nucleotides 91-672 of SEQ ID NO:9, nucleotides 25-1629 of SEQ ID NO:11, nucleotides 91-939 of SEQ ID NO:11, nucleotides 25-1593 of SEQ ID NO:13, nucleotides 91-903 of SEQ ID NO:13, nucleotides 25-1650 of SEQ ID NO:15, nucleotides 91-960 of SEQ ID NO:15, nucleotides 25-1644 of SEQ ID NO:17, nucleotides 91-954 of SEQ ID NO:17, nucleotides 118-1701 of SEQ ID NO:21, nucleotides 184-1011 of SEQ ID NO:21;
(b) sequences which, due to the degeneracy of the genetic code, encode a polypeptide encoded by a nucleic acid of (a); and
(c) sequences that hybridize under conditions of moderate or high stringency to a sequence of (a) or (b) and that encode a polypeptide that retains at least one ADAMdis activity,
wherein the ADAMdis activity is selected from the group consisting of integrin binding activity, inhibition of endothelial cell migration, and inhibition of angiogenesis.
15. The method of claim 11 wherein the ADAM disintegrin domain polypeptide has been produced by culturing a recombinant cell that encodes the ADAM disintegrin domain polypeptide under conditions permitting expression of the ADAM disintegrin domain polypeptide, and recovering the ADAM disintegrin domain polypeptide.
16. The method of claim 11 wherein the ADAM disintegrin domain polypeptide is present in a composition comprising a pharmaceutically acceptable carrier.
17. The method of claim 11 wherein the mammal has a disease or condition mediated by angiogenesis.
18. The method of claim 17 wherein the disease or condition is characterized by ocular neovascularization.
19. The method of claim 17 wherein the disease or condition is a solid tumor.
20. The method of claim 11 wherein the method further comprises treating the mammal with radiation.
21. The method of claim 11 wherein the method further comprises treating the mammal with a second therapeutic agent.
22. The method of claim 21 wherein the second therapeutic agent is selected from the group consisting of alkylating agents, antimetabolites, vinca alkaloids and other plant-derived chemotherapeutics, antitumor antibiotics, antitumor enzymes, topoisomerase inhibitors, platinum analogs, adrenocortical suppressants, hormones and antihormones, antibodies, immunotherapeutics, radiotherapeutics, and biological response modifiers.
23. The method of claim 21 wherein the second therapeutic agent is selected from the group consisting of cisplatin, cyclophosphamide, bleomycin, carboplatin, fluorouracil, 5-fluorouracil, 5-fluorodeoxyuridine, methotrexate, taxol, asparaginase, vincristine, vinblastine, mechloretamine, melphalan, 5-fluorodeoxyuridine, lymphokines and cytokines such as interleukins, interferons (alpha., beta, or delta.) and TNF, chlorambucil, busulfan, carmustine, lomustine, semustine, streptozocin, dacarbazine, cytarabine, mercaptopurine, thioguanine, vindesine, etoposide, teniposide, dactinomycin, daunorubicin, doxorubicin, bleomycin, plicamycin, mitomycin, L-asparaginase, hydroxyurea, methylhydrazine, mitotane, tamoxifen, fluoxymesterone, and COX-2 inhibitors.
24. The method of claim 21 wherein the second therapeutic agent is a polypeptide, including soluble forms thereof, selected from the group consisting of Flt3 ligand, CD40 ligand, interleukin-2, interleukin-12, 4-1BB ligand, anti-4-1BB antibodies, TRAIL, TNF antagonists and TNF receptor antagonists including TNFR/Fc, Tek antagonists, TWEAK antagonists and TWEAK-R antagonists including TWEAK-R/Fc, VEGF antagonists including anti-VEGF antibodies, VEGF receptor antagonists, CD148 binding proteins, and nectin-3 antagonists.
25. The method of claim 11 wherein the ADAM disintegrin domain is administered parenterally.
26. A method for inhibiting the biological activity of an integrin selected from the group consisting of αvβ3, α2β1, α5β1, α6β1, α6β4, and αvβ5 comprising contacting the integrin with an inhibition-effective amount of an ADAM disintegrin domain polypeptide.
27. The method of claim 26 wherein:
(a) the integrin is αvβ3 and the ADAM is ADAM-17, ADAM-20, or ADAM-22;
(b) the integrin is α2β1 and the ADAM is ADAM-23;
(c) the integrin is α5β1 and the ADAM is ADAM-15, ADAM-21, ADAM-22, or ADAM-23;
(d) the integrin is α6β1 or β6β4 and the ADAM is ADAM-0, ADAM-17, ADAM-22, or ADAM-23; or
(e) the integrin is αvβ5 and the ADAM is ADAM-10, ADAM-15, or ADAM-23.
28. A method for identifying a compound that modulates integrin biological activity and/or modulates the interaction between an integrin and an ADAM disintegrin domain comprising:
(a) combining a test compound with an integrin and an ADAM disintegrin domain polypeptide that binds to the integrin; and
(b) determining whether the test compound alters the binding of the ADAM disintegrin domain polypeptide to the integrin.
29. The method of claim 28 wherein the integrin is present on a cell surface.
30. The method of claim 28 wherein the integrin is selected from the group consisting of αvβ3, α2β1, α5β1, α6β1, α6β4, and αvβ5.
31. A method for identifying a compound that inhibits endothelial cell migration and/or angiogenesis comprising:
(a) combining a test compound with endothelial cells and with an ADAM disintegrin domain polypeptide that binds to endothelial cells; and
(b) determining whether the test compound alters the binding of the ADAM disintegrin domain polypeptide to the endothelial cells.
32. The method of claim 30 wherein the ADAM disintegrin domain polypeptide comprises an ADAM disintegrin domain from ADAM-8, ADAM-9, ADAM-10, ADAM-15, ADAM-17, ADAM-20, ADAM-21, ADAM-22, ADAM-23, or ADAM-29.
33. The method of claim 30 wherein the ADAM disintegrin domain polypeptide comprises an ADAM disintegrin domain from ADAM-17, ADAM-20, or ADAM-23.
34. The method of claim 31 wherein the ADAM disintegrin domain polypeptide comprises an ADAM disintegrin domain from ADAM-8, ADAM-9, ADAM-10, ADAM-15, ADAM-17, ADAM-20, ADAM-21, ADAM-22, ADAM-23, or ADAM-29.
35. The method of claim 31 wherein the ADAM disintegrin domain polypeptide comprises an ADAM disintegrin domain from ADAM-17, ADAM-20, or ADAM-23.
US11/392,436 2000-02-25 2006-03-28 Integrin antagonists Abandoned US20060177443A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/392,436 US20060177443A1 (en) 2000-02-25 2006-03-28 Integrin antagonists

