AU1908000A - Inhibition of the formation of vascular hyperpermeability - Google Patents

Inhibition of the formation of vascular hyperpermeability Download PDF

Info

Publication number
AU1908000A
AU1908000A AU19080/00A AU1908000A AU1908000A AU 1908000 A AU1908000 A AU 1908000A AU 19080/00 A AU19080/00 A AU 19080/00A AU 1908000 A AU1908000 A AU 1908000A AU 1908000 A AU1908000 A AU 1908000A
Authority
AU
Australia
Prior art keywords
kdr
compound
tyrosine kinase
edema
vegf
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
AU19080/00A
Inventor
Lee D. Arnold
Peter F. Bousquet
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.)
BASF SE
Original Assignee
BASF SE
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 BASF SE filed Critical BASF SE
Publication of AU1908000A publication Critical patent/AU1908000A/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/71Receptors; Cell surface antigens; Cell surface determinants for growth factors; for growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • 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
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/16Central respiratory analeptics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/02Drugs for disorders of the urinary system of urine or of the urinary tract, e.g. urine acidifiers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • 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
    • A61P19/00Drugs for skeletal disorders
    • 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
    • A61P27/06Antiglaucoma agents or miotics
    • 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
    • A61P27/12Ophthalmic agents for cataracts
    • 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
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • 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
    • A61P7/10Antioedematous agents; Diuretics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/02Non-specific cardiovascular stimulants, e.g. drugs for syncope, antihypotensives
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/14Vasoprotectives; Antihaemorrhoidals; Drugs for varicose therapy; Capillary stabilisers
    • 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
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1137Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against enzymes
    • 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
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1138Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against receptors or cell surface proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • 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
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/11Antisense
    • 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
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/12Type of nucleic acid catalytic nucleic acids, e.g. ribozymes

