US20160296592A1 - Methods and Compositions for the Treatment of Proteinuric Diseases - Google Patents

Methods and Compositions for the Treatment of Proteinuric Diseases Download PDF

Info

Publication number
US20160296592A1
US20160296592A1 US15/093,588 US201615093588A US2016296592A1 US 20160296592 A1 US20160296592 A1 US 20160296592A1 US 201615093588 A US201615093588 A US 201615093588A US 2016296592 A1 US2016296592 A1 US 2016296592A1
Authority
US
United States
Prior art keywords
upar
radical
optionally substituted
radicals
integrin
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US15/093,588
Inventor
Jochen Reiser
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.)
General Hospital Corp
Original Assignee
General Hospital Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by General Hospital Corp filed Critical General Hospital Corp
Priority to US15/093,588 priority Critical patent/US20160296592A1/en
Assigned to THE GENERAL HOSPITAL CORPORATION reassignment THE GENERAL HOSPITAL CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: REISER, JOCHEN
Publication of US20160296592A1 publication Critical patent/US20160296592A1/en
Priority to US15/979,242 priority patent/US20180344803A1/en
Assigned to THE GENERAL HOSPITAL CORPORATION reassignment THE GENERAL HOSPITAL CORPORATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: REISER, JOCHEN
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/12Cyclic peptides, e.g. bacitracins; Polymyxins; Gramicidins S, C; Tyrocidins A, B or C
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/06Tripeptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/07Tetrapeptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/08Peptides having 5 to 11 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • 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
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/22Anxiolytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2839Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the integrin superfamily
    • C07K16/2848Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the integrin superfamily against integrin beta3-subunit-containing molecules, e.g. CD41, CD51, CD61
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6893Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • G01N2333/70546Integrin superfamily, e.g. VLAs, leuCAM, GPIIb/GPIIIa, LPAM
    • G01N2333/70557Integrin beta3-subunit-containing molecules, e.g. CD41, CD51, CD61
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/34Genitourinary disorders
    • G01N2800/347Renal failures; Glomerular diseases; Tubulointerstitial diseases, e.g. nephritic syndrome, glomerulonephritis; Renovascular diseases, e.g. renal artery occlusion, nephropathy

Definitions

  • This invention relates generally to the treatment of proteinuric diseases by ⁇ v ⁇ 3 integrin inhibitors.
  • Kidney podocytes and their foot processes are a key component of the ultrafiltration system in the glomerulus where they comprise the filtration barrier together with endothelial cells and the glomerular basement membrane (GBM).
  • GBM glomerular basement membrane
  • Podocytes are located within the glomerulus of the kidney where they are attached to the GBM via ⁇ 3 ⁇ 1 integrin 2, 3 , and ⁇ / ⁇ dystroglycan 4 .
  • Podocyte FPs are interconnected by the slit diaphragm (SD), a modified adherens junction 5 .
  • SD slit diaphragm
  • Proteinuric kidney diseases are typically associated with various degrees of podocyte membrane remodeling (FP effacement and/or SD disruption) driven by a rearrangement of the podocyte microfilament system 6 .
  • FP effacement and/or SD disruption Recent work has advanced our understanding of the molecular framework underlying podocyte structure largely through the analysis of hereditary proteinuria syndromes and genetic models 7 .
  • a few studies suggest also mechanisms for the far more common acquired proteinuric diseases 8, 9 .
  • An emerging concept for the regulation of podocyte structure and function is the regulation of the podocyte cytoskeleton by proteases such as cathepsin L 8, 10 .
  • Cathepsin L induction in podocytes is accompanied by an increase in cell motility of cultured podocytes 10 .
  • the increased motility of in vitro podocytes 10, 11 is best translated into FP dynamics in vivo where podocytes remain locally attached to the GBM but may have altered FP dynamics resulting in FP fusion.
  • podocytes In some forms of inflammatory glomerular diseases such as crescentic glomerulonephritis, podocytes have been reported to move out of their microenvironment into areas of crescentic glomerular damage 12 .
  • av ⁇ 3 integrin inhibitor in an amount effective to reduce or eliminate the proteinuric disorder.
  • the proteinuric disorder is a kidney proteinuric disorder.
  • the kidney proteinuric disorder is selected from a list comprising of:
  • kidney proteinuric disorder is diabetic nephropathy.
  • the ab ⁇ 3 integrin inhibitor is a ⁇ v ⁇ 3 monoclonal antibody or a peptide which contains a RGD binding sequence.
  • the av ⁇ 3 integrin inhibitor is a ⁇ v ⁇ 3 monoclonal antibody.
  • the ⁇ v ⁇ 3 monoclonal antibody is anti-CD61.
  • the ab ⁇ 3 integrin inhibitor is a peptide which contains a RGD binding sequence.
  • the peptide which contains a RGD binding sequence is cyclo-[Arg-Gly-Asp-D-Phe-Val].
  • the av ⁇ 3 integrin inhibitors is a compound of the formula:
  • the av ⁇ 3 integrin inhibitors is selected from the compounds described herein.
  • FIG. 1A-D is a panel of immunostaining images and a bar graph showing uPAR is induced in podocytes in proteinuric patients and experimental proteinuric models.
  • FIG. 2A-H is a panel of immunostaining images and bar graphs showing uPAR is required in podocytes for the development of foot process effacement and proteinuria.
  • FIG. 3A-D is a panel of immunostaining images and bar graphs showing uPAR mediates podocyte migration.
  • FIG. 4A-C is a panel of a scheme, immunostaining image and bar graph showing uPAR, ⁇ 3 integrin and vitronectin are important for LPS induced proteinuria.
  • FIG. 5A-E is a panel of immunostaining images and a bar graph showing uPAR activates ⁇ 3 integrin.
  • FIG. 6A-G is A panel of immnunostaining images and bar graphs showing the active uPAR- ⁇ v ⁇ 3 integrin complex is lipid dependent and can be targeted pharmacologically to modify proteinuria.
  • FIG. 7A-C is a panel of a scheme, immunostaining images and a bar graph showing induction of uPAR in cultured podocyte in response to LPS and PAN.
  • FIG. 8A-B is a panel of immunostaining images showing Vitronectin is expressed in the human glomerulus and mainly in podocytes.
  • FIG. 9A-D is a panel of immunostaining images and graphs showing stable knockdown of uPAR mRNA using siRNA.
  • FIG. 10A-B is a panel of immunostaining images showing uPAR interacts with ⁇ 3 integrin.
  • FIG. 11 is a bar graph showing the effect of cyclo-RGDfV on recovery of proteinuria.
  • the present invention is directed to methods of treating proteinuric diseases by the administration of av ⁇ 3 integrin inhibitors.
  • the invention is based at least in part on the discovery that induction of urokinase receptor signaling in podocytes leads to foot process effacement and urinary protein loss via a mechanism including lipid dependent activation of av ⁇ 3 integrin.
  • the invention involves, at least in part, the study of molectdes which are associated with cellular motility such as the urokinase plasminogen activator receptor (uPAR) 15, 16 .
  • uPAR is a glycosylphosphatidylinositol (GPI)-anchored protein that has been recognized as a proteinase receptor but has also be involved in non-proteolytic pathways, mainly through its ability to form complexes with other membrane proteins such as integrins for signal transduction 15 .
  • GPI glycosylphosphatidylinositol
  • uPAR plays important roles during wound healing, inflammation, and stem cell mobilization, as well as in severe pathological conditions such as HIV-1 infection, tumor invasion and metastasis 17 .
  • uPAR Besides uPAR's well-established role in the regulation of pericellular proteolysis, it is also involved in cell adhesion, migration, and proliferation through interactions with proteins present in the extracellular matrix, including vitronectin (Vn) 18 . Most recently, the importance of uPAR-Vn interactions was demonstrated by the direct requirement of uPAR to bind Vn for the sufficient induction of downstream signaling aimed at cell motility and morphology 19 .
  • the invention is based on the experimental observations discussed herein that present a mechanism that involves activation of av ⁇ 3 integrin within lipid rafts 24 .
  • the inhibition of uPAR/av ⁇ 3 integrin function in podocytes ameliorated the course of proteinuria and opens novel avenues for pharmacological interventions.
  • Blockade of av ⁇ 3 integrin reduces podocyte motility in vitro and proteinuria in mice. Mice deficient of uPAR are protected from proteinuria unless a constitutively active ⁇ 3 integrin is expressed.
  • Gene transfer of uPAR into podocytes but not into endothelial cells conferred the ability to develop urinary protein loss.
  • uPAR may be required to activate av ⁇ 3 integrin in podocytes which promotes cell motility and activation of small GTPases cdc42 and Rac1.
  • uPAR is required for podocyte migration and LPS-induced FP effacement and proteinuria in mice via a mechanism that includes lipid-dependent activation of ⁇ v ⁇ 3 integrin.
  • uPAR expression is present in all glomerular cells, yet uPAR expression is not required for normal renal function in any of the cells as the uPAR -/- mice behave normal. Surprising is the requirement of uPAR during the development of podocyte FP effacement and proteinuria which suggests that inducible pathways are required for the remodeling of the filtration barrier. uPAR action for the development of proteinuria stems from its action in the podocyte, which was demonstrated using cell-specific promoter elements.
  • methods for treating proteinuria are provided by inhibiting av ⁇ 3 integrin function by inhibiting uPAR function.
  • uPAR is induced during proteinuria and that uPAR-/- mice are protected from the development of proteinurea.
  • the uPAR inhibitors including small molecules, peptides and antibodies can be used to inhibit av ⁇ 3 integtin function.
  • proteinuria refers to any amount of protein passing through a podocyte, such as a podocyte that has suffered podocyte damage.
  • podocyte damage refers to FP effacement and/or cortical actin rearrangement or any other reversible structural or functional change in podocytes that results in proteinuria.
  • proteinuria refers to the presence of excessive amounts of serum protein in the urine.
  • the excessive amount of serum protein in the urine is greater than 50, 100, 150, or 200 mg of serum protein/day. In a preferred embodiment the amount of serum protein in the urine is greater than 150 mg/day.
  • Proteinuria is a characteristic symptom of either renal (kidney), urinary, pancreatic distress, nephrotic syndromes (for example, proteinuria larger than 3.5 grams per day), eclampsia, toxic lesions of kidneys, and it is frequently a symptom of diabetes mellitus. With severe proteinuria general hypoproteinemia can develop and it results in diminished oncotic pressure (ascites, edema, hydrothorax).
  • a proteinuric disorder refers to, but it is not limited to: Diabetic nephropathy, Nephrotic syndromes (i.e. intrinsic renal failure), Nephritic syndromes, Toxic lesions of kidneys, Glomerular diseases, such as membranous glomerulonephritis, Focal segmental glomerulosclerosis (FSGS), IgA nephropathy (i.e., Berger's disease), IgM nephropathy, Membranoproliferative glomerulonephritis, Membranous nephropathy, Minimal change disease, Hypertensive nephrosclerosis and Interstitial nephritis.
  • FSGS Focal segmental glomerulosclerosis
  • IgA nephropathy i.e., Berger's disease
  • IgM nephropathy i.e., Berger's disease
  • IgM nephropathy i.e., Berger's disease
  • a proteinuric disorder refers to: Pre-eclampsia, Eclampsia, Collagen vascular diseases (e.g., systemic lupus erythematosus), Dehydration, Strenuous exercise, Stress, Benign Orthostatic (postural) proteinuria, Sarcoidosis, Alport's syndrome, Diabetes mellitus, Fabry's disease, Infections (e.g., HIV, syphilis, hepatitis, post-streptococcal infection), Aminoaciduria, Fanconi syndrome, Heavy metal ingestion, Sickle cell disease, Hemoglobinuria, Multiple myeloma, Myoglobinuria, Organ rejection, Ebola hemorrhagic fever and Nail Patella Syndrome.
  • Pre-eclampsia e.g., systemic lupus erythematosus
  • Collagen vascular diseases e.g., systemic lupus erythematosus
  • a proteinuric disorder refers to diabetes, hypertension, kidney disease, minimal change disease, membranous glomerulonephritis, focal segmental glomerulosclerosis, diabetic neuropathy, post-infectious glomerulonephritis, mesangioproliferative glomerulonephritis, HIV-associated nephropathy, IgA-nephropathy, and Cardiovascular disease.
  • Podocyte damage can be caused by many conditions and factors including LPS and purine aminonucleoside (PAN).
  • the instant invention is based in part on the surprising discovery of the functional significance of ⁇ v ⁇ 3 integrin activation as a downstream effector for increased podocyte motility and FP effacement.
  • Integrins belong to the family of heterodimeric class I transmembrane receptors, which play an important role in numerous cell-matrix and cell-cell adhesion processes (Tuckwell et al., 1996, Symp. Soc. Exp. Biol. 47). They can be divided roughly into three classes: the ⁇ 1 integrins, which are receptors for the extracellular matrix, the ⁇ 2 integrins, which can be activated on leucocytes and are triggered during inflammatory processes, and the ⁇ v integrins, which influence the cell response in wound-healing and other pathological processes (Marshall and Hart, 1996, Semin. Cancer Biol. 7, 191).
  • the ⁇ b ⁇ 3 integrin also called that the vitronectin receptor, mediates adhesion to a multiplicity of ligand's-plasma proteins, extracellular matrix proteins, cell surface proteins, of which the majority contain the amino acid sequence Arg-Gly-Asp (RGD), such as, for example, fibronectin or vitronectin.
  • Soluble RGD-containing peptides are capable of inhibiting the interaction of each of these integrins with the corresponding natural ligands.
  • Integrin ⁇ v ⁇ 3 antagonistic action has been shown for a multiplicity of compounds, such as anti- ⁇ v ⁇ 3 monoclonal antibodies, peptides which contain the RGD binding sequence, natural, RGD-containing proteins (e.g.
  • Advantageous ⁇ v ⁇ 3 integrin receptor ligands bind to the integrin ⁇ v ⁇ 3 receptor with an increased affinity by at least a factor of 10, preferably at least a factor of 100.
  • a method for treating a proteinuric disorder comprising administering to a patient in need thereof an ⁇ v ⁇ 3 integrin inhibitors.
  • the ⁇ v ⁇ 3 integrin inhibitors are the compounds described in WO 97/08145 A1, WO 00/48996 A2, WO 06043930 A1, US2002072500, US 2005043344, US 2002045645, US 2002099209, US 2005020505, US 2002072518, US 2002077321, US 2004043994, US 2003069236, US 2003144311.
  • the ⁇ v ⁇ 3 integrin inhibitors are peptidic sulfonamides having the formula (I): R 1 -Arg-X-Asp-Leu-Asp-Ser-Leu-Arg-R 2 , in which R 1 denotes H, acetyl or acyl and R 2 denotes —Oh, OR 3 N 2 ,NHR 3 , N(R 3 ) 2 R 3 denotes alkyl, aralkyl, aryl, Het and X denotes an amino acid, in which A denotes (CH 2 ) n R 4 denotes H, alkyl, aralkyl or aryl, and n denotes 1, 2, 3, 4, 5 or 6, and the amino acid is bonded to the adjacent Arg via a peptide bond of the ⁇ -amino group and to the ⁇ -amino group of the adjacent Asp via a peptide bond of the ⁇ -carboxyl group as described in U.
  • R 1 de
  • the ⁇ v ⁇ 3 integrin inhibitors are fluoro-alkyl-cyclopeptide derivatives as described in WO 2004/011487 A2.
  • the ⁇ v ⁇ 3 integrin inhibitors are peptido-mimetic compounds containing an RGD sequence as described in WO 2005/007654 A1 and U.S. Pat. No. 6,451,972.
  • the ⁇ v ⁇ 3 integrin inhibitors are antagonists of the ⁇ v ⁇ 3 integrin receptor based on a bicyclic structural element are described in WO 9906049, WO 9905107, WO 9814192, WO 9724124, WO 9724122 and WO 9626190.
  • the ⁇ v ⁇ 3 integrin inhibitors are compounds to described in U.S. Publication No 2004/0063934.
  • the ⁇ v ⁇ 3 integrin inhibitors of the invention include isolated peptides and proteins.
  • isolated means separated from its native environment and present in sufficient quantity to permit its identification or use. Isolated, when referring to a protein or polypeptide, means, for example: (i) selectively produced by expression cloning or (ii) purified as by chromatography or electrophoresis. Isolated proteins or polypeptides may be, but need not be, substantially pure. The term “substantially pure” means that the proteins or polypeptides are essentially free of other substances with which they may be found in nature or in vivo systems to an extent practical and appropriate for their intended use.
  • Substantially pure polypeptides may be produced by techniques well known in the art. Because an isolated protein may be admixed with a pharmaceutically acceptable carrier in a pharmaceutical preparation, the protein may comprise only a small percentage by weight of the preparation. The protein is nonetheless isolated in that it has been separated from the substances with which it may be associated in living systems, i.e. isolated from other proteins.
  • ⁇ v ⁇ 3 integrin inhibitors of the invention may be produced using any of the methods and techniques known to those skilled in the art.
  • ⁇ v ⁇ 3 integrin inhibitors can be purified from a source which naturally expresses the protein, can be isolated fman a recombinant host which has been altered to express the desired mutant or fragment thereof, or can be synthesized using protein synthesis techniques known in the art.
  • the skilled artisan can readily adapt a variety of techniques in order to obtain ⁇ b ⁇ 3 integrin inhibitors that are peptides, proteins or fragments thereof.
  • agents that are ⁇ v ⁇ 3 integrin inhibitors include but are not limited to small molecules, and other drugs, including known drugs, that prevent (i.e. reduce or inhibit further increase) integrin activation.
  • Such agents can be identified using routine screening methods.
  • ⁇ v ⁇ 3 integrin inhibitors of the present invention can be identified using the methods and assays described herein.
  • the screening may be a random screen or it may be rationally designed.
  • putative inhibitors are selected at random and assayed for their ability to produce the desired physiological effect.
  • the putative modulators may be assayed for the ability to reduce selectively or specifically the amount or rate of ⁇ v ⁇ 3 integrin activation. Any suitable method or technique known to those skilled in the art may be employed to assay putative inhibitors.
  • Methods for screening using rational design employ the same types of screening methods but begin with a set of compounds that has been designed to specifically maximize function.
  • the ⁇ v ⁇ 3 integrin inhibitors may be selected based, for example on the RGD binding domain. Any of the suitable methods and techniques known to those skilled in the art may be employed for rational selection or design.
  • one skilled in the art can readily adapt currently available procedures to generate pharmaceutical agents capable of binding to a specific peptide sequence of ⁇ b ⁇ 3 integrin or uPAR, precluding their interaction and signal transduction and thereby inhibiting ⁇ v ⁇ 3 integrin activity.
  • ⁇ v ⁇ 3 integrin inhibitors include antibodies and antibody fragments which are capable of binding to ⁇ v ⁇ 3 integrin and consequently acting as a ⁇ v ⁇ 3 integrin inhibitors.
  • the antibodies of the present invention include polyclonal and monoclonal antibodies, as well as antibody fragments and derivatives that contain the relevant antigen binding domain of the antibodies. Such antibodies or antibody fragments are preferably used in the diagnostic and therapeutic embodiments of the present invention. In a preferred embodiment the antibody is anti-CD61 antibody.
  • treating a proteinuric disorder includes preventing the development of proteinurea, reducing or inhibiting proteinurea, slowing the progression, and/or any other desired effect on proteinurea.
  • the ⁇ v ⁇ 3 integrin inhibitors can be administered prior to the onset of proteinurea, following the onset of proteinurea, or as part of any therapeutic regimen, for example, including cancer medicaments.
  • the methods of the invention are intended to embrace the use of more than one other medicament along with the ⁇ v ⁇ 3 integrin inhibitor.
  • the ⁇ v ⁇ 3 integrin inhibitor may be administered with both a chemotherapeutic agent, anti-diabetic agent or an immunotherapeutic agent.
  • the invention also encompasses diagnostic assays for determining the presence of a disorder characterized by proteinuria in a subject.
  • This aspect of the invention is based, at least in part, on the discovery that ⁇ v ⁇ 3 integrin activation is reduced in damaged or proteinuric podocytes.
  • an amount of ⁇ v ⁇ 3 integrin activity in a podocyte cell is determined. That amount is compared to a pre-determined threshold or to a control level.
  • a disorder characterized by proteinuria is determined when the amount of ⁇ v ⁇ 3 integrin activity is below the pre-determined threshold.
  • pre-determined threshold or a control level refers to ⁇ v ⁇ 3 integrin activity levels in normal, healthy podocytes, i.e.
  • podocytes not affected by podocyte damage or proteinuria may be within a biological sample.
  • the biological sample may be, for instance, a biopsy sample of proteinuric tissue.
  • activity refers to expression and well as biochemical activity as described herein.
  • ⁇ v ⁇ 3 integrin in podocyte cells can be readily carried out by standard immunostaining or immunocytometric methods, readily known by persons of ordinary skill in the art.
  • immunostaining refers to a technique of applying coloured or fluorescent dyes to tissues in preparation for microscopic examination. The assay may be performed using immunogold election microscopy.
  • the diagnostic assays are performed on cells and/or tissue samples wherein morphological changes of the actin-cytoskeleton can not be readily detected by any other immunocytometric methods.
  • one such disorder would be minimal change disease.
  • minimal change disease comes from the notion that morphological podocyte changes are only visible by electron microscopy. Detection of proteinuria in patients with minimal change disease by immunocytometric methods would be advantageous because it provide ease and speed of detection.
  • the immunocytometric methods may be performed using labeled antibodies.
  • An antibody is said to be “detectably labeled” if the antibody, or fragment thereof, is attached to a molecule which is capable of identification, visualization, or localization using known methods.
  • Suitable detectable labels include radioisotopic labels, enzyme labels, non-radioactive isotopic labels, fluorescent labels, affinity labels, chemiluminescent labels and nuclear magnetic resonance contrast agents.
  • suitable enzyme labels include, but are not limited to, luciferase, malate dehydrogenase, staphyloccal nuclease, delta-5-steroid isomerase, yeast-alcohol dehydrogenase, alpha-glycerol phosphate dehydrogenase, triose phosphate isomerase, horseradish peroxidase, alkaline phosphatase, asparaginase, glucose, oxidase, beta-galactosidase, ribonuclease, urease, catalase, glucose-6-phosphate dehydrogenase, glucoamylase, and acetylcholine esterase.
  • radioisotopic labels include, but are not limited to, 3 H, 111 In, 125 I, 131 I, 32 P, 35 S, 14 C, 51 Cr, 57 To, 58 Co, 59 Fe, 75 Se, 152 Eu, 90 Y, 67 Cu, 217 Ci, 211 At, 212 Pb, 47 Sc, 109 Pd, etc.
  • 111 In is a preferred isotope where in vivo imaging is used since its avoids the problem of dehalogenation of the 125 I or 131 I-labeled monoclonal antibody by the liver.
  • this radionucleotide has a more favorable gamma emission energy for imaging (Perkins et al., Eur. J. Nucl.
  • non-radioactive isotopic labels include, but are not limited to, 157 Gd, 55 Mn, 162 Dy, 52 Tr, and 56 Fe.
  • fluorescent labels include, but are not limited to, an 152 Eu label, a fluorescent protein (including green fluorescent protein (GFP), enhanced green fluorescent protein (EGFP), enhanced yellow fluorescent protein (YFP) and enhanced cyan fluorescent protein (ECFP),), a fluorescein label, an isothiocyanate label, a rhodamine label, a phycoerythrin label, a phycocyanin label, an allophycocyanin label, an o-phthaldehyde label, and a fluorescamine label.
  • GFP green fluorescent protein
  • EGFP enhanced green fluorescent protein
  • YFP enhanced yellow fluorescent protein
  • ECFP enhanced cyan fluorescent protein
  • chemiluminescent labels include a luminal label, an isoluminal label, an aromatic acridinium ester label, an imidazole label, an acridinium salt label, an oxalate ester label, a luciferin label, a luciferase label, and an aequorin label.
  • nuclear magnetic resonance contrasting agents include paramagnetic heavy metal nuclei such as Gd, Mn, and Fe.
  • the coupling of one or more molecules to antibodies is envisioned to include many chemical mechanisms, for instance covalent binding, affinity binding, intercalation, coordinate binding, and complexation.
  • the covalent binding can be achieved either by direct condensation of existing side chains or by the incorporation of external bridging molecules.
  • Many bivalent or polyvalent agents are useful in coupling protein molecules to other proteins, peptides or amine functions, etc.
  • the literature is replete with coupling agents such as carbodiimides, diisocyanate, glutaraldehyde, diazobenzenes, and hexamethylene diamines. This list is not intended to be exhaustive of the various coupling agents known in the art but, rather, is exemplary of the more common coupling agents.
  • the ⁇ v ⁇ 3 integrin inhibitors are administered in pharmaceutically acceptable preparations.
  • the pharmaceutical preparations of the invention are applied in pharmaceutically-acceptable amounts and in pharmaceutically-acceptable compositions.
  • pharmaceutically acceptable means a non-toxic material that does not interfere with the effectiveness of the biological activity of the active ingredients.
  • Such preparations may routinely contain salts, buffering agents, preservatives, compatible carriers, and optionally other therapeutic agents.
  • the salts should be pharmaceutically acceptable, but non-pharmaceutically acceptable salts may conveniently be used to prepare pharmaceutically-aceeptable salts thereof and are not excluded from the scope of the invention.
  • “pharmaceutically acceptable carrier” or “physiologically acceptable carrier” includes any and all salts, solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible.
  • the carrier is suitable for intravenous, intramuscular, subcutaneous, parenteral, per os, spinal or epidermal administration (e.g., by injection or infusion).
  • the active compound i.e., ⁇ v ⁇ 3 integrin inhibitor may be coated in a material to protect the compound from the action of acids and other natural conditions that may inactivate the compound.
  • the carrier is sterile.
  • a salt retains the desired biological activity of the parent compound and does not impart any undesired toxicological effects (see e.g., Berge. S. M., et al. (1977) J. Pharm. Sci, 66: 1-19).
  • Examples of such salts include acid addition salts and base addition salts.
  • Acid addition salts include those derived from nontoxic inorganic acids, such as hydrochloric, nitric, phosphoric, sulfuric, hydrobromic, hydroiodic, phosphorous and the like, as well as from nontoxic organic acids such as aliphatic mono- and dicarboxylic acids, phenyl substituted alkanoic acids, hydroxy alkanoic acids, aromatic acids, aliphatic and aromatic sulfouic acids and the like.
  • Base addition salts include those derived from alkaline earth metals, such as sodium, potassium, magnesium, calcium and the like, as well as from nontoxic organic amines, such as N,N′-dibenzylethylenediamine N-methylglocamine, chioroprocaine, choline, diethanolamine, ethylenediamine, procaine and the like.
  • the pharmaceutical compositions may contain suitable buffering agent, including: acetic acid in a salt; citric acid in a salt; boric acid in a salt; and phosphoric acid in a salt.
  • suitable buffering agent including: acetic acid in a salt; citric acid in a salt; boric acid in a salt; and phosphoric acid in a salt.
  • suitable preservatives such as: benzalkonium chloride; chlorobutanol; parabens and thimerosal.
  • compositions may conveniently be presented in unit dosage form and may be prepared by any of the methods well-known in the art of pharmacy. All methods include the step of bringing the active agent into association with a carrier which constitutes one or more accessory ingredients. In general, the compositions are prepared by uniformly and intimately bringing the active compound into association with a liquid carrier, a finely divided solid carrier, or both, and then, if necessary, shaping the product.
  • compositions suitable for parenteral administration conveniently comprise a sterile aqueous or non-aqueous preparation of the compounds, which is preferably isotonic with the blood of the recipient.
  • This preparation may be formulated according to known methods using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation also may be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example, as a solution in 1,3-butane diol.
  • acceptable vehicles and solvents that may be employed are water. Ringer's solution, and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono-or di-glycerides.
  • fatty acids such as oleic acid may be used in the preparation of injectables.
  • Carrier formulations suitable for oral, subcutaneous, intravenous, intramuscular, etc. administration can be found in Remington's Pharmaceutical Sciences , Mack Publishing Co., Easton, Pa.
  • the active compounds can be prepared with carriers that will protect the compound against rapid release, such as a controlled release fornaulation, including implants, transdermal patches, and microencapsulated delivery systems.
  • Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Many methods for the preparation of such formulations are patented or generally known to those skilled in the art. See, e.g., Sustained and Controlled Release Drug Delivery Systems , J. R. Robinson, ed., Marcel Dekker, Inc., New York, 1978.
  • compositions comprising one or more ⁇ v ⁇ 3 integrin inhibitors.
  • These pharmaceutical compositions may be administered orally, rectally, parenterally, intrathecally, intracisternally, intravaginally, intraperitoneally, topically (as by powders, ointments, drops or transdermal patch), bucally, or as an oral or nasal spray.
  • parenteral refers to modes of administration which include intravenous, intramuscular, intrathecal, intraperitoneal, intrasternal, subcutaneous and intraarticular injection and infusion.
  • a particular mode of administration can be made empirically based upon considerations such as the particular disease state being treated; the type and degree of the response to be achieved; the specific agent or composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration and rate of excretion of the agent or composition; the duration of the treatment; drugs used in combination or coincidental with the specific composition; and like factors well known in the medical arts.
  • compositions of the present invention for parenteral injection may comprise pharmaceutically acceptable sterile aqueous or nonaqueous solutions, dispersions, suspensions or emulsions as well as sterile powders for reconstitution into sterile injectable solutions or dispersions just prior to use.
  • suitable aqueous and nonaqueous carriurs, diluents, solvents or vehicles include, but are not limited to, water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), carboxymethylceuulose and suitable mixtures thereof, vegetable oils (such as olive oil), and injectable organic esters such as ethyl oleate.
  • Proper fluidity can be maintained, for example, by the use of coating materials such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.
  • compositions of the present invention may also contain preservatives, wetting agents, emulsifying agents, and dispersing agents. Prevention of the action of microorganisms may be ensured by the inclusion of various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents such as sugars, sodium chloride, and the like. Prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents which delay absorption such as aluminum monostearate and gelatin.
  • the absorption from subcutaneous or intramuscular injection In some cases, in order to prolong the effect of the therapeutic agent, it is desirable to slow the absorption from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material with poor water solubility. The rate of absorption of the drug then depends upon its rate of dissolution which, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle.
  • Injectable depot forms are made by forming microencapsuled matrices of the drug in biodegradable polymers such as polylactide-polyglycolide. Depending upon the ratio of drug to polymer and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions which are compatible with body tissues.
  • Solid dosage forms for oral administration include, but are not limited to, capsules, tablets, pills, powders, and granules.
  • the active compounds are preferably mixed with at least one pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such as quaternary ammonium compounds, g) wetting agents such as, for example, cetyl
  • compositions of a similar type may also be employed as fillers in soft and hard filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like.
  • the solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner.
  • coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner.
  • opacifying agents include polymeric substances and waxes.
  • the active ⁇ v ⁇ 3 integrin inhibitors can also be in micro-encapsulated form, if appropriate, with one or more of the above-mentioned excipients.
  • Liquid dosage forms for oral administration include, but are not limited to, pharmaceutically acceptable emulsions, solutions, suspensions, syrups and elixirs.
  • the liquid dosage forms may contain inert diluents so commonly used in the art such as, for example, water or other solvents, solubilizing, agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethyl formamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
  • inert diluents so commonly used in the art such as, for example, water or other solvents, solubil
  • the oral compositions may also contain adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
  • adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
  • Suspensions in addition to the active compounds, may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar, and tragacanth, and mixtures thereof.
  • suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar, and tragacanth, and mixtures thereof.
  • the ⁇ v ⁇ 3 integrin inhibitor is administered in the form of liposomes.
  • liposomes are generally derived from phospholipids or other lipid substances. Liposomes are formed by mono- or multi-lamellar hydrated liquid crystals that are dispersed in an aqueous medium. Any non-toxic, physiologically acceptable and metabolizable lipid capable of forming liposomes can be used.
  • the present compositions in liposome form can contain, in addition to the ⁇ v ⁇ 3 integrin inhibitor, stabilizers, preservatives, excipients, and the like.
  • Preferred lipids are phospholipids and phosphatidyl cholines (lecithins), both natural and synthetic. Methods to form liposomes am known in the art. See, e.g., Prescott, ed., METHODS IN CELL BIOLOGY, Volume XIV, Academic Press, New York, N.Y. (1976), p. 33 et seq.
  • therapeutic agents used in the methods of the invention can be determined empirically and may be employed in pure form or, where such forms exist, in pharmaceutically acceptable salt, ester or prodrug form.
  • the therapeutic agents may be administered in compositions in combination with one or more pharmaceutically acceptable excipients. It will be understood that, when administered to a human patient, the total daily usage of the agents and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment.
  • the specific therapeutically effective dose level for any particular patient will depend upon a variety of factors including the type and degree of the response to be achieved; the specific agent or composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the agent or composition; the duration of the treatment; drugs (such as a chemotherapeutic agent) used in combination or coincidental with the specific composition; and like factors well known in the medical arts.
  • dosage determination is well known in the art. For example, satisfactory results are obtained by oral administration of therapeutic dosages on the order of from 0.05 to 10 mg/kg/day, preferably 0.1 to 7.5 mg/kg/day, more preferably 0.1 to 2 mg/kg/day, administered once or, in divided doses, 2 to 4 times per day. On administration parenterally, for example by i.v. drip or infusion, dosages on the order of from 0.01 to 5 mg/kg/day, preferably 0.05 to 1.0 mg/kg/day and more preferably 0.1 to 1.0 mg/kg/day can be used.
  • Suitable daily dosages for patients are thus on the order of from 2.5 to 500 mg p.o., preferably 5 to 250 mg per oral (p.o.), more preferably 5 to 100 mg p.o., or on the order of from 0.5 to 250 mg i.v., preferably 2.5 to 125 mg i.v. and more preferably 2.5 to 50 mg i.v.
  • the administration of the agents of the present invention may be for either prophylactic or therapeutic purpose.
  • the agent When provided prophylactically, the agent is provided in advance of any damage i.e., proteinuria or podocyte damage.
  • the prophylactic administration of the agent serves to prevent or reduce the rate of onset of symptoms.
  • the agent When provided therapeutically, the agent is provided at (or after) the onset of the appearance of symptoms of actual disease.
  • the therapeutic administration of the agent serves to reduce the severity and duration of proteinuria.
  • compositions of the invention are administered in effective amounts.
  • An “effective amount” is that amount of any of the compositions provided herein that alone, or together with further doses, produces the desired response, e.g. reduces or eliminates proteinuria. This may involve only slowing the progression of the disease temporarily, although more preferably, it involves halting the progression of the disease permanently. This can be monitored by routine methods.
  • the desired response to treatment of the disease or condition also can be delaying the onset or even preventing the onset of the disease or condition.
  • An amount that is effective can be the amount of a ⁇ v ⁇ 3 integrin inhibitor alone which produces the desired therapeutic endpoint.
  • An amount that is effective is also the amount of a ⁇ v ⁇ 3 integrin inhibitor in combination with another agent that produces the desired result.
  • Such amounts will depend, of course, on the particular condition being treated, the severity of the condition, the individual patient parameters including age, physical condition, size and weight, the duration of the treatment, the nature of concurrent therapy (if any), the specific route of administration and like factors within the knowledge and expertise of the health practitioner. These factors are well known to those of ordinary skill in the art and can be addressed with no more than routine experimentation. It is generally preferred that a maximum dose of the individual components or combinations thereof be used, that is, the highest safe dose according to sound medical judgment. It will be understood by those of ordinary skill in the art, however, that a patient may insist upon a lower dose or tolerable dose for medical reasons, psychological reasons or for virtually any other reasons.
  • the doses of ⁇ v ⁇ 3 integrin inhibitors administered to a subject can be chosen in accordance with different parameters, in particular in accordance with the mode of administration used and the state of the subject. Other factors include the desired period of treatment. In the event that a response in a subject is insufficient at the initial doses applied, higher doses (or effectively higher doses by a different, more localized delivery route) may be employed to the extent that patient tolerance permits.
  • the dose can be delivered continuously, such as by continuous pump, or at periodic intervals. Desired time internals of multiple doses of a particular composition can be determined without undue experimentation by one skilled in the art.
  • Other protocols for the administration of the compositions provided will be known to one of ordinary skill in the art, in which the dose amount, schedule of administration, site of administration, mode of administration and the like vary from the foregoing.
  • ⁇ v ⁇ 3 integrin inhibitor compositions to mammals other than humans, e.g. for testing purposes or veterinary therapeutic purposes, is carried out under substantially the same conditions as described above.
  • compositions of the present invention have in vitro and in vivo diagnostic and therapeutic utilities.
  • these molecules can be administered to cells in culture, e.g. in vitro or ex vivo, or in a subject, e.g., in vivo, to treat, prevent or diagnose a variety of disorders.
  • the term “subject” is used interchangeably with the term “patient” and is intended to include humans and non-human animals including but not limited to a dog, cat, horse, cow pig, sheep, goat, or primate, e.g., monkey.
  • Preferred patients include a human patient having a proteinuric disorder, including disorders characterized by proteinuria as described herein.
  • a patient in need thereof refers to any patient that is affected with a disorder characterized by proteinuria.
  • a patient in need thereof refers to any patient that may have, or is at risk of having a disorder characterized by proteinuria.
  • a patient in need thereof is a patient that has, may have or is at risk at having cancer, precancer, refractory cancer or metastatic cancer.
  • a patient in need thereof is a patient that has, may have, or is at risk of having a cognitive disorder, such as Alzheimer's disease or dementia.
  • compositions provided of the present invention can be used in conjunction with other therapeutic treatment modalities.
  • Such other treatments include surgery, radiation, cryosurgery, thermotherapy, hormone treatment, chemotherapy, vaccines, and immunotherapies.
  • the invention also relates in some aspects to the identification and testing of candidate agents and molecules that may be ⁇ b ⁇ 3 integrin inhibitors. These molecules are referred to as putative ⁇ v ⁇ 3 integrin inhibitors.
  • putative ⁇ v ⁇ 3 integrin inhibitors can be screened for activity using the same type of assays as described herein (e.g., the assays described in the Examples section). Using such assays, additional ⁇ v ⁇ 3 integrin inhibitors can be can be identified.
  • the invention further provides efficient methods of identitying pharmacological agents or lead compounds as ⁇ v ⁇ 3 integrin inhibitors.
  • the screening methods involve assaying for compounds which inhibit the level of ⁇ v ⁇ 3 integrin activity.
  • the screening methods may measure the level of binding between the molecules directly, such as by using the methods employed in the Examples.
  • screening methods may be utilized that measure a secondary effect of the ⁇ v ⁇ 3 integrin inhibitors, for example the level of proteinuria in a cell or tissue sample.
  • assays for pharmacological agents can be used in accordance with this aspect of the invention, including, labeled in vitro protein-protein binding assays, electrophoretic mobility shift assays, immunoassays, cell-based assays such as two or three-hybrid screens, expression assays, etc.
  • the assay mixture comprises a candidate pharmacological agent.
  • a plurality of assay mixtures are run in parallel with different agent concentrations to obtain a different response to the various concentrations.
  • one of these concentrations serves as a negative control, i.e., at zero concentration of agent or at a concentration of agent below the limits of assay detection.
  • Putative inhibitors useful in accordance with the invention encompass numerous chemical classes, although typically they are organic compounds.
  • the putative modulators are small organic compounds, i.e., those having a molecular weight of more than 50 yet less than about 2500, preferably less than about 1000 and, more preferably, less than about 500.
  • Putative ⁇ v ⁇ 3 integrin inhibitors comprise functional chemical groups necessary for structural interactions with proteins and/or nucleic acid molecules, and typically include at least an amine, carbonyl, hydroxyl or carboxyl group, preferably at least two of the functional chemical groups and more preferably at least three of the functional chemical groups.
  • the putative modulators can comprise cyclic carbon or heterocyclic structure and/or aromatic or polyaromatic structures substituted with one or more of the above-identified functional groups.
  • Putative modulators also can be biomolecules such as peptides, saccharides, fatty acids, sterols, isoprenoids, purines, pyrimidines, derivatives or structural analogs of the above, or combinations thereof and the like.
  • the putative modulator is a nucleic acid molecule
  • the agent typically is a DNA or RNA molecule, although modified nucleic acid molecules as defined herein are also contemplated.
  • cell-based assays as described herein can be performed using cell samples smiler cultured cells. Biopsy cells and tissues as well as cell lines grown in culture are useful in the methods of the invention.
  • Putative ⁇ v ⁇ 3 integrin inhibitors are obtained from a wide variety of sources including libraries of synthetic or natural compounds. For example, numerous means are available for random and directed synthesis of a wide variety of organic compounds and biomolecules, including expression of randomized oligonucleotides, synthetic organic combinatorial libraries, phage display libraries of random peptides, and the like.
  • libraries of natural compounds in the form of bacterial, fungal, plant and animal extracts are available or readily produced.
  • natural and synthetically produced libraries and compounds can be readily be modified through conventional chemical, physical, and biochemical means.
  • known pharmacological agents may be subjected to directed or random chemical modifications such as acylation, alkylation, esterification, amidification, etc. to produce structural analogs of the agents.
  • reagents such as salts, buffers, neutral proteins (e.g., albumin), detergents, etc. which may be used to facilitate optimal protein-protein and/or protein-nucleic acid binding. Such a reagent may also reduce non-specific or background interactions of the reaction components. Other reagents that improve the efficiency of the assay such as antimicrobial agents, and the like may also be used.
  • the prerequisite for producing intact native polypeptides using E. coli is the use of a strong, regulatable promoter and an effective ribosome binding site.
  • Promoters which may be used for this purpose include the temperature sensitive bacteriophage ⁇ pL -promoter, the tac-promoter inducible with IPTG or the T7-promoter.
  • plasmids with suitable promoter structures and efficient ribosome binding sites have been described, such as for example pKC30 ( ⁇ pL ; Shimatake and Rosenberg, Nature 292:128 (1981), pKK173-3 (tac, Amann and Brosius, Gene 40:183 (1985)) or pET-3 (T7-promoter (Studier and Moffat, J. Mol. Biol. 189:113 (1986)).
  • E. coil strains which are specifically tailored to a particular expression vector are known to those skilled in the art (Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory Press (1989)).
  • the experimental performance of the cloning experiments, the expression of the polypeptides in E. coli and the working up and purification of the polypeptides are known and are described, for example, in Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory Press (1989).
  • eukaryotic microorganisms such as yeast may also be used.
  • the plasmid YRp7 (Stinchcomb et al. Nature 282:39 (1979); Kingsman et al., Gene 7:141 (1979); Tschumper et al., Gene 10:157 (1980)) and the plasmid YEp13 (Bwach et al., Gene 8:121-133 (1979)) are used, for example.
  • the plasmid YRp7 contains the TRP1-gene which provides a selection marker for a yeast mutant (e.g., ATCC No. 44076) which is incapable of growing in tryptophan-free medium.
  • TRP1 defect as a characteristic of the yeast strain used then constitutes an effective aid to detecting transformation when cultivation is carried out without tryptophan.
  • plasmid YEp13 which contains the yeast gene LEU-2, which can be used to complete a LEU-2-minus mutant.
  • yeast hybrid vectors also contain a replication start and a marker gene for a bacterial host, particularly E. coli , so that the construction and cloning of the hybrid vectors and their precursors can be carried out in a bacterial host.
  • Other expression control sequences suitable for expression in yeast include, for example, those of PHO3- or PHO5-gene.
  • yeast vectors contain the 5′-flanking region of the genes of ADH I (Ammerer, Methods of Enzymology 101:192-210 (1983)), 3-phosphoglycerate kinase (Hitzeman et al., J Biol. Chem.
  • glycolytic enzymes such as enolase, glycerinaldehyde-3-phosphate-dehydrogenase, hexokinase, pyruvate-decarboxylase, phosphofructokinase, glucose-6-phosphate isomerase, phosphoglucose-isomerase and glucokinase.
  • the termination sequences associated with these genes may also be inserted in the expression vector at the 3′-end of the sequence to be expressed, in order to enable polyadenylation and termination of the mRNA.
  • any vector which contains a yeast-compatible promoter and origin replication and tennination sequences is suitable.
  • hybrid vectors which contain sequences homologous to the yeast 2 ⁇ plasmid DNA may also be used. Such hybrid vectors are incorporated by recombination within the cells of existing 2 ⁇ -plasmids or replicate autonomously.
  • yeasts In addition to yeasts, other eukaryotic systems may, of course, be used to express the polypeptides according to the invention. Since post-translational modifications such as disulphide bridge formation, glycosylation, phosphorylation and/or oligomerization are frequently necessary for the expression of biologically active eukaryotic proteins by means of recombinant DNA, it may be desirable to express the DNA according to the invention not only in mammalian cell lines but also insect cell lines.
  • Functional prerequisites of the corresponding vector systems comprise, in particular, suitable promoter, termination and polyadenylation signals as well as elements which make it possible to carry out replication and selection in mammalian cell lines.
  • particularly suitable promoters are podocyte specific promoters.
  • a podocyte specific promoter is one that is expressed exclusively in podocytes, such as the podocin promoter.
  • Vectors derived fnom viral systems such as SV40, Epstein-Barr-virus etc., include, for example, pTK2, pSV2-dhfv, pRSV-neo, pKO-neo, pHyg, p205, pHEBo, etc. (Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Press, N.Y. (1989)).
  • corresponding transformed cells may be obtained with the aid of selectable markers (thymidine-kinase, dihydrofolate-reductase, green fluorescent protein, etc.) and the corresponding polypeptides are isolated after expression.
  • selectable markers thymidine-kinase, dihydrofolate-reductase, green fluorescent protein, etc.
  • the host cells suitable for the vectors are known, as are the techniques for transfomiation (nricro-injection, electroporation, calcium phosphate method, etc.) as described, for example, in Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Press, N.Y. (1989).
  • the selected vector may be cut, for example, with a restriction endonuclease and, optionally after modification of the linearized vector thus formed, an expression control sequence equipped with corresponding restriction ends is inserted.
  • the expression control sequence contains the recognition sequence of a restriction endonuclease, so that the vector already containing the expression control sequence is digested with the said restriction enzyme and the DNA molecule according, to the invention, provided with ends which fit, can be inserted.
  • the invention also relates to processes for preparing the vectors described herein, particularly expression vectors.
  • These vectors are characterized in that a DNA provided with corresponding ends and coding for a functional derivative or a fragment of the protein is inserted into a vector DNA cut with restriction endonucleases and containing the expression control sequences described by way of example, in such a way that the expression control sequences regulate the expression of the DNA inserted.
  • the peptides and antibody agents of the present invention which are obtained by the expression of recombinant DNA or from the native receptor molecule may, of course, also be derivatized by chemical or enzymatic processes.
  • kits comprising the compositions of the invention and instructions for use.
  • the kits can further contain at least one additional reagent, such as a chemotherapeutic agent.
  • a kit may comprise a carrier being compartmentalized to receive in close confinement therein one or more container means or series of container means such as test tubes, vials, flasks, bottles, syringes, or the like.
  • a first of said container means or series of container means may contain one or more ⁇ b ⁇ 3 integrin inhibitors or recombinant vectors for the expression thereof.
  • a second container means or series of container means may contain a second therapeutic, such as, cytotoxic drug or ⁇ v ⁇ 3 integrin antibodies (or fragment thereof).
  • Kits for use in the therapeutic methods of the invention containing the ⁇ v ⁇ 3 integrin inhibitors conjugated to other compounds or substances can be prepared.
  • the components of the kits can be packaged either in aqueous medium or in lyophilized form.
  • the ⁇ v ⁇ 3 integrin inhibitors or fragments thereof are used in the kits in the form of conjugates in which a label or a therapeutic moiety is attached, such as a radioactive metal ion or a therapeutic drug moiety
  • the components of such conjugates can be supplied either in fully conjugated form, in the form of intermediates or as separate moieties to be conjugated by the user of the kit.
  • uPAR a treatment known to cause FP effacement and proteinuria 9 . It is shown that uPAR is dispensable for normal renal filtration but required in podocytes but not in endothelial cells for loss of renal permselectivity seen in human and rodent kidney diseases.
  • LPS lipopolysaccharide
  • the proteinuric signal originating from uPAR in podocytes is independent of urokinase and is required for podocyte FP effacement and LPS-induced proteinuria.
  • FIG. 1A we first tested if uPAR protein is expressed in human glomeruli ( FIG. 1A ) and found expression in many glomerular cells including podocytes which are identified by synaptopoding labeling 25 .
  • FIG. 1B we investigated the expression of uPAR mRNA in human proteinuric renal diseases ( FIG. 1B ).
  • FIG. 1B We performed quantitative real-time PCR using isolated glomeruli from human kidney biopsies 26 .
  • FSGS focal segmental glomerulosclerosis
  • DN diabetic nephropathy
  • FIG. 1B We found a low level of glomerular uPAR mRNA expression in patients without glomerular disease ( FIG. 1B ). In contrast, patients with biopsy-proven FSGS had a significant increase in glomerular uPAR expression (FSGS 0.35 ⁇ 0.17 vs con 0.14 ⁇ 0.02, **p ⁇ 0.05), ( FIG. 1B ). An even stronger induction of glomerular uPAR mRNA expression (0.69 ⁇ 0.21 vs con 0.14 ⁇ 0.02, *p ⁇ 0.012) was found in patients with diabetic nephropathy ( FIG. 1B ).
  • uPAR was partially localized in podocytes as indicated by double immunofluorescent staining with synaptopodin 25 .
  • expression of uPAR protein in all three proteinuria models was significantly increased in glomerular cells including podocytes as demonstrated by the increased yellow staining pattern resulting from overlap with synaptopodin ( FIG. 1C ).
  • uPAR was preferentially found in nephritic areas of the glomerulus in a more segmental distribution. Such a distribution has been recently reported in marine glomerulonephritis where podocytes can populate cellular crescents 12 .
  • uPAR localization in normal and diabetic animals ( FIG. 1D ), since uPAR mRNA induction is strongest during diabetic nephropathy in humans ( FIG. 1B ).
  • FIG. 1D we carried out semiquantitative immunogold analysis of uPAR and transmission electron microscopy in glomerular walls of normal and 3 months as well as 12 months old diabetic rats in which hyperglycemia was induced by streptozotocin injection 29 .
  • uPAR expression was found in all cell types of the glomerulus (Table 1) including podocytes ( FIG. 1D ).
  • uPAR protein 14 h after uPAR gene delivery, uPAR protein was found in the liver, and at lower expression levels in glomerular extracts ( FIG. 2C ). The restoration of uPAR did change the morphology of podocyte FPs ( FIG. 2D , con).
  • uPAR -/- mice reconstituted with uPAR-cDNA develop heavy proteinuria but like wild type mice only after LPS injection ( FIG. 2E ).
  • the degree of proteinuria was comparable in LPS treated wild type and uPAR-cDNA reconstituted uPAR -/- mice.
  • these data strongly suggest that uPAR is required for the development of LPS-indueed proteinuria in mice.
  • Podocyte PP effacement may represent a motile event resulting in spreading of podocyte FPs on the GBM.
  • uPAR-siRNA was confirmed by both semi-quantitative RT-PCR (FIG. 9 A) and Western blotting ( FIG. 9B ).
  • uPAR Activates ⁇ b ⁇ 3 Integrin in Podocytes
  • uPAR is a GPI-anchored protein without a cytoplasmic tail
  • signal transduction from uPAR involves lateral interactions with membrane proteins such as integrins 16 .
  • membrane proteins such as integrins 16 .
  • Madsen et al. described that uPAR induced cell adhesion and migration required Vn binding which can occur independently of uPAR interactions with integrins 19 .
  • Podocyte motility on Vn is enhanced in a uPAR-dependent fashion ( FIG. 3 ) and Vn is induced in glomeruli during proteinuria ( FIG. 8B ).
  • an uPAR-Vn complex or an uPAR-Vn-integrin complex facilitates podocyte motility and promotes FP effacement in response to LPS.
  • Integrins can be in an inactive or active conformation ( FIG. 4A ). The latter is stimulated by the association with interacting agonists such as domain 3 of uPAR which is important for a5b1 integrin interaction 37, 38 .
  • Degryse and colleagues recently identified an integrin-interacting sequence in domain 2 of uPAR, that acts as a new chemotactic epitope activating ⁇ b ⁇ v3-dependent signaling pathways 39 .
  • Vn receptor ⁇ b ⁇ 3 integrin 36 Given our findings of uPAR and Vn in the glomerulus, we were particulary interested in the Vn receptor ⁇ b ⁇ 3 integrin 36 . Indeed, the localization of ⁇ v ⁇ 3 integrin in podocyte FPs ( FIG. 4B ) 31 and the distribution of uPAR ( FIG. 4B ) was similar. In addition, uPAR interacts with ⁇ 3 integrin ( FIG. 10A ). Thus, we hypothesized that ⁇ v ⁇ 3 integrin may provide a functional link between uPAR, podocyte migration and proteinuria development. The genetic deletion of ⁇ 3 integrin or of the ⁇ v ⁇ 3 integrin ligand Vn resulted in protection from proteinuria when challenged with LPS ( FIG. 4C ).
  • uPAR -/- , ⁇ 3 integrin -/- and Vn -/- mice huge no overt renal phenotypes under normal conditions suggesting that uPAR signaling in podocytes is not required for normal glomerular filtration. On the other hand, all these molecules are required for the development of urinary protein loss. Based on this, we reasoned that changes in activation of ⁇ v ⁇ 3 integrin under disease conditions may be a cause for the increased podocyte motility and FP effacement after the administration of LPS. To explore this idea, we next studied the expression of total and active ⁇ 3 integrin in kidney sections from wild type and uPAR -/- mice.
  • Total podocyte ⁇ 3 integrin expression was visualized by double labeling of ⁇ 3 integrin with the podocyte marker synaptopodin wild type and uPAR -/- mice before and after injection of LPS ( FIG. 5A ).
  • the expression of ⁇ 3 integrin in podocytes was unchanged in wild type and uPAR -/- mice before and after LPS administration ( FIG. 5A ).
  • uPAR and ⁇ 3 integrin can associate with each other.
  • Plasma membrane lipid rafts help to compartmentalize signal transduction events within different regions 24 and uPAR as a GPI-anchored protein is known to be found in lipid rich membrane compartments 16 .
  • uPAR and ⁇ 3 integrin associate together within lipid rafts in podocytes. We therefore performed sucrose density gradient assays of whole cellular extracts of cultured podocytes before and after LPS administration ( FIG. 6A ).
  • uPAR and ⁇ 3 integrin are mainly associated with the non-raft fractions in control podocytes.
  • both uPAR and ⁇ 3 integrin were more enriched within the lipid raft fraction ( FIG. 6A ).
  • the functional association of uPAR and ⁇ 3 integrin within lipid rafts was further confirmed by the observation that disruption of lipid rafts using Methyl- ⁇ -cyclodextrin (MBCD) 47 abrogated the activation of ⁇ 3 integrin in response to LPS in cultured podocytes as revealed by reduced AP5 staining ( FIG. 6B ).
  • MCD Methyl- ⁇ -cyclodextrin
  • ⁇ v ⁇ 3 integrin is an important downstream signal that mediates the uPAR-induced cellular events in podocytes leading to proteinuria
  • expression of a constitutively active ⁇ 3 integain should be sufficient to induce proteinuria even in the absence of uPAR. Therefore, we utilized a ⁇ 3 integrin cDNA which encodes a protein lacking aminoacids 616 to 690 of the carboxy-terminal regions of the ⁇ 3 ectodomain. This mutation confers constitutive activity to the ⁇ 3 protein and is expressed at the cell surface 48 .
  • anti-active ⁇ 3 integrin AP5
  • GTI anti-active ⁇ 3 integrin
  • Fab fragment antigen binding
  • BD Pharmagen anti-CD61 blocking antibody
  • anti-CD31 ER-MP12
  • anti-Vn H-270
  • anti-uPAR FL-290
  • anti-uPAR-1 R&D Systems
  • anti- ⁇ -tubulin Calbiochem
  • anti- ⁇ 3 integrin Chemicon International
  • anti-CD61 Sigma
  • anti-caveolin Sigma
  • anti-GAPDH anti-Flag
  • anti-HA anti-transferrin receptor and anti caveolini
  • anti-synaptopodin mouse monoclonal antibody G1
  • anti-synaptopodin rabbit polyclonal NT
  • Peter Mundel N.Y.
  • uPAR -/- mice and uPA -/- mice on a mixed background of 75% C57BL/6 and 25% 129 Swiss were obtained from University of Leuven, Belgium; Vn -/- mice (C57BL/6) were obtained from Dr. David Ginsburg, University of Michigan; ⁇ 3 integrin -/- mice (C57BL/6) were obtained from Dr. Raghuram Kalluri, Boston.
  • C57BL/6 and 129 Swiss mice were purchased from Jackson Laboratory (Bar Harbor).
  • the LPS mouse model was utilized as previously described 9 . The induction of B7-1 was used as a control for the effectiveness of LPS.
  • Rat PAN nephrosis model was set up by a single intraperitoneal injection of puromycin (15 mg/100 g of body weight, Sigma-Aldrich) into Spragne-Dawley rats as described before 27 .
  • NZB/W F1 mice were purchased from the Jackson Laboratory and analyzed after 20 weeks when proteinuria and Lupus glomerulonephritis were present.
  • As diabetic nephropathy rat model we used Sprague Dawley rats treated with Streptozotocin. Hyperglycaemic state was induced by an intraperitoneal Streptozotocin injection (50-70 mg/kg body weight, in citrate buffer 10 mmol/L, pH 4.5).
  • the hyperglycaemic state developed within 48 h and was maintained during the lifetime of the animals. No insulin administration was required. Glycosuria was evaluated with test strips of the Uriscan TM (YD Diagnostics, VWR, California, QC, Canada) and glycaemia with the AccuSoft Monitoring System (Roche Diagnostics, Laval, QC, Canada). At the end of the study, glycaemia values were 43 ⁇ 0.53 and 8.5 ⁇ 0.7 mmol/L for 3 months-old and 12 months-old normoglycaemic animals, respectively, and 21.2 ⁇ 0.9 and 33.9 ⁇ 4.0 mmol/L for 3 months-old and 12 months-old diabetic animals, respectively.
  • FSGS focal and segmental glomerulosclerosis
  • DN diabetic nephropathy
  • TaqMan real-time RT-PCR was done as previously reported 26 .
  • commercially available predeveloped TaqMan assay reagents (Applied Biosystems) were used for uPAR mRNA analysis.
  • the mRNA expression of uPAR was related to that of synaptopodin, which was used as a podocyte reference gene.
  • the confounding factor of alterations in the proportion of podocyte cell number per total glomerular cells was counterbalanced, and only RNA from the podocyte compartment of the glomerulus was integrated in the analysis, as demonstrated recently 26 .
  • mice were washed with PBS, and H 2 O and mounted for analysis with a confocal microscope (Bio-Rad Laboratories).
  • uPAR -/- mice were injected with ⁇ 3 ⁇ 616-690 or wild type ⁇ 3 integrin constructs. 14 h after iniection, mice were sacrificed and kidney was snap-frozen. Cyro-sections were cut at 4 ⁇ m and fixed with cold acetone for 10 min before incubated with WOW-1 Fab for 1 h. After washing with PBS, the sections were then incubated with the secondary antibody, anti-mouse Fab conjugated with 488 (Invitrogen) for 50 min and analyzed by confocal microscopy.
  • 488 Invitrogen
  • TEM and IEM were performed according to the standard protocols 8 .
  • morphometry of uPAR labeling across the glomerular wall we used renal tissues from 3 and 12 months-old hyperglycaemic animals with the corresponding age-matched normoglycaemic animals (3-4 animals per group). Small pieces of renal cortex were sampled from the anaesthetized animal (urethane, 1 g per kg body weight). The tissue samples were immediately fixed by immersion in periodate-lysine-paraformaldehyde solution, dehydrated in graded methanol and embedded in Lowicryl, following protocols described previously 52 .
  • the grids carrying the ultra thin tissue sections were incubated on a drop of a saturated solution of sodium metaperiodate for 10 min, washed with distilled water, transferred on a drop of 0.15 M glycine for 10 min and washed with PBS. Grids were then incubated on a drop of ovalbumin 1% for 5 min and transferred to the diluted anti-uPAR antibody (1:10) overnight at 4° C. The grids were washed with PBS and incubated on a drop of protein A-gold complex for 30 min at room temperature. The grids were then washed with PBS (3 ⁇ 5 min) and distilled water (1 ⁇ 5min), dried and contrasted with uranyl acetate. Specificity of the immunolabelings was evaluated by control experiments, omitting the specific antibody step, replacing it with PBS.
  • Micrographs of immunolabeled renal glomeruli were recorded (>20 micrographs per animal and timepoint), printed and evaluated through morphometrical techniques 52 .
  • the measurements were performed by direct planimetry and particle counting using an image processing system (Videoplan 2, Carl Zeiss Inc., Toronto, Canada).
  • uPAR-cDNA plasmids were introduced into uPAR -/- mouse using the TransIT in vivo gene delivery system (Mirus) as described previously 32, 8 .
  • the following cDNAs and vector constructs were used for gene delivery: uPAR; mutated uPAR D262A 38 (kind gift from Y Wei and HA Chapman, University of California, San Francisco); ⁇ 3 integrin and the constitutively active form of ⁇ 3 integrin, D616-690 ( ⁇ 3 ⁇ 616-690 ) 48 ; Podocin promoter vector p2.5 33 (a kind gift from Dr. LB Holzman, University of Michigan) for Pod-uPAR; ICAM-2 promoter vector 34 for ICAM-2-uPAR. Tissues were analyzed for expression as described previously 8 .
  • siRNA Mouse uPAR siRNA (sense: CTTCCTGAAGTGTTGCAACTA) was constructed and inserted into a pRNA-H1.2/Neo vector (Genescript). Stable transfection was done with podocytes maintaining at 33° C. by Lipofectamine 2000 (Invitrogen). Positive clones were selected by G418 (Sigma-Aldrich) at 500 ⁇ g/ml. For further experiments with uPAR siRNA, cells were grown under non-permissive conditions for 10-14 days before proceeded with migration experiments.
  • AP5 activity of AP5 in mouse podocytes
  • cells were cultured under non-permissive conditions for 10 days. Then medium was discarded and the cells were washed gently with PBS without Ca 2+ and Mg 2+ . The cells were then exposed to LPS right before they were incubated with various amounts of calcium in the presence of 5 ⁇ g/ml AP5 for 1 h: 1 mM EDTA, PBS without Ca 2+ or Mg 2+ ; 0.1 nM Cacl 2 ; 0.4 mM CaCl 2 ; 1 mM CaCl 2 ; 2 mM CaCl 2 . The cells were then harvested by cell scraper, washed and subjected to flow cytometry analysis.
  • HEK 293 cells were seeded on a 100 mm culture dish and maintained at 37° C. in DMEM with 10% FBS. Upon 90% confluent, cells were transtected with constructs encoding uPAR or ⁇ 3 integrin by Lipofectamine 2000 (Invitrogen). 24 h after transfection, cells were harvested for further experiments. Wild type podocytes were differentiated by culturing at 37° C. for at least 10 days (10 days for transfection and migration assay, 14 days for other experiments).
  • HEK cells were co-transfected with constructs encoding uPAR, and ⁇ 3 integrin for 24 h. Then cells were lysed in RIPA buffer with a cocktail of protease inhibitors. The lysate was incubated with 25 ⁇ l of Flag or HA agarose beads (Sigma) at 4° C., overnight after a pre-cleaning. The beads were then washed 5 times with RIPA buffer and were eluted by heating at 70° C. for 10 min. After a brief centrifugation, the supernatants were ready for Western blotting.
  • Podocyte migration was analyzed using a 12-well chemotaxis chamber (Neuro Probe) according to the manufacturer's protocol. In brief differentiated podocytes were treated with 50 ⁇ g/ml of LPS or PAN for 24 h and then harvested for the migration assay. Bottom plates were coated with vitronectin or type I collagen, while upper plates were loaded with equal number of cells (5 ⁇ 10 4 ) suspended in medium. The chamber was incubated at 37° C. for 4 h before the membrane was taken out and stained with diamidino phenylindole (DAPI). The average number of migrated cells was counted in four fields in six independent experiments.
  • DAPI diamidino phenylindole
  • podocytes were seeded on Vn or type I collagen coated cover-slips and cultured for 10 days at 37° C. before treated with LPS or PAN for 12-48 h.
  • LPS or PAN for 12-48 h.
  • coverslips were washed twice with PBS and incubated in medium. After incubation, cells were fixed with 2% PFA and stained with DAPI for analysis. The cell migratory distance was calculated by averaging the distance from the wound edge to the maximally migrated cell in five distinct border zones.
  • Podocyte lysate was overlaid with a sucrose step gradient and centrifuged for 20 h at 120,000 g at 4° C. in a swing-out rotor. 10 fractions (1 ml each) were collected starting from the top and analyzed by Western blotting with rafts, non-rafts markers, as well as uPAR and ⁇ 3 antibodies.
  • Cyclo-RGDfV (Biomol) was injected in mice at 1, 5 and 20 mg per kg body weight i.v. in mice every 8 h for three times. Control mice received the same amount of PBS instead or the control peptide cyclo-RAD (Biomol), (data not shown). Immediately after the first injection, 200 ⁇ g of LPS was injected intraperitoneally into each mouse to induce proteinuria. 24 h after LPS injection, urine was collected for Braford assay.
  • the antibody was administered 4 h after LPS injection through tail vein, with a final concentration of 10 ⁇ g/ml.
  • Urine was collected at time points 0, 4 and 24 h and analyzed by Bradford assay. Data represented in the figure was based on sample obtained after 24 h.
  • CycloRGDfV also reduces already existing proteinuria ( FIG. 11 ).
  • B6 mice were injected LPS twice (0, 24 h) with LPS to induce and maintain proteinuria.
  • 48 h, 66 h urine was collected for Braford assay.
  • the diagram shows the fold change of urinary protein at timepoint 66 h, *p ⁇ 0.014.
  • S247 also reduces already existing proteinuria.
  • B6 mice are injected twice (0, 24 h) with LPS to induce and maintain proteinuria.
  • 48 h, 66 h urine was collected for Braford assay. Reduction of urinary protein at timepoint 66 h by S247 is observed.
  • uPAR Quantitative analysis of uPAR expression and localization in normal and diabetic glomeruli.
  • uPAR is induced in podocyte foot processes of 3 and 12 months old diabetic rats.
  • Mundel, P. et al. Synaptopodin an actin-associated protein in telencephalic dendrites and renal podocytes. J Cell Biol 139, 193-204 (1997).
  • objects of the present invention are compounds of formula (I), as described above, a process for their preparation, their use as medicaments and pharmaceutical compositions containing them.
  • T is understood as meaning a group COOH, a radical hydrolyzable to COOH or a radical bioisosteric to COOH.
  • a radical hydrolyzable to COOH is understood as meaning a radical with changes into a group COOH after hydrolysis.
  • a group which may be mentioned by way of example as a radical hydrolyzable to COOH is
  • a radical biolsosteric to COOH is understood as meaning radicals which can replace the function of a group COOH in active compounds by equivalent bond donor/acceptor capabilities or by equivalent charge distribution.
  • Radicals which may be mentioned by way of example as radicals bioisosteric to —COOH are those such as described in “The Practice of Medicinal Chemistry, Editor: C. G. Wermuth, Academic Press 1996, pages 125 and 216, in particular the radicals —P ⁇ O(OH) 2 , —SO 3 H, tetrazole or acylsulfonamides.
  • Preferred radicals T are —COOH, —CO—O—C 1 -C 8 -alkyl or —CO—O-benzyl.
  • the radical —U— in the structural element L is a spacer selected from the group —(X L ) a —(CR L 1 R L 2 ) b —, —CR L 1 ⁇ CR L 2 —, ethynylene or 40 ⁇ CR L 1 —.
  • the structural element L is linked to the structural element G via a double bond.
  • X L is a radical CR L 3 R L 4 , NR L 5 , oxygen or sulfur.
  • a halogen radical is understood as meaning, for example, F, Cl, Br or I, preferably F.
  • a branched or unbranched C 1 -C 6 -alkyl radical is understood as meaning, for example, methyl, ethyl, propyl, 1-methylethyl, butyl, 1-methylpropyl, 2-methylpropyl, 1,1-dimethylethyl, pentyl, 1-methylbutyl, 2-methylbutyl, 1,2-dimethylpropyl, 1,1-dimethylpropyl, 2,2-dimethylpropyl, 1-ethylpropyl, hexyl, 1-methylpentyl, 1,2-dimethylbutyl, 1,3-dimethylbutyl, 2,3-dimethylbutyl, 1,1-dimethylbutyl, 2,2-dimethylbutyl, 3,3-dimethylbutyl, 1,1,2-trimethylpropyl, 1,2,2-trimethylpropyl, 1-ethylbutyl, 2-ethyl
  • a branched or unbranhced C 2 -C 6 -alkenyl radical is understood as meaning, for example, vinyl, 2-propenyl, 2-butenyl, 3-butenyl, 1-methyl-2-propenyl, 2-methyl-2-propenyl 2-pentenyl, 3-pentenyl, 4-pentenyl, 1-methyl-2-butenyl, 2-methyl-2-butenyl, 3-methyl-2-butenyl, 1-methyl-3-butenyl, 2-methyl-3-butenyl, 3-methyl-3-butenyl, 1,1-dimethyl-2-propenyl, 1,2-dimethyl-2-propenyl, 1-ethyl-2-propenyl, 2-hexenyl, 3-hexenyl, 4-hexenyl, 5-hexenyl, 1-methyl-2-pentenyl, 2-methyl-2-pentenyl, 3-methyl-2-penteny
  • a branched or unbranched C 2 -C 6 -alkynyl radical is understood as meaning, for example, ethynyl, 2-propynyl, 2-butynyl, 3-butynyl, 1-methyl-2-propynyl, 2-pentynyl, 3-pentynyl, 4-pentynyl, 1-methyl-3-butynyl, 2-methyl-3-butynyl, 1-methyl-2-butynyl, 1,1-dimethyl-2-propynyl, 1-ethyl-2-propynyl, 2-hexynyl, 3-hexynyl, 4-hexynyl, 5-hexynyl, 1-methyl-2-pentynyl, 1-methyl-2-pentynyl, 1-methyl-3-pentynyl, 1-methyl-4-pentynyl, 2-methyl-3-p
  • a branched or unbranched C 3 -C 7 cycloal-kyl radical is understood as meaning, for example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl or cycloheptyl.
  • a branched or unbranched C 1 -C 4 -alkoxy radical is understood as meaning, for example, methoxy, ethoxy, propoxy, 1-methylethoxy, butoxy, 1-methylpropoxy, 2-methylpropoxy or 1,1-dimethylethoxy.
  • the radicals —CO—NH(C 1 -C 6 -alkyl), —CO—N(C 1 -C 6 -alkyl) 2 are secondary or tertiary amides and are composed of the amide bond and the corresponding C 1 -C 6 -alkyl radicals such as described above for R L 1 , R L 2 , R L 3 or R L 4 .
  • radicals R L 1 , R L 2 , R L 3 or R L 4 can furthermore be a radical
  • C 1 -C 2 alkylene-T such as methylene-T or ethylene-T
  • C2-alkenylene-T such as ethenylene-T or C 2 -alkynylene-T, such as ethynylene-T
  • R L 1 and R L 2 or R L 3 and R L 4 or optionally R L 1 and R L 3 can in each case independently of one another together be an optionally substituted 3- to 7-membered saturated or unsaturated carbocycle or heterocycle, which can contain up to three different or identical heteroatoms O, N, S.
  • R L 1 , R L 2 , R L 3 or R L 4 can be optionally substituted.
  • suitable substituents if the substituents are not specified in greater detail, are independently of one another up to 5 substituents, for example selected from the following group:
  • R L 1 , R L 2 , R L 3 or R L 4 are independently of one another hydrogen, halogen, a branched or unbranched, optionally substituted C 1 -C 4 -alkyl, C 1 -C 4 -alkoxy or C 3 -C 7 -cycloalkyl radical or the radical —NR L 6 R L 7 .
  • radicals R L 1 , R L 2 , R L 3 or R L 4 are independently of one another hydrogen, fluorine or a branched or unbranched, optionally substituted C 1 -C 4 -alkyl radical, preferably methyl.
  • radicals R L 5 , R L 6 , R L 7 in structural element L are independently of one another hydrogen, a branched or unbranched, optionally substituted
  • Preferred radicals for R L 6 in structural element L are hydrogen, a branched or unbranched, optionally substituted C 1 -C 4 -alkyl, CO—O—C 1 -C 4 -alkyl, CO—C 1 -C 4 -alkyl or SO 2 —C 1 -C 4 -alkyl radical or an optionally substituted Co-O-benzyl, SO 2 -aryl, SO 2 -alkylenearyl or CO-aryl radical.
  • Preferred radicals for R L 7 in structural element L are hydrogen or a branched or unbranched, optionally substituted C 1 -C 4 -alkyl radical.
  • Preferred structural elements L are composed of the preferred radicals of the structural element.
  • Particularly preferred structural elements L are composed of the particularly preferred radicals of the structural element.
  • G is a structural element of the formula I G
  • Y G in structural element G is CO, CS, C ⁇ NR G 2 or CR G 3 R G 4 , preferably CO, C ⁇ NR G 2 or CR G 3 R G 4 , particularly preferably CO or CR G 3 R G 4 .
  • R G 2 in structural element G is hydrogen, a hydroxyl group, a branched or unbranched, optionally substituted C 1 -C 6 -alkyl, C 1 -C 4 -alkoxy or C 3 -C 7 -cycloalk- yl radical, for example as described above for R L 1 in each case,
  • Preferred radicals R G 2 in structural element G are hydrogen, hydroxyl or a branched or unhranched, optionally substituted C 1 -C 6 -alkyl radical, in particular methyl or C 1 -C 4 -alkoxy radical, in particular methoxy.
  • Branched or unbranched, optionally substituted C 1 -C 6 -alkyl, C2-C6-alkynyl or C 1 -C 4 -alkoxy radicals for R G 3 or R G 4 in structural element G independently of one another are understood as meaning, for example, the corresponding radicals in each case described above for R L 1 .
  • both radicals R G 3 and R G 4 can together form a cyclic acetal, such as —O—CH 2 —CH 2 —O— or —O—CH 2 —O—.
  • both radicals R G 3 and R G 4 can together form an optionally substituted C 3 -C 7 -cycloalkyl radical.
  • radicals for R G 3 or R G 4 are independently of one another hydrogen, C 1 -C 6 -alkyl or C 1 -C 4 -alkoxy, and both radicals R G 3 and R G 4 together form a cyclic acetal, such as —O—CH 2 —CH 2 —O— or —O—CH 2 —O—.
  • Particularly preferred radicals R G 3 or R G 4 are independently of one another hydrogen and both radicals R G 3 and R G 4 together form a cyclic acetal, in particular —O—CH 2 —CH 2 —O— or —O—CH 2 —O—.
  • Branched or unbranched, optionally substituted C 1 -C 6 -alkyl or C 1 -C 4 -alkoxy radicals and optionally substituted aryl or arylalkyl radicals for R G 5 and R G 6 in structural element G independently of one another are, for example, the corresponding radicals in each case described above for R L 1 .
  • both radicals R G 5 and R G 6 can together form an optionally substituted, fused, unsaturated or aromatic 3- to 10-membered carbocycle or heterocycle, which can contain up to three different or identical heteroatoms O, N, S, where in this fused, unsaturated or aromatic 3- to 10-membered carbocycle or heterocycle, as substituents, independently of one another up to four substituents from the group
  • Preferred substituents are halogen, a C 1 -C 4 -alkyl radical, C 1 -C 4 -alkoxy radical or aryl radical.
  • substituents are a C 1 -C 4 -alkyl radical, in particular methyl or ethyl, a C 1 -C 4 -alkoxy radical, in particular methoxy, or F or Cl.
  • R G 5 and R G 6 are independently of one another hydrogen, an optionally substituted C 1 -C 6 -alkyl radical, in particular methyl and ethyl, an optionally substituted aryl radical, in particular optionally substituted phenyl or an optionally substituted arylalkyl radical, in particular an optionally substituted benzyl radical, and in each case both radicals R G 5 and R G 6 together can be an optionally substituted, fused, unsaturated or aromatic 3- to 10-membered carbocycle or heterocycle which can contain up to three different or identical heteroatoms O, N, S.
  • both radicals R G 5 and R G 6 together form an optionally substituted, fused, unsaturated or aromatic 3- to 6-membered carbocycle or heterocycle, for example selected from one of the following doubly bonded structural formulae:
  • W G is a structural element selected from the group of structural elements of the formulae I WG 1 to I WG 4 , where the dashed lines intersect the atomic bonds within the structural element G and the carbon atom substituted by R G 7 and R G 8 is bonded to Y G .
  • W G is a structural element selected from the group of structural elements of the formulae I WG 1 , I WG 2 and I WG 4 , in particular the structural element of the formula IWG 2 .
  • R G 1 in structural element W G is hydrogen, halogen, such as Cl, F, Br or I, a hydroxyl group or a branched or unbranched, optionally substituted C 1 -C 6 -alkyl radical, preferably C 1 -C 4 -alkyl or C 1 -C 4 -alkoxy radical such as in each case described above for R L 1 .
  • Preferred radicals for R G 1 are hydrogen, hydroxyl and optionally substituted C 1 -C 4 -alkyl or C 1 -C 4 -alkoxy radicals.
  • radicals for R G 1 are hydrogen and carboxyl-substituted C 1 -C 4 -alkyl or C 1 -C 4 -alkoxy radicals, in particular the radicals —CH 2 COOH or —O—CH 2 COOH.
  • R G 7 , R G 8 , R G 9 and R G 10 in structural element G are independently of one another hydrogen, a hydroxyl group, CN, halogen, such as F, Cl, Br, I, a branched or unbranched, optionally substituted
  • two radicals R G 7 and R G 9 or R G 8 and R G 10 or R G 7 and R G 8 or R G 9 and R G 1 O can in each case independently of one another together form an optionally substituted, saturated or unsaturated, nonaromatic, 3- to 7-membered carbocycle or heterocycle which can contain up to 3 heteroatoms selected from the group consisting of O, N, S and up to two double bonds.
  • R G 7 , R G 8 , R G 9 and R G 10 in the structural element G are independently of one another hydrogen, halogen, in particular a branched or unbranched, optionally substituted C 1 -C 6 -alkyl, or C 2 -C 6 -alkynyl radical, a branched or unbranched, optionally substituted radical C 1 -C 4 -alkylene-OR G 11 , C 1 -C 4 -alkylene-CO—OR- G 11 , C 1 -C 4 -alkylene-O—CO-R G 11 , C 1 -C 4 -alkylene-CO—NR G 12 R G 13 , C 1 -C 4 -alkylene-O—CO—NR G 12 R G 13 , a radical —O-R G 11 , —CO—OR G 11 , —O—CO—R G 11 , —O—CO—NR G 12 R G 13 , —CO—NR G 12 R
  • radicals for R G 7 R G 8 , R G 9 and R G 10 in the structural element G are independently of one another hydrogen, F, a radical —CO—OR G 11 , —CO—NR G 12 R G 13 , or an optionally substituted aryl radical, as described above in each case.
  • a branched or unbranched, optionally substituted C 1 -C 8 -alkyl radical for R G 11 , R G 12 and R G 13 is understood as meaning independently of one another, for example, the C 1 -C 6 -alkyl radicals mentioned above for R G 1 , plus the radicals heptyl and octyl.
  • Preferred substituents of the branched or unbranched, optionally substituted C 1 -C 8 -alkyl radicals for R G 11 , R G 12 and R G 13 independently of one another are the radicals halogen, hydroxyl, C 1 -C 4 -alkoxy, —CN, —COOH and —CO—O—C 1 -C 4 -alkyl.
  • Preferred branched or unbranched, optionally substituted —C 1 -C 5 -alkylene-C 1 -C 4 -alkoxy radicals for R G 11 , R G 12 and R G 13 are independently of one another methoxymethylene, ethoxymethylene, t-butoxymethylene, methoxyethylene or ethoxyethylene.
  • Preferred branched or unbranched, optionally substituted mono- and bisalkylaminoalkylene or acylaminoalkylene radicals for R G 11 , R G 12 and R G 13 are independently of one another branched or unbranched, optionally substituted radicals —C 1 -C 4 -alkylene-NH(C 1 -C 4 -alkyl), —C 1 -C 4 -alkylene-N(C 1 -C 4 -alkyl) 2 or —C 1 -C 4 -alkylene-NH—CO—C 1 -C 4 -alkyl.
  • Preferred optionally substituted heterocycloalkyl, heterocycloalkenyl, C 1 -C 4 -alkyleneheterocycloalkyl or C 1 -C 4 -alkyleneheterocycloalkenyl radicals for R G 11 , R G 12 and R G 13 are independently of one another the C 3 -C 7 -heterocycloalkyl, C 3 -C 7 -heterocycloalkenyl, C 1 -C 4 -alkylene-C 3 -C 7 -heterocycloalkyl or C 1 -C 4 -alkylene-C 3 -C 7 -heterocycloalkenyl radicals described above for R G 1 .
  • heterocycloalkyl, heterocycloalkenyl, C 1 -C 4 -alkyleneheterocycloalkyl or C 1 -C 4 -alkyleneheterocycloalkenyl radicals for R G 11 , R G 12 and R G 13 are independently of one another the C 3 -C 7 -heterocycloalkyl, C 3 -C 7 -heterocycloalkenyl, C 1 -C 4 --alkylene-C 3 -C 7 -heterocycloalkyl or C 1 -C 4 -alkylene-C 3 -C 7 -heterocycloalkenyl radicals described above for R G 1 , one or two heteroatoms being selected from the group consisting of N, O and S and up to two double bonds being contained in the cyclic moiety.
  • R G 12 and R G 13 can independently of one another be a radical —SO 2 —R G 11 , —CO—O-R G 11 , —CO—NR G 11 R G 11 * or —CO-R G 11 , R G 11 * being a radical R G 11 which is independent of R G 11 .
  • both radicals R G 12 and R G 13 can together form a 5- to 7-membered, preferably saturated nitrogen-containing carbocycle, in the sense of a cyclic amine structure, such as N-pyrrolidinyl, N-piperidinyl, N-hexahydroazepinyl, N-morpholinyl or N-piperazinyl, where in the case of heterocycles which carry free amine protons, such as N-piperazinyl, the free amine protons can be replaced by customary amine protective groups, such as methyl, benzyl, Boc (tert-butoxycarbonyl), Z (benzyloxycarbonyl), tosyl, —SO 2 —C 1 -C 4 -alkyl, —SO 2 -phenyl or —SO 2 -benzyl.
  • a cyclic amine structure such as N-pyrrolidinyl, N-piperidinyl, N-hexahydroazepinyl, N-
  • Particularly preferred radicals for R G 11 are hydrogen or an optionally substituted C 1 -C 4 -alkyl or aryl radical.
  • R G 12 and R G 13 are independently of one another hydrogen or an optionally substituted C 1 -C 4 -alkyl radical.
  • Preffered structural elements G are composed of at least one preferred radical of the strucural element G, while the remaining radicals are widely variable.
  • Particularly preferred structural elements G are composed of the preferred radicals of the structural element G.
  • Very particularly preferred structural elements G are composed of the particularly preferred radicals of the structtnul element G.
  • Structural element B is undentood as meaning a structural element comprising at least one atom which under physiological conditions can form hydrogren bridges as a hydrogen acceptor, at least one hydrogen acceptor atom having a distance of 4 to 15 atom bonds to structural element G along the shortest possible route along the structural element skeleton.
  • the arrangement of the structural skeleton of structural element B is widely variable.
  • Suitable atoms which under physiological conditions can form hydrogen bridges as hydrogen acceptors are, for example, atoms having Lewis base properties, such as the heteroatoms nitrogen, oxygen or sulfur.
  • Physiological conditions is understood as meaning a pH which prevails at the site in a body at which the ligands interact with the receptors.
  • the physiological conditions have a pH of, for example, 5 to 9.
  • structural element B is a structural element of the formula IB
  • the structural element A is a structural element selected from the group of structural elements of the formulae I A 1 to I A 18 ,
  • the structural element A is a structural element of the formula I A 1 I A 4 , I A 7 , I A 8 or I A 9 .
  • a branched or unbranched, optionally substituted C 1 C 6 -alkyl radical for R A 1 or R A 2 independently of one another is understood as meaning, for example, the corresponding radicals described above for R G 1 , preferably methyl or trifluoromethyl.
  • the branched or unbranched optionally substituted radical CO—C 1 -C 6 -alkyl is composed, for example, of the group CO and the branched or unbranched, optionally substituted C 1 -C 6 -alkyl radicals described above for R A 1 or R A 2 .
  • Optionally substituted hetaryl, hetarylalkyl, aryl, arylalkyl or C 3 C 7 -cycloalkyl radicals for R A 1 or R A 2 independently of one another are understood as meaning, for example, the corresponding radicals described above for R G 7 .
  • the optionally substituted radicals Co-O-R A 14 , O-R A 14 , S-R A 14 , NR A 15 R A 16 , CO—NR A 15 R A 16 or SO 2 NR A 15 R A 16 are composed, for example, of the groups CO—O, O, S, N, CO—N or SO 2 —N and the radicals R A 14 , R A 15 or R A 16 described in greater detail below.
  • both radicals R A 1 and R A 2 can together form a fused, optionally substituted, 5- or 6-membered, unsaturated or aromatic carbocycle or heterocycle which can contain up to three heteroatoms selected from the group consisting of O, N and S.
  • R A 13 and R A 13 * are the radicals hydrogen, F, Cl, a branched or unbranched, optionally substituted C 1 -C 5 -alkyl radical, optionally substituted aryl or arylalkyl or a radical Co-O-R A 14 , O-R A 14 , NR A 15 R A 16 , SO 2 -NR A 15 R A 16 or CO—NR A 15 R A 16 .
  • a branched or unbranched, optionally substituted C 1 -C 6 -alkyl, C 3 -C 7 -cycloalkyl, alkylenecycloalkyl, alkylene-C 1 -C 4 -alkoxy, C 2 -C 6 -alkenyl or C 2 -C 6 -alkynyl radical for R A 14 in structural element A is understood as meaning, for example, the corresponding radicals described above for R G 7 .
  • Optionally substituted aryl, arylalkyl, hetaryl or alkylhetaryl radicals for R A 14 in structural element A are understood as meaning, for example, the corresponding radicals described above for R G 7 .
  • Preferred radicals for R A 14 are hydrogen, a branched or unbranched, optionally substituted C 1 -C 6 -alkyl radical and optionally substituted benzyl.
  • a branched or unbranched, optionally substituted C 1 -C 6 -alkyl or arylalkyl radical or an optionally substituted C 3 -C 7 -cycloalkyl, aryl, hetaryl or hetaryalkyl radical for R A 15 or R A 16 independently of one another is understood as meaning, for example, the corresponding radicals described above for R A 14 .
  • the branched or unbranched, optionally substituted CO—C 1 -C 6 -alkyl, SO 2 —C 1 -C 6 -alkyl, COO-C 1 -C 6 -alkyl, CO—NH—C 1 -C 6 -alkyl, COO-alkylenearyl, CO—NH-alkylenearyl, CO—NH-alkylenehetaryl or SO 2 -alkylenearyl radicals or the optionally substituted CO-aryl, SO 2 -aryl, CO—NH-aryl, CO—NH-hetaryl or CO-hetaryl radicals for R A 15 or R A 16 are composed, for example, of the corresponding groups —CO—, SO 2 —, —CO—O—, —CO—NH— and the corresponding branched or unbranched optionally substituted C 1 -C 6 alkyl, hetarylalkyl or arylalkyl radicals or the corresponding optionally substituted aryl or
  • a radical —(CH 2 ) n —(X A ) j —R A 12 for R A 3 or R A 4 independently of one another is understood as meaning a radical which is composed of the corresponding radicals —(CH 2 ) n —, (XA) j and R A 12 .
  • n can be 0, 1, 2 or 3 and j can be: 0 or 1.
  • X A is a doubly bonded radical selected from the group consisting of —CO—, —CO—N(R x 1 )—, —N(R x 1 )—CO—, —N((R x 1 ))-Co-N((R x 1 )*)-, —N(Rx 1 )—CO—O—, —O—, —S—, —SO 2 —, —SO 2 —N(R x 1 )—, —SO 2 —O—, —CO—O—, —O—CO—, —O—CO—N((R x 1 ))—, —N(Rx 1 )— or —N((R x 1 ))—SO 2 —.
  • R A 12 and R x 1 or RX 1 * can together form a saturated or unsaturated C 3 - C 7 -heterocycle which can optionally contain up to two further heteroatoms selected from the group consisting of O, S and N.
  • the radical R A 12 together with the radical R x 1 or R x 1 * forms a cyclic amine as the C 3 -C 7 -heterocycle in the case where the radicals are bonded to the same nitrogen atom, such as N-pyrrolidinyl, N-piperidinyl, N-hexahydroazeptnyl, N-morpholinyl or N-piperazinyl, where in heterocycles which carry free amine protons, such as N-piperazinyl, the free amine protons can be replaced by customary amine protective groups, such as methyl, benzyl, Boc (tert-butoxycarbonyl), Z (benzyloxycarbonyl), tosyl, —SO 2 —C 1 -C 4 -alkyl, —SO 2 -phenyl or —SO 2 -benzyl.
  • customary amine protective groups such as methyl, benzyl, Boc (tert-butoxycarbonyl), Z (benzyl
  • a branched or unbranched, optionally substituted C 1 -C 6 -alkyl, C 2 -C 1 2-alkynyl, preferably C 2 -C 6 -alkynyl or C 2 -C 6 -alkenyl radical, an optionally substituted C 3 -C 7 -cycloalkyl, aryl, arylalkyl or hetaryl radical for Rx 1 and R x 1 * independently of one another is understood as meaning, for example, the corresponding radicals described above for R G 7 .
  • Preferred branched or unbranched, optionally substituted C 1 -C 6 -alkoxyalkyl for R x 1 and R x 1 * are independently of one another methoxymethylene, ethoxymethylene, butoxymethylene, methoxyethylene or ethoxyethylene.
  • Preferred branched or unbranched, optionally substituted radicals CO—C 1 -C 6 -alkyl, CO—O—C 1 -C 6 -alkyl, SO 2 —C 1 -C 6 -alkyl, CO—O-alkylenearyl, CO-alkylenearyl, CO-aryl, SO 2 -aryl, CO-hetaryl or SO 2 -alkylenearyl are preferably composed of the C 1 -C 6 -alkyl, arylalkyl, aryl or hetaryl radicals and the radicals —CO—, —O—, —SO 2 — described above.
  • R x 1 and R x 1 * are independently of one another hydrogen, methyl, cyclopropyl, allyl and propargyl.
  • both radicals R A 6 and R A 6 * in structural element I A 7 can together form an optionally substituted, saturated, unsaturated or aromatic heterocycle which, in addition to the ring nitrogen, can contain up to two further different or identical heteratoms O, N, S.
  • R A 7 is hydrogen, —OH, —CN, —CONH 2 , a branched or unbranched, optionally substituted C 1 -C 4 -alkyl radical, for example as described above for R A 6 , C 1 -C 4 -alkoxy, arylalkyl or C 3 -C 7 -cycloalkyl radical, for example as described above for R L 14 , a branched or unbranched, optionally substituted —O—CO—C 1 -C 4 -alkyl radical, which is composed of the group —O—CO— and, for example, of the C 1 -C 4 -alkyl radicals mentioned above or an optionally substituted —O-alkylenearyl, —O—CO-aryl, —O—CO-alkylenearyl or —O—CO-allyl radical which is composed of the groups —O— or —O—CO— and, for example, of the corresponding radicals described above for R G 7 .
  • both radicals R A 6 and R A 7 can together form an optionally substituted unsaturated or aromatic heterocycle which, in addition to the ring nitrogen, can contain up to two further different or identical heteroatoms O, N, S.
  • a branched or unbranched, optionally substituted C 1 -C 4 -alkyl radical or an optionally substituted aryl or arylalkyl radical is understood as meaning, for example, the corresponding radicals described above for R A 15 , where the radicals CO—C 1 -C 4 -alkyl, SO 2 - C 1 -C 4 -alkyl, CO—O—C 1 -C 4 -alkyl, CO-aryl, SO 2 -aryl, CO—O-aryl, CO-alkylenearyl, SO 2 -alkylenearyl or CO—O-alkylenearyl are composed analogously to the other composed radicals of the group consisting of CO, SO 2 and COO and for example, of the corresponding C 1 -C 4 -alkyl, aryl or arylalkyl radicals described above for R A 15 , and these radicals can be optionally substituted.
  • a branched or unbranched, optionally substituted C 1 -C 6 -alkyl radical or an optionally substituted aryl, arylalkyl, hetaryl or C 3 -C 7 -cycloalkyl radical independently of one another is understood as meaning, for example, the corresponding radicals described above for R A 14 , preferably methyl or trifluoromethyl.
  • a radical CO—O—R A 14 , O—R A 14 , S-R A 14 , SO 2 —NR A 15 R A 16 , NR A 15 R A 16 or CO—NR A 15 R A 16 independently of one another is understood as meaning, for example, the corresponding radicals described above for R A 13 .
  • both radicals R A 9 and R A 10 together in structural element I A 14 can form a 5- to 7-membered saturated, unsaturated or aromatic carbocycle or heterocycle, which can contain up to three different or identical heteroatoms O, N, S and is optionally substituted by up to three identical or different radicals.
  • Substituents in this case are in particular understood as meaning halogen, CN, a branched or unbranched, optionally substituted C 1 -C 4 -alkyl radical, such as methyl or trifluoromethyl or the radicals O—R A 14 , S-R A 14 , NR A 15 R A - 16, CO—NR A 15 R A 16 or —((R A 8 )HN)C ⁇ NR.sub- .A 7 .
  • a branched or unbranched, optionally substituted C 1 -C 6 -alkyl radical or an optionally substituted aryl, arylalkyl, hetaryl, C 3 -C 7 -cycloalkyl radical or a radical CO—O—R A 14 , O—R A 14 , S—R A 14 , NR A 15 R A 16 , SO 2 —NR A 15 R A 16 or CO—NR A 15 R A 16 for R A 11 is understood, for example, as meaning the corresponding radicals described above for R A 9 .
  • both radicals R A 9 and R A 17 together can form a 5- to 7-membered saturated, unsaturated or aromatic heterocycle which, in addition to the ring nitrogen, can contain up to three different or identical heteroatoms O, N, S and is optionally substituted by up to three identical or different radicals.
  • Z 1 , Z 2 , Z 3 , Z 4 are independently of one another nitrogen, C—H, C-halogen, such as C—F, C—Cl, C—Br or C—I or a branched or unbranched, optionally substituted C—C 1 -C 4 -alkyl radical which is composed of a carbon radical and, for example, a C 1 -C 4 -alkyl radical described above for R A 6 or a branched or unbranched optionally substituted C—C 1 -C 4 -alkoxy radical which is composed of a carbon radical and, for example, a C 4 -alkoxy radical described above for R A 7 .
  • C-halogen such as C—F, C—Cl, C—Br or C—I or a branched or unbranched
  • optionally substituted C—C 1 -C 4 -alkyl radical which is composed of a carbon radical and, for example, a C 1 -C 4 -alkyl radical described above
  • Z 5 is oxygen, sulfur or a radical NR A 8 .
  • Preferred structural elements A are composed of at least one preferred radical of the radicals belonging to the structural element A, while the remaining radicals are widely variable.
  • Particularly preferred structural elements A are composed of the preferred radicals of the structural element A.
  • the spacer structural element E is understood as meaning a structural element that consists of a branched or unbranched aliphatic C 2 -C 3 0-hydrocarbon radical which is optionally substituted and contains heteroatoms and/or of a 4-to 20-membered aliphatic or aromatic mono- or polycyclic hydrocarbon radical which is optionally substituted and contains heteroatoms.
  • the spacer structural element E is composed of two to four substructural elements, selected from the group consisting of E 1 and E 2 , where the sequence of linkage of the substructural elements is arbitrary and E 1 and E 2 have the following meanings:
  • the coefficient c is preferably 0 or 1
  • the coefficient d is preferably 1 or 2
  • the coefficients f, g, h independently of one another are preferably 0 or 1
  • K 6 is preferably 0.
  • An optionally substituted 4- to 11-membered mono- or polycyclic aliphatic or aromatic hydrocarbon which can contain up to 6 double bonds and up to 6 identical or different heteroatoms selected from the group consisting of N, O, S, where the ring carbons or ring nitrogens can optionally be substituted, for Q E and X E independently of one another is preferably understood as meaning optionally substituted arylene, such as optionally substituted phenylene or naphthylene or optionally substituted hetarylene such as the radicals
  • XE and QE independently of one another are optionally substituted phenylene, a radical
  • RE 20 is, independently of one another, hydrogen, a branched or unbranched, optionally substituted C 1 -C 6 -alkyl, C 1 -C 6 -alkoxyalkyl, C 3 -C 12 -alkynyl, CO—C 1 -C 6 -alkyl, C0-O—C 1 -C 6 -alkyl or SO 2 —C 1 -C 6 -alkyl radical or an optionally substituted C 3 -C 7 -cycloalkyl, aryl, arylalkyl CO—O-alkylenearyl, CO-alkylenearyl, CO-aryl, SO 2 -aryl, hetaryl, CO-hetaryl or SO 2 alkylenearyl radical, preferably hydrogen or a branched or unbranched, optionally substituted C 1 -C 6 -alkyl radical.
  • a branched or unbranched, optionally substituted C 1 -C 6 -alkyl, C 2 -C 6 -alkenyl or C 2 -C 6 -alkynyl radical or an optionally substituted C 3 -C 7 -cycloalkyl, aryl, arylalkyl, hetaryl or hetarylalkyl radical for RE 13 , RE 14 or RE 15 independendy of one another is understood as meaning, for example, the corresponding radicals described above for RG 7 .
  • a branched or unbranched, optionally substituted C 1 -C 4 -alkoxy radical for RE 13 or RE 14 independently of one another is understood as meaning, for example, the C 1 -C 4 -alkoxy radicals described above for RA 14 .
  • Preferred alkylenecycloalkyl radicals for RE 13 1 RE 14 or RE 15 independently of one another are, for example, the C 1 -C 4 -alkylene-C 3 -C 7 -cycloalkyl radicals described above for RG 7 .
  • a branched or unbranched, optionally substituted C 1 -C 6 -alkyl, C 2 -C 6 -alkenyl, C 2 -C 6 -alkynyl or alkylenecycloalkyl radical or an optionally substituted C 3 -C 7 -cycloalkyl, aryl, arylalkyl, hetaryl or hetarylalkyl radical for RE 1 , RE 2 , RE 3 , RE 4 , RE 5 , RE 6 , RE 7 , RE 8 , RE 9 or RE 10 independently of one another is understood as meaning, for example, the corresponding radicals mentioned above for R G 7 .
  • two radicals RE 3 and RE 4 or RE 5 and RE 6 or RE 7 and RE 8 or RE 9 and RE 10 can in each ease independently of one another together form a 3- to 7-membered, optionally substituted, saturated or unsaturated carbo- or heterocycle, which can contain up to three heteroatoms from the group consisting of O, N and S.
  • the radical —(CH2)x—(WE) Z-RE 17 is composed of a CO—C 4 -alkylene radical, optionally a bonding element WE selected from the group —CO—, —CO—N(R 2 )—, —N(R 12 )—CO—, —N(RW 2 )—CO—N(RW 2 *)-, —N(R 2 )—CO—O—, —O—, —S—, —SO 2 —, —SO 2 —N(RW 2 )—, —SO 2 —O—, —CO—O—, —O—CO—O—CO—N(RW 2 )—, —N(RW 2 )— or —N(RW 2 )—SO 2 —, preferably selected from the group —CO—N(RW 2 )—, —N(RW 2 )—CO—, —O—, —SO 2 —N(RW 2 )—,
  • RE 17 and RW 2 or RW 2 * can together form a saturated or unsaturated C 3 -C 7 -heterocycle which can optionally contain up to two further heteroatoms selected from the group consisting of O, S and N.
  • the radicals RE 17 and RW 2 or RW 2 * together form a cyclic amine as the C 3 -C 7 -heterocycle in the case where the radicals are bonded to the same nitrogen atom, such as N-pyrrolidinyl, N-hexahydroazepinyl, N-morpholinyl or N-piperazinyl where in heterocycles which carry free amine protons, such as N-piperazinyl, the free amine protons can be replaced by customary amine protective groups, such as methyl, benzyl, Boc (tert-butoxycarbonyl), Z (benzyloxycarbonyl), tosyl, —SO 2 —C 1 -C 4 -alkyl, —SO 2 -phenyl or —SO 2 -benzyl.
  • customary amine protective groups such as methyl, benzyl, Boc (tert-butoxycarbonyl), Z (benzyloxycarbonyl), tosyl, —SO 2
  • Preferred radicals for RE 1 , RE 2 , RE 3 , RE 4 , RE 5 , RE 6 , RE 7 , RE 8 , RE 9 or RE 10 are independently of one another hydrogen, halogen, a branched or unbranched, optionally substituted C 1 -C 6 -alkyl radical, optionally substituted aryl or the radical —(CH 2 )x—(WE)z—RE 17 .
  • radicals for RE 1 , RE 2 , RE 3 , RE 4 , RE 5 , RE 6 , RE 7 , RE 8 , RE 9 or RE 10 are independently of one another hydrogen, F, a branched or unbranched, optionally substituted C 1 -C 4 -alkyl radical, in particular methyl.
  • a branched or unbranched, optionally substituted C 1 -C 6 -alkoxyalkyl, C 2 -C 6 -alkenyl, C 2 -C 12 -alkynyl or arylalkyl radical or an optionally substituted aryl, hetaryl or C 3 -C 7 -cycloalkyl for RE 11 and RE 11 * in structural element E independently of one another is understood as meaning, for example, the corresponding radicals described above for RG 7 .
  • the branched or unbranched, optionally substituted radicals CO—C 1 -C 6 -alkyl, CO—O—C 1 -C 6 -alkyl, CO—NH—C 1 -C 6 -alkoxalkyl, CO—NH—C 1 -C 6 -alkyl or SO 2 —C 1 -C 6 -alkyl radical or the optionally substituted radicals Co-O-alkylenearyl, CO—NH-alkylenearyl, CO-alkylenearyl, CO-aryl, CO—NH-aryl, SO 2 -aryl, CO-hetaryl, SO 2 -alkylenearyl, SO 2 -hetaryl or SO 2 -alkylenehetaryl for RE 11 and RE 11 * independently of one another are composed, for example, of the corresponding groups CO, COO, CONH or SO 2 and the corresponding radicals mentioned above.
  • Preferred radicals for RE 11 or RE 11 * are independently of one another hydrogen, a branched or unbranched, optionally substituted C 1 -C 6 -alkyk, C 1 -C 6 -alkoxy, C 2 -C 6 -alkenyl,
  • radicals for RE 11 or RE 11 * are hydrogen, methyl, cyclopropyl, allyl or propargyl.
  • structural element E 1 is a radical —CH 2 —CH 2 —CO—, —CH 2 —CH 2 —CH 2 —CO— or a C 1 -C 5 -alkylene radical.
  • the spacer structural element E used is a structural element of the formula I E1E2
  • Preferred structural elements E are composed of at least one preferred radical of the radicals belonging to structural element E, while the remaining radicals are widely variable.
  • Particularly preferred structural elements E are composed of the preferred radicals of structural element E.
  • Preferred structural elements B are composed either of the preferred structural element A, while E is widely variable or of the preferred structural element E, while A is widely variable.
  • the compounds of the formula I, and also the intermediates for their preparation, can have one or more asymmetric substituted carbon atoms.
  • the compounds can be present as pure enantiomers or pure diastereomers or as a mixture thereof.
  • the use of an enantiomerically pure compound as the active compound is preferred.
  • the compounds of the formula I can also be present in other tautomeric forms.
  • the compounds of the formula I can also be present in the form of physiologically tolerable salts.
  • the compounds of the formula I can also be present as prodrugs in a form in which the compounds of the formula I are liberated under physiological conditions.
  • group T in structural element L which in some cases contains groups which are hydrolyzable to the free carboxylic acid group under physiological conditions.
  • Derivatized structural elements B or A are also suitable which liberate the structural element B or A respectively under physiological conditions.
  • compounds of the formula I in each case two of the three structural elements B, G or L have the preferred range, while the remaining structural elements are widely variable.
  • Preferred compounds of the formula I contain, for example, the preferred structural element G, while the structural elements B and L are widely variable.
  • H Abbreviation edia2 pyma2 pipa2 aepi2 me35thima2 dibema2 edia3 buta aaf 42thiaz2 chex2 bam2 apma2 pdagk mepipe2 prodia2 inda2 35thima2 me25thima2 penta aof hexn mea2 pipeme2 me42thiaz2 a23thima2 a24thima2
  • G Abbreviation 4phaz 3bzlaz mophaz pipmaz cpec mpphec amec dbc pclphec thec mmophec 3bec phaz pymaz 3ipec phec ppec mmphec amaz 4bec dmaphec 4pec pmophec 7eme
  • the compounds of the formula I and the starting substances used for their preparation can generally be prepared by methods of organic chemistry known to the person skilled in the art, such as are described in standard works such as Houben-Weyl (ed.). “Methoden der Organischen Chemie”[Methods of Organic Chemistry], Thieme-Verlag, Stuttgart, Taylor (ed.), “The Chemistry of Heterocyclic Compounds”, Wiley & Sons, New York, or March “Advanced Organic Chemistry”, 4 th Edition, Wiley & Sons. Further methods of preparing specific functional groups are also described in R.
  • the invention relates to novel integrin inhibitors of the formula I
  • the invention relates to novel peptides, of the formula I
  • the present invention also relates to the pharmaceutically usable pro-drugs, derivatives, solvates and stereoisomors of the compounds of the formula I and the salts thereof, including mixtures thereof in all ratios.
  • the invention relates, in particular, to peptidic compounds selected from the group of the formulae Ia)-I o)
  • amino acid radicals stand for the radicals of the following amino acids:
  • amino acids can occur in a plurality of enantiomeric forms, all these forms and also mixtures thereof (for example the DL forms) are included above and below. Furthermore, the amino acids may be provided with corresponding protecting groups known per se.
  • the present invention comprises a class of biphenyl integrin antagonists.
  • the present invention relates to a class of compounds represented by the Formula I:
  • the invention comprises pharmaceutical compositions comprising compounds of the Formula I.
  • Such compounds and compositions are use in selectively inhibiting or antagonizing the ⁇ v ⁇ 3 and/or ⁇ b ⁇ s integrins and therefore in another embodiment the present invention relates to a method of selectively inhibiting or antagonizing the ⁇ v ⁇ 3 and/or ⁇ v ⁇ s integrin.
  • the invention provides methods of treating or inhibiting pathological conditions associated therewith such as osteoporosis, humoral hypercalcemia of malignancy, Paget's disease, rumor metastasis, solid tumor growth (neoplasia), angiogenesis, including tumor angiogenesis, retinopathy including macular degeneration and diabetic retinopathy, arthritis including rheumatoid arthritis and osteoarthritis, periodontal disease, psoriasis, smooth muscle cell migration and restenosis in a mammal in need of such treatment.
  • pathological conditions associated therewith such as osteoporosis, humoral hypercalcemia of malignancy, Paget's disease, rumor metastasis, solid tumor growth (neoplasia), angiogenesis, including tumor angiogenesis, retinopathy including macular degeneration and diabetic retinopathy, arthritis including rheumatoid arthritis and osteoarthritis, periodontal disease, psoriasis, smooth muscle cell migration and restenosis in a
  • the compounds of this invention are 1) ⁇ v ⁇ 3 integrin antagonists; or 2) ⁇ v ⁇ s integrin antagonists; or 3) mixed or dual ⁇ v ⁇ 3 / ⁇ v ⁇ s antagonists.
  • the present invention includes compounds which inhibit the respective integrins and also includes pharmaceutical compositions comprising such compounds.
  • the present invention further provides for methods for treating or preventing conditions mediated by the ⁇ v ⁇ 3 and/or ⁇ v ⁇ s receptors in a mammal in need of such treatment comprising administering a therapeutically effective amount of the compounds of the present invention and pharmaceutical compositions of the present invention.
  • Administration of such compounds and compositions of the present invention inhibits angiogenesis, tumor metastasis, tumor growth, osteoporosis, Paget's disease, humoral hypercalcemia of malignancy, retinopathy, macular degeneration, arthritis, periodontal disease, smooth muscle cell migration, including restenosis and artherosclerosis, and viral diseases.
  • the compounds of the present invention further show greater selectivity for the ⁇ v ⁇ 3 and/or ⁇ v ⁇ s integrin than for the ⁇ v ⁇ 6 integrin. It has been found that the selective antagonism of the ⁇ v ⁇ 3 integrin is desirable in that the ⁇ v ⁇ 6 integrin may play a role in normal physiological processes of tissue repair and cellular turnover that routinely occur in the skin and pulmonary tissue, and the inhibition of this function can be deleterious. Therefore, compounds of the present invention which selectively inhibit the ⁇ v ⁇ 3 integrin as opposed to the ⁇ v ⁇ 6 integrin have reduced side-effects associated with inhibition of the ⁇ v ⁇ 6 integrin.
  • the present invention relates to a class of compounds represented by the Formula I.
  • the compounds according to Formula I can exist in various isomers, enantiomers, tautomers, racemates and polymorphs, and all such forms are meant to be included.
  • Such compounds and compositions are useful in selectively inhibiting or antagoniing the ⁇ v ⁇ 3 and/or ⁇ v ⁇ 3 integrins and therefore in another embodiment the present invention relates to a method of selectively inhibiting or antagonizing the ⁇ v ⁇ 3 and/or ⁇ v ⁇ 5 integrin.
  • the invention further involves treating or inhibiting pathological conditions associated therewith such as osteoporosis, humoral hypercalcemia of malignancy, Paget's disease, tumor metastasis, solid tumor growth (neoplasia), angiogenesis, including tumor angiogenesis, retinopathy including macular degeneration and diabetic retinopathy, arthritis, including rheumatoid arthritis, periodontal disease, psoriasis, smooth muscle cell migration and restenosis in a mammal in need of such treatment. Additionally, such pharmaceutical agents are useful as to antiviral agents, and antimicrobials.
  • pathological conditions associated therewith such as osteoporosis, humoral hypercalcemia of malignancy, Paget's disease, tumor metastasis, solid tumor growth (neoplasia), angiogenesis, including tumor angiogenesis, retinopathy including macular degeneration and diabetic retinopathy, arthritis, including rheumatoid arthritis, periodontal disease, psoriasis, smooth muscle cell migration
  • the compounds of this invention are 1) ⁇ v ⁇ 3 integrin antagonists; or 2) ⁇ v ⁇ 5 integrin antagonists; or 3) mixed or dual ⁇ v ⁇ 3 / ⁇ v ⁇ 5 antagonists.
  • the present invention includes compounds which inhibit the respective integrins and also includes pharmaceutical compositions comprising such compounds.
  • the present invention further provides for methods for treating or preventing conditions mediated by the ⁇ v ⁇ 3 and/or ⁇ v ⁇ 5 receptors in a mammal in need of such treatment comprising administering a therapeutically effective amount of the compounds of the present invention and pharmaceutical compositions of the present invention.
  • Administration of such compounds and compositions of the present invention inhibits angiogenesis, tumor metastasis, tumor growth, osteoporosis, Paget's disease, humoral hypercalcemia of malignancy, retinopathy, macular degeneration, arthritis, periodontal disease, smooth muscle cell migration, including restenosis and artherosclerosis, and viral diseases.
  • the compounds of the present invention further show greater selectivity for the ⁇ v ⁇ 3 and/or ⁇ v ⁇ 5 integrin than for the ⁇ v ⁇ 6 integrin, it has been found that the selective antagonism of the ⁇ v ⁇ 3 integrin is desirable in that the ⁇ v ⁇ 6 integrin may play a role in normal physiological processes of tissue repair and cellular turnover that routinely occur in the skin and pulmonary tissue, and the inhibition of this function can be deleterious. Therefore, compounds of the present invention which selectively inhibit the ⁇ v ⁇ 3 integrin as opposed to the ⁇ v ⁇ 6 integrin have reduced side-effects associated with inhibtion of the ⁇ v ⁇ 6 integrin.
  • the present invention relates to a class of compounds represented by the Formula I.
  • the compounds according to Formula I can exist in various isomers, enantiomers, tautomers, racemates and polymorphs, and all such forms are meant to be included.
  • Such compounds and compositions are useful in selectively inhibiting or antagonizing the ⁇ v ⁇ 3 and/or ⁇ v ⁇ 5 integrins and therefore in another embodiment the present invention relates to a method of selectively inhibiting or antagonizing the ⁇ v ⁇ 3 and/or ⁇ v 5 integrin.
  • the invention further involves treating or inhibiting pathological conditions associated therewith such as osteoporosis, humoral hypercalcemia of malignancy, Paget's disease, tumor metastasis, solid tumor growth (neoplasia), angiogenesis, including tumor angiogenesis, retinopathy including macular degeneration and diabetic retinopathy, arthritis, including rheumatoid arthritis, periodontal disease, psoriasis, smooth muscle cell migration and restenosis in a mammal in need of such treatment. Additionally, such pharmaceutical agents are useful as antiviral agents, and antimicrobials.
  • the compounds of this invention are 1) ⁇ v ⁇ 3 integrin antagonists; or 2) ⁇ v ⁇ 5 integrin antagonists; or 3) mixed or dual ⁇ v ⁇ 3 / ⁇ v ⁇ 5 antagonists.
  • the present invention includes compounds which inhibit the respective integrins and also includes Pharmaceutical compositions comprising such compounds.
  • the present invention further provides for methods for treating or preventing conditions mediated by the ⁇ v ⁇ 3 and/or ⁇ v ⁇ 3 receptors in a mammal in need of such treatment comprising administering a therapeutically effective amount of the compounds of the present invention and pharmaceutical compositions of the present invention.
  • Administration of such compounds and compositions of the present invention inhibits angiogenesis, tumor metastasis, tumor growth, osteoporosis, Paget's disease, humoral hypercalcemia of malignancy, retinopathy, macular degeneration, arthritis, periodontal disease, smooth muscle cell migration, including restenosis and artherosclerosis, and viral diseases.
  • the selective antagonism of the ⁇ v ⁇ 3 integrin is desirable in that the ⁇ v ⁇ 6 integrin may play a role in normal physiological processes of tissue repair and cellular turnover that routinely occur in the skin and pulmonary tissue, and ⁇ v ⁇ 8 may play a role in the regulation of growth in the human pathway. Therefore, compounds which selectively inhibit the ⁇ v ⁇ 3 integrin as opposed to the ⁇ v ⁇ 6 and/or the ⁇ v ⁇ 8 integrin have reduced side-effects associated with inhibition of the ⁇ v ⁇ 6 and/or the ⁇ v ⁇ 8 integrin.
  • the present invention relates to a class of compounds represented by the Formula 1, 2
  • heteroaryls include the ring systems described above.
  • the substituents X 4 and X 5 are selected from the group consisting of H, alkyl, branched alkyl, alkylamino, alkoxyalkylamino, haloalkyl, thioalkyl, halogen, amino, alkoxy, aryloxy, alkoxyalkyl, hydroxy, cyano or acylamino groups.
  • the substituents X4 and X 5 can be methyl, methoxy, amine, methylamine, trifluoromethyl, dimethylamine, hydroxy, chloro, bromo, fluoro and cyano.
  • X 6 may preferentially be H, alkyl, hydroxy, halogen, alkoxy and haloalkyl.
  • the pyridyl ring can be fused with a 4-8 membered ring, optionally saturated or unsaturated.
  • Some examples of these ring systems include tetrahydronaphthyridine, quinoline, tetrahydroquinoline, azaquinoline, morpholinopyridine, imidazo-pyridine and the like.
  • the monocyclic ring systems such as imidazole, thiazole, oxazole, pyrazole, and the like, may contain an amino or alkylamino substituent at any position within the ring.
  • the linkage A 1 -Z 2 of Formula I includes the heterocycle derived ring systems such as: pyridine, imidazole, thiazole, oxazole, benzimidazole, imidazopyridine and the like.
  • heterocycles for A 1 -Z 2 of the present invention include
  • the compounds of the present invention further show greater selectivity for the ⁇ v ⁇ 3 and/or ⁇ v ⁇ 5 integrin than for the ⁇ v ⁇ 6 integrin. It has been found that the selective antagonism of the ⁇ v ⁇ 3 integrin is desirable in that the ⁇ v ⁇ 6 integrin may play a role in normal physiological processes of tissue repair and cellular turnover that routinely occur in the skin and pulmonary tissue, and the inhibition of this function can be deleterious (Huang et al., Am J Respir Cell Mol Biol 1998, 19(4): 636-42). Therefore, compounds of the present invention which selectively inhibit the ⁇ v ⁇ 3 integrin as opposed to the ⁇ v ⁇ 6 integrin have reduced side effects associated with inhibition of the ⁇ v ⁇ 6 integrin.
  • the compounds of the present invention comprise the R-isomers of the carbon of the beta amino acid.
  • Other isomers may result from additional chiral centers, depending on the substitution of the parent structure.
  • the present invention relates to a class of compounds represented by the Formula I:
  • compositions comprising compounds of the Formula I.
  • Such compounds and compositions are useful in selectively inhibiting or antagonizing the ⁇ v ⁇ 3 and/or ⁇ v ⁇ 5 , integrins and therefore in another embodiment the present invention relates to a method of selectively inhibiting or antagonizing the ⁇ v ⁇ 3 and/or ⁇ v ⁇ 5 integrin.
  • the invention further embodies treating or inhibiting pathological conditions associated therewith such as osteoporosis, humoral hypercalcemia of malignancy, Paget's disease, tumor metastasis, solid tumor growth (neoplasia), angiogenesis, including tumor angiogenesis, retinopathy including macular degeneration and diabetic retinopathy, arthritis, including rheumatoid arthritis and osteoarthritis, periodontal disease, psoriasis, smooth muscle cell migration and restenosis in a mammal in need of such treatment.
  • pathological conditions associated therewith such as osteoporosis, humoral hypercalcemia of malignancy, Paget's disease, tumor metastasis, solid tumor growth (neoplasia), angiogenesis, including tumor angiogenesis, retinopathy including macular degeneration and diabetic retinopathy, arthritis, including rheumatoid arthritis and osteoarthritis, periodontal disease, psoriasis, smooth muscle cell migration and restenosis in a ma
  • the compounds of this invention are 1) ⁇ v ⁇ 3 integrin antagonists; or 2) ⁇ v ⁇ 5 integrin antagonists; or 3) mixed or dual ⁇ v ⁇ 3 / ⁇ v ⁇ 5 antagonists.
  • the present invention includes compounds which inhibit the respective integrins and also includes pharmaceutical compositions comprising such compounds.
  • the present invention further provides for methods for treating or preventing conditions mediated by the ⁇ v ⁇ 3 and/or 60 v ⁇ 5 receptors in a mammal in need of such treatment comprising administering a therapeutically effective amount of the compounds of the present invention and pharmaceutical compositions of the present invention.
  • Administration of such compounds and compositions of the present invention inhibits angiogenesis, tumor metastasis, tumor growth, osteoporosis, Paget's disease, humoral hypercalcemia of malignancy, retinopathy, macular degeneration, arthritis, periodontal disease, smooth muscle cell migration, including restenosis and artherosclerosis, and viral diseases.
  • the compounds of the present invention further show greater selectivity for the ⁇ v ⁇ 3 and/or ⁇ v ⁇ 5 integrin than for the ⁇ v ⁇ 6 integrin. It has been found that the selective antagonism of the ⁇ v ⁇ 3 integrin is desirable in that the ⁇ v ⁇ 6 integrin may play a role in normal physiological processes of tissue repair and cellular turnover that routinely occur in the skin and pulmonary tissue, and the inhibition of this function can be deleterious. Therefore, compounds of the present invention which selectively inihibit the ⁇ v ⁇ 3 integrin as opposed to the ⁇ v ⁇ 6 integrin have reduced side-effects associated with inhibition of the ⁇ v ⁇ 6 integrin.
  • the present invention relates to a class of compounds represented by the Formula I
  • the compounds of this invention are 1) ⁇ v ⁇ 3 integrin antagonists; or 2) ⁇ v ⁇ 5 integrin antagonists; or 3) mixed or dual ⁇ v ⁇ 3 / ⁇ v ⁇ 5 antagonists.
  • the present invention includes compounds which inhibit the respective integrins and also includes pharmaceutical compositions comprising such compounds.
  • the present invention further provides for methods for treating or preventing conditions mediated by the ⁇ v ⁇ 3 and/or ⁇ v ⁇ 5 receptors in a mammal in need of such treatment comprising administering a therapeutically effective amount of the compounds of the present invention and pharmaceutical compositions of the present invention.
  • Administration of such compounds and compositions of the present invention inhibits angiogenesis, tumor metastasis, tumor growth, osteoporosis, Paget's disease, humoral hypercalcemia of malignancy, retinopathy, macular degeneration, arthritis, periodontal disease, smooth muscle cell migration, including restenosis and artherosclerosis, and viral diseases.
  • the selective antagonism of the ⁇ v ⁇ 3 integrin is desirable in that the ⁇ v ⁇ 6 integrin may play a role in normal physiological processes of tissue repair and cellular turnover that routinely occur in the skin and pulmonary tissue, and ⁇ v ⁇ 8 may play a role in the regulation of growth in the human pathway. Therefore, compounds which selectively inihibit the ⁇ v ⁇ 3 integrin as opposed to the ⁇ v ⁇ 6 and/or the ⁇ v ⁇ 8 integrin have reduced side-effects associated with inhibition of the ⁇ v ⁇ 6 and/or the ⁇ v ⁇ 8 integrin.
  • the present invention relates to a class of compounds represented by the Formula I
  • a 1 is a 5-9 membered monocyclic ring or 7-12 membered bicyclic heterocycle ring of the formula
  • X or Y are independently selected from the group consisting of —CR g — or —N— wherein R g is selected from the group consisting of H, alkyl, haloalkyl, fluoro, alkoxyalkyl, alkynyl, aryl, heteroaryl, aralkyl, alkylsulfone, heteroaralkyl, hydroxy, alkoxy, hydroxyalkyl, and carboxyalkyl;
  • the group X—X 2 —Y optionally contains a moiety selected from the group consisting of acyl, alkyl, amino, ether, thioether, sulfone, and olefin:
  • Such compounds and compositions are useful in selectively inhibiting or antagonizing the ⁇ v ⁇ 3 and/or ⁇ v ⁇ 5 integrin(s) and therefore in another embodiment the present invention relates to a method of selectively inhibiting or antagonizing the ⁇ v ⁇ 3 and/or ⁇ v ⁇ 5 integrin(s).
  • the invention further involves treating or inhibiting pathological conditions associated therewith such as osteoporosis, immoral hypercalcemia of malignancy, Paget's disease, tumor metastasis, solid tumor growth (neoplasia), angiogenesis, including tumor angiogenesis, retinopathy including macular degeneration and diabetic retinopathy, arthritis, including rheumatoid arthritis. periodontal disease, psoriasis, smooth muscle cell migration including restenosis or atherosclerosis in a mammal in need of such treatment. Additionally, such pharmaceutical agents are useful as antiviral agents, antifungals and antimicrobials. The compounds of the present invention may be used alone or in combination with other pharmaceutical agents.
  • the compounds of this invention are 1) ⁇ v ⁇ 3 integrin antagonists; or 2) ⁇ v ⁇ 5 integrin antagonists; or 3) mixed or dual ⁇ v ⁇ 3 / ⁇ v ⁇ 5 antagonists.
  • the present invention includes compounds which inhibit the respective integrins and also includes pharmaceutical compositions comprising such compounds.
  • the present invention further provides for methods for treating or preventing conditions mediated by the ⁇ v ⁇ 3 and/or ⁇ v ⁇ 5 receptors in a mammal in need of such treatment comprising, administering a therapeutically effective amount of the compounds of the present invention and pharmaceutical compositions of the present invention.
  • Administration of such compounds and compositions of the present invention inhibits angiogenesis, tumor metastasis, tumor growth, osteoporosis, Paget's disease, humoral hypercalcemia of malignancy, retinopathy, macular degeneration, arthritis, periodontal disease, smooth muscle cell migration, including restenosis and artherosclerosis, and viral diseases.
  • the compounds of the present invention further show greater selectivity for the ⁇ v ⁇ 3 and/or ⁇ v ⁇ 5 integrin than for the ⁇ v ⁇ 6 integrin. It has been found that the selective antagonism of the ⁇ v ⁇ 3 integrin is desirable in that the ⁇ v ⁇ 6 integrin may play a role in normal physiological processes of tissue repair and cellular turnover that routinely occur in the skin and pulmonary tissue, and the inhibition of this function can be deleterious.
  • the present invention relates to a class of compounds represented by the Formula I.
  • Such compounds and compositions are useful in selectively inhibiting or antagonizing the ⁇ v ⁇ 3 and/or ⁇ v ⁇ 5 integrins and therefore in another embodiment the present invention relates to a method of selectively inhibiting or antagonizing the ⁇ v ⁇ 3 and/or ⁇ v ⁇ 5 integrin.
  • the invention further involves treating or inhibiting pathological conditions associated therewith such as osteopbrosis, humoral hypercalcemia of malignancy, Paget's disease, tumor metastasis, solid tumor growth (neoplasia), angiogenesis, including tumor angiogenesis, retinopathy including macular degeneration and diabetic retinopathy, arthritis, including rheumatoid arthritis, periodontal disease, psoriasis, smooth muscle cell migration and restenosis in a mammal in need of such treatment.
  • pathological conditions associated therewith such as osteopbrosis, humoral hypercalcemia of malignancy, Paget's disease, tumor metastasis, solid tumor growth (neoplasia), angiogenesis, including tumor angiogenesis, retinopathy including macular degeneration and diabetic retinopathy, arthritis, including rheumatoid arthritis, periodontal disease, psoriasis, smooth muscle cell migration and restenosis in a mammal in need of such treatment.
  • the present invention as a first object provides compounds of the following formula (I)
  • R1 is selected from the group consisting of H, C 1 -C 4 alkyl, C 1 -C 4 alkoxy, OH, halogen, and CF 3 ;
  • the present invention includes within its scope all possible isomers, stereoisomers and optical isomers and their mixtures, and the bioprecursors or metabolites of the compounds of formula (I).
  • Such compounds and compositions are useful in selectively inhibiting or antagonizing the ⁇ v ⁇ 3 integrin and therefore in another embodiment the present invention relates to a method of selectively inhibiting or antagonizing the ⁇ v ⁇ 3 integrin.
  • the invention further involves treating or inhibiting pathological conditions associated therewith such as osteoporosis, humoral hypercalcemia of malignancy, Paget's disease, tumor metastasis, solid tumor growth (neoplasia), angiogenesis, including tumor angiogenesis, retinopathy including macular degeneration and diabetic retinopathy, arthritis, including rheumatoid arthritis, periodontal disease, psoriasis, smooth muscle cell migration and restenosis in a mammal in need of such treatment. Additionally, such pharmaceutical agents are useful as antiviral agents, and antimicrobials.
  • the compounds of this invention include 1) ⁇ v ⁇ 3 integrin antagonists; or 2) ⁇ v ⁇ 5 integrin antagonists; or 3) mixed or dual ⁇ v ⁇ 3 / ⁇ v ⁇ 5 antagonists.
  • the present invention includes compounds which inhibit the respective integrins and also includes pharmaceutical compositions comprising such compounds.
  • the present invention further provides for methods for treating or preventing conditions mediated by the ⁇ v ⁇ 3 and/or ⁇ v ⁇ 5 receptors in a mammal in need of such treatment comprising administering a therapeutically effective amount of the compounds of the present invention and pharmaceutical compositions of the present invention.
  • compositions of the present invention inhibits angiogenesis, tumor metastasis, tumor growth, skeletal malignancy of breast cancer, osteoporosis, Paget's disease, humoral hypercalcemia of malignancy, retinopathy, macular degeneration, arthritis including rheumatoid, periodontal disease, smooth muscle cell migration,
  • the compounds of the present invention can be used, alone or in combination with other therapeutic agents, in the treatment or modulation of various conditions or disease states described above.
  • the compounds of the present invention include selective antagonists of ⁇ v ⁇ 3 over ⁇ v ⁇ 3 .
  • the present invention relates to a class of compounds represented by Formula I
  • the ring A may further contain a carboxamide, sulfone, sulfonamide or an acyl group.
  • the group X-Y can contain a moiety selected from the group consisting of acyl, alkyl, sulfonyl, amino, ether, thioether, carboxamido, sulfonamido, aminosulfonyl and olefins;
  • the compounds of the present invention comprise novel heteroarylalkanoic integrin antagonists.
  • the present invention relates to the following compounds:
  • the present invention may also include the following compounds:
  • heteroaryls include the ring systems described above.
  • the substituents X 4 and X 5 are selected from the group consisting of H, alkyl, branched alkyl, alkylamino, alkoxyalkylamino, haloalkyl, thioalkyl, halogen, amino, alkoxy, aryloxy, alkoxyalkyl, hydroxy, cyano or acylamino groups.
  • the substituents X 4 and X 5 can be methyl, methoxy, amine, methylamine, trifluoromethyl, dimethylamine, hydroxy, chloro, bromo, fluoro and cyano.
  • X 6 may preferentially be H, alkyl hydroxy, halogen, alkoxy and haloalkyl.
  • the pyridyl ring can be fused with a 4-8 membered ring, optionally saturated or unsaturated.
  • Some examples of these ring systems include tetrahydronaphthyridine, quinoline, tetrahydroquinoline, azaquinoline, morpholinopyridine, imidazopyridine and the like.
  • the monocyclic ring systems such as imidazole, thiazole, oxazole, pyrazole, and the like, may contain an amino or alkylamino substituent at any position within the ring.
  • the linkage A 1 -Z 2 of Formula I includes the heterocycle derived ring systems such as: pyridine, imidazole, thiazole, oxazole, benzimidazole, imidazopyrdine and the like.
  • heterocycles for A 1 -Z 2 of the present invention include
  • Y 3 or Y 4 is an aryl or a heteroaryl group selected from phenyl, benzofuran, benzothiophene, indole, quinoline, isoquinoline, benzimidazole, benzoxazole, 1,3-benzodioxole, 1,4-benzodioxane, benzopyran, quinolone, imidazopyridine, tetrahydro-quinoline, benzotriazole, dihydroindole, dihydrobenzofuran, furan, thiophene, phenyl, oxazole, thiazole, isoxazole, pyrazole, imidazole, pyrrole, pyridine, pyrimidine, pyridone, triazole, thiadiazole and the like.
  • the aryl system can be optionally substituted at one or more positions with alkyl, alkoxy, hydroxy, cyano, halogen or haloal
  • Y 3 or Y 4 may be an amine, alkylamine, acylamine, aminosulfone (NHSO 2 R), arylamine, alkoxyalkylamine, aralkylamine, or heterocyclic amine.
  • Y 3 taken together with Y 4 forms a 3-8 membered monocyclic or 7-11 membered bicyclic ring B,
  • X taken together with Y 3 forms a 3-7 membered monocyclic ring C
  • the invention further relates to pharmaceutical compositions containing therapeutically effective amounts of the compounds of Formula I.
  • the present invention as a first object provides novel non-peptide compounds having the following formula (I)
  • the present invention includes within its scope all possible isomers, stereoisomers and optical isomers and their mixtures, and the metabolites and the metabolic precursors or bioprecursors of the compounds of formula (I).
  • a halogen atom is preferably chlorine or fluorine.
  • alkyl, alkoxy, alkenyl and alkynyl groups and the alkylene and alkenylene chains may be branched or straight groups or chains, respectively.
  • a C 5 -C 7 monocyclic heteroaryl ring is preferably a C 5 -C 6 heteromonocyclic ring, in particular selected from pyridine, pyrazine, pyridazine, pyrimidine, thiophene, pyrrole, pyrazole, imidazole, oxazole and isoxazole.
  • An aryl group is e.g., an aromatic C 6 -C 20 mono- or poly-nuclear moiety, typically phenyl, unsubstituted or substituted by one to three substituents independently chosen from halogen, hydroxy, CF 3 , C 1 -C 4 alkyl and C 1 -C 4 alkoxy.
  • an aralkyl group is e.g. benzyl or phenethyl, in which the phenyl ring is optionally substituted by one to three substituents independently chosen from halogen, hydroxy, CF 3 , C 1 -C 4 alkyl and C 1 -C 4 alkoxy.
  • a C 2 -C 4 alkenyl group is preferably an allyl group.
  • a C 1 -C 6 alkyl group is preferably as C 1 -C 4 alkyl group.
  • a C 1 -C 4 alkyl group is preferably a methyl or ethyl group.
  • a C 2 -C 4 alkynyl group is preferably an ethynyl group.
  • a C 2 -C 4 alkoxy group is preferably methoxy, ethoxy, propoxy and butoxy.
  • Examples of pharmaceutically acceptable salts of the compounds of the invention are either those with inorganic bases, such as sodium, potassium, calcium and aluminium hydroxides, or with organic bases, such as lysine, arginine, N-methyl-glucamine, triethylamine, triethanolamine, dibenzylamine, methylbenzylamine, di-(2-ethyl-hexyl)-amine, piperidine, N-ethylpiperidine, N,N-diethylaminoethylamine, N-ethylmorpholine, ⁇ -phenethylamine, N-benzyl- ⁇ -phenethylamine, N-benzyl-N,N-dimethylamine and the other acceptable organic amines, as well as the salts with inorganic acids, e.g.
  • the present invention also includes within its scope pharmaceutically acceptable bio-precursors (otherwise known as pro-drugs) of the compounds of formula (I), i.e. compounds which have a different formula to formula (I) above, but which nevertheless upon administration to a human being are converted directly or indirectly in vivo into a compound of formula (I).
  • Preferred compounds of the invention are those wherein, in formula (I),
  • a further class of preferred compounds of the invention are those wherein, in formula (I),
  • a further object of the present invention is to provide a compound or formula (I)
  • Object of the present invention is also to provide a pharmaceutical composition
  • a pharmaceutical composition comprising a pharmaceutically acceptable carrier and/or diluent and as an active principle, a compound of formula (I), as herein defined, or a pharmaceutically acceptable salt thereof.
  • the present invention also provides the use of a compound of formula (I), a defined above, in the preparation of a medicament having ⁇ v ⁇ 3 integrin inhibiting or antagonizing activity.
  • the present invention also provides a method for treating conditions mediated by the ⁇ v ⁇ 3 integrin in a mammal, including humans, in need of such treatment comprising administering to said mammal an effective ⁇ v ⁇ 3 inhibiting or antagonizing amount of a compound of formula (I)
  • the present invention provides a method for inhibiting bone resorption, treating osteoporosis, inhibiting humoral hypercalcemia of malignancy, treating Paget's disease, inhibiting tumor metastasis, inhibiting neoplasia (solid tumor growth), inhibiting angiogenesis including tumor angiogenesis, treating diabetic retinopathy, inhibiting arthritis, psoriasis and periodontal disease, and inhibiting smooth muscle cell migration including restenosis.
  • the compounds of the invention and the salt thereof can be prepared by and analogy process. Accordingly, the compounds of formula I and the salts thereof, can be for instance obtained by a process which comprises:
  • the optional salification of a compound of formula (I) as well as the conversion of a salt into the free compound and the separation of a mixture of isomers into the single isomers may be carried out by conventional methods.
  • the separation of optical isomers may be carried out by salification with an optically active base or acid and by subsequent fractional crystallization of the diastereoisomeric salts, followed by recovering of the optically active isomeric-acids or, respectively, bases.
  • groups are present which need to be protected before submitting them to the here-above illustrated reactions, they may be protected before the reactions take place and then deprotected, according to well known methods in organic chemistry.
  • the compounds of formulae (I) and the pharmaceutically acceptable, salts thereof are herein defined as the “compounds of the present invention”, the “compounds of the invention” and/or the “active principles of the pharmaceutical compositions of the invention”.
  • the compound 3-( ⁇ 4-[(3- ⁇ [amino(imino)methyl]amino ⁇ benzoyl)amino]-phenyl ⁇ sulfanyl)-3-(3-pyridinyl)propanoic acid (internal code PNU 277262F) can be provided.
  • compound PNU 277362F can be synthesized as bis-trifluoroacetate salt, as depicted in scheme 1, according to the procedures reported.
  • the compound 3- ⁇ [4-( ⁇ 3-[(benzylamino)carbonyl])amino]phenyl ⁇ sulfanyl)-3—(3-pyridinyl) propanoic acid (internal code PHA 515442E) is provided.
  • the compound PNU 515442E can be synthesized as trifluoroacetate salts.