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US18486500P 2000-02-25 2000-02-25
US09/792,200 US7074408B2 (en) 2000-02-25 2001-02-23 Use of integrin antagonists to inhibit angiogenesis
US11/392,436 US20060177443A1 (en) 2000-02-25 2006-03-28 Integrin antagonists

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/792,200 Division US7074408B2 (en) 2000-02-25 2001-02-23 Use of integrin antagonists to inhibit angiogenesis

Publications (1)

Publication Number Publication Date
US20060177443A1 true US20060177443A1 (en) 2006-08-10

Family

ID=22678680

Family Applications (2)

Application Number Title Priority Date Filing Date
US09/792,200 Expired - Fee Related US7074408B2 (en) 2000-02-25 2001-02-23 Use of integrin antagonists to inhibit angiogenesis
US11/392,436 Abandoned US20060177443A1 (en) 2000-02-25 2006-03-28 Integrin antagonists

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US09/792,200 Expired - Fee Related US7074408B2 (en) 2000-02-25 2001-02-23 Use of integrin antagonists to inhibit angiogenesis

Country Status (10)

Country Link
US (2) US7074408B2 (en)
EP (1) EP1259595B1 (en)
JP (1) JP2003523768A (en)
AT (1) ATE358718T1 (en)
AU (2) AU2001247219B2 (en)
CA (1) CA2400040A1 (en)
DE (1) DE60127656T2 (en)
IL (1) IL151032A0 (en)
NZ (1) NZ521437A (en)
WO (1) WO2001062905A2 (en)

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030148924A1 (en) * 2002-07-09 2003-08-07 Tamar Tennenbaum Methods and pharmaceutical compositions of healing wounds
US20040171549A1 (en) * 2001-07-26 2004-09-02 Inserm, A Corporation Of France Method of inhibiting angiogenesis or invasion or formation of metastases
US20060177418A1 (en) * 2003-08-07 2006-08-10 Healor Ltd. Methods for accelerating wound healing by administration of adipokines
EP1970441A1 (en) * 2007-03-06 2008-09-17 BioAlliance Pharma Plasmid containing a sequence encoding a disintegrin domain of metargidin (RDD)
US20080280816A1 (en) * 2001-07-23 2008-11-13 Tamar Tennenbaum Methods and pharmaceutical compositions for healing wounds
US20100129332A1 (en) * 2000-07-31 2010-05-27 Tamar Tennenbaum Methods and pharmaceutical compositions for healing wounds
US20100215634A1 (en) * 2009-02-24 2010-08-26 Tamar Tennenbaum Visfatin therapeutic agents for the treatment of acne and other conditions
US20100310542A1 (en) * 2007-07-30 2010-12-09 Healor Ltd. Pharmaceutical Compositions for treating wouds and related methods
US20110021422A1 (en) * 2005-08-29 2011-01-27 Tamar Tennenbaum Method and compositions for prevention and treatment of diabetic and aged skin
US8349793B2 (en) 2010-01-11 2013-01-08 Heal0r, Ltd. Method for treatment of inflammatory disease and disorder
WO2014066084A1 (en) * 2012-10-24 2014-05-01 Novamedica Limited Liability Company Nucleic acid modulators of alpha2beta1
WO2017001990A1 (en) * 2015-06-28 2017-01-05 Allgenesis Biotherapeutics Inc. Fusion proteins for inhibiting angiogenesis
WO2020080836A1 (en) * 2018-10-19 2020-04-23 아이진 주식회사 Pharmaceutical composition for preventing or treating inflammatory diseases
US11723955B1 (en) 2022-05-13 2023-08-15 Allgenesis Biotherapeutics Inc. VEGFR fusion protein pharmaceutical composition