Description

WO 00/27414 PCTIUS99/25903 -1 5 INHIBITION OF THE FORMATION OF VASCULAR HYPERPERMEABILITY BACKGROUND OF THE INVENTION 10 Edema can be described as an increase in the interstitial fluid volume. This is usually an abnormal condition for which relief is typically sought. This condition quite often arises because fluid leaves the blood vasculature due to an increase in endothelial permeability, often associated with macromolecular extravasation, and finds a new residence in the interstitial spaces. 15 There are a variety of physiological and biochemical mechanisms that underlie edema and the formation of the edematous state in an individual. An important mediator in one or more of these mechanisms is "vascular endothelial cell growth This factor upregulates transport in vascular endothelial cells, and causes an increase in the permeability of numerous vascular beds including the skin, subcutaneous tissues, 20 peritoneal wall, mesentery, diaphragm, trachea, bronchi, duodenum and uterus. Significant diapedesis, alterations in exchange across the endothelium, extravasation and deposition of macromolecules at these sites and prolonged hypotension may accompany these increased permeability effects. These processes are thought to be a facilitating prelude to neovascularization. VEGF is expressed by inflammatory T-cells, 25 macrophages, neutrophils and eosinophils, etc. at sites of inflammation. This factor is upregulated by hypoxia, certain vasopressor hormones, growth factors, reproductive hormones and numerous inflammatory cytokines. VEGF-mediated vascular permeability has been implicated in such disorders as tumor ascites, endometriosis, adult respiratory distress syndrome (ARDS), post cardiopulmonary bypass-related 30 hypotension and hyperpermeability blistering, edematous responses to bums and trauma, endothelial dysfunction in diabetes, ovarian hyperstimulation syndrome complications, and ocular edema. Thus, it is apparent that the inhibition of VEGF production or activity would be beneficial, especially to block the manifestation of the above-listed disorders. In 35 particular, agents that are capable of blocking VEGF mediated hyperpermeability and edema and associated syndromes would be useful for alleviating these disorders.
- WO 00/27414 PCT/US99/25903 -2 5 Protein Tyrosine Kinases. Protein tyrosine kinases (PTKs) comprise a large and diverse class of proteins having enzymatic activity. The PTKs play an important role in the control of cell growth and differentiation (for review, see Schlessinger & Ullrich, 1992, Neuron 9:383-391). Aberrant stimulation, expression or mutations in the PTKs have been shown to 10 lead to either uncontrolled cell proliferation (e.g., malignant tumor growth) or to defects in key developmental, regulatory or reparative processes. Consequently, the biomedical community has expended significant resources to discover the specific biological role of members of the PTK family, their function in differentiation processes, their involvement in tumorigenesis and in other diseases, the biochemical mechanisms is underlying their signal transduction pathways activated upon ligand stimulation and the development of novel drugs. Tyrosine kinases can be of the receptor-type (having extracellular, transmembrane and intracellular domains) or the non-receptor type (being wholly intracellular). 20 Receptor Tyrosine Kinases (RTKs). The RTKs comprise a large family of transmembrane receptors with diverse biological activities. At present, at least nineteen (19) distinct RTK subfamilies have been identified. The receptor tyrosine kinase (RTK) family includes receptors that are crucial for the growth and differentiation of a variety of cell types (Yarden and Ullrich, Ann. Rev. Biochem. 57:433-478, 1988; Ullrich 25 and Schlessinger, Cell 61:243-254, 1990). The intrinsic function of RTKs is activated upon ligand binding, which results in phosphorylation of the receptor and multiple cellular substrates, and subsequently in a variety of cellular responses (Ullrich & Schlessinger, 1990, Cell 61:203-212). Thus, receptor tyrosine kinase mediated signal transduction is initiated by extracellular interaction with a specific growth factor 30 (ligand), typically followed by receptor dimerization, stimulation of the intrinsic protein tyrosine kinase activity and receptor trans-phosphorylation. Binding sites are thereby created for intracellular signal transduction molecules and lead to the formation of complexes with a spectrum of cytoplasmic signaling molecules that facilitate the appropriate cellular response. (e.g., cell division, differentiation, metabolic effects, 35 changes in the extracellular microenvironment) see Schlessinger and Ullrich, 1992, Neuron 9:1-20.
WO 00/27414 PCTIUS99/25903 -3 5 Proteins with SH2 (src homology -2) or phosphotyrosine binding (PTB) domains bind activated tyrosine kinase receptors and their substrates with high affinity to propagate signals into cell. Both of the domains recognize phosphotyrosine. (SH2:Fantle et al., 1992, Cell 69:413-423; Songyant et al., 1994,Mol. Cell. Biol. 14;2777-2785; Songyang et al., 1993, Cell 72:767-778; and Koch et al., 1991, Science 10 252:668-678; Schoelson, Curr. Opin. Chem. Biol. (1997), 1(2), 227-234; Cowburn, Curr. Opin. Struct. Biol. (1997), 7(6), 835-838). Several intercellular substrate proteins that associate with receptor tyrosine kinases (RTKs) have been identified. They may be divided into two principal groups: (1) substrates which have a catalytic domain; and (2) substrates which lack such a domain but serve as adapters and associate with 15 catalytically active molecules (Songyang et al., 1993, Cell 72:767-778). The specificity of the interactions between receptors or proteins and SH2 or PTB domains of their substrates is determined by the amino acid residues immediately surrounding the phosphorylated tyrosine residue. For example, differences in the binding affinities between SH2 domains and the amino acid dsquences surrounding the phosphotyrosine 20 residues on particular receptors correlate with the observed differences in their substrate phosphorylation profiles (Songyang et al., 1993, Cell 72:767-778). Observations suggest that the function of each receptor tyrosine kinase is determined not only by its pattern of expression and ligand availability but also by the array of downstream signal transduction pathways that are activated by a particular receptor as well as the timing 25 and duration of those stimuli. Thus, phosphorylation provides an important regulatory step which determines the selectivity of signaling pathways recruited by specific growth factor receptors, as well as differentiation factor receptors. Several receptor tyrosine kinases, and growth factors that bind thereto, have been suggested to play a role in angiogenesis, although some may promote 30 angiogenesis indirectly (Mustonen and Alitalo, J Cell Biol. 129:895-898, 1995). One such receptor tyrosine kinase, known as "fetal liver kinase 1" (FLK-1), is a member of the type III subclass of RTKs. An alternative designation for human FLK-1 is "kinase insert domain-containing receptor" (KDR) (Terman et al., Oncogene 6:1677-83, 1991). Another alternative designation for FLK-1/KDR is "vascular endothelial cell growth 35 factor receptor 2" (VEGFR-2) since it binds VEGF with high affinity. The murine version of FLK-1/VEGFR-2 has also been called NYK (Oelrichs et al, Oncogene ~ WO 00/27414 PCT/US99/25903 -4 5 8(l):11-15, 1993). DNAs encoding mouse, rat and human FLK-1 have been isolated, and the nucleotide and encoded amino acid sequences reported (Matthews et al., Proc. Natl. Acad. Sci. USA, 88:9026-30, 1991; Terman et al., 1991, supra; Terman et al., Biochem. Biophys. Res. Comm. 187:1579-86, 1992; Sarzani et al., supra; and Millauer et al., Cell 72:835-846, 1993). Numerous studies such as those reported in Millauer et 10 al., supra, suggest that VEGF and FLK-1/KDR/VEGFR-2 are a ligand-receptor pair that play an important role in the proliferation of vascular endothelial cells, and formation and sprouting of blood vessels, termed vasculogenesis and angiogenesis, respectively. Another type III subclass RTK designated fms-like tyrosine kinase-1" (Flt-1) is 15 related to FLK-1/KDR (DeVries et al. Science 255;989-991, 1992; Shibuya et al., Oncogene 5:519-524, 1990). An alternative designation for flt-1 is "vascular endothelial cell growth factor receptor 1" (VEGFR-1). To date, members of the FLK 1/KDR/VEGFR-2 and flt-l/ VEGFR-1 subfamilies have been found expressed primarily on endothelial cells. These subclass members are specifically stimulated by 20 members of the vascular endothelial cell growth factor (VEGF) family of ligands (Klagsburn and D'Amore, Cytokine & Growth Factor Reviews 7: 259-270, 1996). Vascular endothelial cell growth factor (VEGF), binds to Flt- 1 with higher affinity than to FLK-1/KDR and is mitogenic toward vascular endothelial cells (Terman et al., 1992, supra; Mustonen et al. supra; DeVries et al., supra). Flt-1 is believed to be essential 25 for endothelial organization during vascular development. Flt-1 expression is associated with early vascular development in mouse embryos, and with neovascularization during wound healing (Mustonen and Alitalo, supra). Expression of flt-1 in adult organs such as kidney glomeruli suggests an additional function for this receptor that is not related to cell growth (Mustonen and Alitalo, supra). 30 As previously stated, recent evidence suggests that VEGF plays a role in the stimulation of both normal and pathological angiogenesis (Jakeman et al., Endocrinology 133: 848-859, 1993; Kolch et al., Breast Cancer Research and Treatment 36: 139-155, 1995; Ferrara et al., Endocrine Reviews 18(1); 4-25, 1997; Ferrara et al., Regulation ofAngiogenesis (ed. L. D. Goldberg and E.M. Rosen), 209 35 232, 1997). In addition, VEGF has been implicated in the control and enhancement of vascular permeability (Connolly, et al., J. Biol. Chem. 264: 20017-20024, 1989; Brown WO 00/27414 PCTIUS99/25903 -5 5 et al., Regulation of Angiogenesis (ed. L.D. Goldberg and E.M. Rosen), 233-269, 1997). Different forms of VEGF arising from alternative splicing of mRNA have been reported, including the four species described by Ferrara et al. (J. Cell. Biochem. 47:211-218, 1991). Both secreted and predominantly cell-associated species of VEGF 10 have been identified by Ferrara et al. supra, and the protein is known to exist in the form of disulfide linked dimers. Several related homologs of VEGF have recently been identified. However, their roles in normal physiological and disease processes have not yet been elucidated. In addition, the members of the VEGF family are often coexpressed with VEGF in a 15 number of tissues and are, in general, capable of forming heterodimers with VEGF. This property likely alters the receptor specificity and biological effects of the heterodimers and further complicates the elucidation of their specific functions as illustrated below (Korpelainen and Alitalo, Curr. Opin. Cell Biol., 159-164, 1998 and references cited therein). 20 Placenta growth factor (PlGF) has an amino acid sequence that exhibits significant homology to the VEGF sequence (Park et al., J. Biol. Chem. 269:25646-54, 1994; Maglione et al. Oncogene 8:925-31, 1993). As with VEGF, different species of PlGF arise from alternative splicing of mRNA, and the protein exists in dimeric form (Park et al., supra). PlGF-1 and PlGF-2 bind to Flt-1 with high affinity, and PlGF-2 25 also avidly binds to neuropilin-1 (Migdal et al.,J. Biol. Chem. 273 (35): 22272 22278),but neither binds to FLK-1/KDR (Park et al., supra). PlGF has been reported to potentiate both the vascular permeability and mitogenic effect of VEGF on endothelial cells when VEGF is present at low concentrations (purportedly due to heterodimer formation) (Park et al., supra). 30 VEGF-B is produced as two isoforms (167) and 185 residues) that also appear to bind Flt-1/VEGFR-1. It may play a role in the regulation of extracellular matrix degradation, cell adhesion, and migration through modulation of the expression and activity of urokinase type plasminogen activator and plasminogen activator inhibitor 1 (Pepper et al, Proc. Natl. Acad. Sci. U. S. A. (1998), 95(20): 11709-11714). 35 VEGF-C was originally cloned as a ligand for VEGFR-3/Flt-4 which is primarily expressed by lymphatic endothelial cells. In its fully processed form, VEGF- - WO 00/27414 PCTIUS99/25903 -6 s C can also bind KDR/VEGFR-2 and stimulate proliferation and migration of endothelial cells in vitro and angiogenesis in in vivo models(Lymboussaki et al, Am. J. Pathol. (1998), 153(2): 395-403; Witzenbichler et al, Am J. Pathol. (1998), 153(2), 381-394). The overexpression of VEGF-C causes proliferation and enlargement of only lymphatic vessels, while blood vessels are unaffected. Unlike VEGF, the 10 expression of VEGF-C is not induced by hypoxia (Ristimaki et al, J. Biol. Chem. (1998), 273(14),8413-8418). The most recently discovered VEGF-D is structurally very similar to VEGF-C. VEGF-D is reported to bind and activate at least two VEGFRs, VEGFR-3/Flt-4 and KDR/VEGFR-2. It was originally cloned as a c-fos inducible mitogen for fibroblasts 15 and is most prominently expressed in the mesenchymal cells of the lung and skin (Achen et al, Proc. Natl. Acad. Sci. U. S. A. (1998), 95(2), 548-553 and references therein). VEGF-C and VEGF-D have been claimed to induce increases in vascular permeability in vivo in a Miles assay when injected into cutaneous tissue 20 (PCT/US97/14696; W098/07832, Witzenbichler et al., supra). The physiological role and significance of these ligands in modulating vascular hyperpermeability and endothelial responses in tissues where they are expressed remains uncertain. Based upon emerging discoveries of other homologs of VEGF and VEGFRs and the precedents for ligand and receptor heterodimerization, the actions of such 25 VEGF homologs may involve formation of VEGF ligand heterodimers, and/or heterodimerization of receptors, or binding to a yet undiscovered VEGFR (Witzenbichler et al., supra). Also, recent reports suggest the possible involvement of neuropilin- 1 (Migdal et al, supra) or VEGFR-3/Flt-4 (Witzenbichler et al., supra), and that receptors other than KDR/VEGFR-2 are responsible for the induction of vascular 3 0 permeability (Stacker, S.A., Vitali, A., Domagala, T., Nice, E., and Wilks, A.F., "Angiogenesis and Cancer" Conference, Amer. Assoc. Cancer Res., Jan. 1998, Orlando, FL; Williams, Diabetelogia 40: S118-120 (1997)). Development of Compounds to Modulate the PTKs. In view of the surmised importance of PTKs to the control, regulation, and modulation of cell proliferation, as 35 well as the diseases and disorders associated with abnormal cell proliferation, many attempts have been made to identify receptor and non-receptor tyrosine kinase - WO 00/27414 PCTIUS99/25903 -7 5 "inhibitors" using a variety of approaches, including the use of mutant ligands (U.S. Application No. 4,966,849), soluble receptors and antibodies (Application No. WO 94/10202; Kendall & Thomas, 1994, Proc. Natl. Acad. Sci 90:10705-09; Kim et al., 1993, Nature 362:841-844), RNA ligands (Jellinek, et al., Biochemistry 33:10450-56; Takano, et al., 1993, Mol. Bio. Cell 4:358A; Kinsella, et al. 1992, Exp. Cell Res. 10 199:56-62; Wright, et al., 1992, J. Cellular Phys. 152:448-57) and tyrosine kinase inhibitors (WO 94/03427; WO 92/21660; WO 91/15495; WO 94/14808; U.S. Patent No. 5,330,992; Mariani, et al., 1994, Proc. Am. Assoc. Cancer Res. 35:2268). More recently, attempts have been made to identify small molecules which act as tyrosine kinase inhibitors. For example, bis monocyclic, bicyclic or heterocyclic aryl 15 compounds (PCT WO 92/20642) and vinylene-azaindole derivatives (PCT WO 94/14808) have been described generally as tyrosine kinase inhibitors. Styryl compounds (U.S. Patent No. 5,217,999), styryl-substituted pyridyl compounds (U.S. Patent No. 5,302,606), certain quinazoline derivatives (EP Application No. 0 566 266 Al); Expert Opin. Ther. Pat. (1998), 8(4): 475-478, seleoindoles and selenides (PCT 20 WO 94/03427), tricyclic polyhydroxylic compounds (PCT WO 92/21660) and benzylphosphonic acid compounds (PCT WO 91/15495) have been described as compounds for use as tyrosine kinase inhibitors for use in the treatment of cancer. Anilinocinnolines (PCT WO 97/34876) and quinazoline derivative compounds (PCT WO 97/22596; PCT WO 97/42187) have been decribed as inhibitors of angiogenesis 25 and vascular permeability. In addition, attempts have been made to identify small molecules which act as serine/threonine kinase inhibitors. In particular, bis(indolylmaleimide) compounds have been described as inhibiting particular PKC serine/threonine kinase isoforms whose dysfunction is associated with altered vascular permeability in VEGF-related 30 diseases (PCT W097/40830; PCT W097/40831). The identification of effective macromolecules and small organic compounds which specifically inhibit tyrosine signal transduction by modulating the activity of receptor and non-receptor tyrosine kinases to regulate and modulate abnormal or inappropriate cellular function, cell proliferation or differentiation is therefore desirable. 