Abstract

The present invention is directed to methods of treating proteinuric diseases by the administration of avβ3 integrin inhibitors.

Description

    FEDERALLY SPONSORED RESEARCH
  • This invention was made with Government support under NIH grants DK 073495, DK 057683, DK 062472 and George M. O'Brien kidney Centex DK 064236. The Government has certain rights to this invention.
  • FIELD OF INVENTION
  • This invention relates generally to the treatment of proteinuric diseases by αvβ3 integrin inhibitors.
  • BACKGROUND OF INVENTION
  • Urinary protein loss (proteinuria) affects some 100 million people worldwide and is a feature of kidney dysfunction of glomerular origin and itself a risk factor for both renal and extra-renal diseases1. Kidney podocytes and their foot processes (FP) are a key component of the ultrafiltration system in the glomerulus where they comprise the filtration barrier together with endothelial cells and the glomerular basement membrane (GBM). Podocytes are located within the glomerulus of the kidney where they are attached to the GBM via α3β1 integrin2, 3, and α/β dystroglycan4. Podocyte FPs are interconnected by the slit diaphragm (SD), a modified adherens junction5. Proteinuric kidney diseases are typically associated with various degrees of podocyte membrane remodeling (FP effacement and/or SD disruption) driven by a rearrangement of the podocyte microfilament system6. Recent work has advanced our understanding of the molecular framework underlying podocyte structure largely through the analysis of hereditary proteinuria syndromes and genetic models7. A few studies suggest also mechanisms for the far more common acquired proteinuric diseases8, 9. Despite this progress, there are currently no cell-specific therapeutics for podocytes available. An emerging concept for the regulation of podocyte structure and function is the regulation of the podocyte cytoskeleton by proteases such as cathepsin L8, 10. Cathepsin L induction in podocytes is accompanied by an increase in cell motility of cultured podocytes10. The increased motility of in vitro podocytes10, 11 is best translated into FP dynamics in vivo where podocytes remain locally attached to the GBM but may have altered FP dynamics resulting in FP fusion. In some forms of inflammatory glomerular diseases such as crescentic glomerulonephritis, podocytes have been reported to move out of their microenvironment into areas of crescentic glomerular damage12. The concept of dynamic podocyte FPs dates far back into the 1970's where elegant studies of Seiler and colleagues have shown that infusion of polycations such as protamine sulfate can induce rapid changes in FP dynamics and FP effacement13. Moreover, this event could be largely reversed by the infusion of polyanions such as heparin13. Even so it is impossible to image FP dynamics continuously in live animals, results from above studies suggest a highly dynamic podocyte FP system. Moreover the electron-microscopical analysis of normal kidney commonly reveals small areas of FP effacement which probably represents FPs during transition. Cancer cell motility is another example where cells can be hyperdynamic or participate in tissue invasion14.
  • SUMMARY OF INVENTION
  • According to the invention methods for treating a proteinuric disorder are provided comprising administering to a patient in need thereof an avβ3 integrin inhibitor in an amount effective to reduce or eliminate the proteinuric disorder. In one aspect of the invention the proteinuric disorder is a kidney proteinuric disorder. In one aspect of the invention the kidney proteinuric disorder is selected from a list comprising of:
      • Diabetic nephropathy,
      • Nephrotic syndromes (i.e. intrinsic renal failure),
      • Nephritic syndromes,
      • Toxic lesions of kidneys,
      • Glomerular diseases, such as membranous glomerulonephritis,
      • Focal segmental glomerulosclerosis (FSGS),
      • IgA nephropathy (i.e., Berger's disease),
      • IgM nephropathy,
      • Membranoproliferative glomerulonephritis,
      • Membranous nephropathy,
      • Minimal change disease,
      • Hypertensive nephrosclerosis and
  • Interstitial nephritis.
  • In a certain embodiment of the invention the kidney proteinuric disorder is diabetic nephropathy.
  • In a certain other embodiment of the invention the kidney proteinuric disorder is selected from to list comprising of:
      • Pre-eclampsia,
      • Eclampsia,
      • Collagen vascular diseases (e.g., systemic lupus erythematosus),
      • Dehydration,
      • Strenuous exercise,
      • Stress,
      • Benign Orthostatic (postural) proteinuria,
      • Sarcoidosis,
      • Alport's syndrome,
      • Diabetes mellitus,
      • Fabry's disease,
      • Infections (e.g., HIV, syphilis, hepatitis, post-streptococcal infection),
      • Aminoaciduria,
      • Fanconi syndrome,
      • Heavy metal ingestion,
      • Sickle cell disease,
      • Hemoglobinuria,
      • Multiple myeloma,
      • Myoglobinuria,
      • Organ rejection,
      • Ebola hemorrhagic fever and
      • Nail Patella Syndrome.
  • In one aspect of the invention the abβ3 integrin inhibitor is a αvβ3 monoclonal antibody or a peptide which contains a RGD binding sequence. In one aspect of the invention the avβ3 integrin inhibitor is a αvβ3 monoclonal antibody. In one embodiment of the invention the αvβ3 monoclonal antibody is anti-CD61.
  • In one embodiment of the invention the abβ3 integrin inhibitor is a peptide which contains a RGD binding sequence. In a certain embodiment of the invention the peptide which contains a RGD binding sequence is cyclo-[Arg-Gly-Asp-D-Phe-Val].
  • In one aspect of the invention the avβ3 integrin inhibitors is a compound of the formula:
  • Figure US20160296592A1-20161013-C00001
  • or a pharmaceutically acceptable salt thereof.
  • In one aspect of the invention the avβ3 integrin inhibitors is selected from the compounds described herein.
  • BRIEF DESCRIPTION OF DRAWINGS
  • The accompanying drawings are not intended to be drawn to scale. In the drawings, each identical or nearly identical component that is illustrated in various figures is represented by a like numeral. For purposes of clarity, not every component may be labeled in every drawing. In the drawings:
  • FIG. 1A-D is a panel of immunostaining images and a bar graph showing uPAR is induced in podocytes in proteinuric patients and experimental proteinuric models.
      • (A) uPAR protein (green) is expressed in glomeruli of human kidney. Some uPAR is found in podocytes as displayed by double immunofluorescence with the podocyte marker synaptopodin (synpo, red) resulting in a partial yellow overlap. (B) Induction of glomerular uPAR mRNA in proteinuric patients. Quantitative Real-time RT-PCR was performed on the isolated glomeruli from human biopsies. Glomerular uPAR mRNA is upregulated in focal segmental glomerulosclerosis (FSGS, n=14) and diabetic nephropathy (DN, n=20). Values are presented as Mean±SEM. *p<0.05 for FSGS or DN vs con (control patients). (C) Induction of uPAR protein in podocytes in rodent models of proteinuria revealed by immunocytochemistry. As shown by confocal microscopy, uPAR expression (green) is low in control glomeruli (con) from rat or mouse and colocalized partially with the podocyte marker synaptopodin (synpo, red). In podocytes during proteinuria (PAN, LPS, NZB/W F1), uPAR expression is significantly induced in podocytes, resulting in a yellow overlap with synaptopodin (merge). Original magnification, 600×. Note that uPAR expression in New Zealand black/white (NZB/W) F1 mice (Lupus) appears more segmental within the glomerolus. (D) Immunogold analysis of uPAR in glomerular walls of normal (control) and 12 months old diabetic rats. uPAR expression is found in all cells of the glomerulus including podocytes. uPAR expression is significantly induced in foot processes of diabetic rats. Original magnification, 35,000× Black arrows mark uPAR expression in podocyte foot processes. MC=mesangial cell, P=podocyte, End=endothelial cell, GBM=glomerular basement membrane, US=urinary space, CL=capillary lumen.
  • FIG. 2A-H is a panel of immunostaining images and bar graphs showing uPAR is required in podocytes for the development of foot process effacement and proteinuria.
      • (A) The morphology of podocyte foot processes is normal in PBS injected wild type and uPAR-/- mice. LPS treatment leads to foot process effacement in wild type but not in uPAR-/- mice. Original magnification, 31700×. (B) Restoration of uPAR expression in uPAR-/- mice using transient gene transfer of uPAR-cDNA leads to podocyte uPAR expression as shown by partial overlap of uPAR with synaptopodin (merge). Original magnification, 600×. (C) Immunoblot showing the amounts of exogenous uPAR expressed in liver and glomeruli from uPAR-/- mice 14 h after gene delivery. (D) Gene delivery of uPAR-cDNA or empty vector does not change the ultrastructure of podocyte foot processes in uPAR-/- mice. However, concomitant LPS treatment leads to foot process effacement in uPAR-restored uPAR-/- mice, but not in UPAR-/- mice given vector control. Original magnification, 31700×. (E) Urine Bradford assay. Whereas PBS treated control mice and uPAR-/- mice do not have any significant proteinuria, the injection of LPS induces significant proteinuria in wild type mice (0.29±0.21 (wt con); (0.82±0.32 (wt LPS), *p<0.003) but not in uPAR-/- mice (0.25±0.15 (uPAR-/- con); (0.32±0.14 (uPAR-/- LPS) or uPAR-/- mice which received control plasmids (0.14±0.06 (uPAR-/- con); (0.26±0.15 (uPAR-/- LPS). These data indicate that uPAR-/- mice were protected from urinary protein loss. uPAR-/- mice reconstituted with uPAR-cDNA develop heavy proteinuria but like wild type mice only after LPS injection (0.11±0.11 (uPAR-/- uPAR-cDNA); (0.92±0.46 (uPAR-/- uPAR-cDNA LPS), *p<0.002). The degree of proteinuria was comparable in LPS treated wild type and uPAR-cDNA reconstituted uPAR-/- mice, n=15 for each group.
      • (F) Podocyte-uPAR expression in uPAR-/- mice is achieved by gene transfer of podocin-driven uPAR cDNA (Pod-uPAR) and monitored by synaptopodin staining. Note the merge of uPAR and synaptopodin labeling (merge).
      • (G) Endothelial-uPAR expression in uPAR-/- mice achieved by gene transfer of ICAM-2-driven uPAR cDNA (ICAM-2-uPAR) is monitored by staining with the endothelial marker CD31. Note the merge of uPAR and CD31 labeling (merge).
      • (H) Fold-change in urinary protein loss in uPAR-/- mice following gene-transfer of Podocin-uPAR or ICAM-2-uPAR with and without LPS treatment.
  • FIG. 3A-D is a panel of immunostaining images and bar graphs showing uPAR mediates podocyte migration.
      • (A) Multiwell Boyden chamber assay on vitronectin-coated surface under normal conditions and in the presence of LPS or PAN. Podocytes, which migrated randomly through the membrane, were stained with DAPI (blue). (B) When compared to control, LPS and PAN treatment for 24 h promoted podocyte migration (wild type 41±3.7; LPS 72±5.5; PAN 67±4.9). Wild type: *P<0.001 for LPS vs control; *p<0.002 for PAN vs control. Knockdown of uPAR by siRNA strongly reduced podocyte migration in comparison with wild type podocytes under normal conditions and after treatment with LPS or PAN (uPAR-siRNA 16±2.4); (uPAR-siRNA LPS 12±3.6; uPAR-siRNA PAN 15±2.9). Wild type vs uPAR-siRNA *p<0.001. (C-D) Scrape wound assay on vitronectin-coated surface to assess directed podocyte motility. 24 h after scraping, wild type podocytes started to move into the wound track (61.37±8.05). The treatment with LPS (85.34±4.1) or with PAN (79.31±3.72) for 24 h significantly enhanced directed podocyte motility (*p<0.015 for LPS vs control); (*p<0.025 for PAN vs control). uPAR-siRNA expressing podocytes displayed defects in directed motility (uPAR-siRNA 17.24±10.48); (uPAR-siRNA LPS 28.00±7.17); (uPAR-siRNA PAN 22.26±6.92). Compared to wild type podocytes, uPAR-siRNA showed decreased capability to migrate into the wound track within 24 hours (*p<0.00241 for wild type vs uPAR-siRNA). Solid red lines indicate the initial margins of scrape wound. Data were based on six independent experiments. Original magnification, 40×.
  • FIG. 4A-C is a panel of a scheme, immunostaining image and bar graph showing uPAR, β3 integrin and vitronectin are important for LPS induced proteinuria.
      • (A) Schematic depiction of uPAR in complex with avβ3 integrin and vitronectin. Domain 2 of uPAR has been shown to interact with avβ3 integrin but other domains of uPAR may also contribute. The association of uPAR with avβ3 integrin can lead to conformational changes of the integrin consistent with activation that facilitates binding to ligands such as vitronectin. (B) Immunogold analysis of uPAR and β3 integrin in podocyte foot processes shows similarities in their distribution pattern. Both uPAR and β3 integrin are often located in vicinity to the slit diaphragm (black arrows). (C) The absence of uPAR (see FIG. 2E), β3 integrin (β3) and vitronection (Vn) is associated with normal renal permselective function and lack of proteinuria after LPS stimulation. In contrast, mice deficient in urokinase (uPA) have no proteinuria under normal conditions, but readily develop urinary protein loss after LPS administration. The same is observed for LPS treated uPAR-/- mice after gene delivery of UPAR-cDNA (uPAR-WT) or of an uPAR-cDNA encoding for a uPAR which is deficient in binding a3β1 integrin (uPAR-D262A).
  • FIG. 5A-E is a panel of immunostaining images and a bar graph showing uPAR activates β3 integrin.
      • (A) Double-immunofluoresence staining for β3 integrin (green) and the podocyte marker synaptopodin (synpo, red) in glomeruli from wild typo and uPAR-/- mice before and after treatment with LPS using confocal microscopy (original magnification, 600×). LPS treatment does not change podocyte β3 integrin in wild type and uPAR-/- mice. (B) Flow cytometry of AP5 in LPS treated podocytes in the presence of various amounts of Ca2+. Fluorescent intensity units: EDTA: 3020.64±302.06; 0.1 mM Ca++: 3055.76±400; 0.4 nM Ca++: 1644.7±200; 1.0 mM Ca++: 534.52±121; 2.0 mM Ca++: 312.5±53.45. (C) same as (A) but now with the antibody AP5 which recognizes active β3 integrin. LPS treatment induces staining for AP5 in wild type but not in uPAR-/- mice. (D) Immunofluorescent analysis and confocal microscopy of AP5 labeling (red) and uPAR (green) in uPAR downregulated podocytes using siRNA, in control podocytes and in podocytes overexpressing GFP-uPAR. (E) Activity assay of the small GTPases cdc42 and Rac1 isolated glomeruli form wild type and uPAR-/- mice before and after treatment with LPS. In some experiments, wild type mice were treated with LPS and Cyclo-RGDfV which blocks αvβ3 integrin.
  • FIG. 6A-G is A panel of immnunostaining images and bar graphs showing the active uPAR-αvβ3 integrin complex is lipid dependent and can be targeted pharmacologically to modify proteinuria.
      • (A) Sucrose gradient differential centrifugation was performed on podocyte whole cell extracts of normal and LPS treated cells. Each fraction was separated and blotted with raft (anti-caveolin) and non-rafts marker (anti-transferrin receptor), as well as anti-uPAR and anti-β3 integrin. Under normal conditions, uPAR and β3 integrin are preferentially associated with non-rafts fractions. Both, uPAR and β3 integrin are enriched within the lipid raft fraction after LPS treatment. (B) Localization of uPAR and active β3 integrin (AP5-labeling) in podocytes before and after LPS treatment and in the presence of the cholesterol depleting agent Methyl-β-cyclodextrine (MBCD). (C) Immunofluorescent staining of active β3 integrin in glomeruli of uPAR-/- mice (con) and of uPAR-/- mice which received β3 integrin cDNA (β3 integrin) and a cDNA encoding for the β3 integrin with a deletion of aminoacids 616-690 (β3Δ616-690). The latter confers constitutive activity to β3 integrin which is heavily stained by WOW-1 Fab antibody. (D) Gene transfer of cDNA encoding for the constitutively active β3 integrin but not the gene transfer of a cDNA coding for normal β3 integrin is sufficient to cause proteinuria in uPAR-/- mice even in the absence of LPS (con 0.25±0.06); (β3 integrin 0.19±0.05); (β3Δ619-690 integrin 0.43±0.09). *P<0.01 for β3 vs β3Δ616-690 integrin, injected mice, n=6 for each group. (E) Co-injection of LPS and a β3 integrin blocking antibody (anti-CD61) in wild type mice prevents the development of proteinuria (PBS con 0.12±0.03); (LPS+IgG con 0.84±0.14); (LPS+anti-β3 integrin 0.13±0.01). *p<0.001 for LPS+anti-β3 integrin vs IgG con. n=6. (F) Co-administration of LPS and anti-CD61 (anti-β3 integrin) antibody to cultured podocytes reduces podocyte motility (LPS+IgG con 88.56±7.26); (LPS+anti-β3 integrin 54.53±10.15). *p<0.03 for LPS+anti-β3 integrin vs IgG con. (G) The injection of cyclo-[Arg-Gly-Asp-D-Phe-Val] RGDfV blocks αvβ3 integrin and significantly reduces the degree of proteinuria in LPS treated mice. The injection of RGDfV reduces proteinuria in a dose dependent manner (Con, 0.2064±0.038; cyclo-RGDfV, 0.28±0.0535; LPS, 0.924±0.1055; LPS+cyclo-RGDfV (1 mg), 0.712±0.0817; LPS+cyclo-RGDfV (5 mg), 0.5683±0.0729; LPS+cyclo-RGDfV (20 mg), 0.4172±0.0423. *p<0.013 for LPS vs LPS+cyclo-RGD).
  • FIG. 7A-C is a panel of a scheme, immunostaining images and a bar graph showing induction of uPAR in cultured podocyte in response to LPS and PAN.
      • (A) Schematic representing of a kidney glomerulus and the glomerular filtration barrier. (B) Induction of uPAR in cultured podocyte in response to LPS and PAN. Relative quantification of uPAR induction: Con: 0.31±0.06; LPS: 0.49±0.06: PAN: 0.61±0.10. *P<0.025 for Con vs LPS and *P<0.015 for Con vs PAN. (C) Induced uPAR in podocytes localizes preferentially to the leading edge of migrating podocytes, arrows indicate plasma membrane localization.
  • FIG. 8A-B is a panel of immunostaining images showing Vitronectin is expressed in the human glomerulus and mainly in podocytes.
      • (A) Vitronectin (green) is expressed in the human glomerulus and mainly in podocytes, as displayed by double immunofluorescence with the podocyte marker synaptopodin (synpo, red) resulting in a partial yellow overlap. Podocytes are able to produce vitronectin as tested by RT-PCR. The vitronectin primers are as follows:
    Forward: 5′ AGT GGA GCA ACA GGA GGA GA 3′
  • Reverse: 5′ CAA GGC AAA GTG CTC AAA CA 3′ (data not shown). (B) Induction of vitronectin in podocytes in rodent models of proteinuria revealed by immunocytochemistry. As shown by confocal microscopy, vitronectin expression (green) is low in control glomeruli (con) from rat or mouse and colocalized partially with the podocyte marker synaptopodin (synpo, red). In nephrotic conditions however (PAN, LPS, NZB/W F1), vitronectin expression is significantly induced in podocytes, resulting in a yellow overlap with synaptopodin (merge). Original magnification, 600×. Note that vitronectin expression in New Zealand black/white (NZB/W) F1 mice (model for Lupus glomerulonephritis) appears more segmental within the glomerulus.
  • FIG. 9A-D is a panel of immunostaining images and graphs showing stable knockdown of uPAR mRNA using siRNA.
      • (A) Stable knockdown of uPAR mRNA using siRNA. (B) Decreased uPAR protein levels in uPAR siRNA expressing podocytes. (C) Time-course of wound closure in cultured podocytes. The distance migrated by cells from the wound margin was related to the width of the scar at different time points. (D) The addition of urokinase (uPA) to cultured podocytes did not modify podocyte motility.
  • FIG. 10A-B is a panel of immunostaining images showing uPAR interacts with β3 integrin.
      • (A) Heterogeneous Co-IP was performed on the lysates of HEK cells co-transfected with uPAR and β3 integrin, and blotted with HA or Flag antibodies. The results show that uPAR interacts with β3 integrin. (B) Heterogeneous Co-IP was performed on the lysates of HEK cells co-transfected with mouse β1integrin and full length human uPAR or an uPAR cDNA (uPAR D262A) deficient in the binding of β1 integrin. After blotting with HA or Flag antibodies the results show that full length human uPAR but not uPAR D262A interacts with mouse β1 integrin.
  • FIG. 11 is a bar graph showing the effect of cyclo-RGDfV on recovery of proteinuria.
  • DETAILED DESCRIPTION
  • The present invention is directed to methods of treating proteinuric diseases by the administration of avβ3 integrin inhibitors. The invention is based at least in part on the discovery that induction of urokinase receptor signaling in podocytes leads to foot process effacement and urinary protein loss via a mechanism including lipid dependent activation of avβ3 integrin.
  • The invention involves, at least in part, the study of molectdes which are associated with cellular motility such as the urokinase plasminogen activator receptor (uPAR)15, 16. uPAR is a glycosylphosphatidylinositol (GPI)-anchored protein that has been recognized as a proteinase receptor but has also be involved in non-proteolytic pathways, mainly through its ability to form complexes with other membrane proteins such as integrins for signal transduction15. uPAR plays important roles during wound healing, inflammation, and stem cell mobilization, as well as in severe pathological conditions such as HIV-1 infection, tumor invasion and metastasis17. Besides uPAR's well-established role in the regulation of pericellular proteolysis, it is also involved in cell adhesion, migration, and proliferation through interactions with proteins present in the extracellular matrix, including vitronectin (Vn)18. Most recently, the importance of uPAR-Vn interactions was demonstrated by the direct requirement of uPAR to bind Vn for the sufficient induction of downstream signaling aimed at cell motility and morphology19.
  • Although not wishing to be bound by theory the invention is based on the experimental observations discussed herein that present a mechanism that involves activation of avβ3 integrin within lipid rafts24. The inhibition of uPAR/avβ3 integrin function in podocytes ameliorated the course of proteinuria and opens novel avenues for pharmacological interventions. Blockade of avβ3 integrin reduces podocyte motility in vitro and proteinuria in mice. Mice deficient of uPAR are protected from proteinuria unless a constitutively active β3 integrin is expressed. Gene transfer of uPAR into podocytes but not into endothelial cells conferred the ability to develop urinary protein loss. Mechanistically, uPAR may be required to activate avβ3 integrin in podocytes which promotes cell motility and activation of small GTPases cdc42 and Rac1. There is induction of uPAR expression in podocytes during human and rodent proteinuric kidney diseases. uPAR is required for podocyte migration and LPS-induced FP effacement and proteinuria in mice via a mechanism that includes lipid-dependent activation of αvβ3 integrin. These findings define a novel signaling pathway in podocytes which involves uPAR, αvβ3 integrin and Vn. The examples also describe a mechanism whereby widely expressed proteins such as uPAR are utilized in cell-specific manner to regulate basic mechanisms such as kidney filtration. uPAR expression is present in all glomerular cells, yet uPAR expression is not required for normal renal function in any of the cells as the uPAR-/- mice behave normal. Surprising is the requirement of uPAR during the development of podocyte FP effacement and proteinuria which suggests that inducible pathways are required for the remodeling of the filtration barrier. uPAR action for the development of proteinuria stems from its action in the podocyte, which was demonstrated using cell-specific promoter elements. In one aspect of the invention methods for treating proteinuria are provided by inhibiting avβ3 integrin function by inhibiting uPAR function. uPAR is induced during proteinuria and that uPAR-/- mice are protected from the development of proteinurea. In one aspect of the invention the uPAR inhibitors, including small molecules, peptides and antibodies can be used to inhibit avβ3 integtin function.
  • Accordingly, the methods of the present invention are used to treat disorders characterized by proteinuria. As used herein “proteinuria” refers to any amount of protein passing through a podocyte, such as a podocyte that has suffered podocyte damage. For example, the processing of protein by cultured podocytes that have undergone actin-cytoskeleton rearrangment and FP effacement would result in proteinuria. As used herein “podocyte damage” refers to FP effacement and/or cortical actin rearrangement or any other reversible structural or functional change in podocytes that results in proteinuria. In an in vivo system the term “proteinuria” refers to the presence of excessive amounts of serum protein in the urine. In a certain embodiment the excessive amount of serum protein in the urine is greater than 50, 100, 150, or 200 mg of serum protein/day. In a preferred embodiment the amount of serum protein in the urine is greater than 150 mg/day. Proteinuria is a characteristic symptom of either renal (kidney), urinary, pancreatic distress, nephrotic syndromes (for example, proteinuria larger than 3.5 grams per day), eclampsia, toxic lesions of kidneys, and it is frequently a symptom of diabetes mellitus. With severe proteinuria general hypoproteinemia can develop and it results in diminished oncotic pressure (ascites, edema, hydrothorax).
  • As used herein a proteinuric disorder refers to, but it is not limited to: Diabetic nephropathy, Nephrotic syndromes (i.e. intrinsic renal failure), Nephritic syndromes, Toxic lesions of kidneys, Glomerular diseases, such as membranous glomerulonephritis, Focal segmental glomerulosclerosis (FSGS), IgA nephropathy (i.e., Berger's disease), IgM nephropathy, Membranoproliferative glomerulonephritis, Membranous nephropathy, Minimal change disease, Hypertensive nephrosclerosis and Interstitial nephritis.
  • In certain embodiments as used herein, a proteinuric disorder refers to: Pre-eclampsia, Eclampsia, Collagen vascular diseases (e.g., systemic lupus erythematosus), Dehydration, Strenuous exercise, Stress, Benign Orthostatic (postural) proteinuria, Sarcoidosis, Alport's syndrome, Diabetes mellitus, Fabry's disease, Infections (e.g., HIV, syphilis, hepatitis, post-streptococcal infection), Aminoaciduria, Fanconi syndrome, Heavy metal ingestion, Sickle cell disease, Hemoglobinuria, Multiple myeloma, Myoglobinuria, Organ rejection, Ebola hemorrhagic fever and Nail Patella Syndrome.
  • In certain embodiments as used herein a proteinuric disorder refers to diabetes, hypertension, kidney disease, minimal change disease, membranous glomerulonephritis, focal segmental glomerulosclerosis, diabetic neuropathy, post-infectious glomerulonephritis, mesangioproliferative glomerulonephritis, HIV-associated nephropathy, IgA-nephropathy, and Cardiovascular disease.
  • At the cellular level protein loss in the urine is accompanied by a structural re-arrangement of podocyte cells. Renal ultrafiltration is located within the renal glomerulus, a combination of blood vessels and cells. Highly specialized podocyte cells perform the filtering work and are main target cells in kidney disease. Podocytes can reorganize their actin-based cytoskeleton in a highly dynamic fashion. Such a reorganization determines the integrity of the ultrafiltration barrier in the kidney. Reorganization of the actin cytoskeleton in podocyte foot processes from stress fibers into cortical actin leads to podocyte foot processes (FP) effacement and the development of urinary protein loss. Podocyte damage can be caused by many conditions and factors including LPS and purine aminonucleoside (PAN). These alterations lead to ongoing damage of the kidney and over time to a deterioration of the kidney function. The instant invention is based in part on the surprising discovery of the functional significance of αvβ3 integrin activation as a downstream effector for increased podocyte motility and FP effacement.
  • Integrins belong to the family of heterodimeric class I transmembrane receptors, which play an important role in numerous cell-matrix and cell-cell adhesion processes (Tuckwell et al., 1996, Symp. Soc. Exp. Biol. 47). They can be divided roughly into three classes: the β1 integrins, which are receptors for the extracellular matrix, the β2 integrins, which can be activated on leucocytes and are triggered during inflammatory processes, and the αv integrins, which influence the cell response in wound-healing and other pathological processes (Marshall and Hart, 1996, Semin. Cancer Biol. 7, 191).
  • The αbβ3 integrin, also called that the vitronectin receptor, mediates adhesion to a multiplicity of ligand's-plasma proteins, extracellular matrix proteins, cell surface proteins, of which the majority contain the amino acid sequence Arg-Gly-Asp (RGD), such as, for example, fibronectin or vitronectin. Soluble RGD-containing peptides are capable of inhibiting the interaction of each of these integrins with the corresponding natural ligands. Integrin αvβ3 antagonistic action has been shown for a multiplicity of compounds, such as anti-αvβ3 monoclonal antibodies, peptides which contain the RGD binding sequence, natural, RGD-containing proteins (e.g. disintegrins) and low-molecular weight compounds, (FEBS Letts 1991, 291, 50-54; J. Biol. Chem, 1990, 265, 12267-12271; J. Biol. Chem. 1994, 269, 20233-20238; J. Cell Biol 1993, 51, 206-218; J. Biol. Chem. 1987, 262, 17703-17711; Bioorg. Med. Chem. 1998, 6, 1185-1208).
  • Advantageous αvβ3 integrin receptor ligands bind to the integrin αvβ3 receptor with an increased affinity by at least a factor of 10, preferably at least a factor of 100.
  • In one aspect of the invention a method for treating a proteinuric disorder comprising administering to a patient in need thereof an αvβ3 integrin inhibitors is provided. According to one embodiment of the invention the αvβ3 integrin inhibitors are the compounds described in WO 97/08145 A1, WO 00/48996 A2, WO 06043930 A1, US2002072500, US 2005043344, US 2002045645, US 2002099209, US 2005020505, US 2002072518, US 2002077321, US 2004043994, US 2003069236, US 2003144311.
  • In one embodiment of the invention the αvβ3 integrin inhibitors are peptidic sulfonamides having the formula (I): R1-Arg-X-Asp-Leu-Asp-Ser-Leu-Arg-R2, in which R1 denotes H, acetyl or acyl and R2 denotes —Oh, OR3N2,NHR3, N(R3)2 R3 denotes alkyl, aralkyl, aryl, Het and X denotes an amino acid, in which A denotes (CH2)n R4 denotes H, alkyl, aralkyl or aryl, and n denotes 1, 2, 3, 4, 5 or 6, and the amino acid is bonded to the adjacent Arg via a peptide bond of the α-amino group and to the α-amino group of the adjacent Asp via a peptide bond of the α-carboxyl group as described in U.S. patent application No. 2006/0148716 A1.
  • In one embodiment of the invention the αvβ3 integrin inhibitors are fluoro-alkyl-cyclopeptide derivatives as described in WO 2004/011487 A2.
  • In one embodiment of the invention the αvβ3 integrin inhibitors are peptido-mimetic compounds containing an RGD sequence as described in WO 2005/007654 A1 and U.S. Pat. No. 6,451,972.
  • In one embodiment of the invention the αvβ3 integrin inhibitors are antagonists of the αvβ3 integrin receptor based on a bicyclic structural element are described in WO 9906049, WO 9905107, WO 9814192, WO 9724124, WO 9724122 and WO 9626190.
  • In one embodiment of the invention the αvβ3 integrin inhibitors are compounds to described in U.S. Publication No 2004/0063934.
  • The αvβ3 integrin inhibitors of the invention include isolated peptides and proteins. As used herein with respect to polypeptides, proteins or fragments thereof, “isolated” means separated from its native environment and present in sufficient quantity to permit its identification or use. Isolated, when referring to a protein or polypeptide, means, for example: (i) selectively produced by expression cloning or (ii) purified as by chromatography or electrophoresis. Isolated proteins or polypeptides may be, but need not be, substantially pure. The term “substantially pure” means that the proteins or polypeptides are essentially free of other substances with which they may be found in nature or in vivo systems to an extent practical and appropriate for their intended use. Substantially pure polypeptides may be produced by techniques well known in the art. Because an isolated protein may be admixed with a pharmaceutically acceptable carrier in a pharmaceutical preparation, the protein may comprise only a small percentage by weight of the preparation. The protein is nonetheless isolated in that it has been separated from the substances with which it may be associated in living systems, i.e. isolated from other proteins.
  • The αvβ3 integrin inhibitors of the invention may be produced using any of the methods and techniques known to those skilled in the art. For example, αvβ3 integrin inhibitors can be purified from a source which naturally expresses the protein, can be isolated fman a recombinant host which has been altered to express the desired mutant or fragment thereof, or can be synthesized using protein synthesis techniques known in the art. The skilled artisan can readily adapt a variety of techniques in order to obtain αbβ3 integrin inhibitors that are peptides, proteins or fragments thereof.
  • Other agents that are αvβ3 integrin inhibitors include but are not limited to small molecules, and other drugs, including known drugs, that prevent (i.e. reduce or inhibit further increase) integrin activation. Such agents can be identified using routine screening methods. For instance, αvβ3 integrin inhibitors of the present invention can be identified using the methods and assays described herein. The screening may be a random screen or it may be rationally designed. For random screening, putative inhibitors are selected at random and assayed for their ability to produce the desired physiological effect. For instance, the putative modulators may be assayed for the ability to reduce selectively or specifically the amount or rate of αvβ3 integrin activation. Any suitable method or technique known to those skilled in the art may be employed to assay putative inhibitors.
  • Methods for screening using rational design employ the same types of screening methods but begin with a set of compounds that has been designed to specifically maximize function. For rational selection or design, the αvβ3 integrin inhibitors may be selected based, for example on the RGD binding domain. Any of the suitable methods and techniques known to those skilled in the art may be employed for rational selection or design. For example, one skilled in the art can readily adapt currently available procedures to generate pharmaceutical agents capable of binding to a specific peptide sequence of αbβ3 integrin or uPAR, precluding their interaction and signal transduction and thereby inhibiting αvβ3 integrin activity. Illustrative examples of such available procedures are described, for example, in Hurby et al., “Application of Synthetic Peptides: Antisense Peptides,” in Synthetic Peptides, A User's Guide, W. H. Freeman, N.Y., pp. 289-307 (1992); Kaspczak et al., Biochemistry 28:9230 (1989); and Harlow, Antibodies, Cold Spring Harbor Press, N.Y. (1990).
  • In one aspect of the invention αvβ3 integrin inhibitors include antibodies and antibody fragments which are capable of binding to αvβ3 integrin and consequently acting as a αvβ3 integrin inhibitors. The antibodies of the present invention include polyclonal and monoclonal antibodies, as well as antibody fragments and derivatives that contain the relevant antigen binding domain of the antibodies. Such antibodies or antibody fragments are preferably used in the diagnostic and therapeutic embodiments of the present invention. In a preferred embodiment the antibody is anti-CD61 antibody.
  • As used herein, “treating a proteinuric disorder” includes preventing the development of proteinurea, reducing or inhibiting proteinurea, slowing the progression, and/or any other desired effect on proteinurea. According to the invention the αvβ3 integrin inhibitors can be administered prior to the onset of proteinurea, following the onset of proteinurea, or as part of any therapeutic regimen, for example, including cancer medicaments.
  • The methods of the invention are intended to embrace the use of more than one other medicament along with the αvβ3 integrin inhibitor. As an example, where appropriate, the αvβ3 integrin inhibitor may be administered with both a chemotherapeutic agent, anti-diabetic agent or an immunotherapeutic agent.
  • The invention also encompasses diagnostic assays for determining the presence of a disorder characterized by proteinuria in a subject. This aspect of the invention is based, at least in part, on the discovery that αvβ3 integrin activation is reduced in damaged or proteinuric podocytes. In the method an amount of αvβ3 integrin activity in a podocyte cell is determined. That amount is compared to a pre-determined threshold or to a control level. A disorder characterized by proteinuria is determined when the amount of αvβ3 integrin activity is below the pre-determined threshold. As used here in “pre-determined threshold or a control level” refers to αvβ3 integrin activity levels in normal, healthy podocytes, i.e. podocytes not affected by podocyte damage or proteinuria. The podocyte cells may be within a biological sample. The biological sample may be, for instance, a biopsy sample of proteinuric tissue. As used herein, activity refers to expression and well as biochemical activity as described herein.
  • The detection of αvβ3 integrin in podocyte cells can be readily carried out by standard immunostaining or immunocytometric methods, readily known by persons of ordinary skill in the art. As used herein “immunostaining” refers to a technique of applying coloured or fluorescent dyes to tissues in preparation for microscopic examination. The assay may be performed using immunogold election microscopy.
  • In one embodiment the diagnostic assays are performed on cells and/or tissue samples wherein morphological changes of the actin-cytoskeleton can not be readily detected by any other immunocytometric methods. For example one such disorder would be minimal change disease. The term minimal change disease comes from the notion that morphological podocyte changes are only visible by electron microscopy. Detection of proteinuria in patients with minimal change disease by immunocytometric methods would be advantageous because it provide ease and speed of detection.
  • The immunocytometric methods may be performed using labeled antibodies. An antibody is said to be “detectably labeled” if the antibody, or fragment thereof, is attached to a molecule which is capable of identification, visualization, or localization using known methods. Suitable detectable labels include radioisotopic labels, enzyme labels, non-radioactive isotopic labels, fluorescent labels, affinity labels, chemiluminescent labels and nuclear magnetic resonance contrast agents.
  • Illustrative examples of suitable enzyme labels include, but are not limited to, luciferase, malate dehydrogenase, staphyloccal nuclease, delta-5-steroid isomerase, yeast-alcohol dehydrogenase, alpha-glycerol phosphate dehydrogenase, triose phosphate isomerase, horseradish peroxidase, alkaline phosphatase, asparaginase, glucose, oxidase, beta-galactosidase, ribonuclease, urease, catalase, glucose-6-phosphate dehydrogenase, glucoamylase, and acetylcholine esterase.
  • Examples of suitable radioisotopic labels include, but are not limited to, 3H, 111In, 125I, 131I, 32P, 35S, 14C, 51Cr, 57To, 58Co, 59Fe, 75Se, 152Eu, 90Y, 67Cu, 217Ci, 211At, 212Pb, 47Sc, 109Pd, etc. 111In is a preferred isotope where in vivo imaging is used since its avoids the problem of dehalogenation of the 125I or 131I-labeled monoclonal antibody by the liver. In addition, this radionucleotide has a more favorable gamma emission energy for imaging (Perkins et al., Eur. J. Nucl. Med 10:296-301 (1985); Carasquillo et al., J. Nucl. Med. 28:281-287 (1987)). For exaample 111In coupled to monoclonal antibodies with 1-(p-isothiocyanatobenzyl)-DPTA has shown little uptake non-tumorous tissues, particularly the liver, and therefore enhances specificity of tumor localization (Esteban et al., J. Nucl. Med. 28:861870 (1987)).
  • Illustrative examples of suitable non-radioactive isotopic labels include, but are not limited to, 157Gd, 55Mn, 162Dy, 52Tr, and 56Fe.
  • Illustrative examples of suitable fluorescent labels include, but are not limited to, an 152Eu label, a fluorescent protein (including green fluorescent protein (GFP), enhanced green fluorescent protein (EGFP), enhanced yellow fluorescent protein (YFP) and enhanced cyan fluorescent protein (ECFP),), a fluorescein label, an isothiocyanate label, a rhodamine label, a phycoerythrin label, a phycocyanin label, an allophycocyanin label, an o-phthaldehyde label, and a fluorescamine label.
  • Illustrative examples of chemiluminescent labels include a luminal label, an isoluminal label, an aromatic acridinium ester label, an imidazole label, an acridinium salt label, an oxalate ester label, a luciferin label, a luciferase label, and an aequorin label.
  • Illustrative examples of nuclear magnetic resonance contrasting agents include paramagnetic heavy metal nuclei such as Gd, Mn, and Fe.
  • The coupling of one or more molecules to antibodies is envisioned to include many chemical mechanisms, for instance covalent binding, affinity binding, intercalation, coordinate binding, and complexation.
  • The covalent binding can be achieved either by direct condensation of existing side chains or by the incorporation of external bridging molecules. Many bivalent or polyvalent agents are useful in coupling protein molecules to other proteins, peptides or amine functions, etc. For example, the literature is replete with coupling agents such as carbodiimides, diisocyanate, glutaraldehyde, diazobenzenes, and hexamethylene diamines. This list is not intended to be exhaustive of the various coupling agents known in the art but, rather, is exemplary of the more common coupling agents.
  • According to the methods of the invention the αvβ3 integrin inhibitors are administered in pharmaceutically acceptable preparations. When administered, the pharmaceutical preparations of the invention are applied in pharmaceutically-acceptable amounts and in pharmaceutically-acceptable compositions. The term “pharmaceutically acceptable” means a non-toxic material that does not interfere with the effectiveness of the biological activity of the active ingredients. Such preparations may routinely contain salts, buffering agents, preservatives, compatible carriers, and optionally other therapeutic agents. When used in medicine, the salts should be pharmaceutically acceptable, but non-pharmaceutically acceptable salts may conveniently be used to prepare pharmaceutically-aceeptable salts thereof and are not excluded from the scope of the invention.
  • As used herein, “pharmaceutically acceptable carrier” or “physiologically acceptable carrier” includes any and all salts, solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like that are physiologically compatible. Preferably, the carrier is suitable for intravenous, intramuscular, subcutaneous, parenteral, per os, spinal or epidermal administration (e.g., by injection or infusion). Depending on the route of administration, the active compound, i.e., αvβ3 integrin inhibitor may be coated in a material to protect the compound from the action of acids and other natural conditions that may inactivate the compound. Preferably the carrier is sterile.
  • A salt retains the desired biological activity of the parent compound and does not impart any undesired toxicological effects (see e.g., Berge. S. M., et al. (1977) J. Pharm. Sci, 66: 1-19). Examples of such salts include acid addition salts and base addition salts. Acid addition salts include those derived from nontoxic inorganic acids, such as hydrochloric, nitric, phosphoric, sulfuric, hydrobromic, hydroiodic, phosphorous and the like, as well as from nontoxic organic acids such as aliphatic mono- and dicarboxylic acids, phenyl substituted alkanoic acids, hydroxy alkanoic acids, aromatic acids, aliphatic and aromatic sulfouic acids and the like. Base addition salts include those derived from alkaline earth metals, such as sodium, potassium, magnesium, calcium and the like, as well as from nontoxic organic amines, such as N,N′-dibenzylethylenediamine N-methylglocamine, chioroprocaine, choline, diethanolamine, ethylenediamine, procaine and the like.
  • The pharmaceutical compositions may contain suitable buffering agent, including: acetic acid in a salt; citric acid in a salt; boric acid in a salt; and phosphoric acid in a salt. The pharmaceutical compositions also may contain, optionally, suitable preservatives, such as: benzalkonium chloride; chlorobutanol; parabens and thimerosal.
  • The pharmaceutical compositions may conveniently be presented in unit dosage form and may be prepared by any of the methods well-known in the art of pharmacy. All methods include the step of bringing the active agent into association with a carrier which constitutes one or more accessory ingredients. In general, the compositions are prepared by uniformly and intimately bringing the active compound into association with a liquid carrier, a finely divided solid carrier, or both, and then, if necessary, shaping the product.
  • Compositions suitable for parenteral administration conveniently comprise a sterile aqueous or non-aqueous preparation of the compounds, which is preferably isotonic with the blood of the recipient. This preparation may be formulated according to known methods using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation also may be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example, as a solution in 1,3-butane diol. Among the acceptable vehicles and solvents that may be employed are water. Ringer's solution, and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed including synthetic mono-or di-glycerides. In addidon, fatty acids such as oleic acid may be used in the preparation of injectables. Carrier formulations suitable for oral, subcutaneous, intravenous, intramuscular, etc. administration can be found in Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, Pa.
  • The active compounds can be prepared with carriers that will protect the compound against rapid release, such as a controlled release fornaulation, including implants, transdermal patches, and microencapsulated delivery systems. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Many methods for the preparation of such formulations are patented or generally known to those skilled in the art. See, e.g., Sustained and Controlled Release Drug Delivery Systems, J. R. Robinson, ed., Marcel Dekker, Inc., New York, 1978.
  • The present invention therefore provides pharmaceutical compositions comprising one or more αvβ3 integrin inhibitors. These pharmaceutical compositions may be administered orally, rectally, parenterally, intrathecally, intracisternally, intravaginally, intraperitoneally, topically (as by powders, ointments, drops or transdermal patch), bucally, or as an oral or nasal spray. The term “parenteral” as used herein refers to modes of administration which include intravenous, intramuscular, intrathecal, intraperitoneal, intrasternal, subcutaneous and intraarticular injection and infusion. One of ordinary skill will recognize that the choice of a particular mode of administration can be made empirically based upon considerations such as the particular disease state being treated; the type and degree of the response to be achieved; the specific agent or composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration and rate of excretion of the agent or composition; the duration of the treatment; drugs used in combination or coincidental with the specific composition; and like factors well known in the medical arts.
  • Pharmaceutical compositions of the present invention for parenteral injection may comprise pharmaceutically acceptable sterile aqueous or nonaqueous solutions, dispersions, suspensions or emulsions as well as sterile powders for reconstitution into sterile injectable solutions or dispersions just prior to use. Illustrative examples of suitable aqueous and nonaqueous carriurs, diluents, solvents or vehicles include, but are not limited to, water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), carboxymethylceuulose and suitable mixtures thereof, vegetable oils (such as olive oil), and injectable organic esters such as ethyl oleate. Proper fluidity can be maintained, for example, by the use of coating materials such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.
  • The compositions of the present invention may also contain preservatives, wetting agents, emulsifying agents, and dispersing agents. Prevention of the action of microorganisms may be ensured by the inclusion of various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents such as sugars, sodium chloride, and the like. Prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents which delay absorption such as aluminum monostearate and gelatin.
  • In some cases, in order to prolong the effect of the therapeutic agent, it is desirable to slow the absorption from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material with poor water solubility. The rate of absorption of the drug then depends upon its rate of dissolution which, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle.
  • Injectable depot forms are made by forming microencapsuled matrices of the drug in biodegradable polymers such as polylactide-polyglycolide. Depending upon the ratio of drug to polymer and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions which are compatible with body tissues.
  • Solid dosage forms for oral administration include, but are not limited to, capsules, tablets, pills, powders, and granules. In such solid dosage forms, the active compounds are preferably mixed with at least one pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such as quaternary ammonium compounds, g) wetting agents such as, for example, cetyl alcohol and glycerol monostearate, h) absorbents such as kaolin and bentonite clay, and i) lubricants such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof. In the case of capsules, tablets and pills, the dosage form may also comprise buffering agents as appropriate.
  • Solid compositions of a similar type may also be employed as fillers in soft and hard filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like.
  • The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Illustrative examples of embedding compositions which can be used include polymeric substances and waxes.
  • The active αvβ3 integrin inhibitors can also be in micro-encapsulated form, if appropriate, with one or more of the above-mentioned excipients.
  • Liquid dosage forms for oral administration include, but are not limited to, pharmaceutically acceptable emulsions, solutions, suspensions, syrups and elixirs. In addition to the active compounds, the liquid dosage forms may contain inert diluents so commonly used in the art such as, for example, water or other solvents, solubilizing, agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethyl formamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
  • Besides inert diluents, the oral compositions may also contain adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
  • Suspensions, in addition to the active compounds, may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar, and tragacanth, and mixtures thereof.
  • In some embodiments of the invention the αvβ3 integrin inhibitor is administered in the form of liposomes. As is known to those skilled in the art, liposomes are generally derived from phospholipids or other lipid substances. Liposomes are formed by mono- or multi-lamellar hydrated liquid crystals that are dispersed in an aqueous medium. Any non-toxic, physiologically acceptable and metabolizable lipid capable of forming liposomes can be used. The present compositions in liposome form can contain, in addition to the αvβ3 integrin inhibitor, stabilizers, preservatives, excipients, and the like. Preferred lipids are phospholipids and phosphatidyl cholines (lecithins), both natural and synthetic. Methods to form liposomes am known in the art. See, e.g., Prescott, ed., METHODS IN CELL BIOLOGY, Volume XIV, Academic Press, New York, N.Y. (1976), p. 33 et seq.
  • One of ordinary skill will appreciate that effective amounts of the therapeutic agents used in the methods of the invention can be determined empirically and may be employed in pure form or, where such forms exist, in pharmaceutically acceptable salt, ester or prodrug form. The therapeutic agents may be administered in compositions in combination with one or more pharmaceutically acceptable excipients. It will be understood that, when administered to a human patient, the total daily usage of the agents and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment. The specific therapeutically effective dose level for any particular patient will depend upon a variety of factors including the type and degree of the response to be achieved; the specific agent or composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the agent or composition; the duration of the treatment; drugs (such as a chemotherapeutic agent) used in combination or coincidental with the specific composition; and like factors well known in the medical arts.
  • Techniques of dosage determination are well known in the art. For example, satisfactory results are obtained by oral administration of therapeutic dosages on the order of from 0.05 to 10 mg/kg/day, preferably 0.1 to 7.5 mg/kg/day, more preferably 0.1 to 2 mg/kg/day, administered once or, in divided doses, 2 to 4 times per day. On administration parenterally, for example by i.v. drip or infusion, dosages on the order of from 0.01 to 5 mg/kg/day, preferably 0.05 to 1.0 mg/kg/day and more preferably 0.1 to 1.0 mg/kg/day can be used. Suitable daily dosages for patients are thus on the order of from 2.5 to 500 mg p.o., preferably 5 to 250 mg per oral (p.o.), more preferably 5 to 100 mg p.o., or on the order of from 0.5 to 250 mg i.v., preferably 2.5 to 125 mg i.v. and more preferably 2.5 to 50 mg i.v.
  • The administration of the agents of the present invention may be for either prophylactic or therapeutic purpose. When provided prophylactically, the agent is provided in advance of any damage i.e., proteinuria or podocyte damage. The prophylactic administration of the agent serves to prevent or reduce the rate of onset of symptoms. When provided therapeutically, the agent is provided at (or after) the onset of the appearance of symptoms of actual disease. The therapeutic administration of the agent serves to reduce the severity and duration of proteinuria.
  • The compositions of the invention are administered in effective amounts. An “effective amount” is that amount of any of the compositions provided herein that alone, or together with further doses, produces the desired response, e.g. reduces or eliminates proteinuria. This may involve only slowing the progression of the disease temporarily, although more preferably, it involves halting the progression of the disease permanently. This can be monitored by routine methods. The desired response to treatment of the disease or condition also can be delaying the onset or even preventing the onset of the disease or condition. An amount that is effective can be the amount of a αvβ3 integrin inhibitor alone which produces the desired therapeutic endpoint. An amount that is effective is also the amount of a αvβ3 integrin inhibitor in combination with another agent that produces the desired result.
  • Such amounts will depend, of course, on the particular condition being treated, the severity of the condition, the individual patient parameters including age, physical condition, size and weight, the duration of the treatment, the nature of concurrent therapy (if any), the specific route of administration and like factors within the knowledge and expertise of the health practitioner. These factors are well known to those of ordinary skill in the art and can be addressed with no more than routine experimentation. It is generally preferred that a maximum dose of the individual components or combinations thereof be used, that is, the highest safe dose according to sound medical judgment. It will be understood by those of ordinary skill in the art, however, that a patient may insist upon a lower dose or tolerable dose for medical reasons, psychological reasons or for virtually any other reasons.
  • The doses of αvβ3 integrin inhibitors administered to a subject can be chosen in accordance with different parameters, in particular in accordance with the mode of administration used and the state of the subject. Other factors include the desired period of treatment. In the event that a response in a subject is insufficient at the initial doses applied, higher doses (or effectively higher doses by a different, more localized delivery route) may be employed to the extent that patient tolerance permits.
  • Based upon the composition, the dose can be delivered continuously, such as by continuous pump, or at periodic intervals. Desired time internals of multiple doses of a particular composition can be determined without undue experimentation by one skilled in the art. Other protocols for the administration of the compositions provided will be known to one of ordinary skill in the art, in which the dose amount, schedule of administration, site of administration, mode of administration and the like vary from the foregoing.
  • Administration of αvβ3 integrin inhibitor compositions to mammals other than humans, e.g. for testing purposes or veterinary therapeutic purposes, is carried out under substantially the same conditions as described above.
  • The compositions of the present invention have in vitro and in vivo diagnostic and therapeutic utilities. For example, these molecules can be administered to cells in culture, e.g. in vitro or ex vivo, or in a subject, e.g., in vivo, to treat, prevent or diagnose a variety of disorders. As used herein, the term “subject” is used interchangeably with the term “patient” and is intended to include humans and non-human animals including but not limited to a dog, cat, horse, cow pig, sheep, goat, or primate, e.g., monkey. Preferred patients include a human patient having a proteinuric disorder, including disorders characterized by proteinuria as described herein.
  • As used herein “a patient in need thereof” refers to any patient that is affected with a disorder characterized by proteinuria. In one aspect of the invention “a patient in need thereof” refers to any patient that may have, or is at risk of having a disorder characterized by proteinuria. In one embodiment of the invention “a patient in need thereof” is a patient that has, may have or is at risk at having cancer, precancer, refractory cancer or metastatic cancer. In yet another embodiment of the invention “a patient in need thereof” is a patient that has, may have, or is at risk of having a cognitive disorder, such as Alzheimer's disease or dementia.
  • The compositions provided of the present invention can be used in conjunction with other therapeutic treatment modalities. Such other treatments include surgery, radiation, cryosurgery, thermotherapy, hormone treatment, chemotherapy, vaccines, and immunotherapies.
  • The invention also relates in some aspects to the identification and testing of candidate agents and molecules that may be αbβ3 integrin inhibitors. These molecules are referred to as putative αvβ3 integrin inhibitors. The putative αvβ3 integrin inhibitors can be screened for activity using the same type of assays as described herein (e.g., the assays described in the Examples section). Using such assays, additional αvβ3 integrin inhibitors can be can be identified.
  • The invention further provides efficient methods of identitying pharmacological agents or lead compounds as αvβ3 integrin inhibitors. Generally, the screening methods involve assaying for compounds which inhibit the level of αvβ3 integrin activity. As will be understood by one of ordinary skill in the art, the screening methods may measure the level of binding between the molecules directly, such as by using the methods employed in the Examples. In addition, screening methods may be utilized that measure a secondary effect of the αvβ3 integrin inhibitors, for example the level of proteinuria in a cell or tissue sample.
  • A wide variety of assays for pharmacological agents can be used in accordance with this aspect of the invention, including, labeled in vitro protein-protein binding assays, electrophoretic mobility shift assays, immunoassays, cell-based assays such as two or three-hybrid screens, expression assays, etc. The assay mixture comprises a candidate pharmacological agent. Typically, a plurality of assay mixtures are run in parallel with different agent concentrations to obtain a different response to the various concentrations. Typically, one of these concentrations serves as a negative control, i.e., at zero concentration of agent or at a concentration of agent below the limits of assay detection.
  • Putative inhibitors useful in accordance with the invention encompass numerous chemical classes, although typically they are organic compounds. Preferably, the putative modulators are small organic compounds, i.e., those having a molecular weight of more than 50 yet less than about 2500, preferably less than about 1000 and, more preferably, less than about 500. Putative αvβ3 integrin inhibitors comprise functional chemical groups necessary for structural interactions with proteins and/or nucleic acid molecules, and typically include at least an amine, carbonyl, hydroxyl or carboxyl group, preferably at least two of the functional chemical groups and more preferably at least three of the functional chemical groups. The putative modulators can comprise cyclic carbon or heterocyclic structure and/or aromatic or polyaromatic structures substituted with one or more of the above-identified functional groups. Putative modulators also can be biomolecules such as peptides, saccharides, fatty acids, sterols, isoprenoids, purines, pyrimidines, derivatives or structural analogs of the above, or combinations thereof and the like. Where the putative modulator is a nucleic acid molecule, the agent typically is a DNA or RNA molecule, although modified nucleic acid molecules as defined herein are also contemplated.
  • It is contemplated that cell-based assays as described herein can be performed using cell samples smiler cultured cells. Biopsy cells and tissues as well as cell lines grown in culture are useful in the methods of the invention.
  • Putative αvβ3 integrin inhibitors are obtained from a wide variety of sources including libraries of synthetic or natural compounds. For example, numerous means are available for random and directed synthesis of a wide variety of organic compounds and biomolecules, including expression of randomized oligonucleotides, synthetic organic combinatorial libraries, phage display libraries of random peptides, and the like.
  • Alternatively, libraries of natural compounds in the form of bacterial, fungal, plant and animal extracts are available or readily produced. Additionally, natural and synthetically produced libraries and compounds can be readily be modified through conventional chemical, physical, and biochemical means. Further, known pharmacological agents may be subjected to directed or random chemical modifications such as acylation, alkylation, esterification, amidification, etc. to produce structural analogs of the agents.
  • A variety of other reagents also can be included in the mixture. These include reagents such as salts, buffers, neutral proteins (e.g., albumin), detergents, etc. which may be used to facilitate optimal protein-protein and/or protein-nucleic acid binding. Such a reagent may also reduce non-specific or background interactions of the reaction components. Other reagents that improve the efficiency of the assay such as antimicrobial agents, and the like may also be used.
  • The prerequisite for producing intact native polypeptides using E. coli is the use of a strong, regulatable promoter and an effective ribosome binding site. Promoters which may be used for this purpose include the temperature sensitive bacteriophage λpL-promoter, the tac-promoter inducible with IPTG or the T7-promoter. Numerous plasmids with suitable promoter structures and efficient ribosome binding sites have been described, such as for example pKC30 (λpL; Shimatake and Rosenberg, Nature 292:128 (1981), pKK173-3 (tac, Amann and Brosius, Gene 40:183 (1985)) or pET-3 (T7-promoter (Studier and Moffat, J. Mol. Biol. 189:113 (1986)).
  • A number of other suitable vector systems for expressing the DNA according to the invention in E. coil are known from the prior art and are described, for example, in Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory Press (1989)).
  • Suitable E. coil strains which are specifically tailored to a particular expression vector are known to those skilled in the art (Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory Press (1989)). The experimental performance of the cloning experiments, the expression of the polypeptides in E. coli and the working up and purification of the polypeptides are known and are described, for example, in Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory Press (1989). In addition to prokaryotes, eukaryotic microorganisms such as yeast may also be used.
  • For expression in yeast, the plasmid YRp7 (Stinchcomb et al. Nature 282:39 (1979); Kingsman et al., Gene 7:141 (1979); Tschumper et al., Gene 10:157 (1980)) and the plasmid YEp13 (Bwach et al., Gene 8:121-133 (1979)) are used, for example. The plasmid YRp7 contains the TRP1-gene which provides a selection marker for a yeast mutant (e.g., ATCC No. 44076) which is incapable of growing in tryptophan-free medium. The presence of the TRP1 defect as a characteristic of the yeast strain used then constitutes an effective aid to detecting transformation when cultivation is carried out without tryptophan. The same is true with the plasmid YEp13, which contains the yeast gene LEU-2, which can be used to complete a LEU-2-minus mutant.
  • Other suitable marker genes for yeast include, for example, the URA3- and HIS3-gene. Preferably, yeast hybrid vectors also contain a replication start and a marker gene for a bacterial host, particularly E. coli, so that the construction and cloning of the hybrid vectors and their precursors can be carried out in a bacterial host. Other expression control sequences suitable for expression in yeast include, for example, those of PHO3- or PHO5-gene.
  • Other suitable promoter sequences for yeast vectors contain the 5′-flanking region of the genes of ADH I (Ammerer, Methods of Enzymology 101:192-210 (1983)), 3-phosphoglycerate kinase (Hitzeman et al., J Biol. Chem. 255:2073 (1980)) or other glycolytic enzymes (Kawaski and Fraenkel, BBRC 108:1107-1112 (1982)) such as enolase, glycerinaldehyde-3-phosphate-dehydrogenase, hexokinase, pyruvate-decarboxylase, phosphofructokinase, glucose-6-phosphate isomerase, phosphoglucose-isomerase and glucokinase. When constructing suitable expression plasmids, the termination sequences associated with these genes may also be inserted in the expression vector at the 3′-end of the sequence to be expressed, in order to enable polyadenylation and termination of the mRNA.
  • Generally, any vector which contains a yeast-compatible promoter and origin replication and tennination sequences is suitable. Thus, hybrid vectors which contain sequences homologous to the yeast 2 μ plasmid DNA may also be used. Such hybrid vectors are incorporated by recombination within the cells of existing 2 μ-plasmids or replicate autonomously.
  • In addition to yeasts, other eukaryotic systems may, of course, be used to express the polypeptides according to the invention. Since post-translational modifications such as disulphide bridge formation, glycosylation, phosphorylation and/or oligomerization are frequently necessary for the expression of biologically active eukaryotic proteins by means of recombinant DNA, it may be desirable to express the DNA according to the invention not only in mammalian cell lines but also insect cell lines.
  • Functional prerequisites of the corresponding vector systems comprise, in particular, suitable promoter, termination and polyadenylation signals as well as elements which make it possible to carry out replication and selection in mammalian cell lines.
  • In a preferred embodiment of the invention particularly suitable promoters are podocyte specific promoters. A podocyte specific promoter is one that is expressed exclusively in podocytes, such as the podocin promoter.
  • For expression of the DNA molecules according to the invention it is particularly desirable to use vectors which are replicable both in mammalian cells and also in prokaryotes such as E. coil. Vectors derived fnom viral systems such as SV40, Epstein-Barr-virus etc., include, for example, pTK2, pSV2-dhfv, pRSV-neo, pKO-neo, pHyg, p205, pHEBo, etc. (Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Press, N.Y. (1989)).
  • After transformation in suitable host cells, e.g. CHO cells, corresponding transformed cells may be obtained with the aid of selectable markers (thymidine-kinase, dihydrofolate-reductase, green fluorescent protein, etc.) and the corresponding polypeptides are isolated after expression. The host cells suitable for the vectors are known, as are the techniques for transfomiation (nricro-injection, electroporation, calcium phosphate method, etc.) as described, for example, in Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Press, N.Y. (1989).
  • For cloning corresponding DNA fragments in prokaryotic or eukaryotic systems, the selected vector may be cut, for example, with a restriction endonuclease and, optionally after modification of the linearized vector thus formed, an expression control sequence equipped with corresponding restriction ends is inserted. At the 3′-end (in the direction of translation) the expression control sequence contains the recognition sequence of a restriction endonuclease, so that the vector already containing the expression control sequence is digested with the said restriction enzyme and the DNA molecule according, to the invention, provided with ends which fit, can be inserted. It is advantageous to cleave the vector which already contains the expression control sequence with a second restriction endonuclease inside the vector DNA and to insert the DNA molecule provided with the correct ends into the vector fragment produced. The techniques required are described, for example, by Sambrook et al. Molecular Cloning: A Laboratory Manual Cold Spring Harbor Press. N.Y. (1989).
  • Apart from the DNA molecules specified, the invention also relates to processes for preparing the vectors described herein, particularly expression vectors. These vectors are characterized in that a DNA provided with corresponding ends and coding for a functional derivative or a fragment of the protein is inserted into a vector DNA cut with restriction endonucleases and containing the expression control sequences described by way of example, in such a way that the expression control sequences regulate the expression of the DNA inserted. The peptides and antibody agents of the present invention which are obtained by the expression of recombinant DNA or from the native receptor molecule may, of course, also be derivatized by chemical or enzymatic processes.
  • Also within the scope of the invention are kits comprising the compositions of the invention and instructions for use. The kits can further contain at least one additional reagent, such as a chemotherapeutic agent.
  • A kit may comprise a carrier being compartmentalized to receive in close confinement therein one or more container means or series of container means such as test tubes, vials, flasks, bottles, syringes, or the like. A first of said container means or series of container means may contain one or more αbβ3 integrin inhibitors or recombinant vectors for the expression thereof. A second container means or series of container means may contain a second therapeutic, such as, cytotoxic drug or αvβ3 integrin antibodies (or fragment thereof).
  • Kits for use in the therapeutic methods of the invention containing the αvβ3 integrin inhibitors conjugated to other compounds or substances can be prepared. The components of the kits can be packaged either in aqueous medium or in lyophilized form. When the αvβ3 integrin inhibitors or fragments thereof are used in the kits in the form of conjugates in which a label or a therapeutic moiety is attached, such as a radioactive metal ion or a therapeutic drug moiety, the components of such conjugates can be supplied either in fully conjugated form, in the form of intermediates or as separate moieties to be conjugated by the user of the kit.
  • This invention is not limited in its application to the details of construction and the arrangement of components set forth in the following description or illustrated in the drawings. The invention is capable of other embodiments and of being practiced or of being carried out in various ways. Also, the phraseology and terminology used herein is for the purpose of description and should not be regarded as limiting. The use of “including,” “comprising,” or “having,” “containing,” “involving,” and variations thereof herein, is meant to encompass the items listed thereafter and equivalents thereof as well as additional items.
  • The present invention is further illustrated by the following Examples, which in no way should be construed as further limiting. The entire contents of all of the refinences (including literature references, issued patents, published patent applications, and co-pending patent applications) cited throughout this application are hereby expressly incorporated by reference.
  • EXAMPLES Example 1 Modification of Kidney Barrier function by the Urokinase Receptor
  • In this example, the role of uPAR was analyzed in the regulation of structure and function of podocytes during normal and disease conditions. Mice deficient for uPAR20, uPA21, Vn22 and β3 integrin23 were challenged with lipopolysaccharide (LPS), a treatment known to cause FP effacement and proteinuria9. It is shown that uPAR is dispensable for normal renal filtration but required in podocytes but not in endothelial cells for loss of renal permselectivity seen in human and rodent kidney diseases. The proteinuric signal originating from uPAR in podocytes is independent of urokinase and is required for podocyte FP effacement and LPS-induced proteinuria.
  • Results
  • uPAR induction in human and rodent proteinuric diseases
  • We first tested if uPAR protein is expressed in human glomeruli (FIG. 1A) and found expression in many glomerular cells including podocytes which are identified by synaptopoding labeling25. Next, we investigated the expression of uPAR mRNA in human proteinuric renal diseases (FIG. 1B). We performed quantitative real-time PCR using isolated glomeruli from human kidney biopsies26. We analyzed uPAR expression in RNA samples from patients without glomerular disease (con, n=8) and in patients with focal segmental glomerulosclerosis (FSGS, n=14) and diabetic nephropathy (DN, n=20), both conditions with podocyte FP effacement and proteinuria6. We found a low level of glomerular uPAR mRNA expression in patients without glomerular disease (FIG. 1B). In contrast, patients with biopsy-proven FSGS had a significant increase in glomerular uPAR expression (FSGS 0.35±0.17 vs con 0.14±0.02, **p<0.05), (FIG. 1B). An even stronger induction of glomerular uPAR mRNA expression (0.69±0.21 vs con 0.14±0.02, *p<0.012) was found in patients with diabetic nephropathy (FIG. 1B).
  • To test which cells in the glomerulus display increased uPAR expression, we examined the localization of uPAR within the kidney in three different animal models of inducible proteinuria and podocyte FP effacement, including the puromycin aminonucleoside nephrosis model in the rat (PAN)27, the mouse model of LPS-induced transient nephrotic syndrome9 and the NZB/W F1 murine model of systemic lupus erythematosus (SLE)28. Using immunofluorescence and confocal microscopy, we found low expression of uPAR in glomeruli from control rats or mice (FIG. 1C). uPAR was partially localized in podocytes as indicated by double immunofluorescent staining with synaptopodin25. In contrast, expression of uPAR protein in all three proteinuria models was significantly increased in glomerular cells including podocytes as demonstrated by the increased yellow staining pattern resulting from overlap with synaptopodin (FIG. 1C). Of note, in the NZB/W F1 Lupus mouse model, uPAR was preferentially found in nephritic areas of the glomerulus in a more segmental distribution. Such a distribution has been recently reported in marine glomerulonephritis where podocytes can populate cellular crescents12. In addition to glomerular uPAR labeling, we also found expression of uPAR in proximal tubular cells which appeared unchanged under normal and disease conditions (data not shown). Increased uPAR protein expression was also observed in LPS and PAN treated cultured podocytes (FIG. 7B) where uPAR was preferentially located at the cell membrane (FIG. 7C). Analysis of the uPAR ligand Vn revealed a prominent staining of Vn in podocytes of human kidney (FIG. 8A). We detected an induction of Vn labeling in podocytes of PAN rats as well as of LPS and Lupus mice when compared to controls (FIG. 8B). Overall, the expression profiles for Vn in podocytes were similar to the pattern we have observed for uPAR.
  • To define the subcellular localization of uPAR within the glomerular cells including podocytes, we have analyzed uPAR localization in normal and diabetic animals (FIG. 1D), since uPAR mRNA induction is strongest during diabetic nephropathy in humans (FIG. 1B). We carried out semiquantitative immunogold analysis of uPAR and transmission electron microscopy in glomerular walls of normal and 3 months as well as 12 months old diabetic rats in which hyperglycemia was induced by streptozotocin injection29. Under normal conditions, uPAR expression was found in all cell types of the glomerulus (Table 1) including podocytes (FIG. 1D). Morphometrical analysis revealed a homogeneous distribution of uPAR in podocytes, mesangial and endothelial cells (Table 1). 12 months old diabetic rats displayed increased uPAR labeling in all cells of the glomerular wall (FIG. 1E). Endothelial uPAR expression was stronger in luminal and basal membrane aspects when compared to controls. Interestingly, podocyte uPAR was increased in 3 and 12 months diabetic rats but only in basal membrane aspects of the FPs. Mesangial cells expressed high levels of uPAR with no difference under normal and diseased conditions.
  • uPAR in Podocytes is Required for Foot Process Effacement and Proteinuria
  • To explore whether uPAR has a direct role in the regulation of podocyte FP structure and function, we next compared FP morphology of wild type and uPAR-/- mice30 before and after the administration of LPS. Morphologically, there was no diffemace between the structure of podocyte FP in wild type or uPAR-/- mice (FIG. 2A, con). However, 24 h after LPS injection, we found podocyte FP effacement in wild type but not in uPAR-/- mice (FIG. 2A, LPS), suggesting a functional link between the development of podocyte FP effacement and uPAR expression. To test whether the protection from LPS-induced FP effacement could be overcome by restoring glomerular uPAR in uPAR-/- mice, we utilized our previously reported gene delivery protocol to deliver uPAR-cDNA31, 32, 8. 24 h after gene delivery of a plasmid encoding uPAR but not of an empty vector control, we found uPAR expression in glomerular cells including podocytes of uPAR-/- mice, as confirmed by double-immunofluorescent labeling of uPAR and synaptopodin (FIG. 2B). To monitor the expression levels of uPAR after gene delivery, we performed immunoblots. 14 h after uPAR gene delivery, uPAR protein was found in the liver, and at lower expression levels in glomerular extracts (FIG. 2C). The restoration of uPAR did change the morphology of podocyte FPs (FIG. 2D, con).
  • However, concomitant administration of LPS and uPAR-cDNA into uPAR-/- mice resulted in FP effacement (FIG. 2D, LPS), similar to LPS-treated wild type animals (FIG. 2A, LPS). In contrast, we did not find any FP changes in uPAR-/- mice which had received vector control, even after co-administration of LPS (FIG. 2D, LPS).
  • To study the functional consequences of uPAR reconstitution on the development of proteinuria, we analyzed the urine protein excretion of wild type, uPAR-/-, and uPAR-reconstituted uPAR-/- mice before and after LPS injection (FIG. 2E). Whereas PBS treated control mice and uP4R 4- mice did not display any significant proteinuria, the injection of LPS induced significant proteinuria in wild type but not in uPAR-/- mice and uPAR-/- mice which received control plasmids. These data indicate that uPAR-/- mice were protected from urinary protein loss. Most importantly, uPAR-/- mice reconstituted with uPAR-cDNA develop heavy proteinuria but like wild type mice only after LPS injection (FIG. 2E). The degree of proteinuria was comparable in LPS treated wild type and uPAR-cDNA reconstituted uPAR-/- mice. In summary, these data strongly suggest that uPAR is required for the development of LPS-indueed proteinuria in mice.
  • Even so it appears most logical that uPAR exerts its effects on proteinuria development by orchestrating podocyte FP effacement, it seemed still possible that uPAR in glomerular endothelial cells is contributing as well. Thus, we carried out gene-transfer of uPAR cDNA under the control of two different cell-specific promoter allowing cell-specific expression in podocytes (Podocin-uPAR),33 or endothelial cells (ICAM2-uPAR)34 (FIG. 2F). Cell-type specific expression of uPAR was monitored by double-immunofluorescence labeling with synaptopodin or the endothelial marker CD3135 (FIG. 2G). While the expression of uPAR in podocytes was required for LPS-induced proteinuria, the expression of uPAR in endothelial cells was associated with LPS resistance. These results demonstrate that podocyte-uPAR is required and sufficient for LPS induced proteinuria.
  • uPAR Orchestrates Podocyte Motility
  • To begin to understand uPAR function in podocytes, we were considering uPAR's role in cell motility16. Podocyte PP effacement may represent a motile event resulting in spreading of podocyte FPs on the GBM. Thus, we studied podocyte motility in cultured podocytes before and after stable knockdown of uPAR using stable siRNA. The efficiency of uPAR-siRNA was confirmed by both semi-quantitative RT-PCR (FIG. 9A) and Western blotting (FIG. 9B). We then used a modified multi-well Boyden chamber assay to assess the random migration of differentiated podocytes on type I collagen (data not shown) and Vn, a known binding partner of uPAR10 which is induced in proteinuric glomeruli (FIG. 8B). LPS- or PAN-treatment for 24 h significantly promoted migration of wild type podecytes (FIG. 3A, B). In contrast, the knockdown of uPAR significantly decreased the number of migrating podocytes under normal condition and after treatment with LPS or PAN (FIG. 3A, B). These results demonstrate that podocytes migrate in larger number in response to LPS or PAN and that uPAR is necessary for podocyte cell migration.
  • We then analyzed the role of uPAR in the spatial motility of podocytes using a modified scrape wound assay11. Migrated podocytes were counted at different time points and the distance migrated by cells from the wound margin was related to the total width of the scar. Compared to control cells, LPS or PAN treatment significantly promoted podocyte wound closure after 24 h (FIG. 3C, D), a finding which was also obtained in a similar pattern at timepoints 12, 24 and 48 h (FIG. 9C). The addition of external urokinase did not alter podocyte directional migration (FIG. 9D) but the knockdown of uPAR strongly reduced podocyte directed motility before and after administration of LPS or PAN, leaving the wound largely unpopulated (FIG. 3C, D), (FIG. 9C). Together, these data suggest that uPAR is important for random as well as directed podocyte migration,
  • uPAR Activates αbβ3 Integrin in Podocytes
  • Since uPAR is a GPI-anchored protein without a cytoplasmic tail, it is generally believed that signal transduction from uPAR involves lateral interactions with membrane proteins such as integrins16. Most recently, a paper by Madsen et al. described that uPAR induced cell adhesion and migration required Vn binding which can occur independently of uPAR interactions with integrins19. Podocyte motility on Vn is enhanced in a uPAR-dependent fashion (FIG. 3) and Vn is induced in glomeruli during proteinuria (FIG. 8B). Thus, it appeared possible that an uPAR-Vn complex or an uPAR-Vn-integrin complex facilitates podocyte motility and promotes FP effacement in response to LPS. Integrins can be in an inactive or active conformation (FIG. 4A). The latter is stimulated by the association with interacting agonists such as domain 3 of uPAR which is important for a5b1 integrin interaction37, 38. Degryse and colleagues recently identified an integrin-interacting sequence in domain 2 of uPAR, that acts as a new chemotactic epitope activating αbβv3-dependent signaling pathways39. Given our findings of uPAR and Vn in the glomerulus, we were particulary interested in the Vn receptor αbβ3 integrin36. Indeed, the localization of αvβ3 integrin in podocyte FPs (FIG. 4B)31 and the distribution of uPAR (FIG. 4B) was similar. In addition, uPAR interacts with β3 integrin (FIG. 10A). Thus, we hypothesized that αvβ3 integrin may provide a functional link between uPAR, podocyte migration and proteinuria development. The genetic deletion of β3 integrin or of the αvβ3 integrin ligand Vn resulted in protection from proteinuria when challenged with LPS (FIG. 4C). This means that both, Vn and β3 integrin are required for LPS-induced proteinuria but either one is dispensable for normal renal development and function (FIG. 4C). Given the extent of published uPAR interactome and the importance of α3 β1 integrin in podocyte development40, we also analyzed the potential contribution of α3β1 in integrin in the uPAR signaling cascade in podocytes and utilized a cDNA for uPAR which encodes the uPAR mutant D262A that is unable to bind α3β1 integrin in humans38 and mouse (FIG. 10B). The expression of this type of uPAR in podocytes led to the development of LPS-induced proteinuria supporting the idea that uPAR in podocytes preferntially signals through αbβ3 integrin. Since uPAR is involved in pathways dependent and independent of urokinase16, we also explored the contribution of urokinase on uPAR-dependent proteinuria pathways. We utilized UPA-/- mice and treated them with LPS. Interestingly, these mice developed proteinuria suggesting that uPA is not required for the LPS-mediated effects of uPAR on the kidney filtration barrier (FIG. 4C).
  • uPAR-/-, β3 integrin-/- and Vn-/- mice huge no overt renal phenotypes under normal conditions suggesting that uPAR signaling in podocytes is not required for normal glomerular filtration. On the other hand, all these molecules are required for the development of urinary protein loss. Based on this, we reasoned that changes in activation of αvβ3 integrin under disease conditions may be a cause for the increased podocyte motility and FP effacement after the administration of LPS. To explore this idea, we next studied the expression of total and active β3 integrin in kidney sections from wild type and uPAR-/- mice. Total podocyte β3 integrin expression was visualized by double labeling of β3 integrin with the podocyte marker synaptopodin wild type and uPAR-/- mice before and after injection of LPS (FIG. 5A). The expression of β3 integrin in podocytes was unchanged in wild type and uPAR-/- mice before and after LPS administration (FIG. 5A). We next studied the presence of the active form of β3 integrin which can be detected by the use of the well-defined antibody AP5. This antibody recognizes an N-terminal epitope of β3 integrin that is only accessible when the integrin is in its active conformation41. To test if AP5 was capable to detect active β3 integrin in podocytes, we first evaluated the effect of divalent cations on the binding pattern of AP5 to β3 integrin by flow cytometry in normal (data not shown) and LPS treated cultured podocytes (FIG. 5B). We found a similar calcium-dependent binding pattern of AP5 as previously reported for the binding of AP5 to β3 integrin in other cells42. Immunofluorescence of active β3 integrin with AP5 antibody in glomeruli suggest a low baseline activity of β3 integrin in wild type and uPAR-/- mice under normal conditions. This finding suggests that αbβ3 integrin has a low basal activity even in the absence of uPAR. LPS treatment of wild type mice was associated with a strong induction of podocyte AP5 labeling (FIG. 5C, WT+LPS). This induction was absent in LPS treated uPAR-/- mice (FIG. 5C, uPAR-/-+LPS). We also observed colocalization of AP5 labeling at sites of uPAR overexpression in podocytes but not in podocytes in which uPAR was downregulated using siRNA (FIG. 5D). As an additional readout for active β3 integrin, we analyzed the activity of the small GTPases cdc42 and Rac1 in glomerular lysates from wild type and uPAR-/- mice before and after administration of LPS (FIG. 5E)43. The activity of Rac1 was increased in LPS treated wild type but not uPAR-/- mice (FIG. 5E, upper panel). We observed a similar induction pattern for cdc42 activity (FIG. 5E, lower panel). Of note, we could not detect activity for RhoA under normal and disease conditions in wild type or uPAR-/- mice (data not shown). In some experiments we co-injected wild type mice with LPS and cyclo-[Arg-Gly-Asp-D-Phe-Val] RGDfV known from clinical cancer trials as Cilengitide which is used as a specific inhibitor of αvβ3 integrin44. This treatment inhibited the induction of active Rac1 and cdc42 in glomeruli of LPS treated mice (FIG. 5E). Together, these findings support the concept that uPAR is required for the activation of podocyte αvβ3 integrin after LPS treatment.
  • We next explored possible subcellular compartments where uPAR and β3 integrin can associate with each other. Studies suggest that many aspects of the podocyte FP membrane as well as the SD are rich in cholesterol45 and several SD proteins like podocin and nephrin are associated with lipid rafts46. Plasma membrane lipid rafts help to compartmentalize signal transduction events within different regions24 and uPAR as a GPI-anchored protein is known to be found in lipid rich membrane compartments16. We wondered whether uPAR and β3 integrin associate together within lipid rafts in podocytes. We therefore performed sucrose density gradient assays of whole cellular extracts of cultured podocytes before and after LPS administration (FIG. 6A). We found that uPAR and β3 integrin are mainly associated with the non-raft fractions in control podocytes. However, 24 h after LPS administration, both uPAR and β3 integrin were more enriched within the lipid raft fraction (FIG. 6A). The functional association of uPAR and β3 integrin within lipid rafts was further confirmed by the observation that disruption of lipid rafts using Methyl-β-cyclodextrin (MBCD)47 abrogated the activation of β3 integrin in response to LPS in cultured podocytes as revealed by reduced AP5 staining (FIG. 6B). These data suggest that β3 integrin can be activated by uPAR within lipid rich domains of the podocyte plasma membrane.
  • Pharmacological Interference with αvβ3 Integrin Modifies Proteinutia in Mice
  • If the activation of αvβ3 integrin is an important downstream signal that mediates the uPAR-induced cellular events in podocytes leading to proteinuria, expression of a constitutively active β3 integain should be sufficient to induce proteinuria even in the absence of uPAR. Therefore, we utilized a β3 integrin cDNA which encodes a protein lacking aminoacids 616 to 690 of the carboxy-terminal regions of the β3 ectodomain. This mutation confers constitutive activity to the β3 protein and is expressed at the cell surface48. We performed gene transfer of this active β3 integrin construct into uPAR-/- mice. 24 h after gene delivery of active β3 integrin, we monitored activity of β3 integrin in podocytes by immunofluorescent double labeling of synaptopodin and WOW-1 antibody. WOW-1 antibody is known to recognize constitutive active β3 protein49. uPAR-/- mice positive for WOW-1 labeling in the podocyte (FIG. 6D) developed proteinuria (FIG. 6D), while littermates which received normal β3 integrin or vector control had low podocyte labeling with WOW-1 antibody and no proteinuric response (FIG. 6D).
  • We also performed blocking experiments with an antibody inhibiting β3 integrin function. Wild type mice which were co-injected with LPS and a monoclonal anti-β3 integrin antibody failed to develop proteinuria in response to LPS (FIG. 6E). This blocking antibody was also able to reduce podocyte motility significantly during the course of LPS treatment in vitro (FIG. 6D) connecting increased podocyte motility in vitro with the development of proteinuria in vivo.
  • Finally, we used different concentrations of cyclo-[Arg-Gly-Asp-D-Phe-Val] RGDfV (Cilengitide) to specifically block αvβ3 integrin. Cyclo-RGDfV was injected in LPS treated wild type mice (FIG. 6G). While the administration of cyclo-RGDfV in normal mice had no effect, mice which received LPS and cyclo-RGDfV displayed an attenuated course of proteinuria in a dose dependent manner when compared to control mice that had received only LPS. These data show that activity of αvβ3 integrin is important for proteinuria development and opens novel avenues for the modulation of proteinuria by αvβ3 integrin interference in podocytes.
  • Methods Antibodies:
  • The following antibodies were used in this study: anti-active β3 integrin (AP5), (GTI); anti-WOW-1 fragment antigen binding (Fab)49 (kind gift from Sanford Shattil, La Jolla, Calif.); anti-CD61 blocking antibody (BD Pharmagen); anti-CD31 (ER-MP12) and anti-Vn (H-270), anti-uPAR (FL-290) (Santa Cruz Biotechnology, Inc); anti-uPAR-1 (R&D Systems); anti-α-tubulin (Calbiochem); anti-β3 integrin (Chemicon International); anti-CD61 (Sigma); anti-caveolin (Sigma); anti-GAPDH (Sigma); anti-Flag (Sigma); anti-HA (Sigma); anti-transferrin receptor and anti caveolini (Sigma); anti-synaptopodin mouse monoclonal antibody (G1), anti-synaptopodin rabbit polyclonal (NT), (Peter Mundel, N.Y.).
  • Animals and treatments:
  • All animal studies were approved by the Subcommittee on Research Animal Care of the Massachusetts General Hospital. uPAR-/- mice and uPA-/- mice on a mixed background of 75% C57BL/6 and 25% 129 Swiss were obtained from University of Leuven, Belgium; Vn-/- mice (C57BL/6) were obtained from Dr. David Ginsburg, University of Michigan; β3 integrin-/- mice (C57BL/6) were obtained from Dr. Raghuram Kalluri, Boston. C57BL/6 and 129 Swiss mice were purchased from Jackson Laboratory (Bar Harbor). The LPS mouse model was utilized as previously described9. The induction of B7-1 was used as a control for the effectiveness of LPS. Rat PAN nephrosis model was set up by a single intraperitoneal injection of puromycin (15 mg/100 g of body weight, Sigma-Aldrich) into Spragne-Dawley rats as described before27. NZB/W F1 mice were purchased from the Jackson Laboratory and analyzed after 20 weeks when proteinuria and Lupus glomerulonephritis were present. As diabetic nephropathy rat model, we used Sprague Dawley rats treated with Streptozotocin. Hyperglycaemic state was induced by an intraperitoneal Streptozotocin injection (50-70 mg/kg body weight, in citrate buffer 10 mmol/L, pH 4.5). The hyperglycaemic state developed within 48 h and was maintained during the lifetime of the animals. No insulin administration was required. Glycosuria was evaluated with test strips of the Uriscan TM (YD Diagnostics, VWR, Montréal, QC, Canada) and glycaemia with the AccuSoft Monitoring System (Roche Diagnostics, Laval, QC, Canada). At the end of the study, glycaemia values were 43±0.53 and 8.5±0.7 mmol/L for 3 months-old and 12 months-old normoglycaemic animals, respectively, and 21.2±0.9 and 33.9±4.0 mmol/L for 3 months-old and 12 months-old diabetic animals, respectively.
  • Patients: Microdissected glomeruli from 34 patients with proteinuric diseases and 8 control subjects were analyzed. Patients were stratified according to their histological diagnosis into focal and segmental glomerulosclerosis (FSGS; n=14), and diabetic nephropathy (DN, n=20). For control biopsies, renal tissue was derived from pretransplantation kidney biopsies during cold ischemia time from 7 living and 1 cadaveric donors (n=8).
  • Quantitative real-time PCR: TaqMan real-time RT-PCR was done as previously reported26. In brief, commercially available predeveloped TaqMan assay reagents (Applied Biosystems) were used for uPAR mRNA analysis. The mRNA expression of uPAR was related to that of synaptopodin, which was used as a podocyte reference gene. Using this approach, the confounding factor of alterations in the proportion of podocyte cell number per total glomerular cells was counterbalanced, and only RNA from the podocyte compartment of the glomerulus was integrated in the analysis, as demonstrated recently26.
  • Immunohistochemistry: Human glomerular biopsies were fixed in cold acetone and stained with anti-uPAR antibodies, anti-Vn antibody and monoclonal anti-synaptopodin antibody following standard protocols8. Mouse or rat kidneys were harvested and snap-frozen. Fixation and sectioning were performed following standard protocols8. For double-immunofluorescent staining, sections were blocked for 30 min at room temperature, and incubated with antibodies directed against: synaptopodin (G1, NT), β3 integrin (AP5, WOW-1), uPAR, Vn, CD31. After washing with PBS, sections were incubated with appropriate fluorophore conjugated secondary antibodies (Molecular Probes) for 50 min. Thereafter, sections were washed with PBS, and H2O and mounted for analysis with a confocal microscope (Bio-Rad Laboratories). For WOW-1 labeling, uPAR-/- mice were injected with β3Δ616-690 or wild type β3 integrin constructs. 14 h after iniection, mice were sacrificed and kidney was snap-frozen. Cyro-sections were cut at 4 μm and fixed with cold acetone for 10 min before incubated with WOW-1 Fab for 1 h. After washing with PBS, the sections were then incubated with the secondary antibody, anti-mouse Fab conjugated with 488 (Invitrogen) for 50 min and analyzed by confocal microscopy.
  • Cultured podocytes were immunolabeled as described previously8.
  • Transmission electron microscopy (TEM), Immunoelectron microscopy (IEM) and Morphometry: TEM and IEM were performed according to the standard protocols8. For morphometry of uPAR labeling across the glomerular wall, we used renal tissues from 3 and 12 months-old hyperglycaemic animals with the corresponding age-matched normoglycaemic animals (3-4 animals per group). Small pieces of renal cortex were sampled from the anaesthetized animal (urethane, 1 g per kg body weight). The tissue samples were immediately fixed by immersion in periodate-lysine-paraformaldehyde solution, dehydrated in graded methanol and embedded in Lowicryl, following protocols described previously52.
  • For immunogold labeling, the grids carrying the ultra thin tissue sections were incubated on a drop of a saturated solution of sodium metaperiodate for 10 min, washed with distilled water, transferred on a drop of 0.15 M glycine for 10 min and washed with PBS. Grids were then incubated on a drop of ovalbumin 1% for 5 min and transferred to the diluted anti-uPAR antibody (1:10) overnight at 4° C. The grids were washed with PBS and incubated on a drop of protein A-gold complex for 30 min at room temperature. The grids were then washed with PBS (3×5 min) and distilled water (1×5min), dried and contrasted with uranyl acetate. Specificity of the immunolabelings was evaluated by control experiments, omitting the specific antibody step, replacing it with PBS.
  • Micrographs of immunolabeled renal glomeruli were recorded (>20 micrographs per animal and timepoint), printed and evaluated through morphometrical techniques52. We measured the length of the endothelial luminal and abluminal membranes, that of podocyte basal and apical membranes and that of the mesangial cells, the number of gold particles associated to each of those membrane domains was then counted to calculate the density of labeling. Results are expressed as number of gold particles per μm of membrane (mean values±SEM). The measurements were performed by direct planimetry and particle counting using an image processing system (Videoplan 2, Carl Zeiss Inc., Toronto, Canada).
  • GTPase activity assay:
  • Rho family small GTPase activity in glomerular lysates was measured by Rho/Rac/Cdc42 activation assay kit (Cell Biolabs, INC.) following the manufacturer's protocol. Isolated glomeruli from wild type and uPAR-/- mice, some of them treated with LPS (t=0) and with RGDfV every 8 hours for 24 hours were obtained by standard sieve technique25. 2 mg of glomerular lysate were used to prepare for positive or negative control by incubating with 10 μl of GTPγS or GDP respectively. For small GTPase pull-down assay, the same amount (2 mg) of glomerulus lysates from different treatments were incubated with 40 μl of Rhotekin RBD or PAK PBD agarose bead slurry for 1 h at 4° C. After washing 3 times with 1× assay buffer, beads were harvested and resuspended in LDS sample buffer. Samples were heated for 10 min at 70° C. before SDS-PAGE. After electrophoresis and protein transfer, the membranes were blocked with 5% milk, and incubated with anti-Rho, anti-Rac, and anti-Cdc42 antibodies for 1 h, followed by second antibody incubation and detection with ECL (Pierce). For loading control, 100 μg of glomerular lysates from different treatments were applied and immunoblotted with the same anti-Rho, anti-Cdc42 and anti-Rac antibodies as used above.
  • In vivo Gene delivery:
  • uPAR-cDNA, plasmids were introduced into uPAR-/- mouse using the TransIT in vivo gene delivery system (Mirus) as described previously32, 8. The following cDNAs and vector constructs were used for gene delivery: uPAR; mutated uPAR D262A38 (kind gift from Y Wei and HA Chapman, University of California, San Francisco); β3 integrin and the constitutively active form of β3 integrin, D616-690 (β3Δ616-690)48; Podocin promoter vector p2.533 (a kind gift from Dr. LB Holzman, University of Michigan) for Pod-uPAR; ICAM-2 promoter vector34 for ICAM-2-uPAR. Tissues were analyzed for expression as described previously8.
  • siRNA: Mouse uPAR siRNA (sense: CTTCCTGAAGTGTTGCAACTA) was constructed and inserted into a pRNA-H1.2/Neo vector (Genescript). Stable transfection was done with podocytes maintaining at 33° C. by Lipofectamine 2000 (Invitrogen). Positive clones were selected by G418 (Sigma-Aldrich) at 500 μg/ml. For further experiments with uPAR siRNA, cells were grown under non-permissive conditions for 10-14 days before proceeded with migration experiments.
  • Flow cytometry:
  • To determine the activity of AP5 in mouse podocytes, cells were cultured under non-permissive conditions for 10 days. Then medium was discarded and the cells were washed gently with PBS without Ca2+ and Mg2+. The cells were then exposed to LPS right before they were incubated with various amounts of calcium in the presence of 5 μg/ml AP5 for 1 h: 1 mM EDTA, PBS without Ca2+ or Mg2+; 0.1 nM Cacl2; 0.4 mM CaCl2; 1 mM CaCl2; 2 mM CaCl2. The cells were then harvested by cell scraper, washed and subjected to flow cytometry analysis.
  • Cell culture and transient transfection:
  • HEK 293 cells were seeded on a 100 mm culture dish and maintained at 37° C. in DMEM with 10% FBS. Upon 90% confluent, cells were transtected with constructs encoding uPAR or β3 integrin by Lipofectamine 2000 (Invitrogen). 24 h after transfection, cells were harvested for further experiments. Wild type podocytes were differentiated by culturing at 37° C. for at least 10 days (10 days for transfection and migration assay, 14 days for other experiments).
  • Western blot and Co-immunoprecipitation:
  • For Western blotting, podocytes or extracted glomeruli were lysed in RIPA buffer containing a cocktail of protease inhibitors. The lysate was centrifuged for 20 min at 12,000 rpm and the yielded supernatant was further used. Bradford assay was performed to ensure equal amount of loading. Proteins were separated and then transferred to a PVDF membrane. After blocking for 30 min with 5% milk, the membrane was incubated with primary antibody for 1 h, followed by secondary antibody for overnight 4° C. After washing, the membrane was visualized by chemiluminescence immunoblot detection kit (Pierce).
  • For Co-IP, HEK cells were co-transfected with constructs encoding uPAR, and β3 integrin for 24 h. Then cells were lysed in RIPA buffer with a cocktail of protease inhibitors. The lysate was incubated with 25 μl of Flag or HA agarose beads (Sigma) at 4° C., overnight after a pre-cleaning. The beads were then washed 5 times with RIPA buffer and were eluted by heating at 70° C. for 10 min. After a brief centrifugation, the supernatants were ready for Western blotting.
  • Migration assay:
  • Podocyte migration was analyzed using a 12-well chemotaxis chamber (Neuro Probe) according to the manufacturer's protocol. In brief differentiated podocytes were treated with 50 μg/ml of LPS or PAN for 24 h and then harvested for the migration assay. Bottom plates were coated with vitronectin or type I collagen, while upper plates were loaded with equal number of cells (5×104) suspended in medium. The chamber was incubated at 37° C. for 4 h before the membrane was taken out and stained with diamidino phenylindole (DAPI). The average number of migrated cells was counted in four fields in six independent experiments.
  • To study the directional movement of podocytes, a wound healing assay was performed. Briefly, podocytes were seeded on Vn or type I collagen coated cover-slips and cultured for 10 days at 37° C. before treated with LPS or PAN for 12-48 h. To create a scrape wound approximately 0.1 cm wide, the narrow end of a P1000 pipette tip was perpendicularly pushed through the monolayer. Then, coverslips were washed twice with PBS and incubated in medium. After incubation, cells were fixed with 2% PFA and stained with DAPI for analysis. The cell migratory distance was calculated by averaging the distance from the wound edge to the maximally migrated cell in five distinct border zones.
  • Sucrose gradient ultracentrifugation:
  • Podocyte lysate was overlaid with a sucrose step gradient and centrifuged for 20 h at 120,000 g at 4° C. in a swing-out rotor. 10 fractions (1 ml each) were collected starting from the top and analyzed by Western blotting with rafts, non-rafts markers, as well as uPAR and β3 antibodies.
  • Blockade of β3 integrin in animals:
  • Cyclo-RGDfV (Biomol) was injected in mice at 1, 5 and 20 mg per kg body weight i.v. in mice every 8 h for three times. Control mice received the same amount of PBS instead or the control peptide cyclo-RAD (Biomol), (data not shown). Immediately after the first injection, 200 □g of LPS was injected intraperitoneally into each mouse to induce proteinuria. 24 h after LPS injection, urine was collected for Braford assay.
  • For blockade of β3 integrin with the antibody CD61, wild type mice were randomly divided into three groups (n=3 for each group) and injected with PBS alone (control), LPS+anti-CD61 or LPS+IgG isotype. The antibody was administered 4 h after LPS injection through tail vein, with a final concentration of 10 □g/ml. Urine was collected at time points 0, 4 and 24 h and analyzed by Bradford assay. Data represented in the figure was based on sample obtained after 24 h.
  • Statistical analysis:
  • Statistical analyses were performed by using a Student paired or non-paired t-test when appropriate, and the null hypothesis was rejected at the 0.05 level. Values were presented as Mean±SD, unless stated otherwise. Statistical comparisons for the morphometrical experiments were performed using the Student t test and the Mann-Whitney U tests.
  • Example 2: Effect of Cyclo-RGDfV on Recovery of Proteinuria
  • The following example showed that CycloRGDfV also reduces already existing proteinuria (FIG. 11). B6 mice were injected LPS twice (0, 24 h) with LPS to induce and maintain proteinuria. 48 h after first injection cycle-RGD (n=6) was administered (25 mg/kg body weight) through tail vein. Control (n=5) received the same amout of PBS. 0, 48 h, 66 h urine was collected for Braford assay. The diagram shows the fold change of urinary protein at timepoint 66 h, *p<0.014.
  • Example 3: Effect of S247 on Recovery of Proteinuria
  • The following example shows that S247 also reduces already existing proteinuria. B6 mice are injected twice (0, 24 h) with LPS to induce and maintain proteinuria. 48 h after first inection, S247 (n=6) is administered (25 mg/kg body weight) through tail vein. Control (n=5) received the same amout of PBS. 0, 48 h, 66 h urine was collected for Braford assay. Reduction of urinary protein at timepoint 66 h by S247 is observed.
  • Figure US20160296592A1-20161013-C00002
  • Tablet 1
  • Quantitative analysis of uPAR expression and localization in normal and diabetic glomeruli. uPAR is induced in podocyte foot processes of 3 and 12 months old diabetic rats.
  • TABLE 1
    uPAR morphometrical analysis
    Endothelial
    Luminal Endothelial Podocyte Podocyte Mesangial
    Membrane Basal Membrane Luminal Membrane Basal Membrane Membrane
    NORMAL 3 months 0.26 ± 0.06* 0.20 ± 0.05 0.24 ± 0.03 0.19 ± 0.03 0.38 ± 0.05
     (216.01 μm)** (213.89 μm) (576.86 μm) (258.95 μm) (196.62 μm)
    NORMAL 12 months 0.23 ± 0.06  0.18 ± 0.04 0.27 ± 0.05 0.19 ± 0.05 0.30 ± 0.06
    (144.63 μm) (141.51 μm) (288.68 μm) (138.03 μm) (240.94 μm)
    DIABETIC 3 months 0.38 ± 0.05§ 0.27 ± 0.06 0.33 ± 0.03  0.29 ± 0.04§ 0.42 ± 0.06
    (212.17 μm) (210.15 μm) (519.05 μm) (267.88 μm) (213.83 μm)
    DIABETIC 12 months 0.36 ± 0.09§  0.37 ± 0.08§ 0.24 ± 0.06  0.27 ± 0.03§ 0.41 ± 0.08
    (219.79 μm) (234.75 μm) (586.03 μm) (256.68 μm) (253.73 μm)
    CONTROL of 0.02 ± 0.01  0.05 ± 0.03 0.05 ± 0.04 0.04 ± 0.03 0.07 ± 0.03
    SPECIFICITY  (95.06 μm)  (89.29 μm) (133.65 μm)  (75.57 μm) (172.70 μm)
    *particles per μm of membrane
    **total length of membrane measured
    §Significantly different from corresponding value of normal animals *p < 0.005, n = 3 animals for each time point
  • REFERENCES
  • 1. Zandi-Nejad, K., Eddy, A. A., Glassock, R. J. & Brenner, B. M. Why is proteinuria an ominous biomarker of progressive kidney disease? Kidney Int Suppl, S76-89 (2004).
  • 2. Kerjaschki, D. et al. A. beta 1-integrin receptor for fibronectin in human kidney glomeruli. Am J Pathol 134, 481-9 (1989).
  • 3. Kreidberg, J. A. Functions of alpha3beta1 integrin. Curr Opin Cell Biol 12, 548-53. (2000).
  • 4. Regele, H. M. et al. Glomerular expression of dystroglycans is reduced in minimal change nephrosis but not in focal segmental glomerulosclerosis. J Am Soc Nephrol 11, 403-12 (2000).
  • 5. Reiser, J., Kriz, W., Kretzler, M. & Mundel, P. The glomerular slit diaphragm is a modified adherens junction. J Am Soc Nephrol 11, 1-8 (2000).
  • 6. Durvasula, R. V. & Shankland, S. J. Podocyte injury and targeting therapy: an update. Curr Opin Nephrol Hypertens 15, 1-7 (2006).
  • 7. Tryggvason, K., Patrakka, J. & Wartiovnara, J. Hereditary proteinuria syndromes and mechanisms of proteinuria. N Engl J Med 354, 1387-401 (2006).
  • 8. Sever, S. et al. Proteolytic processing of dynamin by cytoplasmic cathepsin L is a mechanism for proteinuric kidney disease. J Clin Invest 117, 2095-2104 (2007).
  • 9. Reiser, J. et al. Induction of B7-1 in podocytes is associated with nephrotic syndrome J Clin Invest 113, 1390-7 (2004).
  • 10. Reiser, J. et al. Podocyte Migration during Nephrotic Syndrome Requires a Coordinated Interplay between Cathepsin L and {alpha}3 Integrin. J Biol Chem 279, 34827-34832 (2004).
  • 11. Asanuma, K. et al. Synaptopodin orchestrates satin organization and cell motility via regulation of RhoA signalling. Nat Cell Biol (2006).
  • 12. Moeller, M. J. et al. Podocytes populate cellular crescents in a murine model of inflammatory glomerulonephritis. J Am Soc Nephrol 15, 61-7 (2004).
  • 13. Seiler, M. W., Venkatachalam, M. A. & Cotran, R. S. Glomerular epithelium: structural alterations induced by polycations. Science 189, 390-3 (1975).
  • 14. Gadea, G., de Toledo, M., Anguille, C. & Roux, P. Loss of p53 promotes RhoA-ROCK-dependent cell migration and invasion in 3D matrices. J Cell Biol 178, 23-30 (2007).
  • 15, Wei, Y. et al. Regulation of integrin function by the urokinase receptor. Science 273, 1551-5 (1996).
  • 16. Blasi, F. & Carmeliet, P. uPAR: a versatile signalling orchestrator. Nat Rev Mol Cell Biol 3, 932-43 (2002).
  • 17. Alfano, M., Sidenius, N., Panzeri, B., Blasi, F. & Poli, G. Urokinase-urokinase receptor interaction mediates an inhibitory signal for HIV-1 replication. Proc Natl Acad Sci U S A 99., 8862-7(2002).
  • 18. Wei, Y. et al. Identification of the urokinase receptor as an adhesion receptor for vitronectin. J Biol Chem 269, 32380-8 (1994).
  • 19. Madsen, C. D., Ferraris, G. M. , Andolfo, A., Cunningham, O. & Sidenius, N. uPAR-induced cell adhesion and migration: vitronectin provides the key. J Cell Biol 177, 927-39 (2007).
  • 20. Dewerchin, M. et al. Generation and characterization of urokinase receptor-deficient mice. J Clin Invest 97, 870-8 (1996).
  • 21. Carmeliet P. et al. Biological effects of disruption of the tissue-type plasminogen activator, urokinase-type plasminogen activator, and plasminogen activator inhibitor-1 genes in mice, Ann N Y Acad Sci 748, 367-81; discussion 381-2 (1995).
  • 22. Zheng X., Saunders, T. L., Camper, S. A., Samuelson, L. C. & Ginsburg, D. Vitronectin is not essential for normal mammalian development and fertility. Proc Natl Acad Sci USA 92, 12426-30 (1995).
  • 23. Sund, M. et al. Function of endogenous inhibitors of angiogenesis as endothelium-specific tumor suppressors. Proc Natl Acad Sci U S A 102, 2934-9 (2005).
  • 24. Simons, K. & Toomre, D. Lipid Rafts and Signal Transduction, Nat Rev Mol Cell Biol 1, 31-39. (2000).
  • 25. Mundel, P. et al. Synaptopodin: an actin-associated protein in telencephalic dendrites and renal podocytes. J Cell Biol 139, 193-204 (1997).
  • 26. Schmid, H. et al. Validation of endogenous controls for gene expression analysis in microdissected human renal biopsies. Kidney Int 64, 356-60 (2003).
  • 27. Nakamura, T., Ebihara, I., Shirato, I., Tomino, Y. & Koide H. Modulation of basement membrane component gene expression in glomeruli of aminonucleoside nephrosis. Lab Invest 64, 640-7 (1991).
  • 28. Kelley V. E. & Cavallo, T. An ultrastructural study of the glomerular slit diaphragm in New Zealand black/white mice. Lab Invest 35,213-20 (1976).
  • 29. Mauer, S. M. et al. Effects of kidney and pancreas transplantation on streptozotocin-induced malignant kidney tumors in rats. Cancer Res 34, 1643-5 (1974).
  • 30. Rijneveld, A. W. et al. Urokinase receptor is necessary for adequate host defense against pnemuccoccal pneumonia. J Immunol 168, 3507-11 (2002).
  • 31. Mayer, G., Boileau, G. & Bendayan, M. Furin interacts with proMT1-MMP and integrin alphaV at specialized domains of renal cell plasma membrane. J Cell Sci 116, 1763-73 (2003).
  • 32. Moller, C. C. et al. Induction of TRPC6 Channel in Acquired Forms of Proteinuric Kidney Disease. J Am Soc Nephrol (2006).
  • 33. Moeller, M. J., Sanden, S. K., Soofi, A., Wiggins, R. C. & Holzman, L. B. Podocyte-specific expression of cre recombinase in transgenic mice. Genesis 35, 39-42 (2003).
  • 34. Velasco, B. et al. Vascular gene transfer driven by endoglin and ICAM-2 endothelial-specific promoters. Gene Ther 8, 897-904 (2001).
  • 35. Zeisberg, E. M. et al. Endothelial-to-mesenchymal transition contributes to cardiac fibrosis. Nat Med 13, 952-61 (2007).
  • 36. Arnaout, M. A., Mahalingam, B. & Xiong, J. P. Integrin structure, allostery, and bidirectional signaling. Annu Rev Cell Dev Biol 21, 381-410 (2005).
  • 37. Chaurasia, P. et al. A region in urokinase plasminogen receptor domain III controlling a functional association with alpha5beta1 integrin and tumor growth. J Biol Chem 281, 14852-63 (2006).
  • 38. Wei, Y. et al. Urokinase receptors are required for alpha 5 beta 1 integrin-mediated signaling in tumor cells. J Biol Chem 282, 3929-39 (2007).
  • 39. Degryse, B., Resnati, M., Czekay, Loskutoff D. J. & Blasi, F. Domain 2 of the urokinase receptor contains an integrin-interacting epitope with intrinsic signaling activity: generation of a new avβ3 integrin inhibitors. J Biol Chem 280, 24792-803 (2005).
  • 40. Kreidberg, J. A. et al. Alpha 3 beta 1 integrin has a crucial role in kidney and lung organogenesis. Development 122, 3537-47 (1996).
  • 41. Pelletier, A. J., Kunicki, T. & Quaranta, V. Activation of the integrin alpha v beta 3 involves a discrete cation-binding site that regulates confirmation. J Biol Chem 271, 1364-70(1996).
  • 42. Honda, S. et al. Topography of ligand-induced binding sites, including a novel cation-sensitive epitope (AP5) at the amino terminus, of the human integrin beta 3 subunit. J Biol Chem 270, 11947-54 (1995).
  • 43. Dormond, O., Foletti, A., Paroz, C. & Ruegg, C. NSAIDs inhibit alpha V to 3 integrin-mediated and Cdc42/Rac-dependent endothelial-cell spreading, migration and angiogenesis. Nat Med 7, 1041-7 (2001).
  • 44. Cai, W. & Chen, X. Anti-angiogenic cancer therapy based on integrin alphavbeta3 antagonism. Anticancer Agents Med Chem 6, 407-28 (2006).
  • 45. Orci, L., Singh, A. Amherdt, M., Brown, D. & Perrelet, A. Microheterogeneity of protein and sterol content in kidney podocyte membrane. Nature 293, 646-7 (1981).
  • 46. Schwarz, K. et al. Podocin, a raft-associated component of the glomerular slit diaphragm, interacts with CD2AP and nephrin. J Clin Invest 108, 1621-9 (2001).
  • 47. Keller, P. & Simons, K. Cholesterol is required for surface transport of influenza virus hemagglutinin. J Cell Biol 140, 1357-67 (1998).
  • 48. Butta, N. et al. Disruption of the beta3 663-687 disulfide bridge confers constitutive activity to beta3 integrins. Blood 102, 2491-7 (2003).
  • 49. Pampori, N. et al. Mechanisms and consequences of affinity modulation of integrin alpha(V)beta(3) detected with a novel patch-engineered monovalent ligand. J Biol Chem 274, 21609-16 (1999).
  • 50. de Jong, P. E. & Brenner, B. M. From secondary to primary prevention of progressive renal disease: the case for screening for albuminuria. Kidney Int 66, 2109-18 (2004).
  • 51. Silberman, S., Janulis, M. & Schultz, R. M. Characterization of downstream Ras signals that induce alternative protease-dependent invasive phenotypes. J Biol Chem 272, 5927-35 (1997).
  • 52. Gugliucci, A. & Bendayan, M. Reaction of advanced glycation endproducts with renal tissue from normal and streprozotocin-induced diabetic rats: an ultrastructural study using colloidal gold cytochemistry. J Histochem Cytochem 43, 591-600 (1995).
  • It has now been found that compounds of formula (I)
      • cyclo {NX1—R1—CO—NX2—R2—CO—NX3—R3—CO—NX4—R4—CO—NX5—R5—CO]
        where:
      • R1 is selected from:
      • CH(CH2)3NHC(NH)NH2; C[CHnFm](CH2)3NHC(NH)NH2
      • R2 is the group CH2; CH2-CH2;
  • Figure US20160296592A1-20161013-C00003
      • R3 is selected from CHCH2COOH; C[CHnFm]CH2-COOH;
      • R4 is selected from CH-CH2-Ph; C[CHnFm]CH2-Ph; CH-CH2-(4-OH)Ph;
      • CH-CH2-(4-OMe)Ph; CH-CH2-(4-F)Ph; CH-CH(OH)-Ph; C(CH3)2; CH-C(CH3)3; CH-CH2-COOH;
  • Figure US20160296592A1-20161013-C00004
      • R5 is selected from CH-CH2-Ph; C[CHnFm]CH2-Ph; CH-CH(CH3)2; C[CHnFm]CH(CH3)2; CH-C(CH3)3;
      • or, the NX4-R4-Co-NX5-R5CO group is 3-aminomethyl-benzoyl n+m=3
      • X1-X5, which may be the same or diffetent, are H, (CH2)n-CH3; (CH2)n-
  • Figure US20160296592A1-20161013-C00005
      • CHF2; (CH2)n-CH2F; (CH2)n-CFs where n=0-3;
      • with the proviso that at least one α-fluoroalkylated amino acid is present in the formula (I) compound.
      • where each NX-R-CO amino acid can have an absolute type R or type S configuration; their individual enantiomers, diastereoisomers, the related mixtures, the pharmaceutically acceptable salts are selective inhibitors of the avβ3 and/or avβ5 integrin receptors.
  • Therefore, objects of the present invention are compounds of formula (I), as described above, a process for their preparation, their use as medicaments and pharmaceutical compositions containing them.
  • Accordingly, we have found that this object is achieved by compounds of the formula I
      • B-G-L I
      • where B, G and L have the following meanings:
      • L is a structural element of the formula IL
      • —U-T IL
      • where
      • T is a group COOH, a radical hydrolyzable to COOH or a radical bioisosteric to COOH and
      • —U— is —(XL)a—(CRL 1RL 2)b—, —CRL 1═CRL 2—, ethynylene or ═CRL 1—,
        where
      • a is 0 or 1,
      • b is 0, 1 or 2,
      • XL is CRL 3RL 4, NRL 5, oxygen or sulfur,
      • RL 1, RL 2, RL 3, RL 4
      • independently of one another are hydrogen, —T, —OH, —NRL 6RL 7, —CO—NH2, a halogen radical, a branched or unbranched, optionally substituted C1-C6-alkyl, C2-C6-alkenyl, C2-C6-alkynyl, C3-C7-cycloal-kyl, —CO—NH(C1-C6-alkyl), —CO—N(C1-C6-alkly)2 or C1-C4-alkoxy radical, an optionally substituted radical C1-C2-alkylene-T, C2-alkenylene-T or C2-alkynylene-T, an optionally substituted aryl or arylalkyl radical or in each case independently of one another are two radicals RL 1 RL 2 or RL 3 and RL 4 or optionally RL 1 and RL 3 together are an optionally substituted 3- to 7-membered saturated or unsaturated carbocycle or heterocycle, which can contain up to three identical or different heteroatoms O, N, S,
      • RL 5, RL 6, RL 7
      • independently of one another are hydrogen, a branched or unbranched, optionally substituted C1-6-alkyl, C3-C7-cycloalkyl, CO—O—C1-C6-alkyl, SO2—C1-C6-alkyl or CO—C1-C6-alkyl radical or an optionally substituted CO—O-alkylenearyl, SO2-aryl, CO-aryl, SO2-alkylenearyl or CO-alkylertearyl radical,
      • G is a structural element of the formula IG
  • Figure US20160296592A1-20161013-C00006
  • where
      • the structural element B is bonded via the ring nitrogen and the structural element L is bonded to the structural element G via WG,
      • YG is CO, CS, C═NRG 2 CRG 3RG-su-p.4,
      • RG 2 is hydrogen, a hydroxyl group, a branched or unbranehed, optionally substituted C1-C6-alkyl, C1-C4-alkoxy, C3-C7-cycloalkyl or —O—C3-C7-cycloalkyl radical or an optionally substituted aryl, —O-aryl, arylalkyl or —O-alkylenearyl radical,
      • RG 3, RG 4
      • independently of one another are hydrogen or a branched or unbranched, optionally substituted C1-C6-alkyl, C2-C6-alkyenyl, C2-C6-alkynyl or C1-C4-alkoxy radical or both radicals RG 3 and RG 4 together are a cyclic acetal —O—CH2—CH2—O— or —O—CH2—O— or both radicals RG 3 and RG 4 together are an optionally substituted C3-C7-cycloalkyl radical,
      • RG 5 and RG 6
      • independently of one another are hydrogen, a branched or unbranched, optionally substituted C1-C6-alkyl or C1-C4-alkoxy radical, an optionally substituted aryl or arylalkyl radical or both radicals RG 5 and RG 6 together are an optionally substituted, fused, unsaturated or aromatic 3- to 10-membered carbocycle or heterocycle, which can contain up to three different or identical heteroatoms O, N, S,
      • where in this fused, unsaturated or aromatic 3- to 10-membered carbocycle or heterocycle, as substituents independently of one another up to four substituents from the group consisting of
      • hydroxyl, halogen or a branched or unbranched, optionally halogen-substituted C1-C4-aloxy, C1-C4-thioalkyl or C1-C4-alkyl radical or an optionally halogen-substituted aryl, hetaryl or C3-C7-cycloalkyl radical or an optionally halogen-substituted radical —SO2—C1-C4-alkyl, SO2—C1-C4-alkyl, —SO—C1-C4-alkyl, —SO2—C1-C4- alkylenearyl, —SO—C1-C4-alkylenearyl, —SO2-aryl or —SO-aryl are selected,
      • WG is a structural element selected front the group of structural elements of the formulae IWG1 to IWG4, 2
  • Figure US20160296592A1-20161013-C00007
      • RG1 is hydrogen, halogen, a hydroxyl group or a branched or unbranched, optionally substituted C1-C6-alkyl or C1-C4-alkoxy radical,
      • RG 7, RG 8, RG 9 RG 10
      • independently of one another are hydrogen, a hydrogxl group, —CN, halogen, a branched or unbranched, optionally substituted C1-C6-alkyl, C2-C6-alkenyl, C2-C6-alkynyl, C1-C4-alkylene-C3-C7-cycloalkyl, C1-C4-alkylene-C3-C7-heterocycloalkyl or C1-C4-alkylene-C3-C7-heterocycloalkenyl radical, a branched or unbranched, optionally substituted radical C1-C4-alkylene-ORG 11, C1-C4-alkylene-CO—OR-G 11, C1-C4-alkylene-O—CO—RG 11, C1-C4-alkylene-CO-RG 11, C1-C4-alkylene-SO2—NRG 12RG 13, C1-C4-alkylene-CO—NRG-12RG 13, C1-C4-alkylene-O—CO—NRG 12R.su-b.G13, C1-C4-alkylene-NRG 12RG 13 or C1-C4-alkylene-SRG 11, C1-C4-alkylene-SO—R.-sub.G11, a radical —S-RG 11, —O-RG 11, —SO-RG 11, —SO2—RG 11, —CO—ORG 11, —O—CO-RG 11, —O—CO—NRG 12RG 13, —SO2—NRG 12G13, —CO—NRG 12RG 13, —NRG 12RG 13 or CO-RG 11, an optionally substituted C3-C7-cycloalkyl, aryl, hetaryl, arylalkyl or hetarylalkyl radical or in each case independently of one another two radicals RG 7 and RG 9 or RG 9 and RG 10 or RG 7 and RG 8 or RG 9 and RG 10 together are an optionally substituted, saturated or unsaturated, nonaromatic, 3 to 7-membered carbocycle or heterocycle which can contain up to 3 heteroatoms selected from the group O, N, S and up to two double bonds,
      • RG 11 is hydrogen, a branched or unbranched, optionally substituted C1-C8-alkyl, C2-C6-alkenyl, C2-C6-alkynyl, C1-C5-alkylene-C1-C4-alkoxy, mono- and bis-alkylaminoalkylene or acylaminoalkylen radical or an optionally substituted aryl, heterocycloalkyl, heterocycloalkenyl, hetaryl, C3-C7-cycloalkyl, C1-C4-alkylene-C3-C.s-ub.7-cycloalkyl, arylalkyl, C1-C4-alkyleneheterocycloalkyl, C1-C4-alkyleneheterocycloalkenyl or hetarylalkyl radical,
      • RG 12, RG 13
      • independently of one another are hydrogen, a branched or unbranched, optionally substituted C1-C8-alkyl, C2-C6-alkenyl, C2-C6-alkynyl, C1-C5-alkylen-e-C1-C4-alkoxy, mono- and bis-alkylaminoalkylene or acylaminoalkylene radical or an optionally substituted aryl, heterocycloalkyl, heterocycloalkenyl, hetaryl, C3-C7cycloalkyl-, C1-C4-alkylene-C3-C7-cycloalkyl, arylalkyl, C1-C4-alkyleneheterocycloalkyl, C1-C4-alkyleneheteroc-ycloalkenyl or hetaryalkyl radical, or a radical —SO2—RG 11, —CO—ORG 11, —CO—NRG 11RG 11* or —CO-RG 11 or both radicals RG 12 and RG 13 together form a 5-to 7-membered nitrogen-containing carbocycle and
      • RG 11 is a radical RG 11 which is independent of RG 11
      • B is a structural element containing at least one atom which, under physiological conditions, as a hydrogen acceptor can form hydrogen bridges, where at least one hydrogen acceptor atom has a distance of 4 to 15 atom bonds to structural element G along the shortest possible route along the structural element skeleton,
      • and the physiologically tolerable salts, prodrugs and the enantiomerically pure or diastereomerically pure and tautomeric forms.
  • In the structural element L, T is understood as meaning a group COOH, a radical hydrolyzable to COOH or a radical bioisosteric to COOH.
  • A radical hydrolyzable to COOH is understood as meaning a radical with changes into a group COOH after hydrolysis.
  • A group which may be mentioned by way of example as a radical hydrolyzable to COOH is
  • Figure US20160296592A1-20161013-C00008
      • in which RT 1 has the following meanings:
      • a) OM, where M can be a metal cation, such as an alkali metal cation, such as lithium, sodium, potassium, the equivalent of an alkaline earth metal cation, such as calcium, magnesium and barium, or an environmentally tolerable organic ammonium ion such as primary, secondary, tertiary or quaternary C1-C4-alkylammonium or ammonium ion, such as ONa, OK or OLi,
      • b) a branched or unbranched, optionally halogen-substituted C1-C8-alkoxy radical, such as methoxy, ethoxy, propoxy, 1-methylethoxy, butoxy, 1-methylpropoxy, 2-methylpropoxy 1,1-dimethylethoxy, in particular methoxy, ethoxy, 1-methylethoxy, pentoxy, hexoxy, heptoxy, octoxy, difluoromethoxy, trifluoromethoxy, chlorodifluoromethoxy, 1-fluoroethoxy, 2-fluoroethoxy, 2,2-difluoroethoxy, 1,1,2,2-tetrafluoroethoxy, 2,2,2-trifluoroethoxy, 2-chloro-1,1,2-trifluoroethoxy or pentafluoroethoxy,
      • c) a branched or unbranched, optionally halogen-substituted C1-C4-alkylthio radical such as methylthio, ethylthio, propylthio, 1 -methylethylthio, butylthio, 1-methylpropylthio, 2-methylpropylthio or 1,1-dimethylethylthio radical,
      • d) an optionally substituted —O-alkylenearyl radical, such as —O-benzyl,
      • e) RT 1 is further a radical —(O)m—N(R18)(R19), in which m is 0 or 1 and R18 and R19, which can be identical or different, have the following meanings:
      • hydrogen,
      • a branched or unbranched, optionally substituted
      • C1-C6-alkyl radical, such as methyl, ethyl, propyl, 1 -methylethyl, butyl, 1-methylpropyl, 2-methylpropyl, 1,1-dimethylethyl, pentyl, 1-methylbutyl, 2-methylbutyl, 1,2-dimethylpropyl, 1,1-dimethylpropyl, 2,2-dimethylpropyl, 1-ethylpropyl, hexyl, 1-methylpentyl, 1,2-dimethylbutyl, 1,3-dimethylbutyl, 2,3-dimethylbutyl, 1,1-dimethylbutyl 2,2-dimethylbutyl, 3,3-dimethylbutyl, 1,1,2-trimethylpropyl, 1,2,2-trimethylpropyl, 1-ethylbutyl, 2-ethylbutyl or 1 -ethyl-2-methylpropyl or the corresponding substituted radicals, preferably methyl, ethyl, propyl, butyl or i-butyl,
      • C2-C6-alkenyl radical, such as vinyl, 2-propenyl, 2-butenyl, 3-butenyl, 1-methyl-2-propenyl, 2-methyl-2-propenyl, 2-pententyl, 3-pentenyl, 4-pentenyl, 1-methyl-2-butenyl, 2-methyl-2-butenyl, 3-methyl-2-butenyl, 1-methyl-3-butenyl, 2-methyl-3-butenyl, 3-methyl-3-butenyl, 1,1-dimethyl-2-propenyl, 1,2-dimethyl-2-propenyl, 1-ethyl-2-propenyl, 2-hexenyl, 3-hexenyl, 4-hexenyl, 5-hexenyl, 1-methyl-2-pentenyl, 2-methyl-2-pentenyl, 3-methyl-2-pentenyl, 4-methyl-2-pentenyl, 3-methyl-3-pentenyl, 4-methyl-3-pentenyl, 1-methyl-4-pentenyl, 2-methyl-4-pentenyl, 3-methyl-4-pentenyl, 4-methyl-4-pentenyl, 1,1-dimethyl-2 -butenyl, 1,1-dimethyl-3-butenyl, 1,2-dimethyl-2-butenyl, 1,2-dimethyl-3-butenyl, 1,3-dimethyl-2-butenyl, 1,3-dimethyl-3-butenyl, 2,2-dimethyl-3-butenyl, 2,3-dimethyl-2-butenyl, 2,3-dimethyl-3-butenyl, 1-ethyl-2-butenyl, 1-ethyl-3-butenyl, 2-ethyl-2-butenyl, 2-ethyl-3-butenyl, 1,1,2-trimethyl-2-propenyl, 1-ethyl-1-methyl-2-propenyl and 1-ethyl-2-methyl-2-propenyl, in particular 2-propenyl, 2-butenyl, 3-methyl-2-butenyl or 3-methyl-2-pentenyl or the corresponding substituted radicals,
      • C2-C6-alkynyl radical, such as ethynyl, 2-propynyl, 2-butynyl, 3-butynyl, 1-methyl-2propynyl, 2-pentynyl, 3-pentynyl, 4-pentynyl, 1-methyl-3-butynyl, 2-methyl-3-butynyl, 1-methyl-2-butynyl, 1,1-dimethyl-2-propynyl, 1-ethyl-2-propynyl, 2-hexynyl, 3-hexynyl, 4-hexynyl, 5-hexynyl, 1-methyl-2-pentynyl, 1-methyl-2-pentynyl, 1-methyl-3-pentynyl, 1-methyl-4-pentynyl, 2-methyl-3-pentynyl, 2-methyl-4-pentynyl, 3-methyl-4-pentynyl, 4-methyl-2-pentynyl, 1,1-dimethyl-2-butynyl, 1,1-dimethyl-3-butynyl, 1,2-dimethyl-3-butynyl, 2,2-dimethyl-3-butynyl, 1-ethyl-2-butynyl, 1-ethyl-3-butynyl, 2-ethyl-3--butynyl and 1-ethyl-1-methyl-2-propynyl, preferably 2-propynyl 2-butynyl, 1-methyl-2-propynyl or 1-methyl-2-butynyl or the corresponding substituted radicals,
      • C3-C8-cycloalkyl, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl, cyclooctyl or the corresponding substituted radicals,
      • or a phenyl radical, optionally mono- or polysubstituted, for example mono- to trisubstituted, by halogen, nitro, cyano, C1-C4-alkyl, C1-C4-haloalkyl, C1-C4-alkoxy, C1-C4-haloalkoxy or C1-C4-alkylthio such as 2-fluorophenyl, 3-chlorophenyl, 4-bromophenyl, 2-methylphenyl, 3-nitrophenyl, 4-cyanophenyl, 2-trifluoromethylphenyl, 3-methoxyphenyl, 4-trifluoroethoxyphenyl, 2-methylthiphenyl, 2,4-dichlorophenyl, 2-methoxy-3-methylphenyl, 2,4-dimethoxyphenyl, 2-nitro-5-cyanophenyl, 2,6-difluorophenyl,
      • or R18 and R19 together form an optionally substituted, e.g. C1-C4-alkyl-substituted, C4-C7-alkylene chain closed to give a cycle which can contain a heteroatom selected from the group consisting of oxygen, sulfur and nitrogen, such as —(CH2)4—, —(CH2)5—, —(CH2)6—, —(CH2)7—, —(CH2)2—O—(CH2)2—, —CH2—S—(CH2)3—, —(CH2)2—O—(CH2)3—, —NH—(CH2)3—, —CH2—NH—(CH2)2—, —CH2—CH.═CH—CH2—, —CH═CH—(CH2)3—, —CO—(CH2)2—CO— or —CO—(CH2)3—CO—.
  • A radical biolsosteric to COOH is understood as meaning radicals which can replace the function of a group COOH in active compounds by equivalent bond donor/acceptor capabilities or by equivalent charge distribution.
  • Radicals which may be mentioned by way of example as radicals bioisosteric to —COOH are those such as described in “The Practice of Medicinal Chemistry, Editor: C. G. Wermuth, Academic Press 1996, pages 125 and 216, in particular the radicals —P═O(OH)2, —SO3H, tetrazole or acylsulfonamides.
  • Preferred radicals T are —COOH, —CO—O—C1-C8-alkyl or —CO—O-benzyl.
  • The radical —U— in the structural element L is a spacer selected from the group —(XL)a—(CRL 1RL 2)b—, —CRL 1═CRL 2—, ethynylene or 40═CRL 1—. In the case of the radical ═CRL 1—, the structural element L is linked to the structural element G via a double bond.
  • XL is a radical CRL 3RL 4, NRL 5, oxygen or sulfur.
  • Preferred radicals —U— are the radicals —CRL 1═CR.s-ub.L2—, ethynylene or (XL)a—(CRL 1RL 2)-b—, where XL is preferably CRL 3RL 4 (a=0 or 1) or oxygen (a=1).
  • Particularly preferred radicals —U— are the radicals —(XL)a—(CRL 1RL 2)b—, where XL is preferably CRL 3RL 4 (a=0 or 1) or oxygen (a=1).
  • Under RL 1, RL 2, RL 3 or RL 4 in structural element L, a halogen radical is understood as meaning, for example, F, Cl, Br or I, preferably F.
  • Under RL 1, RL 2, RL 3 or RL 4 in structural element L, a branched or unbranched C1-C6-alkyl radical is understood as meaning, for example, methyl, ethyl, propyl, 1-methylethyl, butyl, 1-methylpropyl, 2-methylpropyl, 1,1-dimethylethyl, pentyl, 1-methylbutyl, 2-methylbutyl, 1,2-dimethylpropyl, 1,1-dimethylpropyl, 2,2-dimethylpropyl, 1-ethylpropyl, hexyl, 1-methylpentyl, 1,2-dimethylbutyl, 1,3-dimethylbutyl, 2,3-dimethylbutyl, 1,1-dimethylbutyl, 2,2-dimethylbutyl, 3,3-dimethylbutyl, 1,1,2-trimethylpropyl, 1,2,2-trimethylpropyl, 1-ethylbutyl, 2-ethylbutyl or 1-ethyl-2-methylpropyl, preferably branched or unbranched C1-C4-alkyl radicals such as methyl, ethyl, propyl, 1-methylethyl, butyl, 1-methylpropyl, 2-methylpropyl or 1,1-dimethylethyl, particularly preferably methyl.
  • Under RL 1, RL 2, RL 3 or RL 4 in structural element L, a branched or unbranhced C2-C6-alkenyl radical is understood as meaning, for example, vinyl, 2-propenyl, 2-butenyl, 3-butenyl, 1-methyl-2-propenyl, 2-methyl-2-propenyl 2-pentenyl, 3-pentenyl, 4-pentenyl, 1-methyl-2-butenyl, 2-methyl-2-butenyl, 3-methyl-2-butenyl, 1-methyl-3-butenyl, 2-methyl-3-butenyl, 3-methyl-3-butenyl, 1,1-dimethyl-2-propenyl, 1,2-dimethyl-2-propenyl, 1-ethyl-2-propenyl, 2-hexenyl, 3-hexenyl, 4-hexenyl, 5-hexenyl, 1-methyl-2-pentenyl, 2-methyl-2-pentenyl, 3-methyl-2-pentenyl, 4-methyl-2-pentenyl, 3-methyl-3-pentenyl, 4-methyl-3-pentenyl, 1-methyl-4-pentenyl, 2-methyl-4-pentenyl, 3-methyl-4-pentenyl, 4-methyl-4-pentenyl, 1,1 -dimethyl-2-butenyl, 1,1-dimethyl-3-butenyl, 1,2-dimethyl-2-butenyl, 1,2-dimethyl-3-butenyl, 1,3-dimethyl-2-butenyl, 1,3-dimethyl-3-butenyl, 2,2-dimethyl-3-butenyl, 2,3-dimethyl-2-butenyl, 2,3-dimethyl-3-butenyl, 1-ethyl-2-butenyl, 1-ethyl-3-butenyl, 2-ethyl-2-butenyl, 2-ethyl-3-butenyl, 1,1,2-trimethyl-2-propenyl, 1-ethyl-1-methyl-2-propenyl and 1-ethyl-2-methyl-2-propenyl, in particular 2-propenyl, 2-butenyl, 3-methyl-2-butenyl or 3-methyl-2-pentenyl.
  • Under RL 1, RL 2, RL 3 or RL 4 in structural element L, a branched or unbranched C2-C6-alkynyl radical is understood as meaning, for example, ethynyl, 2-propynyl, 2-butynyl, 3-butynyl, 1-methyl-2-propynyl, 2-pentynyl, 3-pentynyl, 4-pentynyl, 1-methyl-3-butynyl, 2-methyl-3-butynyl, 1-methyl-2-butynyl, 1,1-dimethyl-2-propynyl, 1-ethyl-2-propynyl, 2-hexynyl, 3-hexynyl, 4-hexynyl, 5-hexynyl, 1-methyl-2-pentynyl, 1-methyl-2-pentynyl, 1-methyl-3-pentynyl, 1-methyl-4-pentynyl, 2-methyl-3-pentynyl, 2-methyl-4-pentynyl, 3-methyl-4-pentynyl, 4-methyl-2-pentynyl, 1,1-dimethyl-2-butynyl, 1,1-dimethyl-3-butynyl, 1,2-dimethyl-3-butynyl, 2,2-dimethyl-3-butynyl, 1-ethyl-2-butynyl, 1-ethyl-3-butynyl, 2-ethyl-3-butynyl and 1-ethyl-1-methyl-2-propryl, preferably ethynyl, 2-propynyl, 2-butynyl, 1-methyl-2-propynyl or 1-methyl-2-butynyl.
  • Under RL 1, RL 2, RL 3 or RL 4 in structural element L, a branched or unbranched C3-C7cycloal-kyl radical is understood as meaning, for example, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl or cycloheptyl.
  • Under RL 1, RL 2, RL 3 or RL 4 in structural element L, a branched or unbranched C1-C4-alkoxy radical is understood as meaning, for example, methoxy, ethoxy, propoxy, 1-methylethoxy, butoxy, 1-methylpropoxy, 2-methylpropoxy or 1,1-dimethylethoxy.
  • The radicals —CO—NH(C1-C6-alkyl), —CO—N(C1-C6-alkyl)2 are secondary or tertiary amides and are composed of the amide bond and the corresponding C1-C6-alkyl radicals such as described above for RL 1, RL 2, RL 3 or RL 4.
  • The radicals RL 1, RL 2, RL 3 or RL 4 can furthermore be a radical
  • C1-C2alkylene-T, such as methylene-T or ethylene-T, C2-alkenylene-T, such as ethenylene-T or C2-alkynylene-T, such as ethynylene-T,
      • an aryl radical, such as phenyl, 1-naphthyl or 2-naphthyl or
      • an arylalkyl radical, such as benzyl or ethylenephenyl (homobenzyl),
      • where the radicals can optionally be substituted.
  • Furthermore, two radicals RL 1 and RL 2 or RL 3 and RL 4 or optionally RL 1 and RL 3 can in each case independently of one another together be an optionally substituted 3- to 7-membered saturated or unsaturated carbocycle or heterocycle, which can contain up to three different or identical heteroatoms O, N, S.
  • All radicals for RL 1, RL 2, RL 3 or RL 4 can be optionally substituted. For the radicals RL 1, RL 2, RL 3 or RL 4 and all further substituted radicals of the description below, suitable substituents, if the substituents are not specified in greater detail, are independently of one another up to 5 substituents, for example selected from the following group:
      • —NO2, —NH2, —OH, —CN, —COOH, —O—CH2—COOH, halogen, a branched or unbranched, optionally substituted C1-C4-alkyl radical, such as methyl, CF3, C2F5 or CH2F, —CO—O—C1-C4-alkyl, C3-C6-cycloalkyl, C1-C4-alkoxy, C1-C4-thioalkyl, —NH—CO—O—C1-C4-alkyl, —O—CH2—COO—C1-C4-a-lkyl, —NH—CO—C1-C4-alkyl, —CO—NH—C1-C4-alkyl, —NH—SO2—C1-C4-alkyl, —SO2—NH—C1-C4-al-kyl, —N(C1-C4-alkyl)2, 20-NH—C1-C4-alkyl, or —SO2—C1-C4-alkyl radical, such as —SO2—CF3, an optionally substituted —NH—CO-aryl, —CO—NH-aryl, —NH—CO—O-aryl, —NH—CO—O-alkylenearyl, —NH—SO2-aryl, —SO2—NH-aryl, —CO—NH-benzyl, —NH—SO2-benzyl or —SO2—NH-benzyl radical, an optionally substituted radical —SO2—NR5 2R5 3 or —CO—NR5 2RS3 where the radicals R2 and R3 independently of one another can have the meaning RL 5 as below or both radicals R2 and R3 together can be a 3- to 6-membered, optionally substituted, saturated, unsaturated or aromatic heterocycle which, in addition to the ring nitrogen, can contain up to three further different or identical heteroatorns O, N, S, and optionally two radicals substituted on this heterocycle can together be a fused, saturated, unsaturated or aromatic carbocycle or heterocycle which can contain up to three different or identical heteroatoms O, N, S and the cycle can be optionally substituted or a further, optionally substituted cycle can be fused to this cycle.
  • If not specified in greater detail, in all terminally bonded, substituted hetaryl radicals of the description, two substituents can form a fused 5- to 7-membered, unsaturated or aromatic carbocycle.
  • Preferred radicals RL 1, RL 2, RL 3 or RL 4 are independently of one another hydrogen, halogen, a branched or unbranched, optionally substituted C1-C4-alkyl, C1-C4-alkoxy or C3-C7-cycloalkyl radical or the radical —NRL 6RL 7.
  • Particularly preferred radicals RL 1, RL 2, RL 3 or RL 4 are independently of one another hydrogen, fluorine or a branched or unbranched, optionally substituted C1-C4-alkyl radical, preferably methyl.
  • The radicals RL 5, RL 6, RL 7 in structural element L are independently of one another hydrogen, a branched or unbranched, optionally substituted
      • C1-C6-alkyl radical, for example as described above for RL 1,
      • C3-C7-cycloalkyl radical, for example as described above for RL 1,
      • CO—O—C1-C6-alkyl, SO2—C1-C6-alkyl or CO—C1-C6-alkyl radical, which is composed of the group CO—O, SO2 or CO and, for example, of the C1-C6-alkyl radicals described above for RL 1,
      • or an optionally substituted CO—O—-alkylenearyl, SO2-aryl, SO2-alkylenearyl or CO-alkylenearyl radical, which is composed of the group CO—O, SO2 or CO and, for example, of the aryl or arylalkyl radicals described above for RL 1.
  • Preferred radicals for RL 6 in structural element L are hydrogen, a branched or unbranched, optionally substituted C1-C4-alkyl, CO—O—C1-C4-alkyl, CO—C1-C4-alkyl or SO2—C1-C4-alkyl radical or an optionally substituted Co-O-benzyl, SO2-aryl, SO2-alkylenearyl or CO-aryl radical.
  • Preferred radicals for RL 7 in structural element L are hydrogen or a branched or unbranched, optionally substituted C1-C4-alkyl radical.
  • Preferred structural elements L are composed of the preferred radicals of the structural element.
  • Particularly preferred structural elements L are composed of the particularly preferred radicals of the structural element.
  • G is a structural element of the formula IG
  • Figure US20160296592A1-20161013-C00009
      • where the incorporation of the structural element G can take place in both orientations. Preferably, the incorporation of the structural element G into the compounds of the formula I can take place such that the structural element B is bonded via the ring nitrogen and the structural element L is bonded via WG to the structural element G, optionally via a double bond.
  • YG in structural element G is CO, CS, C═NRG 2 or CRG 3RG 4, preferably CO, C═NRG 2 or CRG 3RG 4, particularly preferably CO or CRG 3RG 4.
  • RG 2 in structural element G is hydrogen, a hydroxyl group, a branched or unbranched, optionally substituted C1-C6-alkyl, C1-C4-alkoxy or C3-C7-cycloalk- yl radical, for example as described above for RL 1 in each case,
      • an optionally substituted —O—C3-C7-cycloalkyl radical, which is composed of an ether group and, for example, of the C3-C7-cycloalkyl radical described above for RL 1,
      • an optionally substituted aryl or arylalkyl radical, for example as described above for RL 1 in each ease or
      • an optionally substituted —O—aryl or —O-alkylenearyl radical, which is composed of a group —O— and, for example, of the aryl or arylalkyl radicals described above for RL 1.
  • Preferred radicals RG 2 in structural element G are hydrogen, hydroxyl or a branched or unhranched, optionally substituted C1-C6-alkyl radical, in particular methyl or C1-C4-alkoxy radical, in particular methoxy.
  • Branched or unbranched, optionally substituted C1-C6-alkyl, C2-C6-alkynyl or C1-C4-alkoxy radicals for RG 3 or RG 4 in structural element G independently of one another are understood as meaning, for example, the corresponding radicals in each case described above for RL 1.
  • Further, both radicals RG 3 and RG 4 can together form a cyclic acetal, such as —O—CH2—CH2—O— or —O—CH2—O—.
  • Furthermore, both radicals RG 3 and RG 4 can together form an optionally substituted C3-C7-cycloalkyl radical.
  • Preferred radicals for RG 3 or RG 4 are independently of one another hydrogen, C1-C6-alkyl or C1-C4-alkoxy, and both radicals RG 3 and RG 4 together form a cyclic acetal, such as —O—CH2—CH2—O— or —O—CH2—O—. Particularly preferred radicals RG 3 or RG 4 are independently of one another hydrogen and both radicals RG 3 and RG 4 together form a cyclic acetal, in particular —O—CH2—CH2—O— or —O—CH2—O—.
  • Branched or unbranched, optionally substituted C1-C6-alkyl or C1-C4-alkoxy radicals and optionally substituted aryl or arylalkyl radicals for RG 5 and RG 6 in structural element G independently of one another are, for example, the corresponding radicals in each case described above for RL 1.
  • Further, both radicals RG 5 and RG 6 can together form an optionally substituted, fused, unsaturated or aromatic 3- to 10-membered carbocycle or heterocycle, which can contain up to three different or identical heteroatoms O, N, S, where in this fused, unsaturated or aromatic 3- to 10-membered carbocycle or heterocycle, as substituents, independently of one another up to four substituents from the group
      • hydroxyl, halogen, such as F or Cl or a branched or unbranched, optionally halogen-substituted C1-C4-alkoxy radical, such as methoxy, C1-C4-thioalkyl or C1-C4-alkyl radical, such as methyl, ethyl, propyl or butyl, or an optionally halogen-substituted aryl radical, such as phenyl, hetaryl, such as described below for RG7, or C3-C7-cycloalkyl radical, such as described below for RG 7, or an optionally halogen-substituted radical —SO2—C1-C4-alkyl, —SO—C1-C4-alkyl, —SO2—C1-C4-alkylenearyl, —SO—C1-C4-alkylenea- ryl, —SO2-aryl or —SO-aryl are selected.
  • Preferred substituents are halogen, a C1-C4-alkyl radical, C1-C4-alkoxy radical or aryl radical.
  • Particularly preferred substituents are a C1-C4-alkyl radical, in particular methyl or ethyl, a C1-C4-alkoxy radical, in particular methoxy, or F or Cl.
  • Preferred radicals for RG 5 and RG 6 are independently of one another hydrogen, an optionally substituted C1-C6-alkyl radical, in particular methyl and ethyl, an optionally substituted aryl radical, in particular optionally substituted phenyl or an optionally substituted arylalkyl radical, in particular an optionally substituted benzyl radical, and in each case both radicals RG 5 and RG 6 together can be an optionally substituted, fused, unsaturated or aromatic 3- to 10-membered carbocycle or heterocycle which can contain up to three different or identical heteroatoms O, N, S.
  • In particularly preferred radicals for RG 5 and RG 6, both radicals RG 5 and RG 6 together form an optionally substituted, fused, unsaturated or aromatic 3- to 6-membered carbocycle or heterocycle, for example selected from one of the following doubly bonded structural formulae:
  • Figure US20160296592A1-20161013-C00010
      • in particular selected from one of the following, doubly bonded structural formulae:
  • Figure US20160296592A1-20161013-C00011
  • WG is a structural element selected from the group of structural elements of the formulae IWG 1 to IWG 4, where the dashed lines intersect the atomic bonds within the structural element G and the carbon atom substituted by RG 7 and RG 8 is bonded to YG.
  • Figure US20160296592A1-20161013-C00012
  • In a preferred embodiment, WG is a structural element selected from the group of structural elements of the formulae IWG 1, IWG 2 and IWG 4, in particular the structural element of the formula IWG2.
  • RG 1 in structural element WG is hydrogen, halogen, such as Cl, F, Br or I, a hydroxyl group or a branched or unbranched, optionally substituted C1-C6-alkyl radical, preferably C1-C4-alkyl or C1-C4-alkoxy radical such as in each case described above for RL 1.
  • Preferred radicals for RG 1 are hydrogen, hydroxyl and optionally substituted C1-C4-alkyl or C1-C4-alkoxy radicals.
  • Particularly preferred radicals for RG 1 are hydrogen and carboxyl-substituted C1-C4-alkyl or C1-C4-alkoxy radicals, in particular the radicals —CH2COOH or —O—CH2COOH.
  • RG 7, RG 8, RG 9 and RG 10 in structural element G are independently of one another hydrogen, a hydroxyl group, CN, halogen, such as F, Cl, Br, I, a branched or unbranched, optionally substituted
      • C1-C6-alkyl radical, such as optionally substituted methyl, ethyl, propyl, 1-methylethyl, butyl, 1-methylpropyl, 2-methylpropyl, 1,1-dimethylethyl, pentyl, 1-methylbutyl, 2-methylbutyl, 1,2-dimethylpropyl, 1,1-dimethylpropyl, 2,2-dimethylpropyl, 1-ethylpropyl, hexyl, 1-methylpentyl, 1,2-dimethylbutyl, 1,3-dimethylbutyl, 2,3-dimethylbutyl, 1,1-dimethylbutyl, 2,2-dimethylbutyl, 3,3-dimethylbutyl, 1,1,2-trimethylpropyl, 1,2,2-trimethylpropyl, 1-ethylbutyl, 2-ethylbutyl or 1-ethyl-2-methylpropyl,
      • C2-C6-alkenyl radical, such as optionally substituted vinyl, 2-propenyl, 2-butenyl, 3-butenyl, 1-methyl-2-propenyl, 2-methyl-2-propenyl, 2-pentenyl, 3-pentenyl, 4-pentenyl, 1-methyl-2-butenyl, 2-methyl-2-butenyl, 3-methyl-2-butenyl, 1-methyl-3-butenyl, 2-methyl-3-butenyl, 3-methyl-3-butenyl, 1,1-dimethyl-2-propenyl, 1,2-dimethyl-2-propenyl, 1-ethyl-2-propenyl, 2-hexenyl, 3-hexenyl, 4-hexenyl, 5-hexenyl, 1-methyl-2-pentenyl, 2-methyl-2-pentenyl, 3-methyl-2-pentenyl, 4-methyl-2-pentenyl, 3-methyl-3-pentenyl, 4-methyl-3-pentenyl, 1-methyl-4-pentenyl, 2-methyl-4-pentenyl, 3-methyl-4-pentenyl, 4-methyl-4-pentenyl, 1,1-dimethyl-2-butenyl, 1,1-dimethyl-3-butenyl, 1,2-dimethyl-2-butenyl, 1,2-dimethyl-3-butenyl, 1,3-dimethyl-2-butenyl, 1,3-dimethyl-3-butenyl, 2,2-dimethyl-3-butenyl, 2,3-dimethyl-2-butenyl, 2,3-dimethyl-3-butenyl, 1-ethyl-2-butenyl, 1-ethyl-3-butenyl, 2-ethyl-2-butenyl, 2-ethyl-3-butenyl, 1,1,2-trimethyl-2-propenyl, 1-ethyl-1-methyl-2-propeny-1 or 1-ethyl-2-methyl-2-propenyl,
      • C2-C6-alkynyl radical, such as optionally substituted ethynyl, 2-propynyl, 2-butynyl, 3-butynyl, 1-methyl-2-propynyl, 2-pentynyl, 3-pentynyl, 4-pentynyl, 1-methyl-3-butynyl, 2-methyl-3-butynyl, 1-methyl-2-butynyl, 1,1-dimethyl-2-propynyl, 1-ethyl-2-propynyl, 2-hexynyl, 3-hexynyl, 4-hexynyl, 5-hexynyl, 1-methyl-2-pentynyl, 1-methyl-3-pentynyl, 1-methyl-4-pentynyl, 2-methyl-3-pentynyl, 2-methyl-4-pentynyl, 3-methyl-4-pentynyl, 4-methyl-2-pentynyl, 1,1-dimethyl-2-butynyl, 1,1-dimethyl-2-butynyl, 1,1-dimethyl-3-butynyl, 1,2-dimethyl-3-butynyl, 2,2-dimethyl-3-butynyl, 1-ethyl-2-butynyl, 1-ethyl-3-butynyl, 2-ethyl-3-butynyl or 1-ethyl-1-methyl-2-propynyl,
        an optionally substituted
      • C3-C7-cycloalkyl radical, such as optionally substituted cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl or cycloheptyl,
      • C3-C7-heterocycloalkyl radical, such as optionally substituted aziridinyl, diaziridinyl, oxiranyl, oxaziridinyl, oxetanyl, thiiranyl, thietanyl, pyrrolidinyl, piperazinyl, morpholinyl, piperidinyl, tetrahydrofuranyl, tetrahydropyranyl, 1,4-dioxanyl, hexahydroazepinyl, oxepanyl, 1,2-oxathiolanyl or oxazolidinyl,
      • C3-C7-heterocycloalkenyl radical, such as optionally substituted azirinyl, diazirinyl, thiirenyl, thietyl, pyrrolinyls, oxazolinyls, azepinyl, oxepinyl, α-pyranyl, β-pyranyl, γ-pyranyl, dihydropyranyls, 2,5-dihydropyrrolinyl or 4,5-dihydrooxazolyl, a branched or unbranched, optionally substituted C1-C4-alkylene-C3-C7-cycloalkyl radical, which is composed, for example, of branched or unbranched C1-C4-alkylene radicals such as methylene, ethylene, propylene, n-butylene, isobutylene or t-butylene and, for example, the above mentioned C3-C7-cycloalkyl radicals,
      • a branched or unbranched optionally substituted C1-C4-alkylene-C3-C7-heterocycloalkyl or C1-C4-alkylene-C3-C7-heterocycloalkenyl radical, which is composed of optionally substituted C1-C4-alkylene radicals, such as methylene, ethylene, propylene, n-butylene, isobutylene or t-butylene and, for example, the above-mentioned C3-C7-heterocycloalkyl or C3-C7-heterocycloalkenyl radicals, the radicals being preferred which in the cyclic moiety contain one or two heteroatoms selected from the group consisting of N, O and S and up to two double bonds,
      • a branched or unbranched, optionally substituted radical C1-C4-alkylene-O-RG 11, C1-C4-alkylene-CO-OR-G11, C1-C4-alkylene-O—CO-RG 11, C1-C4-alkylene-CO-RG 11, C1-C4-alkylene-SO2—NRG 12RG 13, C1-C4-alkylene-CO—NRG 12, RG 3, C1-C4-alkylene-O—CO—NRG 12RG 13, C1-C4-alkylene-NRG 12RG 13, C1-C4-alkylene-SRG 11 or C1-C4-alkylene-SO—RG 11 which is composed of branched or unbranched, optionally substituted C1-C4-alkylene radicals, such as methylene, ethylene, propylene, n-butylene, isobutylene or t-butylene, the corresponding groups —O—, —CO—, —S—, —N and the terminal radicals RG 11, RG 12 and RG 13 described below,
        an optionally substituted
      • aryl radical, preferably optionally substituted phenyl, 1-naphthyl or 2-naphthyl,
      • arylalkyl radical, preferably optionally substituted benzyl or ethylenephenyl (homobenzyl),
      • hetaryl radical, preferably optionally substituted 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-furyl, 3-furyl, 2-pyrrolyl, 3-pyrrolyl, 2-thienyl, 3-thienyl, 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, 2-oxazolyl, 4-oxazolyl, 5-oxazolyl, 2-pyrimidyl, 4-pyrimidyl, 5-pyrimidyl, 6-pyrimidyl, 3-pyrazolyl, 4-pyrazolyl, 5-pyrazolyl, 3-isothiazolyl, 4-isothiazolyl, 5-isothiazolyl, 2-imidazolyl, 4-imidazolyl, 5-imidazolyl, 3-pyridazinyl, 4-pyridazinyl, 5-pyridazinyl, 6-pyridazinyl, 3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, thiadiazolyl, oxadiazolyl or triazinyl or their fused derivatives such as indazolyl, indolyl, benzothiophenyl, benzofuranyl, indolinyl, benzimidazolyl, benzothiazolyl, benzoxazolyl, quinolinyl or isoquinolinyl,
      • hetarylalkyl radical, preferably optionally substituted —CH2-2-pyridyl, —CH2-3-pyridyl, —CH2-4-pyridyl, —CH2-2-thienyl, —CH2-3-thienyl, —CH2-2-thiazolyl, —CH2-4-thiazolyl, —CH2-5-thiazolyl, —CH2—CH2-2-py- ridyl, —CH2—CH2-3-pyridyl, —CH2—CH2-4-pyridyl, —CH2—CH2-2-thienyl, —CH2—CH2-3-thienyl, 10-CH2—CH2-2-thiazolyl, —CH2—CH2-4-thiazolyl or —CH2—CH2-5-thiazolyl or
      • a radical —S—R11, —O—RG 11, —SO-RG 11, —SO2—RG 11, —CO—ORG 11, —O—CO-RG 11, —O—CO—NRG 12RG 13, —SO2—NRG 12RG 13, —CO—NRG 12RG 13, —NRG 12RG 13, CO—RG 11.
  • Further, two radicals RG 7 and RG 9 or RG 8 and RG 10 or RG 7 and RG 8 or RG 9 and RG 1O can in each case independently of one another together form an optionally substituted, saturated or unsaturated, nonaromatic, 3- to 7-membered carbocycle or heterocycle which can contain up to 3 heteroatoms selected from the group consisting of O, N, S and up to two double bonds.
  • Preferred radicals for RG 7, RG 8, RG 9 and RG 10 in the structural element G are independently of one another hydrogen, halogen, in particular a branched or unbranched, optionally substituted C1-C6-alkyl, or C2-C6-alkynyl radical, a branched or unbranched, optionally substituted radical C1-C4-alkylene-ORG 11, C1-C4-alkylene-CO—OR-G 11, C1-C4-alkylene-O—CO-RG 11, C1-C4-alkylene-CO—NRG 12RG 13, C1-C4-alkylene-O—CO—NRG 12RG 13, a radical —O-RG 11, —CO—ORG 11, —O—CO—RG 11, —O—CO—NRG 12RG 13, —CO—NRG 12RG-13, —NRG 12RG 13 or CO—RG 11, an optionally substituted aryl, hetaryl or arylalkyl radical, as described above in each case.
  • Particularly preferred radicals for RG 7 RG 8, RG 9 and RG 10 in the structural element G are independently of one another hydrogen, F, a radical —CO—ORG 11, —CO—NRG 12RG 13, or an optionally substituted aryl radical, as described above in each case.
  • A branched or unbranched, optionally substituted C1-C8-alkyl radical for RG 11, RG 12 and RG 13 is understood as meaning independently of one another, for example, the C1-C6-alkyl radicals mentioned above for RG 1, plus the radicals heptyl and octyl.
  • Preferred substituents of the branched or unbranched, optionally substituted C1-C8-alkyl radicals for RG 11, RG 12 and RG 13 independently of one another are the radicals halogen, hydroxyl, C1-C4-alkoxy, —CN, —COOH and —CO—O—C1-C4-alkyl.
  • A branched or unbranched, optionally substituted C2-C6-alkenyl, C2-C6-alkynyl or C1-C4-alkylene-C3-C7-cycloalkyl radical, an optionally substituted C3-C7-cycloalkyl, aryl, arylalkyl, hetaryl or hetarylalkyl radical for RG 11, RG 12 and RG 13 independently of one another is understood as meaning, for example, the corresponding radicals mentioned above for RG 1.
  • Preferred branched or unbranched, optionally substituted —C1-C5-alkylene-C1-C4-alkoxy radicals for RG 11, RG 12 and RG 13 are independently of one another methoxymethylene, ethoxymethylene, t-butoxymethylene, methoxyethylene or ethoxyethylene.
  • Preferred branched or unbranched, optionally substituted mono- and bisalkylaminoalkylene or acylaminoalkylene radicals for RG 11, RG 12 and RG 13 are independently of one another branched or unbranched, optionally substituted radicals —C1-C4-alkylene-NH(C1-C4-alkyl), —C1-C4-alkylene-N(C1-C4-alkyl)2or —C1-C4-alkylene-NH—CO—C1-C4-alkyl.
  • Preferred optionally substituted heterocycloalkyl, heterocycloalkenyl, C1-C4-alkyleneheterocycloalkyl or C1-C4-alkyleneheterocycloalkenyl radicals for RG 11, RG 12 and RG 13 are independently of one another the C3-C7-heterocycloalkyl, C3-C7-heterocycloalkenyl, C1-C4-alkylene-C3-C7-heterocycloalkyl or C1-C4-alkylene-C3-C7-heterocycloalkenyl radicals described above for RG 1.
  • Particularly preferred, optionally substituted heterocycloalkyl, heterocycloalkenyl, C1-C4-alkyleneheterocycloalkyl or C1-C4-alkyleneheterocycloalkenyl radicals for RG 11, RG 12 and RG 13 are independently of one another the C3-C7-heterocycloalkyl, C3-C7-heterocycloalkenyl, C1-C4--alkylene-C3-C7-heterocycloalkyl or C1-C4-alkylene-C3-C7-heterocycloalkenyl radicals described above for RG 1, one or two heteroatoms being selected from the group consisting of N, O and S and up to two double bonds being contained in the cyclic moiety.
  • Furher, RG 12 and RG 13 can independently of one another be a radical —SO2—RG 11, —CO—O-RG 11, —CO—NRG 11RG 11* or —CO-RG 11, RG 11* being a radical RG 11 which is independent of RG 11.
  • Furthermore, both radicals RG 12 and RG 13 can together form a 5- to 7-membered, preferably saturated nitrogen-containing carbocycle, in the sense of a cyclic amine structure, such as N-pyrrolidinyl, N-piperidinyl, N-hexahydroazepinyl, N-morpholinyl or N-piperazinyl, where in the case of heterocycles which carry free amine protons, such as N-piperazinyl, the free amine protons can be replaced by customary amine protective groups, such as methyl, benzyl, Boc (tert-butoxycarbonyl), Z (benzyloxycarbonyl), tosyl, —SO2—C1-C4-alkyl, —SO2-phenyl or —SO2-benzyl.
  • Particularly preferred radicals for RG 11 are hydrogen or an optionally substituted C1-C4-alkyl or aryl radical.
  • Particularly preferred for RG 12 and RG 13 are independently of one another hydrogen or an optionally substituted C1-C4-alkyl radical.
  • Preffered structural elements G are composed of at least one preferred radical of the strucural element G, while the remaining radicals are widely variable.
  • Particularly preferred structural elements G are composed of the preferred radicals of the structural element G.
  • Very particularly preferred structural elements G are composed of the particularly preferred radicals of the structtnul element G.
  • Structural element B is undentood as meaning a structural element comprising at least one atom which under physiological conditions can form hydrogren bridges as a hydrogen acceptor, at least one hydrogen acceptor atom having a distance of 4 to 15 atom bonds to structural element G along the shortest possible route along the structural element skeleton. The arrangement of the structural skeleton of structural element B is widely variable.
  • Suitable atoms which under physiological conditions can form hydrogen bridges as hydrogen acceptors are, for example, atoms having Lewis base properties, such as the heteroatoms nitrogen, oxygen or sulfur.
  • Physiological conditions is understood as meaning a pH which prevails at the site in a body at which the ligands interact with the receptors. In the present case, the physiological conditions have a pH of, for example, 5 to 9.
  • In a preferred embodiment, structural element B is a structural element of the formula IB