Families Citing this family (126)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7129061B1 (en) * 1996-08-07 2006-10-31 Biogen Idec Ma Inc. Tumor necrosis factor related ligand
CA2308110A1 (en) * 1997-10-30 1999-05-14 Immunex Corporation Svph1-26 dna and polypeptides
NZ506817A (en) * 1998-02-11 2003-08-29 Immunex Corp Metalloprotease-disintegrin SVPH3-13 and SVPH3-17 nucleic acids, and their corresponding polypeptides and methods for their production
US6265199B1 (en) * 1998-07-10 2001-07-24 Zymogenetics, Inc. Disintegrin homologs
KR20010102978A (en) * 1999-01-15 2001-11-17 아스트루 마이클 제이 Antagonists of tweak and of tweak receptor and their use to treat immunological disorders
CA2356681A1 (en) * 1999-01-21 2000-07-27 Immunex Corporation Metalloproteinase-disintegrin family members: svph dnas and polypeptides
CA2378182C (en) * 1999-07-28 2014-09-23 Genentech, Inc. Compositions and methods for the treatment of tumors
PT1234031T (en) 1999-11-30 2017-06-26 Mayo Foundation B7-h1, a novel immunoregulatory molecule
EP1268756A2 (en) * 2000-04-03 2003-01-02 Bristol-Myers Squibb Company Methods and compositions for modulating integrin-mediated cell-cell interactions
US20020168363A1 (en) * 2000-04-21 2002-11-14 Amgen Inc. Integrin/adhesion antagonists
AU5951901A (en) * 2000-05-08 2001-11-20 Biogen Inc Method for promoting neovascularization
US6458552B1 (en) * 2000-06-06 2002-10-01 Ortho-Mcneil Pharmaceutical, Inc. Metalloprotease peptide substrates and methods
US7208151B2 (en) * 2001-09-12 2007-04-24 Biogen Idec Ma Inc. Tweak receptor agonists as anti-angiogenic agents
AU2001295038A1 (en) * 2000-09-18 2002-03-26 Merck & Co., Inc. Treatment of inflammation with a combination of a cyclooxygenase-2 inhibitor and an integrin alpha-V antagonist
US6294368B1 (en) 2001-03-22 2001-09-25 Applera Corporation Isolated human metalloprotease proteins, nucleic acid molecules encoding human protease proteins, and uses thereof
AU2002364935A1 (en) 2001-10-09 2003-06-23 Mayo Foundation For Medical Education And Research Enhancement of immune responses by 4-1bb-binding agents
SI1997512T1 (en) 2002-04-09 2014-03-31 Biogen Idec Ma Inc. Methods for treating TWEAK-related conditions
BR0313491A (en) * 2002-08-16 2007-08-14 Wyeth Corp compositions and methods for treating rage-associated disorders
EP1601330A4 (en) * 2003-03-04 2008-05-07 Ghc Res Dev Corp Antitumor agents comprising a targeting portion and an immune response triggering portion
EP2594304B1 (en) 2003-04-11 2014-12-24 Ambu A/S Laryngeal mask and method for the manufacture thereof
JP5105874B2 (en) 2003-07-18 2012-12-26 アムジエン・インコーポレーテツド Specific binding factor for hepatocyte growth factor
US20060051345A1 (en) 2004-06-04 2006-03-09 Genentech, Inc. Method for treating multiple sclerosis
US7507748B2 (en) * 2004-07-22 2009-03-24 Amgen Inc. Substituted aryl-amine derivatives and methods of use
MX2007004176A (en) 2004-10-06 2007-06-15 Mayo Foundation B7-h1 and methods of diagnosis, prognosis, and treatment of cancer.
CA2596273C (en) * 2005-02-02 2017-11-14 Raven Biotechnologies, Inc. Adam-9 modulators
US20090124993A1 (en) 2005-02-17 2009-05-14 Burkly Linda C Treating neurological disorders
TW200714289A (en) * 2005-02-28 2007-04-16 Genentech Inc Treatment of bone disorders
WO2006122187A2 (en) 2005-05-10 2006-11-16 Biogen Idec Ma Inc. Treating and evaluating inflammatory disorders
WO2006138219A2 (en) 2005-06-13 2006-12-28 Biogen Idec Ma Inc. Methods of diagnosis / prognosis of inflammatory conditions
US20070048325A1 (en) * 2005-08-24 2007-03-01 Dennis Van Epps Combination therapies for inhibiting integrin-extracellular matrix interactions
JP2009512423A (en) * 2005-09-12 2009-03-26 ザ・ジョンズ・ホプキンス・ユニバーシティ Composition having anti-angiogenic activity and use thereof
MY149159A (en) 2005-11-15 2013-07-31 Hoffmann La Roche Method for treating joint damage
US7354904B2 (en) * 2005-12-08 2008-04-08 The United States Of America As Represented By The Secretary Of Agriculture Bacteriocin inducer peptides
US9101581B2 (en) * 2008-10-27 2015-08-11 Eyegene Inc. Method for treating vascular-related disease
CN101379082A (en) * 2006-01-19 2009-03-04 爱吉恩公司 Pharmaceutical composition for treating vascular-related diseases comprising peptide
KR101201886B1 (en) * 2006-01-19 2012-11-15 아이진 주식회사 Pharmaceutical composition for treating vascular-related diseases comprising peptide
KR100797567B1 (en) 2006-04-27 2008-01-24 연세대학교 산학협력단 Protein for inhibiting secretion of platelet granules
PE20121506A1 (en) 2006-07-14 2012-11-26 Amgen Inc TRIAZOLOPYRIDINE COMPOUNDS AS C-MET INHIBITORS
US8217177B2 (en) 2006-07-14 2012-07-10 Amgen Inc. Fused heterocyclic derivatives and methods of use
WO2008103277A2 (en) 2007-02-16 2008-08-28 Amgen Inc. Nitrogen-containing heterocyclyl ketones and their use as c-met inhibitors
US8398977B2 (en) 2007-06-14 2013-03-19 Galactica Pharmaceuticals, Inc. Rage fusion proteins