35 In particular, the identification of methods and compounds that specifically inhibit the function of a tyrosine kinases which is essential for the formation of vascular WO 00/27414 PCT/US99/25903 -8 5 hyperpermeability leading to edema, effusions, exudates, and macromolecular extravasation and deposition as well as associated disorders would be beneficial. SUMMARY OF THE INVENTION This invention is directed to the inhibition of vascular hyperpermeability by 10 inhibiting the cellular signaling function of KDR tyrosine kinase. This invention also provides a method of inhibiting vascular hyperpermeability by selectively disrupting the catalytic kinase response of KDR/VEGFR-2 without significantly affecting the activity of Flt-l/VEGFR1 or other tyrosine kinases. Agents which function according to this method have a distinct pharmacological advantage over current therapeutic approaches 15 encompassing materials such as steroids which are prone to numerous undesirable side effects. These methods of the present invention are also preferred over the use of less specific kinase inhibitors, including those which inhibit multiple VEGF receptors, since these methods will not directly perturb the inportant normal physiologic function of the other kinases. As a result of the inhibition of the hyperpermeability of the vascular 20 endothelium, the subsequent formation of edema, associated diapedesis, alterations in trans-endothelial molecular exchange, extravasation, exudates and effusions are also inhibited by the suppression of the tyrosine kinase activity of KDR. Since these latter events often lead to excessive matrix deposition, aberrant stromal proliferation and organ dysfunction, the inhibition of KDR tyrosine kinase is also useful in the treatment 25 of numerous non-cancerous disorders that share these etiologic features. In addition, vascular hypotension that can be caused by a VEGF-related activating ligand binding to KDR tyrosine kinase receptor is also minimized by inhibiting the activity of KDR tyrosine kinase. This invention also provides a therapeutic approach to the inhibition of vascular 30 hyperpermeability and of the formation of edema in individuals by administering a compound that specifically inhibits the activity of KDR tyrosine kinase. DETAILED DESCRIPTION OF THE INVENTION This invention discloses a method of inhibiting vascular hyperpermeability by 35 inhibiting the cellular signaling function of KDR tyrosine kinase. This invention also discloses a method of inhibiting vascular hyperpermeability through the utilization of - WO 00/27414 PCT/US99/25903 -9 5 agents which selectively inhibit the cellular signaling function of KDR. Through the identification and utilization of highly selective KDR inhibitors which effectively block KDR cellular signaling, and subsequently vascular hyperpermeability, according to the methods of this invention, the essential role of KDR in mediating the vascular permeability response to VEGF has been established. Such highly selective KDR 10 inhibitors have demonstrated efficacy in modulating vascular permeability, without the need to inhibit the function of the higher affinity receptor, VEGFR-1/Flt-1. This property should afford better toleration to therapy than current therapies or treatment with agents that less selectively disrupt the function of other non-KDR kinases KDR tyrosine kinase is activated when vascular endothelial cell growth factor 15 (VEGF) or another activating ligand (such as HIV Tat protein, VEGF-C or VEGF-D) binds to a KDR tyrosine kinase receptor which lies on the surface of vascular endothelial cells. Although naturally occurring kinase-activating mutations and truncations have not yet been identified for KDR, they have been reported for EGFR and Tie-2 receptor kinases. Hence instances of constitutive activation of KDR are also 20 anticipated. The KDR tyrosine kinase may also be referred to as FLK-1 tyrosine kinase, NYK tyrosine kinase or VEGFR-2 tyrosine kinase. In addition to stimulating angiogenesis, and endothelial cell migration and proliferation, VEGF induces hypermeability of the blood vessels. As a result, fluid moves from the blood stream past the blood vessel walls into the interstitial spaces, 25 thereby forming an area of edema. Diapedesis also often accompanies this response. Similarly, excessive vascular hyperpermeability can disrupt normal molecular exchange across the endothelium in critical tissues and organs (e.g., lung and kidney), thereby causing organ dysfunction, macromolecular extravasation, and matrix deposition often with promoted stromal proliferation. When occurring in confined compartments, the 3 0 edema (e.g., cerebral edema) may lead to impaired organ function and damage. KDR cellular signaling function can be inhibited by a number of approaches: either by blocking the production of the activating ligand, by blocking the activating ligand binding to the KDR tyrosine kinase receptor, by preventing receptor dimerization and transphosphorylation, by inhibiting the enzyme activity of the KDR tyrosine kinase 35 (inhibiting the phosphorylation capacity of the enzyme) or by some other mechanism that interrupts its downstream signaling (D. Mukhopedhyay et al., Cancer Res. 58:1278- - WO 00/27414 PCTIUS99/25903 -10 5 1284 (1998) and references therein). According to the method disclosed herein, such approaches which are selective for disrupting KDR cellular signaling function will reduce vascular hyperpermeability, as well as associated extravasation, subsequent edema formation and matrix deposition. There are a variety of compounds that have the requisite KDR tyrosine kinase 10 inhibition property. Among these compounds are antibodies (hereafter meant to include single chain antibody constructs) that bind the extracellular KDR receptor domain or cellular kinase enzyme portion or, alternatively, that bind VEGF itself. These antibodies interfere with VEGF binding to the KDR tyrosine kinase receptor and/or, importantly, with KDR tyrosine kinase cellular signaling function. Antibodies that bind 15 to the KDR tyrosine kinase may act as VEGF antagonists or, more generally, VEGF activator antagonists. Alternatively these antibodies may block functional receptor dimerization or they may be KDR tyrosine kinase inhibitors. Antibodies that bind to VEGF or an activating ligand are neutralizing antibodies of VEGF or activating ligand. It should be noted that such VEGF neutralizing antibodies may block VEGF responses 20 through both the KDR and the Flt-1 receptors and, typically, are specific for a single activating ligand. In most instances, the blocking of VEGF responses through the Flt-l receptors is not necessary nor desirable. Since these VEGFRs have been reported to recognize different epitopes on VEGF, the desired specific blockade of KDR activation can be achieved through the use of antibodies that specifically bind to and "mask" the 25 KDR-binding epitope of VEGF or other activating ligand. Other compounds that can inhibit KDR tyrosine kinase activity, and thereby minimize vascular hyperpermeability and the formation of edema, include peptides and organic molecules. Among the peptides are the soluble extracellular domain of KDR and KDR binding fragments. Other useful peptides are mutants of VEGF or VEGF 30 related growth factors (e.g., VEGF-C, VEGF-D or HIV Tat protein and fusion proteins thereof) which bind and block further ligand binding to this receptor but do not stimulate dimerization, activation or KDR tyrosine kinase transphosphorylation. Such mutants may act as monomers or nonfunctional heterodimers, thereby blocking the binding of native dimeric VEGF or activating ligand. Similarly, other peptides or small 35 molecules that block receptor dimerization and/or activation can be successfully employed. These compounds act, also, as antagonists of activating ligands or are - WO 00/27414 PCT/US99/25903 5 inhibitors of KDR tyrosine kinase activity. Preferred compounds are small organic molecules. In addition, molecules such as KDR-specific ribozymes, antisense polynucleotides (such as antisense mRNA) or intracellular single chain antibodies (SF) that inhibit the biosynthesis or proper presentation of active, functional KDR tyrosine 10 kinase will effectively block KDR-mediated responses to VEGF. These molecules can be introduced into cells preformed or their production can be induced intracellularly (e.g., through the use of appropriate adenoviral, retroviral or baculoviral vectors). The preferred compounds of this invention have the property of inhibiting the cellular signaling function of KDR without significantly inhibiting the cellular signaling 15 function Flt-1 (Flt-1 tyrosine kinase is also referred to as VEGFR-1 tyrosine kinase). Both KDR tyrosine kinase and fit-I tyrosine kinase are activated by VEGF binding to KDR tyrosine kinase receptors and to Flt-1 tyrosine kinase receptors, respectively. Since Flt-1 tyrosine kinase activity may mediate important events in endothelial maintenance and vascular function, an inhibition of this enzyme activity or associated 20 transduced signals may lead to toxic or adverse effects. At the very least, such inhibition is unnecessary for blocking the induction of vascular hyperpermeability and the formation of edema, so it is wasteful and of no value to the individual. The preferred compounds of this invention are unique because they inhibit the activity of one VEGF-receptor tyrosine kinase (KDR) that is activated by activating ligands but do 25 not inhibit other receptor tyrosine kinases, such as Flt-1, that are also activated by certain activating ligands. The most preferred compounds of this invention are, therefore, selective in their tyrosine kinase inhibitory activity. VEGF is known to contribute to vascular hyperpermeability and the formation of edema. VEGF is expressed by inflammatory T-cells, macrophages, neutrophils and 30 eosinophils, etc. at sites of inflammation. The production of this factor is quickly upregulated by hypoxia, certain vasopressor hormones, growth factors, reproductive hormones and numerous inflammatory cytokines. Indeed, vascular hyperpermeability and edema that is associated with the expression or administration of many other growth factors appears to be mediated via VEGF production. Vascular hyperpermeability, 35 associated edema, altered transendothelial exchange and macromolecular extravasation, which is often accompanied by diapedesis, can result in excessive matrix deposition, WO 00/27414 PCTIUS99/25903 -12 5 aberrant stromal proliferation, fibrosis, etc. Hence, VEGF-mediated hyperpermeability can significantly contribute to disorders with these etiologic features. For example: (1) VEGF is markedly increased in the epidermis of lesional psoriatic skin. This factor potently stimulates the dermal endothelial cell proliferation and microvascular hyperpermeability associated with psoriasis. 10 (2) Following burns and severe scalds, main organs are often damaged. This appears to be manifested by an uncontrollable "mediator disease" resulting from ischemic-reperfusion damage, swelling and edema of visceral tissues, i.e., endothelial cell damage. For bum victims, inhalation injury is one of the primary causes of mortality. The tracheobronchial epithelium sloughs and 15 combines with a protein rich exudate to form casts of the airways that can lead to obstruction of these airways. The combination of inhalation bum and hypoxia followed by exposure to a high concentration of oxygen (in an attempt to aid the individual) can worsen the situation by causing progressive changes in the lung, such as diffuse exudative formation, hemorrhage into the trachea and edematic 20 changes in the wall of the blood vessels. Circulating serum VEGF levels are dramatically increased (up to twentyfold) in victims following bums and multiple trauma and may be a prime mediator of these complications (Grad et al, Clin. Chem. Lab. Med. 36:379-383, 1998). (3) Sunburns are also associated with the formation of edema. VEGF production is 25 also known to be upregulated following UV radiation exposure. Other skin disorders where edema is produced include blistering symptomatic erythedema (acrodynia), persistent acrodema and bullous diseases such as erythema multiforme, bullous pemphigoid and dermatitis herpetiformis (i.e., conditions of acute or chronic inflammation). Edematous macules and roseacea such as that 30 associated with telangiectasia are further disorders where edema is manifested. (4) Enhanced microvascular permeability and edema are common characteristics of inflammatory and neoplastic disorders. Brain tumors such as gliomas, where tumoral and peritumoral brain edema and fluid filled cysts are formed, and meningiomas, with accompanying massive cerebral edema, are examples of such 35 disorders. Locally high levels of VEGF are associated with these disorders. Induction of malignant ascites fluid and tumor effusions (especially malignant WO 00/27414 PCTIUS99/25903 -13 5 pleural and pericardial effusions) are further examples of such edema-producing disorders and are known to involve VEGF production. Additionally, edema resulting from head trauma can produce concussions and impaired brain functions. Similarily, communicating hydrocephalus has been shown to involve cytokines such as IGF-1 and TGF-,81 known to modulate VEGF production. 