EP0831795A2 - Phosphatase inhibitoren - Google Patents

Phosphatase inhibitoren

Info

Publication number
EP0831795A2
EP0831795A2 EP96918441A EP96918441A EP0831795A2 EP 0831795 A2 EP0831795 A2 EP 0831795A2 EP 96918441 A EP96918441 A EP 96918441A EP 96918441 A EP96918441 A EP 96918441A EP 0831795 A2 EP0831795 A2 EP 0831795A2
Authority
EP
European Patent Office
Prior art keywords
compounds
compound
receptor
cells
cell
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.)
Withdrawn
Application number
EP96918441A
Other languages
English (en)
French (fr)
Inventor
Gerald Mcmahon
Klaus P. Hirth
Peng Cho Tang
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.)
Sugen LLC
Original Assignee
Sugen LLC
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 Sugen LLC filed Critical Sugen LLC
Publication of EP0831795A2 publication Critical patent/EP0831795A2/de
Withdrawn legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/38Heterocyclic compounds having sulfur as a ring hetero atom
    • A61K31/381Heterocyclic compounds having sulfur as a ring hetero atom having five-membered rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • A61K31/137Arylalkylamines, e.g. amphetamine, epinephrine, salbutamol, ephedrine or methadone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • A61K31/22Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acyclic acids, e.g. pravastatin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • A61K31/22Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acyclic acids, e.g. pravastatin
    • A61K31/222Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acyclic acids, e.g. pravastatin with compounds having aromatic groups, e.g. dipivefrine, ibopamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/34Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide
    • A61K31/341Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having five-membered rings with one oxygen as the only ring hetero atom, e.g. isosorbide not condensed with another ring, e.g. ranitidine, furosemide, bufetolol, muscarine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/35Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom
    • A61K31/352Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom condensed with carbocyclic rings, e.g. methantheline 
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/42Oxazoles
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/425Thiazoles
    • A61K31/4261,3-Thiazoles
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/433Thidiazoles
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • 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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • 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
    • A61K31/53Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with three nitrogens as the only ring hetero atoms, e.g. chlorazanil, melamine
    • 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
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole

Definitions

  • the present invention relates to compounds capable of modulating and/or regulating the activity of phosphotyrosine phosphatases that regulate signal transduction.
  • the present invention relates to the use of such compounds for the treatment of diseases caused by dysfunctional signal transduction.
  • Cellular signal transduction is a fundamental mechanism whereby external stimuli that regulate diverse cellular processes are relayed to the interior of cells.
  • the biochemical pathways through which signals are relayed within cells comprise a circuitry of directly or functionally connected interactive proteins.
  • One of the key biochemical mechanisms of signal transduction involves the reversible phosphorylation of tyrosine residues on proteins.
  • the phosphorylation state of a protein may affect its conformation and/or enzymic activity as well as its cellular location.
  • the phosphorylation state of a protein is modified through the reciprocal actions of protein tyrosine kinases (PTKs) and protein tyrosine phosphatases (PTPs) at various specific tyrosine residues.
  • PTKs protein tyrosine kinases
  • PTPs protein tyrosine phosphatases
  • a common mechanism by which receptors regulate cell function is through an inducible tyrosine kinase activity which is either endogenous to the receptor or is imparted by other proteins that become associated with the receptor.
  • Protein tyrosine kinases comprise a large family of transmembrane receptor and intracellular enzymes with multiple functional domains (Taylor et al . , 1992 Ann. Rev. Cell Biol. 8:429-62).
  • the binding of ligand allosterically transduces a signal across the cell membrane where the cytoplasmic portion of the PTKs initiates a cascade of molecular interactions that disseminate the signal throughout the cell and into the nucleus.
  • RPTKs receptor protein tyrosine kinase
  • EGFR epidermal growth factor receptor
  • PDGFR platelet-derived growth factor receptor
  • CPTKs Cytoplasmic protein tyrosine kinases
  • Src kinases e . g. , src, lck, fyn
  • cytokines e.g., IL-2, IL-3, IL-6, erythropoietin
  • interferons e.g., IL-6, erythropoietin
  • antigen receptors e.g., IL-6, erythropoietin
  • the protein tyrosine phosphatases comprise a family of transmembrane and cytoplasmic enzymes, possessing at least an approximately 230 amino acid catalytic domain containing a highly conserved active site with the consensus motif [I/V]HCXAGXXR[S/T]G.