CL2008002444A1 (en) 2007-08-21 2009-09-04 Amgen Inc Antibody or fragment thereof that binds to human c-fms protein; nucleic acid molecule that encodes it; vector and host cell; production method; pharmaceutical composition comprising it; and its use to treat or prevent a condition associated with c-fms in a patient.
JP5526407B2 (en) * 2008-02-14 2014-06-18 株式会社ジーンテクノサイエンス Anti-ADAM-15 antibody and use thereof
TW201014605A (en) * 2008-09-16 2010-04-16 Genentech Inc Methods for treating progressive multiple sclerosis
WO2010075249A2 (en) 2008-12-22 2010-07-01 Genentech, Inc. A method for treating rheumatoid arthritis with b-cell antagonists
US8663210B2 (en) 2009-05-13 2014-03-04 Novian Health, Inc. Methods and apparatus for performing interstitial laser therapy and interstitial brachytherapy
EP2353607A1 (en) * 2010-02-04 2011-08-10 BioAlliance Pharma Use of disintegrin domain of an adamalysin for the treatment of psoriasis
US20110319335A1 (en) 2010-06-23 2011-12-29 Xiaodong Feng Combined administration of integrin receptor antagonists for anti-angiogenic therapy
DK2688887T3 (en) 2011-03-23 2015-06-29 Amgen Inc DEHYDRATED tricyclic DUALINHIBITORER OF CDK 4/6 AND FLT3
US9745288B2 (en) 2011-08-16 2017-08-29 Indiana University Research And Technology Corporation Compounds and methods for treating cancer by inhibiting the urokinase receptor
US8946159B2 (en) * 2011-12-22 2015-02-03 California Northstate College Of Pharmacy, Llc Administration of an antagonist of α5β1 for anti-angiogenesis and cancer treatment
WO2013101771A2 (en) 2011-12-30 2013-07-04 Genentech, Inc. Compositions and method for treating autoimmune diseases
WO2013116287A1 (en) 2012-01-31 2013-08-08 Genentech, Inc. Anti-ig-e m1' antibodies and methods using same
AR090263A1 (en) 2012-03-08 2014-10-29 Hoffmann La Roche COMBINED ANTIBODY THERAPY AGAINST HUMAN CSF-1R AND USES OF THE SAME
JP6200437B2 (en) 2012-03-16 2017-09-20 ユニバーシティ ヘルス ネットワーク Methods and compositions for modulating TOSO activity
US9505749B2 (en) 2012-08-29 2016-11-29 Amgen Inc. Quinazolinone compounds and derivatives thereof
US9302005B2 (en) 2013-03-14 2016-04-05 Mayo Foundation For Medical Education And Research Methods and materials for treating cancer
EP3029071B1 (en) * 2013-07-30 2018-07-11 Industry-Academic Cooperation Foundation, Yonsei University Saxatilin-fc fusion protein and use thereof
TW201605896A (en) 2013-08-30 2016-02-16 安美基股份有限公司 GITR antigen binding proteins
US10259875B2 (en) 2013-10-01 2019-04-16 Mayo Foundation For Medical Education And Research Methods for treating cancer in patients with elevated levels of BIM
US10302653B2 (en) 2014-05-22 2019-05-28 Mayo Foundation For Medical Education And Research Distinguishing antagonistic and agonistic anti B7-H1 antibodies
US10517875B2 (en) 2014-07-23 2019-12-31 Mayo Foundation for Medical Engineering and Research Targeting DNA-PKcs and B7-H1 to treat cancer
WO2016108669A1 (en) * 2014-12-31 2016-07-07 주식회사 휴온스 Composition, containing rgd motif-containing peptide or fragment thereof, for treating burns and glaucoma, alleviating skin wrinkles, and promoting hair growth
IL293708A (en) 2015-10-06 2022-08-01 Genentech Inc Method for treating multiple sclerosis
US10875923B2 (en) 2015-10-30 2020-12-29 Mayo Foundation For Medical Education And Research Antibodies to B7-H1
MA43814A (en) 2016-03-08 2018-11-28 Janssen Biotech Inc ANTI-GITR ANTIBODIES, METHODS AND USES
US20180164221A1 (en) 2016-12-07 2018-06-14 Progenity Inc. Gastrointestinal tract detection methods, devices and systems
US20200253506A1 (en) 2016-12-14 2020-08-13 Progenity, Inc. Treatment of a disease of the gastrointestinal tract with an integrin inhibitor
RS62456B1 (en) 2016-12-22 2021-11-30 Amgen Inc Benzisothiazole, isothiazolo[3,4-b]pyridine, quinazoline, phthalazine, pyrido[2,3-d]pyridazine and pyrido[2,3-d]pyrimidine derivatives as kras g12c inhibitors for treating lung, pancreatic or colorectal cancer
CN107137722B (en) * 2017-05-20 2020-06-02 青岛科技大学 Nano drug delivery system (DOX-RGD-BSA @ AuNCs) drug delivery and biological imaging
JOP20190272A1 (en) 2017-05-22 2019-11-21 Amgen Inc Kras g12c inhibitors and methods of using the same
SG11202001499WA (en) 2017-09-08 2020-03-30 Amgen Inc Inhibitors of kras g12c and methods of using the same
US11045484B2 (en) 2018-05-04 2021-06-29 Amgen Inc. KRAS G12C inhibitors and methods of using the same
EP3788038B1 (en) 2018-05-04 2023-10-11 Amgen Inc. Kras g12c inhibitors and methods of using the same
WO2019217691A1 (en) 2018-05-10 2019-11-14 Amgen Inc. Kras g12c inhibitors for the treatment of cancer
US11096939B2 (en) 2018-06-01 2021-08-24 Amgen Inc. KRAS G12C inhibitors and methods of using the same
EP3802537A1 (en) 2018-06-11 2021-04-14 Amgen Inc. Kras g12c inhibitors for treating cancer
MX2020012261A (en) 2018-06-12 2021-03-31 Amgen Inc Kras g12c inhibitors encompassing a piperazine ring and use thereof in the treatment of cancer.
EP3810085A1 (en) 2018-06-20 2021-04-28 Progenity, Inc. Treatment of a disease of the gastrointestinal tract with an integrin inhibitor
JP2020090482A (en) 2018-11-16 2020-06-11 アムジエン・インコーポレーテツド Improved synthesis of key intermediate of kras g12c inhibitor compound