10 (5) Edema occurs in some types of chronic inflammation such as the formation of nasal polyps, uterine cervical polyps and gastric hyperplastic polyps. In such cases, inflammatory cells have been shown to play an important role in the development of these edematous states, at least in part through the production of VEGF. 15 (6) Cytokine-activated eosinophils can be an important source of VEGF, thereby contributing to tissue edema formation at sites of allergic inflammation. Edema and exudates are common complications that arise during allergic and delayed type hypersensitivity reactions; also often including anaphylaxis. VEGF is implicated especially in those reactions that are not responsive to antihistamines 20 or aspirin, and its upregulation has been observed in cases of poison ivy, and contact dermatitis. In addition, tuberculosis, certain viral infections, angioedema, urticaria (hives) and exercise-induced anaphylaxis are examples of such allergic and delayed-type hypersensitivity reactions which may also involve VEGF. Edema is also often formed as a result of drug sensitivity or 25 hypersensitivity reactions, or in response to the administration of VEGF upregulating growth factor or cytokines (e.g., IGF-1, FGF-2, or IL-2). Radioanaphylaxis and radiodermatitis is associated with vascular hyperpermeability. (7) VEGF is involved in ocular neovascularization leading to diabetic retinopathy 30 and microangiopathy, blindness due to age-related macular degeneration and neonatal blindness resulting from hyperoxic exposure. In many instances, these conditions are preceded by macular or other ocular edema. VEGF has been identified as a prime mediator of iris, corneal and retinal neovascularization in cases of ocular ischemia and vascular edema. VEGF induced vascular 35 hyperpermeability contributes to blood-retinal barrier breakdown in a variety of ocular disorders with extravasation and matrix deposition laying the foundations - WO 00/27414 PCTIUS99/25903 -14 5 for subsequent angiogenesis. Corneal neovascularization is a major outcome following chemical bums, corneal inflammation and edema. Recent evidence implicates VEGF in the processes following such ocular trauma. The iron chelator deferoxamine has been used in clinical treatment of cancer patients. However, this treatment often induces macular edema. The concentrations of 10 this iron chelator that are achieved in the patients induce a 3-5 fold increase in VEGF mRNA expression in all normal and tumor cell lines studied, implicating VEGF as a likely mediator of the edema formation. Increased intraocular pressures caused by VEGF overproduction and edema can lead to inappropriate matrix depositions, ocular distortions, changes in the optic disk, defects in the 15 field of vision and can result in glaucoma. Vascular hyperpermeability is also often associated with conjunctivitis. (8) Chronic lung disease in neonates and adults results from both lung injury and inadequate repair processes. The production of VEGF has been reported in several animal models of lung injury. Destruction of pulmonary endothelial 20 cells is also characteristic of hyperoxic lung injury. During recovery from hyperoxia, VEGF is upregulated by alveolar type II cells and subsequently causes pulmonary endothelial cells to proliferate and regenerate. However, this result can cause disrupted exchange across pulmonary endothelia and pulmonary edema. Asthma and bronchitis often involve bronchial vascular dilation, 25 vascular engorgement, edema of the bronchial wall and exudates which result in thickening of airway mucosa and narrowing of the bronchial lumen. Edema with protein exudates and aberrant stromal growth are typically intertwined with these phenomena. By related processes, pulmonary edema is formed during adult respiratory distress syndrome. Causes of adult respiratory distress 30 syndrome typically include pneumonia, inhalation of noxious substances, lung contusions, near drowning and aspiration of gastric contents. (9) Corticosteroids, such as cortisone, hydrocortisone, dexamethasone or prednisolone, are among the most widely used therapeutants for edematous conditions. They are potent inhibitors of VEGF expression. This property is 35 now believed to significantly contribute to the well-known antiedematous efficacy of such steroids. However, their pluripotent biological activities are - WO 00/27414 PCTIUS99/25903 -15 5 also responsible for their undesirable side-effects. Steroid hormones and their agonists and antagonists also dramatically affect the production of VEGF, especially in reproductive tissues. Endometritis and endometriosis can occur during pregnancy, the menstrual cycle or sex hormone therapy. Swelling and cramps of menstruation is associated with vascular hyperpermeability. 10 Tamoxifen, an agent that reduces the risk of breast cancer, also increases uterine cell proliferation and tumor incidence. This steroid analog, as well as estradiol, causes uterine edema and cell proliferation which have been shown to involve local increases in VEGF production. Ovarian hyperstimulation syndrome is a serious complication affecting ovulation induction. The most severe 15 manifestation of these syndromes take the form of massive ovarian enlargement and multiple cysts, ascites, hemoconcentration and third-space accumulation of fluid. The increased capillary permeability triggered by the release of VEGF secreted by luteinized granulosa cells, etc. of the ovaries following stimulation with human chorionic gonadotropin is believed to play a key role in these 20 syndromes. It has been demonstrated that VEGF is overexpressed in the hyperthecotic ovarian stroma of polycystic ovaries in the Stein-Leventhal syndrome. (10) A rapid differential induction of VEGF gene expression in both neuronal and pial cells after transient middle cerebral artery occlusion has been demonstrated 25 in animal models of stroke. VEGF may contribute to the recovery of brain cells from ischemic insult, such as from stroke, head trauma or cerebral infarct, by potentiating neovascularization, but brain damage can also be exacerbated by the concomitant formation of brain edema. Malaria can also induce edema as a result of VEGF-induced cerebral hypoxia. Brain tumor-associated cerebral 30 edema and fluid-filled cysts arise because tumor capillaries lack normal blood brain barrier function. VEGF released by glioma cells in situ most likely accounts for the pathognomonic histopathology and clinical features of glioblastoma tumors in patients including increased cerebral edema. Carpal tunnel syndrome is accompanied by enhanced nerve hydration and, often, by 35 subsequent increased extracellular matrix deposition (entrapment neuropathy). Increased VEGF levels in the tissues which surround the nerve can cause the -WO 00/27414 PCT/US99/25903 -16 5 nerve entrapment by inducing vascular permeability, fluid efflux and stromal deposition into the perineural tissues. (11) VEGF production in tissues is dramatically upregulated in response to hypoxia. Hence, the observation that in regions of necrosis, ischemia, infarct, occlusion, anemia, circulatory impairments, or other oxygen deprivation, VEGF levels are 10 increased and vascular hyperpermeability, edema and extravasation are common. Lower oxygen pressure that is responsible for "altitude sickness" also induces rapid VEGF production which is the likely cause of life threatening cerebral and pulmonary edema (HACE and HAPE) that can occur if a person is unacclimatized. 15 (12) VEGF overproduction is likewise implicated in pericardial and pleural effusions caused by vascular hyperpermeability that results from radiation injury, rheumatoid diseases, lupus, myocardial infarction, trauma or drug reaction. Not surprisingly, VEGF overproduction in association with pericardial and pleural effusions is commonly observed at autopsy in patients with lung or breast 20 carcinomas, lymphomas and leukemias. VEGF amounts are also significantly elevated in the synovial fluid of swollen joints of individuals with rheumatoid arthritis. Sprains and fractures, although associated with some swelling and vascular hyperpermeability that is beneficial in promoting angiogenesis and healing, can be accompanied by painful and excessive, undesirable edema. 25 Similarly, VEGF involvement is anticipated in conditions such as synovitis or meniscus injury with effusion (e.g., "water-on-the-knee"). (13) Ulcerations associated with circulatory restriction (e.g. decubitus, gravitational and varicose ulcers) are also often accompanied by edema and protein exudates. Diabetic complications often arise as a result of elevational circulating glucose 30 levels (hyperglycemia) and the formation of advanced glycation endproducts (AGE), often accompanied by impaired circulation. These conditions, either alone or in combination, are known to stimulate VEGF production and, hence, vascular hyperpermeability which can lead to numerous diabetic complications. (14) Due to the significant constitutive production of endogenous VEGF by the 35 kidney podocytes and the known vascular hyperpermeability effects of elevated VEGF levels, renal disorders such as microalbuminuria, proteinuria, oliguria, WO 00/27414 PCTIUS9-9/25903 -17 5 electrolyte imbalance (often encountered as diabetic complications) and nephrotic syndrome (especially when hypoxia-induced following bums, shock or trauma) may be treated according to the method of this invention. (15) Protein extravasation and diapedesis, that commonly accompanies edema and leads to excessive matrix deposition and stromal proliferation, contribute to the 10 progression of other disorders. These disorders include hyperviscosity syndrome, liver cirrhosis, fibroses, keloid and formation of undesired scar tissue. Inhibition of VEGF-mediated hyperpermeability will impede such disease progression. (16) Significant amounts of VEGF isoforms are known to be stored in platelets, mast 15 cells, etc. and in extracellular matrices. In certain situations, these stores of VEGF/VPF can be rapidly liberated and thereby contribute to acute vascular hyperpermeability. From these diverse examples, it is readily apparent that edema occurs under a 20 variety of physiological conditions and VEGF/VPF or a related analog is strongly implicated in edema formation and extravasation. The compounds of this invention minimize the edematous state associated with macular edema, aphakic/pseudoaphakic cystoid macular edema, retinoblastoma, ocular ischemia, ocular inflammatory disease or infection, choroidal melanoma, edematous side-effects induced by iron chelation 25 therapy, pulmonary edema, pleural effusion, pericardial effusion, myocardial infarction, rheumatoid diseases, tissue edema at sites of trauma or allergic inflammation, polyp edema at sites of chronic inflammation, cerebral edema, brain tumor fluid-filled cysts, communicating hydrocephalus, edema associated with organ damage resulting from a bum and edema resulting from an inhalation burn injury. The compounds of this 30 invention also minimize the edematous state associated with skin bums, blisters, erythema multiforme, edematous macules and other skin disorders, brain tumors, ascites and various effusions associated with cancers, carpal tunnel syndrome, altitude "sickness", allergies and hypersensitivity reactions, radioanaphylaxis, radiodermatitis, glaucoma, conjunctivitis, adult respiratory distress syndrome, asthma, bronchitis, 35 ovarian hyperstimulation syndrome, polycystic ovary syndrome, menstrual swelling and cramps, stroke, head trauma, cerebral infarct or occlusion, ulcerations, sprains, fractures, WO 00/27414 PCT/US99/25903 -18 5 effusions associated with synovitis, diabetic complications, liver cirrhosis and the administration of growth factors. The compounds of this invention can also be used to treat microalbuminuria, proteinuria, oliguria, electrolyte imbalance, nephrotic syndrome, hyperviscosity syndrome, exudates, fibroses, keloid and formation of undesired scar tissue. 10 The compounds of this invention can be administered in combination with one or more additional pharmaceutical agents that inhibit or prevent the production of VEGF, attenuate intracellular responses to VEFG, inhibit vascular hyperpermeability, reduce inflammation or inhibit or prevent the formation of edema. The compounds of the invention can be administered prior to, subsequent to or simultaneously with the 15 additional pharmaceutical agent, whichever course of administration is appropriate. The additional pharmaceutical agents include but are not limited to anti-edemic steroids, NSAIDS, ras inhibitors, anti-TNF agents, anti-IL1 agents, antihistamines, PAF antagonists, COX-1 inhibitors, COX-2 inhibitors, NO synthase inhibitors, PKC inhibitors and PI 3 kinase inhibitors. The compounds of the invention and the additional 20 pharmaceutical agents act either additively or synergistically. Thus, the administration of such a combination of substances that inhibit vascular hyperpermeability and/or inhibit the formation of edema can provide greater relief from the deletrious effects of vascular hyperpermeability or edema than the administration of either substance alone. Since the formation of edema often results from the extravasation of fluid from 25 the bloodstream, hypotension often occurs as extravasation takes place. Hypotension can also occur as a result of VEGF or VEGF activator binding to VEGF receptors on vascular endothelial cells. The compounds of this invention minimize the development of hypotension by, it appears, inhibiting the cellular signaling function of KDR that is a consequence of VEGF (or other activating ligand) binding to this receptor. The 30 compounds of this invention inhibit hypotension in individuals when they are administered to the individual.