  • the substrates of PTPs may be PTKs which possess phosphotyrosine residues or the substrates of PTKs.
  • Transmembrane or receptor-like PTPs possess an extracellular domain, a single transmembrane domain, and one or two catalytic domains followed by a short cytoplasmic tail.
  • the extracellular domains of these RPTPs are highly divergent, with small glycosylated segments (e .g. , RPTP ⁇ , RPTPe) , tandem repeats of immunoglobulin-like and/or fibronectin type III domains (e . g. , LAR) or carbonic anhydrase like domains (e . g. , RPTP ⁇ , RPTP ⁇ ) .
  • Intracellular or cytoplasmic PTPs such as PTP1C, PTPID
  • CPTPs Intracellular or cytoplasmic PTPs
  • PTP1C a hemopoietic cell CPTP is characterized by two Src- homology 2 (SH2) domains that recognize short peptide motifs bearing phosphotyrosine (pTyr) .
  • SH2 Src- homology 2
  • SH2- containing proteins are able to bind pTyr sites in activated receptors and cytoplasmic phosphoproteins.
  • Another conserved domain known as SH3 binds to proteins with proline-rich regions.
  • a third type known as pleckstrin-ho ology (PH) domain has also been identified.
  • These modular domains have been found in both CPTKs and CPTPs as well as in non- catalytic adapter molecules, such as Grbs (Growth factor Receptor Bound) , which mediate protein-protein interactions between components of the signal transduction pathway (Skolnik et al . , 1991, Cell 65:83-90; Pawson, 1995, Nature 373:573-580) .
  • Multiprotein signaling complexes comprising receptor subunits, kinases, phosphatases and adapter molecules are assembled in subcellular compartments through the specific and dynamic interactions between these domains with their binding motifs.
  • Such signaling complexes integrate the extracellular signal from the ligand-bound receptor and relay the signal to other downstream signaling proteins or complexes in other locations inside the cell or in the nucleus (Koch et al . , 1991, Science 252:668-674; Pawson,
  • the levels of tyrosine phosphorylation required for normal cell growth and differentiation at any time are achieved through the coordinated action of PTKs and PTPs.
  • these two types of enzymes may either antagonize or cooperate with each other during signal transduction.
  • An imbalance between these enzymes may impair normal cell functions leading to metabolic disorders and cellular transformation.
  • insulin binding to the insulin receptor which is a PTK
  • triggers a variety of metabolic and growth promoting effects such as glucose transport, biosynthesis of glycogen and fats, DNA synthesis, cell division and differentiation.
  • Diabetes mellitus which is characterized by insufficient or a lack of insulin signal transduction can be caused by any abnormality at any step along the insulin signaling pathway.
  • PTKs such as HER2
  • HER2 can play a decisive role in the development of cancer (Slamon et al . , 1987, Science 235:77-82) and that antibodies capable of blocking the activity of this enzyme can abrogate tumor growth (Drebin et al . , 1988, Oncogene 2:387-394).
  • Blocking the signal transduction capability of tyrosine kinases such as Flk-1 and the PDGF receptor have been shown to block tumor growth in animal models (Millauer et al . , 1994, Nature 367:577; Ueno et al . , Science, 252:844-848).
  • PTPs may play a role in human diseases.
  • ectopic expression of RPTP ⁇ produces a transformed phenotype in embryonic fibroblasts (Zheng et al . , Nature 359:336-339), and overexpression of RPTP ⁇ in embryonal carcinoma cells causes the cells to differentiate into a cell type with neuronal phenotype (den Hertog et al . , EMBO J 12:3789-3798).
  • the gene for human RPTP ⁇ has been localized to chromosome 3p21 which is a segment frequently altered in renal and small lung carcinoma.
  • Mutations may occur in the extracellular segment of RPTP ⁇ which renders a RPTP that no longer respond to external signals (LaForgia et al . , Wary et al . , 1993, Cancer Res 52:478-482). Mutations in the gene encoding PTP1C (also known as HCP, SHP) are the cause of the motheaten phenotype in mice which suffer severe immunodeficiency, and systemic autoimmune disease accompanied by hyperproliferation of macrophages (Schultz et al . , 1993, Cell 73:1445-1454).