JP7377679B2 (en) 2018-11-19 2023-11-10 アムジエン・インコーポレーテツド Combination therapy comprising a KRASG12C inhibitor and one or more additional pharmaceutically active agents for the treatment of cancer
CN116726361A (en) 2018-11-19 2023-09-12 比奥拉治疗股份有限公司 Methods and devices for treating diseases with biologic therapeutic agents
CA3117222A1 (en) 2018-11-19 2020-05-28 Amgen Inc. Kras g12c inhibitors and methods of using the same
MA54547A (en) 2018-12-20 2022-03-30 Amgen Inc HETEROARYL AMIDES USEFUL AS KIF18A INHIBITORS
MA54546A (en) 2018-12-20 2022-03-30 Amgen Inc HETEROARYL AMIDES USEFUL AS KIF18A INHIBITORS
PE20211475A1 (en) 2018-12-20 2021-08-05 Amgen Inc KIF18A INHIBITORS
ES2953821T3 (en) 2018-12-20 2023-11-16 Amgen Inc KIF18A inhibitors
JP2022522777A (en) 2019-03-01 2022-04-20 レボリューション メディシンズ インコーポレイテッド Bicyclic heteroaryl compounds and their use
US20230096028A1 (en) 2019-03-01 2023-03-30 Revolution Medicines, Inc. Bicyclic heterocyclyl compounds and uses thereof
EP3738593A1 (en) 2019-05-14 2020-11-18 Amgen, Inc Dosing of kras inhibitor for treatment of cancers
EP3972973A1 (en) 2019-05-21 2022-03-30 Amgen Inc. Solid state forms
WO2021026098A1 (en) 2019-08-02 2021-02-11 Amgen Inc. Kif18a inhibitors
CN114401953A (en) 2019-08-02 2022-04-26 美国安进公司 KIF18A inhibitors
AU2020324963A1 (en) 2019-08-02 2022-02-24 Amgen Inc. KIF18A inhibitors
US20220372018A1 (en) 2019-08-02 2022-11-24 Amgen Inc. Kif18a inhibitors
WO2021081212A1 (en) 2019-10-24 2021-04-29 Amgen Inc. Pyridopyrimidine derivatives useful as kras g12c and kras g12d inhibitors in the treatment of cancer
IL292438A (en) 2019-10-28 2022-06-01 Merck Sharp & Dohme Small molecule inhibitors of kras g12c mutant
US20230023023A1 (en) 2019-10-31 2023-01-26 Taiho Pharmaceutical Co., Ltd. 4-aminobut-2-enamide derivatives and salts thereof
TW202132314A (en) 2019-11-04 2021-09-01 美商銳新醫藥公司 Ras inhibitors
CA3159559A1 (en) 2019-11-04 2021-05-14 Revolution Medicines, Inc. Ras inhibitors
EP4054719A1 (en) 2019-11-04 2022-09-14 Revolution Medicines, Inc. Ras inhibitors
PE20230249A1 (en) 2019-11-08 2023-02-07 Revolution Medicines Inc BICYCLIC HETEROARYL COMPOUNDS AND THEIR USES
AR120456A1 (en) 2019-11-14 2022-02-16 Amgen Inc ENHANCED SYNTHESIS OF KRAS G12C INHIBITOR COMPOUND
WO2021097212A1 (en) 2019-11-14 2021-05-20 Amgen Inc. Improved synthesis of kras g12c inhibitor compound
JP2023505100A (en) 2019-11-27 2023-02-08 レボリューション メディシンズ インコーポレイテッド Covalent RAS inhibitors and uses thereof
WO2021106231A1 (en) 2019-11-29 2021-06-03 Taiho Pharmaceutical Co., Ltd. A compound having inhibitory activity against kras g12d mutation
WO2021119482A1 (en) 2019-12-13 2021-06-17 Progenity, Inc. Ingestible device for delivery of therapeutic agent to the gastrointestinal tract
BR112022010086A2 (en) 2020-01-07 2022-09-06 Revolution Medicines Inc SHP2 INHIBITOR DOSAGE AND CANCER TREATMENT METHODS
WO2021185719A1 (en) * 2020-03-15 2021-09-23 Ac Bioscience Sa Rgd-containing peptide and use thereof for cancer treatment
US20230181536A1 (en) 2020-04-24 2023-06-15 Taiho Pharmaceutical Co., Ltd. Anticancer combination therapy with n-(1-acryloyl-azetidin-3-yl)-2-((1h-indazol-3-yl)amino)methyl)-1h-imidazole-5-carboxamide inhibitor of kras-g12c
US20230174518A1 (en) 2020-04-24 2023-06-08 Taiho Pharmaceutical Co., Ltd. Kras g12d protein inhibitors
CN115916194A (en) 2020-06-18 2023-04-04 锐新医药公司 Methods for delaying, preventing and treating acquired resistance to RAS inhibitors
AU2021308045A1 (en) 2020-07-15 2023-02-23 Taiho Pharmaceutical Co., Ltd. Pyrimidine compound-containing combination to be used in tumor treatment
AU2021344830A1 (en) 2020-09-03 2023-04-06 Revolution Medicines, Inc. Use of SOS1 inhibitors to treat malignancies with SHP2 mutations
KR20230067635A (en) 2020-09-15 2023-05-16 레볼루션 메디슨즈, 인크. Indole derivatives as RAS inhibitors in the treatment of cancer
CN117396472A (en) 2020-12-22 2024-01-12 上海齐鲁锐格医药研发有限公司 SOS1 inhibitors and uses thereof
CN117500811A (en) 2021-05-05 2024-02-02 锐新医药公司 Covalent RAS inhibitors and uses thereof
CR20230570A (en) 2021-05-05 2024-01-22 Revolution Medicines Inc Ras inhibitors
WO2022235870A1 (en) 2021-05-05 2022-11-10 Revolution Medicines, Inc. Ras inhibitors for the treatment of cancer
EP4347041A1 (en) 2021-05-28 2024-04-10 Taiho Pharmaceutical Co., Ltd. Small molecule inhibitors of kras mutated proteins
AR127308A1 (en) 2021-10-08 2024-01-10 Revolution Medicines Inc RAS INHIBITORS
TW202340214A (en) 2021-12-17 2023-10-16 美商健臻公司 Pyrazolopyrazine compounds as shp2 inhibitors
EP4227307A1 (en) 2022-02-11 2023-08-16 Genzyme Corporation Pyrazolopyrazine compounds as shp2 inhibitors
WO2023172940A1 (en) 2022-03-08 2023-09-14 Revolution Medicines, Inc. Methods for treating immune refractory lung cancer
WO2023240263A1 (en) 2022-06-10 2023-12-14 Revolution Medicines, Inc. Macrocyclic ras inhibitors
WO2024081916A1 (en) 2022-10-14 2024-04-18 Black Diamond Therapeutics, Inc. Methods of treating cancers using isoquinoline or 6-aza-quinoline derivatives