  • A-E-tm IB
      • where A and E have the following meanings:
      • A is a structural element selected from the group:
      • a 4- to 8-membered monocyclic saturated, unsaturated or aromatic hydrocarbon which can contain up to 4 heteroatoms selected from the group O, N and S, where, in each case independently of one another, the ring nitrogen optionally contained or the carbons can be substituted, with the proviso that at least one heteroatom selected from the group O, N and S contained in the structural element A, or
      • a 9- to 14-membered polycyclic, saturated, unsaturated or aromatic hydrocarbon which can contain up to 6 heteroatoms selected from the group N, O and S, where, in each case independently of one another, the ring nitrogen optionally contained or the carbons can be substituted, with the proviso that at least one heteroatom selected from the group O, N and S is contained in the structural element A,
      • a radical
  • Figure US20160296592A1-20161013-C00013
  • where
      • Z A 1 is oxygen, sulfur or optionally substituted nitrogen and
      • Z A 2 is optionally substituted nitrogen, oxygen or sulfur,
      • or a radical
  • Figure US20160296592A1-20161013-C00014
  • where
      • RA18, RA 19
      • independently of one another are hydrogen, a branched or unbranched, optionally substituted C1-C8-alkyl, C2-C6-alkenyl, C2-C6-alkynyl, C1-C5-alkylen-e-C1-C4-alkoxy, mono- and bisalkylaminoalkylene or acylaminoalkylene radical or an optionally substituted aryl, heterocycloalkyl, heterocycloalkenyl, hetaryl, C3-C7-cycloalkyl-, C1-C4-alkylene-C3-C7-cycloalkyl, arylalkyl, C1-C4-alkyleneheterocycloalkyl, C1-C4-alkyleneheteroc- ycloalkenyl or hetarylalkyl radical, or a radical —SO2—RG 11, —CO—ORG 11, —CO-NR11RG 11* or —CO—RG 11 and
      • E is a spacer structural clement which covalentily bonds the structural element A to the structural element G, where the number of atom bonds along the shortest possible route along the structural element skeleton E is 3 to 14.
  • In a particularly preferred embodiment, the structural element A is a structural element selected from the group of structural elements of the formulae IA1 to IA 18,
  • Figure US20160296592A1-20161013-C00015
    Figure US20160296592A1-20161013-C00016
    Figure US20160296592A1-20161013-C00017
  • where
      • independently of one another are 1, 2 or 3,
      • independently of one another are hydrogen, CN, halogen, a branched or unbranched, optionally substituted C1-C1-6-alkyl or CO—C1-C6-alkyl radical or an optionally substituted aryl, arylalkyl, hetaryl, hetarylalkyl or C3-C7-cycloalkyl radical or a radical CO—O-RA 14, O-RA 14, S-RA 14, NRA 15RA 16, CO—NRA 15RA 16 or SO2NRA 15RA 16 or both radicals RA 1 and RA 2 together are a fused, optionally substituted, 5- or 6-membered, unsaturated or aromatic carbocycle or heterocycle which can contain up to three heteroatoms selected from the group O, N, and S,
      • RA 13, RA 13*
      • independently of one another are hydrogen, CN, halogen, a branched or unbranched, optionally substituted C1-C6-alkyl radical or an optionally substituted aryl, arylalkyl, hetaryl, C3-C7-cycloalk- yl radical or a radical CO—O-RA 14, O-RA 14, S-RA 14, NRA 15RA 16, SO2—NRA 1-5RA 16 or CO—NRA 15RA 16,
        where
      • RA 14 is hydrogen, a branched or unbranched, optionally substituted C1-C6-alkyl, alkylene-C1-C4-alkoxy, C2-C6-alkenyl, C2-C6-alkynyl or C1-C6-alkylene-C3-C7-cycloalkyl radical or an optionally substituted C3-C7-cycloalkyl, aryl, arylalkyl, hetaryl or hetarylalkyl radical,
      • RA 15, RA 16,
      • independently of one another are hydrogen, a branched or unbranched, optionally substituted C1-C6-alkyl, CO—C1-C6-alkyl, SO2—C1-C6-alkyl, COO—C1-C6-alkyl, CO—NH—C1-C6-alkyl, arylalkyl, COO-alkylenearyl, SO2-alkylenearyl, CO—NH-alkylenearyl, CO—NH-alkylenehetaryl or hetarylalkyl radical or an optionally substituted C3C7-cycloalkyl, aryl, CO-aryl, CO—NH-aryl, SO2-aryl, hetaryl, CO—NH-hetaryl or Co-hetaryl radical,
      • RA 3, RA 4
      • independently of one another are hydrogen, —(CH2)n—(XA)j—RA 12, or both radicals together are a 3- to 8-membered, saturated, unsaturated or aromatic N-heterocycle which can additionally contain two further, identical or different heteroatoms O, N or S, where the cycle is optionally substituted or a further, optionally substituted, saturated, unsaturated or aromatic cycle can be fused to this cycle,
        where
      • n is 0, 1, 2 or 3,
      • j is 0 or 1,
      • XA is —CO—, —CO—N(Rx 1)—, —N(Rx 1)-CO—, —N(Rx 1)—CO—N(Rx 1*)—, —N(Rx 1)—CO—O—, —O—, —S—, —SO2—, —SO2—N(Rx 1)—, —SO2—O—, —CO—O—, —O—CO—, —O—CO—N(Rx 1)—, —N(Rx 1)— or —N(Rx 1)—SO2—,
      • RA 12 is hydrogen, a branched or unbranched, optionally substituted C1-C6-alkyl radical, an optionally C1-C4-alkyl- or aryl-substituted C2-C6-alkynyl or C2-C6-alkenyl radical or a 3- to 6-membered, saturated or unsaturated heterocycle, substituted by up to three identical or different radicals, which can contain up to three different or identical heteroatoms O, N, S, a C3-C7-cycloalkyl, aryl or hetaryl radical, where two radicals together can be a fused, saturated, unsaturated or aromatic carbocycle or heterocycle which can contain up to three different or identical heteroatoms O, N, S and the cycle can optionally be substituted or a further, optionally substituted, saturated, unsaturated or aromatic cycle can be fused to this cycle, or the radical RA 12, together with RX1 or RX1* forms a saturated or unsaturated C3-C7-heterocycle which can optionally contain up to two further heteroatoms selected from the group O, S and N,
      • Rx 1, Rx 1*
      • independently of one another are hydrogen, a branched or unbranched, optionally substituted C1-C6-alkyl, C1-C6-alkoxyalkyl, C2-C6-alkenyl, C2-C12-alkynyl, CO—C1-C6-alkyl, CO—O—C1-C6-alkyl or SO2—C1-C6-alkyl radical or an optionally substituted C3-C7-cycloalkyl, aryl, arylalkyl, CO—O-alkylenearyl, CO-alkylenearyl, CO-aryl, SO2-aryl, hetaryl, CO-hetaryl or SO2-alkylenearyl radical,
      • RA 6, RA 6*
      • are hydrogen, a branched or unbranched, optionally substituted C1-C4-alkyl, —CO—O—C1-C4-alkyl, arylalkyl, —CO—O-alkylenearyl, —CO—O-allyl, —CO—C1-C4-alkyl, —CO-alkylenearyl, C3-C7-cycloalkyl or —CO-allyl radical or in structural element IA 7 radicals RA 6 and RA 6* together are an optionally substituted, saturated, unsaturated or aromatic heterocycle which, in addition to the ring nitrogen, can contain up to two further different or identical heteroatoms O, N, S,
      • RA 7 is hydrogen, —OH, —CN, —CONH2, a branched or unbranched, optionally substituted C1-C4-alkyl, C1-C4-alkoxy, C3-C7-cycloalkyl or —O—CO—C1-C4-alkyl radical, or an optionally substituted arylalkyl, —O-alkylenearyl, —O—CO-aryl, —O—CO-alkylenearyl or radical, or both radicals RA 6 and RA 7 together are an optionally substituted, unsaturated or aromatic heterocycle which, in addition to the ring nitrogen, can contain up to two further different or identical heteroatorns O, N, S,
      • RA 8 is hydrogen, a branched or unbranched, optionally substituted C1-C4-alkyl, CO—C1-C4-alkyl, SO2—C1-C4-alkyl or CO—O—C1-C4-alkyl radical or an optionally substituted aryl, CO-aryl, SO2-aryl, CO—O-aryl, CO-alkylenearyl, SO2-alkylenearyl, CO—O-alkylenearyl or alkylenearyl radical,
      • RA 9, RA 10
      • independently of one another are hydrogen, —CN, halogen, a branched or unbranched, optionally substituted C1-C6-alkyl radical or an optionally substituted aryl, arylalkyl, hetaryl, C3-C7-cycloalkyl radical or a radical CO-O-RA 14, O-RA 14, S-RA 14, NRA 15RA 16, SO2—NRA 15RA 16 or CO—NRA 15RA.sup-.16, or both radicals RA 9 and RA 10 together in structural element IA 14 are a 5- to 7-membered saturated, unsaturated or aromatic carbocycle or heterocycle which can contain up to three different or identical heteroatoms O, N, S and is optionally substituted by up to three identical or different radicals,
      • RA 11 is hydrogen, —CN, halogen, a branched or unbranched, optionally substituted C1-C6-alkyl radical or an optionally substituted aryl, arylalkyl, hetaryl, C3-C7-cycloalk- yl radical or a radical CO-o-RA 14, O-RA 14, S-RA 14, NRA 15RA 16, SO2—NRA 1-5RA 16 or CO—NRA 15RA 16,
      • RA 17 is hydrogen o,r in structural element IA 16, both radicals RA 9 and RA 17 together are a 5 to 7-membered saturated, unsaturated or aromatic heterocycle which, in addition to the ring nitrogen, can contain up to three different or identical heteroatoms O, N, S and is optionally substituted by up to three identical or different radicals,
      • RA 18, RA 19
      • independently of one another are hydrogen, a branched or unbranched, optionally substituted C1-C8-alkyl, C2-C6-alkenyl, C2-C6-alkynyl, C1-C5-alkylen- e-C1-C4-alkoxy, mono- and bisalkylaminoalkylene or acylaminoalkylene radical or an optionally substituted aryl, heterocycloalkyl, heterocycloalkenyl, hetaryl, C3-C7-cycloalkyl-, C1-C4-alkylene-C3-C7-cycloalkyl, arylalkyl, C1-C4-alkyleneheterocycloalkyl, C1-C4-alkyleneheteroc- ycloalkenyl or hetarylalkyl radical, or a radical —SO2—RG- 11, —CO—ORG11, —CO—NRG 11RG 11* or —CO-RG 11 which is independent of RG 11
      • Z1, Z2, Z3, Z4,
      • independently of one another are nitrogen, C—H, C-halogen or a branched or unbranched optionally substituted C—C1-C4-alkyl or C—C1-C4-alkoxy radical,
      • Z5 is NRA 8, oxygen or sulfur.
  • In a further very particularly preferred embodiment, the structural element A is a structural element of the formula IA 1 IA 4, IA 7, IA 8 or IA 9.
  • A branched or unbranched, optionally substituted C1C6-alkyl radical for RA 1 or RA 2 independently of one another is understood as meaning, for example, the corresponding radicals described above for RG 1, preferably methyl or trifluoromethyl.
  • For RA 1 or RA 2 in the structural elements IA 1, I2 2, IA 3 IA 17 the branched or unbranched optionally substituted radical CO—C1-C6-alkyl is composed, for example, of the group CO and the branched or unbranched, optionally substituted C1-C6-alkyl radicals described above for RA 1 or RA 2.
  • Optionally substituted hetaryl, hetarylalkyl, aryl, arylalkyl or C3C7-cycloalkyl radicals for RA 1 or RA 2 independently of one another are understood as meaning, for example, the corresponding radicals described above for RG 7.
  • For RA 1 or RA 2, the optionally substituted radicals Co-O-RA 14, O-RA 14, S-RA 14, NRA 15RA 16, CO—NRA 15RA 16 or SO2NRA 15RA 16 are composed, for example, of the groups CO—O, O, S, N, CO—N or SO2—N and the radicals RA 14, RA 15 or RA 16 described in greater detail below.
  • Further, both radicals RA 1 and RA 2 can together form a fused, optionally substituted, 5- or 6-membered, unsaturated or aromatic carbocycle or heterocycle which can contain up to three heteroatoms selected from the group consisting of O, N and S.
      • RA 13 and RA 13* are independently of one another hydrogen, CN,
      • halogen, such as fluorine, chlorine, bromine or iodine,
      • a branched or unbranched, optionally substituted C1-C6-alkyl radical, such as described above for RG 1, preferably methyl or trifluoromethyl or
      • an optionally substituted aryl, arylalkyl, hetaryl or C3-C7-cycloalkyl radical or a radical Co-O-RA 14, O-RA 14, S-RA 14, NRA 15RA 16, SO2NRA 15RA 16 or CO—NRA 15RA 16 as in each case described above for RA 1.
  • Preferred radicals for RA 13 and RA 13* are the radicals hydrogen, F, Cl, a branched or unbranched, optionally substituted C1-C5-alkyl radical, optionally substituted aryl or arylalkyl or a radical Co-O-RA 14, O-RA 14, NRA 15RA 16, SO2-NRA 15RA 16 or CO—NRA 15RA 16.
  • A branched or unbranched, optionally substituted C1-C6-alkyl, C3-C7-cycloalkyl, alkylenecycloalkyl, alkylene-C1-C4-alkoxy, C2-C6-alkenyl or C2-C6-alkynyl radical for RA 14 in structural element A is understood as meaning, for example, the corresponding radicals described above for RG 7.
  • Optionally substituted aryl, arylalkyl, hetaryl or alkylhetaryl radicals for RA 14 in structural element A are understood as meaning, for example, the corresponding radicals described above for RG 7.
  • Preferred radicals for RA 14 are hydrogen, a branched or unbranched, optionally substituted C1-C6-alkyl radical and optionally substituted benzyl.
  • A branched or unbranched, optionally substituted C1-C6-alkyl or arylalkyl radical or an optionally substituted C3-C7-cycloalkyl, aryl, hetaryl or hetaryalkyl radical for RA 15 or RA 16 independently of one another is understood as meaning, for example, the corresponding radicals described above for RA 14.
  • The branched or unbranched, optionally substituted CO—C1-C6-alkyl, SO2—C1-C6-alkyl, COO-C1-C6-alkyl, CO—NH—C1-C6-alkyl, COO-alkylenearyl, CO—NH-alkylenearyl, CO—NH-alkylenehetaryl or SO2-alkylenearyl radicals or the optionally substituted CO-aryl, SO2-aryl, CO—NH-aryl, CO—NH-hetaryl or CO-hetaryl radicals for RA 15 or RA 16 are composed, for example, of the corresponding groups —CO—, SO2—, —CO—O—, —CO—NH— and the corresponding branched or unbranched optionally substituted C1-C6alkyl, hetarylalkyl or arylalkyl radicals or the corresponding optionally substituted aryl or hetaryl radicals described above.
  • A radical —(CH2)n—(XA)j—RA 12 for RA 3 or RA 4 independently of one another is understood as meaning a radical which is composed of the corresponding radicals —(CH2)n—, (XA)j and RA 12. Here, n can be 0, 1, 2 or 3 and j can be: 0 or 1.
  • XA is a doubly bonded radical selected from the group consisting of —CO—, —CO—N(Rx 1)—, —N(Rx 1)—CO—, —N((Rx 1))-Co-N((Rx 1)*)-, —N(Rx1)—CO—O—, —O—, —S—, —SO2—, —SO2—N(Rx 1)—, —SO2—O—, —CO—O—, —O—CO—, —O—CO—N((Rx 1))—, —N(Rx1)— or —N((Rx 1))—SO2—.
      • RA 12 is hydrogen,
      • a branched or unbranched, optionally substituted C1-C6-alkyl radical, as described above for RG 7,
      • a C2-C6-alkynyl or C2-C6-alkenyl radical optionally substituted by C1-C4-alkyl or aryl,
      • or a 3- to 6-membered, saturated or unsaturated heterocycle which is substituted by up to three identical or different radicals and can contain up to three different or identical heteroatoms O, N, S, such as optionally substituted 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-furyl, 3-furyl, 2-pyrrolyl, 3-pyrrolyl, 2-thienyl, 3-thienyl, 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, 2-oxazolyl, 4-oxazolyl, 5-oxazolyl, 2-pyrimidyl, 4-pyrimidyl, 5-pyrimidyl, 6-pyrimidyl, 3-pyrazolyl, 4-pyrazolyl, 5-pyrazolyl, 3-isothiazolyl, 4-isothiazolyl, 5-isothiazolyl, 2-imidazolyl, 4-imidazolyl, 5-imidazolyl, 3-pyridazinyl, 4-pyridazinyl, 5-pyridazinyl, 6-pyridazinyl, 2-(1,3,4-thiadiazolyl), 2-(1,3,4)-oxadiazolyl, 3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, triazinyl.
  • Further, RA 12 and Rx 1 or RX1* can together form a saturated or unsaturated C3- C7-heterocycle which can optionally contain up to two further heteroatoms selected from the group consisting of O, S and N.
  • Preferably, the radical RA 12 together with the radical Rx 1 or Rx 1* forms a cyclic amine as the C3-C7-heterocycle in the case where the radicals are bonded to the same nitrogen atom, such as N-pyrrolidinyl, N-piperidinyl, N-hexahydroazeptnyl, N-morpholinyl or N-piperazinyl, where in heterocycles which carry free amine protons, such as N-piperazinyl, the free amine protons can be replaced by customary amine protective groups, such as methyl, benzyl, Boc (tert-butoxycarbonyl), Z (benzyloxycarbonyl), tosyl, —SO2—C1-C4-alkyl, —SO2-phenyl or —SO2-benzyl.
  • A branched or unbranched, optionally substituted C1-C6-alkyl, C2-C12-alkynyl, preferably C2-C6-alkynyl or C2-C6-alkenyl radical, an optionally substituted C3-C7-cycloalkyl, aryl, arylalkyl or hetaryl radical for Rx1 and Rx 1* independently of one another is understood as meaning, for example, the corresponding radicals described above for RG 7.
  • Preferred branched or unbranched, optionally substituted C1-C6-alkoxyalkyl for Rx 1 and Rx 1* are independently of one another methoxymethylene, ethoxymethylene, butoxymethylene, methoxyethylene or ethoxyethylene.
  • Preferred branched or unbranched, optionally substituted radicals CO—C1-C6-alkyl, CO—O—C1-C6-alkyl, SO2—C1-C6-alkyl, CO—O-alkylenearyl, CO-alkylenearyl, CO-aryl, SO2-aryl, CO-hetaryl or SO2-alkylenearyl are preferably composed of the C1-C6-alkyl, arylalkyl, aryl or hetaryl radicals and the radicals —CO—, —O—, —SO2— described above.
  • Preferred radicals for Rx 1and Rx 1* are independently of one another hydrogen, methyl, cyclopropyl, allyl and propargyl.
      • RA 3 and RA 4 can further together form a 3- to 8-membered saturated, unsaturated or aromatic N heterocycle which can additionally contain two further, identical or different heteroatoms O, N or S, where the cycle can be optionally substituted or a further, optionally substituted, saturated, unsaturated or aromatic cycle can be fused to this cycle,
      • RA 5 is a branched or unbranched, optionally substituted C1-C6-alkyl, arylalkyl, C1-C4-alkyl-C3-C7-c- ycloolkyl or C3-C7-cycloalkyl radical or an optionally substituted aryl, hetaryl, heterocycloalkyl or heterocycloalkenyl radical, such as described above for RG 7.
      • RA 6 and RA 6* are independently of one another hydrogen, a branched or unbranched, optionally substituted
      • C1-C4-alkyl radical, such as optionally substituted methyl, ethyl, propyl 1-methylethyl, butyl, 1-methylpropyl, 2-methylpropyl or 1,1-dimethylethyl,
      • —CO—O—C1-C4-alkyl or —CO—C1-C4-alkyl radical, such as composed of the group —CO—O— or —CO— and the C1-C4-alkyl radicals described above,
      • arylalkyl radical, as described above for RG 7,
      • —CO—O-alkylenearyl or —CO-alkylenearyl radical such as composed of the group —CO—O— or —CO— and the arylalkyl radicals described above, —CO—O-allyl or —CO-allyl radical, or C3-C7-cycloalkyl radical, such as described above for RG 7.
  • Further, both radicals RA 6 and RA 6* in structural element IA 7 can together form an optionally substituted, saturated, unsaturated or aromatic heterocycle which, in addition to the ring nitrogen, can contain up to two further different or identical heteratoms O, N, S.
  • RA 7 is hydrogen, —OH, —CN, —CONH2, a branched or unbranched, optionally substituted C1-C4-alkyl radical, for example as described above for RA 6, C1-C4-alkoxy, arylalkyl or C3-C7-cycloalkyl radical, for example as described above for RL 14, a branched or unbranched, optionally substituted —O—CO—C1-C4-alkyl radical, which is composed of the group —O—CO— and, for example, of the C1-C4-alkyl radicals mentioned above or an optionally substituted —O-alkylenearyl, —O—CO-aryl, —O—CO-alkylenearyl or —O—CO-allyl radical which is composed of the groups —O— or —O—CO— and, for example, of the corresponding radicals described above for RG 7.
  • Further, both radicals RA 6 and RA 7 can together form an optionally substituted unsaturated or aromatic heterocycle which, in addition to the ring nitrogen, can contain up to two further different or identical heteroatoms O, N, S.
  • For RA 8 in structural element A, a branched or unbranched, optionally substituted C1-C4-alkyl radical or an optionally substituted aryl or arylalkyl radical is understood as meaning, for example, the corresponding radicals described above for RA 15, where the radicals CO—C1-C4-alkyl, SO2- C1-C4-alkyl, CO—O—C1-C4-alkyl, CO-aryl, SO2-aryl, CO—O-aryl, CO-alkylenearyl, SO2-alkylenearyl or CO—O-alkylenearyl are composed analogously to the other composed radicals of the group consisting of CO, SO2 and COO and for example, of the corresponding C1-C4-alkyl, aryl or arylalkyl radicals described above for RA 15, and these radicals can be optionally substituted.
  • In each case, for RA 9 or RA 10, a branched or unbranched, optionally substituted C1-C6-alkyl radical or an optionally substituted aryl, arylalkyl, hetaryl or C3-C7-cycloalkyl radical independently of one another is understood as meaning, for example, the corresponding radicals described above for RA 14, preferably methyl or trifluoromethyl.
  • In each case, for RA 9 or RA 10, a radical CO—O—RA 14, O—RA 14, S-RA 14, SO2—NRA 15RA 16, NRA 15RA 16or CO—NRA 15RA 16 independently of one another is understood as meaning, for example, the corresponding radicals described above for RA 13.
  • Further, both radicals RA 9 and RA 10 together in structural element IA 14 can form a 5- to 7-membered saturated, unsaturated or aromatic carbocycle or heterocycle, which can contain up to three different or identical heteroatoms O, N, S and is optionally substituted by up to three identical or different radicals.
  • Substituents in this case are in particular understood as meaning halogen, CN, a branched or unbranched, optionally substituted C1-C4-alkyl radical, such as methyl or trifluoromethyl or the radicals O—RA 14, S-RA 14, NRA 15RA- 16, CO—NRA 15RA 16 or —((RA 8)HN)C═NR.sub- .A7.
  • A branched or unbranched, optionally substituted C1-C6-alkyl radical or an optionally substituted aryl, arylalkyl, hetaryl, C3-C7-cycloalkyl radical or a radical CO—O—RA 14, O—RA 14, S—RA 14, NRA 15RA 16, SO2—NRA 15RA 16 or CO—NRA 15RA 16 for RA 11 is understood, for example, as meaning the corresponding radicals described above for RA 9.
  • Further, in structural element IA 16, both radicals RA 9 and RA 17 together can form a 5- to 7-membered saturated, unsaturated or aromatic heterocycle which, in addition to the ring nitrogen, can contain up to three different or identical heteroatoms O, N, S and is optionally substituted by up to three identical or different radicals.
  • A branched or unbranched, optionally substituted C1-C8-alkyl, C2-C6-alkenyl, C2-C6-alkynyl, C1-C5-alkylene-C1-C4-alkoxy, mono- and bisalkylaminoalkylene or acylaminoalkylene radical or an optionally substituted aryl, heterocycloalkyl, heteroeycloalkenyl, hetaryl, C3-C7-cycloalkyl, C1-C4-alkylene-C3-C7-cycl- oalkyl, arylalkyl, C1-C4-alkyleneheterocycloalkyl, C1-C4-alkyleneheterocycloalkenyl or hetarylalkyl radical, or a radical —SO2—RG 11, —CO—ORG 11, —CO—NRG 11RG 11* or —CO-RG 1 for RA 18 and RA 19 independently of one another is understood as meaning, for example, the radicals described above for RG 12, preferably hydrogen or a branched or unbranched, optionally substituted C1-C8-alkyl radical.
  • Z1, Z2, Z3, Z4 are independently of one another nitrogen, C—H, C-halogen, such as C—F, C—Cl, C—Br or C—I or a branched or unbranched, optionally substituted C—C1-C4-alkyl radical which is composed of a carbon radical and, for example, a C1-C4-alkyl radical described above for RA 6 or a branched or unbranched optionally substituted C—C1-C4-alkoxy radical which is composed of a carbon radical and, for example, a C4-alkoxy radical described above for RA 7.
  • Z5 is oxygen, sulfur or a radical NRA 8.
  • Preferred structural elements A are composed of at least one preferred radical of the radicals belonging to the structural element A, while the remaining radicals are widely variable.
  • Particularly preferred structural elements A are composed of the preferred radicals of the structural element A.
  • In a preferred embodiment, the spacer structural element E is understood as meaning a structural element that consists of a branched or unbranched aliphatic C2-C30-hydrocarbon radical which is optionally substituted and contains heteroatoms and/or of a 4-to 20-membered aliphatic or aromatic mono- or polycyclic hydrocarbon radical which is optionally substituted and contains heteroatoms.
  • In a further preferred embodiment, the spacer structural element E is composed of two to four substructural elements, selected from the group consisting of E1 and E2, where the sequence of linkage of the substructural elements is arbitrary and E1 and E2 have the following meanings:
      • E1 is a substructural element of the formula IE1

  • —(YE)k1—(CRE 1RE 2)c-(QE)k2-(-CRE 3RE 4)d-IE1
  • and
      • E2 is a substructural element of the formula IE2