  • PTPID also known as Syp or PTP2C
  • PTP2C has been shown to bind through SH2 domains to sites of phosphorylation in PDGFR, EGFR and insulin receptor substrate 1 (IRS-1) . Reducing the activity of PTPID by microinjection of anti-PTPID antibody has been shown to block insulin or EGF-induced mitogenesis (Xiao et al . , 1994, J Biol Chem 269:21244-21248).
  • Vanadates and pervanadates are known to be non-specific phosphatase inhibitors.
  • this class of compounds is toxic because each compound contains a heavy metal (U.S. Patent No. 5,155,031; Fantus et al . , 1989, Biochem., 28:8864-71; Swarup et al . , 1982, Biochem. Biophys. Res. Commun. 107:1104-9).
  • the present invention is directed to compounds capable of modulating and/or regulating receptor-mediated signal transduction by inhibiting the activity of protein tyrosine phosphatases (PTPs) .
  • PTPs protein tyrosine phosphatases
  • the compounds of the invention can inhibit the dephosphorylation of phosphotyrosine residues of a substrate wherein the substrate relays signals in a signaling pathway.
  • the compounds may also functionally mimic the effect on a signal transduction of ligand binding to a signaling receptor.
  • the invention is also directed to methods of inhibiting protein tyrosine phosphatase activity by contacting cells with an effective amount of a compound of the present invention or a pharmaceutically acceptable salt thereof.
  • the invention encompasses methods of treating disease states in mammals, including humans, which are ameliorated by modulating and/or regulating signal transduction through the inhibition of protein tyrosine phosphatase activity.
  • disease states or disorders include but are not limited to diabetes, cancer, immune and neurological disorders.
  • the compounds of the present invention inhibit phosphatase activity in cells, so that dephosphorylation of various tyrosine kinases, or other phosphatase substrates involved in the signaling pathway is decreased. As a result, the appearance of phosphotyrosine residues on such substrates will increase.
  • a number of different types of compounds, which may or may not share certain structural features are encompassed within the invention. Although not limited to any theory or explanation, the compounds of the invention are thought to demonstrate such activity by mimicking the transition state of phosphates departing phosphotyrosines.
  • Two features believed to be involved in conferring such activities are (a) an aryl group that mimics the structure of tyrosine and/or tyrosine backbone, and (b) a highly diffuse negatively charged moiety that mimics the negatively charged phosphate binding of divalent metal ions, such as Mg ++ , Mn ++ , Zn ++ and the like.
  • the negatively charged moiety is located sufficiently close to the aryl group so that the negative charge could diffuse into the aryl moiety.
  • Functional groups which contribute to the negatively charged moiety include, but are not limited to hydroxy-ketone and acidic groups; examples of such groups are shown in Formula IV, R 2 and R 3 and in Formula VII, R 6 , infra.
  • An aryl moiety that mimics tyrosine is exemplified by the substituted five membered ring shown in Formula III infra.
  • the compounds of the present invention are heterocyclic nitrogen containing compounds of formula I:
  • the nitrogen containing heterocyclic nucleus identified by the term "Q" is preferably nitropyridine or nitrothiazole.
  • the present invention encompasses pharmaceutically acceptable salts or analogs of the above compounds.
  • Preferred compounds of the present invention include those of formula II:
  • A represents (i) a substituted or unsubstituted monocyclic five or six membered ring having 1-4 hetero ring atoms, at least one of which is nitrogen, the remainder of which are selected from nitrogen, oxygen or sulfur, e .g.
  • indole quinoxaline, quinazoline, quinoline, isoquinoline, purine; or (iii) a substituted or unsubstituted monocyclic or fused polycyclic saturated or unsaturated ring having three to 15 atoms, which are carbon, sulfur, nitrogen or oxygen.
  • the heterocyclic rings defined above may be saturated or unsaturated.
  • the unsaturated rings or heteroaromatic group may, if desired, bear one or more substituents which do not substantially adversely affect the activity of the compound of formula I.