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5731288A (en) * 1993-10-22 1998-03-24 University Of Southern California Compositions containing contortrostatin and methods for the use thereof
US5814609A (en) * 1993-10-22 1998-09-29 University Of Southern California Compositions containing a disintegrin and methods for its use in preventing metastasis and other conditions
US5830742A (en) * 1995-06-08 1998-11-03 Immunex Corporation TNF-α converting enzyme
US5922546A (en) * 1997-08-25 1999-07-13 Smithkline Beecham Corporation Human disintegrin metalloprotease KUZ gene
US5935578A (en) * 1994-06-20 1999-08-10 Merck & Co., Inc. Contraceptive vaccine
US5935792A (en) * 1996-08-29 1999-08-10 The Regents Of The University Of California KUZ, a novel family of metalloproteases

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6177475B1 (en) 1992-06-30 2001-01-23 Billings Pharmaceuticals, Inc. Methods of using integrin modulators for treatment of inflammation
US6255064B1 (en) 1996-03-01 2001-07-03 The Procter & Gamble Company Disintegrin metalloprotease and its use
US6140098A (en) 1996-08-30 2000-10-31 Schering Corporation Nucleic acids encoding mammalian proteinases; related reagents
GB9716755D0 (en) 1997-08-07 1997-10-15 Glaxo Group Ltd Invention
CA2308110A1 (en) 1997-10-30 1999-05-14 Immunex Corporation Svph1-26 dna and polypeptides
NZ506277A (en) 1998-01-14 2004-07-30 Immunex Corp Testis-specific human SVPH1-8 proteinase
NZ506817A (en) 1998-02-11 2003-08-29 Immunex Corp Metalloprotease-disintegrin SVPH3-13 and SVPH3-17 nucleic acids, and their corresponding polypeptides and methods for their production
US6265199B1 (en) 1998-07-10 2001-07-24 Zymogenetics, Inc. Disintegrin homologs
CA2356681A1 (en) 1999-01-21 2000-07-27 Immunex Corporation Metalloproteinase-disintegrin family members: svph dnas and polypeptides
WO2000043493A2 (en) 1999-01-22 2000-07-27 Human Genome Sciences, Inc. Metalloproteinase adam 22
EP1268756A2 (en) 2000-04-03 2003-01-02 Bristol-Myers Squibb Company Methods and compositions for modulating integrin-mediated cell-cell interactions