  • —(NRE 11)k3—(CRE 5RE 6)f-(ZE)k4-(CRE 7RE 8)g-(XE)kS—(CRE 9RE 10)h-(NRE 11)k6-IE2,
  • where
    c, d, f, g, h
      • independently of one another are 0, 1 or 2,
        k1, k2, k3, k4, k5, k6
      • independently of one another are 0 or 1,
    XE, QE
      • independently of one another are an optionally substituted 4- to 11-membered mono- or polycyclic, aliphatic or aromatic hydrocarbon which can contain up to 6 double bonds and up to 6 identical or different heteroatoms selected from the group N, O and S, where the ring carbons and/or the ring nitrogens can optionally be substituted,
    YE, ZE
      • independently of one another are CO, CO—NRE 12, NRE 13-CO, sulfur, SO, SO2, SO2—NRE 12, NRE 12—SO2, CS, CS—NRE 12, NRE 12—CS, CS—O, O—CS, CO—O, O—CO, oxygen, ethynylene, CRE 13-O—CR.sub- .E14, C(═CRE 13RE 14), CRE 13═CRE-14, —CRE 13(CRE 15)—CHRE 14- or —CHRE 13-CRE 14(0RE 15)-,
      • RE 1, RE 2, RE 3, RE 4, RE 5, RE 6, RE 7, RE 8, RE 9, RE 10
      • independently of one another are hydrogen, halogen, a hydroxyl group, a branched or unbranched, optionally substituted C1-C6-alkyl, C2-C6-alkenyl, C2-C6-alkynyl or alkylenecycloalkyl radical, a radical —(CH2)x—(WE).su- b.z- RE 17, an optionally substituted C3-C7-cycloalkyl, aryl, arylalkyl, hetaryl or hetarylalkyl radical or independently of one another in each case two radicals RE 1 and RE 2 or RE 3 and Re 4 or RE 5 and RE 6 or RE 7 and RE 8 or RE 9 and RE 10 together are, a 3- to 7-membered, optionally substituted, saturated or unsaturated carbocycle or heterocycle which can contain up to three heteroatoms selected from the group O, N and S,
      • x is 0, 1, 2, 3 or 4,
      • z is 0 or 1,
      • WE is —CO—, —CO—N(RW 2)—, —N(RW 2)—Co—, —N(RW 2)—CO—N(RW 2*)—, —N(RW 2)—CO—O—, —O—, —S—, —SO2—, —SO2—N(RW 2)—, —SO2—O—, —CO—O—, —O—CO—, —O—CO—N(RW 2)—, —N(RW 2)— or —N(RW 2)—SO2—,
      • RW 2, RW 2*
      • independently of one another are hydrogen, a branched or unbranched, optionally substituted C1-C6-alkyl, C2-C6-alkenyl, C2-C8-alkynyl, CO—C1-C6-alk- yl, CO—O—C1-C6-alkyl or SO2—C1-C6-alkyl radical or an optionally substituted hetaryl, hetarylalkyl, arylalkyl, C3-C7-cycloalkyl, CO—O-alkylenearyl, CO-alkylenearyl, CO-aryl, SO2-aryl, CO-hetaryl or SO2-alkylenearyl radical,
      • RE 17 is hydrogen, a hydroxyl group, CN, halogen, a branched or unbranched, optionally substituted C1-C6-alkyl radical, an optionally substituted C3-C7-cycloalkyl, aryl, hetaryl or arylalkyl radical, a C2-C6-alkynyl or C2-C6-alkenyl radical optionally substituted by C1-C4alkyl or aryl, an optionally substituted C6-C12-bicycloalkyl, C1-C6-alkylene-C6-C 12—bicycloalkyl, C7-C20-tricycloalkyl or C1-C6-alkylene-C- 7-C20-tricycloalkyl radical, or a 3- to 8-membered, saturated or ausatutated heterocycle substituted by up to three identical or different radicals, which can contain up to three different or identical heteroatoms O, N, S, where two radicals together can be a fused, saturated, unsaturated or aromatic carbocycle or heterocycle which can contain up to three different or identical heteroatoms O, N, S and the cycle can optionally be substituted or a further, optionally substituted, saturated, unsaturated or aromatic cycle can be fused to this cycle, or the radical RE 17 forms, together with RW 2 or RW 2* a saturated or unsaturated C3-C7-heterocycle which can optionally contain up to two further heteroatoms selected from the group O, S and N,
      • RE 11, RE 11*
      • independently of one another are hydrogen, a branched or unbranched, optionally substituted C1-C6-alkyl, C1-C6-alkoxyalkyl, C2-C6-alkenyl, C2-C12-alkynyl, CO—C1-C6-alkyl, CO—O—C1-C6-alkyl, CO—NH—C1-C6-alkoxyalkyl, CO—NH—C1-C6-alkyl or SO2—C1-C6-alkyl radical or an optionally substituted hetaryl, arylalkyl, C3-C7-cycloalkyl, CO—O-alkylenearyl, CO—NH-alkylenearyl, CO-alkylenearyl, CO-aryl, CO—NH-aryl, SO2-aryl, CO-hetaryl, SO2-alkylenearyl, SO2-hetaryl or SO2-alkylenehetaryl radical,
      • RE 12 is hydrogen, a branched or unbranched, optionally substituted C1-C6-alkyl, C2-C6-alkenyl, C2-C8-alkynyl radical, an optionally substituted C3-C7-cycloalkyl, hetaryl, arylalkyl or hetarylalkyl radical or a radical CO—RE 16, COORE 16 or SO2—RE 16,
      • RE 13, RE 14
      • independently of one another are hydrogen, a hydroxyl group, a branched or unbranched, optionally substituted C1-C6-alkyl, C1-C4-alkoxy, C2-C6-alkenyl, C2-C6-alkynyl or alkylenecycloalkyl radical or an optionally substituted C3-C7-cycloalkyl, aryl, arylalkyl, hetaryl or hetarylalkyl radical,
      • RE 15 is hydrogen, a branched or unbranched, optionally substituted C1-C6-alkyl, C2-C6-alkenyl, C2-C6-alkynyl or alkylenecycloalkyl radical or an optionally substituted C3-C7-cycloalkyl, aryl, arylalkyl, hetaryl or hetarylalkyl radical,
      • RE 16 is hydrogen, a hydroxyl group, a branched or unbranched, optionally substituted C1-C6-alkyl, C2-C6-alkenyl, C2-C6-alkynyl or C1-C5-alkylene-C1-C4-alkoxy radical, or an optionally substituted aryl, heterocycloalkyl, heterocycloalkenyl, hetaryl, C3-C7-cycloalkyl, C1-C4-alkylene-C3-C7-cycl- oalkyl, arylalkyl, C1-C4-alkylene-C3-C7-heterocycloalk- yl C1-C4-alkylene-C3-C7-heterocycloalkenyl or hetarylalkyl radical.
  • The coefficient c is preferably 0 or 1, the coefficient d is preferably 1 or 2, the coefficients f, g, h independently of one another are preferably 0 or 1 and K6 is preferably 0.
  • An optionally substituted 4- to 11-membered mono- or polycyclic aliphatic or aromatic hydrocarbon which can contain up to 6 double bonds and up to 6 identical or different heteroatoms selected from the group consisting of N, O, S, where the ring carbons or ring nitrogens can optionally be substituted, for QE and XE independently of one another is preferably understood as meaning optionally substituted arylene, such as optionally substituted phenylene or naphthylene or optionally substituted hetarylene such as the radicals
  • Figure US20160296592A1-20161013-C00018
  • and their substituted or fused derivatives, or radicals of the formulae IE 1 to IE 11,
  • Figure US20160296592A1-20161013-C00019
    Figure US20160296592A1-20161013-C00020
      • where the incorporation of the radicals can take place in both orientations. Aliphatic hydrocarbons are understood as meaning, for example, saturated and unsaturated hydrocarbons.
      • Z6 and Z7 are independently of one another CH or nitrogen.
      • Z8 is oxygen, sulfur or NH,
      • Z9 is oxygen, sulfur or NRE 20.
      • r1, r2, r3 and t are independently of one another 0, 1, 2 or 3.
      • s and u are independently of one another 0, 1 or 2.
  • Particularly preferably, XE and QE independently of one another are optionally substituted phenylene, a radical
  • Figure US20160296592A1-20161013-C00021
      • and their substituted or fused derivatives, or radicals of the formulae IE1, IE2, IE3, IE4 and IE7, where the incorporation of the radicals can take place in both orientations.
      • RE18 and RE19 are independently of one another hydrogen, —NO2, —NH2, —CN, —COOH, a hydroxyl group, halogen, a branched or unbranched, optionally substituted C1-C6-alkyl, C1-C4-alkoxy, C2-C6-alkenyl, C2-C6-alkynyl or alkylenecycloalkyl radical or an optionally substituted C3-C7-cycloalkyl, aryl, arylalkyl, hetaryl or hetarylalkyl radical, as in each ease described above.
  • RE20 is, independently of one another, hydrogen, a branched or unbranched, optionally substituted C1-C6-alkyl, C1-C6-alkoxyalkyl, C3-C12-alkynyl, CO—C1-C6-alkyl, C0-O—C1-C6-alkyl or SO2—C1-C6-alkyl radical or an optionally substituted C3-C7-cycloalkyl, aryl, arylalkyl CO—O-alkylenearyl, CO-alkylenearyl, CO-aryl, SO2-aryl, hetaryl, CO-hetaryl or SO2alkylenearyl radical, preferably hydrogen or a branched or unbranched, optionally substituted C1-C6-alkyl radical.
      • YE and ZE are independently of one another CO, CO—NRE12, NRE12-CO, sulfur, SO, SO2, SO2—NRE12, NRE12-SO2, CS, CS—NRE12, NRE12-CS, CS—O, O—CS, CO—O, O—CO, oxygen, ethynylene, CRE13-O—CRE14, C(═CRE13RE14), CRE13═CRE14, —CRE13(ORE15)—CHRE14—or —CHRE13-CRE14(ORE15)-, preferably CO, SO2 and oxygen.
      • RE12 is hydrogen, a branched or unbranched, optionally substituted C1-C6-alkyl, C2-C6-alkenyl or C2-C8-alkynyl radical or an optionally substituted C3-C7-cycloalkyl, hetaryl, arylalkyl or hetarylalkyl radical, such as correspondingly described above for RG7 or a radical CO-RE16, COORE16 or SO2—RE16, preferably hydrogen, methyl, allyl propargyl and cyclopropyl.
  • A branched or unbranched, optionally substituted C1-C6-alkyl, C2-C6-alkenyl or C2-C6-alkynyl radical or an optionally substituted C3-C7-cycloalkyl, aryl, arylalkyl, hetaryl or hetarylalkyl radical for RE13, RE14 or RE15 independendy of one another is understood as meaning, for example, the corresponding radicals described above for RG7.
  • A branched or unbranched, optionally substituted C1-C4-alkoxy radical for RE13 or RE14 independently of one another is understood as meaning, for example, the C1-C4-alkoxy radicals described above for RA14.
  • Preferred alkylenecycloalkyl radicals for RE13 1 RE14 or RE15 independently of one another are, for example, the C1-C4-alkylene-C3-C7-cycloalkyl radicals described above for RG7.
  • A branched or unbranched, optionally substituted C1-C6-alkyl, C2-C6-alkenyl, C2-C6-alkynyl or C1-C5-alkylene-C1-C4-alkoxy radical, or an optionally substituted aryl, heterocycloalkyl, heterocycloalkenyl, hetaryl, C3-C7-cycloalkyl, C1-C4-alkylene-C3-C.s-ub.7-cycloalkyl, arylalkyl, C1-C4-akylene-C3-C7-heter- ocycloalkyl, C1-C4-alkylene-C3-C7-heterocycloalkenyl hetarylalkyl radical for RE16 is understood as meaning, for example, the corresponding radicals described above for RG11.
  • A branched or unbranched, optionally substituted C1-C6-alkyl, C2-C6-alkenyl, C2-C6-alkynyl or alkylenecycloalkyl radical or an optionally substituted C3-C7-cycloalkyl, aryl, arylalkyl, hetaryl or hetarylalkyl radical for RE1, RE2, RE3, RE4, RE5, RE6, RE7, RE8, RE9 or RE10 independently of one another is understood as meaning, for example, the corresponding radicals mentioned above for RG 7.
  • Further, two radicals RE3 and RE4 or RE5 and RE6 or RE7 and RE8 or RE9 and RE10 can in each ease independently of one another together form a 3- to 7-membered, optionally substituted, saturated or unsaturated carbo- or heterocycle, which can contain up to three heteroatoms from the group consisting of O, N and S.
  • The radical —(CH2)x—(WE) Z-RE17 is composed of a CO—C4-alkylene radical, optionally a bonding element WE selected from the group —CO—, —CO—N(R2)—, —N(R12)—CO—, —N(RW2)—CO—N(RW2*)-, —N(R2)—CO—O—, —O—, —S—, —SO2—, —SO2—N(RW2)—, —SO2—O—, —CO—O—, —O—CO—O—CO—N(RW2)—, —N(RW2)— or —N(RW2)—SO2—, preferably selected from the group —CO—N(RW2)—, —N(RW2)—CO—, —O—, —SO2—N(RW2)—, —N(RW2)— or —N(RW2)—SO2, and the radical RE17, where
      • RW2 and RW2*
      • independently of one another are hydrogen, a branched or unbranched, optionally substituted C1-C6-alkyl, C2-C6-alkenyl, C2-C8-alkynyl, CO—C1-C6-alk- yl, CO—O—C1-C6-alkyl or SO2—C1-C6-alkyl radical or an optionally substituted hetaryl, hetarylalkyl, arylalkyl, C3-C-7-cycloalkyl, CO—O-alkylenearyl, CO-alkylenearyl, CO-aryl, SO2-aryl, CO-hetaryl or SO2-alkylenearyl radical, preferably independently of one another are hydrogen, methyl, cyclopropyl, ally, propargyl, and
      • RE17
      • is hydrogen, a hydroxyl group, CN, halogen, a branched or unbranched, optionally substituted C1-C6-alkyl radical, an optionally substituted C3-C7-cycloalkyl, aryl, hetaryl or arylalkyl radical, a C2C6alkynyl or C2-C6-alkenyl radical optionally substituted by C1-C4alkyl or aryl, an optionally substituted C6-C12-bicycloalkyl, C1-C6-alkylene-C6-C12-bicycloalkyl, C7-C20-tricycloalkyl or C1-C6-alkylene-C7-C- 20-tricycloalkyl radical, or a 3- to 8-membered, saturated or unsaturated heterocycle which is substituted by up to three identical or different radicals and can contain up to three different or identical heteroatoms O, N, S, where two radicals together can be a fused, saturated, unsaturated or aromatic carbocycle or heterocycle which can contain up to three different or identical heteroatoms O, N, S, and the cycle can be optionally substituted or a further, optionally substituted, saturated, unsaturated or aromatic cycle can be fused to this cycle, such as optionally substituted 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-furyl, 3-furyl, 2-pyrrolyl, 3-pyrrolyl, 2-thienyl, 3-thienyl, 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, 2-oxazolyl, 4-oxazolyl, 5-oxazolyl, 2-pyrimidyl, 4-pyrimidyl, 5-pyrimidyl, 6-pyrimidyl, 3-pyrazolyl, 4-pyrazolyl, 5-pyrazolyl, 3-isothiazolyl, 4-isothiazolyl, 5-isothiazolyl, 2-imidazolyl, 4-imidazolyl, 5-imidazolyl, 3-pyridazinyl, 4-pyridazinyl, 5-pyridazinyl, 6-pyridazinyl, 2-(1,3,4-thiadiazolyl), 2-(1,3,4)-oxadiazolyl, 3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl or triazinyl.
  • Further, RE17 and RW2 or RW2* can together form a saturated or unsaturated C3-C7-heterocycle which can optionally contain up to two further heteroatoms selected from the group consisting of O, S and N.
  • Preferably, the radicals RE17 and RW2 or RW2* together form a cyclic amine as the C3-C7-heterocycle in the case where the radicals are bonded to the same nitrogen atom, such as N-pyrrolidinyl, N-hexahydroazepinyl, N-morpholinyl or N-piperazinyl where in heterocycles which carry free amine protons, such as N-piperazinyl, the free amine protons can be replaced by customary amine protective groups, such as methyl, benzyl, Boc (tert-butoxycarbonyl), Z (benzyloxycarbonyl), tosyl, —SO2—C1-C4-alkyl, —SO2-phenyl or —SO2-benzyl.
  • Preferred radicals for RE1, RE2, RE3, RE4, RE5, RE6, RE7, RE8, RE9 or RE10 are independently of one another hydrogen, halogen, a branched or unbranched, optionally substituted C1-C6-alkyl radical, optionally substituted aryl or the radical —(CH2)x—(WE)z—RE17.
  • Particularly preferred radicals for RE1, RE2, RE3, RE4, RE5, RE6, RE7, RE8, RE9 or RE10 are independently of one another hydrogen, F, a branched or unbranched, optionally substituted C1-C4-alkyl radical, in particular methyl.
  • A branched or unbranched, optionally substituted C1-C6-alkoxyalkyl, C2-C6-alkenyl, C2-C12-alkynyl or arylalkyl radical or an optionally substituted aryl, hetaryl or C3-C7-cycloalkyl for RE11 and RE11* in structural element E independently of one another is understood as meaning, for example, the corresponding radicals described above for RG7.
  • The branched or unbranched, optionally substituted radicals CO—C1-C6-alkyl, CO—O—C1-C6-alkyl, CO—NH—C1-C6-alkoxalkyl, CO—NH—C1-C6-alkyl or SO2—C1-C6-alkyl radical or the optionally substituted radicals Co-O-alkylenearyl, CO—NH-alkylenearyl, CO-alkylenearyl, CO-aryl, CO—NH-aryl, SO2-aryl, CO-hetaryl, SO2-alkylenearyl, SO2-hetaryl or SO2-alkylenehetaryl for RE11 and RE11* independently of one another are composed, for example, of the corresponding groups CO, COO, CONH or SO2 and the corresponding radicals mentioned above.
  • Preferred radicals for RE11 or RE11* are independently of one another hydrogen, a branched or unbranched, optionally substituted C1-C6-alkyk, C1-C6-alkoxy, C2-C6-alkenyl,
      • C2-C12-alkynyl or arylalkyl radical, or an optionally substituted hetaryl or C3-C7-cycloalkyl radical.
  • Particularly preferred radicals for RE11 or RE11* are hydrogen, methyl, cyclopropyl, allyl or propargyl.
  • In a particularly preferred embodiment of structural element E1, structural element E1 is a radical —CH2—CH2—CO—, —CH2—CH2—CH2—CO— or a C1-C5-alkylene radical.
  • In a particularly preferred embodiment of structural element E, the spacer structural element E used is a structural element of the formula IE1E2

  • -E2-E1-  IE1E2
      • where the structural elements E2 and E1 have the meanings described above.
  • Preferred structural elements E are composed of at least one preferred radical of the radicals belonging to structural element E, while the remaining radicals are widely variable.
  • Particularly preferred structural elements E are composed of the preferred radicals of structural element E.
  • Preferred structural elements B are composed either of the preferred structural element A, while E is widely variable or of the preferred structural element E, while A is widely variable.
  • The compounds of the formula I, and also the intermediates for their preparation, can have one or more asymmetric substituted carbon atoms. The compounds can be present as pure enantiomers or pure diastereomers or as a mixture thereof. The use of an enantiomerically pure compound as the active compound is preferred.
  • The compounds of the formula I can also be present in other tautomeric forms.
  • The compounds of the formula I can also be present in the form of physiologically tolerable salts.
  • The compounds of the formula I can also be present as prodrugs in a form in which the compounds of the formula I are liberated under physiological conditions. By way of exammple, reference may be made here to the group T in structural element L, which in some cases contains groups which are hydrolyzable to the free carboxylic acid group under physiological conditions. Derivatized structural elements B or A are also suitable which liberate the structural element B or A respectively under physiological conditions.
  • In preferred compounds of the formula I, in each case one of the three structural elements B, G or L has the preferred range, while the remaining structural elements are widely variable.
  • In particularly preferred, compounds of the formula I, in each case two of the three structural elements B, G or L have the preferred range, while the remaining structural elements are widely variable.
  • In very particularly preferred compounds of the formula I, in each case all three structural elements B, G or L have the preferred range, while the remaining structural element is widely variable.
  • Preferred compounds of the formula I contain, for example, the preferred structural element G, while the structural elements B and L are widely variable.
  • In particularly preferred compounds of the formula I, for example, B is replaced by the structural element A-E- and the compounds contain, for example, the preferred structural element G and the preferred structural element A, while the structural elements E and L are widely variable.
  • Further particularly preferred compounds of the formula I contain, for example, the preferred structural element G and the preferred structural element A, while the structural elements E and L are widely variable.
  • Very particularly preferred compounds of the formula I in which A-E- is B- are listed below, the number before the text block being the number of an individualized compound of the formula I, and in the text block A-E-G-L the abbreviations being separated by a bonding dash in each case for an individual structural element A, E, G or L and the meaning of the abbreviations of the structural elements being explained after the table.
      • No. A-E-G-L
      • 1 bhs-dibema2-mmophec-es
      • 2 gua-mepipe2-phec-es
      • 3 gua-35thima2-4phaz-es
      • 4 bhs-apma2-pclphec-es gua-a23thima2-4bec-es
      • 6 bim-dibema2-4bec-es
      • 7 2py-bam2-pipmaz-es
      • 8 bim-bam2-mmphec-es
      • 9 2py-a23thima2-thec-es gua-pipa2-4pec-es
      • 11 dhim-35thima2-thec-es
      • 12 gua-a24thima2-amaz-es
      • 13 bim-pyma2-phec-es
      • 14 gua-a24thima2-3bzlaz-es bhs-inda2-thec-es
      • 16 2py-a24thima2-3bec-nes
      • 17 gua-a24thima2-phaz-es
      • 18 gua-bam2-pymaz-es
      • 19 gua-me35thima2-phec-es
      • 2py-dibema2-4pec-es
      • 21 bhs-35thima2-thec-gs
      • 22 bhs-aaf-3bec-es
      • 23 im-35thima2-thec-es
      • 24 bhs-a23thima2-3ipec-es bim-pipa2-4pec-es
      • 26 bhs-mea2-thec-es
      • 27 gua-dibema2-7 cmc-es
      • 28 2py-apma2-phaz-es
      • 29 bhs-apma2-7 cmc-es thpym-bam2-4pec-es
      • 31 bim-me35thima2-4pec-es
      • 32 bim-a24thima2-3bec-es
      • 33 bhs-me42thiaz2-phaz-es
      • 34 2py-42thiaz2-thec-es
      • 2py-pipa2-cpec-es
      • 36 bim-35thima2-pymaz-es
      • 37 bhs-a23thima2-3bec-es
      • 38 2py-apma2-ppec-es
      • 39 bhs-35thima2-pclphec-es
      • 40 2py-buta-3bec-es
      • 41 bim-a23thima2-7 cmc-gs
      • 42 bhs-hexa-thec-es
      • 43 bim-a23thima2-4pec-f2es
      • 44 2py-35thima2-7 cmc-es
      • 45 gua-chex2-4pec-es
      • 46 bhs-edia2-thec-es
      • 45 47 bhs-bam2-phaz-es
      • 48 amim-35thima2-3bec-es
      • 49 clim-apma2-3bec-es
      • 50 gua-pipa2-phec-mals
      • 51 am2py-a24thima2-thec-es
      • 52 bhs-apma2-phaz-gs
      • 53 2py-inda2-thec-es
      • 54 bim-35thima2-mmophec-es
      • 55 2py-inda2-3bec-es
      • 56 2py-mepipe2-thec-es
      • 57 bim-bam2-thec-es
      • 58 bim-bam2-4phaz-es
      • 59 2py-apma2-3bec-es
      • 60 bhs-a24thima2-pmophec-es
      • 61 bim-dibema2-thec-es
      • 62 mam2py-a24thima2-phaz-es
      • 63 2py-mea2-3bec-es
      • 64 bim-penta-4pec-es
      • 65 gua-prodia2-7 cmc-es
      • 66 bhs-dibema2-cpec-es
      • 67 2py-hexa-phaz-es
      • 68 gua-apma2-3ipec-es
      • 69 bim-apma2--phec-ms
      • 70 gua-35thima2-phec-ps
      • 71 bim-pipa2-3bec-es
      • 72 gua-a23thima2-phec-es
      • 73 2py-42thiaz2-phaz-es
      • 74 bim-me35thima2-7 cmc-es
      • 75 bhs-bam2-mpphec-es
      • 76 gua-dibema2-thec-es
      • 77 clim-bam2-thec-es
      • 78 dimethpym-a23thima2-thec-es
      • 79 gua-dibema2-4pec-cs
      • 80 bhs-apma2-3bzlaz-es
      • 81 gua-a24thima2-4pec-es
      • 82 bhs-pyma2-phaz-es
      • 83 gua-apma2-7 cmc-es
      • 84 bhs-a23thima2-4phaz-es
      • 85 bhs-penta-3bec-es
      • 86 gua-aof-7 cmc-es
      • 87 2py-a23thima2-phaz-ms
      • 88 bim-dibema2-phaz-es
      • 40 89 bim-35thima2-phec-as
      • 90 bim-apma2-cpec-es
      • 91 bhs-pipa2-3bec-nes
      • 92 2py-pipa2-mmophec-es
      • 93 bhs-35thima2-3bec-es
      • 45 94 bhs-dibema2-phaz-es
      • 95 gua-pipa2-3bec-es
      • 96 bim-pipa2-phec-es
      • 97 gua-42thiaz2-phec-es
      • 98 pippy-a24thima2-4pec-es
      • 99 2py-35thima2-thec-es
      • 100 2py-bam2-7 cmc-es
      • 101 2py-35thima2-pmophec-es
      • 102 bhs-dibema2-thec-es
      • 103 bim-aof-4pec-es
      • 104 bim-hexa-phec-es
      • 105 2py-a24thima2-7 cmc-es
      • 106 gua-a24thima2-phec-gs
      • 107 gua-me25thima2-7 cmc-es
      • 108clim-a24thima2-7 cmc-es
      • 109 gua-apma2-4pec-es
      • 110 bim-35thima2-cpec-es
      • 111 2py-me35thima2-thec-es
      • 112 bhs-a24thima2-dbc-es
      • 113 bim-bam2-4pec-es
      • 114 amim-a24thima2-4pec-es
      • 115 2py-dibema2-amec-es
      • 116 2py-a23thima2-dbc-es
      • 117 bim-bam2-4pec-ps
      • 118 2py-ba m2-mmophec-es
      • 119 bim-apma2-3bec-es
      • 120 bhs-pdagk-thec-es
      • 121 gua-42thiaz2-7 cmc-es
      • 122 gua-a23thima2-thec-es
      • 123 bim-apma2-4pec-es
      • 124 thpym--35thima2-phec-es
      • 125 bim-bam2-7 cmc-es
      • 126 mam2py-bam2-4pec-es
      • 127 bhs-edia2-3bec-es
      • 128 bhs-a23thima2-amec-es
      • 129 gua-dibema2-3bec-es
      • 130 bim-me42thiaz2-7 cmc-es
      • 131 bhs-a23thima2-phec-es
      • 132 bim-diberma2-mpphec-es
      • 133 2py-prodia2-thec-es
      • 134 bhs-bam2-mophaz-es
      • 135 bhs-a24thima2-7 cmc-es
      • 40 136 im-dibema2-4pec-es
      • 137 imhs-a24thima2-thec-es
      • 138 bhs-a24thima2-dmaphec-es
      • 139 2py-pipa2-dmaphec-es
      • 140 2py-a24thima2-4pec-es
      • 141 2py-dibema2-7 cmc-es
      • 142 bhs-apma2-phaz-es
      • 143 gua-pipa2-mophaz-es
      • 144 dhim-dibema2-4pec-es
      • 145 gua-pipa2-mpphec-es
      • 146 bim-a23thima2-4pec-es
      • 147 2py-dibema2-4phaz-es
      • 148 bim-42thiaz2-4pec-es
      • 149 am2py-dibema2-3bec-es
      • 150 bim-pipa2-7 cmc-es
      • 151 gua-bam2-dmaphec-es
      • 152 bhs-pipa2-amec-es
      • 153 2py-apma2-mpphec-es
      • 154 2py-hexa-3bec-es
      • 155 bim-apma2-7 cmc-es
      • 156 bim-a23thima2-pclphec-es
      • 157 gua-a24thima2-pclphec-es
      • 158 bim-a23thima2-phec-es
      • 159 bim-a24thima2-4pec-es
      • 160 bhs-a23thima2-7 cmc-es
      • 161 dimethpym-dibema2-phaz-es
      • 162 2py-me25thima2-3bec-es
      • 163 bhs-aof-thec-es
      • 164 gua-dibema2-phec-f2es
      • 165 amim-a23thima2-phec-es
      • 166 2py-bam2-pclphec-es
      • 167 bhs-pyma2-thec-es
      • 168 2py-a24thima2-3bec-es
      • 169 bim-bam2-phec-es
      • 170 bim-35thima2-7 cmc-es
      • 171 bhs-35thima2-pipmaz-es
      • 172 bim-prodia2-phec-es
      • 173 bim-35thima2-phec-es
      • 174 gua-edia3-4pec-es
      • 175 gua-a23thima2-ppec-es
      • 176 gua-pipeme2-phec-es
      • 177 gua-dibema2-phaz-es
      • 178 2py-bam2-3bec-es
      • 179 bhs-bam2-3bec-mals
      • 180 mam2py-apma2-7 cmc-es
      • 181 bhs-bam2-pmophec-es
      • 182 gua-bam2-7 cmc-es
      • 183 gua-buta-phec-es
      • 184 bim-pyma2-7 cmc-es
      • 185 2py-pipa2-thec-ms
      • 186 bhs-dibema2-dmaphec-es
      • 187 bim-a24thima2-ppec-es
      • 188 am2py-bam2-7 cmc-es
      • 189 bim-buta-7cmc-es
      • 190 im pipa2-phec-es
      • 191 gua-dibema2-4pec-gs
      • 192 2py-buta-thec-es
      • 193 2py-pipa2-7 cmc-es
      • 194 2py-apma2-phec-es
      • 195 bim-pipa2-phec-gs
      • 196 bim-me25thima2-phec-es
      • 191 2py-pyma2-3bec-es
      • 198 gua-bam2-pmophec-es
      • 199 gua-35thima2-4pec-es
      • 200 2py-pipeme2-thec-es
      • 201 bhs-35thima2-phaz-f2es
      • 202 bhs-edia3-phaz-es
      • 203 2py-apma2-thec-pms
      • 204 im apma2-phaz-es
      • 205 bim-chex2-phec-es
      • 206 bhs-35thima2-4pec-es
      • 207 gua-a23thima2-phaz-es
      • 208 2py-me25thima2-phaz-es
      • 209 2py-a23thima2-pmophec-es
      • 210 bhs-chex2-3bec-es
      • 211 2py-dibema2-3ipec-es
      • 212 2py-bam2-phec-es
      • 213 bhs-dibema2-phec-es
      • 214 bim-a24thima2-thec-es
      • 215 bim-pipa2-thec-es
      • 216 bhs-buta-phaz-es
      • 217 bhs-mepipe2-phaz-es
      • 218 gua-buta-4pec-es
      • 219 am2py-a23thima2-phaz-es
      • 220 gua-bam2-thec-es
      • 221 gua-pdagk-4pec-es
      • 222 bim-pdagk-phec-es
      • 223 2py-35thima2-phec-es
      • 224 gua-35thima2-7 cmc-es
      • 225 gua-bam2-3bec-es
      • 226 bhs-bam2-3bec-es
      • 227 gua-a23thima2-7 cmc-es
      • 228 bhs-aepi2-thec-es
      • 229 clim-pipa2-7 cmc-es
      • 230 2py-a23thima2-3bec-es
      • 231 bim-a23thima2-3bzlaz-es
      • 232 bhs-pipa2-3bec-es
      • 233 bim-pipa2-mmphec-es
      • 234 clim-dibema2-phec-es
      • 235 bhs-aepi2-3bec-es
      • 236 2py-apma2-4pec-es
      • 237 dhim-a23thima2-7 cmc-es
      • 238 bim-pipa2-pclphec-es
      • 239 gua-mepipe2-7 cmc-es
      • 240 gua-35thima2-3ipec-es
      • 241 bhs-chex2-thec-es
      • 242 bim-inda2-7 cmc-es
      • 243 bhs-pipa2-phaz-es
      • 244 imhs-pipa2-thec-es
      • 245gua-apma2-4phaz-es
      • 246 gua-me25thima2-4pec-es
      • 247 gua-35thima2-phec-es
      • 248 bim-pipa2-amaz-es
      • 249 2py-a24thima2-4phaz-es
      • 250 2py-me42thiaz2-3bec-es
      • 251 imhs-apma2-phec-es
      • 252 bhs-pipeme2-thec-es
      • 253 dhim-a24thima2-phec-es
      • 254 2py-a23thima2-7 cmc-es
      • 255 2py-pipa2-pymaz-es
      • 256 2py-me35thima2-3bec-es
      • 257 bim-apma2-7 cmc-as
      • 258 bhs-35thima2-amaz-es
      • 259 mam2py-dibema2-thec-es
      • 260 dimethpym-apma2-4pec-es
      • 261 bhs-bam2-4bec-es
      • 262 2py-a23thima2-cpec-es
      • 263 mam2py-35thima2-phec-es
      • 264 am2py-apma2-phec-es
      • 265 gua-a23thima2-4pec-es
      • 266 bim-a24thima2-phec-es
      • 267 2py-pipa2-thec-es
      • 268 2py-dibema2-thec-es
      • 269 pippy-pipa2-4pec-es
      • 270 bim-dibema2-7 cmc-es
      • 271 bim-dibema2-phec-es
      • 272 gua-pdagk-7 cmc-es
      • 273 bhs-35thima2-thec-es
      • 274 bhs-a23thima2-mmphec-es
      • 275 bhs-a23thima2-thec-nes
      • 276 bim-me25thima2-7 cmc-es
      • 277 2py-a24thima2-phec-es
      • 278 gua-barn2-dbc-es
      • 279 amim-dibema2-7 cmc-es
      • 280 2py-a23thima2-4pec-es
      • 281 thpym-dibema2-thec-es
      • 282 2py-pipa2-phec-es
      • 283 bhs-a24thima2-pymaz-es
      • 284 gua-dibema2-amaz-es
      • 285 dhim-bam2-3bec-es
      • 286 gua-bam2-7 cmc-ms
      • 287 bhs-edia3-thec-es
      • 288 bim-a24thima2-phec-mals
      • 289 bim-a24thima2-mophaz-es
      • 290 gua-dibema2-phec-es
      • 291 bhs-pipa2-4pec-es
      • 292 bhs-apma2-pipmaz-es
      • 293 gua-dibema2-pipmaz-es
      • 294 gua-aepi2-4pec-es
      • 295 gua-pipa2-ppec-es
      • 296 bim-mea2-7 cmc-es
      • 297 gua-pipa2-pmophec-es
      • 298 imhs-bam2-7 cmc-es
      • 299 gua-a24thima2-7 cmc-f2es
      • 300 thpym-a23thima2-3bec-es
      • 301 bim-mepipe2-7 cmc-es
      • 302 thpym-pipa2-phaz-es
      • 303 bim-aaf-7 cmc-es
      • 304 bim-edia3-phec-es
      • 305 2py-a24thima2-thec-es
      • 306 bim-pipa2-phaz-es
      • 307 dimethpym-bam2-phec-es
      • 308 bim-a24thima2-phaz-es
      • 309 bhs-bam2-phaz-pms
      • 310 2py-35thima2-3bec-es
      • 311 2py-35thima2-mophaz-es
      • 312 gua-apma2-phaz-es
      • 313 bim-apma2-phaz-es
      • 314 gua-35thima2-7 cmc-nes
      • 315 bhs-pipa2-phec-es
      • 316 bhs-mepipe2-3bec-es
      • 317 gua-pipa2-phaz-es
      • 318 2py-a23thima2-phec-es
      • 319 2py-pipa2-4pec-es
      • 320 gua-apma2-mmphec-es
      • 321 2py-apma2-7 cmc-es
      • 322 bhs-a24thima2-phec-es
      • 323 bhs-a23thima2-4pec-es
      • 324 bim-35thima2-phaz-es
      • 325 bim-pipeme2-7 cmc-es
      • 326 bhs-42thiaz2-3bec-es
      • 327 pippy-a23thima2-phec-es
      • 328 2py-aof-thec-es
      • 329 2py-pdagk-phaz-es
      • 330 gua-aepi2-7 cmc-es
      • 331 dimethpym-pipa2-3bec-es
      • 332 gua-35thima2-amec-es
      • 333-bhs-inda2-phaz-es
      • 334 2py-pipeme2-3bec-es
      • 335 gua-apma2-4pec-nes
      • 336 gua-edia2-4pec-es
      • 337 gua-a24thima2-phec-es
      • 338 gua-apma2-3bec-es
      • 339 gua-aaf-phec-es
      • 340 gua-apma2-thec-es
      • 341 bim-apma2-pymaz-es
      • 342 im a24thima2-phec-es
      • 343 2py-a24thima2-ames-es
      • 344 bim-pdagk-7 cmc-es
      • 345 bim-pipa2-3bzlaz-es
      • 346 2py-mea2-phaz-es
      • 347 amim-bam2-phaz-es
      • 348 2py-pipa2-3bec-es
      • 349 dhim-apma2-phaz-es
      • 350 2py-35thima2-4pec-es
      • 351 bhs-aof-3bec-es
      • 352 2py-dibema2-phaz-nes
      • 353 gua-a24thima2-3bec-es
      • 354 bhs-dibema2-pymaz-es
      • 335 bim-a24thima2-4bec-es
      • 356 bhs-bam2-4pec-es
      • 357 bim-35thima2-thec-es gua-penta-phec-es
      • 358 bim-buta-4pec-es
      • 359 bhs-apma2-amaz-es
      • 360 dimethpym-a24thima2-3bec-es
      • 361 gua-a2-3thima2-7 cmc-mals
      • 362 gua-dibema2-3bzlaz-es
      • 363 2py-edia2-3bec-es
      • 364 2py-aaf-thec-es
      • 364 gua-a24thima2-7 cmc-es
      • 365 2py-dibema2-mmphec-es
      • 366 hs-apma2-3bec-es
      • 357 im-dibema2-ppec-es
      • 368 ua-35thima2-phaz-es
      • 369 py-me42thiaz2-thec-es
      • 370 im-35thima2-dbc-es
      • 371 hs-prodia2-3bec-es
      • 372 ua-35thima2-mmphec-es
      • 373 hs-hexa-3bec-es
      • 374 hs-penta-phaz-es
      • 375 him-pipa2-phec-es
      • 376 ua-bam2-phec-es
      • 377 dhim-pipa2-phec-es
      • 378 gua-bam2-phec-es
      • 379 2py-apma2-phaz-mals
      • 380 bim-apma2-dbc-es
      • 381 gua-inda2-phec-es
      • 382 2py-bam2thec-es
      • 383 gua-pipa2-4bec-es
      • 384 am2py-35thima2-4pec-es
      • 383 bim-a24thima2-mpphec-es
      • 386 2py-35thima2-4bec-es
      • 387 bhs-pipa2-7 cmc-es
      • 388 amim-pipa2-4pec-es
      • 389 bhs-apma2-4pec-es
      • 390 gua-a23thima2-phec-pms
      • 391 bim-35thima2-4pec-es
      • 392 bhs-a24thima2-thec-es
      • 393 thpym-a24thima2-phaz-es
      • 394 bim-mea2-phec-es
      • 395 bim-a23thima2-thec-es
      • 396 pippy-apma2-thec-es
      • 397 2py-35thima2-ppec-es
      • 398 im a23thima2-7 cmc-es
      • 399 gua-mea2-4pec-es
      • 400 gua-edia2-7 cmc-es
      • 401 mam2py-pipa2-phaz-es
      • 402 bhs-apma2-3bec-f2es
      • 403 bim-aepi2-phec-es
      • 404 2py-aepi2-phaz-es
      • 405 2py-35thima2-thec-mals
      • 406 2py-bam2-phaz-es
      • 407 am2py-pipa2-thec-es
      • 408 bhs-bam2-ppec-es
      • 409 2py-dibema2-thec-ps
      • 410 gua-pipa2-7 cmc-es
      • 411 gua-bam2-4pec-as
      • 412 bhs-apma2-thec-es
      • 413 clim-35thima2-phaz-es
      • 414 2py-bam2-amaz-es
      • 415 bhs-pipa2-phaz-ps
      • 416 gua-bam2-phaz-es
      • 417 bhs-apma2-mmophec-es
      • 418 gua-a24 thima2-thec-es
      • 419 gua-chex2-7 cmc-es
      • 420 2py-penta-thec-es
      • 421 2py-edia2-phaz-es
      • 422 gua-pipa2-phec-es
      • 423 bim-chec2-4pec-es
      • 424 gua-dibema2-mmophec-es
      • 425 2py-35thima2-phaz-es
      • 426 bim-dibema2-mophaz-es
      • 427 bim-me42thiaz2-4pec-es
      • 428 2py-pyma2-phaz-es
      • 429 bhs-a24thima2-3bec-es
      • 430 2py-penta-phaz-es
      • 431 bim-dibema2-pmophec-es
      • 432 gua-pipa2-4pec-pms
      • 433 bim-a23thima2-mmophec-es
      • 434 2py-dibema2-phec-es
      • 435 gua-a24thima2-pipmaz-es
      • 436 bim-apma2-phec-es
      • 437 bhs-pipa2-mpphec-es
      • 438 gua-a23thima2-3bec-es
      • 439 bim-a23thima2-amaz-es
      • 440 bhs-dibema2-4pec-es
      • 441 imhs-35thima2-4pec-es
      • 442 imhs-a23thima2-phaz-es
      • 443 bim-bam2-phec-nes
      • 444 bhs-dibema2-3bec-es
      • 445 bhs-a24thima2-phaz-es
      • 446 gua-apma2-7 cmc-ps
      • 447 amim-apma2-thec-es
      • 448 bim-edia3-7 cmc-es
      • 449 gua-bam2-cpec-es
      • 450 gua-inda2-4pec-es
      • 451 gua-edia3-phec-es
      • 452 2py-pipa2-dbc-es
      • 453 2py-a24thima2-mmphec-es
      • 454 bim-pipa2-pipmaz-es
      • 455 2py-a23thima2-dmaphec-es
      • 456 bim-a23thima2-3bec-es
      • 457 2py-pdagk-3bec-es
      • 458 bim-dibema2-3bec-es
      • 459 bim-apma2-thec-es
      • 460 2py-bam2-4pec-es
      • 461 bhs-me35thima2-3bec-es
      • 462 gua-35thima2-3bec-es
      • 463 pippy-35thima2-3bec-es
      • 464 2py-bam2-3bec-gs
      • 465 2py-bam2-3bzlaz-es
      • 466 bhs-pipeme2-phaz-es
      • 467 bim-mepipe2-4pec-es
      • 468 bhs-dibema2-thec-as
      • 469 2py-apma2thec-es
      • 470 bim-35thima2-3bec-es
      • 471 bhs-mea2-phaz-es
      • 472 bim-prodia2-4pec-es
      • 473 bhs-mea2-phaz-es
      • 474 gua-a24thima2-mmophec-es
      • 475 gua-pipeme2-4pec-es
      • 476 bim-a23thima2-phaz-es
      • 477 gua-prodia2-phec-es
      • 478 gua-dilbema2-pclphec-es
      • 479 bhs-aaf-phaz-es
      • 480 2py-chex2-phaz-es
      • 481 bim-35thima2-dmaphec-es
      • 482 imhs-dibema2-3bec-es
      • 483 2py-bam2-thec-f2es
      • 484 bhs-35thima2-phec-es
      • 485 bim-a23thima2-7 cmc-es
      • 486 bhs-apma2-phec-es
      • 487 bim-apma2-pmophec-es
      • 488 bim-dibema2-7 cmc-pms
      • 489 gua-35thima2-thec-es
      • 490 bhs-pipa2-4phaz-es
      • 491 2py-dibema2-phaz-es
      • 492 bim-apma2-dmaphec-es
      • 493 bim-edia2-phec-es
      • 494 2py-dibema2-3bec-es
      • 495 bhs-35thima2-mmphec-es
      • 496 gua-apma2-phec-es
      • 497 bim-bam2-amec-es
      • 498 gua-apma2-amec-cs
      • 499 bhs-35thima2-7 cmc-es
      • 500 bhs-me25thima2-thec-es
      • 501 bhs-dibema2-7 cmc-es
      • 502 gua-hexa-4pec-es
      • 503 bim-bam2-3bec-es
      • 504 bhs-pipa2-3ipec-es
      • 505 2py-apma2-4bec-es
      • 506 dimethpym-35thima2-7 cmc-es
      • 507 bhs-bam2-phec-es
      • 508 bhs-dibema2-3bec-ms
      • 509 bhs-35thima2-3bzlaz-es
      • 510 gua-penta-7 cmc-es
      • 511 bhs-a23thima2-thec-es
      • 512 clim-a23thima2-4pec-es
      • 513 bhs-me42thiaz2-3bec-es
      • 514 bhs-35thima2-phaz-es
      • 515 bhs-a24thima2-4pec-es
      • 516 bhs-a23thima2-phaz-es
      • 517 bhs-bam2-thec-es
      • 518 2py-35thima2-mpphec-es
      • 519 bhs-dibema2-dbc-es
      • 520 2py-35thima2-3bec-pms
      • 521 2py-a24thima2-phaz-es
      • 522 gua-aaf-7 cmc-es
      • 523 gua-me42thiaz2-phec-es
      • 524 bim-a23thima2-ipmaz-es
      • 525 bim-a24thima2-7 cmc-es
      • 526 im-bam2-3bec-es
      • 527 bhs-a24thima2-cpec-es
      • 528 bim-bam2-phaz-es
      • 529 2py-apma2-mophaz-es
      • 530 bim-pipa2-7 cmc-[2es
      • 531 gua-a23thima2-mpphec-es
      • 532 2py-a23thima2-3bec-as
      • 533 gua-pyma2-4pec-es
      • 534 2py-pipa2-phaz-es
      • 535 2py-edia3-3bec-es
      • 536 mam2py-a23thima2-3bec-es
      • 537 2py-a24thima2-3ipec-es
      • 538 2py-aof-phaz-es
      • 539 gua-hexa-7 cmc-es
      • 540 bhs-a23thima2-3bec-ps
      • 541 bim-a24thima2-4pec-pms
      • 542 bim-aaf-4pec-es
      • 543 bhs-pipa2-thec-es
      • 544 pippy-dibema2-7 cmc-es
      • 545 gua-pipa2-thec-es
      • 546 bhs-bam2-7 cmc-es
      • 547 gua-bam2-4pec-es
      • 548 bim-aepi2-4pec-es
      • 549 2py-prodia2-phaz-es
      • 550 2py-a23thima2-phaz-es
      • 551 bim-35thima2-4pec-ms
      • 552 bim-dibema2-4pec-mals
      • 553 bhs-a24thima2-thec-ms
      • 554 bim-42thiaz2-phec-es
      • 555 2py-a24thima2-phaz-ps
      • 556 bim-aof-phec-es
      • 557 2py-a23thima2-pymaz-es
      • 558 gua-a23thima2-mophaz-es
      • 559 thpym-apma2-7 cmc-es
      • 560 bim-bam2-3ipec-es
      • 561 pippy-bam2-phaz-es
      • 562im-dibema2-4pec-es
  • In the above list, the following abbreviations are used for the structural units A, E, G and L.
  • A = Abbreviation
    Figure US20160296592A1-20161013-C00022
    2py
    Figure US20160296592A1-20161013-C00023
    dbim
    Figure US20160296592A1-20161013-C00024
    bim
    Figure US20160296592A1-20161013-C00025
    imhs
    Figure US20160296592A1-20161013-C00026
    dimethpym
    Figure US20160296592A1-20161013-C00027
    mam2py
    Figure US20160296592A1-20161013-C00028
    am2py
    Figure US20160296592A1-20161013-C00029
    thpym
    Figure US20160296592A1-20161013-C00030
    bhs
    Figure US20160296592A1-20161013-C00031
    gua
    Figure US20160296592A1-20161013-C00032
    amim
    Figure US20160296592A1-20161013-C00033
    clim
    Figure US20160296592A1-20161013-C00034
    im
    Figure US20160296592A1-20161013-C00035
    pippy
  • H = Abbreviation
    Figure US20160296592A1-20161013-C00036
    edia2
    Figure US20160296592A1-20161013-C00037
    pyma2
    Figure US20160296592A1-20161013-C00038
    pipa2
    Figure US20160296592A1-20161013-C00039
    aepi2
    Figure US20160296592A1-20161013-C00040
    me35thima2
    Figure US20160296592A1-20161013-C00041
    dibema2
    Figure US20160296592A1-20161013-C00042
    edia3
    Figure US20160296592A1-20161013-C00043
    buta
    Figure US20160296592A1-20161013-C00044
    aaf
    Figure US20160296592A1-20161013-C00045
    42thiaz2
    Figure US20160296592A1-20161013-C00046
    chex2
    Figure US20160296592A1-20161013-C00047
    bam2
    Figure US20160296592A1-20161013-C00048
    apma2
    Figure US20160296592A1-20161013-C00049
    pdagk
    Figure US20160296592A1-20161013-C00050
    mepipe2
    Figure US20160296592A1-20161013-C00051
    prodia2
    Figure US20160296592A1-20161013-C00052
    inda2
    Figure US20160296592A1-20161013-C00053
    35thima2
    Figure US20160296592A1-20161013-C00054
    me25thima2
    Figure US20160296592A1-20161013-C00055
    penta
    Figure US20160296592A1-20161013-C00056
    aof
    Figure US20160296592A1-20161013-C00057
    hexn
    Figure US20160296592A1-20161013-C00058
    mea2
    Figure US20160296592A1-20161013-C00059
    pipeme2
    Figure US20160296592A1-20161013-C00060
    me42thiaz2
    Figure US20160296592A1-20161013-C00061
    a23thima2
    Figure US20160296592A1-20161013-C00062
    a24thima2
  • The bond from structural element G to structural unit L should be understood as meaning a double bond in the compound where L=as.
  • G = Abbreviation
    Figure US20160296592A1-20161013-C00063
    4phaz
    Figure US20160296592A1-20161013-C00064
    3bzlaz
    Figure US20160296592A1-20161013-C00065
    mophaz
    Figure US20160296592A1-20161013-C00066
    pipmaz
    Figure US20160296592A1-20161013-C00067
    cpec
    Figure US20160296592A1-20161013-C00068
    mpphec
    Figure US20160296592A1-20161013-C00069
    amec
    Figure US20160296592A1-20161013-C00070
    dbc
    Figure US20160296592A1-20161013-C00071
    pclphec
    Figure US20160296592A1-20161013-C00072
    thec
    Figure US20160296592A1-20161013-C00073
    mmophec
    Figure US20160296592A1-20161013-C00074
    3bec
    Figure US20160296592A1-20161013-C00075
    phaz
    Figure US20160296592A1-20161013-C00076
    pymaz
    Figure US20160296592A1-20161013-C00077
    3ipec
    Figure US20160296592A1-20161013-C00078
    phec
    Figure US20160296592A1-20161013-C00079
    ppec
    Figure US20160296592A1-20161013-C00080
    mmphec
    Figure US20160296592A1-20161013-C00081
    amaz
    Figure US20160296592A1-20161013-C00082
    4bec
    Figure US20160296592A1-20161013-C00083
    dmaphec
    Figure US20160296592A1-20161013-C00084
    4pec
    Figure US20160296592A1-20161013-C00085
    pmophec
    Figure US20160296592A1-20161013-C00086
    7eme
  • L = Abbreviation
    Figure US20160296592A1-20161013-C00087
    es
    Figure US20160296592A1-20161013-C00088
    gs
    Figure US20160296592A1-20161013-C00089
    pms
    Figure US20160296592A1-20161013-C00090
    f2es
    Figure US20160296592A1-20161013-C00091
    mals
    Figure US20160296592A1-20161013-C00092
    ps
    Figure US20160296592A1-20161013-C00093
    ms
    Figure US20160296592A1-20161013-C00094
    nes
    Figure US20160296592A1-20161013-C00095
    as
  • The compounds of the formula I and the starting substances used for their preparation can generally be prepared by methods of organic chemistry known to the person skilled in the art, such as are described in standard works such as Houben-Weyl (ed.). “Methoden der Organischen Chemie”[Methods of Organic Chemistry], Thieme-Verlag, Stuttgart, Taylor (ed.), “The Chemistry of Heterocyclic Compounds”, Wiley & Sons, New York, or March “Advanced Organic Chemistry”, 4th Edition, Wiley & Sons. Further methods of preparing specific functional groups are also described in R. Larock, “Comprehensive Organic Transformations”, Weinheim 1989, in particular the preparation of alkenes, alkynes, halides, amines, ethers, alcohols, phenols, aldehydes, ketones, nitrites, carboxylic acids, esters, amides and acid chlorides.
  • Inhibitors of Integrin αvβ6
  • The invention relates to novel integrin inhibitors of the formula I
  • Figure US20160296592A1-20161013-C00096
  • in which
      • R1, R1′ and R1″ are H, A, Ar, Het′, Hal, NO2, CN, OR4, COA, NHCOA, NH(CHO), NR4, COOR4 or CONHR4 2,
      • R2 is A, Ar, (CH2)mXA, (CH2)mOH, (CH2)mNH2, (CH2)mNHA, (CH2)mNA2, (CH2)mNHCOA, (CH2) NO2, (CH2) COOR1, (CH2)mCONH2, (CH2)mX(CH2)oAr, (CH2)mX(CH2)oCHAr2, (CH2)mX(CH2)oCAr3, (CH2)mXCOYA, (CH2)mXCOY(CH2)oAr, (CH2)mX (CH2)Het1, (CH2)mX (CH2)CHHet1 2, (CH2)mX(CH2)oCHet1 3, (CH2)mX(CH2)oYA, (CH2)mX (CH2)oNHCOA, (CH2)mNHCONHR2, (CH2)mCH2A,| (CH2)mCHA2, (CH2)mCA3, (CH2)mAr, (CH2)mCHAr2, (CH2)mCAr3, (CH2)mHet1, (CH2)mCHHet1 2, (CH2)mChet1 3, (CH2)mcycloalkyl, (CH2)m-NH-C (═NH)-NH2 or (CH2)m-(HN═) C-NH2,
      • where X and Y, independently of one another, may be S, O, S═O, SO2 or NH, where, if R2=(CH2)mXSCOYA or (CH2)mXCOY (CH2)oAr, X and Y cannot be S═O or SO2,
      • R2′ is H or A,
      • R2 and R2′ together may alternatively be —(CH2)p—,0
      • R3 is H2N—C (═NH)—, H2N—C (═NH)—NH—, A—C (═NH)—NH—, Het2— or Het2—NH—, where the primary amino groups may also be provided with conventional amino-protecting groups,
      • R4 is H, A, Het1, Hal, NO2 or CN,
      • A is alkyl having from 1 to 8 carbon atoms,
      • Ar is phenyl, naphthyl, anthranyl or biphenyl, each of which is unsubstituted or monosubstituted or polysubstituted by Hal, A, OA, OH, CO—A CN, COOA, COOH, CONH2, CONHA, CONA2, CF3, OCF3 or NO2,
      • Het1 is an aromatic monocyclic or bicyclic heterocyclic radical having from 1 to 3 N, O and/or S atoms, with may be unsubstitutod or monosubstituted or disubstituted by F, Cl, Br, A, OA, SA, OCF3,—CO—A, CN, COOA, CONH2, CONHA, CONA2, NA2 or NO2,
      • Het2 is a monocyclic or bicyclic heterocyclic radical having from 1 to 4 N atoms, which may be unsubstituted or mono substituted or disubstituted by NH2 or NHA,
      • m is 0, 1,2,3,4,5,6 or 8,
      • n is 1, 2,3,4,5 or 6,
      • o is 0, 1, 2 or 3,
      • p is 2, 3,4 or 5,
        to their stereoisomers, and to their physiologically acceptable salts and solvates.
  • The invention relates to novel peptides, of the formula I