  • substituents are alkyl, alkoxy, phenoxy, alkenyl, alkynyl, phenylalkyl, hydroxyalkyi, haloalkyl, aryl, arylalkyi, alkyloxy, alkylthio, alkenylthio, phenylalkylthio, hydroxyalkyl-thio, alkylthiocarbbamylthio, phenyl, cyclohexyl, pyridyl, piperidinyl, alkylamino, amino, nitro, mercapto, cyano, hydroxyl, a halogen atom, an oxygen atom (forming a ketone or N-oxide) or a sulphur atom (
  • alkyl as used herein is meant a straight or branched chain saturated hydrocarbon group having from 1 to 20 carbons such as methyl, ethyl, isopropyl, n-butyl, s- butyl, t-butyl, n-amyl, isoamyl, n-hexyl, n-octyl and n- decyl.
  • alkenyl and alkynyl are used to mean straight or branched chain hydrocarbon groups having from 2 to 10 carbons and unsaturated by a double or triple bond respectively, such as vinyl, allyl, propargyl, 1-methylvinyl, but-l-enyl, but-2-enyl, but-2-ynyl, 1 methylbut-2-enyl, pent- 1-enyl, pent-3-enyl, 3-methylbut-l-ynyl, 1,1-dimethylallyl, hex-2-enyl and 1-methyl-l-ethylallyl.
  • phenylalkyl means the aforementioned alkyl groups substituted by a phenyl group such as benzyl, phenethyl, phenopropyl, 1-benzylethyl, phenobutyl and 2-benzylpropyl.
  • aryl as used herein is meant to include a monocyclic, bicyclic, tricyclic or other polycyclic compounds, wherein at least one ring is aromatic including aromatic hydrocarbons or hetero-aromatic hydrocarbons having heteroaromatic atoms such as nitrogen, sulfur and oxygen.
  • hydroxy-alkyl means the aforementioned alkyl groups substituted by a single hydroxyl group such as 2-hydroxyethyl, 2-hydroxypropyl, 3- hydroxypropyl, 4-hydroxybutyl, 1-hydroxybutyl and 6- hydroxyhexyl.
  • alkylthio, alkenylthio, alkynylthio, alkylthio, hydroxy-alkylthio and phenyl- alkylthio as used herein mean the aforementioned alkyl, alkenyl, alkynyl, hydroxy-alkyl and phenyl-alkyl groups linked through a sulfur atom to group R.
  • substituted means that the group in question, e . g. , alkyl group, aryl group, etc., may bear one or more substituents including but not limited to halogen, hydroxy, cyano amino, nitro, mercapto, carboxy and other substituents known to those skilled in the art.
  • saturated as used herein means an organic compound with neither double or triple bonds.
  • unsaturated as used herein means an organic compound containing either double or triple bonds.
  • Figure 1 Dose response effect of compound 10 on the level of phosphotyrosine (pTyr) residues on insulin receptor over time.
  • Figure 2. Dose response effect of 2-methyl-5,6,8- trihydroxy-9-(5,6,8-trihydroxy-2-methyl-4H- naphtho[2,3,b]pyrano-4-one-9-yl)4H-naphtho[2,3,b]pyran-4-one on the uptake of 2-deoxy- 3 H-glucose in 3T3-L1 cells, a preadipocyte cell line.
  • the present invention is directed to compounds capable of inhibiting the activity of protein tyrosine phosphatases (PTPs) for modulating or triggering signal transduction.
  • PTPs protein tyrosine phosphatases
  • the compounds of the invention can inhibit the dephosphorylation of phosphotyrosine residues of a substrate wherein the substrate relays signals in a signaling pathway.
  • the compounds may also functionally mimic the effect on a signal transduction of ligand binding to a signaling receptor.
  • the invention is further directed to the use of such compounds in modulating or triggering signal transduction, and in the regulation of cellular processes that are controlled by signal transduction through the inhibition of the activity of PTPs by the compounds.
  • the invention further provides for the use of such compounds in the treatment of a subject having a disorder caused by dysfunctional signal transduction.
  • the compounds of the invention are capable of inhibiting the activity of protein tyrosine phosphatases, that are transmembrane or intracellular, and that may have one or more characteristic catalytic domains.
  • the amino acid sequences of the PTPs in the catalytic domains may include but are not limited to [I/V]HCXAGXXR[S/T]G (single-letter amino acid code; X is any amino acid) .
  • the PTPs may possess one or more modular conserved domains, which include but are not limited to, SH2, SH3 and PH domains.