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5731288A (en) * 1993-10-22 1998-03-24 University Of Southern California Compositions containing contortrostatin and methods for the use thereof
US5814609A (en) * 1993-10-22 1998-09-29 University Of Southern California Compositions containing a disintegrin and methods for its use in preventing metastasis and other conditions
US5935578A (en) * 1994-06-20 1999-08-10 Merck & Co., Inc. Contraceptive vaccine
US5830742A (en) * 1995-06-08 1998-11-03 Immunex Corporation TNF-α converting enzyme
US6013466A (en) * 1995-06-08 2000-01-11 Immunex Corporation TNF-α converting enzyme
US5935792A (en) * 1996-08-29 1999-08-10 The Regents Of The University Of California KUZ, a novel family of metalloproteases
US5922546A (en) * 1997-08-25 1999-07-13 Smithkline Beecham Corporation Human disintegrin metalloprotease KUZ gene

Cited By (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100129332A1 (en) * 2000-07-31 2010-05-27 Tamar Tennenbaum Methods and pharmaceutical compositions for healing wounds
US8093211B2 (en) 2000-07-31 2012-01-10 Bar-Ilan University Methods and pharmaceutical compositions for healing wounds
US20090192074A1 (en) * 2000-07-31 2009-07-30 Bar Ilan University Methods and pharmaceutical compositions for healing wounds
US20100167987A9 (en) * 2000-07-31 2010-07-01 Bar Ilan University Methods and pharmaceutical compositions for healing wounds
US20080280816A1 (en) * 2001-07-23 2008-11-13 Tamar Tennenbaum Methods and pharmaceutical compositions for healing wounds
US20040171549A1 (en) * 2001-07-26 2004-09-02 Inserm, A Corporation Of France Method of inhibiting angiogenesis or invasion or formation of metastases
US8207137B2 (en) * 2001-07-26 2012-06-26 Inserm Method of inhibiting angiogenesis or invasion or formation of metastases
US20030148924A1 (en) * 2002-07-09 2003-08-07 Tamar Tennenbaum Methods and pharmaceutical compositions of healing wounds
US8507431B2 (en) 2003-08-07 2013-08-13 Healor Ltd. Methods for accelerating wound healing by administration of a preadipocyte modulator or an adipocyte modulator
US20080159978A1 (en) * 2003-08-07 2008-07-03 Liora Braiman-Wiksman Methods for accelerating wound healing by administration of adipokines
US20060177418A1 (en) * 2003-08-07 2006-08-10 Healor Ltd. Methods for accelerating wound healing by administration of adipokines
US7638484B2 (en) 2003-08-07 2009-12-29 Healor Ltd. Methods for accelerating wound healing by administration of adipokines
US8367606B2 (en) 2005-08-29 2013-02-05 Healor Ltd. Method and compositions for prevention and treatment of diabetic and aged skin
US20110021422A1 (en) * 2005-08-29 2011-01-27 Tamar Tennenbaum Method and compositions for prevention and treatment of diabetic and aged skin
EP1970441A1 (en) * 2007-03-06 2008-09-17 BioAlliance Pharma Plasmid containing a sequence encoding a disintegrin domain of metargidin (RDD)
US20100197769A1 (en) * 2007-03-06 2010-08-05 Celine Bouquet Plasmid containing a sequence encoding a disintegrin domain of metargidin (rdd)
US20100310542A1 (en) * 2007-07-30 2010-12-09 Healor Ltd. Pharmaceutical Compositions for treating wouds and related methods
US20100215634A1 (en) * 2009-02-24 2010-08-26 Tamar Tennenbaum Visfatin therapeutic agents for the treatment of acne and other conditions
US8349793B2 (en) 2010-01-11 2013-01-08 Heal0r, Ltd. Method for treatment of inflammatory disease and disorder
WO2014066084A1 (en) * 2012-10-24 2014-05-01 Novamedica Limited Liability Company Nucleic acid modulators of alpha2beta1
WO2017001990A1 (en) * 2015-06-28 2017-01-05 Allgenesis Biotherapeutics Inc. Fusion proteins for inhibiting angiogenesis
TWI698447B (en) * 2015-06-28 2020-07-11 新源生物科技股份有限公司 Fusion proteins for inhibiting angiogenesis
US11192927B2 (en) 2015-06-28 2021-12-07 Allgenesis Biotherapeutics Inc. Fusion proteins for inhibiting angiogenesis
WO2020080836A1 (en) * 2018-10-19 2020-04-23 아이진 주식회사 Pharmaceutical composition for preventing or treating inflammatory diseases
US11723955B1 (en) 2022-05-13 2023-08-15 Allgenesis Biotherapeutics Inc. VEGFR fusion protein pharmaceutical composition