  • R1-Arg-X-Asp-Leu-Asp-Ser-Leu-Arg-R2  I
  • in which
      • R1 denotes H, acetyl or acyl and
      • R2 denotes —OH, OR3 NH2,NHR3, N(R3)2
      • R3 denotes alkyl, aralkyl, aryl, Het and
      • X denotes an amino acid of the formula II
  • Figure US20160296592A1-20161013-C00097
  • in which
      • A denotes (CH2)n
      • R4 denotes H, alkyl, aralkyl or aryl, and
      • n denotes 1, 2, 3, 4, 5 or 6,
      • and the amino acid of the formula II is bonded to the adjacent Arg via a peptide bond of the α-amino group and to the α-amino group of the adjacent Asp via a peptide bond of the α-carboxyl group.
  • The present invention also relates to the pharmaceutically usable pro-drugs, derivatives, solvates and stereoisomors of the compounds of the formula I and the salts thereof, including mixtures thereof in all ratios.
  • The invention relates, in particular, to peptidic compounds selected from the group of the formulae Ia)-I o)
      • I a) Ac-Arg-Dap(Psa)-Asp-Leu-Asp-Ser-Leu-Arg-NH2,
      • I b) Ac-Arg-Dap(F5-PSA)-Asp-Leu-Asp-Ser-Leu-Arg-NH2,
      • I c) Ac-Arg-Dap(2-NO2-PSA)-Asp-Leu-Asp-Ser-Leu-Arg-NH2,
      • I d) Ac-Arg-Dap(4-NO2-PSA)-Asp-Leu-Asp-Ser-Leu-Arg-NH2,
      • I e) Ac-Arg-Dap(2,4-NO2-PSA)-Asp-Leu-Asp-Ser-Leu-Arg-NH2,
      • I f) Ac-Arg-Dap(6-OMe-PSA)-Asp-Leu-Asp-Ser-Leu-Arg-NH2,
      • I g) Ac-Arg-Dap(2-CF3-PSA)-Asp-Leu-Asp-Ser-Leu-Arg-NH2,
      • I h) Ae-Arg-Dap(3-CF3-PSA.)-Asp-Leu-Asp-Ser-Lt.-m-Arg-NH2,
      • I a) Ac-Arg-Dap(Me5-PSA)-Asp-Leu-Asp-Ser-Leu-Arg-NH2,
      • I j) Ac-Arg-Dap(4-tBu-PSA)-Asp-Leu-Asp-Ser-Leu-Arg-N H2,
      • I k) Ac-Arg-Dap(BSA)-Asp-Leu-Asp-Ser-Leu-Arg-NH2,
      • I l) Ac-Arg-Dap(iPrs)-Asp-Leu-Asp-Ser-Leu-Arg-NH2,
      • I m) Ac-Arg-Dap(1-Nap)-Asp-Leu-Asp-Ser-Leu-Arg-NH2,
      • I n) Ac-Arg-Dap(2-Nap)-Asp-Leu-Asp-Ser-Leu-Arg-NH2,
      • I o) Ac-Arg-Dap(4-Ph-Psa)-Asp-Leu-Asp-Ser-Leu-Arg-NH2,
      • and pharmaceutically usable prodrugs, derivatives, solvates and stereoisomers thereof, and salts thereof, including mixtures thereof in all ratios, where the abbreviations used in brackets in the formulae, I a)-I o) stand for the following radicals:
  • Psa phenylsulfonyl radical
    F5-PSA pentafluorophenylsulfonyl radical
    2-NO2-PSA 2-nitrophenylsulfonyl radical
    4-NO2-PSA 4-nitrophenylsulfonyl radical
    2,4-NO2-PSA 2,4-dinitrophenylsulfonyl radical
    6-OMe-PSA 6-methoxyphenylsulfonyl radical
    2-CF3-PSA 2-trifluoromethylphenylsulfonyl radical
    3-CF3-PSA 3-trifluoromethylphenylsulfonyl radical
    Me5-PSA pentamethylphenylsulfonyl radical
    4-tBu-PSA 4-tert-butylphenylsulfonyl radical
    Bsa benzylsulfonyl radical
    iPrs isopropylsulfonyl radical
    1-Nap 1-naphthylsulfonyl radical
    2-Nap 2-naphthylsulfonyl radical
    4-Ph-Psa 4-phenylphenylsulfonyl radical
  • The abbreviations mentioned above and below for amino acid radicals stand for the radicals of the following amino acids:
  • Asp or D aspartic acid
    Arg or R arginine
    Dap
    2,3-diaminopropionic acid
    Leu or L leucine
    Ser or S serine
    Thr or T threonine
  • Furthermore, the following abbreviations above and below have the following meanings:
  • Ac acetyl
    BOC tert-butoxycarbonyl
    BSA bovine serum albumin
    CBZ or Z benzyloxycarbonyl
    DCCI dicyclohexylcarbodiimide
    DCM dichloromethane
    DIEA diisopropylamine
    DMA N,N-dimethylacetamide
    DMF dimethylformamide
    EDCI N-ethyl-N,N′-(dimethylaminopropyl)carbodiimide
    Et ethyl
    FCA fluoresceincarboxylic acid
    FITC fluorescein isothiocyanate
    Fmoc 9-fluorenylmethoxycarbonyl
    Fmoc-Dap(ivDde) N-α-Fmoc-N-γ-1-(4,4-dimethyl-2,6-
    dioxocyclohex-1-yli-dene)-3-
    methylbutyldiaminopropionic acid
    FTH fluoresceinthiourea
    HOBt 1-hydroxybenzotriazole
    Me methyl
    MBHA 4-methylbenzhydrylamine
    Mtr 4-methoxy-2,3,6-trimethylphenylsulfonyl
    NMP N-methylpyrrolidone
    HONSu N-hydroxysuccinimide
    OBut tent-butyl ester
    Oct octanoyl
    OMe methyl ester
    OEt ethyl ester
    Pbf
    2,2,4,6,7-pentamethyldihydrobenzofuran-5- sulfonyl
    Pmc
    2,2,5,7,8-pentamethylchroman-6-sulfonyl
    POA phenoxyacetyl
    Sal salicyloyl
    TBS++ Tris buffered saline with divalent cations
    TBSA TBS + BSA
    TBTU 2-(1H-benzotriazol-1-yl)-1,1,3-tetramethyluronium
    tetrafluoroborate
    TFA trifluoroacetic acid
    TIS triisopropylamine
    Trt trityl(triphenylmethyl).
  • If the above-mentioned amino acids can occur in a plurality of enantiomeric forms, all these forms and also mixtures thereof (for example the DL forms) are included above and below. Furthermore, the amino acids may be provided with corresponding protecting groups known per se.
  • As evidenced by the continuing research in integrin antagonists and by the shortcomings of the compounds and methods of the art, there still remains a need for small-molecule, non-peptidic selective αvβ3 and/or αvβ5 antagonists that display decreased side-effects, improved potency, pharmacodynamic and pharmacokinetic properties, such as oral bioavailability and duration of action, over already described compounds. Such compounds would prove to be useful for the treatment, prevention, or suppression of various pathologies enumerated above that are mediated by αvβ3 and/or αvβ5 receptor binding and cell adhesion and activation.
  • In one embodiment, the present invention comprises a class of biphenyl integrin antagonists.
  • The present invention relates to a class of compounds represented by the Formula I:
  • Figure US20160296592A1-20161013-C00098
  • or a pharmaceutically acceptable salt, ester or tautomer thereof, wherein:
      • A and B are phenyl;
      • n is an integer from 1 to 3;
      • X1 is selected from O, NR, S, SO, SO2, CHR, and CH2, wherein:
        • R is selected from the group consisting of hydrogen, aryl, and heterocyclyl;
      • X2 is selected from the group consisting of alkyl, alkylamino, aminoalkyl, alkylaminoalkyl, alklthio, thioalkyl, alkylthioalkyl, alkylsulfonyl, sulfonylalkyl, alkylsulfonylalkyl, oxyalkyl, alkoxyalkyl, and alkoxy;
      • X3 is C1-C6 alkyl;
      • R1 is selected from the group consisting of pyridinyl and napthhyridinyl, wherein:
        • either is optionally substituted with a substituent selected from the group consisting of hydrogen, alkyl, halo, and amino;
      • R2, R3, R4, R5, R6, R7, R8 and R9 are independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkoxy, alkoxyalkyl, alkoxycarbonylalkyl, alkylamino, alkylcarbonyl, alkylheteroaryl, alkylsulfonylalkyl, alkylthio, alkynyl, aminocarbonylalkyl, cyano, dialkylamino, halo, haloalkoxy, haloalkyl, hydroxy and hydroxyalkyl; and
      • X3 is independently meta- or para- to the X1 of the B ring, and wherein further X3 is ortho-, meta-, or para- to the carboxylic acid chain of the A ring.
  • In another embodiment, the invention comprises pharmaceutical compositions comprising compounds of the Formula I. Such compounds and compositions are use in selectively inhibiting or antagonizing the αvβ3 and/or αbβs integrins and therefore in another embodiment the present invention relates to a method of selectively inhibiting or antagonizing the αvβ3 and/or αvβs integrin.
  • In another embodiment the invention provides methods of treating or inhibiting pathological conditions associated therewith such as osteoporosis, humoral hypercalcemia of malignancy, Paget's disease, rumor metastasis, solid tumor growth (neoplasia), angiogenesis, including tumor angiogenesis, retinopathy including macular degeneration and diabetic retinopathy, arthritis including rheumatoid arthritis and osteoarthritis, periodontal disease, psoriasis, smooth muscle cell migration and restenosis in a mammal in need of such treatment. Additionally, such pharmaceutical agents are useful as antiviral agents and antimicrobials. The compounds of the present invention may be used alone or in combination with other pharmaceutical agents.
  • The compounds of this invention are 1) αvβ3 integrin antagonists; or 2) αvβs integrin antagonists; or 3) mixed or dual αvβ3vβs antagonists. The present invention includes compounds which inhibit the respective integrins and also includes pharmaceutical compositions comprising such compounds. The present invention further provides for methods for treating or preventing conditions mediated by the αvβ3 and/or αvβs receptors in a mammal in need of such treatment comprising administering a therapeutically effective amount of the compounds of the present invention and pharmaceutical compositions of the present invention. Administration of such compounds and compositions of the present invention inhibits angiogenesis, tumor metastasis, tumor growth, osteoporosis, Paget's disease, humoral hypercalcemia of malignancy, retinopathy, macular degeneration, arthritis, periodontal disease, smooth muscle cell migration, including restenosis and artherosclerosis, and viral diseases.
  • The compounds of the present invention further show greater selectivity for the αvβ3 and/or αvβs integrin than for the αvβ6 integrin. It has been found that the selective antagonism of the αvβ3 integrin is desirable in that the αvβ6 integrin may play a role in normal physiological processes of tissue repair and cellular turnover that routinely occur in the skin and pulmonary tissue, and the inhibition of this function can be deleterious. Therefore, compounds of the present invention which selectively inhibit the αvβ3 integrin as opposed to the αvβ6 integrin have reduced side-effects associated with inhibition of the αvβ6 integrin.
  • The present invention relates to a class of compounds represented by the Formula I.
  • Figure US20160296592A1-20161013-C00099
  • or a pharmaceutically acceptable salts thereof wherein
      • X is
  • Figure US20160296592A1-20161013-C00100
      • Y is selected from the group consisting of N—R1, O, and S;
      • A is N or C;
      • R1 is selected from the group consisting of H, alkyl, aryl, hydroxy, alkoxy, cyano, nitro, amino, alkenyl, alkynyl, amido, alkylcarbonyl, arylcarbonyl, alkoxycarbonyl, aryloxycarbonyl, haloalkylcarbonyl, haloalkoxycarbonyl, alkylthiocarbonyl, arylthiocarbonyl, acyloxymethoxycarbonyl, alkyl optionally substituted with one or more substituent selected from lower alkyl, halogen, hydroxyl, haloalkyl, cyano, nitro, carboxyl, amino, alkoxy, aryl or aryl optionally substituted with one or more halogen, haloalkyl, lower alkyl, alkoxy, cyano, alkylsulfonyl, alkylthio, nitro, carboxyl, amino, hydroxyl, sulfonic acid, sulfonamide, aryl, fused aryl, monocycle heterocycles, or fused monocyclic heterocycles, aryl optionally substituted with one or more substituent selected from halogen, haloalkyl, hydroxy, lower alkyl, alkoxy, methylenedioxy, ethylenedioxy, cyano, nitro, alkylthio, alkylsulfonyl, sulfonic acid, sulfonamide, carboxyl derivatives, amino, aryl, fused aryl, monocyclic heterocycles and fused monocyclic heterocycle, monocyclic heterocycles, and monocyclic heterocycles optionally substituted with one or more substituent selected from halogen, haloalkyl, lower alkyl, alkoxy, amino, nitro, hydroxy, carboxyl derivatives, cyano, alkylthio, alkylsulfonyl, sulfonic acid, sulfonamide, aryl or fused aryl; or
      • R1 taken together with R8 forms a 4-12 membered dinitrogen containing heterocycle optionally substituted with one or more substituent selected from the group consisting of lower hydroxy, keto, alkoxy, halo, phenyl, amino, carboxyl or carboxyl ester, and fused phenyl; or
      • R1 taken together with R8 forms a 5 membered heteroaromatic ring optionally substituted with one or more substituent selected from lower alkyl, phenyl and hydroxy; or
      • R1 taken together with R8 forms a 5 membered heteroaromatic ring fused with a phenyl group;
      • R8 (when not taken together with R1) and R9 are independently selected from the group consisting of H, alkyl, alkenyl, alkynyl, aralkyl, amino, alkylamino, hydroxy, alkoxy, arylamino, amido, alkylcarbonyl, arylcarbonyl, alkoxycarbonyl, aryloxy, aryloxycarbonyl, haloalkylcarbonyl, haloalkoxy-carbonyl, alkylthiocarbonyl, arylthiocarbonyl, acyloxymethoxycarbonyl, cycloalkyl, bicycloalkyl, aryl, acyl, benzoyl, alkyl optionally substituted with one or more substituent selected from lower alkyl, halogen, hydroxy, haloalkyl, cyano, nitro, carboxyl derivatives, amino, alkoxy, thio, sulfonyl, aryl, aralkyl, aryl optionally substituted with one or more substituent selected from halogen, haloalkyl, lower alkyl, alkoxy, methylenedioxy, ethylenedioxy, alkylthio, haloalkylthio, thio, hydroxy, cyano, nitro, carboxyl derivatives, aryloxy, amido, acylamino, amino, alkylamino, dialkylamino, trifluoroalkoxy, trifluoromethyl, sulfonyl, alkylsulfonyl, haloalkylulfonyl, sulfonic acid, sulfonamide, aryl, fused aryl, monocyclic heterocycles, fused monocyclic heterocycles, aryl optionally substituted with one or more substituent selected from halogen, haloalkyl, lower alkyl, alkoxy, methylenedioxy, ethylenedioxy, alkylthio, haloalkylthio, thio, hydroxy, cyano, nitro, carboxyl derivatives, aryloxy, amido, acylamino, amino, alkylamino, dialkylamino, trifluoroalkoxy, trifluoromethylsulfonyl, alkylsulfonyl, sulfonic acid, sulfonamide, aryl, fused aryl, monocyclic heterocycles, or fused monocyclic heterocycles, monocyclic heterocycles, monocyclic heterocycles optionally substituted with one or more substituent selected from halogen, haloalkyl, lower alkyl, alkoxy, aryloxy, amino, nitro, hydroxy, carboxyl derivatives, cyano, alkylthio, alkylsulfonyl, aryl, fused aryl, monocyclic and bicyclic heterocyclicalkyls, —SO2R10 wherein R10 is selected from the group consisting of alkyl, aryl and monocyclic heterocycles, all optionally substituted with one or more substituent selected from the group consisting of halogen, haloalkyl, alkyl, alkoxy, cyano, nitro, amino, acylamino, trifluoroalkyl, amido, alkylaminosulfonyl, alkylsulfonyl, alkylsulfonylamino, alkylamino, dialkylamino, trifluoromethylthio, trifluoroalkoxy, trifluoromethylsulfonyl, aryl, aryloxy, thio, alkylthio, and monocyclic heterocycles; and
  • Figure US20160296592A1-20161013-C00101
      • wherein R10 is defined as above; or
      • NR8 and R9 taken together form a 4-12 membered mononitrogen containing monocyclic or bicyclic ring optionally substituted with one or more substituent selected from lower alkyl, carboxyl derivatives, aryl or hydroxy and wherein said ring optionally contains a heteroatom selected from the group consisting of O, N and S; or
      • X is
  • Figure US20160296592A1-20161013-C00102
      • wherein Y′ is selected from the group consisting of alkyl, cycloalkyl, bicycloalkyl, aryl, monocyclic heterocycles, alkyl optionally substituted with aryl which can also be optionally substituted with one or more substituent selected from halogen, haloalkyl, alkyl, nitro, hydroxy, alkoxy, aryloxy, aryl, or fused aryl, aryl optionally substituted with one or more substituent selected from halogen, haloalkyl, hydroxy, alkoxy, aryloxy, aryl, fused aryl, nitro, methylenedioxy, ethylenedioxy, or alkyl, alkynyl, alkenyl, —S—R11 and —OR11 wherein R11 is selected from the group consisting of H, alkyl, aralkyl, aryl, alkenyl, and alkynyl, or R11 taken together with R8 forms a 4-12 membered mononitrogen and monosulfur or monooxygen containing heterocyclic ring optionally substituted with lower alkyl, hydroxy, keto, phenyl, carboxyl or carboxyl ester, and fused phenyl, or R11 taken together with R8 is thiazole, oxazole, benzoxazole, or benzothiazole;
      • R8 is defined as above; or
      • Y1 (when Y1 is carbon) taken together with R6 forms a 4-12 membered mononitrogen or dinitrogen containing ring optionally substituted with alkyl, aryl, keto or hydroxy; or
      • X is
  • Figure US20160296592A1-20161013-C00103
      • wherein R1 and R8 taken together form a 5-8 membered dinitrogen containing heterocycle optionally substituted with one or more substituent selected from the group consisting of lower alkyl, hydroxy, keto, phenyl, or carboxyl derivatives; and R9 is selected from the group consisting of alkylcarbonyl, arylcarbonyl, alkoxycarbonyl, aryloxycarbonyl, haloalkyl-carbonyl, haloalkoxycarbonyl, alkylthiocarbonyl, arylthiocarbonyl, or acyloxymethoxycarbonyl; or
      • X is
  • Figure US20160296592A1-20161013-C00104
      • wherein R1 and R8 taken together form a 5-8 membered dinitrogen containing heterocycle optionally substituted with hydroxy, keto, phenyl, or alkyl; and
      • R9 are both selected from the group consisting of alkylcarbonyl, arylcarbonyl, alkoxycarbonyl, aryloxycarbonyl, haloalkylcarbonyl, haloalkoxycarbonyl, alkylthiocarbonyl, arylthiocarbonyl and acyloxymethoxycarbonyl;
      • R2, R3 nd R4 are independently selected from one or more substituent selected from the group consisting of H, alkyl, hydroxy, alkoxy, aryloxy, halogen, haloalkyl, haloalkoxy, nitro, amino, alkylamino, acylamino, dialkylamino, cyano, alkylthio, alkylsulfonyl, carboxyl derivatives, trihaloacetamide, acetamide, aryl, fused aryl, cycloalkyl, thio, monocyclic heterocycles, fused monocyclic heterocycles, and X, wherein X is defined as above; R5, R6 and R7 are independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, aryl, carboxyl derivatives, haloalkyl, cycloalkyl, monocyclic heterocycles, monocyclic heterocycles optionally substituted with alkyl, halogen, haloalkyl, cyano, hydroxy, aryl, fused aryl, nitro, alkoxy, aryloxy, alkylsulfonyl, arylsulfonyl, sulfonamide, thio, alkylthio, carboxyl derivatives, amino, amido, alkyl optionally substituted with one or more of halo, haloalkyl, hydroxy, alkoxy, aryloxy, thio, alkylthio, alkynyl, alkenyl, alkyl, arylthio, alkylssulfoxide, alkylsulfonyl, arylsulfoxide, arylsulfonyl, cyano, nitro, amino, alkylamino, dialkylamino, alkylsulfonamide, arylsulfonamide, acylamide, carboxyl derivatives, sulfonamide, sulfonic acid, phosphonic acid derivatives, phosphinic acid derivatives, aryl, arylthio, arylsulfoxide, or arylsulfone all optionally substituted on the aryl ring with halo alkyl, haloalkyl, cyano, nitro, hydroxy, carboxyl derivatives, alkoxy, aryloxy, amino, alkylamino, dialkylamino, amido, aryl, fused aryl, monocyclic heterocycles, and fused monocyclic heterocycles, monocyclic heterocyclicthio, monocyclic heterocyclicsulfoxide, and monocyclic heterocyclic sulfone, which can be optionally substituted with halo, haloalkyl, nitro, hydroxy, alkoxy, fused aryl, or alkyl, alkylcarbonyl, haloalkylcarbonyl, and arylcarbonyl, aryl optionally substituted in one or more positions with halo, haloalkyl, alkyl, alkoxy, aryloxy, methylenedioxy, ethylenedioxy, alkylthio, haloalkylthio, thio, hydroxy, cyano, nitro, acyloxy, carboxyl derivatives, carboxyalkoxy, amido, acylamino, amino, alkylamino, dialkylamino, trifluoroalkoxy, trifluoromethylsulfonyl, alkylsulfonyl, sulfonic acid, sulfonamide, aryl, fused aryl, monocyclic heterocycles and fused monocyclic heterocycles.
  • The compounds according to Formula I can exist in various isomers, enantiomers, tautomers, racemates and polymorphs, and all such forms are meant to be included.
  • It is another object of the invention to provide pharmaceutical compositions comprising compounds of the Formula I. Such compounds and compositions are useful in selectively inhibiting or antagoniing the αvβ3 and/or αvβ3 integrins and therefore in another embodiment the present invention relates to a method of selectively inhibiting or antagonizing the αvβ3 and/or αvβ5 integrin. The invention further involves treating or inhibiting pathological conditions associated therewith such as osteoporosis, humoral hypercalcemia of malignancy, Paget's disease, tumor metastasis, solid tumor growth (neoplasia), angiogenesis, including tumor angiogenesis, retinopathy including macular degeneration and diabetic retinopathy, arthritis, including rheumatoid arthritis, periodontal disease, psoriasis, smooth muscle cell migration and restenosis in a mammal in need of such treatment. Additionally, such pharmaceutical agents are useful as to antiviral agents, and antimicrobials.
  • The compounds of this invention are 1) αvβ3 integrin antagonists; or 2) αvβ5 integrin antagonists; or 3) mixed or dual αvβ3vβ5 antagonists. The present invention includes compounds which inhibit the respective integrins and also includes pharmaceutical compositions comprising such compounds. The present invention further provides for methods for treating or preventing conditions mediated by the αvβ3 and/or αvβ5 receptors in a mammal in need of such treatment comprising administering a therapeutically effective amount of the compounds of the present invention and pharmaceutical compositions of the present invention. Administration of such compounds and compositions of the present invention inhibits angiogenesis, tumor metastasis, tumor growth, osteoporosis, Paget's disease, humoral hypercalcemia of malignancy, retinopathy, macular degeneration, arthritis, periodontal disease, smooth muscle cell migration, including restenosis and artherosclerosis, and viral diseases.
  • The compounds of the present invention further show greater selectivity for the αvβ3 and/or αvβ5 integrin than for the αvβ6 integrin, it has been found that the selective antagonism of the αvβ3 integrin is desirable in that the αvβ6 integrin may play a role in normal physiological processes of tissue repair and cellular turnover that routinely occur in the skin and pulmonary tissue, and the inhibition of this function can be deleterious. Therefore, compounds of the present invention which selectively inhibit the αvβ3 integrin as opposed to the αvβ6 integrin have reduced side-effects associated with inhibtion of the αvβ6 integrin.
  • The present invention relates to a class of compounds represented by the Formula I.
  • Figure US20160296592A1-20161013-C00105
      • or a pharmaceutically acceptable salts thereof wherein
      • X is
  • Figure US20160296592A1-20161013-C00106
      • Y is selected from the group consisting of N—R1, O, and S;
      • y and z are independently selected from an integer selected form 0, 1, 2 and 3;
      • A is N or C;
      • R1 is selected from the group consisting of H, alkyl, aryl, hydroxy, alkoxy, cyano, nitro, amino, alkenyl, alkynyl, amido, alkylcarbonyl, arylcarbonyl, alkoxycarbonyl, aryloxycarbonyl holaalkylcarbonyl, haloalkoxycarbonyl, alkylthiocarbonyl, arylthiocarbonyl, acyloxymethoxycarboyl, alkyl optionally substituted with one or more substituent selected from lower alkyl, halogen, hydroxyl, haloalkyl, cyano, nitro, carboxyl, amino, alkoxy, aryl or aryl optionally substituted with one or more halogen, haloalkyl, lower alkyl, alkoxy, cyano, alkylsulfonyl, alkylthio, nitro, carboxyl, amino, hydroxyl, sulfonic acid, sulfonamide, aryl, fused aryl, monocyclic heterocycles, or fused monocyclic heterocycles, aryl optionally substituted with one or more substituent selected from halogen, haloalkyl, hydroxy, lower alkyl, alkoxy, methylenedioxy, ethylenedioxy, cyano, nitro, alkylthio, alkylsufonyl, sulfonic acid, sulfonamide, carboxyl derivatives, amino, aryl, fused aryl, monocyclic heterocycles and fused monocyclic heterocycle, monocyclic heterocycles, and monocyclic heterocycles optionally substituted with one or more substituent selected from halogen, haloalkyl, lower alkyl, alkoxy, amino, nitro, hydroxy, carboxyl derivatives, cyano, alkylthio, alkylsulfonyl, sulfonic acid, sulfonamide, aryl or fused aryl; or
      • R1 taken together with R8 forms a 4-12 membered dinitrogen containing heterocycle optionally substituted with one or more substituent selected from the group consisting of lower alkyl, hydroxy, keto, alkoxy, halo, phenyl, amino, carboxyl or carboxyl ester, and fused phenyl; or
      • R1 taken together with R8 forms a 5 membered heteroaromatic ring optionally substituted with one or more substituent selected from lower alkyl, phenyl and hydroxy; or
      • R1 taken together with R8 forms a 5 membered heteroaromatic ring fused with a phenyl group;
      • R8 (when not taken together with R1) and R9 are independently selected from the group consisting of H, alkyl, alkenyl, alkynyl, aralkyl, amino, alkylamino, hydroxy, alkoxy, arylamino, amido, alkylcarbonyl, arylcarbonyl, alkoxycarbonyl, aryloxy, aryloxycarbonyl, haloalkylcarbonyl, haloalkoxycarbonyl, alkylthiocarbonyl, arylthiocarbonyl, acyloxymethoxycarbonyl, cycloalkyl, bicycloalkyl, aryl, acyl, benzoyl, alkyl optionally substituted with one or more substituent selected from lower alkyl, halogen, hydroxy, haloalkyl, cyano, nitro, carboxyl derivatives, amino, alkoxy, thio, alkylthio, sulfonyl, aryl, aralkyl, aryl optionally substituted with one or more substituent selected from halogen, haloalkyl, lower alkyl, alkoxy, methylenedioxy, ethylenedioxy, alkylthio, haloalkylthio, thio, hydroxy, cyano, nitro, carboxyl derivatives, aryloxy, amido, acylamino, amino, alkylamino, dialkylamino, trifluoroalkoxy, trifluoromethyl, sulfonyl, alkylsulfonyl, haloalkylsulfonyl, sulfonic acid, sulfonamide, aryl, fused aryl, monocyclic heterocycles, fused monocyclic heterocycles, aryl optionally substituted with one or more substituent selected from halogen, haloalkyl, lower alkyl, alkoxy, methylenedioxy, ethylenedioxy, alkylthio, haloalkylthio, thio, hydroxy, cyano, nitro, carboxyl derivatives, aryloxy, amido, acylamino, amino, alkylamino, dialkylamino, trifluoroalkoxy, trifluoromethylsulfonyl, alkylsulfonyl, sulfonic acid, sulfonamide, aryl, fused aryl, monoyclic heterocycles, or fused monocyclic heterocycles, monocyclic heterocycles, monocyclic heterocycles optionally substituted with one or more substituent selected from halogen, haloalkyl, lower alkyl, alkoxy, aryloxy, amino, nitro, hydroxy, carboxyl derivatives, cyano, alkylthio, alkylsulfonyl, aryl, fused aryl, monocyclic and bicyclic heterocyclicalkyls, —SO2R10 wherein R10 is selected from the group consisting of alkyl, aryl and monocyclic heterocycles, all optionally substituted with one or more substituent selected faun the group consisting of halogen, haloalkyl, alkyl, alkoxy, cyano, nitro, amino, acylamino, trifluoroalkyl, amido, alkylaminosulfonyl, alkylsulfonyl, alkylsulfonylamino, alkylamino, dialkylamino, trifluoromethylthio, Trifluoroalkoxy, trifluoromethylsulfonyl, aryl, aryloxy, thio, and monocyclic Heterocycles; and
  • Figure US20160296592A1-20161013-C00107
      • wherein R10 is defined as above; or
      • NR8 and R9 taken together form a 4-12 membered mononitrogen containing monocyclic or bicyclic ring optionally substituted with one or more substituent selected from lower alkyl, carboxyl derivatives, aryl or hydroxy and wherein said ring optionally contains a heteroatom selected from the group consisting of O, N and S; or
      • X is
  • Figure US20160296592A1-20161013-C00108
      • wherein Y′ is selected from the group consisting of alkyl, cycloalkyl, bicycloalkyl, aryl, monocyclic heterocycles, alkyl optionally substituted with aryl which can also be optionally substituted with one or more substituent selected from halo, haloalkyl, alkyl, nitro, hydroxy, alkoxy, aryloxy, aryl, or fused aryl, aryl optionally substituted with one or more substituent selected from halo, haloalkyl, hydroxy, alkoxy, aryloxy, aryl, fused aryl, nitro, methylenedioxy, ethylenedioxy, or alkyl, alkynyl, alkenyl, —SR—R11 and —OR11 wherein R11 is selected from the group consisting of H, aralkyl, aryl, alkenyl, and alkynyl, or R11 taken together with R8 forms a 4-12 membered mononitrogen and monosulfur or monooxygen containing heterocyclic ring optionally substituted with lower alkyl, hydroxy, keto, phenyl, carboxyl or carboxyl ester, and fused phenyl, or R11 taken together with R8 is thiazole, oxazole, benzoxazole, or benzothiazole;
      • R8 is defined as above; or
      • Y1 (when Y1 is carbon) taken together with R8 forms a 4-12 membered mononitrogen or dinitrogen containing ring optionally substituted with alkyl, aryl, keto or hydroxy; or
      • X is
  • Figure US20160296592A1-20161013-C00109
      • wherein R1 and R8 taken together form a 5-8 membered dinitrogen containing heterocycle optionally substituted with one or more substituent selected from the group consisting of lower alkyl, hydroxy, keto, phenyl, or carboxyl derivatives; and R9 is selected from the group consisting of alkylcarbonyl, arylcarbonyl, alkoxycarbornyl, aryloxycarbonyl, haloalkylcarbonyl, haloalkoxycarbonyl, alkylthiocarbonyl, arylthiocarbonyl, or acyloxymethoxycarbonyl; or
      • X is
  • Figure US20160296592A1-20161013-C00110
      • wherein R1 and R8 taken together form a 5-8 membered dinitrogen containing heterocycle optionally substituted with hydroxy, keto, phenyl, or alkyl; and R9 are both selected from the group consisting of alkylcarbonyl, arylcarbonyl, alkoxycarbonyl, aryloxycarbonyl, haloalkylcarbonyl, haloalkoxycarbonyl, alkylthiocarbonyl, arylthiocarbonyl and acyloxymethoxycarbonyl;
      • R2, R3 and R4 are independently selected from one or more substituent selected from the group consisting of H, alkyl, hydroxy, alkoxy, aryloxy halogen, haloalkyl, haloalkoxy, nitro, amino, alkylamino, acylamino, dialkylamino, cyano, alkylthio, alkylsulfonyl, carboxyl derivatives, trihaloacetamide, acetamide, aryl, fused aryl, cycloalkyl, thio, monocyclic heterocycles, fused monocyclic heterocycles, and X, wherein X is defined as above;
      • R5, R6 and R7 are independently selected from the group consisting of hydrogen, alkyl, alkenyl, alkynyl, aryl, carboxyl derivatives, haloalkyl, cycloalkyl, monocyclic heterocycles, monocycli hetemeyeles optionally substituted with alkyl, halogen, halonikyl, cyano, hydroxy, aryl, fused aryl, nitro, alkoxy, aryloxy alkylsulthnyl aylsulfortyl, staltbnamide, thio, alkylthio carboxyl derivatives, amino, amido, alkyl optionally substituted with one or more of halo, haloalkyl, hydroxy, alkoxy, aryloxy, thio, alkylthio, alkynyl, alkenyl, alkyl, arylthio, alkylsulfoxide, alkylaulfonyl, arylsulfoxide, arylsulfonyl, cyano, nitro, amino, alkylamino, dialkylamino, alkylsulfonamide, arylsufonamide, acylamide, carboxyl derivatives, sulfonamide, Sulfonic acid, phosphonic acid derivatives phosphinic acid derivatives, aryl, arylthio, arylsulfoxide, or arylsulfone all optionally substituted on the aryl ring with halo, alkyl, haloalkyl, cyano, nitro, hydroxy, carboxyl derivatives, alkoxy, aryloxy, amino, alkylamino, dialkylamino, amido, aryl, fused aryl, monocyclic heterocycles, and fused monocyclic heterocycles, monocyclic heterocyclicthio, monocyclic heterocyclicsulfoxide, and monocyclic heterocyclic sulfone, which can be optionally substituted with halo, haloalkyl, nitro, hydroxy, alkoxy, fused aryl, or alkyl, alkylcarbonyl, haloalkylcarbonyl, and arylcarbonyl, aryl optionally substituted in one or more positions with halo, haloalkyl, alkyl, alkoxy, aryloxy, methylenedioxy, Ethylenedioxy, alkylthio, haloalkythio, thio, hydroxy, cyano, nitro, acyloxy, carboxyl derivatives, carboxyalkoxy, amido, acylamino, amino, alkylamino, dialkylamino, trifluoroalkoxy, trifluoromethylsulfonyl, alkylsulfonyl, sulfonic acid, sulfonamide, aryl, fused aryl, monocyclic heterocycles and fused monocyclic heterocycles.
  • The compounds according to Formula I can exist in various isomers, enantiomers, tautomers, racemates and polymorphs, and all such forms are meant to be included.
  • It is another object of the invention to provide pharmaceutical compositions comprising compounds of the Formula I. Such compounds and compositions are useful in selectively inhibiting or antagonizing the αvβ3 and/or αvβ5 integrins and therefore in another embodiment the present invention relates to a method of selectively inhibiting or antagonizing the αvβ3 and/or αv 5 integrin. The invention further involves treating or inhibiting pathological conditions associated therewith such as osteoporosis, humoral hypercalcemia of malignancy, Paget's disease, tumor metastasis, solid tumor growth (neoplasia), angiogenesis, including tumor angiogenesis, retinopathy including macular degeneration and diabetic retinopathy, arthritis, including rheumatoid arthritis, periodontal disease, psoriasis, smooth muscle cell migration and restenosis in a mammal in need of such treatment. Additionally, such pharmaceutical agents are useful as antiviral agents, and antimicrobials.
  • The compounds of this invention are 1) αvβ3 integrin antagonists; or 2) αvβ5 integrin antagonists; or 3) mixed or dual αvβ3vβ5 antagonists. The present invention includes compounds which inhibit the respective integrins and also includes Pharmaceutical compositions comprising such compounds. The present invention further provides for methods for treating or preventing conditions mediated by the αvβ3 and/or αvβ3 receptors in a mammal in need of such treatment comprising administering a therapeutically effective amount of the compounds of the present invention and pharmaceutical compositions of the present invention. Administration of such compounds and compositions of the present invention inhibits angiogenesis, tumor metastasis, tumor growth, osteoporosis, Paget's disease, humoral hypercalcemia of malignancy, retinopathy, macular degeneration, arthritis, periodontal disease, smooth muscle cell migration, including restenosis and artherosclerosis, and viral diseases.
  • Further, it has been found that the selective antagonism of the αvβ3 integrin is desirable in that the αvβ6 integrin may play a role in normal physiological processes of tissue repair and cellular turnover that routinely occur in the skin and pulmonary tissue, and αvβ8 may play a role in the regulation of growth in the human pathway. Therefore, compounds which selectively inhibit the αvβ3 integrin as opposed to the αvβ6 and/or the αvβ8 integrin have reduced side-effects associated with inhibition of the αvβ6 and/or the αvβ8 integrin. It is therefore another object of the present invention to provide compounds that are selective antagonists of αvβ3 and/or αvβ5 as opposed to αvβ6, and it is yet another object of the present invention to provide compounds that are selective antagonists of CVD3 and/or αvβ5 as opposed to αvβ8.
  • The present invention relates to a class of compounds represented by the Formula 1, 2
  • Figure US20160296592A1-20161013-C00111
      • or a pharmaceutically acceptable salt thereof, wherein 3
  • Figure US20160296592A1-20161013-C00112
      • is a 4-8 membered monocyclic or a 7-12 membered bicyclic ring, optionally containing 1 to 4 heteroatoms, selected from the group consisting of O, N or S; optionally saturated or unsaturated, optionally substituted with one or more substituent selected from the group consisting of alkyl, haloalkyl, aryl, heteroaryl, halogen, alkoxyalkyl, aminoalkyl, hydroxy, nitro, alkoxy, hydroxyalkyl, thioalkyl, amino, alkylamino, arylamino, alkylsulfonamide, acyl, acylamino, sulfone, sulfonamide, allyl, alkenyl, methylenedioxy, ethylenedioxy, alkynyl, carboxamide, cyano, and —(CH2)nCOR wherein n is 0-2 and R is hydroxy, alkoxy, alkyl or amino;
      • A1 is a 5-9 membered monocyclic or 7-14 membered polycyclic heterocycle of the formula 4
  • Figure US20160296592A1-20161013-C00113
      • containing at least one nitrogen atom and optionally 1 to 4 heteroatoms or groups selected from O, N, S, SO2 or CO; optionally saturated or unsaturated; optionally substituted by one or more Rk selected from the group consisting of hydroxy, alkyl, alkoxy, alkoxyalkyl, thioalkyl, haloalkyl, cyano, amino, alkylamino, halogen, acylamino, sulfonamide and —COR wherein R is hydroxy, alkoxy, alkyl or amino; 5
  • Figure US20160296592A1-20161013-C00114
      • include the following heterocyclic ring systems containing at least one nitrogen atom:
  • Figure US20160296592A1-20161013-C00115
      • wherein Zn is H, alkyl, alkoxy, hydroxy, amine, alkylamine, dialkylamine, carboxyl, alkoxycarbonyl, hydroxyalkyl, halogen or haloalkyl and R1 is H, alkyl, alkoxyalkyl, acyl, haloalkyl or alkoxycarbonyl. More specifically some examples of embodiments include pyridylamino, imidazolylamino, morpholinopyridine, tetrahydronaphthyridine, oxazolylamino, thiazolylamino, pyrimidinylamino, quinoline, isoquinoline, tetrahydroquinoline, imidazopyridine, benzimidazole, pyridone quinolone.
  • The following heteroaryls include the ring systems described above.
  • Figure US20160296592A1-20161013-C00116
    Figure US20160296592A1-20161013-C00117
  • For the pyridyl derived heterocycle, the substituents X4 and X5 are selected from the group consisting of H, alkyl, branched alkyl, alkylamino, alkoxyalkylamino, haloalkyl, thioalkyl, halogen, amino, alkoxy, aryloxy, alkoxyalkyl, hydroxy, cyano or acylamino groups. In another embodiment of the invention, the substituents X4 and X5 can be methyl, methoxy, amine, methylamine, trifluoromethyl, dimethylamine, hydroxy, chloro, bromo, fluoro and cyano. X6 may preferentially be H, alkyl, hydroxy, halogen, alkoxy and haloalkyl. Alternately, the pyridyl ring can be fused with a 4-8 membered ring, optionally saturated or unsaturated. Some examples of these ring systems include tetrahydronaphthyridine, quinoline, tetrahydroquinoline, azaquinoline, morpholinopyridine, imidazo-pyridine and the like. The monocyclic ring systems such as imidazole, thiazole, oxazole, pyrazole, and the like, may contain an amino or alkylamino substituent at any position within the ring.
  • In another embodiment of the present invention, when Z1 of Formula I is CO or SO2, the linkage A1-Z2 of Formula I includes the heterocycle derived ring systems such as: pyridine, imidazole, thiazole, oxazole, benzimidazole, imidazopyridine and the like.
  • Other heterocycles for A1-Z2 of the present invention include
  • Figure US20160296592A1-20161013-C00118
      • wherein X4 is as defined above. or
      • A1 is
  • Figure US20160296592A1-20161013-C00119
      • wherein Y1 is selected from the group consisting of N—R2, O, and S;
      • R2 is selected from the group consisting of H; alkyl; aryl; hydroxy; alkoxy; cyano; alkenyl; alkynyl; amido; alkylcarbonyl; arylcarbonyl; alkoxycarbonyl; aryloxycarbonyl; haloalkylcarbonyl; haloalkoxycarbonyl; alkylthiocarbonyl; arylthiocarbonyl; acyloxymethoxycarbonyl;
      • R2 taken together with R7 forms a 4-12 membered dinitrogen containing heterocycle optionally substituted with one or more substituent selected from the group consisting of lower alkyl, thioalkyl, alkylamino, hydroxy, keto, alkoxy, halo, phenyl, amino, carboxyl or carboxyl ester, and fused phenyl; or
      • R2 taken together with R7 forms a 4-12 membered heterocycle containing one or more heteroatom selected from O, N and S optionally unsaturated; or
      • R2 taken together with R7 forms a 5 membered heteroaromatic ring fused with a aryl or heteroaryl ring;
      • R7 (when not taken together with R2) and R8 are independently selected from the group consisting of H; alkyl; alkenyl; alkynyl; aralkyl; amino; alkylamino; hydroxy; alkoxy; arylamino; amido, alkylcarbonyl, arylcarbonyl; alkoxycarbonyl; aryloxy; aryloxycarbonyl; haloalkylcarbonyl; haloalkoxycarbonyl; alkylthiocarbonyl; arylthiocarbonyl; acyloxymethoxycarbonyl; cycloalkyl; bicycloalkyl; aryl; acyl; benzoyl; or
      • NR7 and R8 taken together form a 4-12 membered mononitrogen containing monocyclic or bicyclic ring optionally substituted with one or more substituent selected from lower alkyl, carboxyl derivatives, aryl or hydroxy and wherein said ring optionally contains a heteroatom selected from the group consisting of O, N and S;
      • R5 is selected from the group consisting of H and alkyl.
      • As evidenced by the continuing research in integrin antagonists and by the shortcomings of the compounds and methods of the art, there still remains a need for non-peptidic selective αvβ3 and/or αvβ5 antagonist that displays decreased side-effects, and improved potency, pharmacodynamic, and pharmacokinetic properties, such as oral bioavailability and duration of action, over already described compounds. Such compounds would prove to be useful for the treatment, prevention, or suppression of various pathologies enumerated above that are mediated by αvβ3 and/or αvβ5 receptor binding and cell adhesion and activation.
  • The compounds of the present invention further show greater selectivity for the αvβ3 and/or αvβ5 integrin than for the αvβ6 integrin. It has been found that the selective antagonism of the αvβ3 integrin is desirable in that the αvβ6 integrin may play a role in normal physiological processes of tissue repair and cellular turnover that routinely occur in the skin and pulmonary tissue, and the inhibition of this function can be deleterious (Huang et al., Am J Respir Cell Mol Biol 1998, 19(4): 636-42). Therefore, compounds of the present invention which selectively inhibit the αvβ3 integrin as opposed to the αvβ6 integrin have reduced side effects associated with inhibition of the αvβ6 integrin.
  • The compounds of the present invention comprise the R-isomers of the carbon of the beta amino acid. Other isomers may result from additional chiral centers, depending on the substitution of the parent structure.
  • The present invention relates to a class of compounds represented by the Formula I:
  • Figure US20160296592A1-20161013-C00120
      • or a pharmaceutically acceptable salt or tautomer thereof;
      • wherein X has the structure of formula Ia:
  • Figure US20160296592A1-20161013-C00121
      • and wherein X is optionally substituted with one or more substituents independently selected from the group consisting of OH, alkyl, alkenyl, alkynyl, haloalkyl, alkylaryl, arylalkyl, alkoxy, dialkylamino, thioalkyl, cycloalkyl, CN, NO2, and halogen; or, in an alternative embodiment, X is a monocyclic heterocycle containing a N as shown, optionally substituted with one to ten, or alternatively 1-3 substituents independently selected from the group consisting of H, OH, alkyl, CN, NO2, aminoalkyl, halogen, haloalkyl, and alkoxy;
      • Y is a six-membered aryl; or alternatively, a six-membered heterocycyl ring containing 1 to 2 heteroatoms, selected from the group consisting of O, N or S; wherein the six-membered ring is optionally substituted with one or more substitutents independently selected from the group consisting of OH, alkyl, alkoxy, NO2, NH2, CN, NHCOCF3, COCF3, haloalkyl, aryl, methylenedioxy, ethylenedioxy, heterocycyl, halogen, alkoxyalkyl, aminoalkyl, hydroxyalkyl, thioalkyl, alkylamino, arylamino, alkylsulfonamido, acyl, acylamino, alkylsulfone, sulfonamido, allyl, alkenyl, alkynyl, carboxamide NHCOCF3, and —(CH2)mCOR2;
      • m is a number from 0 to 2;
      • R2 is hydroxy, alkoxy, or amino;
      • Z is a 5 to 6-membered monocyclic, or a 9 to 12-membered bicyclic, aryl or heterocycyl ring; optionally containing 1 to 5 heteroatoms selected from the group consisting of O, N or S; optionally saturated or unsaturated, optionally substituted with one or more substituents selected from the group consisting of alkyl, haloalkyl, aryl, heterocycyl, arylalkyl, aryloxy, phenethyl, arylsulfone, halogen, alkoxyalkyl, aminoalkyl, cycloalkyl, hydroxy, nitro, alkoxy, hydroxyalkyl, thioalkyl, amino, alkylamino, arylamino, alkylsulfonamido, acyl, acylamino, alkylsulfone, sulfonamido, allyl, alkenyl, methylenedioxy, ethylenedioxy, alkynyl, carboxamide, cyano, NHCOCF3, and —(CH2)mCOR2; wherein the aryl and heterocycyl substituents are also optionally substituted with one or more substituents selected from the group consisting of alkyl, cycloalkyl, haloalkyl, halogen, alkoxyalkyl, aminoalkyl, hydroxy, nitro, alkoxy, hydroxyalkyl, thioalkyl, amino, alkylamino, arylamino, alkylsulfonamido, acyl, acylamino, alkylsulfone, sulfonamido, allyl, alkenyl, methylenedioxy, ethylenedioxy, alkynyl, carboxamide, cyano, and —(CH2)mCOR2;
      • Q is NH or CH2;
      • R is selected from the group consisting of OH, alkoxy, and NHR3;
      • R3 is H or an alkyl group;
      • R1 is H, CN, NO2, acyl, haloalkyl, alkenyl, alkynyl, or alkyl;
      • n is 0, 1, or 2,
      • and carbon atom 3 is in the (R) conformation.
  • It is another embodiment of the invention to provide pharmaceutical compositions comprising compounds of the Formula I. Such compounds and compositions are useful in selectively inhibiting or antagonizing the αvβ3 and/or αvβ5, integrins and therefore in another embodiment the present invention relates to a method of selectively inhibiting or antagonizing the αvβ3 and/or αvβ5 integrin. The invention further embodies treating or inhibiting pathological conditions associated therewith such as osteoporosis, humoral hypercalcemia of malignancy, Paget's disease, tumor metastasis, solid tumor growth (neoplasia), angiogenesis, including tumor angiogenesis, retinopathy including macular degeneration and diabetic retinopathy, arthritis, including rheumatoid arthritis and osteoarthritis, periodontal disease, psoriasis, smooth muscle cell migration and restenosis in a mammal in need of such treatment. Additionally, such pharmaceutical agents are useful as antiviral agents, and antimicrobials. The compounds of the present invention may be used alone or in combination with other pharmaceutical agents.
  • The compounds of this invention are 1) αvβ3 integrin antagonists; or 2) αvβ5 integrin antagonists; or 3) mixed or dual αvβ3vβ5 antagonists. The present invention includes compounds which inhibit the respective integrins and also includes pharmaceutical compositions comprising such compounds. The present invention further provides for methods for treating or preventing conditions mediated by the αvβ3 and/or 60vβ5 receptors in a mammal in need of such treatment comprising administering a therapeutically effective amount of the compounds of the present invention and pharmaceutical compositions of the present invention. Administration of such compounds and compositions of the present invention inhibits angiogenesis, tumor metastasis, tumor growth, osteoporosis, Paget's disease, humoral hypercalcemia of malignancy, retinopathy, macular degeneration, arthritis, periodontal disease, smooth muscle cell migration, including restenosis and artherosclerosis, and viral diseases.
  • The compounds of the present invention further show greater selectivity for the αvβ3 and/or αvβ5 integrin than for the αvβ6 integrin. It has been found that the selective antagonism of the αvβ3 integrin is desirable in that the αvβ6 integrin may play a role in normal physiological processes of tissue repair and cellular turnover that routinely occur in the skin and pulmonary tissue, and the inhibition of this function can be deleterious. Therefore, compounds of the present invention which selectively inihibit the αvβ3 integrin as opposed to the αvβ6 integrin have reduced side-effects associated with inhibition of the αvβ6 integrin.
  • The present invention relates to a class of compounds represented by the Formula I
  • Figure US20160296592A1-20161013-C00122
      • or a pharmaceutically acceptable salt thereof, wherein
      • A1 is a 5-9 membered monocyclic or 7-12 membered bicyclic heterocycle of the formula
  • Figure US20160296592A1-20161013-C00123
      • containing at least one nitrogen atom and optionally 1 to 3 heteroatoms, selected from the group consisting of O, N or S; optionally saturated or unsaturated; optionally substituted by one or more Rk selected from the group consisting of hydroxy, alkyl, cycloalkyl, alkoxy, alkoxyalkyl, thioalkyl, cyano, amino, alkylamino, halogen, acylamino, sulfonamide and —COR wherein R is hydroxy, alkoxy, alkyl or amino;
      • or A1 is
  • Figure US20160296592A1-20161013-C00124
      • wherein Y1 is selected from the group consisting of N—R2, O, and S;
      • R2 is selected from the group consisting of H; alkyl; cycloalkyl; aryl; hydroxy; alkoxy; cyano; alkenyl; alkynyl; amido; alkylcarbonyl; arylcarbonyl; alkoxycarbonyl; aryloxycarbonyl; haloalkylcarbonyl; haloalkoxycarbonyl; alkylthiocarbonyl; arylthiocarbonyl; acyloxymethoxycarbonyl;
      • R2 taken together with R7 forms a 4-12 membered dinitrogen containing heterocycle optionally substituted with one or more substituent selected from the group consisting of lower alkyl, thioalkyl, alkylamino, hydroxy, keto, alkoxy, halo, phenyl, amino, carboxyl or carboxyl ester, and fused phenyl;
      • or R2 taken together with R7 forms a 4-12 membered heterocycle containing one or more heteroatom selected from O, N and S optionally unsaturated;
      • or R2 taken together with R7 forms a 5 membered heteroaromatic ring fused with a aryl or heteroaryl ring;
      • R7 (when not taken together with R2) and R8 are independently selected from the group consisting of H; alkyl; alkenyl; alkynyl; aralkyl; amino; alkylamino; hydroxy; alkoxy; arylamino; amido, alkylcarbonyl, arylcarbonyl; alkoxycarbonyl; aryloxy; aryloxycarbonyl; haloalkylcarbonyl; haloalkoxycarbonyl; alkylthiocarbonyl; arylthiocarbonyl; acyloxymethoxycarbonyl; cycloalkyl; bicycloalkyl; aryl; acyl; benzoyl;
      • or NR7 and R8 taken together form a 4-12 membered mononitrogen containing monocyclic or bicyclic ring optionally substituted with one or more substituent selected from lower alkyl, carboxyl derivatives, aryl or hydroxy and wherein said ring optionally contains a heteroatom selected from the group consisting of O, N and S;
      • R5 is selected from the group consisting of H, cycloalkyl and alkyl; 5
      • A1 is
  • Figure US20160296592A1-20161013-C00125
      • wherein Y2 is selected from the group consisting of alkyl; cycloalkyl; bicycloalkyl; aryl; monocyclic heterocycles;
      • Z1 is selected from the group consisting of CH2, O, CH2O, NRk, CO, S, SO, CH(OH) and SO2, wherein Rk is selected from H or lower alkyl;
      • Z2 is a 1-5 carbon linker optionally containing one or more heteroatom selected from the group consisting of O, S and N; alternatively Z1-Z2 may further contain a carboxamide, sulfone, sulfonamide, alkenyl, alkynyl, or acyl group;
      • wherein the carbon and nitrogen atoms of Z1-Z2 are optionally substituted by alkyl, cycloalkyl, alkoxy, thioalkyl, alkylsulfone, aryl, alkoxyalkyl, hydroxy, alkylamino, heteroaryl, alkenyl, alkynyl, carboxyalkyl, halogen, haloalkyl or acylamino;
      • n is an integer 0, 1 or 2;
      • Rc is selected from the group consisting of hydrogen; alkyl; cycloalkyl; halogen, hydroxy, nitro, alkoxy, amino, haloalkyl, aryl, heteroaryl, alkoxyalkyl, aminoalkyl, hydroxyalkyl, thioalkyl, alkylamino, arylamino, alkylsulfonylamino, acyl, acylamino, sulfonyl, sulfonamide, allyl, alkenyl, methylenedioxy, ethylenedioxy, alkynyl, alkynylalkyl, carboxy, alkoxycarbonyl, carboxamido, cyano, and —(CH2)nCOR wherein n is 0-2 and R is selected from hydroxy, alkoxy, alkyl and amino;
      • X is selected from the group consisting of O, CO, SO2, NRm and (CHRp)n; wherein Rp and Rm are H or alkyl;
      • Rb is X3—Rh wherein X3is selected from the group consisting of O, S and NR1 wherein Rh and Rj are independently selected from the group consisting of H, alkyl, acyl, aryl, aralkyl and alkoxyalkyl; and
      • It is another object of the invention to provide pharmaceutical compositions comprising compounds of the Formula I. Such compounds and compositions are useful in selectively inhibiting or antagonizing the αv β3 and/or αv β5 integrin(s) and therefore in another embodiment the present invention relates to a method of selectively inhibiting or antagonizing the αv β3 and/or αv β5 integrin(s). The invention further involves treating or inhibiting pathological conditions associated therewith such as osteoporosis, humoral Hypercalcemia of malignancy, Paget's disease, tumor metastasis, solid tumor growth (neoplasia), angiogenesis, including tumor angiogenesis, retinopathy including macular degeneration and diabetic retinopathy, arthritis, including rheumatoid arthritis, periodontal disease, psoriasis, smooth muscle cell migration including restenosis or atherosclerosis in a mammal in need of such treatment. Additionally, such pharmaceutical agents are useful as antiviral agents, antifungals and antimicrobials. The compounds of the present invention may be used alone or in combination with other pharmaceutical agents.
  • The compounds of this invention are 1) αvβ3 integrin antagonists; or 2) αvβ5 integrin antagonists; or 3) mixed or dual αvβ3 vβ5 antagonists. The present invention includes compounds which inhibit the respective integrins and also includes pharmaceutical compositions comprising such compounds. The present invention further provides for methods for treating or preventing conditions mediated by the αvβ3 and/or αvβ5 receptors in a mammal in need of such treatment comprising administering a therapeutically effective amount of the compounds of the present invention and pharmaceutical compositions of the present invention. Administration of such compounds and compositions of the present invention inhibits angiogenesis, tumor metastasis, tumor growth, osteoporosis, Paget's disease, humoral hypercalcemia of malignancy, retinopathy, macular degeneration, arthritis, periodontal disease, smooth muscle cell migration, including restenosis and artherosclerosis, and viral diseases.
  • Further, it has been found that the selective antagonism of the αvβ3 integrin is desirable in that the αvβ6 integrin may play a role in normal physiological processes of tissue repair and cellular turnover that routinely occur in the skin and pulmonary tissue, and αvβ8 may play a role in the regulation of growth in the human pathway. Therefore, compounds which selectively inihibit the αvβ3 integrin as opposed to the αvβ6 and/or the αvβ8 integrin have reduced side-effects associated with inhibition of the αvβ6 and/or the αvβ8 integrin. It is therefore another object of the present invention to provide compounds that are selective antagonists of αvβ3 as opposed to αvβ6, and it is yet another object of the present invention to provide compounds that are selective antagonists of αvβ3 and/or αvβ5 as opposed to αvβ8.
  • The present invention relates to a class of compounds represented by the Formula I
  • Figure US20160296592A1-20161013-C00126
      • or a pharmaceutically acceptable salt thereof, wherein
  • Figure US20160296592A1-20161013-C00127
      • is a 4-8 membered monocyclic ring or a 7-12 membered bicyclic ring, which ring is optionally saturated or unsaturated; which ring is optionally substituted with one or more substituent selected from the group consisting of alkyl, haloalkyl, aryl, heteroaryl, halogen, alkoxyalkyl, aminoalkyl, hydroxy, nitro, alkoxy, hydroxyalkyl, thioalkyl, amino, alkylamino, arylamino, alkylsulfonamide, acyl, acylamino, alkylsulfone, sulfonamide, allyl, alkenyl, methylenedioxy, ethylenedioxy, alkynyl, carboxamide, cyano, and —(C2)n COR wherein n is 0-2 and R is hydroxy, alkoxy, alkyl or amino;
  • A1 is a 5-9 membered monocyclic ring or 7-12 membered bicyclic heterocycle ring of the formula
  • Figure US20160296592A1-20161013-C00128
      • containing at least one nitrogen atom and optionally containing 1 to 4 heteroatoms selected from the group consisting of O, N, S, SO2 and CO; optionally saturated or unsaturated; optionally substituted by one or more Rk is selected from the group consisting of hydroxy, alkyl, alkoxy, alkoxyalkyl, thioalkyl, cyano, amino, alkylamino, haloalkyl, halogen, acylamino, sulfonamide and —COR wherein R is hydroxy, alkoxy, alkyl or amino;
      • or A1 is
  • Figure US20160296592A1-20161013-C00129
      • wherein Y1 is selected from the group consisting of N—R2, O, and S;
      • R2 is selected from the group consisting of H; alkyl; aryl; hydroxy; alkoxy; cyano; amido; alkylcarbonyl; arylcarbonyl; alkoxycarbonyl; aryloxycarbonyl; haloalkylcarhonyl; haloalkoxycarbonyl; alkylthiocarbonyl, arylthiocarbonyl; acyloxymethoxycarbonyl;
      • R2taken together with R7 forms a 4-12 membered dinitrogen containing heterocycle optionally substituted with one or more substituent selected from the group consisting of lower alkyl, thioalkyl, alkylamino, hydroxy, keto, alkoxy, halo, phenyl, amino, carboxyl or carboxyl ester;
      • or R2 taken together with R7 forms a 4-12 membered heterocycle containing one or more heteroatom selected from O, N and S optionally unsaturated;
      • or R2 taken together with R7 forms a 5 membered heterearomatic ring fused with a aryl or heteroaryl ring;
      • R7 (when not taken together with R2) and R8 are independently selected from the group consisting of H; alkyl; aralkyl; amino; alkylamino; hydroxy; alkoxy; arylamino; amido, alkylcarbonyl, arylcarbonyl; alkoxycarbonyl; aryloxy; aryloxycarbonyl; haloalkylcarbonyl; haloalkoxycarbonyl; alkylthiocarbonyl; arylthiocarbonyl; acyloxymethoxycarbonyl; cycloalkyl; bicycloalkyl; aryl; acyl; benzoyl;
      • or NR7 and R8 taken together form a 4-12 membered mononitrogen containing monocyclic or bicyclic ring optionally substituted with one or more substituent selected from lower alkyl, carboxyl derivatives, aryl or hydroxy and wherein said ring optionally contains a heteroatom selected from the group consisting of O, N and S; R5 is selected from the group consisting of H, and alkyl; or
      • A is
  • Figure US20160296592A1-20161013-C00130
      • wherein Y is selected from the group consisting of alkyl; cycloalkyl; bicycloalkyl; aryl; monocyclic heterocycles;
      • Z1 is selected from the group consisting of CH2, CH2O, O, NH, NRk, CO, S, SO, CH(OH), and SO2, wherein Rk (is selected from H or lower alkyl;
      • Z2 is a 1-5 carbon linker optionally containing one or more heteroatom selected from the group consisting of O, S and N; alternatively Z1-Z2 may further contain a carboxamide, sulfone, oxime, sulfonamide, alkenyl, alkynyl, or acyl group;
      • wherein the carbon and nitrogen atoms of Z1-Z2 are optionally substituted by alkyl, alkoxy, thioalkyl, alkylsulfone, aryl, alkoxyalkyl, hydroxy, alkylamino, heteroaryl, alkenyl, alkynyl, carboxyalkyl, halogen, haloalkyl or acylamino;
      • wherein Z2-Z1 is attached to
  • Figure US20160296592A1-20161013-C00131
      • at the para or meta position relative to the X1 substituent;
      • n is an integer 0, 1 or 2;
      • Rc is selected from the group consisting of hydrogen; alkyl; halogen, hydroxy nitro, alkoxy, amino, haloalkyl, aryl, heteroaryl, alkoxyalkyl, aminoalkyl, hydroxyalkyl, thioalkyl, alkylamino, arylamino, alkylsulfonylamino, acyl, acylamino, sulfonyl, sulfonamide, allyl, alkenyl, methylenedioxy, ethylenedioxy, alkynyl alkynylalkyl, carboxy, alkoxycarbonyl, carboxamido, cyano and —(CH2)n—COR wherein n is 0-2 and R is selected from hydroxy, alkoxy, alkyl and amino;
      • X1 is selected from the group consisting of —O—, CO, SO2, NRm and (CHRp)q; wherein Rm is H or alkyl; Rp is alkoxy or hydroxy and q is 0 or 1;
      • X2 is selected from the group consisting of —CHRc—, CO, SO2, O, NRf and S;
      • Rc is selected from the group consisting of H, alkyl, hydroxy and alkoxy; Rf is H or alkyl;
  • X or Y are independently selected from the group consisting of —CRg— or —N— wherein Rg is selected from the group consisting of H, alkyl, haloalkyl, fluoro, alkoxyalkyl, alkynyl, aryl, heteroaryl, aralkyl, alkylsulfone, heteroaralkyl, hydroxy, alkoxy, hydroxyalkyl, and carboxyalkyl;
  • The group X—X2—Y optionally contains a moiety selected from the group consisting of acyl, alkyl, amino, ether, thioether, sulfone, and olefin:
  • Figure US20160296592A1-20161013-C00132
      • forms a 3-8 membered monocyclic ring system; or an 8-11 membered bicyclic system; optionally saturated or unsaturated; the monocyclic ring system optionally containing 1-2 heteroatoms selected from N, O and S; the bicyclic ring system optionally containing 1-4 heteroatoms selected from N, O, S or optionally containing the group such as SO2 or CO); and optionally substituted with one or more substituent selected from the group consisting of alkyl, halogen, cyano, carboalkoxy, haloalkyl, alkoxyalkyl, alkylsulfone, aryl, heteroaryl, aralkyl, heteroaralkyl or alkoxy;
      • Rb is X3-Rh wherein X3 is selected from the group consisting of O, S and NRj wherein Rh and Rj are independently selected from the group consisting of H, alkyl, acyl, aryl, aralkyl and alkoxyalkyl; and
      • and n is 0, 1 or 2.
  • It is another object of the invention to provide pharmaceutical compositions comprising compounds of the Formula I. Such compounds and compositions are useful in selectively inhibiting or antagonizing the αvβ3 and/or αvβ5 integrin(s) and therefore in another embodiment the present invention relates to a method of selectively inhibiting or antagonizing the αvβ3 and/or αvβ5 integrin(s). The invention further involves treating or inhibiting pathological conditions associated therewith such as osteoporosis, immoral hypercalcemia of malignancy, Paget's disease, tumor metastasis, solid tumor growth (neoplasia), angiogenesis, including tumor angiogenesis, retinopathy including macular degeneration and diabetic retinopathy, arthritis, including rheumatoid arthritis. periodontal disease, psoriasis, smooth muscle cell migration including restenosis or atherosclerosis in a mammal in need of such treatment. Additionally, such pharmaceutical agents are useful as antiviral agents, antifungals and antimicrobials. The compounds of the present invention may be used alone or in combination with other pharmaceutical agents.
  • The compounds of this invention are 1) αvβ3 integrin antagonists; or 2) αvβ5 integrin antagonists; or 3) mixed or dual αvβ3vβ5 antagonists. The present invention includes compounds which inhibit the respective integrins and also includes pharmaceutical compositions comprising such compounds. The present invention further provides for methods for treating or preventing conditions mediated by the αvβ3 and/or αvβ5 receptors in a mammal in need of such treatment comprising, administering a therapeutically effective amount of the compounds of the present invention and pharmaceutical compositions of the present invention. Administration of such compounds and compositions of the present invention inhibits angiogenesis, tumor metastasis, tumor growth, osteoporosis, Paget's disease, humoral hypercalcemia of malignancy, retinopathy, macular degeneration, arthritis, periodontal disease, smooth muscle cell migration, including restenosis and artherosclerosis, and viral diseases.
  • The compounds of the present invention further show greater selectivity for the αvβ3 and/or αvβ5 integrin than for the αvβ6 integrin. It has been found that the selective antagonism of the αvβ3 integrin is desirable in that the αvβ6 integrin may play a role in normal physiological processes of tissue repair and cellular turnover that routinely occur in the skin and pulmonary tissue, and the inhibition of this function can be deleterious. Therefore, compounds of the present invention which selectively inihibit the αvβ3 integrin as opposed to the αvβ6 integrin have reduced side-effects associated with inhibtion of the αvβ αvβ6 integrin.
  • The present invention relates to a class of compounds represented by the Formula I.
  • Figure US20160296592A1-20161013-C00133
      • or a pharmaceutically acceptable salt thereof, wherein
  • Figure US20160296592A1-20161013-C00134
      • is a 4-8 membered monocyclic or a 7-12 membered bicyclic ring, optionally saturated or unsaturated, optionally substituted with one or more substituent selected from the group consisting of alkyl, haloalkyl, aryl, heteroaryl, halogen, alkoxyalkyl, aminoalkyl, hydroxy, nitro, alkoxy, hydroxyalkyl, thioalkyl, amino, alkylamino, arylamino, alkylsulfonamide, acyl, acylamino, alkylsulfone, sulfonamide, alkylsulfoxide, allyl, alkenyl, methylenedioxy, ethylenedioxy, alkynyl, carboxamide, cyano, and —(CH2)n COR wherein n is 0-2 and R is hydroxy, alkoxy, alkyl or amino;
      • A1 is a 5-9 membered monocyclic or 7-12 membered bicyclic heterocycle of the formula
  • Figure US20160296592A1-20161013-C00135
      • containing at least one nitrogen atom and optionally 1 to 3 additional heteroatoms, selected from the group consisting of O, N, S, CO, or SO2 optionally saturated or unsaturated; optionally substituted by one or more Rk selected from the group consisting of hydroxy, alkyl, cycloalkyl, alkoxy, alkoxyalkyl, thioalkyl, cyano, amino, alkylamino, halogen, acylamino, sulfonamide and —COR wherein R is hydroxy, alkoxy, alkyl or amino;
      • or A1 is
  • Figure US20160296592A1-20161013-C00136
      • wherein Y1 is selected from the group consisting of N—R2, O, and S;
      • R2 is selected from the group consisting of H; alkyl; cycloalkyl; aryl; hydroxy; alkoxy; cyano; alkenyl; alkynyl; amino; alkylcarbonyl; arylcarbonyl; alkoxycarbonyl; aryloxycarbonyl; haloalkylcarbonyl; haloalkoxycarbonyl; alkylthiocarbonyl; arylthiocarbonyl; acyloxymethoxycarbonyl;
      • R2 taken together with R7 forms a 4-12 membered dinitrogen containing heterocycle optionally substituted with one or more substituent selected from the group consisting of lower alkyl, thioalkyl, alkylamino, hydroxy, keto, alkoxy, halo, phenyl, amino, carboxyl or carboxyl ester, and fused phenyl;
      • or R2 taken together with R7 forms a 4-12 membered heterocycle containing one or more heteroatom selected from O, N and S optionally unsaturated;
      • or R2 taken together with R7 forms a 5 membered heteroaromatic ring fused with an aryl or heteroaryl ring;
      • R7 (when not taken together with R2) and R8 are independently selected from the group consisting of H; alkyl; alkenyl; alkynyl; aralkyl; amino; alkylamino; hydroxy; alkoxy; arylamino; amido, alkylcarbonyl; arylcarbonyl; alkoxycarbonyl, aryloxy, aryloxycarbonyl; haloalkylcarbonyl; haloalkoxycarbonyl; alkylthiocarbonyl; arylthiocarbonyl; acyloxymethoxycarbonyl; cycloalkyl; bicyclocalkyl; aryl; acyl; benzoyl;
      • or NR7 and R8 taken together form a 4-12 membered mononitrogen containing monocyclic or bicyclic ring optionally substituted with one or more substituent selected from lower alkyl, carboxyl derivatives, aryl or hydroxy and wherein said ring optionally contins a heteroatom selected from the group consisting of O, N and S;
      • R5 is selected from the group consisting of H, hydroxy, alkoxy, cycloalkyl, and alkyl; or
  • Figure US20160296592A1-20161013-C00137
      • wherein Y2 is selected from the ggroup consisting of alkyl; cycloalkyl; bicycloalkyl; aryl; monocyclic heterocycles;
      • Z1 is selected from the group consisting of CH2, O, CH2O, NRk, CO, S, SO, CH(OH) and SO2, wherein Rk is selected from H or lower alkyl;
      • Z2 is a 1-5 carbon linker optionally containing one or more heteroatom selected from the group consisting of O, S and N; alternatively Z1-Z2 may further contain a Carboxamide, sulfone, sulfonamide, alkenyl, alkynyl, or acyl group;
      • wherein the carbon and nitrogen atoms of Z1-Z2 are optionally substituted by alkyl, cycloalkyl, alkoxy, thioalkyl, alkylsulfone, aryl, arylsulfone, alkoxyalkyl, hydroxy, alkylamino, heteroaryl, alkenyl, alkynyl, carboxyalkyl, halogen, haloalkyl, or acylamino;
      • n is an integer 1 or 2;
      • Rc is selected from the group consisting of hydrogen, alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl, halogen, hydroxy, nitro, alkoxy, amino, haloalkyl, aryl, heteroaryl, alkoxyalkyl, aminoalkyl, hydroxyalkyl, thioalkyl, alkylamino, arylamino, alkylsulfonylamino, acyl, acylamino, sulfonyl, sulfonamide, allyl, alkenyl, methylenedioxy, ethylenedioxy; alkynyl, alkynylalkyl, carboxy, alkoxycarbonyl, carboxamido, cyano, and —(CH2)nCOR wherein n is 0-2 and R is selected from hydroxy, alkoxy, alkyl and amino;
      • X is selected from the group consisting of —CHRe—, —NRf—, —O—, —S—, —SO2—, and —CO— wherein Rc is H, lower alkyl, alkoxy, cycloalkyl, alkoxyalkyl, hydroxy, alkynyl, alkenyl, haloalkyl, thioalkyl or aryl; wherein when Re is hydroxy, the hydroxy group can optionally form a lactone with the carboxylic acid function of the chain; wherein Rf is selected from the group consisting of H, alkyl, heteroalkyl, aryl, heteroaryl, aralkyl, arakylheteroaryl, and haloalkyl;
      • Y is selected from the group consisting of (CH2)p, —CR9—, —NR9, CO and SO2, wherein R9 is selected from the group consisting of H, alkyl, haloalkyl, alkoxyalkyl, alkynyl, aryl, heteroaryl, aralkyl, hydroxyl, hydroxyalkyl, alkoxy, and carboxyalkyl; wherein p is 0 or 1;
      • optionally the group X-Y can contain a moiety selected from the group consisting of acyl, alkyl, sulfonyl, amino, ether, thioether, carboxamido, sulfonamido, aminosulfonyl and olefins;
      • Y3 and Y4 are independently selected from the group consisting of alkyl, haloalkyl, hydroxy, alkoxy, cyano, halogen, aralkyl, heteroaralkyl, alkoxyalkyl, hydroxyalkyl, aryloxyalkyl, alkylsulfone, alkene or alkyne; wherein the alkyl group optionally contains one or more heteroatoms selected from the group consisting of N, O, and S;
      • alternately, when Y3 is an aryl or a heteroaryl, Y4 may be an aryl, heteroaryl, alkene, alkyne, alkoxy, hydroxy, cyano, alkoxyalkyl or an alkylsulfone;
      • Y5 is C;
      • Optionally, Y3, Y4 and Y5 may form a sulfone (SO2) group;
      • or Y3 taken together with Y4 forms a 3-8 membered monocyclic or a 7-11 membered bicyclic ring, optionally containing one or mare double bonds, optionally containing one or more heteroatom or functional group selected from O, NR9, S, CO or SO2, optionally substituted with one or more substituent selected from the group consisting of alkyl, heteroalkyl, hydroxy, halogen, lialoalk,O, alkoxy, alkyne, cyano, alkylsulfone, sulfonamide, aryl, heteroaryl, arakylaryl, heteroarakylarylcarboalkoxy and carboxyalkyl;
      • Rb is X2—Rh wherein X2 is selected from the group consisting of O, S and NR1 wherein Rh and Rj are independently selected from the group consisting of H, alkyl, aryl, aralkyl, heteroalkyl, heteroaryl, heteroarakylaryl, acyl, and alkoxyalkyl;
  • It is another object of the invention to provide pharmaceutical compositions comprising compounds of the Formula I. Such compounds and compositions are useful in selectively inhibiting or antagonizing the αvβ3 and/or αvβ5 integrins and therefore in another embodiment the present invention relates to a method of selectively inhibiting or antagonizing the αvβ3 and/or αvβ5 integrin. The invention further involves treating or inhibiting pathological conditions associated therewith such as osteopbrosis, humoral hypercalcemia of malignancy, Paget's disease, tumor metastasis, solid tumor growth (neoplasia), angiogenesis, including tumor angiogenesis, retinopathy including macular degeneration and diabetic retinopathy, arthritis, including rheumatoid arthritis, periodontal disease, psoriasis, smooth muscle cell migration and restenosis in a mammal in need of such treatment. Additionally, such pharmaceutical agents are useful as antiviral agents, and antimicrobials. The compounds of the present invention may be used alone or in combination with other pharmaceutical agents.
  • The present invention as a first object provides compounds of the following formula (I)
  • Figure US20160296592A1-20161013-C00138
      • or a pharmaceutically acceptable salt, prodrug or ester thereof, wherein
      • G is selected from the group consisting of:
  • Figure US20160296592A1-20161013-C00139
      • wherein Q is NH or O and Q′ is H, C1-C6alkyl, phenyl, or phenyl-C1-C4-alkyl;
  • Figure US20160296592A1-20161013-C00140
      • wherein R′ and R″ are independently H or C1-C-4 alkyl;
      • B is a C1C4 alkyl or a C2-C4 alkenyl;
      • A is CH2, O, S(O)n wherein n is zero, 1 or 2, NH, a group CON(R′″) or N(R′″)CO wherein R′″ is hydrogen or CH3;
  • R1 is selected from the group consisting of H, C1-C4 alkyl, C1-C4 alkoxy, OH, halogen, and CF3;
      • X is C═O or completes a single bond;
      • R2 is selected from the group consisting of H, C1-C4 alkyl, C3-C7 cycloalkyl, C1-C4-alkylcycloalkyl; aryl unsubstituted or optionally substituted by one to three Substituents independently selected from halogen, CF3, C1-C4 alkyl, hydroxy and C1-C4 alkoxy; aralkyl; and C5-C7monocyclic heteroaryl ring containing one to three heteroatoms chosen from O, S and N, unsubstituted or optionally substituted by one to three substituents independently selected from halogen, CF3, C1-C4 alkyl, hydroxy and C1-C4 alkoxy;
      • Y is (CH2)n wherein n is 1 or 2;
      • R is hydrogen, C1-C6 alkyl, C2-C4 alkenyl, C2-C4alkynyl, aryl or aryl-C1-C4 alkyl.
  • With the proviso that m can not be 0 when G is:
  • Figure US20160296592A1-20161013-C00141
      • wherein Q′ is H and Q is O and X is (C═O)m.
  • The present invention includes within its scope all possible isomers, stereoisomers and optical isomers and their mixtures, and the bioprecursors or metabolites of the compounds of formula (I).
  • It is another object of the invention to provide pharmaceutical compositions comprising compounds of the Formula I. Such compounds and compositions are useful in selectively inhibiting or antagonizing the αvβ3 integrin and therefore in another embodiment the present invention relates to a method of selectively inhibiting or antagonizing the αvβ3 integrin. The invention further involves treating or inhibiting pathological conditions associated therewith such as osteoporosis, humoral hypercalcemia of malignancy, Paget's disease, tumor metastasis, solid tumor growth (neoplasia), angiogenesis, including tumor angiogenesis, retinopathy including macular degeneration and diabetic retinopathy, arthritis, including rheumatoid arthritis, periodontal disease, psoriasis, smooth muscle cell migration and restenosis in a mammal in need of such treatment. Additionally, such pharmaceutical agents are useful as antiviral agents, and antimicrobials.
  • As evidenced by the continuing research in integrin antagonists and by the shortcomings of the compounds and methods of the art, there still remains a need for small-molecule, non-peptidic selective αvβ3 and/or αvβ5 antagonist that displays decreased side-effects, and improved potency, pharmacodynamic, and pharmacokinetic properties, such as oral bioavailability and duration of action, over already described compounds. Such compounds would prove to be useful for the treatment, prevention, or suppression of various pathologies enumerated above that are mediated by αvβ3 and/or αvβ5 receptor binding and cell adhesion and activation.
  • The compounds of this invention include 1) αvβ3 integrin antagonists; or 2) αvβ5 integrin antagonists; or 3) mixed or dual αvβ3vβ5 antagonists. The present invention includes compounds which inhibit the respective integrins and also includes pharmaceutical compositions comprising such compounds. The present invention further provides for methods for treating or preventing conditions mediated by the αvβ3 and/or αvβ5receptors in a mammal in need of such treatment comprising administering a therapeutically effective amount of the compounds of the present invention and pharmaceutical compositions of the present invention. Administration of such compounds and compositions of the present invention inhibits angiogenesis, tumor metastasis, tumor growth, skeletal malignancy of breast cancer, osteoporosis, Paget's disease, humoral hypercalcemia of malignancy, retinopathy, macular degeneration, arthritis including rheumatoid, periodontal disease, smooth muscle cell migration,
  • Including restenosis and artherosclerosis, and microbial or viral diseases.
  • The compounds of the present invention can be used, alone or in combination with other therapeutic agents, in the treatment or modulation of various conditions or disease states described above.
  • In order to prevent bleeding side effects associated with the inhibition of αvβ3, it would be beneficial to have a high selectivity ratio of αvβ3 and αvβ5over αvβ3. The compounds of the present invention include selective antagonists of αvβ3 over αvβ3.
  • The present invention relates to a class of compounds represented by Formula I
  • Figure US20160296592A1-20161013-C00142
      • or a pharmaceutically acceptable salt thereof, wherein
  • Figure US20160296592A1-20161013-C00143
      • is a 4-8 membered monocyclic or a 7-12 membered bicyclic ring, containing 1 to 5 heteroatoms, selected from the group consisting of O, N or S; optionally saturated or unsaturated, optionally substituted with one or more substituents selected from the group consisting of alkyl, haloalkyl, aryl, heteroaryl, halogen, alkoxyalkyl, aminoalkyl, hydroxy, nitro, alkoxy, hydroxyalkyl, thioalkyl, amino, alkylamino, arylamino, alkylsulfonamide, acyl, acylamino, alkylsullone, sulfonamide, allyl, alkenyl, methylenedioxy, ethylenedioxy, alkynyl, carboxamide, cyano, and —(CH2)mCOR wherein m is 0-2 and R is hydroxy, alkoxy, alkyl or amino; with the proviso that when Y4 in formula I is H, the ring A may not be an oxazole, with X-Y containing side-chain connected at the carbon-2 as in
  • Figure US20160296592A1-20161013-C00144
  • The ring A may further contain a carboxamide, sulfone, sulfonamide or an acyl group.
      • A1 is a 5-9 membered monocyclic or 8-14 membered poly-cyclic heterocycle of the formula
  • Figure US20160296592A1-20161013-C00145
      • containing at least one nitrogen atom and optionally 1 to 4 heteroatoms or groups, selected from O, N, S, SO2 or CO; optionally saturated or unsaturated; optionally substituted by one or more Rk selected from the group consisting of hydroxy, alkoxy, alkoxyalkyl, thioalkyl, haloalkyl, cyano, amino, alkylamino, halogen, acylamino, sulfonamide and —COR wherein R is hydroxy, alkoxy, alkyl or amino; or A1 is
  • Figure US20160296592A1-20161013-C00146
      • wherein Y1 is selected from the group consisting of N—R2, O, and S;
      • R2 is selected from the group consisting of H; alkyl; aryl; hydroxy; alkoxy; cyano; amido; alkylcarbonyl; arylcarbonyl; alkoxycarbonyl; aryloxycarbonyl; haloalkylcarbonyl; haloalkoxycarbonyl; alkylthiocarbonyl; arylthiocarbonyl; acyloxymethoxycarbonyl;
      • R2 taken together with R7 forms a 4-12 membered dinitrogen containing heterocycle optionally substituted with one or more substituent selected from the group consisting of lower alkyl, thioalkyl, alkylamino, hydroxy, keta, alkoxy, halo, phenyl, amino, carboxyl or carboxyl ester; or
      • R2 taken together with R7 forms a 4-12 membered heterocycle containing one or more heteroatom selected from O, N and S optionally unsaturated; or
      • R2 taken together with R7 forms a 5 membered heteroaromatic ring fused with an aryl or heteroaryl ring;
      • R7 (when not taken together with R2) and R8 are independently selected from the group consisting of H; alkyl; aralkyl; amino; alkylamino; hydroxy; alkoxy; arylamino; amido, alkylcarbonyl, arylcarbonyl; alkoxycarbonyl; aryloxy; aryloxycarbonyl; haloalkylcarbonyl; haloalkoxycarbonyl; alkylthiocarbonyl; arylthiocarbonyl; acyloxymethoxycarbonyl; cycloalkyl; bicycloalkyl; aryl; acyl; benzoyl; or
      • NR7 and R8 taken together form a 412 membered mononitrogen containing monocyclic or bicyclic ring optionally substituted with one or more substituent selected from lower alkyl, carboxyl derivatives, aryl or hydroxy and wherein said ring optionally contains a heteroatom selected from the group consisting of O, N and S;
      • R5 is selected from the group consisting of H and alkyl; or
      • A1 is
  • Figure US20160296592A1-20161013-C00147
      • wherein Y2 is selected from the group consisting of alkyl; cycloalkyl; bicycloalkyl; aryl; monocyclic heterocycles;
      • Z1 is selected from the group consisting of CH2, CH2O, O, NH, CO, S, SO, CH(OH) and SO2;
      • Z2 is a 1-5 carbon linker optionally containing one or more heteroatom selected from the group consisting of O, S and N;
      • alternatively Z1-Z2 may further contain a carboxamide, sulfone, sulfonamide, alkenyl, alkynyl, or acyl group;
      • wherein the carbon and nitrogen atoms of Z1-Z2 are optionally substituted by alkyl, alkoxy, thioalkyl, alkylsulfone, aryl, alkoxyalkyl, hydroxy, alkylamino, heteroaryl, alkenyl, alkynyl, carboxyalkyl, halogen, haloalkyl or acylamino;
      • Additionally, Z1-Z2 may contain a 5- or 6-membered aryl or heteroaryl ring optionally substituted with Rc, wherein the heteroaryl ring may contain 1-3 heteroatoms selected from the group consisting of O, N and S; Rc is selected from the group consisting of H, alkyl, haloalkyl, aryl, heteroaryl, halogen, alkoxyalkyl, aminoalkyl, hydroxy, alkoxy, carboxamide, or cyano.