  • the compounds of the invention can be used to inhibit the phosphatase activity of PTP1B (Charbonneau et al., 1989, Proc. Natl Acad Sci USA, 86: 5252-5256), T-cell PTP (Cool et al., 1989, Proc Natl Acad Sci USA, 86: 5257- 5 5261, PTP1C (Shen et al., 1991, Nature, 352: 736-739), PTPID (Vogel et al., 1993, Science 259: 1611-1614), RPTP ⁇ , RPTP ⁇ , RPTP ⁇ (Kaplan et al., 1990, Proc Natl Acad Sci USA, 87: 7000- 7004), RPTP ⁇ (Yan et al., 1993, J Biol Chem 268: 24880- 24886), RPTPK (Jiang et al., 1993, Mol Cell Biol, 13: 2942-
  • the PTPs preferred in the invention are of human origin. Inhibition of phosphatase activity that is substantially specific to a PTP or a set of PTPs in a signaling pathway is preferred.
  • signal transduction is not limited to transmembrane signaling, and includes the multiple pathways that branch off throughout the cell and into the nucleus. Such signaling pathways may include but are not limited to the Ras pathway (Schlessinger, 1994, Curr Opin
  • the term "modulation” or “modulating” shall mean upregulation or downregulation of a signaling pathway.
  • 25 under the control of signal transduction may include, but are not limited to, transcription of specific genes; normal cellular functions, such as metabolism, proliferation, differentiation, adhesion, apoptosis and survival; as well as abnormal processes, such as transformation, blocking of
  • a signal may be triggered by the binding of a ligand to its receptor on the cell surface, and the signal is relayed and propagated by the phosphorylation or dephosphorylation of specific tyrosine residues on various substrates inside the
  • a substrate may contain one or more tyrosine residues that are specifically phosphorylated or dephosphorylated by PTKs or PTPs in the signaling pathway.
  • substrates may include the receptor and its subunits, molecules associated with or recruited to the receptor such as cytoplasmic kinases, cytoplasmic phosphatases, adapter molecules, cytoskeletal proteins and transcription factors.
  • receptor as used herein may include, but is not limited to, insulin receptor, members of the insulin-like growth factor receptor family, epidermal growth factor receptor family, fibroblast growth factor receptor family, hepatocyte growth factor receptor family, vascular endothelial growth factor receptor family, neurotrophin receptor (trk) family, the T-cell receptor, the B cell receptor and members of the Type I-IV cytokine receptor families (Heldin, 1995, Cell. 80: 213-223; Taniguchi, 1995, Science, 268: 251-255).
  • Adapter molecules that are substrates may include the Grb proteins, IRS-l, Zap-70 and She (Pawson et al., 1995, Nature 373: 573-580). Cytoskeletal proteins such as actin and transcription factors such as the STAT proteins (Ihle et al . , Trends Biochem Sci, 19:222-227) may also serve as substrates.
  • ligand is synonymous with extracellular signaling molecules, and includes but is not limited to growth factors such as insulin, EGF, PDGF, fibroblast growth factors, vascular endothelial growth factor, and neurotrophins; and cytokines such as growth hormone, erythropoietin, tumor necrosis factor, interleukins and interferons.
  • growth factors such as insulin, EGF, PDGF, fibroblast growth factors, vascular endothelial growth factor, and neurotrophins
  • cytokines such as growth hormone, erythropoietin, tumor necrosis factor, interleukins and interferons.
  • the term ligand is not limited to soluble molecules, and includes; for example, extracellular matrix proteins, cell adhesion molecules as well as antigenic peptides associated with the major histocompatibility complex proteins on the surface of an antigen-presenting cell.
  • the compounds of the invention can be used to trigger or upregulate signal transduction in cells so that the effect of ligand binding to a receptor is enhanced, or mimicked if the ligand is not present.
  • the compounds exert the effect by inhibiting or diminishing the activity of a phosphatase in the signaling pathway which normally acts negatively toward signaling.
  • PTPs normally downregulate signal transduction involves the dephosphorylation of specific phosphotyrosine residues (pTyr) on PTKs and their substrates since many PTKs require phosphorylation of some of its own tyrosine residues in order to become optimally active in the signaling pathway.
  • the compounds of the invention can be used to prevent the dephosphorylation of pTyr residues on receptors or their subunits which normally becomes phosphorylated upon ligand binding, thereby enhancing the extent and duration of PTK phosphorylation.