WO 00/27414 PCT/US99/25903 -19 5 Pharmaceutical Formulations The compounds of this invention can be administered to a human patient by themselves or in pharmaceutical compositions where they are mixed with suitable carriers or excipient(s) at doses to treat or ameliorate vascular hyperpermeability, edema and associated disorders. Mixtures of these compounds can also be administered to the 10 patient as a simple mixture or in suitable formulated pharmaceutical compositions. A therapeutically effective dose further refers to that amount of the compound or compounds sufficient to result in the prevention of edema, VEGF-associated hyperpermeability and/or VEGF-related hypotension progression. Techniques for formulation and administration of the compounds of the instant application may be 15 found in "Remington's Pharmaceutical Sciences," Mack Publishing Co., Easton, PA, latest edition. Routes of Administration Suitable routes of administration may, for example, include oral, eyedrop, rectal, 20 transmucosal, topical, or intestinal administration; parenteral delivery, including intramuscular, subcutaneous, intramedullary injections, as well as intrathecal, direct intraventricular, intravenous, intraperitoneal, intranasal, or intraocular injections. Alternatively, one may administer the compound in a local rather than a systemic manner, for example, via injection of the compound directly into an edematous 25 site, often in a depot or sustained release formulation. Furthermore, one may administer the drug in a targeted drug delivery system, for example, in a liposome coated with endothelial cell-specific antibody. Composition/Formulation 30 The pharmaceutical compositions of the present invention may be manufactured in a manner that is itself known, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or lyophilizing processes. Pharmaceutical compositions for use in accordance with the present invention 35 thus may be formulated in a conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries which facilitate processing of WO 00/27414 PCT/US99/25903 -20 5 the active compounds into preparations which can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen. For injection, the agents of the invention may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hanks's solution, Ringer's solution, or physiological saline buffer. For transmucosal administration, 10 penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art. For oral administration, the compounds can be formulated readily by combining the active compounds with pharmaceutically acceptable carriers well known in the art. Such carriers enable the compounds of the invention to be formulated as tablets, pills, 15 dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient to be treated. Pharmaceutical preparations for oral use can be obtained by combining the active compound with a solid excipient, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. Suitable excipients are, in 20 particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl cellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone (PVP). If desired, disintegrating agents may be added, such as the cross-linked polyvinyl 25 pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate. Dragee cores are provided with suitable coatings. For this purpose, concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. Dyestuffs or pigments may 30 be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses. Pharmaceutical preparations which can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. The push-fit capsules can contain the active ingredients in 35 admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active WO 00/27414 PCTIUS99/25903 -21 5 compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers may be added. All formulations for oral administration should be in dosages suitable for such administration. For buccal administration, the compositions may take the form of tablets or 10 lozenges formulated in conventional manner. For administration by inhalation, the compounds for use according to the present invention are conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebuliser, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon 15 dioxide or other suitable gas. In the case of pressurized aerosol, the dosage unit may be determined by providing a valve to deliver a metered amount. Capsules and cartridges of e gelatin for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch. The compounds may be formulated for parenteral administration by injection, 20 e bolus injection or continuous infusion. Formulations for injection may be presented in unit dosage form, e, in ampoules or in multi-dose containers, with an added preservative. The compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents. 25 Pharmaceutical formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form. Additionally, suspensions of the active compounds may be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection 30 suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions. Alternatively, the active ingredient may be in powder form for constitution with 35 a suitable vehicle, e, sterile pyrogen-free water, before use.
- WO 00/27414 PCTIUS99/25903 -22 5 The compounds may also be formulated in rectal compositions such as suppositories or retention enemas, e, containing conventional suppository bases such as cocoa butter or other glycerides. In addition to the formulations described previously, the compounds may also be formulated as a depot preparation. Such long acting formulations may be administered 10 by implantation (for example subcutaneously or intramuscularly or by intramuscular injection). Thus, for example, the compounds may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt. 15 An example of a pharmaceutical carrier for the hydrophobic compounds of the invention is a cosolvent system comprising benzyl alcohol, a nonpolar surfactant, a water-miscible organic polymer, and an aqueous phase. The cosolvent system may be the VPD cosolvent system. VPD is a solution of 3% w/v benzyl alcohol, 8% w/v of the nonpolar surfactant polysorbate 80, and 65% w/v polyethylene glycol 300, made up to 20 volume in absolute ethanol. The VPD cosolvent system (VPD:5W) consists of VPD diluted 1:1 with a 5% dextrose in water solution. This cosolvent system dissolves hydrophobic compounds well, and itself produces low toxicity upon systemic administration. Naturally, the proportions of a cosolvent system may be varied considerably without destroying its solubility and toxicity characteristics. Furthermore, 25 the identity of the cosolvent components may be varied: for example, other low-toxicity nonpolar surfactants may be used instead of polysorbate 80; the fraction size of polyethylene glycol may be varied; other biocompatible polymers may replace polyethylene glycol, e.g. polyvinyl pyrrolidone; and other sugars or polysaccharides may substitute for dextrose. 30 Alternatively, other delivery systems for hydrophobic pharmaceutical compounds may be employed. Liposomes and emulsions are well known examples of delivery vehicles or carriers for hydrophobic drugs. Certain organic solvents such as dimethysulfoxide also may be employed, although usually at the cost of greater toxicity. Additionally, the compounds may be delivered using a sustained-release system, such 35 as semipermeable matrices of solid hydrophobic polymers containing the therapeutic agent. Various sustained-release materials have been established and are well known by - WO 00/27414 PCT/US99/25903 -23 5 those skilled in the art. Sustained-release capsules may, depending on their chemical nature, release the compounds for a few weeks up to over 100 days. Depending on the chemical nature and the biological stability of the therapeutic reagent, additional strategies for protein stabilization may be employed. The pharmaceutical compositions also may comprise suitable solid or gel phase 10 carriers or excipients. Examples of such carriers or excipients include but are not limited to calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin, and polymers such as polyethylene glycols. Many of the organic molecule compounds of the invention may be provided as salts with pharmaceutically compatible counterions. Pharmaceutically compatible salts 15 may be formed with many acids, including but not limited to hydrochloric, sulfuric, acetic, lactic, tartaric, malic, succinic, etc. Salts tend to be more soluble in aqueous or other protonic solvents than are the corresponding free base forms. Effective Dosage 20 Pharmaceutical compositions suitable for use in the present invention include compositions wherein the active ingredients are contained in an effective amount to achieve its intended purpose. More specifically, a therapeutically effective amount means an amount effective to prevent development of or to alleviate the existing symptoms of the subject being treated. Determination of the effective amounts is well 25 within the capability of those skilled in the art. The effective dose of the compound inhibits the cellular signaling function of KDR sufficiently to suppress vascular hyperpermeability without causing significant adverse effects due to inhibition of the Flt-1 or other tyrosine kinase functions. Certain compounds which have such activity can be identified by in vitro assays that determine 30 the dose-dependent inhibition of KDR tyrosine kinase. Preferred compounds have an
IC
5 0 versus KDR that is significantly lower than the IC 5 0 against Flt-1 or other PTK's determined under similar conditions of [ATP]/K.(ATP) and substrate (ideally, -1 00x selective for KDR tyrosine kinase).
WO 00/27414 PCTIUS99/25903 -24 5 For any compound used in the method of the invention, the therapeutically effective dose can be estimated initially from cellular assays. For example, a dose can be formulated in cellular and animal models to achieve a circulating concentration range that includes the IC 5 0 as determined in cellular assays (ie, the concentration of the test compound which achieves a half-maximal inhibition of the cellular signaling function 10 KDR, usually in response to VEGF or another activating stimulus). The determination of the cellular IC 5 0 in the presence of 3 to 5% serum albumin may approximate the binding effects of plasma protein on the compound. Such information can be used to more accurately determine useful doses in humans. Further, the most preferred compounds for systemic administration effectively inhibit the cellular signaling function 15 KDR in intact cells at levels that are safely achievable in plasma. A therapeutically effective dose refers to that amount of the compound that results in amelioration of symptoms in a patient. Toxicity and therapeutic efficacy of such compounds can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e, for determining the maximum tolerated dose 20 (MTD) and the ED,, (effective dose for 50% maximal response). The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio between MTD and ED,,. Compounds which exhibit high therapeutic indices are preferred. The data obtained from these cell culture assays and animal studies can be used in formulating a range of dosage for use in humans. The dosage of such 25 compounds lies preferably within a range of circulating concentrations that include the
ED
50 with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized. The exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition. (See e Fingl et al., 1975, in "The 30 Pharmacological Basis of Therapeutics", Ch. 1 p1). In the treatment of crises, the administration of an acute bolus or an infusion approaching the MTD may be required to obtain a rapid response. Dosage amount and interval may be adjusted individually to provide plasma levels of the active moiety which are sufficient to maintain the KDR modulating effects, 35 or minimal effective concentration (MEC). The MEC will vary for each compound but can be estimated from in vitro data; 2 the concentration necessary to achieve 50-90% - WO 00/27414 PCT/US99/25903 -25 5 inhibition of KDR tyrosine kinase using the assays described herein. Dosages necessary to achieve the MEC will depend on individual characteristics and route of administration. However, HPLC assays or bioassays can be used to determine plasma concentrations. Dosage intervals can also be determined using the MEC value. Compounds 10 should be administered using a regimen which maintains plasma levels above the MEC for 10-90% of the time, preferably between 30-90% and most preferably between 50 90% until the desired amelioration of symptoms is achieved. In cases of local administration or selective uptake, the effective local concentration of the drug may not be related to plasma concentration. 15 The amount of composition administered will, of course, be dependent on the subject being treated, on the subject's weight, the severity of the affliction, the manner of administration and the judgment of the prescribing physician. Packaging 20 The compositions may, if desired, be presented in a pack or dispenser device which may contain one or more unit dosage forms containing the active ingredient. The pack may for example comprise metal or plastic foil, such as a blister pack. The pack or dispenser device may be accompanied by instructions for administration. Compositions comprising a compound of the invention formulated in a compatible pharmaceutical 25 carrier may also be prepared, placed in an appropriate container, and labeled for treatment of an indicated condition. Suitable conditions indicated on the label may include treatment of an edema, inhibition of vascular hyperpermeability and extravasation, stromal deposition, minimization of VEGF-related hypotension, and the like. 30 EXEMPLIFICATIONS I. In Vitro PTK Assays The following in vitro assays may be used to determine the level of activity and effect of the different compounds of the present invention on one or more of the PTKs. 35 Similar assays can be designed along the same lines for other tyrosine kinases using techniques well known in the art.
WO 00/27414 PCT/US99/25903 -26 5 A. KDR Tyrosine Kinase Production Using Baculovirus System: The coding sequence for the human KDR intra-cellular domain (aa789-1354) was generated through PCR using cDNAs isolated from HUVEC cells. A poly-His, sequence was introduced at the N-terminus of this protein as well. This fragment was 10 cloned into transfection vector pVL1393 at the Xba 1 and Not 1 site. Recombinant baculovirus (BV) was generated through co-transfection using the BaculoGold Transfection reagent (PharMingen). Recombinant BV was plaque purified and verified through Western analysis. For protein production, SF-9 cells were grown in SF-900-H medium at 2 x 10*/ml, and were infected at 0.5 plaque forming units per cell (MOI). 15 Cells were harvested at 48 hours post infection. B. Purification of KDR SF-9 cells expressing (His) 6 KDR(aa789-1354) were lysed by adding 50 ml of Triton X-100 lysis buffer (20 mM Tris, pH 8.0, 137 mM NaCl, 10% glycerol, 1% Triton 20 X-100, 1mM PMSF, 10j g/ml aprotinin, 1 j.g/ml leupeptin) to the cell pellet from 1L of cell culture. The lysate was centrifuged at 19,000 rpm in a Sorval SS-34 rotor for 30 min at 4*C. The cell lysate was applied to a 5 ml NiCl 2 chelating sepharose column, equilibrated with 50 mM HEPES, pH7.5, 0.3 M NaCl. KDR was eluted using the same buffer containing 0.25 M imidazole. Column fractions were analyzed using SDS-PAGE 25 and an ELISA assay (below) which measures kinase activity. The purified KDR was exchanged into 25mM HEPES, pH7.5, 25mM NaCl, 5 mM DTT buffer and stored at -80 0 C. C. Human Tie-2 Kinase Production and Purification 30 The coding sequence for the human Tie-2 intra-cellular domain (aa775-1124) was generated through PCR using cDNAs isolated from human placenta as a template. A poly-His, sequence was introduced at the N-terminus and this construct was cloned into transfection vector pVL 1939 at the Xba 1 and Not 1 site. Recombinant BV was generated through co-transfection using the BaculoGold Transfection reagent 35 (PharMingen). Recombinant BV was plaque purified and verified through Western analysis. For protein production, SF-9 insect cells were grown in SF-900-II medium at WO 00/27414 PCT/US99125903 -27 5 2 x 10'/ml, and were infected at MOI of 0.5. Purification of the His-tagged kinase used in screening was analogous to that described for KDR. D. Human Fit-1 Tyrosine Kinase Production and Purification The baculoviral expression vector pVL1393 (Phar Mingen, Los Angeles, CA) 10 was used. A nucleotide sequence encoding poly-His 6 was placed 5' to the nucleotide region encoding the entire intracellular kinase domain of human Flt-1 (amino acids 786 1338). The nucleotide sequence encoding the kinase domain was generated through PCR using cDNA libraries isolated from HUVEC cells. The histidine residues enabled affinity purification of the protein as a manner analogous to that for KDR (Part B.) and 15 ZAP70 (Part F.) SF-9 insect cells were infected at a 0.5 multiplicity and harvested 48 hours post infection. E. Lck and EGFR Tyrosine Kinase Sources Lck or truncated forms of Lck were commercially obtained (e.g. Upstate 20 Biotechnology Inc., Saranac Lake, NY or Santa Cruz Biotechnology, Inc., Santa Cruz, CA) or were purified from known natural or recombinant sources using conventional methods. EGFR was purchased from Sigma (Cat # E-3641; ~ 500 units/50 pl) and the EGF ligand was acquired from Oncogene Research Products/Calbiochem (Cat # PF011 100). 25 F. ZAP70 Tyrosine Kinase Production The Baculoviral expression vector pVL 1393 (Phar Mingen, Los Angeles, CA) was used. A nucleotide sequence encoding poly-His, was placed 5' to the nucleotide 30 region encoding the entire ZAP70 (amino acids 1-619). The nucleotide sequence encoding the ZAP70 coding region was generated through PCR using cDNA libraries isolated from Jurkat immortalized T-cells. The histidine residues enabled affinity purification of the protein (see Part B.). The LVPRGS bridge constituted a recognition sequence for proteolytic cleavage by thrombin, thereby enabling removal of the affinity 35 tag from the enzyme. SF-9 insect cells were infected at a 0.5 multiplicity and harvested 48 hours post infection.