      • X is selected from the group consisting of —CHRe—, —NRf—, —O—, —S—, —SO2—, and —CO— wherein Rc is H, lower alkyl, alkoxy, cycloalkyl, alkoxyalkyl, hydroxy, alkynyl, alkenyl, haloalkyl, thioalkyl or aryl; wherein when Rc is hydroxy, the hydroxy group can optionally form a lactone with the carboxylic acid function of the chain; wherein Rf is selected from the group consisting of H, alkyl, aryl, aralkyl, and haloalkyl;
      • Y is selected from the group consisting of (CH2)p, —CHRg—, —NRg—, CO and SO2, wherein Rg is selected from the group consisting of H, alkyl, haloalkyl, alkoxyalkyl, alkynyl, aryl, heteroaryl, aralkyl, hydroxy, alkoxy, and carboxyalkyl; wherein p is 0 or 1.
  • Optionally the group X-Y can contain a moiety selected from the group consisting of acyl, alkyl, sulfonyl, amino, ether, thioether, carboxamido, sulfonamido, aminosulfonyl and olefins;
      • Y3 and Y4 are independently selected from the group consisting of H, alkyl, haloalkyl, halogen, aryl, aralkyl, heteroaryl, heteroaryl, hydroxyalkyl, alkenes, and alkyne; wherein the alkyl chain may be straight or branched and optionally containing one or more heteroatoms selected from the group consisting of N, O, and S, and may further contain a sulfone, sulfonamide, nitrile, carboxamide, carboalkoxy or carboxyl group; wherein aryl and heteroaryl rings may be monocyclic or bicyclic optionally containing 1-5 heteroatoms and wherein said ring may be saturated or unsaturated, and such rings may optionally be substituted by one or more substituent selected front the group consisting of alkyl, haloalkyl, aryl, heteroaryl, halogen, alkoxyalkyl, aminoalkyl, hydroxy, nitro, alkoxy, hydroxyalkyl, thioalkyl, amino, alkylamino, arylamino, alkylsulfonamide, acyl, acylamino, alkylsulfone, sulfonamide, allyl, alkenyl, methylenedioxy, ethylenedioxy, alkynyl, carboxamide, cyano, and —(CH2)mCOR wherein m is 0-2 and R is hydroxy, alkoxy, alkyl or amino; with the proviso that when Y3 or Y4 is H, Y5 may be C or N, otherwise Y5 is C; or
      • Y3 taken together with Y4 forms a 38 membered monocyclic or a 7-11 membered bicyclic ring B,
  • Figure US20160296592A1-20161013-C00148
      • optionally containing one or more double bonds, optionally containing one or more heteroatom or functional group selected from O, NR8, S, CO or SO2, optionally substituted with one or more substituent selected from the group consisting of hydroxy, halogen, haloalkyl, alkoxy, alkyne, cyano, alkylsulfone, sulfonamide, carboalkoxy and carboxyalkyl; or
      • X taken together with Y3 forms a 3-7 membered monocyclic ring C,
  • Figure US20160296592A1-20161013-C00149
      • optionally containing one or more double bonds, optionally containing one or more heteroatom or functional group selected from O, NRg, S, CO or SO2, optionally substituted with one or more substituent selected from the group consisting of alkyl, halogen, alkoxy, haloalkyl, hydroxyalkyl, or alkoxyalkyl; and
      • Rb is X2—Rh wherein X2 is selected from the group consisting of O, S and NRj wherein Rh and Ri are independently selected from the group consisting of H, alkyl, aryl, aralkyl, acyl and alkoxyalkyl.
  • The compounds of the present invention comprise novel heteroarylalkanoic integrin antagonists.
  • The present invention relates to the following compounds:
      • 3-(3,5-ditert-butylphenyl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphthyr- idin-2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid (TFA salt);
      • 3-(3-tert-butyl-5-iodophenyl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8-napht- hyridin-2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid;
      • 3-(3-tert-butyl-5-bromophenyl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naph- thyridin-2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid;
      • 3-(5-tert-Butyl-2-hydroxyphenyl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8-na- phthyridin-2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid;
      • 3-[3,5-Ditert-butyl-2-(carboxymethoxy)phenyl]-4-{3-[3-(5,6,7,8-tetr- ahydro-1,8-naphthyridin-2-yl)propyl]-1,2,4-oxadiaol-5-yl}butanoic acid;
      • 3-(5-tert-Butyl-2-methoxyphenyl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8-na- phthyridin-2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid;
      • 3-(3,5-Ditert-butyl-4-methoxyphenyl)-4-{3-[3-(5,6,7,8-tetrahydro-1,-8-naphthyridin-2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid;
      • 3-{3-tert-Butyl-5-[2,2,2-trifluoro-1-hydroxy-1-(trifluoromethyl)eth- yl]-phenyl}-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)propyl]-1,2,-4-oxadiazol-5-yl}butanoic acid;
      • 3-(3,4-Dicholophenyl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin--2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid trifluoroacetate;
      • 3-(3-Fluoro-4-methylphenyl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphthy- ridin-2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid hydrochloride;
      • 3-(4-Phenoxyphenyl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin-2—yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid trifluoroacetate;
      • 3-(1-Benzofuran-2-yl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid trifluoroacetate;
      • 3-[4-(Benzyloxy)phenyl]4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin--2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid trifluoroacetate;
      • 3-[4-(Methylsulfonyl)phenyl]-4-[3-[3-(5,6,7,8-tetrahydro-1,8-naphth- yridin-2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid trifluoroacetate;
      • 4-{3-[3-(5,6,7,8-Tetrahydro-1,8-naphthyridin-2-yl)propyl]-1,2,4-oxa- diazol-5-yl}-3-[4-(trifluoromethoxy)phenyl]butanoic acid trifluoroacetate;
      • 3-(3-Furyl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)prop- l]-1,2,4-oxadiazol-5-yl}butanoic acid trifluoroacetate;
      • 4-{3-[3-(5,6,7,8-Tetrahydro-1,8-naphthyridin-2-yl)propyl]-1,2,4-oxa- diazol-5-yl}-3-thien-3-ylbutanoic acid trifluoroacetate;
      • 3-(2,3-Dihydro-1,4-benzodioxin-6-yl)-4-{3-[3-(5,6,7,8-tetrahydro-1,-8-naphthyridin-2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid hydrochloride;
      • 4-{3-[3-(5,6,7,8-Tetrahydro-1,8-naphthyridin-2-yl)propyl]-1,2,4-oxa- diazol-5-yl}-3-[3-(trifluoromethoxy)phenyl]butanoic acid hydrochloride;
      • 4-{3-[3-(5,6,7,8-Tetrahydro-1,8-naphthyridin-2-yl)propyl]-1,2,4-oxa- diazol-5-yl}-3-(3,4,5-trifluorophenyl)butanoic acid hydrochloride;
      • 3-(2,2-Difluoro-1,3-benzodioxol-5-yl)-4-{3-[3-(5,6,7,8-tetrahydro-1- ,8-naphthyridin-2-yl)propyl]1,2,4-oxadiazol-5-yl}butanoic acid hydrochloride;
      • 3-[3-Fluoro-5-(trifluoromethyl)phenyl]4-{3-[3-(5,6,7,8-tetrahydro-1- ,8-naphthyridin-2-yl)proply]-1,2,4-oxadiazol-5-yl}butanoic acid hydrochloride;
      • 3-(6-Methoxy-2-naphthyl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphthyrid- in-2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid hydrochloride;
      • 3-(6-Methoxypyridin-3-yl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphthyri- din-2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid;
      • 3-(4-Cyanophenyl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl-)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid;
      • 3-(3-Cyanophenyl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl-)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid;
      • 3-benzyl-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)propyl]— 1,2,4-oxadiazol-5-yl}butanoic acid trifluoroacetate;
      • 3-(4-fluoro-3-methoxyphenyl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphth- yridin-2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid trifluoroacetate;
      • 3-(3-Fluoro-5-methoxyphenyl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphth- yridin-2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid hydrochloride;
      • 3-(2-Methyl-1,3-benzothiazol-5-yl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8—naphthyridin-2-yl)propyl]-1,2,-4-oxadiazol-5-yl}butanoic acid hydrochloride;
      • 3-[2-(4-Chlorophenyl)-1,3-thiazol-5-yl]-4-{3-[3-(5,6,7,8-tetrahydro--1,8-naphthyridin-2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid hydrochloride;
      • 3-[2-(4-Methoxyphenyl)-1,3-thiazol-5-yl]4-[3-[3-(5,6,7,8-tetrahydro--1,8-naphthyridin-2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid hydrochloride;
      • 3-(2-Methyl-1,3-benzothiazol-5-yl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8—naphthyridin-2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid hydrochloride;
      • 3-[2-(4-Fluorophenyl)-1,3-thiazol-5-yl]4-{3-[3-(5,6,7,8-tetrahydro— 1,8-naphthyridin-2-yl)propyl]1,2,4-oxadiazol-5-yl}butanoic acid hydrochloride;
      • 3-[2-(3,5-Difluorophenyl)-1,3-thiazol-5-yl]-4-{3-[3-(5,6,7,8-tetrah- ydro-1,8-naphthyridin-2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid hydrochloride;
      • 3-[2-(3,4-Difluorophenyl)-1,3-thiazol-5-yl]4-{3-[3-(5,6,7,8-tetrahy- dro-1,8-naphthyridin-2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid hydrochloride;
      • 3-[2-(2-Furyl)-1,3-thiazol-5-yl]4-{3-[3-(5,6,7,8-tetrahydro-1,8-nap- hthyridin-2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid hydrochloride;
      • 3-(3,4-Dimethoxyphenyl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphthyridi- n-2-yl)propyl]1,2,4-oxadiazol-5-yl}butanoic acid trifluoroacetate;
      • 3-(3,5-Dimethoxyphenyl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphthyridi- n-2-yl)propyl]1,2,4-oxadiazol-5-yl}butanoic acid trifluoroacetate;
      • 3-(3,5-Dichlorophenyl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin--2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid trifluoroacetate;
      • 3-(3,5-Difluorophenyl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin--2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid trifluoroacetate;
      • 3-(3-Fluoro-4-methoxyphenyl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphth- yridin-2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid trifluoroacetate;
      • 4-{3-[3-(5,6,7,8-Tetrahydro-1,8-naphthyridin-2-yl)propyl]-1,2,4-oxa- diazol-5-yl}-3-[4-(trifluoromethyl)phenyl]butanoic acid trifluoroacetate;
      • 3-(2-Methyl-1,3-thiazol-5-yl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8-napht- hyridin-2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid trifluoroacetate;
      • 3-(1-Phenyl-1H-pyrazol-4-yl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphth- yridin-2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid trifluoroacetate;
      • 3-(1-Benzofuran-6-yl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin— 2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid hydrochloride;
      • 3-(2,3-dihydro-1-benzofuran-6-yl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8-n-aphthyridin-2-yl)propyl]1,2,4-oxadiazol-5-yl}butanoic acid hydrochloride;
      • 3-(1,3-Benzodioxol-5-yl)-4-(3-{3-[(pyridin-2-ylamino)methyl]phenyl}--1,2,4-oxadiazol-5-yl)butanoic acid hydrochloride;
      • 3-(7-Fluoro-1,3-benzodioxol-5-yl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8-n-aphthyridin-2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid trifluoroacetate;
      • 3-(1,3-Benzoxazol-6-yl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphthyridi- n-2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid hydrochloride;
      • 3-(3-Methyl-1,2,4-oxadiazol-5-yl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8-n-aphthyridin-2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid trifluoroacetate;
      • 3-(3-Ethyl-1,2,4-oxadiazol-5-yl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8-na- phthyridin-2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid trifluoroacetate;
      • 3-(3-Phenyl-1,2,4-oxadiazol-5-yl)-4-(3-[3-(5,6,7,8-tetrahydro-1,8-n- aphthyridin-2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid trifluoroacetate;
      • [1-Benzoyl-4-({3-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)propy-1]-1,2,4-oxadiazol-5-yl}methyl)piperdin-4-yl]acetic acid trifluoroacetate;
      • [1-Benzoyl-4-({3-[4-(pyridin-2-ylamino)butyl]-1,2,4-oxadiazol-5-yl}-methyl)piperidin-4-yl]acetic acid trifluoroacetate;
      • [1-(tert-Butoxycarbonyl)-4-({3-[3-(5,6,7,8-tetrahydro-1,8-naphthyri- din-2-yl)propyl]-1,2,4-oxadiazol-5-yl]methyl)piperidin-4-yl]acetic acid trifluoroacetate;
      • [1-(tert-Butoxycarbonyl)-4-({3-[4-(pyridin-2-ylamino)butyl]-1,2,4-o-xadiazol-5-yl{methyl)piperdin-4-yl]acetic acid trifluoroacetate;
      • 3-(4-Methylphenyl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-y-l)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid trifluoroacetate;
      • 3-(3-Chlorophenyl)-4-{-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-y-l)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid hydrochloride;
      • 3-(4-Methoxy-3-methylphenyl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphth-yridin-2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid hydrochloride;
      • 3-[4-(Methylthio)phenyl]-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphthyrid-in-2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid trifluoroacetate;
      • 3-[4-(Methylthio)phenyl]-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphthyrid-in-2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid trifluoroacetate;
      • 3-(1-Methyl-1H-indol-3-yl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphthyr-idin-2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid trifluoroacetate;
      • 3-(1,1′-Biphenyl-4-yl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin--2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid hydrochloride;
      • 3-(3-Bromophenyl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl-)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid trifluoroacetate;
      • 3-(4-Bromophenyl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl-)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid hydrochloride;
      • 3-(3-Phenoxyphenyl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphthridin-2—yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid trifluoroacetate;
      • 3-[3-Benzyloxy)phenyl]4{3-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin—2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid hydrochloride;
      • 3-(3-Bromo-4-methoxyphenyl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphthy- ridin-2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid trifluoroacetate;
      • 4-{3-[3-(5,6,7,8-Tetrahydro-1,8-naphthyridin-2-yl)propyl]-1,2,4-oxa- diazol-5-yl}-3-(3,4,5-trimethoxyphenyl)butanoic acid trifluoroacetate;
      • 3-(2-Napthyl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)pr- opyl]-1,2,4-oxadiazol-5-yl}butanoic acid hydrochloride;
      • 3-(3-Nitrophenyl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl-)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid hydrochloride;
      • 3-(3-Methylphenyl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-y-l)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid hydrochloride;
      • 3-(2-Fu ryl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)prop- yl]-1,2,4-oxadiazol-5-yl}butanoic acid hydrochloride;
      • 3-(2-Methylphenyl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-y-l)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid hydrochloride;
      • 3-(1,3-benzodioxol-5-yl)-4-{3-[3-(3,4-dihydro-2H-pyrido[3,2-b][1,4]- oxazin-6-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid, TFA;
      • 4-{3-[3-(3,4-dihydro-2H-pyrido[3,2-b][1,4]oxazin-6-yl)propyl]-1,2,4--oxadiazol-5-yl}-3-(3,5-dimethoxyphenyl)butanoic acid, TFA;
      • 3-Benzo[1,3]dioxol-5-yl-4-{3-[3-(6,7,8,9-tetrahydro-5-oxa-1,9-diaza--benzocyclohepten-2-yl)-propyl]-[1,2,4]oxadiazol-5-yl}- butryic acid;
      • 3-(3-Fluoro-4-methoxyphenyl)-4-{3-[3-(6,7,8,9-tetrahydro-5-oxa-1,9— diaza-benzocyclohepten-2-yl)-propyl]-[1,2,4]oxadiazol-5-yl}-butyric acid;
      • 3-(3,5-Difluorophenyl)-4-{3-[3-(6,7,8,9-tetrahydro-5-oxa-1,9-diaza—benzocyclohepten-2-yl)-propyl]-[1,2,4]oxadiazol-5-yl}-butyric acid;
      • 3-(3,5-Dimethoxyphenyl)-4-{3-[3-(6,7,8,9-tetrahydro-5-oxa-1,9-diaza--benzocyclohepten-2-yl)-propyl]-[1,2,4]oxadiazol-5-yl}-butryic acid;
      • 3-(2-Methylbenzothiazol-5-yl)-4-{3-[3-(6,7,8,9-tetrahydro-5-oxa-1,9--diaza-benzocyclohepten-2-yl)-propyl]-[1,2,4]oxadiazol-5-yl}- butryic acid;
      • 3-(1,3-benzodioxol-5-yl)-4-{3-[3-(1,2,3,5-tetrahydropyrido[2,3-e][1-,4]oxazepin-8-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid, TFA;
      • 3-(3,5-dimethoxyphenyl)-4-{3-[3-(1,2,3,5-tetrahydropyrido[2,3-e][1,-4]oxazepin-8-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid, TFA;
      • 3-(1,3-Benzodioxol-5-yl)-4-(3-{3-[6-(methylamino)pyridin-2-yl]propy-l}-1,2,4-oxadiazol-5-yl)butanoic acid hydrochloride;
      • 3-(3-Fluorophenyl)-4-(3-{3-[6-(methylamino)pyridin-2-yl]propyl}-1,2- ,4-oxadiazol-5-yl)butanoic acid trifluoroacetate;
      • 3-(1,3-benzodioxol-5-yl)-4-(3-3-[6-(ethylamino)pyridin-2-yl]propyl— 1,2,4-oxadiazol-5-yl)butanoic acid trifluoroacetate;
      • 3-(3-Fuorophenyl)-4-(3-{3-[6-(methylamino) pyridin-2-yl]propyl-1,2,-4-oxadiazol-5-yl)butanoic acid trifluoroacetate;
      • 3-(1,3-Benzodioxol-5-yl)-4-(3-{4-[(4-methylpyridin-2-yl)amino]butyl- }-1,2,4-oxadiazol-5-yl)butanoic acid;
      • 3-(1,3-benzodixoxol-5-yl)-4-(3-{4-[(6-methylpyridin-2-yl)amino]butyl- }-1,2,4-oxadiazol-5-yl)butanoic acid;
      • (2-{6-[2-(5,6,7,8-Tetrahydro-1,8-naphthyridin-2-yl)ethoxy]pyridin-3--yl}cyclopropyl)acetic acid;
      • 3-Methyl-4-{6-[2-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)ethoxy]p- yridin-3-yl}butanoic acid;
      • 3-(1,3-benzodioxol-5-yl)-4-{5-[3-(5,6,7,8-tetrahydro-1,8-naphthyrid- in-2-yl)propyl]-1,3,4-oxadiazol-2-yl}butanoic acid trifluoroacetate;
      • 3-(3-fluorophenyl)-4-{5-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-y-l)propyl]-1,3,4-oxadiazol-2-yl}butanoic acid trifluoroacetate;
      • 3-(3-Fluoro-4-methoxyphenyl)-4-{5-[3-(5,6,7,8-tetrahydro-1,8-naphth- yridin-2-yl)propyl]-1,3,4-oxadiazol-2-yl}butanoic acid trifluoroacetate;
      • 3-(3,5-Dimethoxyphenyl)-4-{5-[3-(5,6,7,8-tetrahydro-1,8-naphthyridi- n-2-yl)propyl]-1,3,4-oxadiazol-2-yl}butanoic acid trifluoroacetate;
      • 3-(2-Methyl-1,3-thiazol-5-yl)-4-{5-[3-(5,6,7,8-tetrahydro-1,8-napht- hyridin-2-yl)propyl]-1,3,4-oxadiazol-2-yl}butanoic acid trifluoroacetate;
      • 3-(4-Fluorophenyl)-4-{5-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-y-l)propyl]-1,3,4-oxadiazol-2-yl}butanoic acid trifluoroacetate;
      • 3-(3,5-Difluoropehnyl)-4-{5-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin--2-yl)propyl]-1,3,4-oxadiazol-2-yl}butanoic acid trifluoroacetate;
      • 3-(3,5-Difluorophenyl)-4-{5-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin--2-yl)propyl]1,3,4-thiadiazol-2-yl}butanoic acid trifluoroacetate;
      • 3-(4-Fluorophenyl)-4{5-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl-)propyl]-1,3,4-thiadiazol-2-yl}butanoic acid trifluoroacetate;
      • 3-(2-Methyl-1,3-thiazol-5-yl)-4-{5-[3-(5,6,7,8-tetrahydro-1,8-napht- hyridin-2-yl)propyl]-1,3,4-thiadiazol-2-yl}butanoic acid trifluoroacetate;
      • 3-(1,3-Benzodioxol-5-yl)-4-{5-[3-(5,6,7,8-tetrahydro-1,8-naphthyrid- in-2-yl)propyl]-1,3,4-thiadiazol-2-yl}butanoic acid trifluoroacetate;
      • 3-(1,3-benzodioxol-5-yl)-4-{3-[2-(5,6,7,8-tetrahydro-1,8-naphthyrid- in-2-yl)ethoxy]isoxazol-5-yl}butanoic acid;
      • 3-(1,3-benzodioxol-5-yl)-4-{5-[3-(5,6,7,8-tetrahydro-1,8-naphthyrid- in-2-yl)propyl]-2H-tetraazol-2-yl}butanoic acid;
      • 3-(1,3-benzodioxol-5-yl)-4-{5-[3-(5,6,7,8-tetrahydro-1,8-naphthyrid- in-2-yl)propyl]-1H-tetraazol-1-yl}butanoic acid;
      • 3-(1,3-benzodioxol-5-yl)-4-{3-[2-(5,6,7,8-tetrahydro-1,8-naphthyrid- in-2-yl)ethoxy]-1H-pyrazol-5-yl}butanoic acid;
      • 3-(1,3-benzodioxol-5-yl)-4-{3-[3-(4,5-dihydro-1H-imidazol-2-ylamino-)propoxy]isoxazol-5-yl}butanoic acid;
      • 3-[2-(4-chlorophenyl)-1,3-thiazol-5-yl]4-{3-[2-(5,6,7,8-tetrahydro— 1,8-naphthyridin-2-yl)ethoxy]isoxazol-5-yl}butanoic acid;
      • 3-Benzo[1,3]dioxol-5-yl-4-{3-[2-(6,7,8,9-tetrahydro-5-oxa-1,9-diaza--benzocyclohepten-2-yl)-ethoxy]-isoxazol-5-yl}-butyric acid;
      • 3-Benzo[1,3]dioxol-5-yl-4-{3-oxo-2-[2-(6,7,8,9-tetrahydro-5-oxa-1,9--diaza-benzocyclohepten-2-yl)-ethyl]-2,3-dihydro-isoxazol-5- yl}-butyric acid;
      • 3-(1,3-benzodioxol-5-yl)-4-{3-[2-(3,4-dihydro-2H-pyrido[3,2-b][1,4]- oxazin-6-yl)ethoxy]isoxazol-5-yl}butanoic acid, TFA;
      • 3-(1,3-benzodioxol-5-yl)-4-{2-[2-(3,4-dihydro-2H-pyrido[3,2-b][1,4]-oxazin-6-yl)ethyl]-3-oxo-2,3-dihydroisoxazol-5- yl}butanoic acid, TFA;
      • 3-(1,3-benzodioxol-5-yl)-4-{3-[2-(1,2,3,5-tetrahydropyrido[2,3-e][1-,4]oxazepin-8-yl)ethoxy]isoxazol-5-yl}butanoic acid, TFA;
      • 3-(1,3-benzodioxol-5-yl)-4-{3-oxo-2-[2-(1,2,3,5-tetrahydropyrido[2,-3-e][1,4]oxazepin-8-yl)ethyl]-2,3-dihydroisoxazol-5-yl}butanoic acid, TFA;
      • 3-(1,3-benzodioxol-5-yl)-4-(3-{2-[5-(methoxymethyl)-6-(methylamino)- pyridin-2-yl]ethoxy}isoxazol-5-yl)butanoic acid, TFA;
      • 3-(1,3-Benzodioxol-5-yl)-4-{3-[3-(5,5-dimethyl-5,6,7,8-tetrahydro-1- ,8-naphthyridin-2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid;
      • 3-(1,3-benzodioxol-5-yl-4-{3-[3-(1-methyl-1,2,3,4-tetrahydropyrido- [2,3-b]pyrazin-6-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid;
      • 3-(2-methyl-1,3-benzothizol-5-yl)-4-{3-[3-(1-methyl-1,2,3,4-tetrah-ydropyrido[2,3-b]pyrazin-6-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid;
      • 3-(3-fluoro-4-methoxyphenyl)-4-{3-[3-(1-methyl-1,2,3,4-tetrahydropy- rido[2,3-b]pyrazin-6-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid;
      • 3-(6-methoxypyridin-3-yl)-4-{3-[3-(1-methyl-1,2,3,4-tetrahydropyrid-o[2,3-b]pyrazin-6-yl)propyl]-1,2,4-oxydiazol-5-yl}butanoic acid;
      • 3-(1,3-benzodioxol-5-yl)-4-(3-{[2-(5,6,7,8-tetrahydro-1,8-naphthyri- din-2-yl)ethyl]thio}-1H-1,2,4-triazol-5-yl)butanoic acid;
      • 3-(1,3-benzodioxol-5-yl)-4-(1-methyl-5-{[2-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)ethyl]thio}-1H-1,2,4-triazol-3-yl)butanoic acid;
      • 3-(1,3-benzodioxol-5-yl)-4-(4-methyl-5-{[2-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)ethyl]thio}4H-1,2,4-triazol-3-yl)butanoic acid;
      • 3-(1,3-benzodioxol-5-yl)-4-{3-[2-(1-methyl-1,2,3,4-tetrahydropyrido- [2,3-b]pyrazin-6-yl)ethoxy]isoxazol-5-yl}butanoic acid;
      • 3-(1,3-benzodioxol-5-yl)-4-(3-{2-[6-(methylamino)pyridine-2-yl]ethox-y}isoxazol-5-yl)butanoic acid; and
      • 3-(6-methoxypyridin-3-yl)-4-{3-[2-(5,6,7,8-tetrahydro-1,8-naphthyri- din-2-yl)ethoxy]isoxazol-5-yl}butanoic acid.
  • In another embodiment, the present invention may also include the following compounds:
      • 3-methyl-4-(3-{3-[(pyridin-2-ylamino)methyl]phenyl}-1,2,4-oxadiazol--5-yl)butanoic acid;
      • 3-methyl-4-(3-{4-[(pyridin-2-ylamino)methyl]phenyl}-1,2,4-oxadiazol--5-yl)butanoic acid;
      • 3,3-dimethyl-4-{4-[4-(pyridin-2-ylamino)butyl]-1,3-thiazol-2-yl}butanoic acid;
      • [1-({3-[4-(pyridin-2-ylamino)butyl]-1,2,4-oxadiazol-5-yl}methyl)cyclo-pentyl]-acetic acid;
      • 4-phenyl-4-{3-[4-(pyridin-2-ylamino)butyl]-1,2,4-oxadiazol-5-yl}-butanoic acid;
      • 2-phenyl-4-{3-[4-(pyridin-2-ylamino)butyl]-1,2,4-oxadiazol-5-yl}-butanoic acid;
      • 3,3-dimethyl-4-{3-[2-(2-methyl-5,6,7,8-tetrahydro-1,8-naphthyridin-3-yl)ethyl]1,2,4-oxadiazol-5-yl}butanoic acid;
      • [1-({3-[2-(2-methyl-5,6,7,8-tetrahydro-1,8-naphthyridin-3-yl)ethyl--1,2,4-oxadiazol-5-yl}methyl)cyclopentyl]acetic acid;
      • 4-{3-[2-(2-methyl-5,6,7,8-tetrahydro-1,8-naphthyridin-3-yl)ethyl]-1-,2,4-oxadiazol-5-yl}4-phenylbutanoic acid;
      • 4-{3-[2-(2-methyl-5,6,7,8-tetrahydro-1,8-naphthyridin-3-yl)ethyl]-1-,2,4-oxadiazol-5-yl}2-phenylbutanoic acid;
      • 4-{3-[2-(2-methyl-5,6,7,8-tetrahydro-1,8-naphthyridin-3-yl)ethyl]-1-,2,4-oxadiazol-5-yl}4-phenylbutanoic acid;
      • 3,3-dimethyl-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid;
      • [1-({3-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)propyl]-1,2,4-oxadiazol-5-yl}methyl)cyclopentyl]acetic acid;
      • 4-phenyl-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid;
      • 2-phenyl-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid;
      • 3-(1,3-benzodioxol-5-yl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid;
      • 3-(1,3-benzodioxol-5-yl)-4-{3-[4(pyridine-2-ylamino)butyl]-1,2,4-oxadiazol-5-yl}butanoic acid;
      • 3-quinolin-3-yl-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)-propyl]-1,2,4-oxadiazol-5-yl}butanoic acid;
      • 3-quinolin-3-yl-4-{3-[4-(pyridine-2-ylamino)butyl]-1,2,4-oxadiazol-5--yl}-butanoic acid;
      • 3-(3-methoxyphenyl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid;
      • 3-(3-methoxyphenyl)-4-{3-[4-(pyridine-2-ylamino)butyl]-1,2,4-oxadiazol-5-yl}butanoic acid;
      • 3-(3-methoxyphenyl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid;
      • 3-(3-methoxyphenyl)-4-{3-[4-(pyridine-2-ylamino)butyl]-1,2,4-oxadiazol-5-yl}butanoic acid;
      • 3-(3-fluorophenyl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid;
      • 3-(3-fluorophenyl)-4-{3-[4-(pyridine-2-ylamino)butyl]-1,2,4-oxadiazol-1-5-yl}butanoic acid;
      • 3-(4-fluorophenyl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid;
      • 3-(4-fluorophenyl)-4-{3-[4-(pyridine-2-ylamino)butyl]-1,2,4-oxadiazol-1-5-yl}butanoic acid;
      • 4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)propyl]-1,2,4-oxadiazol-5-yl}-3-[3-(trifluoromethyl)phenyl]butanoic acid;
      • 4-{3-[4-(pyridine-2-ylamino)butyl]-1,2,4-oxadiazol-5-yl}-3-[3-(trifluoro-methyl)-phenyl]butanoic acid;
      • 3-(3-hydroxyphenyl)-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid;
      • 3-(3-hydroxyphenyl)-4-{3-[4-(pyridin-2-ylamino)butyl]-1,2,4-oxadiazol-5-yl}butanoic acid;
      • 3-pyridin-3-yl-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)-propyl]-1,2,4-oxadiazol-5-yl}butanoic acid;
      • 3-pyridin-3-yl-4-{3-[4-(pyridin-2-ylamino)butyl]-1,2,4-oxadiazol-5-yl}-butanoic acid;
      • 3-phenyl-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)-propyl]--1,2,4-oxadiazol-5-yl}butanoic acid;
      • 3-phenyl-4-{3-[4-(pyridin-2-ylamino)butyl]-1,2,4-oxadiazol-5-yl}butanoic acid;
      • 3-methyl-3-({3-[4-(pyridin-2-ylamino)butyl]-1,2,4-oxadiazol-5-yl}-methyl)pentanoic acid;
      • [1-({3-[4-(pyridin-2-ylamino)butyl]-1,2,4-oxadiazol-5-yl}methyl-cyclohexyl]acetic acid;
      • 3-methyl-3-{3-[4-(pyridin-2-ylamino)butyl]-1,2,4-oxadiazol-5-yl}-methyl)-hexanoic acid;
      • 3,4-dimethyl-3-({3-[4-(pyridin-2-ylamino)butyl]-1,2,4-oxadiazol-5-yl}methyl)-pentanoic acid;
      • 3-ethyl-3-({3-[4-(pyridin-2-ylamino)butyl]-1,2,4-oxadiazol-5-yl}methyl)-pentanoic acid;
      • 4-{3-[4-(pyridin-2-ylamino)butyl]-1,2,4-oxadiazol-5-yl}butanoic acid;
      • 3-methyl-3-phenyl-4-{3-[4-(pyridin-2-ylamino)butyl]-1,2,4-oxadiazol--5-yl}butanoic acid;
      • 3-Methyl-3-{3-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)propyl-]-[1,2,4]oxadiazol-5-ylmethyl}-pentanoic acid;
      • 3-Methyl-3-{3-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)propyl-]-[1,2,4]oxadiazol-5-ylmethyl}-hexanoic acid;
      • 3,4-Dimethyl-3-{3-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)propyl]-[1,2,4]oxadiazol-5-ylmethyl}-pentanoic acid;
      • 3-Ethyl-3-{3-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)propyl]--[1,2,4]oxadiazol-5-ylmethyl}-pentanoic acid;
      • 3-Methyl-3-phenyl-4-(3-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)propyl]-[1,2,4]oxadiazol-5-yl}-butyric acid;
      • 3-Phenyl-3-{3-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)propyl-]-[1,2,4]oxadiazol-5-ylmethyl}-pentanoic acid;
      • 3-Phenyl-3-{3-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)propyl-]-[1,2,4]oxadiazol-5-ylmethyl}-hexanoic acid;
      • 4-{3-[3-(5,6,7,8-Tetrahydro-[1,8]naphthyridin-2-yl)-propyl]-[1,2,4]-oxa-diazol-5-yl)-butyric acid;
      • 3-Methyl-3-pyridin-3-yl-4-{3-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propyl]-[1,2,4]oxadiazol-5-yl}-butyric acid;
      • (1-Acetyl-4-{3-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)propyl]-[1,2,4]oxadiazol-5-ylmethyl}-piperidin-4-yl)-acetic acid;
      • 1-{3-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)propyl]-[1,2,4]oxadiazol-5-ylmethyl}-cyclohexyl)-acetic acid;
      • 3-Methyl-3-pyridin-3-yl-4-{3-[4-(pyridin-2-ylamino)butyl]-[1,2,4]oxadiazol-5-yl}-butyric acid;
      • 4-(benzyloxy)-3-({3-[4-(pyridin-2-ylamino)butyl]-1,2,4-oxadiazol-5-yl}methyl)-butanoic acid;
      • 4-[4-(N-pyridin-2-yl-beta-alanyl)piperazin-1-yl]butanoic acid;
      • 4-{4-[3-(pyridin-2-ylamino)propyl]piperazin-1-yl}butanoic acid;
      • 2-methyl-6-[3(2-pyridylamino)propoxy)-3-pyridinebutanoic acid;
      • β,β-dimethyl-3-[5-(2-pyridinylamino)pentyl]-1,2,4-oxadiazole-5-butanoic acid;
      • β,β-dimethyl-3-[4-(2-pyridinylamino)butyl]-1,2,4-oxadiazole-5-butanoic acid;
      • β,β-dimethyl-3-[[[2-(2-pyridinylamino)ethyl]thio]methyl-1,2,4-oxadiazole-5-butanoic acid;
      • 4-Carboxymethyl-4-{3-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)--propyl]-[1,2,4]oxadiazol-5-ylmethyl}-piperidine-1-carboxylic acid tert-butyl ester;
      • (1-Benzoyl-4-{3-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propyl]-[1,2,4]oxadiazol-5-ylmethyl}-piperidin-4-yl)-acetic acid;
      • [4-{3-[3-(5,6,7,8-Tetrahydro-[1,8]naphthyridin-2-yl)-propyl]-[1,2,4-]oxadiazol-5-ylmethyl}-1-(2,2,2-trifluoroacetyl)piperidine-4-yl]-acetic acid;
      • 4-(phenylthio)3-({3-[4-(pyridine-2-ylamino)butyl]-1,2,4-oxadiazol-5--yl}methyl)butanoic acid;
      • 4-(phenylthio)3-({3-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)propyl]-1,2,4-oxadiazol-5-yl}methyl)butanoic acid;
      • 3-methyl-4-{3-[4-(pyridin-2-ylamino)butyl]-1,2,4-oxadiazol-5-yl}butanoic acid hydrochloride;
      • 3-methyl-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)propyl]-1,2,4-oxadiazol-5-yl}butanoic acid;
      • ((1S,2R)-2-(3-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)propyl]-1,2,4-oxadiazol-5-yl}cyclepropyl)acetic acid;
      • ((1S,2S)-2-(3-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)propyl]-1,2,4-oxadiazol-5-yl}cyclepropyl)acetic acid;
      • 3-Pyridin-3-yl-4-{5-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)-- propyl]4H-[1,2,4]triazol-3-yl}butyric acid;
      • 3-Benzo[1,3]dioxol-5-yl-4-{5-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propyl]-tetrazol-2-yl}-butyric acid;
      • (2-{5-[3-(5,6,7,8-Tetrahydro-1,8-naphthyridin-2-yl)-propyl]-[1,3,4]oxa-diazol-2-yl}-cyclopropyl)-acetic acid;
      • 3-Phenyl-4-{5-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propyl-]-[1,3,4]oxadiazol-2-yl)butyric acid;
      • 3-(2,3-Dihydro-benzofuran-6-yl)-4-{5-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propyl]-[1,3,4]oxadiazol-2-yl}butyric acid;
      • 3-(3-Fluoro-phenyl)-4-(5-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin-2--yl)- propyl]-[1,3,4]oxadiazol-2-yl}-butyric acid;
      • 3-Benzo-[1,3]dioxol-5-yl-4-{5-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)- propyl]-[1,3,4]oxadiazol-2-yl}butyric acid;
      • (2-(2-[3-(5,6,7,8-Tetrahydro-[1,8]naphthyridin-2-yl)-propyl]-2H-tetrazol-5-yl}-cyclopropyl)acetic acid;
      • 3-Phenyl-4-{2-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propyl-]-2H-tetrazol-5-yl}butyric acid;
      • 3-(2,3-Dihydro-benzofuran-6-yl)-4-{2-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-2H-tetrazol-5-yl}-butyric acid;
      • 3-(2,3-Dihydro-benzofuran-6-yl)-4-{2-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-2H-tetrazol-5-yl}-butyric acid;
      • 3-(3-Fluoro-phenyl)-4-{2-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2--yl)-propyl]-2H-tetrazol-5-yl}-butyric acid;
      • 3-Pyridin-3-yl-4-(2-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propyl]-2H-tetrazol-5-yl}-butyric acid;
      • 3-Benzo[1,3]dioxol-5-yl-4-(2-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propyl]-2H-tetrazol-5-yl}-butyric acid;
      • (2-{5-[3-(5,6,7,8-Tetrahydro-[1,8]naphthyridin-2-yl)-propyl]-isoxazol-3-yl}-cyclopropyl)-acetic acid;
      • 3-Phenyl-4-(5-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propyl-]-isoxazol-3-yl}-butyric acid;
      • 3-(2,3-Dihydro-benzofuran-6-yl)-4-(5-[3-(5,6,7,8-tetrahydro-[1,8]-naphthyridin-2-yl)-propyl]-isoxazol-3-yl-butyric acid;
      • 3-(3-Fluoro-phenyl)-4-(5-[3-(5,6,7,8-tetrahydro-[1,8]-naphthyridin-2--yl)- propyl]-isoxazol-3-yl}-butyric acid;
      • 3-Pyridin-3-yl-4-{5-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propyl]-isoxazol-3-yl}butyric acid;
      • 3-Benzo[1,3]dioxyl-5-yl-4-{5-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)- propyl]-isoxazol-3-yl}-butyric acid;
      • 3-(2,3-Dihydro-benzofuran-6-yl)-4-(3-[3-(5,6,7,8-tetrahydro-[1,8]-naphthyridin-2-yl)-propyl]-isoxazol-5-yl-butyric acid;
      • 3-(3-Fluoro-phenyl)-4-{3-[3-(5,6,7,8-tetrahydro-[1,8]-naphthyridin-2--yl)-propyl]-isoxazol-5-yl}-butyric acid;
      • 3-Pyridin-3-yl-4-{3-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propyl]-isoxazol-5-yl}butyric acid;
      • 3-Benzo-[1,3]dioxol-5-yl-4-{3-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)- propyl]-isoxazol-5-yl}-butyric acid;
      • 3-Phenyl-4-{5-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propyl-]-1H-pyrazol-3-yl}butyric acid;
      • 3-(2,3-Dihydro-benzofuran-6-yl)-4-{5-[3-(5,6,7,8-tetrahydro-[1,8]-naphthyridin-2-yl)-propyl]-1H-pyrazol-3-yl-butyric acid;
      • 3-(3-Fluoro-phenyl)-4-{5-[3-(5,6,7,8-tetrahydro-[1,8]-naphthyridin-2--yl propyl]-1H-pyrazol-3-yl}-butyric acid;
      • 3-Pyridin-3-yl-4-{5-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propyl]-1H-pyrazol-3-yl}butyric acid;
      • 3-Benzo-[1,3]dioxol-5-yl-4-{5-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)- propyl]-1H-pyrazol-3-yl}-butyric acid;
      • (2-{3-[3-(5,6,7,8-Tetrahydro-[1,8]naphthyridin-2-yl)-propyl]-isoxazol-5-yl}-cyclopropyl)-acetic acid;
      • (2-{5-[3-(5,6,7,8-Tetrahydro-[1,8]naphthyridin-2-yl)-propyl]-1H-pyrazol-3-yl}-cyclopropyl)-acetic acid;
      • (2-{4-[3-(5,6,7,8-Tetrahydro-[1,8]naphthyridin-2-yl)-propyl]-thiazo-1-2-yl}-cyclopropyl)-acetic acid; 3-Phenyl-4-{4-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propyl]-thiazol-2-yl}-butyric acid;
      • 3-(2,3-Dihydro-benzofuran-6-yl)-4-{4-[3-(5,6,7,8-tetrahydro-[1,8]-naphthyridin-2-yl)-propyl-thiazol-2-yl}-butyric acid;
      • 3-(3-Fluoro-phenyl)-4-{4-[3-(5,6,7,8-tetrahydro-[1,8]-naphthyridin-2--yl)-propyl)-thiazol-2-yl)-butyric acid;
      • 3-Pyridin-3-yl-4-{4-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propyl]-thiazol-2-yl}butyric acid;
      • 3-Benzo[1,3]dioxol-5-yl-4-{4-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propyl]-thiazol-2-yl}-butyric acid;
      • 3-Phenyl-4-{3-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propyl- }-pyrazol-1-yl}-butyric acid;
      • 3-(2,3-Dihydro-benzofuran-6-yl)-4-{3-[3-(5,6,7,8-tetrahydro-[1,8]-naphthyridin-2-yl)-propyl]-pyrazol-1-yl}-butyric acid;
      • 3-(3-Fluoro-phenyl)-4-{3-[3-(5,6,7,8-tetrahydro-[1,8]-naphthyridin-2--yl)-propyl]-pyrazol-1-yl}-butyric acid;
      • 3-Pyridin-3-yl-4-{3-[3-(5,6,7,8-tetrahydro-[1,8]-naphthyridin-2-yl)-propyl]-pyrazol-1-yl}-butyric acid;
      • 3-Benzo[1,3]dioxol-5-yl-4-{3-[3-(5,6,7,8-tetrahydro-[1,8]naphthyrid-in-2-yl)-propyl]-pyrazol-1-yl)-butyric acid;
      • 3-Phenyl-4-{4-[3-(5,6,7,8-tetrahydro-[1,8]-naphthyridin-2-yl)-propyl-]-imidazol-1-yl}-butyric acid;
      • 3-(2,3-Dihydro-benzofuran-6-yl)-4-{4-[3-(5,6,7,8-tetrahydro-[1,8]-naphthyridin-2-yl)-propyl]-imidazol-1-yl}-butyric acid;
      • 3-(3-Fluoro-phenyl)-4-{4-[3-(5,6,7,8-tetrahydro-[1,8]-naphthyridin-2--yl)-propyl]-imidazol-1-yl}-butyric acid;
      • 3-Pyridin-3-yl-4-{4-[3-(5,6,7,8-tetrahydro-[1,8]-naphthyridin-2-yl)-propyl]-imidazol-1-yl}-butyric acid;
      • 3-Benzo[1,3]dioxol-5-yl-4-{4-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propyl]-imidazol-1-yl}-butyric acid3-Phenyl-4-{3-[2-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-ethoxy]-isoxazol-5-yl}-butyric acid;
      • 3-(2,3-Dihydro-benzofuran-6-yl)-4-{3-[2-(5,6,7,8-tetrahydro-[1,8]-naphthyridin-2-yl)-ethoxy]-isoxazol-5-yl}-butyric acid;
      • 3-(3-Fluoro-phenyl)-4-{3-[2-(5,6,7,8-tetrahydro-[1,8]-naphthyridin-2--yl)-ethoxy]-isoxazol-5-yl}-butyric acid;
      • 3-Pyridin-3-yl-4-{3-[2-(5,6,7,8-tetrahydro-[1,8]-naphthyridin-2-yl)-ethoxy]-isoxazol-5-yl}-butyric acid;
      • 3-Benzol[1,3]dioxol-5-yl-4-{3-[2-(5,6,7,8-tetrahydro-[1,8]naphthyrid-in-2-yl)-ethoxy]-isoxazol-5-yl)-butyric acid;
      • 3-(3-Fluoro-phenyl)-4-{5-[2-(5,6,7,8-tetrahydro-[1,8]-naphthyridin-2--yl)-ethoxy]-2H-pyrazol-3-yl}-butyric acid;
      • 3-(2,3-Dihydro-benzofuran-6-yl)-4-{5-[2-(5,6,7,8-tetrahydro-[1,8]-naphthyridin-2-yl)-ethoxy]-2H-pyrazol-3-yl}-butyric acid;
      • 3-Phenyl-4-{5-[2-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-ethoxy-]-2H-pyrazol-3-yl}-butyric acid;
      • 3-Pyridin-3-yl-4-{5-[2-(5,6,7,8-tetrahydro-[1,8]-naphthyridin-2-yl)-ethoxy]-2H-pyrazol-3-yl}-butyric acid;
      • 3-Benzo[1,3]dioxol-5-yl-4-{5-[2-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-ethoxy]-2H-pyrazol-3-yl)-butyric acid;
      • 3-Phenyl-4-[4-(2-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propiony-1)-imidazol-1-yl]-butyric acid;
      • 3-(2,3-Dihydro-benzofuran-6-yl)-4-[4-(3-5,6,7,8-tetrahydro-[1,8]-naphthyridin-2-yl-propionyl imidazole-1-yl]-butyric acid;
      • 3-(3-Fluoro-phenyl)-4-[4-(3-5,6,7,8-tetrahydro-[1,8]-naphthyridin-2--yl-propionyl)-imidzol-1-yl]-butyric acid;
      • 3-Pyridin-3-yl-4-[4-(3-5,6,7,8-tetrahydro-[1,8]-naphthyridin-2-yl-propionyl)-imidazol-1-yl]-butyric acid;
      • 3-Benzo[1,3]dioxol-5-yl-4-[4-(3-5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl-propionyl)-imidazol-1-yl]-butyric acid;
      • 4-{4-[1-Hydroxy-3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propyl]-imidazol-1-yl}-3-phenyl-butyric acid;
      • 3-(2,3-Dihydro-benzofuran-6-yl)-4-{4-[1-hydroxy-3-(5,6,7,8-tetrahydro-[1,8]-naphthyridin-2-yl)-propyl]-imidazol-1-yl}-butyric acid;
      • 3-(3-Fluoro-phenyl)-4-{4-[1-hydroxy-3-(5,6,7,8-tetrahydro-[1,8]-naphthyridin-2-yl]-propyl]-imidzol-1-yl}-butyric acid;
      • 4-{4-[1-Hydroxy-3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propyl]-imidazol-1-yl}-3-pyridin-3-yl-butyric acid;
      • 4-{4-[1-Hydroxy-3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propyl]-imidazol-1-yl}-3-phenyl-butyric acid;
      • 4-{4-[1-Hydroxy-3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-prop-1-ynyl]-imidazol-1-yl}-3-phenyl-butyric acid;
      • 3-(2,3-Dihydro-benzofuran-6-yl)-4-{4-[3-hydroxy-3-(5,6,7,8-tetrahydro-[1,8]-naphthyridin-2-yl)-prop-1-ynyl]-imidazol-1-yl}-butyric acid;
      • 3-(3-Fluoro-phenyl)-4-{4-(3-hydroxy-3-(5,6,7,8-tetrahydro-[1,8]-naphthyridin-2-yl)-prop-1-ynyl]-imidzol-1-yl]-butyric acid;
      • 4-{4-[3-Hydroxy-3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-prop-1-ynyl]-imidazol-1-yl}-3-pyridin-3-yl-butyric acid;
      • 3-(2,3-Dihydro-benzofuran-6-yl)-4-{2-[3-(5,6,7,8-tetrahydro-[1,8]-naphthyridin-2-yl)-propyl]-2H-tetrazol-5-yl}-butyric acid;
      • 3-(3-Fluoro-phenyl)-4-{2-[3-(5,6,7,8-tetrahydro-[1,8]-naphthyridin-2--yl)-propyl]-2H-tetrazol-5-yl}-butyric acid;
      • 3-Pyridin-3-yl-4-{2-[3-(5,6,7,8-tetrahydro-[1,8]-naphthyridin-2-yl)-propyl]-2H-tetrazol-5-yl}-butyric acid;
      • 3-Benzo[1,3]dioxol-5-yl-4-{2-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propyl]-2H-tetrazol-5-yl}-butyric acid;
      • (2-{5-[3-(5,6,7,8-Tetrahydro-[1,8]naphthyridin-2-yl)-propyl]-isoxazol-3-yl}-cyclopropyl)-acetic acid;
      • 3-Phenyl-4-{5-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propyl-]-isoxazol-3-yl}-butyric acid;
      • 3-(2,3-Dihydro-benzofuran-6-yl)-4-{5-[3-(5,6,7,8-tetrahydro-[1,8]-naphthyridin-2-yl)-propyl]-isoxazol-3-yl}-butyric acid;
      • 3-(3-Fluoro-phenyl)-4-{5-[3-(5,6,7,8-tetrahydro-[1,8]-naphthyridin-2--yl)-propyl]-isoxazol-3-yl}-butyric acid;
      • 3-Pyridin-3-yl-4-{5-[3-(5,6,7,8-tetrahydro-[1,8]-naphthyridin-2-yl)-propyl]-isoxazol-3-yl}-butyric acid;
      • 3-Benzo[1,3]dioxol-5-yl-4-{5-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propyl]-isoxazol-3-yl}-butyric acid;
      • 3-(2,3-Dihydro-benzofuran-6-yl)-4-{3-[3-(5,6,7,8-tetrahydro-[1,8]-naphthyridin-2-yl)-propyl]-isoxazol-5-yl}-butyric acid;
      • 3-(3-Fluoro-phenyl)-4-{3-(3-(5,6,7,8-tetrahydro-[1,8]-naphthyridin-2--yl)-propyl]-isoxazol-5-yl}-butyric acid;
      • 3-Pyridin-3-yl-4-{3-[3-(5,6,7,8-tetrahydro-[1,8]-naphthyridin-2-yl)-propyl]-isoxazol-5-yl}-butyric acid;
      • 3-Benzo[1,3]dioxol-5-yl-4-{3-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propyl]-isoxazol-5-yl}-butryic acid;
      • 3-Phenyl-4-{5-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propyl-]-1H-pyrazol-3-yl}-butryic acid;
      • 3-(2,3-Dihydro-benzofuran-6-yl)-4-{5-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propyl]-1H-pyrazol-3-yl≢-butryic acid;
      • 3-(3-Fluoro-phenyl)-4-{5-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2--yl)-propyl]-1H-pyrazol-3-yl}-butryic acid;
      • 3-Pyridin-3-yl-4-{5-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)—propyl]-1H-pyrazol-3-yl}-butyric acid;
      • 3-Benzo[1,3]dioxol-5-yl-4-{5-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propyl]-1H-pyrazol-3-yl}-butryic acid;
      • (2-{3-[3-(5,6,7,8-Tetrahydro-[1,8]naphthyridin-2-yl}-propyl]-isoxazol-5-yl}-cyclopropyl)-acetic acid;
      • 2-{5-[3-(5,6,7,8-Tetrahydro-[1,8]naphthyridin-2-yl}-propyl]-1H-pyrazol-3-yl}-cyclopropyl)-acetic acid;
      • 2-{4-[3-(5,6,7,8-Tetrahydro-[1,8]naphthyridin-2-yl}-propyl]-thiazo-1-2-yl}-cyclopropyl)-acetic acid;
      • 3-Phenyl-4-{-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propyl]--thiazol-2-yl}butyric acid;
      • 3-(2,3-Dihydro-benzofuran-6-yl)-4-{4-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propyl]-thiazol-2-yl}-butyric acid;
      • 3-(3-Fluoro-phenyl)-4-{4-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2- yl)-propyl]-thiazol-2-yl}-butyric acid;
      • 3-Pyridin-3-yl-4-{4-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)—propyl]-thiazol-2-yl}-butyric acid;
      • 3-Benzo[1,3]dioxol-5-yl-4-{4-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propyl]-thiazol-2-yl}-butyric acid;
      • 3-Phenyl-4-{3-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propyl- ]-pyrazol-1-yl}-butryic acid;
      • 3-(2,3-Dihydro-benzofuran-6-yl)-4-≡3-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propyl]-pyrazol-1-yl}-butyric acid;
      • 3-(3-Fluoro-phenyl)-4-{3-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2--yl)-propyl]-pyrazol-1-yl≢-butryic acid;
      • 3-Pyridin-3-yl-4-{3-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)—propyl]-pyrazol-1-yl}-butyric acid;
      • 3-Pyridin-3-yl-4-{3-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)—propyl]-pyrazol-1-yl}-butyric acid;
      • 3-Benzo[1,3]dioxol-5-yl-4-{3-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propyl]-pyrazol-1-yl}-butyric acid;
      • 3-Phenyl-4-{4-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propyl- ]-imidazol-1-yl}-butryic acid;
      • 3-(2,3-Dihydro-benzofuran-6-yl)-4-{5-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propyl]-imidazol-1-yl}-butyric acid;
      • 3-(3-Fluoro-phenyl)-4-{4-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2--yl)-propyl]-imidazol-1-yl}-butyric acid;
      • 3-Pyridin-3-yl-4-{4-ο3-(5,6,7,8-tetrahydro-[1,8]naphthryidin-2-yl)—propyl]-imidazol-1-yl}-butyric acid;
      • 3-Benzo[1,3]dioxol-5-yl-4-{4-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propyl]-imidazol-1-yl}-butryic acid3-Phenyl-4-{3-[2-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-ethoxy]-isoxazol-5-yl}butryic acid;
      • 3-(2,3-Dihydro-benzofuran-6-yl)-4-{3-[2-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-ethoxy]-isoxazol-5-yl}-butyric acid;
      • 3-(3-Fluoro-phenyl)-4-{3-[2-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2--yl)-ethoxy]-isoxazol-5-yl}-butyric acid;
      • 3-Pyridin-3-yl-4-{3-[2-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)—ethoxy]-isoxazol-5-yl}-butyric acid;
      • 3-Benzo[1,3]dioxol-5-yl-4-{3-]2-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-ethoxy]-isoxazol-5-yl}-butyric acid;
      • 3-(3-Fluoro-phenyl)-4-{5-[2-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2--yl)-ethoxy]-2H-pyrazol-3-yl}-butyric acid;
      • 3-(2,3-Dihydro-benzofuran-6-yl)-4-{5-[2-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-ethoxy]-2H-pyrazol-3-yl}-butyric acid;
      • 3-Phenyl-4-{5-[2-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-ethoxy- ]-2H-pyrazol-3-yl}-butyric acid;
      • 3-Pyridin-3-yl-4-{5-[2-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)—ethoxy]-2H-pyrazol-3-yl}-butyric acid;
      • 3-Benzo[1,3]dioxol-5-yl-4-{5-[2-(5,6,7,8-tetrahydro-[1,8]naphthryidin-2-yl)-ethoxy]-2H-pyrazol-3-yl}-butryic acid;
      • 3-Phenyl-4-[4-(3-5,6,7,8-tetrahydro-[1,8]naphthryidin-2-yl-propionyl)-imidazol-1-yl]-butryic acid;
      • 3-(2,3-Dihydro-benzofuran-6-yl)-4-[4-(3-5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl-propionyl)-imidazol-1-yl]- butryic acid;
      • 3-(3-Fluoro-phenyl)-4-[4-(3-5,6,7,8-tetrahydro-[1,8]naphthryidin-2—yl-propionyl)-imidazol-1-yl]-butryic acid;
      • 3-Pyridin-3-yl-4-[4-(3-5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl-propionyl)-imidazol-1-yl]-butyric acid;
      • 3-Benzo[1,3]dioxol-5-yl-4-]4-(3-5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl-propionyl)-imidazol-1-yl]butryic acid;
      • 4-{4-[1-Hydroxy-3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)propyl]-imidazol-1-yl}-3-phenyl-butryic acid;
      • 3-(2,3-Dihydro-benzofuran-6-yl)-4-{4-[1-hydroxy-3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propyl]-imidazol-1-yl}-butryic acid;
      • 3-(3-Fluoro-phenyl)-4-{4-[1-hydroxy-3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)propyl]-imidazol-1-yl}butryic acid;
      • 4-{5-[1-Hydroxy-3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl propyl]-imidazol-1-yl}-3-pyridin-3-yl-butyric acid;
      • 4-{5-[1-Hydroxy-3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propyl]-imidazol-1-yl}-3-pyridin-3-yl-butyric acid;
      • 4-{5-[1-Hydroxy-3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propylynyl]-imidazol-1-yl}-3-phenyl-butyric acid;
      • 3-(2,3-Dihydro-benzofuran-6-yl)-4-{4-[3-hydroxy-3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-prop-1-ynyl]-imidazol-1-yl}-butyric acid;
      • 3-(3-Fluoro-phenyl)-4-{4-[3-hydroxy-3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-prop-1-ynyl]-imidazol-1-yl}-butyric acid;
      • 4-{4-[3-Hydroxy-3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-prop-1-ynyl]-imidazaol-1-yl}-3-pyridin-3-yl-butyric acid;
      • 3-Benzo[1,3]dioxol-5-yl-4-{4-[3-hydroxy-3-(5,6,7,8-tetrahydro-[1,8]-naphthyridin-2-yl)-prop-1-ynyl]-imidazol-1-yl}-butyric acid;
      • 4-{4-[3-Hydroxy-3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-prop-1-ynyl]-pyrazol-1-yl}-3-phenyl-butyric acid;
      • 3-(2,3-Dihydro-benzofuran-6-yl)-4-{4-[3-hydroxy-3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-prop-1-ynyl]-pyrazol-1-yl}-butyric acid;
      • 3-(3-Fluoro-phenyl)-4-{-[3-hydroxy-3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-prop-1-ynyl]-pyrazol-1-yl}butyric acid;
      • 4-{4-[3-Hydroxy-3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-prop-1-ynyl]-pyrazol-1-yl}-3-pyridin-3-yl-butyric acid3-Benzo[1,3]dioxol-5-yl-4--{4-[3-hydroxy-3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-prop-1-ynyl]—pyrazol-1-yl}butyric acid4-{4-[3-Hydroxy-3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propenyl]-pyrazol-1-yl}-3-phenyl-butyric acid;
      • 3-(2,3-Dihydro-benzofuran-6-yl)-4-{4-[3-hydroxy-3-(5,6,7,8-tetrahydro[1,8]naphthyridin-2-yl)-propenyl]-pyrazol-1-yl}-butyric acid3-(3-Fluoro-phenyl)-4-{4-[3-hydroxy-3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propenyl]-pyrazol-1-yl≢-butyric acid;
      • 4-{4-[3-Hydroxy-3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)propenyl]-pyrazol-1-yl}-3-pyridin-3-yl-butyric acid;
      • 3-Benzo[1,3]dioxol-5-yl-4-{4-[3-hydroxy-3-(5,6,7,8-tetrahydro-[1,8]-naphthyridin-2-yl)-propenyl]-pyrazol-1-yl}-butyric acid;
      • 3-Benzo[1,3]dioxol-5-yl-4-{4-[3-hydroxy-3-(5,6,7,8-tetrahydro-[1,8]-naphthyridin-2-yl)-propenyl]-pyrazol-1-yl}-butyric acid;
      • 4-{4-[3-Hydroxy-3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propenyl]-imidazol-1-yl}-3-phenyl-butyric acid;
      • 3-(2,3-Dihydro-benzofuran-6-yl)-4-{4-[3-hydroxy-3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propenyl]-imidazol-1-yl}-butyric acid;
      • 3-(3-Fluoro-phenyl)-4-{4-[3-hydroxy-3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propenyl]-imidazol-1-yl}-butyric acid;
      • 4-{4-[3-Hydroxy-3-(5,6,7,8-tetrahydro-[1,8]-naphthyridin-2-yl)-propenyl]-imidazol-1-yl}-3-pyridin-3-yl-butyric acid;
      • 3-Benzo[1,3]dioxol-5-yl-4-{4-[3-hydroxy-3-(5,6,7,8-tetrahydro-[1,8]-naphthyridin-2-yl)-propenyl]-imidazol-1-yl}-butyric acid;
      • 4-{4-[3-Hydroxy-3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propyl]-imidazol-1-yl}-3-phenyl-butyric acid;
      • 3-(2,3-Dihydro-benzofuran-6-yl)-4-{4-[3-hydroxy-3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propyl]-imidazol-1-yl}-butyric acid;
      • 3-(3-Fluoro-phenyl)-4-{4-[3-hydroxy-3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propyl]-imidazol-1-yl}-butyric acid;
      • 4-{4-[3-Hydroxy-3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propyl]-imidazol-1-yl}-3-pyridin-3-yl-butyric acid;
      • 3-Benzo[1,3]dioxol-5-yl]-{4-[3-hydroxy-3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propyl]-imidazol-1-yl}-butyric acid;
      • 4-{4-[3-Hydroxy-3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propyl]-pyrazol-1-yl}-3-phenyl-butyric acid;
      • 3-(2,3-Dihydro-benzofuran-6-yl)-4-(4-[3-hydroxy-3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propyl]-pyrazol-1-yl}-butyric acid;
      • 3-(3-Fluoro-phenyl)-4-{4-[3-hydroxy-3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propyl]-pyrazol-1-yl}-butyric acid;
      • 3-Benzo[1,3]dioxol-5-yl-4-{4-[3-hydroxy-3-(5,6,7,8-tetrahydro-[1,8]-naphthyridin-2-yl)-propyl]-pyrazol-1-yl}-butyric acid;
      • 4-{4-[3-Hydroxy-3-(5,6,7,8-tetrahydro-[1,8]-naphthyridin-2-yl)-propyl]-pyrazol-1-yl}-3-pyridin-3-yl-butryic acid;
      • 3-(3-Fluoro-phenyl)-4-{4-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2--yl)-propenyl]-imidazol-1-yl}-butyric acid;
      • 3-Phenyl-4-{4-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propenyl]-imidazol-1-yl}-butyric acid;
      • 3-(2,3-Dihydro-benzofuran-6-yl)-4-{4-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propenyl]-imidazol-1-yl}-butyric acid;
      • 3-Pyridin-3-yl-4-{4-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)—propenyl]-imidazol-1-yl}-butyric acid;
      • 3-Benzo[1,3]dioxol-5-yl-4-{4-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propenyl]-imidazol-1-yl}-butyric acid;
      • 3-Benzo[1,3]dioxol-5-yl-4-{4-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propenyl]-pyrazol-1-yl}-butyric acid;
      • 3-Pyridin-3-yl-4-{4-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)—propenyl]-pyrazol-1-yl}-butyric acid;
      • 3-Phenyl-4-{4-[3-(5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)propenyl]-pyrazol-1-yl}-butyric acid;
      • 3-hydroxy-4-{3-[4-(pyridine-2-ylamio)butyl]-1,2,4-oxadiazol-5-yl}butanoic acid;
      • 3-hydroxy-4-{3-[3-(5,6,7,8-tetrahydro-1,8-naphthyridin-2-yl)propyl]--1,2,4-oxadiazol-5-yl}butanoic acid;
      • 3-Benzo[1,3]dioxol-5-yl-4-{3-[4-(1H-imidazol-3-ylamino)-butyl]-[1,2-,4]oxadiazol-5-yl}-butyric acid;
      • 3-(3-Fluoro-phenyl)-4-{3-[4-(1H-imidazol-2-ylamino)-butyl]-[1,2,4]oxadiazol-5-yl}-butyric acid;
      • 3-Benzo[1,3]dioxol-5-yl-4-{3-[4-(2H-pyrazol-3-ylamino)-butyl]-1,2,-4]oxadiazol-5-yl}-butyric acid;
      • 3-(3-Fluoro-phenyl)-4-{3-[4-(2H-pyrazol-3-ylamino)-butyl]-[1,2,4]oxadiazol-5-yl}-buutyric acid;
      • 3-Benzo]1,3]dioxol-5-yl-4-{3-[4-(3H-imidazol-4-ylamino)-butyl]-[1,2-,4]oxadiazol-5-yl≢-butyric acid;
      • 3-(3-Fluoro-phenyl)-4-{3-[4-(3H-imidazol-4-ylamino)-butyl]-[1,2,4]oxadiazol-5-yl}-butyric acid;
      • 3-Benzo[1,3]dioxol-5-yl-4-{3-[3-(6-methylamino-pyridin-2-yl)propyl]-[1,2,4]oxadiazol-5-yl}-butyric acid;
      • 3-(3-Fluoro-phenyl)-4-{3-[3-(6-methylamino-pyridin-2-yl)-propyl]-[1,2,4]oxadiazol-5-yl}-butyric acid;
      • 4-{3-[3-(6-Ethylamino-pyridin-2-yl)-propyl]-[1,2,4]oxadiazol-5-yl≢—3-(3-fluoro-phenyl)-butyric acid;
      • 3-(3-Fluoro-phenyl)-4-(3-{3-[6-(2-methoxy-ethylamino)-pyridin-2-yl]--propyl}-[1,2,4]oxadiazol-5-yl)-butyric acid;
      • 3-(3-Fluoro-phenyl)-4-(3-{3-[6-(3-methoxy-propylamino)-pyridin-2-yl- ]-propyl}-[1,2,4]oxadiazol-5-yl)butyric acid;
      • 3-(3-Fluoro-phenyl)-4-(3-{3-[6-(2,2,2-trifluoro-ethylamino)-pyridin--2-yl]-propyl}-[1,2,4]oxadiazol-5-yl)-butyric acid;
      • 3-(3-Fluoro-phenyl)-4-{3-[3-(5-oxo-5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)-propyl]-[1,2,4]oxadiazol-5-yl}-butyric acid;
      • 4-{3-[3-(5,5-Dimethyl-5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)propyl]-[1,2,4]oxadiazol-5-yl}-3-(3-fluoro-phenyl)-butyric acid;
      • 4-{3-[3-(5,5-Difluoro-5,6,7,8-tetrahydro-[1,8]naphthyridin-2-yl)propyl]-[1,2,4]oxadiazol-5-yl}-3-(3-fluoro-phenyl)butyric acid; and
      • 3-(1,3-benzodioxol-5-yl)-4-{3-{(5,6,7,8-tetrahydro-1,8-naphthyridin--2-ylmethoxy)methyl]-1,2,4-oxadiazol-5-yl}butanoic acid;
  • In another embodiment of the present invention
  • Figure US20160296592A1-20161013-C00150
      • is a heteroaryl substituted by one or more substituents selected from lower alkyl, alknyl, alkenyl, halogen, alkoxy, hydroxy, cyano, amino, alkylamino, dialkylamino or methylsulfonamide. More specifically, some examples of heteroaryl include oxadiazole, pyrimidine, imidazole, thiadiazole, triazole, tetrazole, pyrazole, isoxazole, and thiazole.
  • Other embodiments of
  • Figure US20160296592A1-20161013-C00151
      • include the following heterocyclic ring systems containing at least one nitrogen atom:
  • Figure US20160296592A1-20161013-C00152
      • wherein Zn is H, alkyl, alkoxy, hydroxy, amine, alkylamine, dialkylamine, carboxyl, alkoxycarbonyl, hydroxyalkyl, halogen or haloalkyl and R1 is H, alkyl, alkoxyalkyl, acyl, haloalkyl or alkoxycarbonyl. More specifically some examples include pyridylamino, imidazolylamin, morpholinopyridine, tetrahydronaphthyridine, oxazolylamino, thiazolylamino, pyrimidinylamino, quinoline, tetrahydroquinoline, imidazopyridine, benzimidazole, pyridone or quinolone.
  • The following heteroaryls include the ring systems described above.
  • Figure US20160296592A1-20161013-C00153
    Figure US20160296592A1-20161013-C00154
  • RECTIFIED SHEET (RULE 91)
  • RECTIFIED SHEET (RULE 91)
  • For the pyridyl derived heterocycle, the substituents X4 and X5 are selected from the group consisting of H, alkyl, branched alkyl, alkylamino, alkoxyalkylamino, haloalkyl, thioalkyl, halogen, amino, alkoxy, aryloxy, alkoxyalkyl, hydroxy, cyano or acylamino groups.
  • In another embodiment of the invention, the substituents X4 and X5 can be methyl, methoxy, amine, methylamine, trifluoromethyl, dimethylamine, hydroxy, chloro, bromo, fluoro and cyano. X6 may preferentially be H, alkyl hydroxy, halogen, alkoxy and haloalkyl. Alternately, the pyridyl ring can be fused with a 4-8 membered ring, optionally saturated or unsaturated. Some examples of these ring systems include tetrahydronaphthyridine, quinoline, tetrahydroquinoline, azaquinoline, morpholinopyridine, imidazopyridine and the like. The monocyclic ring systems such as imidazole, thiazole, oxazole, pyrazole, and the like, may contain an amino or alkylamino substituent at any position within the ring.
  • In another embodiment of the present invention, when Z1 of Formula I is CO or SO2, the linkage A1-Z2 of Formula I includes the heterocycle derived ring systems such as: pyridine, imidazole, thiazole, oxazole, benzimidazole, imidazopyrdine and the like.
  • Other heterocycles for A1-Z2 of the present invention include
  • Figure US20160296592A1-20161013-C00155
      • wherein X4 is as defined above.
  • In another embodiment, Y3 or Y4 is an aryl or a heteroaryl group selected from phenyl, benzofuran, benzothiophene, indole, quinoline, isoquinoline, benzimidazole, benzoxazole, 1,3-benzodioxole, 1,4-benzodioxane, benzopyran, quinolone, imidazopyridine, tetrahydro-quinoline, benzotriazole, dihydroindole, dihydrobenzofuran, furan, thiophene, phenyl, oxazole, thiazole, isoxazole, pyrazole, imidazole, pyrrole, pyridine, pyrimidine, pyridone, triazole, thiadiazole and the like. The aryl system can be optionally substituted at one or more positions with alkyl, alkoxy, hydroxy, cyano, halogen or haloalkyl.
  • In another embodiment of the present invention, Y3 or Y4 may be an amine, alkylamine, acylamine, aminosulfone (NHSO2R), arylamine, alkoxyalkylamine, aralkylamine, or heterocyclic amine.
  • In another embodiment of the present invention, Y3 taken together with Y4 forms a 3-8 membered monocyclic or 7-11 membered bicyclic ring B,
  • Figure US20160296592A1-20161013-C00156
      • optionally containing one or more double bonds, optionally containing one or more heteroatoms or functional groups selected from O, NR8, S, CO or SO2, optionally substituted with one or more substituent selected from the group consisting of alkyl, haloalkyl, halogen, haloalkyl, alkoxy, alkyne, cyano, alkylsulfone, sulfonamide, carboalkoxy and carboxyalkyl; wherein Rg is selected from the group consisting of alkyl, haloalkyl, alkoxyalkyl, aryl, heteroaryl, aralkyl and carboxyalkyl.
  • In another embodiment of the present invention, X taken together with Y3 forms a 3-7 membered monocyclic ring C,
  • Figure US20160296592A1-20161013-C00157
      • optionally containing one or more double bonds, optionally containing one or more heteroatom or functional group selected from O, NR8, S, CO or SO2, optionally substituted with one or more substituent selected from the group consisting of alkyl, halogen, alkoxy, haloalkyl, hydroxyalkyl, or alkoxyalkyl; wherein Rg is selected from the group consisting of H, alkyl, haloalkyl, alkoxyalkyl, aryl, heteroaryl, aralkyl, and carboxyalkyl.
  • The invention further relates to pharmaceutical compositions containing therapeutically effective amounts of the compounds of Formula I.
  • The present invention as a first object provides novel non-peptide compounds having the following formula (I)
  • Figure US20160296592A1-20161013-C00158
      • wherein:
      • G is a group selected among:
  • Figure US20160296592A1-20161013-C00159
      • wherein Q is NH or O and Q′ is H C1-C6 alkyl, phenyl, or phenyl-C1-C4-alkyl;
  • Figure US20160296592A1-20161013-C00160
      • X is a direct linkage, CH2—CONH, —(CH2)m— or (CH2)m—X′ wherein X′ is O, S or NH and m is an integer of 1 to 4;
      • B is CONH, CH2—CONH, —(CH2)m— or C2-C4 alkylene or C2-C4 alkenylene chain or —(CH2)m—X′, wherein X′ and m are as defined above;
      • A is a phenyl or pyridine ring, unsubstituted or optionally substituted by one to to three substituents chosen independently from halogen, CF3, C1-C4 alkyl, OH and C1-C4 alkoxy;
      • Y is selected from O or S(O)n wherein n is zero, 1 or 2;
      • R is C1-C6 alkyl or a phenyl or C5-C7 monocyclic heteroaryl ring containing one to three heteroatoms chosen from O, S, and N unsubstituted or optionally substituted by one to three substituents chosen independently from halogen, CF3, C1-C4 alkyl, hydroxy and C1-C4 alkoxy;
      • R′ is hydrogen, C1 -C6 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, aryl or aryl-C1-C-4 alkyl;
      • and the pharmaceutically acceptable salts thereof.
  • The present invention includes within its scope all possible isomers, stereoisomers and optical isomers and their mixtures, and the metabolites and the metabolic precursors or bioprecursors of the compounds of formula (I).
  • A halogen atom is preferably chlorine or fluorine.
  • The alkyl, alkoxy, alkenyl and alkynyl groups and the alkylene and alkenylene chains may be branched or straight groups or chains, respectively.
  • A C5-C7 monocyclic heteroaryl ring is preferably a C5-C6 heteromonocyclic ring, in particular selected from pyridine, pyrazine, pyridazine, pyrimidine, thiophene, pyrrole, pyrazole, imidazole, oxazole and isoxazole.
  • An aryl group is e.g., an aromatic C6-C20 mono- or poly-nuclear moiety, typically phenyl, unsubstituted or substituted by one to three substituents independently chosen from halogen, hydroxy, CF3, C1-C4 alkyl and C1-C4 alkoxy.
  • Accordingly an aralkyl group is e.g. benzyl or phenethyl, in which the phenyl ring is optionally substituted by one to three substituents independently chosen from halogen, hydroxy, CF3, C1-C4alkyl and C1-C4 alkoxy.
  • A C2-C4 alkenyl group is preferably an allyl group.
  • A C1-C6 alkyl group is preferably as C1-C4 alkyl group.
  • A C1-C4 alkyl group is preferably a methyl or ethyl group.
  • A C2-C4 alkynyl group is preferably an ethynyl group.
  • A C2-C4 alkoxy group is preferably methoxy, ethoxy, propoxy and butoxy.
  • Examples of pharmaceutically acceptable salts of the compounds of the invention are either those with inorganic bases, such as sodium, potassium, calcium and aluminium hydroxides, or with organic bases, such as lysine, arginine, N-methyl-glucamine, triethylamine, triethanolamine, dibenzylamine, methylbenzylamine, di-(2-ethyl-hexyl)-amine, piperidine, N-ethylpiperidine, N,N-diethylaminoethylamine, N-ethylmorpholine, β-phenethylamine, N-benzyl-β-phenethylamine, N-benzyl-N,N-dimethylamine and the other acceptable organic amines, as well as the salts with inorganic acids, e.g. hydrochloric, hydrobromic and sulphuric acids and with organic acids, e.g. citric, tartaric, maleic, malic, fumaric, trifluoroacetic, methanesulphonic and ethanesulphonic acids. As stated above the present invention also includes within its scope pharmaceutically acceptable bio-precursors (otherwise known as pro-drugs) of the compounds of formula (I), i.e. compounds which have a different formula to formula (I) above, but which nevertheless upon administration to a human being are converted directly or indirectly in vivo into a compound of formula (I).
  • Preferred compounds of the invention are those wherein, in formula (I),
      • G is as defined above;
      • X is a direct linkage or (CH2)m—X′, in which m is 1 or 2 and X′ is as defined above;
      • B is CONH or CH═CH or CH2—X′, wherein X is as defined above;
      • A is a phenyl ring unsubstituted or optionally substituted by one or two substituents selected from halogen CF3, C1-C4 alkyl, OH, and C1-C4 alkoxy;
      • Y is as defined above;
      • R is a phenyl, pyridine, pyrazine, pyridazine, pyrimidine, thiophene, pyrrole, pyrazole, imidazole, oxazole or isoxazole ring, unsubstituded or optionally substituted by one or two substituents selected from halogen, CF3, C1-C4 alkyl, OH and C1-C4 alkoxy;
      • R′ is hydrogen, C1-C6 alkyl; and the pharmaceutically acceptable salts thereof.
  • A further class of preferred compounds of the invention are those wherein, in formula (I),
      • G and Y are as defined above;
      • X is CH2-CONH or (CH2)m—X′, in which m and X′ are as defined above;
      • A is a phenyl or pyridine ring, unsubstituted or optionally substituted by one to three substituents chosen independently from halogen, CF3, C1-C4 alkyl, OH and C1-C4 alkoxy;
      • B is CONH or CH2—CONH;
      • R is a phenyl, pyridine, pyrazine, pyridazine, pyrimidine, thiophene, pyrrole, pyrazole, imidazole oxazole or isoxazole ring, unsubstituded or optionally substituted by one or two substituents selected from halogen, CF3, C1-C4 alkyl, OH and C1-C4 alkoxy;
      • R′ is hydrogen, C1-C6 alkyl; and the pharmaceutically acceptable salts thereof.
  • Most preferred compounds of the invention are those wherein, in formula (I),
      • G and Y are as defined above;
      • X is a direct linkage or (CH2)m—X′, in which m is 1 or 2 and X′ is as defined above;
      • B is CONH or CH═CH or CH2—X′, where X′ is as defined above;
      • A is a phenyl ring unsubstituted or optionally substituted by one or two substituents selected from halogen, CF3, C1-C4 alkyl, OH and C1-C4 alkoxy;
      • R is a phenyl or pyridinehding, unsubstituded or optionally substituted by one or two substituents selected from halogen, CF3, C1-C4 alkyl, OH and C1-C4alkoxy;
      • R′ is hydrogen, C1-C6 alkyl; and the pharmaceutically acceptable salts thereof. Examples of specific preferred compounds according to the invention are the following:
      • 3-({2-[(3-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfanyl)--3-(3-pyridinyl)propanoic acid;
      • 3-({3-[(3-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfanyl)--3-(3-pyridinyl)propanoic acid;
      • 3-({4-[(3-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfanyl)--3-(3-pyridinyl)propanoic acid;
      • 3-({2-[(3-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfanyl)-3-(3-pyridinyl)propanoic acid;
      • 3-({3-[(3-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfanyl)-3-(3-pyridinyl)propanoic acid;
      • 3-({4-[(3-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfanyl)-3-(3-pyridinyl)propanoic acid;
      • 3-{[2-({3-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfanyl}-3-(3--pyridinyl)propanoic acid;
      • 3-{[3-({3-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfanyl}-3-(3--pyridinyl)propanoic acid;
      • 3-{[4-({3-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfanyl}-3-(3--pyridinyl)propanoic acid;
      • 3-({2-[(3-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfanyl)-3-(3-pyridinyl)propanoic acid;
      • 3-({3-[(3-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfanyl)-3-(3-pyridinyl)propanoic acid;
      • 3-({4-[(3-{[(benzylamino)carbonyl]amino}benzoyl)amino[phenyl}sulfanyl)-3-(3-pyridinyl)propanoic acid;
      • 3-[(2-{[3-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfanyl]-3-(3-pyridinyl)propanoic acid;
      • 3-[(3-{[3-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfanyl]-3-(3-pyridinyl)propanoic acid;
      • 3-[(4-{[3-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfanyl]-3-(3-pyridinyl)propanoic acid;
      • 3-[(2-{[3-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfanyl]-3-(-3-pyridinyl)propanoic acid;
      • 3-[(3-{[3-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfanyl]-3-(-3-pyridinyl)propanoic acid;
      • 3-[(4-{[3-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfanyl]-3-(-3-pyridinyl)propanoic acid;
      • 3-(3-pyridinyl)-3-[(2-{[3-(2-pyridinylamino)benzoyl]amino}phenyl)sulfanyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(3-{[3-(2-pyridinylamino)benzoyl]amino}phenyl)sulfanyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(4-{[3-(2-pyridinylamino)benzoyl]amino}phenyl)sulfanyl]propanoic acid;
      • 3-({2-[(4-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfanyl)--3-(3-pyridinyl)propanoic acid;
      • 3-({3-[(4-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfanyl)--3-(3-pyridinyl)propanoic acid;
      • 3-({4-[(4-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfanyl)--3-(3-pyridinyl)propanoic acid;
      • 3-({2-[(4-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfanyl)-3-(3-pyridinyl)propanoic acid;
      • 3-({3-[(4-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfanyl)-3-(3-pyridinyl)propanoic acid;
      • 3-({4-[(4-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfanyl)-3-(3-pyridinyl)propanoic acid;
      • 3-{[2-({4-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfanyl}-3-(3-pyridinyl)propanoic acid;
      • 3-{[3-({4-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfanyl}-3-(3-pyridinyl)propanoic acid;
      • 3-{[4-({4-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfanyl}-3-(3-pyridinyl)propanoic acid;
      • 3-({2-[(4-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfanyl)-3-(3-pyridinyl)propanoic acid;
      • 3-({3-[(4-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfanyl)-3-(3-pyridinyl)propanoic acid;
      • 3-({4-[(4-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfanyl)-3-(3-pyridinyl)propanoic acid;
      • 3-[(2-{[4-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfanyl]-3-(3pyridinyl)propanoic acid;
      • 3-[(3-{[4-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfanyl]-3-(3pyridinyl)propanoic acid;
      • 3-[(4-{[4-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfanyl]-3-(3pyridinyl)propanoic acid;
      • 3-[(2-{[4-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfanyl]-3-(-3-pyridinyl)propanoic acid;
      • 3-[(3-{[4-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfanyl]-3-(-3-pyridinyl)propanoic acid;
      • 3-[(4-{[4-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfanyl]-3-(-3-pyridinyl)propanoic acid;
      • 3-(3-pyridinyl)-3-[(2-{[4-(2-pyridinylamino)benzoyl]amino}phenyl)sulfanyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(3-{[4-(2-pyridinylamino)benzoyl]amino}phenyl)sulfanyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(4-{[4-(2-pyridinylamino)benzoyl]amino}phenyl)sulfanyl]propanoic acid;
      • 3-({3-[(3-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfanyl)--3-phenylpropanoic acid;
      • 3-({2-[(3-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfanyl)--3-phenylpropanoic acid;
      • 3-({4-[(3-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfanyl)--3-phenylpropanoic acid;
      • 3-({2-[(3-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfanyl-3-phenylpropanoic acid;
      • 3-({3-[(3-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfanyl-3-phenylpropanoic acid;
      • 3-({4-[(3-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfanyl-3-phenylpropanoic acid;
      • 3-{[2-({3-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfanyl}-3-phenylpropanoic acid;
      • 3-{[3-({3-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfanyl}-3-phenylpropanoic acid;
      • 3-{[4-({3-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfanyl}-3-phenylpropanoic acid;
      • 3-({2-[(3-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfanyl)-3-phenylpropanoic acid;
      • 3-({3-[(3-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfanyl)-3-phenylpropanoic acid;
      • 3-({4-[(3-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfanyl)-3-phenylpropanoic acid;
      • 3-[(2-{[3-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfanyl]-3-phenylpropanoic acid;
      • 3-[(3-{[3-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfanyl]-3-phenylpropanoic acid;
      • 3-[(4-{[3-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfanyl]-3-phenylpropanoic acid;
      • 3-[(2-{[3-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfanyl]-3-phenylpropanoic acid;
      • 3-[(3-{[3-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfanyl]-3-phenylpropanoic acid;
      • 3-[(4-{[3-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfanyl]-3-phenylpropanoic acid;
      • 3-phenyl-3-[(2-{[3-(2-pyridinylamino)benzoyl]amino}phenyl)sulfanyl]-propanoic acid;
      • 3-phenyl-3-[(3-{[3-(2-pyridinylamino)benzoyl]amino}phenyl)sulfanyl]-propanoic acid;
      • 3-phenyl-3-[(4-{[3-(2-pyridinylamino)benzoyl]amino}phenyl)sulfanyl]-propanoic acid;
      • 3-({2-[(4-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfanyl)--3-phenylpropanoic acid;
      • 3-({3-[(4-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfanyl)--3-phenylpropanoic acid;
      • 3-({4-[(4-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfanyl)--3-phenylpropanoic acid;
      • 3-({2-[(4-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl)sulfanyl-3-phenylpropanoic acid;
      • 3-({3-[(4-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl)sulfanyl-3-phenylpropanoic acid;
      • 3-({4-[(4-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl)sulfanyl-3-phenylpropanoic acid;
      • 3-{[2-({4-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfanyl}-3-phenylpropanoic acid;
      • 3-{[3-({4-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfanyl}-3-phenylpropanoic acid;
      • 3-{[4-({4-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfanyl}-3-phenylpropanoic acid;
      • 3-({2-[(4-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfanyl)-3-phenylpropanoic acid;
      • 3-({3-[(4-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfanyl)-3-phenylpropanoic acid;
      • 3-({4-[(4-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfanyl)-3-phenylpropanoic acid;
      • 3-[(2-{[4-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfanyl]-3-phenylpropanoic acid;
      • 3-[(3-{[4-(4,5-dihydro-1H- imidazol-2-ylamino)benzoyl]amino}phenyl)sulfanyl]-3-phenylpropanoic acid;
      • 3-[(4-{[4-(4,5-dihydro-1H- imidazol-2-ylamino)benzoyl]amino}phenyl)sulfanyl]-3-phenylpropanoic acid;
      • 3-[(2-{[4-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfanyl]-3-phenylpropanoic acid;
      • 3-[(3-{[4-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfanyl]-3-phenylpropanoic acid;
      • 3-[(4-{[4-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfanyl]-3-phenylpropanoic acid;
      • 3-phenyl-3-[(2-{[4-(2-pyridinylamino)benzoyl]amino}phenyl)sulfanyl]-propanoic acid;
      • 3-phenyl-3-[(3-{[4-(2-pyridinylamino)benzoyl]amino}phenyl)sulfanyl]-propanoic acid;
      • 3-phenyl-3-[(4-{[4-(2-pyridinylamino)benzoyl]amino}phenyl)sulfanyl]-propanoic acid;
      • 3-{2-[(4-{[amino(imino)methyl]amino}benzoyl)amino]phenoxy}-3-(3-pyridinyl)propanoic acid;
      • 3-{3-[(4-{[amino(imino)methyl]amino}benzoyl)amino]phenoxy}-3-(3-pyridinyl)propanoic acid;
      • 3-{4-[(4-{[amino(imino)methyl]amino}benzoyl)amino]phenoxy}-3-(3-pyridinyl)propanoic acid;
      • 3-{2-[(4-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenoxy}—3-(3-pyridinyl)propanoic acid;
      • 3-{3-[(4-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenoxy}—3-(3-pyridinyl)propanoic acid;
      • 3-{4-[(4-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenoxy}—3-(3-pyridinyl)propanoic acid;
      • 3-[2-({4-[(aminocarbonyl)amino]benzoyl}amino)phenoxyl]-3-(3-pyridinyl)propanoic acid
      • 3-[3-({4-[(aminocarbonyl)amino]benzoyl}amino)phenoxyl]-3-(3-pyridinyl)propanoic acid;
      • 3-[2-({4-[(aminocarbonyl)amino]benzoyl}amino)phenoxyl]-3-(3-pyridinyl)propanoic acid;
      • 3-{2-[(4-{[(benzylamino)carbonyl]amino)benzoyl)amino]phenoxy}-3-(3—pyridinyl)propanoic acid;
      • 3-{3-[(4-{[(benzylamino)carbonyl]amino)benzoyl)amino]phenoxy}-3-(3—pyridinyl)propanoic acid;
      • 3-{4-[(4-{[(benzylamino)carbonyl]amino)benzoyl)amino]phenoxy}-3-(3—pyridinyl)propanoic acid;
      • 3-(2-{[4-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenoxy)—3-(3-pyridinyl)propanoic acid;
      • 3-(3-{[4-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenoxy)—3-(3-pyridinyl)propanoic acid;
      • 3-(4-{[4-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenoxy)—3-(3-pyridinyl)propanoic acid;
      • 3-(2-{[4-(1H-imidazol-2-ylamino)benzoyl]amino}phenoxy)-3-(3-pyridinyl)propanoic acid;
      • 3-(3-{[4-(1H-imidazol-2-ylamino)benzoyl]amino}phenoxy)-3-(3-pyridinyl)propanoic acid;
      • 3-(4-{[4-(1H-imidazol-2-ylamino)benzoyl]amino}phenoxy)-3-(3-pyridinyl)propanoic acid;
      • 3-(3-pyridinyl)-3-(2-{[4-(2-pyridinylamino)benzoyl]amino}phenoxy)propanoic acid;
      • 3-(3-pyridinyl)-3-(3-{[4-(2-pyridinylamino)benzoyl]amino}phenoxy)propanoic acid;
      • 3-(3-pyridinyl)-3-(4-{[4-(2-pyridinylamino)benzoyl]amino}phenoxy)propanoic acid;
      • 3-{2-[(3-{[amino(imino)methyl]amino}benzoyl)amino]phenoxy}-3-(3-pyridinyl)propanoic acid;
      • 3-{3-[(3-{[amino(imino)methyl]amino}benzoyl)amino]phenoxy}-3-(3-pyridinyl)propanoic acid;
      • 3-{4-[(3-{[amino(imino)methyl]amino}benzoyl)amino]phenoxy}-3-(3-pyridinyl)propanoic acid;
      • 3-{2-[(3-{[(benzylamino)(imino)methyl]aminobenzoyl)amino]phenoxy)—3-(3-pyridinyl)propanoic acid;
      • 3-{3-[(3-{[(benzylamino)(imino)methyl]aminobenzoyl)amino]phenoxy)—3-(3-pyridinyl)propanoic acid;
      • 3-{4-[(3-{[(benzylamino)(imino)methyl]aminobenzoyl)amino]phenoxy)—3-(3-pyridinyl)propanoic acid;
      • 3-[2-({3-[(aminocarbonyl)amino]benzoyl}amino)phenoxy]-3-(3-pyridinyl)propanoic acid;
      • 3-[3-({3-[(aminocarbonyl)amino]benzoyl}amino)phenoxy]-3-(3-pyridinyl)propanoic acid;
      • 3-[4-({3-[(aminocarbonyl)amino]benzoyl}amino)phenoxy]-3-(3-pyridinyl)propanoic acid;
      • 3-{2-[(3-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenoxy}-3-(3—pyridinyl)propanoic acid;
      • 3-{3-[(3-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenoxy}-3-(3—pyridinyl)propanoic acid;
      • 3-{4-[(3-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenoxy}-3-(3—pyridinyl)propanoic acid;
      • 3-(2-{[3-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenoxy)—3-(3-pyridinyl)propanoic acid;
      • 3-(3-{[3-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenoxy)—3-(3-pyridinyl)propanoic acid;
      • 3-(4-{[3-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenoxy)—3-(3-pyridinyl)propanoic acid;
      • 3-(2-{[3-(1H-imidazol-2-ylamino)benzoyl]amino}phenoxy)-3-(3-pyridinyl)propanoic acid;
      • 3-(3-{[3-(1H-imidazol-2-ylamino)benzoyl]amino}phenoxy)-3-(3-pyridinyl)propanoic acid;
      • 3-(4-{[3-(1H-imidazol-2-ylamino)benzoyl]amino}phenoxy)-3-(3-pyridinyl)propanoic acid;
      • 3-(3-pyridinyl)-3-(2-{[3-(2-pyridinylamino)benzoyl]amino}phenoxy)propanoic acid;
      • 3-(3-pyridinyl)-3-(3-{[3-(2-pyridinylamino)benzoyl]amino}phenoxy)propanoic acid;
      • 3-(3-pyridinyl)-3-(4-{[3-(2-pyridinylamino)benzoyl]amino}phenoxy)propanoic acid;
      • 3-{2-[(3-{[amino(imino)methyl]amino}benzoyl)amino]phenoxy)-3-phenyl-propanoic acid;
      • 3-{3-[(3-{[amino(imino)methyl]amino}benzoyl)amino]phenoxy)-3-phenyl-propanoic acid;
      • 3-{4-[(3-{[amino(imino)methyl]amino}benzoyl)amino]phenoxy)-3-phenyl-propanoic acid;
      • 3-{2-[(3-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenoxy}—3-phenylpropanoic acid;
      • 3-{3-[(3-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenoxy}—3-phenylpropanoic acid;
      • 3-{4-[(3-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenoxy}—3- phenylpropanoic acid;
      • 3-[2-({3-[(aminocarbonyl)amino]benzoyl}amino)phenoxyl]-3-phenylpropanoic acid;
      • 3-[3-({3-[(aminocarbonyl)amino]benzoyl}amino)phenoxyl]-3-phenylpropanoic acid;
      • 3-[4-({3-[(aminocarbonyl)amino]benzoyl}amino)phenoxyl]-3-phenylpropanoic acid;
      • 3-{2-[(3-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenoxy-3-phenylpropanoic acid;
      • 3-{3-[(3-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenoxy-3-phenylpropanoic acid;
      • 3-{4-[(3-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenoxy-3-phenylpropanoic acid;
      • 3-(2-{[3-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenoxy)—3- phenylpropanoic acid;
      • 3-(3-{[3-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenoxy)—3-phenylpropanoic acid;
      • 3-(4-{[3-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenoxy)—3- phenylpropanoic acid;
      • 3-(2-{[3-(1H-imidazol-2-ylamino)benzoyl]amino}phenoxy)-3-phenylpropanoic acid;
      • 3-(3-{[3-(1H-imidazol-2-ylamino)benzoyl]amino}phenoxy)-3-phenylpropanoic acid;
      • 3-(4-{[3-(1H-imidazol-2-ylamino)benzoyl]amino}phenoxy)-3-phenylpropanoic acid;
      • 3-phenyl-3-(2-{[3-(2-pyridinylamino)benzoyl]amino}phenoxy)propanoic acid;
      • 3-phenyl-3-(3-{[3-(2-pyridinylamino)benzoyl]amino}phenoxy)propanoic acid;
      • 3-phenyl-3-(4-{[3-(2-pyridinylamino)benzoyl]amino}phenoxy)propanoic acid;
      • 3-{2-[(4-{[amino(imino)methyl]amino}benzoyl)amino]phenoxy}-3-phenyl-propanoic acid;
      • 3-{3-[(4-{[amino(imino)methyl]amino}benzoyl)amino]phenoxy}-3-phenyl-propanoic acid;
      • 3-{4-[(4-{[amino(imino)methyl]amino}benzoyl)amino]phenoxy}-3-phenyl-propanoic acid;
      • 3-{2-[(4-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenoxy}—3- phenylpropanoic acid;
      • 3-{3-[(4-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenoxy}—3- phenylpropanoic acid;
      • 3-{4-[(4-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenoxy}—3- phenylpropanoic acid;
      • 3-[2-({4-[(aminocarbonyl)amino]benzoyl}amino)phenoxy]-3-phenylpropanoic acid;
      • 3-[3-({4-[(aminocarbonyl)amino]benzoyl}amino)phenoxy]-3-phenylpropanoic acid;