Also Published As

Publication number Publication date
NZ521437A (en) 2004-04-30
DE60127656T2 (en) 2007-12-20
CA2400040A1 (en) 2001-08-30
EP1259595B1 (en) 2007-04-04
ATE358718T1 (en) 2007-04-15
WO2001062905A2 (en) 2001-08-30
IL151032A0 (en) 2003-04-10
US7074408B2 (en) 2006-07-11
WO2001062905A3 (en) 2002-03-21
US20020042368A1 (en) 2002-04-11
JP2003523768A (en) 2003-08-12
AU2001247219B2 (en) 2007-01-04
EP1259595A2 (en) 2002-11-27
DE60127656D1 (en) 2007-05-16
AU4721901A (en) 2001-09-03

Similar Documents

Publication Publication Date Title
US7074408B2 (en) Use of integrin antagonists to inhibit angiogenesis
AU2001247219A1 (en) Integrin antagonists
EP1187918B2 (en) Tek antagonists
US6521424B2 (en) Recombinant expression of Tek antagonists
AU6411599A (en) Inhibitors of platelet activation and recruitment
US7482430B2 (en) Compositions and methods relating to multimeric and oligomeric soluble fragments of the TWEAK receptor
JP4326022B2 (en) TNF-α convertase
US6406901B1 (en) TNF-a converting enzyme
US20020002277A1 (en) Inhibitors of platelet activation and recruitment
EP1803810A1 (en) Integrin antagonists
JP2002503472A (en) DNA and polypeptide of metalloprotease-disintegrin SVPH3-13 and SVPH3-17
AU2007201172A1 (en) Integrin antagonists
US7264809B1 (en) Methods of inhibiting platelet activation and recruitment
JP2002534983A (en) Metalloproteinase disintegrin family members: SVPH DNA and polypeptide
AU1757500A (en) TNF-alpha converting enzyme
MXPA06000653A (en) Compositions and methods relating to multimeric and oligomeric soluble fragments of the tweak receptor

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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