- WO 00/27414 PCT/US99125903 -28 5 G. Purification of ZAP70 SF-9 cells were lysed in a buffer containing 20 mM Tris, pH 8.0, 137 mM NaCl, 10% glycerol, 1% Triton X-100, 1 mM PMSF, 1 ptg/ml leupeptin, 10 pg/ml aprotinin and 1 mM sodium orthovanadate. The soluble lysate was applied to a chelating 1o Sepharose Hi Trap column (Pharmacia) equilibrated in 50 mM HEPES, pH 7.5, 0.3 M NaCl. The fusion protein was eluted with 250 mM imidazole. The recovered enzyme was stored in buffer containing 50 mM HEPES, pH 7.5, 50 mM NaCl and 5 mM DTT. H. Enzyme Linked Immunosorbent Assay (ELISA) For RTKs 15 Enzyme linked immunosorbent assays (ELISA) were used to detect and measure the presence of tyrosine kinase activity. The ELISA were conducted according to known protocols which are described in, for example, Voller, et al., 1980, "Enzyme Linked Immunosorbent Assay," In: Manual of Clinical Immunology, 2d ed., edited by Rose and Friedman, pp 359-371 Am. Soc. of Microbiology, Washington, D.C. 20 The disclosed protocol was adapted for determining activity with respect to a specific RTK. For example, a preferred protocol for conducting the ELISA experiments for KDR is provided below. Adaptation of this protocol for determining a compound's activity for other members of the RTK family, as well as non-receptor tyrosine kinases, are well within the abilities of those in the art. For purposes of determining inhibitor 25 selectivity, a universal PTK substrate (e.g., random copolymer of poly (Glu 4 Tyr), 20,000-50,000 MW) was employed together with ATP (typically 5 pM) at concentrations approximately twice the apparent Km in the assay. KDR IN VITRO ELISA 30 The following procedure was used to assay the inhibitory effect of compounds of this invention on KDR tyrosine kinase activity: Buffers and Solutions: PGT: Poly (Glu,Tyr) 4:1 - WO 00/27414 PCTIUS99/25903 -29 5 Store powder at -20 0 C. Dissolve powder in phosphate buffered saline (PBS) for 50mg/ml solution. Store 1ml aliquots at -20'C. When making plates, dilute to 250p.g/ml in Gibco PBS. Reaction Buffer: 10 100mM Hepes, 20mM MgCl 2 , 4mM MnCl 2 , 5mM DTT, 0.02%BSA, 200pM NaVO 4 , pH 7.10 ATP: Store aliquots of 100mM at -20*C. Dilute to 20pM in water 15 Washing Buffer: PBS with 0.1% Tween 20 Antibody Diluting Buffer: 0.1% bovine serum albumin (BSA) in PBS 20 TMB Substrate: mix TMB substrate and peroxide solutions 9:1 just before use or use K-Blue Substrate from Neogen 25 Stop Solution: IM Phosphoric Acid Procedure 30 1. Plate Preparation: Dilute PGT stock (50mg/ml, frozen) in PBS to a 250 g/ml. Add 125d per well of Corning modified flat bottom high affinity ELISA plates (Corning #25805 96). Add 125l PBS to blank wells. Cover with sealing tape and incubate overnight 37"C. Wash lx with 250pl washing buffer and dry for about 2hrs in 35 37 0 C dry incubator.
WO 00/27414 PCT/US99/25903 -30 5 Store coated plates in sealed bag at 4*C until used. 2. Tyrosine Kinase Reaction: -Prepare inhibitor solutions at a 4x concentration in 20% DMSO in water. -Prepare reaction buffer 10 -Prepare enzyme solution so that desired units are in 50pd, e.g. for KDR make to 1 ng/pl for a total of 50ng per well in the reactions. Store on ice. -Make 4x ATP solution to 20pM from 100mM stock in water. Store on ice -Add 50d of the enzyme solution per well (typically 5-50 ng 15 enzyme/well depending on the specific activity of the kinase) -Add 25p4l 4x inhibitor -Add 25pd 4x ATP for inhibitor assay 20 -Incubate for 10 minutes at room temperature -Stop reaction by adding 50 Ll 0.05N HCl per well 25 -Wash plate **Final Concentrations for Reaction: ATP: 5p M 5% DMSO 30 3. Antibody Binding -Dilute lmg/ml aliquot of PY20-HRP (Pierce) antibody (a phosphotyrosine antibody) to 50ng/ml in 0.1% BSA in PBS by a 2 step dilution (100x, then 200x) 35 -Add 100 i Ab per well. Incubate 1 hr at room temp. Incubate lhr at 4C.
WO 00/27414 PCTIUS99/25903 -31 5 -Wash 4x plate 4. Color reaction -Prepare TMB substrate and add 100pl per well 10 -Monitor OD at 650nn until 0.6 is reached -Stop with 1M Phosphoric acid. Shake on plate reader. -Read OD immediately at 450nm 15 Optimal incubation times and enzyme reaction conditions vary slightly with enzyme preparations and are determined empirically for each lot. Analogous assay conditions were used for Flt-1, Tie-2, EGFR and ZAP70. For Lck, the Reaction Buffer utilized was 100 mM MOPSO, pH 6.5, 4 mM MnCl 2 , 20 mM MgCl 2 , 5 mM DTT, 0.2% BSA, 200 mM NaVO 4 under the analogous assay conditions. 20 PKC kinase source The catalytic subunit of PKC may be obtained commercially (Calbiochem). 25 PKC kinase assay A radioactive kinase assay was employed following a published procedure (Yasuda, I., Kirshimoto, A., Tanaka, S., Tominaga, M., Sakurai, A., Nishizuka, Y. Biochemical and Biophysical Research Communication 3:166, 1220-1227 (1990)). Briefly, all reactions were performed in a kinase buffer consisting of 50 mM Tris-HCl 30 pH7.5, 10mM MgCl 2 , 2mM DTT, 1mM EGTA, 100 pM ATP, 8 pM peptide, 5% DMSO and 3 P ATP (8Ci/mM). Compound and enzyme were mixed in the reaction vessel and the reaction initiated by addition of the ATP and substrate mixture. Following termination of the reaction by the addition of 10 pl stop buffer (5 mM ATP in 75mM phosphoric acid), a portion of the mixture was spotted on phosphocellulose 35 filters. The spotted samples were washed 3 times in 75 mM phosphoric acid at room WO 00/27414 PCT/US99/25903 -32 5 temperature for 5 to 15 minutes. Incorporation of radiolabel was quantified by liquid scintillation counting. Estrogen Receptor Binding Assay Binding of 1 nM radiolabeled 17p-estradiol to human estrogen receptor in the 10 cytosol of MCF-7 mammary carcinoma cells was determined following incubation for 20 h at 4'C using the reaction conditions of Shein et al., Cancer Res. 45:4192 (1985) (herein incorporated by reference). Following incubation, the cytosol fractions were mixed with a suspension of dextran-coated charcoal for 10 min at 4 C, centrifuged, and the supernatants collected. Bound radioactivity remaining in the charcoal supernatant 15 was measured with a scintillation counter (LS 6000, Beckman) using a liquid scintillation cocktail (Formula 989 Packard). The compounds were tested simultaneously at eight concentrations in duplicate to obtain a competition curve in order to quantify the inhibitory activity. The specific radioligand binding to the estrogen receptor was defined as the difference between total binding and nonspecific 20 binding determined in the presence of an excess of unlabeled 17p-estradiol (6 tM). Results The following inhibitory concentrations of a representative compound with the structural formula: 25 WO 00/27414 PCT/US99/25903 -33 0 P N+H NH N 5 were obtained: WO 00/27414 PCT/US99/25903 -34 5 Assay IC50 for Compound KDR 0.20 iM Flt-1 > 50.0 pM Lck > 50.0 pM TIE2 > 50.0 4M ZAP70 > 50.0 pM EGFR > 50.0 pM PKC >20 pM Estrogen Receptor >10.0 pM (< 10% inh @ 10 p.M) These results demonstrate that compounds of the present invention and exemplified herein have notable inhibitory activity for KDR tyrosine kinase and are particularly selective as KDR tyrosine kinase inhibitors. 10 II. Cellular RTK Assays The following cellular assay was used to determine the level of activity and effect of the different compounds of the present invention on KDR. Similar assays can be designed along the same lines for other tyrosine kinases using appropriate antibody 15 reagents and techniques such as immunoprecipitations and Western blotting well known in the art.
- WO 00/27414 PCT/US99/25903 -35 5 A. VEGF-Induced KDR Phosphorvlation in Human Umbilical Vein Endothelial Cells (HUVEC) as Measured by Western Blots. 1. HUVEC cells (from pooled donors) were purchased from Clonetics (San Diego, CA) and cultured according to the manufacturer directions. Only early passages (3-8) 10 were used for this assay. Cells were cultured in 100 mm dishes (Falcon for tissue culture; Becton Dickinson; Plymouth, England) using complete EBM media (Clonetics). 2. For evaluating a compound's inhibitory activity, cells were trypsinized and 15 seeded at 0.5-1.0 x 10' cells/well in each well of 6-well cluster plates (Costar; Cambridge, MA). 3. 3-4 days after seeding, plates were 90-100% confluent. Medium was removed from all the wells, cells were rinsed with 5-10ml of PBS and incubated 18-24h with 5ml 20 of EBM base media with no supplements added (i.e., serum starvation). 4. Serial dilutions of inhibitors were added in 1ml of EBM media (25p.M, 5pM, or 1 LM final concentration to cells and incubated for one hour at 370 C. Human recombinant VEGF 6 5 ) ( R & D Systems) was then added to all the wells in 2 ml of 25 EBM medium at a final concentration of 50ng/ml and incubated at 37'C for 10 minutes. Control cells untreated or treated with VEGF only were used to assess background phosphorylation and phosphorylation induction by VEGF. 5. All wells were then rinsed with 5-1 Oml of cold PBS containing 1mM sodium 30 orthovanadate (Sigma) and cells were lysed and scraped in 20041 of RIPA buffer (50mM Tris-HC1) pH7, 150mM NaCl, 1% NP-40, 0.25% sodium deoxycholate, 1mM EDTA) containing protease inhibitors (PMSF 1mM, aprotinin 1 pg/ml, pepstatin 1Lpg/ml, leupeptin 1pg/ml, Na vanadate 1mM, Na fluoride 1mM) and 1 g/ml of DNase (all chemicals from Sigma Chemical Company, St Louis, MO). The lysate was spun at 35 14,000 rpm for 30min, to eliminate nuclei.
- WO 00/27414 PCT/US99/25903 -36 5 6. Equal amounts of proteins were then precipitated by addition of cold (-20*C) ethanol (2 volumes) for a minimum of 1 hour or a maximum of overnight. Pellets were reconstituted in Laemli sample buffer containing 5% p-mercaptoethanol (BioRad; Hercules, CA) and boiled for 5min. The proteins were resolved by polyacrylamide gel electrophoresis (6%, 1.5mm Novex, San Deigo, CA) and transferred onto a 10 nitrocellulose membrane using the Novex system. After blocking with bovine serum albumin (3%), the proteins were probed overnight with anti-KDR polyclonal antibody (C20, Santa Cruz Biotechnology; Santa Cruz, CA) or with anti-phosphotyrosine monoclonal antibody (4G10, Upstate Biotechnology, Lake Placid, NY) at 4'C. After washing and incubating for 1 hour with HRP-conjugated F(ab)2 of goat anti-rabbit or 15 goat-anti-mouse IgG the bands were visualized using the emission chemiluminescience (ECL) system (Amersham Life Sciences, Arlington Height, IL). Results The inhibitory concentrations of a representative compound I with the structural 20 formula: 0--0 N+H N were: WO 00/27414 PCT/US99/25903 -37 5 HUVEC Cellular IC 50 for Compound KDR phosphorylation 5-10 1 .M This compound also has demonstrated KDR tyrosine kinase selectivity (See section I). These results demonstrate that suitable compounds of the present invention have notable inhibitory activity for VEGF-induced tyrosine phosphorylation of KDR tyrosine 10 kinase in endothelial cells. III. Uterine Edema Model This assay measures the capacity of compounds to inhibit the acute increase in uterine weight in mice which occurs in the first few hours following estrogen 15 stimulation. This early onset of uterine weight increase is known to be due to edema caused by increased permeability of uterine vasculature. Cullinan-Bove and Koss (Endocrinology (1993), 133:829-837) demonstrated a close temporal relationship of estrogen-stimulated uterine edema with increased expression of VEGF mRNA in the uterus. These results have been confirmed by the use of neutralizing monoclonal 20 antibody to VEGF which significantly reduced the acute increase in uterine weight following estrogen stimulation (WO 97/42187). Hence, this system can serve as an in vivo model for inhibition of VEGF-mediated hyperpermeability and edema. Materials: All hormones were purchased from Sigma (St. Louis, MO) or Cal 25 Biochem (La Jolla, CA) as lyophilized powders and prepared according to supplier instructions. Vehicle components (DMSO, Cremaphor EL) were purchased from Sigma (St. Louis, MO). Mice (Balb/c, 8-12 weeks old) were purchased from Taconic 30 (Germantown, NY) and housed in a pathogen-free animal facility in accordance with institutional Animal Care and Use Committee Guidelines.
WO 00/27414 PCT/US99/25903 -38 5 Method: Day 1: Balb/c mice were given an intraperitoneal (i.p.) injection of 12.5 units of pregnant mare's serum gonadotropin (PMSG). 10 Day 3: Mice received 15 units of human chorionic gonadotropin (hCG) i.p. Day 4: Mice were randomized and divided into groups of 5-10. Test compounds were administered by i.p., i.v. or p.o. routes depending on solubility and vehicle at doses ranging from 1-200 mg/kg. Vehicle control group 15 received vehicle only and two groups were left untreated. Typically thirty minutes later, experimental, vehicle and one of the untreated groups were given an i.p. injection of 17p-estradiol (500 pg/kg). After 2-3 hours, the animals were sacrificed by CO 2 inhalation. 20 Following a midline incision, each uterus was isolated and removed by cutting just below the cervix and at the junctions of the uterus and oviducts. Fat and connective tissue were removed with care not to disturb the integrity of the uterus prior to weighing. Mean weights of treated groups were compared to untreated or vehicle treated groups. 25 Significance was determined by Student's test. Non-stimulated control group was used to monitor estradiol response. Results The percent inhibition of uterine edema following estradiol stimulation for a 30 representative compound with the structural formula: WO 00/27414 PCTIUS99/25903 -39 5 0 o N+H N H N was obtained for three routes of administration at 100 mg/kg doses. Administration % Inhibition p Value Route p.o. 17 ns i.v. 57 0.01 i.p. 52 0.04 10 Compound I has been demonstrated herein to be KDR-selective for inhibition of kinase activity in vitro and efficacious in blocking cellular autophosphorylation of KDR in response to a VEGF-stimulus. These results demonstrate that suitable compounds of the present invention such 15 as Compound I which selectively inhibit KDR function effectively block the formation of edema. The results also demonstrate that i.v. and i.p. administration are particularly WO 00/27414 PCTIUS99/25903 -40 5 effective for this compound. Importantly, similar anti-edemic efficacy results have also been obtained with numerous structurally-distinct selective inhibitors of KDR function. EQUIVALENTS While this invention has been particularly shown and described with references 10 to preferred embodiments thereof, it will be understood by those skilled in the art that various changes in form and details may be made therein without departing from the spirit and scope of the invention as defined by the appended claims. Those skilled in the art will recognize or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention is described specifically herein. Such equivalents are intended to be encompassed in the scope of the claims.