      • 3-[4-({4-[(aminocarbonyl)amino]benzoyl}amino)phenoxy]-3-phenylpropanoic acid;
      • 3-{2-[(4-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenoxy}-3-phenylpropanoic acid;
      • 3-{3-[(4-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenoxy}-3-phenylpropanoic acid;
      • 3-{4-[(4-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenoxy}-3-phenylpropanoic acid;
      • 3-(2-{[4-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenoxy)—3- phenylpropanoic acid;
      • 3-(3-{[4-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenoxy)—3- phenylpropanoic acid;
      • 3-(4-{[4-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenoxy)—3- phenylpropanoic acid;
      • 3-(2-{[4-(1H-imidazol-2-ylamino)benzoyl]amino}phenoxy)-3-phenylpropanoic acid;
      • 3-(3-{[4-(1H-imidazol-2-ylamino)benzoyl]amino}phenoxy)-3-phenylpropanoic acid;
      • 3-(4-{[4-(1H-imidazol-2-ylamino)benzoyl]amino}phenoxy)-3-phenylpropanoic acid;
      • 3-phenyl-3-(2-{[4-(2-pyridinylamino)benzoyl]amino}phenoxy)propanoic acid;
      • 3-phenyl-3-(3-{[4-(2-pyridinylamino)benzoyl]amino}phenoxy)propanoic acid;
      • 3-phenyl-3-(4-{[4-(2-pyridinylamino)benzoyl]amino}phenoxy)propanoic acid;
      • 3-({2-[(3-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfinyl)--3-(3-pyridinyl)propanoic acid;
      • 3-({3-[(3-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfinyl)--3-(3-pyridinyl)propanoic acid;
      • 3-({4-[(3-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfinyl)--3-(3-pyridinyl)propanoic acid;
      • 3-({2-[(3-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfinyl)-3-(3-pyridinyl)propanoic acid;
      • 3-({3-[(3-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfinyl)-3-(3-pyridinyl)propanoic acid;
      • 3-({4-[(3-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfinyl)-3-(3-pyridinyl)propanoic acid;
      • 3-{[2-({3-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfinyl}-3-(3--pyridinyl)propanoic acid;
      • 3-{[3-({3-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfinyl}-3-(3--pyridinyl)propanoic acid;
      • 3-({[4-({3-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfinyl}-3-(-3-pyridinyl)propanoic acid;
      • 3-({2-[(3-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfinyl)-3-(3-pyridinyl)propanoic acid;
      • 3-({3-[(3-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfinyl)-3-(3-pyridinyl)propanoic acid;
      • 3-({4-[(3-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfinyl)-3-(3-pyridinyl)propanoic acid;
      • 3-[(2-{[3-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfinyl]-3-(3-pyridinyl)propanoic acid;
      • 3-[(3-{[3-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfinyl]-3-(3-pyridinyl)propanoic acid;
      • 3-[(4-{[3-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfinyl]-3-(3-pyridinyl)propanoic acid;
      • 3-[(2-{[3-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfinyl]-3-(-3-pyridinyl)propanoic acid;
      • 3-[(3-{[3-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfinyl]-3-(-3-pyridinyl)propanoic acid;
      • 3-[(4-{[3-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfinyl]-3-(-3-pyridinyl)propanoic acid;
      • 3-(3-pyridinyl)-3-[(2-{[3-(2-pyridinylamino)benzoyl]amino}phenyl)sulfinyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(3-{[3-(2-pyridinylamino)benzoyl]amino}phenyl)sulfinyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(4-{[3-(2-pyridinylamino)benzoyl]amino}phenyl)sulfinyl]propanoic acid;
      • 3-({2-[4-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfinyl)--3-(3-pyridinyl)propanoic acid;
      • 3-({3-[4-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfinyl)--3-(3-pyridinyl)propanoic acid;
      • 3-({4-[4-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfinyl)--3-(3-pyridinyl)propanoic acid;
      • 3-({2-[(4-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfinyl)-3-a(3-pyridinyl)propanoic acid;
      • 3-({3-[(4-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfinyl)-3-(3-pyridinyl)propanoic acid;
      • 3-({4-[(4-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfinyl)-3-(3-pyridinyl)propanoic acid;
      • 3-{[2-({4-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfinyl}-3-(3--pyridinyl)propanoic acid;
      • 3-{[3-({4-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfinyl}-3-(3--pyridinyl)propanoic acid;
      • 3-{[4-({4-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfinyl}-3-(3--pyridinyl)propanoic acid;
      • 3-({2-[(4-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfinyl)-3-(3-pyridinyl)propanoic acid;
      • 3-({3-[(4-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfinyl)-3-(3-pyridinyl)propanoic acid;
      • 3-({4-[(4-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfinyl)-3-(3-pyridinyl)propanoic acid;
      • 3-[(2-{[4-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfinyl]-3-(3-pyridinyl)propanoic acid;
      • 3-[(3-{[4-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfinyl]-3-(3-pyridinyl)propanoic acid;
      • 3-[(4-{[4-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfinyl]-3-(3-pyridinyl)propanoic acid;
      • 3-[(2-{[4-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfinyl]-3-(-3-pyridinyl)propanoic acid;
      • 3-[(3-{[4-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfinyl]-3-(-3-pyridinyl)propanoic acid;
      • 3-[(4-{[4-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfinyl]-3-(-3-pyridinyl)propanoic acid;
      • 3-(3-pyridinyl)-3-[(2-{[4-(2-pyridinylamino)benzoyl]amino}phenyl)sulfinyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(3-{[4-(2-pyridinylamino)benzoyl]amino}phenyl)sulfinyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(4-{[4-(2-pyridinylamino)benzoyl]amino}phenyl)sulfinyl]propanoic acid;
      • 3-({2-[(3-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfonyl)--3-(3-pyridinyl)propanoic acid;
      • 3-({3-[(3-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfonyl)--3-(3-pyridinyl)propanoic acid;
      • 3-({4-[(3-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfonyl)--3-(3-pyridinyl)propanoic acid;
      • 3-({2-[(3-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfonyl)-3-(3-pyridinyl)propanoic acid;
      • 3-({3-[(3-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfonyl)-3-(3-pyridinyl)propanoic acid;
      • 3-({4-[(3-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfonyl)-3-(3-pyridinyl)propanoic acid;
      • 3-{[2-({3-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfonyl}-3-(3--pyridinyl)propanoic acid;
      • 3-{[3-({3-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfonyl}-3-(3--pyridinyl)propanoic acid;
      • 3-{[4-({3-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfonyl}-3-(3--pyridinyl)propanoic acid;
      • 3-({2-[(3-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfon- yl)-3-(3-pyridinyl)propanoic acid;
      • 3-({3-[(3-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfon- yl)-3-(3-pyridinyl)propanoic acid;
      • 3-({4-[(3-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfonyl)-3-(3-pyridinyl)propanoic acid;
      • 3-[(2-{[3-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfonyl]-3-(3-pyridinyl)propanoic acid;
      • 3-[(3-{[3-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfonyl]-3-(3-pyridinyl)propanoic acid;
      • 3-[(4-{[3-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfonyl]-3-(3-pyridinyl)propanoic acid;
      • 3-[(2-{[3-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfonyl]-3-(-3-pyridinyl)propanoic acid;
      • 3-[(3-{[3-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfonyl]-3-(-3-pyridinyl)propanoic acid;
      • 3-[(4-{[3-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfonyl]-3-(-3-pyridinyl)propanoic acid;
      • 3-(3-pyridinyl)-3-[(2-{[3-(2-pyridinylamino)benzoyl]amino}phenyl)sulfonyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(3-{[3-(2-pyridinylamino)benzoyl]amino}phenyl)sulfonyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(4-{[3-(2-pyridinylamino)benzoyl]amino}phenyl)sulfonyl]propanoic acid;
      • 3-({2-[(4-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfonyl)--3-(3-pyridinyl)propanoic acid;
      • 3-({3-[(4-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfonyl)--3-(3-pyridinyl)propanoic acid;
      • 3-({4-[(4-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfonyl)--3-(3-pyridinyl)propanoic acid;
      • 3-({2-[(4-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfonyl)-3-(3-pyridinyl)propanoic acid;
      • 3-({3-[(4-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfonyl)-3-(3-pyridinyl)propanoic acid;
      • 3-({4-[(4-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfonyl)-3-(3-pyridinyl)propanoic acid;
      • 3-{[2-({4-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfonyl}-3-(3--pyridinyl)propanoic acid;
      • 3-{[3-({4-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfonyl}-3-(3--pyridinyl)propanoic acid;
      • 3-{[4-({4-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfonyl}-3-(3-- pyridinyl)propanoic acid;
      • 3-({2-[(4-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfonyl)-3-(3-pyridinyl)propanoic acid;
      • 3-({3-[(4-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfonyl)-3-(3-pyridinyl)propanoic acid;
      • 3-({4-[(4-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfonyl)-3-(3-pyridinyl)propanoic acid;
      • 3-[(2{[4-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino)phenyl)sulfonyl]-3-(3-pyridinyl)propanoic acid;
      • 3-[(3{[4-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino)phenyl)sulfonyl]-3-(3-pyridinyl)propanoic acid;
      • 3-[(4{[4-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino)phenyl)sulfonyl]-3-(3-pyridinyl)propanoic acid;
      • 3-[(2-{[4-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfonyl]-3-(-3- pyridinyl)propanoic acid;
      • 3-[(3-{[4-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfonyl]-3-(-3- pyridinyl)propanoic acid;
      • 3-[(4-{[4-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfonyl]-3-(-3- pyridinyl)propanoic acid;
      • 3-(3-pyridinyl)-3-[(2-{[4-(2-pyridinylamino)benzoyl]amino}phenyl)sulfonyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(3-{[4-(2-pyridinylamino)benzoyl]amino}phenyl)sulfonyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(4-{[4-(2-pyridinylamino)benzoyl]amino}phenyl)sulfonyl]propanoic acid;
      • 3-({3-[(3-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfinyl)--3-phenylpropanoic acid;
      • 3-({2-[(3-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfinyl)--3-phenylpropanoic acid;
      • 3-({4-[(3-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfinyl)--3phenylpropanoic acid;
      • 3-({2-[(3-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfinyl)-3-phenylpropanoic acid;
      • 3-({3-[(3-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfinyl)-3-phenylpropanoic acid;
      • 3-({4-[(3-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfinyl)-3-phenylpropanoic acid;
      • 3-{[2-({3-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfinyl}-3-phenylpropanoic acid;
      • 3-{[3-({3-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfinyl}-3-phenylpropanoic acid;
      • 3-{[4-({3-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfinyl}-3-phenylpropanoic acid;
      • 3-({2-[(3-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfinyl)-3-phenylpropanoic acid;
      • 3-({3-[(3-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfinyl)-3-phenylpropanoic acid;
      • 3-({4-[(3-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfinyl)-3-phenylpropanoic acid;
      • 3-[(2-{[3-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfinyl]-3-phenylpropanoic acid;
      • 3-[(3-{[3-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfinyl]-3-phenylpropanoic acid;
      • 3-[(4-{[3-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfinyl]-3-phenylpropanoic acid;
      • 3-[(2-{[3-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfinyl]-3-phenylpropanoic acid;
      • 3-[(3-{[3-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfinyl]-3-phenylpropanoic acid;
      • 3-[(4-{[3-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfinyl]-3-phenylpropanoic acid;
      • 3-phenyl-3-[(2-{[3-(2-pyridinylamino)benzoyl]amino}phenyl)sulfinyl]-propanoic acid;
      • 3-phenyl-3-[(3-{[3-(2-pyridinylamino)benzoyl]amino}phenyl)sulfinyl]-propanoic acid;
      • 3-phenyl-3-[(4-{[3-(2-pyridinylamino)benzoyl]amino}phenyl)sulfinyl]-propanoic acid;
      • 3-({2-[(4-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfinyl)--3-phenylpropanoic acid;
      • 3-({3-[(4-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfinyl)--3-phenylpropanoic acid;
      • 3-({4-[(4-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfinyl)--3-phenylpropanoic acid;
      • 3-({2-[(4-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfinyl)-3-phenylpropanoic acid;
      • 3-({3-[(4-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfinyl)-3-phenylpropanoic acid;
      • 3-({4-[(4-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfinyl)-3-phenylpropanoic acid;
      • 3-{[2-({4-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfinyl}-3-phenylpropanoic acid;
      • 3-{[3-({4-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfinyl}-3-phenylpropanoic acid;
      • 3-{[4-({4-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfinyl}-3-phenylpropanoic acid;
      • 3-({2-[(4-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfin- yl)-3-phenylpropanoic acid;
      • 3-({3-[(4-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfin- yl)-3-phenylpropanoic acid;
      • 3-({4-[(4-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfin- yl)-3-phenylpropanoic acid;
      • 3-[(2-{[4-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfinyl]-3-phenylpropanoic acid;
      • 3-[(3-{[4-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfinyl]-3-phenylpropanoic acid;
      • 3-[(4-{[4-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfinyl]-3-phenylpropanoic acid;
      • 3-[(2-{[4-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfinyl]-3-phenylpropanoic acid;
      • 3-[(3-{[4-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfinyl]-3-phenylpropanoic acid;
      • 3-[(4-{[4-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfinyl]-3-phenylpropanoic acid;
      • 3-phenyl-3-[(2-{[4-(2-pyridinylamino)benzoyl]amino}phenyl)sulfinyl]-propanoic acid;
      • 3-phenyl-3-[(3-{[4-(2-pyridinylamino)benzoyl]amino}phenyl)sulfinyl]-propanoic acid;
      • 3-phenyl-3-[(4-{[4-(2-pyridinylamino)benzoyl]amino}phenyl)sulfinyl]-propanoic acid;
      • 3-({3-[(3-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfonyl)--3-phenylpropanoic acid;
      • 3-({2-[(3-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfonyl)--3-phenylpropanoic acid;
      • 3-({4-[(3-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfonyl)--3phenylpropanoic acid;
      • 3-({2-[(3-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfonyl)-3-phenylpropanoic acid;
      • 3-({3-[(3-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfonyl)-3-phenylpropanoic acid;
      • 3-({4-[(3-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfonyl)-3-phenylpropanoic acid;
      • 3-{[2-({3-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfonyl}-3-phenylpropanoic acid;
      • 3-{[3-({3-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfonyl}-3-phenylpropanoic acid;
      • 3-{[4-({3-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfonyl}-3-phenylpropanoic acid;
      • 3-({2-[(3-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfon- yl)-3-phenylpropanoic acid;
      • 3-({3-[(3-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfon- yl)-3-phenylpropanoic acid;
      • 3-({4-[(3-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfon- yl)-3-phenylpropanoic acid;
      • 3-[(2-{[3-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfonyl]-3-phenylpropanoic acid;
      • 3-[(3-{[3-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfonyl]-3-phenylpropanoic acid;
      • 3-[(4-{[3-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfonyl]-3-phenylpropanoic acid;
      • 3-[(2-{[3-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfonyl]-3-phenylpropanoic acid;
      • 3-[(3-{[3-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfonyl]-3-phenylpropanoic acid;
      • 3-[(4-{[3-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfonyl]-3-phenylpropanoic acid;
      • 3-phenyl-3-[(2-{[3-(2-pyridinylamino)benzoyl]amino}phenyl)sulfonyl]-propanoic acid;
      • 3-phenyl-3-[(3-{[3-(2-pyridinylamino)benzoyl]amino}phenyl)sulfonyl]-propanoic acid;
      • 3-phenyl-3-[(4-{[3-(2-pyridinylamino)benzoyl]amino}phenyl)sulfonyl]-propanoic acid;
      • 3-({2-[(4-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfonyl)--3-phenylpropanoic acid;
      • 3-({3-[(4-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfonyl)--3-phenylpropanoic acid;
      • 3-({4-[(4-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfonyl)--3-phenylpropanoic acid;
      • 3-({2-[(4-{[(benzylamino)imino)methyl]amino}benzoyl)amino]phenyl}sulfonyl)-3-phenylpropanoic acid;
      • 3-({3-[(4-{[(benzylamino)imino)methyl]amino}benzoyl)amino]phenyl}sulfonyl)-3-phenylpropanoic acid;
      • 3-({4-[(4-{[(benzylamino)imino)methyl]amino}benzoyl)amino]phenyl}sulfonyl)-3-phenylpropanoic acid;
      • 3-{[2-({4-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfonyl}-3-phenylpropanoic acid;
      • 3-{[3-({4-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfonyl}-3-phenylpropanoic acid;
      • 3-{[4-({4-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfonyl}-3-phenylpropanoic acid;
      • 3-({2-[(4-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfonyl)-3-phenylpropanoic acid;
      • 3-({3-[(4-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfonyl)-3-phenylpropanoic acid;
      • 3-({4-[(4-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfonyl)-3-phenylpropanoic acid;
      • 3-[(2-{[4-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfonyl]-3-phenylpropanoic acid;
      • 3-[(3-{[4-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfonyl]-3-phenylpropanoic acid;
      • 3-[(4-{[4-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfonyl]-3-phenylpropanoic acid;
      • 3-[(2-{[4-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfonyl]-3-phenylpropanoic acid;
      • 3-[(3-{[4-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfonyl]-3-phenylpropanoic acid;
      • 3-[(4-{[4-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfonyl]-3-phenylpropanoic acid;
      • 3-phenyl-3-[(2-{[4-(2-pyridinylamino)benzoyl]amino}phenyl)sulfonyl]-propanoic acid;
      • 3-phenyl-3-[(3-{[4-(2-pyridinylamino)benzoyl]amino}phenyl)sulfonyl]-propanoic acid;
      • 3-phenyl-3-[(4-{[4-(2-pyridinylamino)benzoyl]amino}phenyl)sulfonyl]-propanoic acid;
      • 3-phenyl-3-[(2-{[4-(2-pyridinylamino)benzoyl]amino}sulfonyl]-propanoic acid;
      • 3-phenyl-3-[(3-{[4-(2-pyridinylamino)benzoyl]amino}sulfonyl]-propanoic acid;
      • 3-phenyl-3-[(4-{[4-(2-pyridinylamino)benzoyl]amino}sulfonyl]-propanoic acid;
      • 3-(3-pyridinyl)-3-(2-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenoxy)propanoic acid;
      • 3-(3-pyridinyl)-3-(3-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenoxy)propanoic acid;
      • 3-(3-pyridinyl)-3-(4-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenoxy)propanoic acid;
      • 3-(3-pyridinyl)-3-(2-{[4-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenoxy)propanoic acid;
      • 3-(3-pyridinyl)-3-(3-{[4-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenoxy)propanoic acid;
      • 3-(3-pyridinyl)-3-(4-{[4-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenoxy)propanoic acid;
      • 3-phenyl-3-(2-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenoxy)propanoic acid;
      • 3-phenyl-3-(3-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenoxy)propanoic acid;
      • 3-phenyl-3-(4-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenoxy)propanoic acid;
      • 3-phenyl-3-(4-{[4-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenoxy)propanoic acid;
      • 3-phenyl-3-(3-{[4-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenoxy)propanoic acid;
      • 3-phenyl-3-(4-{[4-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenoxy)propanoic acid;
      • 3-(3-pyridinyl)-3-[(2-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfanyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(3-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfanyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(4-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfanyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(2-{[4-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfanyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(3-{[4-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfanyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(4-{[4-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfanyl]propanoic acid;
      • 3-phenyl-3-[(2-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfanyl]propanoic acid;
      • 3-phenyl-3-[(3-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfanyl]propanoic acid;
      • 3-phenyl-3-[(4-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfanyl]propanoic acid;
      • 3-phenyl-3-[(2-{[4-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfanyl]propanoic acid;
      • 3-phenyl-3-[(3-{[4-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfanyl]propanoic acid;
      • 3-phenyl-3-[(4-{[4-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfanyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(2-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfinyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(3-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfinyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(4-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfinyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(2-{[4-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfinyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(3-{[4-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfinyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(4-{[4-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfinyl]propanoic acid;
      • 3-phenyl-3-[(2-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfinyl]propanoic acid;
      • 3-phenyl-3-[(3-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfinyl]propanoic acid;
      • 3-phenyl-3-[(4-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfinyl]propanoic acid;
      • 3-phenyl-3-[(2-{[4-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfinyl]propanoic acid;
      • 3-phenyl-3-[(3-{[4-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfinyl]propanoic acid;
      • 3-phenyl-3-[(4-{[4-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfinyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(2-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfonyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(3-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfonyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(4-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfonyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(2-{[4-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfonyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(3-{[4-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfonyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(4-{[4-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfonyl]propanoic acid;
      • 3-phenyl-3-[(2-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfonyl]propanoic acid;
      • 3-phenyl-3-[(3-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfonyl]propanoic acid;
      • 3-phenyl-3-[(4-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfonyl]propanoic acid;
      • 3-phenyl-3-[(2-{[4-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfonyl]propanoic acid;
      • 3-phenyl-3-[(3-{[4-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfonyl]propanoic acid;
      • 3-phenyl-3-[(4-{[4-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfonyl]propanoic acid;
      • either as a single isomer or as a mixture thereof, or a pharmaceutically acceptable salt thereof, in particular the hydrochloride or the trifluoroacetate.
  • A further object of the present invention is to provide a compound or formula (I)
  • Figure US20160296592A1-20161013-C00161
      • wherein:
      • G is a group selected among:
  • Figure US20160296592A1-20161013-C00162
      • wherein Q is NH or O and Q′ is H, C1-C6 alkyl, phenyl, or phenyl-C1-C4-alkyl;
  • Figure US20160296592A1-20161013-C00163
      • X is a direct linkage, CH2—CONH, —(CH2)m—X′ wherein X′ is O, S, or NH and m is an integer of 1 to 4;
      • B is CONH, CH2—CONH, a C2-C4 alkylene or C2-C4 alkenylene chain or —(CH2)m—X′, wherein X′ and m are as defined above;
      • A is a phenyl or pyridine ring, unsubstituted or optionally substituted by one to three substituents chosen independently from halogen, CF3, C1-C4 alkyl, OH and C1-C4 alkoxy;
      • Y is selected from O or S(O)n wherein n is zero, 1 or 2;
      • R is C1-C6 alkyl or a phenyl or C5-C7 monocyclic heteroaryl ring containing one to three heteroatoms chosen from O, S, and N, unsubstituted or optionally substituted by one to three substituents chosen independently from halogen, CF3, C1-C4 alkyl, hydroxy and C1-C4 alkoxy;
      • R′ is hydrogen, C1-C6 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, aryl or aryl-C1-C4 alkyl;
      • or a pharmaceutically acceptable salt thereof, for use in a method of treatment of the human or animal body by therapy, in particular for treating conditions mediated by the αvβ3 integrin.
  • Object of the present invention is also to provide a pharmaceutical composition comprising a pharmaceutically acceptable carrier and/or diluent and as an active principle, a compound of formula (I), as herein defined, or a pharmaceutically acceptable salt thereof.
  • The present invention also provides the use of a compound of formula (I), a defined above, in the preparation of a medicament having αvβ3 integrin inhibiting or antagonizing activity.
  • The present invention also provides a method for treating conditions mediated by the αvβ3 integrin in a mammal, including humans, in need of such treatment comprising administering to said mammal an effective αvβ3 inhibiting or antagonizing amount of a compound of formula (I)
  • Figure US20160296592A1-20161013-C00164
      • wherein:
      • G is a group selected among:
  • Figure US20160296592A1-20161013-C00165
      • wherein Q is NH or O and Q′ is H, C1-C6 alkyl, phenyl, or phenyl-C1-C4-alkyl;
  • Figure US20160296592A1-20161013-C00166
      • X is a direct linkage, CH2—CONH, —(CH2)m— or (CH2)m—X′ wherein X′ is O, S or NH and m is an integer of 1 to 4;
      • B is CONH, CH2—CONH, a C2-C4 alkylene or C2-C4 alkenylene chain or —(CH2)m—X′, wherein X′ and m are as defined above;
      • A is a phenyl or pyridine ring, unsubstituted or optionally substituted by one to three substituents chosen independently from halogen, CF3, C1-C4 alkyl, OH and C1-C4 alkoxy;
      • Y is selected from O or S(O)n wherein n is zero, 1 or 2;
      • R is C1-C6 alkyl or a phenyl or C5-C7 monocyclic heteroaryl ring containing one to three heteroatoms chosen from O, S, and N, unsubstituted or optionally substituted by one to three substituents chosen independently from halogen, CF3, C1-C4 alkyl, hydroxy and C1-C4 alkoxy;
      • R′ is hydrogen, C1-C6 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, aryl or aryl-C1-C4 alkyl;
      • or a pharmaceutically acceptable salt thereof.
  • More specifically, the present invention provides a method for inhibiting bone resorption, treating osteoporosis, inhibiting humoral hypercalcemia of malignancy, treating Paget's disease, inhibiting tumor metastasis, inhibiting neoplasia (solid tumor growth), inhibiting angiogenesis including tumor angiogenesis, treating diabetic retinopathy, inhibiting arthritis, psoriasis and periodontal disease, and inhibiting smooth muscle cell migration including restenosis.
  • The compounds of the invention and the salt thereof can be prepared by and analogy process. Accordingly, the compounds of formula I and the salts thereof, can be for instance obtained by a process which comprises:
      • a) reacting a compound of formula II
  • Figure US20160296592A1-20161013-C00167
      • where q is zero or 1, y′ is a reactive function, preferably halogen, and G and X are as defined above, with a compound of formula III
  • Figure US20160296592A1-20161013-C00168
      • where A, Y, R, and R′ are as defined above, thus obtaining a compound of formula I where B is CONH or CH2—CONH; or
      • b) reacting a compound of formula IV
  • Figure US20160296592A1-20161013-C00169
      • where y′ is a reactive function, preferably halogen and G, X and m are as defined above, with a compound of formula V
  • Figure US20160296592A1-20161013-C00170
      • where X′, A, Y, R, and R′ are as defined above, thus obtaining a compound of formula I where B is —(CH2)m—X′, wherein X′ and m are as defined above; or
      • c) reacting a compound of formula VI
  • Figure US20160296592A1-20161013-C00171
      • where X, B, A, Y, R, and R′ are as defined above, and Y′ is NH2, with a suitable guanilating agent such as di-Boc-trifyl-guanidine, thus obtaining a compound of formula I where G is a goanidino group; or
      • d) reacting a compound of formula VI where X, B, A, Y, R, R′ and Y′ are as defined above, with a benzyl cyanate or a cyanate salt, as e.g. a ammonium or sodium or potassium salt, thus obtaining a compound of formula I where G is an urea group; or
      • e) reacting a compound of formula VI where X, B, A, Y, R, R′ and Y′ are as defined above, with an isocyanate of formula Q′NCO, where Q′ is a group selected among C1-C6 alkyl, Phenyl, Phenyl-C1-C4-alkyl, thus obtaining a compound of formula I where G is a Q′NH(CO)NH— group, in which Q′ is a group selected among C1-C6 alkyl, Phenyl, Phenyl-C1-C4-alkyl;
      • f) reacting a compound of formula VII
  • Figure US20160296592A1-20161013-C00172
      • wherein G, X, B and A, are as defined above and Y″ is a thiol group, with a compound of formula VIII,
  • Figure US20160296592A1-20161013-C00173
      • wherein R and R′ are as defined above, thus obtaining a compound of formula (I) wherein Y is —S—; or
      • g) reacting a compound of formula IX 1
  • Figure US20160296592A1-20161013-C00174
      • wherein G, X, B, A, are as defined above and Y″ is a group Y—H with a compound of formula X,
  • Figure US20160296592A1-20161013-C00175
      • wherein R, R′ and y′ are as defined above, thus obtaining a compound of formula I;
      • h) hydrolyzing, preferably in acidic conditions, a carboxylic ester compound of formula I wherein G, X, B, A, Y, R are as defined above and R′ is different from hydrogen, thus obtaining a compound of formula I wherein R′ is hydrogen;
      • i) oxidizing with a suitable agent such as NaIO4 or oxone or H2O2, a compound of formula I wherein G, X, B, A, R and R′ are as defined above and Y is sulfur, thus obtaining a compound of formula 1 wherein G, X, B, A, R and R′ are as defined above and Y is S(O)n wherein n is 1 or 2, i.e. Y is a sulfoxide or sulfone group;
      • l) cleaving a compound of formula I, wherein G, X, B, A, Y and R are as defined above and the group R is represented by Wang (p-bezyloxybenzylalcohol) resin, with TFA in CH2Cl2, thus obtaining a compound of formula I wherein R′ is hydrogen; and, if desired, salifying a compound of formula (I) and/or, if desired, converting a salt of a compound of formula (I) into a free compound and/or, if desired, separating as mixture of isomers of a compound of formula (I) into the single isomers.
    Process-Variants a) to I), Respectively, Can for Instance be Carried out at Follows:
      • a) In a typical procedure the acid was added to as large excess of thionyl chloride (25 equivalents) kept under stirring at about 0 degree. C. under nitrogen atmosphere, the mixture heated at about 60 degree. C. for 1 hour, brought to room temperature and thoroughly evaporated in vacuo. The acid chloride II was dissolved in DMF and added to a mixture of the intermediate required (III) (1 equivalent) and triethylamine (1 equivalent) in DMF or pyridine. The mixture was stirred at room temperature for 48-72 hours, the solvent removed, the residue taken with brine and extracted with dichloromethane. The compounds thus obtained were purified by silica gel flash chromatography eluting with CH2Cl2/MeOH 9:1.
      • b) In a typical procedure compounds of formula IV were added to a solution of V in solvents like DMF, acetone, in presence of a base (NaH, K2CO3, KHMSA) the temperature varying between room temperature and reflux, the time of reaction varying between 24-72 hours. The mixture filtered the solvent evaporated and the residue taken with brine and extracted with dichloromethane or ethyl acetate. The compounds were purified by silica gel flash chromatography eluting with CH2Cl2/MeOH 9:1 or Petroleum ether 40-60/Ethyl acetate 1:1.
      • c) In a typical procedure the amino derivatives of formula VI reacted with the guanilating agent, such as di-Boc-triflyl-guanidine, in dichloromethane in presence of stechiometric amounts of base such as triethylamine or diusopropylethylamine, the time of reaction varying between 24-72 hours. The compounds thus obtained did not request further purification. (FEICHTINGER, K.; ZAPF, C.; SINGS, H. L.; GOODMAN, M.; J Org 1998, 63 (12), 3804-3805). The protection groups were removed according to standard procedures, with TFA.
      • d) In a typical procedure the amino derivatives of formula VI reacted with a cyanate salt, as e.g. a ammonium or sodium or potassium salt, in solvent such as AcOH, H2O (Org. Synth. 1963, IV, 49) PNAS 1993, 90, 6909-6913) the temperature varying between 50-100 degree. C., the time of reaction varying
      • e)In a typical procedure the amino derivatives of formula VI reacted with isocyanates in presence of triethylamine refuxing in solvents such as dichloromethane, acetonitrile, toluene or dioxane (JMC 1996, 39 (22) 4382-4395; Eur J Med Chem 1997, 32 (10), 795-804.) to give the subtituted ureas, the temperature varying between room temperature and reflux, the time of reaction varying between 8-24 hours.
      • f) in a typical procedure to a stirred solution of the acids VIII (cinnamic or trans-3-(3-pyridyl)acrylic) as ethyl esters or polymer supported, in dichloromethane kept under vigorous stirring in nitrogen atmosphere, the amiothiophenols VII (2eq) and DBU(0.1 equivalent) were added, the mixture was stirred at room temperature for about 24 hours, the solvent evaporated and the residue purified by silica gel flash chromatography eluting with petroleum ether 40-60/ethyl acetate 1:1, to give the intermediates required.
      • g In a typical procedure compounds of formula X were added to a solution of IX in solvents like DMF, acetone, in presence of a base (K2CO3, NaH, KHMSA) the temperature varying between room temperature and reflux, time of reaction varying between 8-24 hours
      • h) The ester compounds of formula I, were hydrolysed by treatment with a mixture of HCl 4N and ethanol overnight. Solvents were evaporated and the final compounds, obtained as hydrochlorides, purified by crystallisation with methanol and ether.
      • i) The sulfur functions of formula I were oxidised to solfones with 30% aq H2O2 (RAVIKUMAR, K. S.; BEGUE, J.-P.; BONNET-DELPON, D.; Tetrahedron Lett 1998, 39 (20), 3141-3144.) or with NaIO4, in water and methanol, or acetone and water, according to a standard procedure (BEIER, C.; SCHAUMANN, E.; Synthesis 1997, (11), 1296-1300; LE MERRER, Y.; FUZIER, M.; DOSBAA, I.; FOGLIETTI, M. -J.; DEPEZAY, J.-C.; Tetrahedron 1997, 53 (49), 16731-16746). The sulfur functions were oxidised to solfoxides according to a standard procedure by reacting with oxone in a mixture methanol/water as a solvent (HINTERBERGER, S.; HOFER, O.; GREGER, H.; Tetrahedron 1998, 54 (3), 487-496.)
      • l) Compounds of formula 1 linked to the Wang resin were cleaved as a final step with a mixture trifluoroacetic acid/dichloromethane, the time of reaction varying between 15-30 minutes.
  • Also the optional salification of a compound of formula (I) as well as the conversion of a salt into the free compound and the separation of a mixture of isomers into the single isomers may be carried out by conventional methods. For example, the separation of optical isomers may be carried out by salification with an optically active base or acid and by subsequent fractional crystallization of the diastereoisomeric salts, followed by recovering of the optically active isomeric-acids or, respectively, bases. When in the compound of formula (I), and in the intermediate products thereof, groups are present which need to be protected before submitting them to the here-above illustrated reactions, they may be protected before the reactions take place and then deprotected, according to well known methods in organic chemistry. The compounds of formulae (I) and the pharmaceutically acceptable, salts thereof are herein defined as the “compounds of the present invention”, the “compounds of the invention” and/or the “active principles of the pharmaceutical compositions of the invention”.
  • For instance, according to process variant a) above, and according the indications herein provided, the compound 3-({4-[(3-{[amino(imino)methyl]amino}benzoyl)amino]-phenyl}sulfanyl)-3-(3-pyridinyl)propanoic acid (internal code PNU 277262F) can be provided. In particular compound PNU 277362F can be synthesized as bis-trifluoroacetate salt, as depicted in scheme 1, according to the procedures reported.
  • Figure US20160296592A1-20161013-C00176
      • 3-{4-[(3-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfanyl)}--3-(3-pyridinyl)propanoic acid bis-trifluoroacetate (PNU 277362F)
      • 1H-NMR (DMSO-d6): 10.3 (s, 1 H); 9.8 (s, 1 H); 8.5 (m, 2 H); 7.85 (m, 2 H); 7.78 (m, 1 H); 7.7 (d, 2 H) 7.6-7.4 (m, 5 H); 7.3 (d, 2 H), 4.8 (t, 1 H); 2.9 (m, 2 H); Elemental Analysis: Theoretic: C, 47.06%; H, 3.49%; F, 17.18%; N, 10.55%; S, 4.83%; Found: C, 44.32%; H, 3.49%; F, 16.70%; N, 9.95%; S, 4.44%
  • Figure US20160296592A1-20161013-C00177
      • Z is
  • Figure US20160296592A1-20161013-C00178
      • wherein
      • Pol=polymer of Wang resin; DIC=diisopropylcarbodiimide; DMAP=4-dimethylminopyridine; DBUcat=1,8-diazabicyclo[5,4,0]9 undec-7-ene in catalytic amount; DMF=dimethylformamide; TFA=trifluoro acetic acid.
  • Analogously, according to process variant d) above, the compound 3-{[4-({3-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfanyl)-3-(3-pyridinyl)propanoic acid (internal code PNU 515440) is provided.
  • Figure US20160296592A1-20161013-C00179
      • 3-{[4-({3-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfanyl}-3-(3--pyridinyl) propanoic acid (PNU 515440)
      • MS; m/z 437 (M+H+). 1H-NMR (400 MHz), .delta. (DMSO-d6): 2.82 (m, 2 H, CH2, 4.65 (m, 1 H, CHS), 5.91 (s, 2 H, CONH2), 7.20-7.70 (m, 9 H, ArH), 7.85 (m, 1 H, ArH), 8.38 (m, 2 H, pyridine hydrogens), 8.77 (s, 1 H, NHCO), 10.24 (s, 1 H, NHCO).
  • According to process variant e) above, the compound 3-{[4-({3-[(benzylamino)carbonyl])amino]phenyl}sulfanyl)-3—(3-pyridinyl) propanoic acid (internal code PHA 515442E) is provided. The compound PNU 515442E can be synthesized as trifluoroacetate salts.
  • Figure US20160296592A1-20161013-C00180
      • 3-{[4-({3-[(benzylamino)carbonyl])amino}benzoyl)amino]phenyl}sulfanyl)-3-(3-pyridinyl) propanoic acid (PHA 515442E)
      • MS: m/z 527 (M+H+). 1H-NMR (400 MHz), .delta. (DMSO-d6): 2.97 (m, 2 H, CH2), 4.29 (d, 2 H, J=5.7, CH2NH), 4.71 (m, 1 H, CHS), 7.20-7.70 (m, 14 H, ArH) 7.88 (m, 1 H, ArH), 8.53 (m, 2 H, pyridine hydrogens), 8.77 (s, 1 H, NHCO), 10.27 (s, 1 H, NHCO).
  • In analogy to process variants a) to l), the following compounds can be obtained:
      • 3-({2-[(3-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfanyl)--3-(3-pyridinyl)propanoic acid;
      • 3-({3-[(3-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfanyl)--3-(3-pyridinyl)propanoic acid;
      • 3-({2-[(3-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfanyl)-3-(3-pyridinyl)propanoic acid;
      • 3-({3-[(3-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfanyl)-3-(3-pyridinyl)propanoic acid;
      • 3-{[2-({3-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfanyl}-3-(3--pyridinyl)propanoic acid;
      • 3-{[3-({3-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfanyl}-3-(3--pyridinyl)propanoic acid;
      • 3-({2-[(3-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfan- yl)-3-(3-pyridinyl)propanoic acid;
      • 3-({3-[(3-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfan- yl)-3-(3-pyridinyl)propanoic acid;
      • 3-({4-[(3-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfan- yl)-3-(3-pyridinyl)propanoic acid;
      • 3-[(2-{[3-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfanyl]-3-(3-pyridinyl)propanoic acid;
      • 3-[(3-{[3-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfanyl]-3-(3-pyridinyl)propanoic acid;
      • 3-[(4-{[3-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfanyl]-3-(3-pyridinyl)propanoic acid;
      • 3-[(2-{[3-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfanyl]-3-(-3-pyridinyl)propanoic acid;
      • 3-[(3-{[3-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfanyl]-3-(-3-pyridinyl)propanoic acid;
      • 3-[(4-{[3-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfanyl]-3-(-3-pyridinyl)propanoic acid;
      • 3-(3-pyridinyl)-3-[(2-{[3-(2-pyridinylamino)benzoyl]amino}phenyl)sulfanyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(3-{[3-(2-pyridinylamino)benzoyl]amino}phenyl)sulfanyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(4-{[3-(2-pyridinylamino)benzoyl]amino}phenyl)sulfanyl]propanoic acid;
      • 3-({2-[(4-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfanyl)--3-(3-pyridinyl)propanoic acid;
      • 3-({3-[(4-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfanyl)--3-(3-pyridinyl)propanoic acid;
      • 3-({4-[(4-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfanyl)--3-(3-pyridinyl)propanoic acid;
      • 3-({2-[(4-{[benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfanyl-3-(3-pyridinyl)propanoic acid;
      • 3-({3-[(4-{[benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfanyl-3-(3-pyridinyl)propanoic acid;
      • 3-({4-[(4-{[benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfanyl-3-(3-pyridinyl)propanoic acid;
      • 3-{[2-({4-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfanyl}-3-(3--pyridinyl)propanoic acid;
      • 3-{[3-({4-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfanyl}-3-(3--pyridinyl)propanoic acid;
      • 3-{[4-({4-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfanyl}-3-(3--pyridinyl)propanoic acid;
      • 3-({2-[(4-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfanyl)-3-(3-pyridinyl)propanoic acid;
      • 3-({3-[(4-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfanyl)-3-(3-pyridinyl)propanoic acid;
      • 3-({4-[(4-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfanyl)-3-(3-pyridinyl)propanoic acid;
      • 3-[(2-{[4-(4,5-dihyrdo-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfanyl]-3-(3-pyrdinyl)propanoic acid;
      • 3-[(3-{[4-(4,5-dihyrdo-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfanyl]-3-(3-pyrdinyl)propanoic acid;
      • 3-[(4-{[4-(4,5-dihyrdo-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfanyl]-3-(3-pyrdinyl)propanoic acid;
      • 3-[(2-{[4-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfanyl]-3-(-3-pyridinyl)propanoic acid;
      • 3-[(3-{[4-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfanyl]-3-(-3-pyridinyl)propanoic acid;
      • 3-[(4-{[4-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfanyl]-3-(-3-pyridinyl)propanoic acid;
      • 3-(3-pyridinyl-3-[(2-{[4-(2-pyridinylamino)benzoyl]amino}phenyl)sulfanyl]propanoic acid;
      • 3-(3-pyridinyl-3-[(3-{[4-(2-pyridinylamino)benzoyl]amino}phenyl)sulfanyl]propanoic acid;
      • 3-(3-pyridinyl-3-[(4-{[4-(2-pyridinylamino)benzoyl]amino}phenyl)sulfanyl]propanoic acid;
      • 3-({3-[(3-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfanyl)--3-phenylpropanoic acid;
      • 3-({2-[(3-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfanyl)--3-phenylpropanoic acid;
      • 3-({4-[(3-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfanyl)--3phenylpropanoic acid;
      • 3-({2-[(3-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfanyl)-3-phenylpropanoic acid;
      • 3-({3-[(3-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfanyl)-3-phenylpropanoic acid;
      • 3-({4-[(3-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfanyl)-3-phenylpropanoic acid;
      • 3-{[2-({3-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfanyl}-3-phenylpropanoic acid;
      • 3-{[3-({3-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfanyl}-3-phenylpropanoic acid;
      • 3-{[4-({3-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfanyl}-3-phenylpropanoic acid;
      • 3-({2-[(3-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfanyl)-3-phenylpropanoic acid;
      • 3-({3-[(3-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfanyl)-3-phenylpropanoic acid;
      • 3-({4-[(3-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfanyl)-3-phenylpropanoic acid;
      • 3-[(2-{[3-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfanyl]-3-phenylpropanoic acid;
      • 3-[(3-{[3-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfanyl]-3-phenylpropanoic acid;
      • 3-[(4-{[3-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfanyl]-3-phenylpropanoic acid;
      • 3-[(2-{[3-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfanyl]-3-phenylpropanoic acid;
      • 3-[(3-{[3-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfanyl]-3-phenylpropanoic acid;
      • 3-[(4-{[3-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfanyl]-3-phenylpropanoic acid;
      • 3-phenyl-3-[(2-{[3-(2-pyridinylamino)benzoyl]amino}phenyl)sulfanyl]-propanoic acid;
      • 3-phenyl-3-[(3-{[3-(2-pyridinylamino)benzoyl]amino}phenyl)sulfanyl]-propanoic acid;
      • 3-phenyl-3-[(4-{[3-(2-pyridinylamino)benzoyl]amino}phenyl)sulfanyl]-propanoic acid;
      • 3-({2-[(4-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfanyl)--3-phenylpropanoic acid;
      • 3-({3-[(4-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfanyl)--3-phenylpropanoic acid;
      • 3-({4-[(4-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfanyl)--3-phenylpropanoic acid;
      • 3-({2-[(4-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfanyl)-3-phenylpropanoic acid;
      • 3-({3-[(4-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfanyl)-3-phenylpropanoic acid;
      • 3-({4-[(4-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfanyl)-3-phenylpropanoic acid;
      • 3-{[2-({4-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfanyl}-3-phenylpropanoic acid;
      • 3-{[3-({4-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfanyl}-3-phenylpropanoic acid;
      • 3-{[4-({4-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfanyl}-3-phenylpropanoic acid;
      • 3-({2-[(4-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfanyl-3-phenylpropanoic acid;
      • 3-({3-[(4-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfanyl-3-phenylpropanoic acid;
      • 3-({4-[(4-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfanyl-3-phenylpropanoic acid;
      • 3-[(2-{[4-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfanyl]-3-phenylpropanoic acid;
      • 3-[(3-{[4-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfanyl]-3-phenylpropanoic acid;
      • 3-[(4-{[4-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfanyl]-3-phenylpropanoic acid;
      • 3-[(2-{[4-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfanyl]-3-phenylpropanoic acid;
      • 3-[(3-{[4-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfanyl]-3-phenylpropanoic acid;
      • 3-[(4-{[4-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfanyl]-3-phenylpropanoic acid;
      • 3-phenyl-3-[(2-{[4-(2-pyridinylamino)benzoyl]amino}phenyl)sulfanyl]-propanoic acid;
      • 3-phenyl-3-[(3-{[4-(2-pyridinylamino)benzoyl]amino}phenyl)sulfanyl]-propanoic acid;
      • 3-phenyl-3-[(4-{[4-(2-pyridinylamino)benzoyl]amino}phenyl)sulfanyl]-propanoic acid;
      • 3-{2-[(4-{[amino(imino)methyl]amino}benzoyl)amino]phenoxy}-3-(3-pyridinyl)propanoic acid;
      • 3-{3-[(4-{[amino(imino)methyl]amino}benzoyl)amino]phenoxy}-3-(3-pyridinyl)propanoic acid;
      • 3-{4-[(4-{[amino(imino)methyl]amino}benzoyl)amino]phenoxy}-3-(3-pyridinyl)propanoic acid;
      • 3-{2-[(4-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenoxy}—3-(3-pyridinyl)propanoic acid;
      • 3-{3-[(4-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenoxy}—3-(3-pyridinyl)propanoic acid;
      • 3-{4-[(4-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenoxy}—3-(3-pyridinyl)propanoic acid;
      • 3-[2-({4-[(aminocarbonyl)amino]benzoyl}amino)phenoxy]-3-(3-pyridinyl)propanoic acid;
      • 3-[3-({4-[(aminocarbonyl)amino]benzoyl}amino)phenoxy]-3-(3-pyridinyl)propanoic acid;
      • 3-[4-({4-[(aminocarbonyl)amino]benzoyl}amino)phenoxy]-3-(3-pyridinyl)propanoic acid;
      • 3-{2-[(4-{[benzylamino)carbonyl]amino}benzoyl)amino]phenoxy}-3-(3—pyridinyl)propanoic acid;
      • 3-{3-[(4-{[benzylamino)carbonyl]amino}benzoyl)amino]phenoxy}-3-(3—pyridinyl)propanoic acid;
      • 3-{4-[(4-{[benzylamino)carbonyl]amino}benzoyl)amino]phenoxy}-3-(3—pyridinyl)propanoic acid;
      • 3-(2-{[4-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenoxy)—3-(3-pyridinyl)propanoic acid;
      • 3-(3-{[4-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenoxy)—3-(3-pyridinyl)propanoic acid;
      • 3-(4-{[4-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenoxy)—3-(3-pyridinyl)propanoic acid;
      • 3-(2-{[4-(1H-imidazol-2-ylamino)benzoyl]amino}phenoxy)-3-(3-pyridinyl)propanoic acid;
      • 3-(3-{[4-(1H-imidazol-2-ylamino)benzoyl]amino}phenoxy)-3-(3-pyridinyl)propanoic acid;
      • 3-(4-{[4-(1H-imidazol-2-ylamino)benzoyl]amino}phenoxy)-3-(3-pyridinyl)propanoic acid;
      • 3-(3-pyridinyl)-3-(2-{[4-(2-pyridinylamino)benzoyl]amino}phenoxy)propanoic acid;
      • 3-(3-pyridinyl)-3-(3-{[4-(2-pyridinylamino)benzoyl]amino}phenoxy)propanoic acid;
      • 3-(3-pyridinyl)-3-(4-{[4-(2-pyridinylamino)benzoyl]amino}phenoxy)propanoic acid;
      • 3-{2-[(3-{[amino(imino)methyl]amino}benzoyl)amino]phenoxy]-3-(3-pyridinyl)propanoic acid;
      • 3-{3-[(3-{[amino(imino)methyl]amino}benzoyl)amino]phenoxy]-3-(3-pyridinyl)propanoic acid;
      • 3-{4-[(3-{[amino(imino)methyl]amino}benzoyl)amino]phenoxy]-3-(3-pyridinyl)propanoic acid;
      • 3-{2-[(3-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenoxy}—3-(3-pyridinyl)propanoic acid;
      • 3-{3-[(3-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenoxy}—3-(3-pyridinyl)propanoic acid;
      • 3-{4-[(3-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenoxy}—3-(3-pyridinyl)propanoic acid;
      • 3-[2-({3-[(aminocarbonyl)amino]benzoyl}amino)phenoxy]-3-(3-pyridinyl)propanoic acid;
      • 3-[3-({3-[(aminocarbonyl)amino]benzoyl}amino)phenoxy]-3-(3-pyridinyl)propanoic acid;
      • 3-[4-({3-[(aminocarbonyl)amino]benzoyl}amino)phenoxy]-3-(3-pyridinyl)propanoic acid;
      • 3-{2-[(3-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenoxy}-3-(3—pyridinyl)propanoic acid;
      • 3-{3-[(3-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenoxy}-3-(3—pyridinyl)propanoic acid;
      • 3-{4-[(3-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenoxy}-3-(3—pyridinyl)propanoic acid;
      • 3-(2-{[3-(4,5-dihydro-1H-imidzaol-2-ylamino)benzoyl]amino}phenoxy—3-(3-pyridinyl)propanoic acid;
      • 3-(3-{[3-(4,5-dihydro-1H-imidzaol-2-ylamino)benzoyl]amino}phenoxy—3-(3-pyridinyl)propanoic acid;
      • 3-(4-{[3-(4,5-dihydro-1H-imidzaol-2-ylamino)benzoyl]amino}phenoxy—3-(3-pyridinyl)propanoic acid;
      • 3-(2-{[3-(1H-imidazol-2-ylamino)benzoyl]amino}phenoxy)-3-(3-pyridinyl)propanoic acid;
      • 3-(3-{[3-(1H-imidazol-2-ylamino)benzoyl]amino}phenoxy)-3-(3-pyridinyl)propanoic acid;
      • 3-(4-{[3-(1H-imidazol-2-ylamino)benzoyl]amino}phenoxy)-3-(3-pyridinyl)propanoic acid;
      • 3-(3-pyridinyl)-3-(2-{[3-(2-pyridinylamino)benzoyl]amino}phenoxy)propanoic acid;
      • 3-(3-pyridinyl)-3-(3-{[3-(2-pyridinylamino)benzoyl]amino}phenoxy)propanoic acid;
      • 3-(3-pyridinyl)-3-(4-{[3-(2-pyridinylamino)benzoyl]amino}phenoxy)propanoic acid;
      • 3-{2-[(3-{[amino(imino)methyl]amino}benzoyl)amino]phenoxy}-3-phenyl-propanoic acid;
      • 3-{3-[(3-{[amino(imino)methyl]amino}benzoyl)amino]phenoxy}-3-phenyl-propanoic acid;
      • 3-{4-[(3-{[amino(imino)methyl]amino}benzoyl)amino]phenoxy}-3-phenyl-propanoic acid;
      • 3-{2-[(3-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenoxy}—3-phenylpropanoic acid;
      • 3-{3-[(3-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenoxy}—3-phenylpropanoic acid;
      • 3-{4-[(3-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenoxy}—3-phenylpropanoic acid;
      • 3-[2-({3-[(aminocarbonyl)amino]benzoyl}amino)phenoxy]-3-phenylpropanoic acid;
      • 3-[3-({3-[(aminocarbonyl)amino]benzoyl}amino)phenoxy]-3-phenylpropanoic acid;
      • 3-[4-({3-[(aminocarbonyl)amino]benzoyl}amino)phenoxy]-3-phenylpropanoic acid;
      • 3-{2-[(3-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenoxy}-3-phenylpropanoic acid;
      • 3-{3-[(3-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenoxy}-3-phenylpropanoic acid;
      • 3-{4-[(3-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenoxy}-3-phenylpropanoic acid;
      • 3-(2-{[3-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenoxy)—3-phenylpropanoic acid;
      • 3-(3-{[3-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenoxy)—3-phenylpropanoic acid;
      • 3-(4-{[3-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenoxy)—3-phenylpropanoic acid;
      • 3-(2-{[3-(1H-imidazol-2-ylamino)benzoyl]amino}phenoxy)-3-phenylpropanoic acid;
      • 3-(3-{[3-(1H-imidazol-2-ylamino)benzoyl]amino}phenoxy)-3-phenylpropanoic acid;
      • 3-(4-{[3-(1H-imidazol-2-ylamino)benzoyl]amino}phenoxy)-3-phenylpropanoic acid;
      • 3-phenyl-3-(2-{[3-(2-pyridinylamino)benzoyl]amino}phenoxy)propanoic acid;
      • 3-phenyl-3-(3-{[3-(2-pyridinylamino)benzoyl]amino}phenoxy)propanoic acid;
      • 3-phenyl-3-(4-{[3-(2-pyridinylamino)benzoyl]amino}phenoxy)propanoic acid;
      • 3-{2-[(4-{[amino(imino)methyl]amino}benzoyl)amino]phenoxy}-3-phenyl-propanoic acid;
      • 3-{3-[(4-{[amino(imino)methyl]amino}benzoyl)amino]phenoxy}-3-phenyl-propanoic acid;
      • 3-{4-[(4-{[amino(imino)methyl]amino}benzoyl)amino]phenoxy}-3-phenyl-propanoic acid;
      • 3-{2-[(4-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenoxy}—3-phenylpropanoic acid;
      • 3-{3-[(4-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenoxy}—3-phenylpropanoic acid;
      • 3-{4-[(4-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenoxy}—3-phenylpropanoic acid;
      • 3-[2-({4-[(aminocarbonyl)amino]benzoyl}amino)phenoxy]-3-phenylpropanoic acid;
      • 3-[3-({4-[(aminocarbonyl)amino]benzoyl}amino)phenoxy]-3-phenylpropanoic acid;
      • 3-[4-({4-[(aminocarbonyl)amino]benzoyl}amino)phenoxy]-3-phenylpropanoic acid;
      • 3-{2-[(4-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenoxy}-3-phenylpropanoic acid;
      • 3-{3-[(4-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenoxy}-3-phenylpropanoic acid;
      • 3-{4-[(4-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenoxy}-3-phenylpropanoic acid;
      • 3-(2-{[4-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenoxy)—3-phenylpropanoic acid;
      • 3-(3-{[4-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenoxy)—3-phenylpropanoic acid;
      • 3-(4-{[4-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenoxy)—3-phenylpropanoic acid;
      • 3-(2-{[4-(1H-imidazol-2-ylamino)benzoyl]amino}phenoxy)-3-phenylpropanoic acid;
      • 3-(3-{[4-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenoxy)—3-phenylpropanoic acid;
      • 3-(4-{[4-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenoxy)—3-phenylpropanoic acid;
      • 3-phenyl-3-(2-{[4-(2-pyridinylamino)benzoyl]amino}phenoxy)propanoic acid;
      • 3-phenyl-3-(3-{[4-(2-pyridinylamino)benzoyl]amino}phenoxy)propanoic acid;
      • 3-phenyl-3-(4-{[4-(2-pyridinylamino)benzoyl]amino}phenoxy)propanoic acid;
      • 3-({2-[(3-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfinyl)--3-(3-pyridinyl)propanoic acid;
      • 3-({3-[(3-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfinyl)--3-(3-pyridinyl)propanoic acid;
      • 3-({4-[(3-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfinyl)--3-(3-pyridinyl)propanoic acid;
      • 3-({2-[(3-{[(benzylamino)imino)methyl]amino}benzoyl)amino]phenyl}sulfinyl-3-(3-pyridinyl)propanoic acid;
      • 3-({3-[(3-{[(benzylamino)imino)methyl]amino}benzoyl)amino]phenyl}sulfinyl-3-(3-pyridinyl)propanoic acid;
      • 3-({4-[(3-{[(benzylamino)imino)methyl]amino}benzoyl)amino]phenyl}sulfinyl-3-(3-pyridinyl)propanoic acid;
      • 3-{[2-({3-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfinyl}-3-(3--pyridinyl)propanoic acid;
      • 3-{[3-({3-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfinyl}-3-(3--pyridinyl)propanoic acid;
      • 3-{[4-({3-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfinyl}-3-(3--pyridinyl)propanoic acid;
      • 3-({2-[(3-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfinyl)-3-(3-pyridinyl)propanoic acid;
      • 3-({3-[(3-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfinyl)-3-(3-pyridinyl)propanoic acid;
      • 3-({4-[(3-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfinyl)-3-(3-pyridinyl)propanoic acid;
      • 3-[(2-{[3-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfinyl]-1-3-pyridinyl)propanoic acid;
      • 3-[(3-{[3-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfinyl]-1-3-pyridinyl)propanoic acid;
      • 3-[(4-{[3-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfinyl]-1-3-pyridinyl)propanoic acid;
      • 3-[(2-{[3-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfinyl]-3-(-3-pyridinyl)propanoic acid;
      • 3-[(3-{[3-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfinyl]-3-(-3-pyridinyl)propanoic acid;
      • 3-[(4-{[3-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfinyl]-3-(-3-pyridinyl)propanoic acid;
      • 3-(3-pyridinyl)-3-[(2-{[3-(2-pyridinylamino)benzoyl]amino]phenyl)sulfinyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(3-{[3-(2-pyridinylamino)benzoyl]amino]phenyl)sulfinyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(4-{[3-(2-pyridinylamino)benzoyl]amino]phenyl)sulfinyl]propanoic acid;
      • 3-({2-[(4-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfinyl)--3-(3-pyridinyl)propanoic acid;
      • 3-({3-[(4-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfinyl)--3-(3-pyridinyl)propanoic acid;
      • 3-({4-[(4-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfinyl)--3-(3-pyridinyl)propanoic acid;
      • 3-({2-[(4-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfinyl)-3-(3-pyridinyl)propanoic acid;
      • 3-({3-[(4-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfinyl)-3-(3-pyridinyl)propanoic acid;
      • 3-({4-[(4-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfinyl)-3-(3-pyridinyl)propanoic acid;
      • 3-{[2-({4-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfinyl}-3-(3--pyridinyl)propanoic acid;
      • 3-{[3-({4-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfinyl}-3-(3--pyridinyl)propanoic acid;
      • 3-{[4-({4-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfinyl}-3-(3--pyridinyl)propanoic acid;
      • 3-({2-[(4-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfinyl-3-(3-pyridinyl)propanoic acid;
      • 3-({3-[(4-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfinyl-3-(3-pyridinyl)propanoic acid;
      • 3-({4-[(4-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfinyl-3-(3-pyridinyl)propanoic acid;
      • 3-[(2-{[4-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfinyl]-3-(3-pyridinyl)propanoic acid;
      • 3-[(3-{[4-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfinyl]-3-(3-pyridinyl)propanoic acid;
      • 3-[(4-{[4-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfinyl]-3-(3-pyridinyl)propanoic acid;
      • 3-[(2-{[4-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfinyl]-3-(-3-pyridinyl)propanoic acid;
      • 3-[(3-{[4-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfinyl]-3-(-3-pyridinyl)propanoic acid;
      • 3-[(4-{[4-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfinyl]-3-(-3-pyridinyl)propanoic acid;
      • 3-(3-pyridinyl)-3-[(2-{[4-(2-pyridinylamino)benzoyl]amino}phenyl)sulfinyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(3-{[4-(2-pyridinylamino)benzoyl]amino}phenyl)sulfinyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(4-{[4-(2-pyridinylamino)benzoyl]amino}phenyl)sulfinyl]propanoic acid;
      • 3-({2-[(3-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfonyl)--3-(3-pyridinyl)propanoic acid;
      • 3-({3-[(3-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfonyl)--3-(3-pyridinyl)propanoic acid;
      • 3-({4-[(3-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfonyl)--3-(3-pyridinyl)propanoic acid;
      • 3-({2-[(3-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfonyl)-3-(3-pyridinyl)propanoic acid;
      • 3-({3-[(3-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfonyl)-3-(3-pyridinyl)propanoic acid;
      • 3-({4-[(3-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfonyl)-3-(3-pyridinyl)propanoic acid;
      • 3-{[2-({3-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfonyl}-3-(3--pyridinyl)propanoic acid;
      • 3-{[3-({3-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfonyl}-3-(3--pyridinyl)propanoic acid;
      • 3-{[4-({3-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfonyl}-3-(3--pyridinyl)propanoic acid;
      • 3-({2-[(3-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfonyl)-3-(3-pyridinyl)propanoic acid;
      • 3-({3-[(3-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfonyl)-3-(3-pyridinyl)propanoic acid;
      • 3-({4-[(3-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfonyl)-3-(3-pyridinyl)propanoic acid;
      • 3-[(2-{[3-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfonyl]-3-(3-pyridinyl)propanoic acid;
      • 3-[(3-{[3-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfonyl]-3-(3-pyridinyl)propanoic acid;
      • 3-[(4-{[3-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfonyl]-3-(3-pyridinyl)propanoic acid;
      • 3-[(2-{[3-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfonyl]-3-(-3-pyridinyl)propanoic acid;
      • 3-[(3-{[3-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfonyl]-3-(-3-pyridinyl)propanoic acid;
      • 3-[(4-{[3-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfonyl]-3-(-3-pyridinyl)propanoic acid;
      • 3-(3-pyridinyl)-3-[(2-{[3-(2-pyridinylamino)benzoyl]amino}phenyl)sulfonyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(3-{[3-(2-pyridinylamino)benzoyl]amino}phenyl)sulfonyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(4-{[3-(2-pyridinylamino)benzoyl]amino}phenyl)sulfonyl]propanoic acid;
      • 3-({2-[(4-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfonyl)--3-(3-pyridinyl)propanoic acid;
      • 3-({3-[(4-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfonyl)--3-(3-pyridinyl)propanoic acid;
      • 3-({4-[(4-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfonyl)--3-(3-pyridinyl)propanoic acid;
      • 3-({2-[(4-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfonyl)-3-(3-pyridinyl)propanoic acid;
      • 3-({3-[(4-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfonyl)-3-(3-pyridinyl)propanoic acid;
      • 3-({4-[(4-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfonyl)-3-(3-pyridinyl)propanoic acid;
      • 3-{[2-({4-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfonyl}-3-(3--pyridinyl)propanoic acid;
      • 3-{[3-({4-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfonyl}-3-(3--pyridinyl)propanoic acid;
      • 3-{[4-({4-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfonyl}-3-(3--pyridinyl)propanoic acid;
      • 3-({2-[(4-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfonyl)-3-(3-pyridinyl)propanoic acid;
      • 3-({3-[(4-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfonyl)-3-(3-pyridinyl)propanoic acid;
      • 3-({4-[(4-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfonyl)-3-(3-pyridinyl)propanoic acid;
      • 3-[(2-{[4-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfonyl]-3-(3-pyridinyl)propanoic acid;
      • 3-[(3-{[4-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfonyl]-3-(3-pyridinyl)propanoic acid;
      • 3-[(4-{[4-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfonyl]-3-(3-pyridinyl)propanoic acid;
      • 3-[(2-{[4-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfonyl]-3-(-3-pyridinyl)propanoic acid;
      • 3-[(3-{[4-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfonyl]-3-(-3-pyridinyl)propanoic acid;
      • 3-[(4-{[4-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfonyl]-3-(-3-pyridinyl)propanoic acid;
      • 3-(3-pyridinyl)-3-[(2-{[4-(2-pyridinylamino)benzoyl]amino}phenyl)sulfonyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(3-{[4-(2-pyridinylamino)benzoyl]amino}phenyl)sulfonyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(4-{[4-(2-pyridinylamino)benzoyl]amino}phenyl)sulfonyl]propanoic acid;
      • 3-({3-[(3-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfinyl)--3-phenylpropanoic acid;
      • 3-({2-[(3-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfinyl)--3-phenylpropanoic acid;
      • 3-({4-[(3-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfinyl)--3-phenylpropanoic acid;
      • 3-({2-[(3-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfinyl)-3-phenylpropanoic acid;
      • 3-({3-[(3-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfinyl)-3-phenylpropanoic acid;
      • 3-({4-[(3-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfinyl)-3-phenylpropanoic acid;
      • 3-{[2-({-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfinyl}-3-phenylpropanoic acid;
      • 3-{[3-({-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfinyl}-3-phenylpropanoic acid;
      • 3-{[4-({-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfinyl}-3-phenylpropanoic acid;
      • 3-({2-[(3-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfinyl)-3-pheynylpropanoic acid;
      • 3-({3-[(3-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfinyl)-3-pheynylpropanoic acid;
      • 3-({4-[(3-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfinyl)-3-pheynylpropanoic acid;
      • 3-[(2-{[3-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfinyl]-3-phenylpropanoic acid;
      • 3-[(3-{[3-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfinyl]-3-phenylpropanoic acid;
      • 3-[(4-{[3-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfinyl]-3-phenylpropanoic acid;
      • 3-[(2-{[3-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfinyl]-3-phenylpropanoic acid;
      • 3-[(3-{[3-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfinyl]-3-phenylpropanoic acid;
      • 3-[(4-{[3-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfinyl]-3-phenylpropanoic acid;
      • 3-phenyl-3-[(2-{[3-(2-pyridinylamino)benzoyl]amino}phenyl)sulfinyl]-propanoic acid;
      • 3-phenyl-3-[(3-{[3-(2-pyridinylamino)benzoyl]amino}phenyl)sulfinyl]-propanoic acid;
      • 3-phenyl-3-[(4-{[3-(2-pyridinylamino)benzoyl]amino}phenyl)sulfinyl]-propanoic acid;
      • 3-({2-[(4-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfinyl)--3-phenylpropanoic acid;
      • 3-({3-[(4-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfinyl)--3-phenylpropanoic acid;
      • 3-({4-[(4-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfinyl)--3-phenylpropanoic acid;
      • 3-({2-[(4-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfinyl)-3-phenylpropanoic acid;
      • 3-({3-[(4-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfinyl)-3-phenylpropanoic acid;
      • 3-({4-[(4-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfinyl)-3-phenylpropanoic acid;
      • 3-{[2-({4-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfinyl}-3-phenylpropanoic acid;
      • 3-{[3-({4-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfinyl}-3-phenylpropanoic acid;
      • 3-{[4-({4-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfinyl}-3-phenylpropanoic acid;
      • 3-({2-[(4-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfinyl)-3-phenylpropanoic acid;
      • 3-({3-[(4-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfinyl)-3-phenylpropanoic acid;
      • 3-({4-[(4-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfinyl)-3-phenylpropanoic acid;
      • 3-[(2-{[4-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfinyl]-3-phenylpropanoic acid;
      • 3-[(3-{[4-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfinyl]-3-phenylpropanoic acid;
      • 3-[(4-{[4-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfinyl]-3-phenylpropanoic acid;
      • 3-[(2-{[4-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfinyl]-3-phenylpropanoic acid;
      • 3-[(3-{[4-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfinyl]-3-phenylpropanoic acid;
      • 3-[(4-{[4-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfinyl]-3-phenylpropanoic acid;
      • 3-phenyl-3-[(2-{[4-(2-pyridinylamino)benzoyl]amino}phenyl)sulfinyl]-propanoic acid;
      • 3-phenyl-3-[(3-{[4-(2-pyridinylamino)benzoyl]amino}phenyl)sulfinyl]-propanoic acid;
      • 3-phenyl-3-[(4-{[4-(2-pyridinylamino)benzoyl]amino}phenyl)sulfinyl]-propanoic acid;
      • 3-({3-[(3-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfonyl)--3-phenylpropanoic acid;
      • 3-({2-[(3-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfonyl)--3-phenylpropanoic acid;
      • 3-({4-[(3-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfonyl)--3-phenylpropanoic acid;
      • 3-({2-[(3-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfonyl)-3-phenylpropanoic acid;
      • 3-({3-[(3-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfonyl)-3-phenylpropanoic acid;
      • 3-({4-[(3-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfonyl)-3-phenylpropanoic acid;
      • 3-{[2-({3-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfonyl}-3-phenylpropanoic acid;
      • 3-{[3-({3-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfonyl}-3-phenylpropanoic acid;
      • 3-{[4-({3-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfonyl}-3-phenylpropanoic acid;
      • 3-({2-[(3-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfonyl)-3-phenylpropanoic acid;
      • 3-({3-[(3-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfonyl)-3-phenylpropanoic acid;
      • 3-({4-[(3-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfonyl)-3-phenylpropanoic acid;
      • 3-[(2-{[3-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfonyl]-3-phenylpropanoic acid;
      • 3-[(3-{[3-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfonyl]-3-phenylpropanoic acid;
      • 3-[(4-{[3-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfonyl]-3-phenylpropanoic acid;
      • 3-[(2-{[3-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfonyl]-3-phenylpropanoic acid;
      • 3-[(3-{[3-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfonyl]-3-phenylpropanoic acid;
      • 3-[(4-{[3-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfonyl]-3-phenylpropanoic acid;
      • 3-phenyl-3-[(2-{[3-(2-pyridinylamino)benzoyl]amino}phenyl)sulfonyl]-propanoic acid;
      • 3-phenyl-3-[(3-{[3-(2-pyridinylamino)benzoyl]amino}phenyl)sulfonyl]-propanoic acid;
      • 3-phenyl-3-[(4-{[3-(2-pyridinylamino)benzoyl]amino}phenyl)sulfonyl]-propanoic acid;
      • 3-({2-[(4-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfonyl)--3-phenylpropanoic acid;
      • 3-({3-[(4-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfonyl)--3-phenylpropanoic acid;
      • 3-({4-[(4-{[amino(imino)methyl]amino}benzoyl)amino]phenyl}sulfonyl)--3-phenylpropanoic acid;
      • 3-({2-[(4-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfonyl)-3-phenylpropanoic acid;
      • 3-({3-[(4-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfonyl)-3-phenylpropanoic acid;
      • 3-({4-[(4-{[(benzylamino)(imino)methyl]amino}benzoyl)amino]phenyl}sulfonyl)-3-phenylpropanoic acid;
      • 3-{[2-({4-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfonyl}-3-phenylpropanoic acid;
      • 3-{[3-({4-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfonyl}-3-phenylpropanoic acid;
      • 3-{[4-({4-[(aminocarbonyl)amino]benzoyl}amino)phenyl]sulfonyl}-3-phenylpropanoic acid;
      • 3-({2-[(4-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfonyl)-3-phenylpropanoic acid;
      • 3-({3-[(4-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfonyl)-3-phenylpropanoic acid;
      • 3-({4-[(4-{[(benzylamino)carbonyl]amino}benzoyl)amino]phenyl}sulfonyl)-3-phenylpropanoic acid;
      • 3-[(2-{[4-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfonyl]-3-phenylpropanoic acid;
      • 3-[(3-{[4-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfonyl]-3-phenylpropanoic acid;
      • 3-[(4-{[4-(4,5-dihydro-1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfonyl]-3-phenylpropanoic acid;
      • 3-[(2-{[4-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfonyl]-3-phenylpropanoic acid;
      • 3-[(3-{[4-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfonyl]-3-phenylpropanoic acid;
      • 3-[(4-{[4-(1H-imidazol-2-ylamino)benzoyl]amino}phenyl)sulfonyl]-3-phenylpropanoic acid;
      • 3-phenyl-3-[(2-{[4-(2-pyridinylamino)benzoyl]amino}phenyl)sulfonyl]-propanoic acid;
      • 3-phenyl-3-[(3-{[4-(2-pyridinylamino)benzoyl]amino}phenyl)sulfonyl]-propanoic acid;
      • 3-phenyl-3-[(4-{[4-(2-pyridinylamino)benzoyl]amino}phenyl)sulfonyl]-propanoic acid;
      • 3-phenyl-3-[(2-{[4-(2-pyridinylamino)benzoyl]amino}phenyl)sulfonyl]-propanoic acid;
      • 3-phenyl-3-[(3-{[4-(2-pyridinylamino)benzoyl]amino}phenyl)sulfonyl]-propanoic acid;
      • 3-phenyl-3-[(4-{[4-(2-pyridinylamino)benzoyl]amino}phenyl)sulfonyl]-propanoic acid;
      • 3-(3-pyridinyl)-3-(2-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenoxy)propanoic acid;
      • 3-(3-pyridinyl)-3-(3-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenoxy)propanoic acid;
      • 3-(3-pyridinyl)-3-(4-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenoxy)propanoic acid;
      • 3-(3-pyridinyl)-3-(2-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenoxy)propanoic acid;
      • 3-(3-pyridinyl)-3-(3-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenoxy)propanoic acid;
      • 3-(3-pyridinyl)-3-(4-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenoxy)propanoic acid;
      • 3-phenyl-3-(2-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenoxy)propanoic acid;
      • 3-phenyl-3-(3-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenoxy)propanoic acid;
      • 3-phenyl-3-(4-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenoxy)propanoic acid;
      • 3-phenyl-3-(2-{[4-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenoxy)propanoic acid;
      • 3-phenyl-3-(3-{[4-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenoxy)propanoic acid;
      • 3-phenyl-3-(4-{[4-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenoxy)propanoic acid;
      • 3-(3-pyridinyl)-3-[(2-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfanyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(3-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfanyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(4-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfanyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(2-{[4-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfanyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(3-{[4-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfanyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(4-{[4-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfanyl]propanoic acid;
      • 3-phenyl-3-[(2-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfanyl]propanoic acid;
      • 3-phenyl-3-[(3-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfanyl]propanoic acid;
      • 3-phenyl-3-[(4-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfanyl]propanoic acid;
      • 3-phenyl-3-[(2-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfanyl]propanoic acid;
      • 3-phenyl-3-[(3-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfanyl]propanoic acid;
      • 3-phenyl-3-[(4-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfanyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(2-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfinyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(3-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfinyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(4-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfinyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(2-{[4-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfinyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(3-{[4-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfinyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(4-{[4-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfinyl]propanoic acid;
      • 3-phenyl-3-[(2-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfinyl]propanoic acid;
      • 3-phenyl-3-[(3-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfinyl]propanoic acid;
      • 3-phenyl-3-[(2-{[4-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfinyl]propanoic acid;
      • 3-phenyl-3-[(2-{[4-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfinyl]propanoic acid;
      • 3-phenyl-3-[(3-{[4-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfinyl]propanoic acid;
      • 3-phenyl-3-[(4-{[4-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfinyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(2-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfonyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(3-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfonyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(4-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfonyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(2-{[4-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfonyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(3-{[4-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfonyl]propanoic acid;
      • 3-(3-pyridinyl)-3-[(4-{[4-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfonyl]propanoic acid;
      • 3-phenyl-3-[(2-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfonyl]propanoic acid;
      • 3-phenyl-3-[(3-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfonyl]propanoic acid;
      • 3-phenyl-3-[(4-{[3-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfonyl]propanoic acid;
      • 3-phenyl-3-[(2-{[4-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfonyl]propanoic acid;
      • 3-phenyl-3-[(3-{[4-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfonyl]propanoic acid;
      • 3-phenyl-3-[(4-{[4-(1,4,5,6-tetrahydro-2-pyrimidinylamino)benzoyl]amino}phenyl)sulfonyl]propanoic acid;
      • either as a free acid or a salt thereof, in particular the hydrochloride or the trifluroacetate.
  • Having thus described several aspects of at least one embodiment of this invention, it is to be appreciated various alterations, modifications, and improvements will readily occur to those skilled in the art. Such alterations, modifications, and improvements are intended to be part of this disclosure, and are intended to be within the spirit and scope of the invention. Accordingly, the foregoing description and drawings are by way of example only.
  • All of the references cited herein, including published patents and patent applications are hereby incorporated in their entirety.

Claims (13)

What is claimed is:
1-11. (canceled)
12. A method for treating a proteinuric disorder comprising:
administering to a patient in need thereof, a peptide αvβ3 integrin inhibitor comprising an RGD binding sequence, in an amount effective to treat the proteinuric disorder.
13. The method of claim 12, wherein the proteinuric disorder is a kidney proteinuric disorder.
14. The method of claim 13, wherein the kidney proteinuric disorder is selected from the group consisting of:
Diabetic nephropathy,
Nephrotic syndromes (i.e. intrinsic renal failure),
Nephritic syndromes,
Toxic lesions of kidneys,
Glomerular diseases, such as membranous glomerulonephritis,
Focal segmental glomerulosclerosis (FSGS),
IgA nephropathy,
IgM nephropathy,
Membranoproliferative glomerulonephritis,
Membranous nephropathy,
Minimal change disease,
Hypertensive nephrosclerosis, and
Interstitial nephritis.
15. The method of claim 14, wherein the kidney proteinuric disorder is diabetic nephropathy.
16. The method of claim 13, wherein the kidney proteinuric disorder is selected from the group consisting of:
Pre-eclampsia,
Eclampsia,
Collagen vascular diseases,
Dehydration,
Strenuous exercise,
Stress,
Benign Orthostatic (postural) proteinuria,
Sarcoidosis,
Alport's syndrome,
Diabetes mellitus,
Fabry's disease,
Infections,
Aminoaciduria,
Fanconi syndrome,
Heavy metal ingestion,
Sickle cell disease,
Hemoglobinuria,
Multiple myeloma,
Myoglobinuria,
Organ rejection,
Ebola hemorrhagic fever, and
Nail Patella Syndrome.
17. The method of claim 12, wherein the peptide that contains an RGD binding sequence is cyclo-[Arg-Gly-Asp-D-Phe-Val].
18. The method of claim 12, wherein the patient has proteinuria associated with podocytes exhibiting increased uPAR-induced αvβ3 integrin activation.
19. The method of claim 18, further comprising detecting increased podocyte αvβ3 integrin activation.
20. The method of claim 19, wherein detecting increased podocyte αvβ3 integrin activation is performed using an antibody-based assay.
21. The method of claim 12, further comprising measuring the patient's urinary protein prior to and after the administration of the αvβ3 integrin inhibitor, wherein a reduction in the amount of urinary protein indicates that the proteinuria is reduced.
22. The method of claim 21, wherein measuring the patient's urinary protein prior to administration of the peptide comprises detecting 150 mg or more of protein in the urine of the subject.
23. The method of claim 12, wherein the peptide is administered to the subject intravenously.
US15/093,588 2007-11-08 2016-04-07 Methods and Compositions for the Treatment of Proteinuric Diseases Abandoned US20160296592A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US15/093,588 US20160296592A1 (en) 2007-11-08 2016-04-07 Methods and Compositions for the Treatment of Proteinuric Diseases
US15/979,242 US20180344803A1 (en) 2007-11-08 2018-05-14 Methods and Compositions For the Treatment of Proteinuric Diseases

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US244507P 2007-11-08 2007-11-08
PCT/US2008/012541 WO2009061448A2 (en) 2007-11-08 2008-11-07 Methods and compositions for the treatment of proteinuric diseases
US74009010A 2010-08-10 2010-08-10
US15/093,588 US20160296592A1 (en) 2007-11-08 2016-04-07 Methods and Compositions for the Treatment of Proteinuric Diseases

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
US12/740,090 Division US9345739B2 (en) 2007-11-08 2008-11-07 Methods and compositions for the treatment of proteinuric diseases
PCT/US2008/012541 Division WO2009061448A2 (en) 2007-11-08 2008-11-07 Methods and compositions for the treatment of proteinuric diseases

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/979,242 Division US20180344803A1 (en) 2007-11-08 2018-05-14 Methods and Compositions For the Treatment of Proteinuric Diseases

Publications (1)

Publication Number Publication Date
US20160296592A1 true US20160296592A1 (en) 2016-10-13

Family

ID=40626390

Family Applications (3)

Application Number Title Priority Date Filing Date
US12/740,090 Active US9345739B2 (en) 2007-11-08 2008-11-07 Methods and compositions for the treatment of proteinuric diseases
US15/093,588 Abandoned US20160296592A1 (en) 2007-11-08 2016-04-07 Methods and Compositions for the Treatment of Proteinuric Diseases
US15/979,242 Abandoned US20180344803A1 (en) 2007-11-08 2018-05-14 Methods and Compositions For the Treatment of Proteinuric Diseases

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US12/740,090 Active US9345739B2 (en) 2007-11-08 2008-11-07 Methods and compositions for the treatment of proteinuric diseases

Family Applications After (1)

Application Number Title Priority Date Filing Date
US15/979,242 Abandoned US20180344803A1 (en) 2007-11-08 2018-05-14 Methods and Compositions For the Treatment of Proteinuric Diseases

Country Status (5)

Country Link
US (3) US9345739B2 (en)
EP (2) EP2217238B1 (en)
JP (3) JP5637855B2 (en)
ES (1) ES2457822T3 (en)
WO (1) WO2009061448A2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022125776A2 (en) 2020-12-09 2022-06-16 Siwa Corporation Methods and compositions for treating kidney diseases

Families Citing this family (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9345739B2 (en) * 2007-11-08 2016-05-24 The General Hospital Corporation Methods and compositions for the treatment of proteinuric diseases
US20110212083A1 (en) * 2008-11-06 2011-09-01 University Of Miami Office Of Technology Transfer Role of soluble upar in the pathogenesis of proteinuric kidney disease
DE102009000854A1 (en) * 2009-02-13 2010-08-19 Ernst-Moritz-Arndt-Universität Greifswald Osteopontin-receptor-ligand for binding osteopontin receptors of podocytes of the renal corpuscles, useful for treating chronic renal failure
WO2012027745A1 (en) * 2010-08-27 2012-03-01 University Of Miami Treatment of renal diseases
US9867923B2 (en) 2011-05-09 2018-01-16 University Of Miami Reducing soluble urokinase receptor in the circulation
MX2015002465A (en) 2012-08-31 2015-11-06 Univ North Carolina Monoclonal antibodies for enhancing or inhibiting insulin-like growth factor 1 (igf-1).
WO2017053976A1 (en) * 2015-09-25 2017-03-30 The General Hospital Corporation DIAGNOSTIC ASSAYS FOR SUPAR-β3 INTEGRIN DRIVEN KIDNEY DISEASES
EP3294716B1 (en) 2015-12-30 2020-04-15 Saint Louis University Meta-azacyclic amino benzoic acid derivatives as pan integrin antagonists
GB201916071D0 (en) * 2019-11-05 2019-12-18 Univ Bristol Treatment for nephrotic syndrome
EP4114429A2 (en) * 2020-03-06 2023-01-11 Allegro Pharmaceuticals, LLC Treatments for improving or lessening impairment of mitochondrial function

Family Cites Families (66)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE3920358A1 (en) 1989-06-22 1991-01-17 Behringwerke Ag BISPECIFIC AND OLIGO-SPECIFIC, MONO- AND OLIGOVALENT ANTI-BODY CONSTRUCTS, THEIR PRODUCTION AND USE
WO1996026190A1 (en) 1995-02-22 1996-08-29 Smithkline Beecham Corporation Integrin receptor antagonists
CA2230209A1 (en) 1995-08-30 1997-03-06 G.D. Searle & Co. Meta-guanidine, urea, thiourea or azacyclic amino benzoic acid derivatives as integrin antagonists
KR19990076877A (en) 1995-12-29 1999-10-25 스티븐 베네티아너 Bitronectin receptor antagonist
KR19990076876A (en) 1995-12-29 1999-10-25 스티븐 베네티아너 Bitronectin receptor antagonist
UA60311C2 (en) 1996-10-02 2003-10-15 Смітклайн Бічам Корпорейшн Vitronectin receptor antagonists
US5942492A (en) 1996-11-12 1999-08-24 Angstrom Pharmaceuticals, Inc. Cyclic peptides that bind to urokinase-type plasminogen activator receptor
US6794358B1 (en) 1997-03-28 2004-09-21 Brigham And Women's Hospital Peptide ligands of the urokinase receptor
CA2297910A1 (en) 1997-07-25 1999-02-04 Smithkline Beecham Corporation Vitronectin receptor antagonist
JP2001511452A (en) 1997-08-04 2001-08-14 スミスクライン・ビーチャム・コーポレイション Integrin receptor antagonist
DE69930723T2 (en) 1998-01-23 2006-11-16 Merck Patent Gmbh USE OF ANTIBODY 271.14D9.F8 (DSM ACC2331) TO REDUCE BINDING BETWEEN ALPHAVBETA6 INTEGRIN AND FIBRONECTIN IN VITRO
US6852318B1 (en) 1998-05-08 2005-02-08 The Regents Of The University Of California Methods for detecting and inhibiting angiogenesis
US6492325B1 (en) 1998-05-22 2002-12-10 Boys Town National Research Hospital Use of α1β1 integrin receptor inhibitors and TGF-β1 inhibitors in the treatment of kidney disease
IT1299575B1 (en) 1998-07-23 2000-03-16 Demetrio Biancucci SOLE FOR FOOTWEAR EQUIPPED WITH RAMPINI OR HALF NAILS ABLE TO PASS FROM A POSITION IN CONTACT WITH THE GROUND TO A POSITION
EP0982036A1 (en) 1998-08-28 2000-03-01 Wilex Biotechnology GmbH Modulation of Beta2 -integrin-mediated cell adhesion
US6451972B1 (en) 1998-11-16 2002-09-17 Sigma-Tau Industrie Farmaceutiche Riunite S.P.A. Peptido-mimetic compounds containing RGD sequence useful as integrin inhibitors
PT1153014E (en) 1999-02-20 2005-02-28 Merck Patent Gmbh BETA-ALANINE DERIVATIVES
ES2274781T3 (en) 1999-04-13 2007-06-01 Wilex Ag DIAGNOSTIC AND THERAPEUTIC EMPLOYMENT OF ANTIBODIES AGAINST THE RECEIVER OF UROCINASE.
DZ3263A1 (en) 1999-06-02 2000-12-07 Merck & Co Inc ALPHA V INTEGRIN RECEPTOR ANTAGONISTS
AU7099900A (en) 1999-09-03 2001-04-10 Brigham And Women's Hospital Peptides that bind to urokinase receptor
EP1700606A1 (en) 1999-09-14 2006-09-13 Biogen Idec MA, Inc. Therapies for chronic renal failure using one or more integrin antagonists
US6955808B2 (en) 1999-09-24 2005-10-18 Uab Research Foundation Capsid-modified recombinant adenovirus and methods of use
DE10028575A1 (en) 2000-06-14 2002-03-14 Basf Ag integrin
GB9928882D0 (en) 1999-12-08 2000-02-02 Zeneca Ltd Assay method
US6825167B1 (en) 2000-04-03 2004-11-30 Regents Of The University Of Minnesota Genetic modification of endostatin
GB0011817D0 (en) 2000-05-16 2000-07-05 Pharmacia & Upjohn Spa Antagonists of integrin receptors
AU2001269821A1 (en) 2000-06-15 2001-12-24 Barbara Chen Cycloalkyl alkanoic acids as integrin receptor antagonists
AU2001268490A1 (en) 2000-06-15 2001-12-24 Pharmacia Corporation Dihydrostilbene alkanoic acid derivatives useful as vitronectin antagonists
US6649597B1 (en) 2000-06-22 2003-11-18 The University Of Iowa Research Foundation Targeting vector to the urokinase plasminogen activator receptor
GB0016791D0 (en) 2000-07-07 2000-08-30 Novartis Ag Organic compounds
US7163681B2 (en) * 2000-08-07 2007-01-16 Centocor, Inc. Anti-integrin antibodies, compositions, methods and uses
WO2002018377A1 (en) 2000-08-29 2002-03-07 Pharmacia Corporation Compounds containing a bicyclic ring system useful as alpha v beta 3 antagonists
US6531494B1 (en) 2001-08-29 2003-03-11 Pharmacia Corporation Gem-substituted αvβ3 antagonists
US6720327B2 (en) 2000-09-27 2004-04-13 Pharmacia Corporation Lactone integrin antagonists
US20020072500A1 (en) 2000-09-27 2002-06-13 Thomas Rogers Hydroxy acid integrin antagonists
EP1197553A1 (en) 2000-10-12 2002-04-17 A3D GmbH, Antisense Design &amp; Drug Development Antisense nucleic acid against alphaV integrin
DE10112771A1 (en) 2001-03-16 2002-09-26 Merck Patent Gmbh New 3-acylamino-3-phenyl-propionic acid derivatives, are integrin inhibitors useful e.g. for treating thrombosis, cardiac infarction, angina pectoris, tumor diseases, inflammation, osteoporosis or infections
US6794385B2 (en) 2001-08-08 2004-09-21 Pharmacia & Upjohn, S.P.A. Benzoxazine derivatives useful as integrin receptor antagonists
DE10204789A1 (en) * 2002-02-06 2003-08-14 Merck Patent Gmbh Inhibitors of the integrin alpha¶v¶beta6
US7138230B2 (en) 2002-12-06 2006-11-21 Renovar, Inc. Systems and methods for characterizing kidney diseases
ITRM20020402A1 (en) 2002-07-29 2004-01-29 Sigma Tau Ind Farmaceuti FLUORO-ALCHIL-CYCLOPEPTIDES DERIVATIVES WITH ANTI-INTEGRINE ACTIVITIES.
AU2003259891A1 (en) * 2002-08-16 2004-03-19 Janssen Pharmaceutica N.V. Piperidinyl compounds that selectively bind integrins
AU2003299807A1 (en) 2002-12-20 2004-07-22 Pharmacia Corporation Heteroarylalkanoic acids as integrin receptor antagonists
CA2510050A1 (en) 2002-12-20 2004-07-22 Pharmacia Corporation The r-isomer of beta amino acid compounds as integrin receptor antagonists derivatives
CN1747966A (en) 2003-02-06 2006-03-15 默克专利有限公司 Peptidic sulfonamides
KR100568755B1 (en) 2003-04-03 2006-04-07 주식회사 리젠 바이오텍 Agent for inhibiting angiogenesis, containing peptides which comprise YH motif as effective component
JP2006522158A (en) * 2003-04-03 2006-09-28 アルナイラム ファーマシューティカルズ インコーポレイテッド iRNA complex
US7560244B2 (en) * 2003-06-04 2009-07-14 Joslin Diabetes Center, Inc. Method of evaluating a subject for risk or predisposition of reduced renal function over time
US7365154B2 (en) 2003-06-23 2008-04-29 Centocor, Inc. Peptides antagonistic to an anti-angiogenic antibody and uses therefor
ITMI20031476A1 (en) 2003-07-18 2005-01-19 Univ Degli Studi Milano PEPTIDO-MIMETIC COMPOUNDS WITH A AZABICICLOALCANIC STRUCTURE INCLUDING THE RGD SEQUENCE
DE602005020137D1 (en) 2004-05-07 2010-05-06 Univ North Carolina METHOD FOR REINFORCING OR INHIBITING THE INSULINARY GROWTH FACTOR-I
US8187595B2 (en) 2004-05-07 2012-05-29 The University Of North Carolina At Chapel Hill Monoclonal antibodies for enhancing or inhibiting insulin-like growth factor-I
PL1765397T3 (en) 2004-05-25 2013-04-30 Tactic Pharma Llc Inhibitory antibodies binding the complex of urokinase-type plasminogen activator (uPA) and its receptor (uPAR)
MX2007004535A (en) 2004-10-14 2007-06-08 Pharmacia Corp Biphenyl integrin antagonists.
US7670817B2 (en) 2005-11-08 2010-03-02 The General Hospital Corporation Dynamin mediated diseases and associated methods and products
US20070244046A1 (en) 2006-03-29 2007-10-18 Margarita Gutova Identification and characterization of cancer stem cells and methods of use
TW200813091A (en) 2006-04-10 2008-03-16 Amgen Fremont Inc Targeted binding agents directed to uPAR and uses thereof
US20080020979A1 (en) * 2006-06-09 2008-01-24 Rapraeger Alan C Peptides of Syndecan-1 For Inhibiting Angiogenesis
US8815519B2 (en) 2006-12-22 2014-08-26 Hvidovre Hospital Method for predicting cancer and other diseases
US20100158858A1 (en) * 2007-04-13 2010-06-24 Liangxian Cao Administration of carboline derivatives useful in the treatment of cancer and other diseases
AU2008316793A1 (en) 2007-10-26 2009-04-30 Genentech, Inc. Inhibition of urokinase-type plasminogen activator (uPA) activity
US9345739B2 (en) * 2007-11-08 2016-05-24 The General Hospital Corporation Methods and compositions for the treatment of proteinuric diseases
US8236319B2 (en) 2008-04-30 2012-08-07 Immunogen, Inc. Cross-linkers and their uses
US20110212083A1 (en) 2008-11-06 2011-09-01 University Of Miami Office Of Technology Transfer Role of soluble upar in the pathogenesis of proteinuric kidney disease
WO2012027745A1 (en) 2010-08-27 2012-03-01 University Of Miami Treatment of renal diseases
MX2015002465A (en) * 2012-08-31 2015-11-06 Univ North Carolina Monoclonal antibodies for enhancing or inhibiting insulin-like growth factor 1 (igf-1).

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022125776A2 (en) 2020-12-09 2022-06-16 Siwa Corporation Methods and compositions for treating kidney diseases

Also Published As

Publication number Publication date
ES2457822T3 (en) 2014-04-29
JP2014205690A (en) 2014-10-30
US20100297139A1 (en) 2010-11-25
EP2217238B1 (en) 2014-03-12
EP2730282A1 (en) 2014-05-14
JP5637855B2 (en) 2014-12-10
US20180344803A1 (en) 2018-12-06
JP2011503065A (en) 2011-01-27
EP2217238A4 (en) 2011-06-29
US9345739B2 (en) 2016-05-24
WO2009061448A9 (en) 2009-08-13
EP2217238A2 (en) 2010-08-18
JP2016104767A (en) 2016-06-09
WO2009061448A2 (en) 2009-05-14

Similar Documents

Publication Publication Date Title
US20180344803A1 (en) Methods and Compositions For the Treatment of Proteinuric Diseases
US10131658B2 (en) Anti-alphavbeta1 integrin compounds and methods
KR102449308B1 (en) Compositions and methods for the treatment of cardiovascular disease
US11685717B2 (en) Polypeptide integrin antagonists
JP2021193106A (en) Complement component c5 antibodies
RU2547592C2 (en) αvβ3 INTEGRIN SELECTIVE POLYPEPTIDES CONJUGATED WITH VARIANT OF HUMAN SERUM ALBUMIN (HSA), AND THEIR PHARMACEUTICAL APPLICATIONS
JP2021530223A (en) Tissue-specific Wnt signaling-enhancing molecule and its use
JP2020078304A (en) Complement factor Bb antibody
CN105263948A (en) Inhibition of pulmonary fibrosis with Nutlin-3a and peptides
AU2843900A (en) Proteins that bind angiogenesis-inhibiting proteins, compositions and methods of use thereof
JP2014065747A (en) METHOD OF TREATING FIBROSIS USING ANTAGONIST OF INTEGRIN α-4 SUBUNIT
KR20070115884A (en) Anti-alpha9 integrin antibody and the use thereof
US20210292436A1 (en) Methods of Inhibiting MASP-2 for the Treatment and/or Prevention of Coronavirus-induced Acute Respiratory Distress Syndrome
JP2004519215A (en) Claudin polypeptide
CN101990548B (en) Synthetic scfv analogue to the 6313/g2 (anti angiotensin ii type 1 receptor) monoclonal antibody variable regions
US20120237504A1 (en) Compositions and methods for treating inflammation and fibrosis
EP3745862A1 (en) Methods of treating fibrotic pathologies
WO2021021922A1 (en) Compounds and methods for treating fibrotic pathologies
US20210309704A1 (en) Wnt signaling agonist molecules
US8080252B2 (en) Compounds and methods of modulating angiogenesis
CN110914286B (en) Autophagy inhibitor
Nanthakumar et al. Targeting the αv Integrins in Fibroproliferative Disease
Vicari Evaluation of VEGF peptide mimics as inhibitors of angiogenesis
Pirazzoli Role of vitronectin interaction in the biology of the urokinase receptor
EA045598B1 (en) METHODS FOR INHIBITION OF ANGIOGENESIS IN A PATIENT

Legal Events

Date Code Title Description
AS Assignment

Owner name: THE GENERAL HOSPITAL CORPORATION, MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:REISER, JOCHEN;REEL/FRAME:039162/0687

Effective date: 20110527

STCB Information on status: application discontinuation

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

AS Assignment

Owner name: THE GENERAL HOSPITAL CORPORATION, MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:REISER, JOCHEN;REEL/FRAME:046358/0333

Effective date: 20110527