Claims (30)

1. A method of inhibiting vascular hyperpermeability in an individual comprising 10 the inhibition of the cellular signaling function of KDR.
2. The method of Claim 1 wherein said inhibition of the cellular signaling function of KDR is selective for the KDR signaling function. 15
3. The method of Claim 1 wherein said cellular signaling function of KDR is stimulated by the binding of an activating ligand to the receptor portion of KDR.
4. The method of Claim 3 wherein said inhibition of the cellular signaling function of KDR is selective for the KDR signaling function. 20
5. The method of Claim I wherein said inhibition of the cellular signaling function of KDR is a process selected from the group consisting of blocking the production of an activating ligand, modulating the binding of the activating ligand to the KDR tyrosine kinase receptor, disrupting the dimerization of the 25 receptor, blocking KDR trans-phosphorylation, inhibiting the activity of the KDR tyrosine kinase, impairing the recruitment of intracellular substrates of KDR, and interrupting the downstream signaling initiated by the phosphorylation activity of the KDR tyrosine kinase. 30
6. The method of Claim 5 wherein said inhibition of the cellular signaling function of KDR is selective for the KDR signaling function.
7. The method of Claim 1 wherein said inhibition occurs by the administration of a compound to said individual. 35 WO 00/27414 PCT/US99/25903 -42 5
8. The method of Claim 7 wherein said compound inhibits the catalytic kinase activity of said KDR.
9. The method of Claim 7 wherein said compound is an antagonist of KDR tyrosine kinase activation. 10
10. The method of Claim 7 wherein said compound selectively inhibits the phosphorylation of KDR kinase substrates. 15
11. The method of Claim 7 wherein said compound is selective for said KDR tyrosine kinase.
12. The method of Claim 11 wherein said compound is selected from the group consisting of peptides, antibodies and organic molecules, wherein said 20 compound binds to said KDR tyrosine kinase.
13. The method of Claim 12 wherein the administration of said compound inhibits the formation of a disease state selected from the group consisting of macular edema, aphakic/pseudoaphakic cystoid macular edema, retinoblastoma, ocular 25 ischemia, ocular inflammatory disease or infection, choroidal melanoma, edematous side-effects induced by iron chelation therapy, pulmonary edema, myocardial infarction, rheumatoid diseases, anaphylaxis, tissue edema at sites of trauma and allergic inflammation, allergies, hypersensitive reactions, polyp edema at sites of chronic inflammation, cerebral edema, brain tumor fluid-filled 30 cysts, communicating hydrocephalus, carpal tunnel syndrome, organ damage resulting from a bum, inhalation bum injury, skin bums, blistering associated with sunburn, irritation or infection, erythema multiforme, edematous macules and other skin disorders, brain tumors, tumor effusions, lung or breast carcinomas, ascites, pleural effusions, pericardial effusions, high altitude 35 "sickness", radioanaphylaxis, radiodermatitis, glaucoma, conjunctivitis, choroidal melanoma, adult respiratory distress syndrome, asthma, bronchitis, WO 00/27414 PCT/US99/25903 -43 5 ovarian hyperstimulation syndrome, polycystic ovary syndrome, menstrual swelling, menstrual cramps, stroke, head trauma, cerebral infarct or occlusion, hypotension, ulcerations, sprains, fractures, effusions associated with synovitis, diabetic complications, hyperviscosity syndrome, liver cirrhosis, microalbuminuria, proteinuria, oliguria, electrolyte imbalance, nephrotic 10 syndrome, exudates, fibroses, keloid, and the administration of growth factors.
14. The method of Claim 11 wherein adverse effects associated with an alteration in the cellular signaling function of tyrosine kinases other than KDR are avoided when said compound is administered. 15
15. The method of Claim 7 wherein said compound is selected from the group consisting of single-chain antibodies, KDR-specific ribozymes and anti-sense polynucletodies, wherein said compound is introduced or produced intracellularly thereby inhibiting the proper presentation of functional KDR 20 tyrosine kinase.
16. The method of Claim 7 wherein said compound is administered in combination with a pharmaceutical agent selected from the group consisting of an anti endemic steroid, a Ras inhibitor, anti-TNF agents, anti-IL1 agents, an 25 antihistamine, a PAF-antagonist, a COX-1 inhibitor, a COX-2 inhibitor, a NO synthase inhibitor, a nonsteroidal anti-inflammatory agent (NSAID), a PKC inhibitor and a P1 3 kinase inhibitor.
17. A method of inhibiting a physiological process or state in an individual, said 30 physiological process or state selected from the group consisting of edema formation, diapedesis, extravasation, effusion, exudation, ascites formation, matrix deposition and vascular hypotension, wherein said inhibiting comprises the administration of a compound that inhibits the cellular signaling function of KDR. 35 WO 00/27414 PCT/US99/25903 -44 5
18. The method of Claim 17 wherein said compound is selective for said KDR tyrosine kinase.
19. The method of Claim 18 wherein said compound is selected from the group consisting of peptides, antibodies and organic molecules, wherein said 10 compound binds to said KDR tyrosine kinase.
20. The method of Claim 19 wherein the administration of said compound inhibits the formation of a disease state selected from the group consisting of macular edema, aphakic/pseudoaphakic cystoid macular edema, retinoblastoma, ocular 15 ischemia, ocular inflammatory disease or infection, choroidal melanoma, edematous side-effects induced by iron chelation therapy, pulmonary edema, myocardial infarction, rheumatoid diseases, anaphylaxis, tissue edema at sites of trauma and allergic inflammation, allergies, hypersensitive reactions, polyp edema at sites of chronic inflammation, cerebral edema, brain tumor fluid-filled 20 cysts, communicating hydrocephalus, carpal tunnel syndrome, organ damage resulting from a bum, inhalation bum injury, skin burns, blistering associated with sunburn, irritation or infection, erythema multiforme, edematous macules and other skin disorders, brain tumors, tumor effusions, lung or breast carcinomas, ascites, pleural effusions, pericardial effusions, high altitude 25 "sickness", radioanaphylaxis, radiodermatitis, glaucoma, conjunctivitis, choroidal melanoma, adult respiratory distress syndrome, asthma, bronchitis, ovarian hyperstimulation syndrome, polycystic ovary syndrome, menstrual swelling, menstrual cramps, stroke, head trauma, cerebral infarct or occlusion, hypotension, ulcerations, sprains, fractures, effusions associated with synovitis, 30 diabetic complications, hyperviscosity syndrome, liver cirrhosis, microalbuminuria, proteinuria, oliguria, electrolyte imbalance, nephrotic syndrome, exudates, fibroses, keloid, and the administration of growth factors.
21. The method of Claim 17 wherein said compound inhibits the catalytic kinase 35 activity of said KDR. WO 00/27414 PCT/US99/25903 -45. 5
22. The method of Claim 17 wherein said compound is an antagonist of KDR tyrosine kinase activation.
23. The method of Claim 17 wherein said compound selectively inhibits the phosphorylation of KDR kinase substrates. 10
24. The method of Claim 17 wherein said compound is selective for said KDR tyrosine kinase.
25. The method of Claim 17 wherein said cellular signaling function of KDR is 15 stimulated by the binding of an activating ligand to the receptor portion of KDR.
26. The method of Claim 25 wherein said compound is selective for said KDR tyrosine kinase. 20
27. The method of Claim 17 wherein said compound is selected from the group consisting of single-chain antibodies, KDR-specific ribozymes and anti-sense polynucletodies, wherein said compound is introduced or produced intracellularly thereby inhibiting the proper presentation of functional KDR tyrosine kinase. 25
28. The method of Claim 17 wherein said inhibition of the cellular signaling function of KDR is a process selected from the group consisting of blocking the production of an activating ligand, modulating the binding of the activating ligand to the KDR tyrosine kinase receptor, disrupting the dimerization of the 30 receptor, blocking KDR trans-phosphorylation, inhibiting the activity of the KDR tyrosine kinase, impairing the recruitment of intracellular substrates of KDR, and interrupting the downstream signaling initiated by the phosphorylation activity of the KDR tyrosine kinase. WO 00/27414 PCTIUS99/25903 -46 5
29. The method of Claim 17 wherein adverse effects associated with an alteration in the cellular signaling function of tyrosine kinases other than KDR are avoided when said compound is administered.
30. The method of Claim 17 wherein said compound is administered in combination 10 with a pharmaceutical agent selected from the group consisting of an anti endemic steroid, a Ras inhibitor, anti-TNF agents, anti-ILl agents, an antihistamine, a PAF-antagonist, a COX-1 inhibitor, a COX-2 inhibitor, a NO synthase inhibitor, a nonsteroidal anti-inflammatory agent (NSAID), a PKC inhibitor and a PI, kinase inhibitor. 15
AU19080/00A 1998-11-06 1999-11-03 Inhibition of the formation of vascular hyperpermeability Abandoned AU1908000A (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US10746298P 1998-11-06 1998-11-06
US60107462 1998-11-06
PCT/US1999/025903 WO2000027414A2 (en) 1998-11-06 1999-11-03 Inhibition of the formation of vascular hyperpermeability

Publications (1)

Publication Number Publication Date
AU1908000A true AU1908000A (en) 2000-05-29

Family

ID=22316735

Family Applications (1)

Application Number Title Priority Date Filing Date
AU19080/00A Abandoned AU1908000A (en) 1998-11-06 1999-11-03 Inhibition of the formation of vascular hyperpermeability

Country Status (19)

Country Link
EP (1) EP1126842A2 (en)
JP (1) JP2002529421A (en)
KR (1) KR20010080952A (en)
CN (1) CN1342077A (en)
AR (1) AR023912A1 (en)
AU (1) AU1908000A (en)
BG (1) BG105476A (en)
BR (1) BR9915139A (en)
CA (1) CA2347916A1 (en)
CO (1) CO5150183A1 (en)
CZ (1) CZ20011564A3 (en)
HU (1) HUP0104302A3 (en)
ID (1) ID29063A (en)
IL (1) IL142583A0 (en)
NO (1) NO20012218L (en)
PL (1) PL348163A1 (en)
SK (1) SK5052001A3 (en)
TR (1) TR200102278T2 (en)
WO (1) WO2000027414A2 (en)

Families Citing this family (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP4731016B2 (en) 1998-12-22 2011-07-20 ジェネンテック, インコーポレイテッド Vascular endothelial growth factor antagonists and their uses
IL144144A0 (en) 1999-01-13 2002-05-23 Bayer Ag Omega-carboxy aryl substituted diphenyl ureas as p38 kinase inhibitors
US8124630B2 (en) 1999-01-13 2012-02-28 Bayer Healthcare Llc ω-carboxyaryl substituted diphenyl ureas as raf kinase inhibitors
ATE392215T1 (en) * 1999-12-22 2008-05-15 Scripps Research Inst MODULATORS AND INHIBITORS OF ANGIOGENESIS AND VASCULAR PERMEABILITY
DE10021246A1 (en) * 2000-04-25 2001-10-31 Schering Ag New N-substituted benzamide derivatives are tyrosine kinase KDR and FLT inhibitors useful e.g. for treating tumors, psoriasis, rheumatoid arthritis, diabetic retinopathy or liver sclerosis
CA2454976C (en) * 2001-07-26 2011-05-10 Santen Pharmaceutical Co., Ltd. Therapeutic agent for glaucoma comprising compound having pi3 kinase inhibitory action as active ingredient
PT1478358E (en) 2002-02-11 2013-09-11 Bayer Healthcare Llc Sorafenib tosylate for the treatment of diseases characterized by abnormal angiogenesis
US8623822B2 (en) 2002-03-01 2014-01-07 Bracco Suisse Sa KDR and VEGF/KDR binding peptides and their use in diagnosis and therapy
US7794693B2 (en) 2002-03-01 2010-09-14 Bracco International B.V. Targeting vector-phospholipid conjugates
US7261876B2 (en) 2002-03-01 2007-08-28 Bracco International Bv Multivalent constructs for therapeutic and diagnostic applications
AU2003278807A1 (en) 2002-03-01 2004-08-13 Bracco International B.V. Kdr and vegf/kdr binding peptides and their use in diagnosis and therapy
ES2440284T3 (en) * 2002-11-14 2014-01-28 Thermo Fisher Scientific Biosciences Inc. SiRNA directed to tp53
WO2006006948A2 (en) 2002-11-14 2006-01-19 Dharmacon, Inc. METHODS AND COMPOSITIONS FOR SELECTING siRNA OF IMPROVED FUNCTIONALITY
HUE039154T2 (en) 2003-03-03 2018-12-28 Dyax Corp Peptides that specifically bind HGF receptor (cMet) and uses thereof
ATE366108T1 (en) 2003-05-20 2007-07-15 Bayer Pharmaceuticals Corp DIARYL UREAS FOR PDGFR-MEDIATED DISEASES
PL1663978T3 (en) 2003-07-23 2008-04-30 Bayer Healthcare Llc Fluoro substituted omega-carboxyaryl diphenyl urea for the treatment and prevention of diseases and conditions
RU2542471C2 (en) 2006-11-15 2015-02-20 Коуда Терапьютикс, Инк. Improved methods and compositions for wound healing
AU2011282902B2 (en) 2010-07-28 2015-01-29 Alcon Research, Ltd. siRNA targeting VEGFA and methods for treatment in vivo
KR101565886B1 (en) 2013-10-23 2015-11-05 전남대학교산학협력단 Method and Kit for Diagnosting of Ovarian Hyperstimulation Syndrome
US10465188B2 (en) 2014-08-22 2019-11-05 Auckland Uniservices Limited Channel modulators
CN106381330A (en) * 2016-08-30 2017-02-08 张建华 Primer and kit for detecting susceptibility of communicating hydrocephalus
CN112055718A (en) * 2018-03-19 2020-12-08 药物抗体公司 anti-VEGFR-2 antibodies
CN109718253B (en) * 2019-01-16 2021-09-24 中国人民解放军总医院 Use of bacteria capable of metabolizing to produce histamine for preventing or treating altitude sickness

Family Cites Families (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BE789948A (en) * 1971-10-13 1973-04-11 Sandoz Sa NEW DERIVATIVES OF PYRAZOLE, THEIR PREPARATION AND THEIR APPLICATION AS MEDICINAL PRODUCTS
US3843664A (en) * 1973-02-20 1974-10-22 Sandoz Ag Substituted naphtho pyrazoles
US3843665A (en) * 1973-04-11 1974-10-22 Sandoz Ag Process for preparing substituted indeno,naphtho and cyclohepta pyrazoles
US3843666A (en) * 1973-05-29 1974-10-22 Sandoz Ag Process for preparing substituted indeno,naphtho and cyclohepta pyrazoles
US5712395A (en) * 1992-11-13 1998-01-27 Yissum Research Development Corp. Compounds for the treatment of disorders related to vasculogenesis and/or angiogenesis
WO1998033917A1 (en) * 1994-11-14 1998-08-06 The Ludwig Institute For Cancer Research Vascular endothelial growth factor c (vegf-c) protein and gene, mutants thereof, and uses thereof
US6346398B1 (en) * 1995-10-26 2002-02-12 Ribozyme Pharmaceuticals, Inc. Method and reagent for the treatment of diseases or conditions related to levels of vascular endothelial growth factor receptor
FR2742662B1 (en) * 1995-12-21 1998-01-23 Centre Nat Rech Scient ANTI-IDIOTYPIC ANTIBODIES OF VASCULAR ENDOTHELIAL GROWTH FACTOR AND THEIR USE AS MEDICAMENTS
US6100071A (en) * 1996-05-07 2000-08-08 Genentech, Inc. Receptors as novel inhibitors of vascular endothelial growth factor activity and processes for their production
DE19638745C2 (en) * 1996-09-11 2001-05-10 Schering Ag Monoclonal antibodies against the extracellular domain of the human VEGF receptor protein (KDR)
WO1998058053A1 (en) * 1997-06-18 1998-12-23 Merck & Co., Inc. Human receptor tyrosine kinase, kdr
DE69818083T2 (en) * 1997-10-06 2004-07-08 Abbott Gmbh & Co. Kg INDENO [1,2-c] -, NAPHTHO [1,2-C] AND BENZO [6,7] CYCLOHEPTA [1,2-c] PYRAZOLE DERIVATIVES
EP1021182A1 (en) * 1997-10-06 2000-07-26 Basf Aktiengesellschaft Indeno[1,2-c] pyrazole derivatives for inhibiting tyrosine kinase activity
CN1311678A (en) * 1998-04-30 2001-09-05 巴斯福股份公司 Substituted tricyclic pyrazole derivatives with protein kinase activity

Also Published As

Publication number Publication date
AR023912A1 (en) 2002-09-04
CN1342077A (en) 2002-03-27
CZ20011564A3 (en) 2002-04-17
KR20010080952A (en) 2001-08-25
HUP0104302A3 (en) 2002-11-28
JP2002529421A (en) 2002-09-10
EP1126842A2 (en) 2001-08-29
BR9915139A (en) 2001-08-07
HUP0104302A2 (en) 2002-03-28
PL348163A1 (en) 2002-05-06
NO20012218L (en) 2001-06-18
BG105476A (en) 2002-02-28
NO20012218D0 (en) 2001-05-04
CO5150183A1 (en) 2002-04-29
SK5052001A3 (en) 2002-10-08
TR200102278T2 (en) 2001-12-21
CA2347916A1 (en) 2000-05-18
WO2000027414A3 (en) 2000-09-08
ID29063A (en) 2001-07-26
WO2000027414A2 (en) 2000-05-18
IL142583A0 (en) 2002-03-10

Similar Documents

Publication Publication Date Title
AU1908000A (en) Inhibition of the formation of vascular hyperpermeability
EP1023063B1 (en) INDENO[1,2-c], NAPHTHO[1,2-c]- AND BENZO[6,7]CYCLOHEPTA[1,2-c]PYRAZOLE DERIVATIVES
Wright et al. Erythropoietin receptor expression in adult rat cardiomyocytes is associated with an acute cardioprotective effect for recombinant erythropoietin during ischemia‐reperfusion injury
EP1268481B1 (en) Kinase inhibitors as therapeutic agents
US7189745B1 (en) Compounds
US6534655B1 (en) Indeno[1,2-C]pyrazole derivatives for inhibiting tyrosine kinase activity
EP1073435B1 (en) Substituted tricyclic pyrazole derivatives with protein kinase activity
KR20020015034A (en) Substituted 1,4-dihydroindeno[1,2-c]pyrazoles as inhibitors of tyrosine kinase
US7005434B2 (en) Compounds and uses thereof
MXPA01004438A (en) Inhibition of the formation of vascular hyperpermeability
MXPA00003359A (en) Indeno[1,2-c]pyrazole derivatives for inhibiting tyrosine kinase activity
MXPA00003360A (en) INDENO[1,2-c]-, NAPHTHO[1,2-c]- AND BENZO[6,7]CYCLOHEPTA[1,2-c]PYRAZOLE DERIVATIVES

Legal Events

Date Code Title Description
MK5 Application lapsed section 142(2)(e) - patent request and compl. specification not accepted