  • the compounds of the invention can also be used to prevent the dephosphorylation of PTKs in which the tyrosine residues become autophosphorylated or transphosphorylated due to its basal activity.
  • a signal may be triggered by the compounds of the invention in the absence of ligand binding since the basal activity of PTKs is sufficient to promote a signal if constitutive PTP activity is inhibited or diminished by the compounds.
  • the compounds of the invention may be used to enhance or sustain insulin receptor signal transduction by inhibiting the constitutive dephosphorylation of the pTyr sites on the activated insulin receptor. This would allow the insulin receptor to remain phosphorylated, thus enhancing or sustaining the insulin signal. Furthermore, since it has been shown that insulin receptor is phosphorylated at a low level even in the absence of insulin (Goldstein, 1992, J.
  • the compounds of the invention can be used to trigger a signal, even in the absence of insulin, by allowing the tyrosine residues on the receptor to become self-phosphorylated.
  • Another mechanism by which PTPs may exert a negative effect on signaling is through the dephosphorylation of specific pTyr sites to which SH2-containing molecules bind during signaling. The absence of such pTyr sites would prevent the recruitment of SH2-containing molecules to specific subcellular compartments to form multiprotein signaling complexes, thereby, preventing the further propagation of the signal.
  • the compounds of the invention can be used to upregulate or prolong signal transduction by preventing the dephosphorylation of pTyr sites on substrate proteins that normally serve as binding sites for SH2-containing proteins which promote signaling.
  • the compounds of the invention may be used to prevent the dephosphorylation of specific pTyr residues on any substrate, which pTyr residues are essential to the relay or propagation of the signal.
  • the compounds of the invention may be used to prevent the dephosphorylation of specific pTyr residues on any substrate, which pTyr residues are inhibitory to signal transduction.
  • the compounds of the invention can also be used to suppress or downregulate signal transduction in cells so that the effect of ligand binding to a receptor is abolished or attenuated.
  • the compounds can inhibit a phosphatase in a signaling pathway which normally acts positively toward signaling.
  • PTPs promote signaling through the activation of members of the Src family of PTKs.
  • Src family PTKs have an inhibitory site of phosphorylation in their carboxy termini which by dephosphorylation activates the kinase activity.
  • the compounds of the invention can be used to prevent the dephosphorylation of the inhibitory pTyr in the carboxy termini of kinases which function normally to promote signal transductions.
  • Src family PTKs may include Src, Fyn, Lck, Lyn, Blk, Hck, Fgr and Yrk.
  • Other kinases which may be similarly regulated by a phosphatase may include Fak and Csk (Taniguchi, 1995, Science 268: 251-255).
  • Various procedures known in the art may be used for identifying, evaluating or assaying the inhibition of activity of protein tyrosine phosphatases by the compounds of the invention.
  • assays involve exposing target cells in culture to the compounds, and a) biochemically analyzing cell lysates to assess the level and/or identity of tyrosine phosphorylated proteins; or (b) scoring phenotypic or functional changes in treated cells as compared to control cells that were not exposed to the test substance.
  • the cells are exposed to the compound of the invention and compared to positive controls which are exposed only to the natural ligand, and to negative controls which were not exposed to either the compound or the natural ligand.
  • the assay may be carried out in the absence of the ligand.
  • the cells are exposed to the compound of the invention in the presence of the natural ligand and compared to controls which are not exposed to the compound of the invention.
  • the assays described hereinbelow may be used as a primary screen to evaluate the phosphatase inhibition activity of the compounds of the invention.
  • the assays may also be used to assess the relative potency of a compound by testing a range of concentrations, in a range from lOO ⁇ M to 1 pM, for example, and computing the concentration at which the amount of phosphorylation or signal transduction is reduced or increased by 50% (IC50) compared to controls.
  • Target cells having a substrate molecule that is phosphorylated or dephosphorylated on a tyrosine residue during signal transduction are exposed to the compounds of the invention and radiolabelled phosphate, and thereafter, lysed to release cellular contents, including the substrate of interest.
  • the substrate may be analyzed by separating the protein components of the cell lysate using a sodium dodecyl sulphate-polyacrylamide gel electrophoresis (SDS-PAGE) technique, in either one or two dimensions, and detecting the presence of phosphorylated proteins by exposing to X-ray film.
  • SDS-PAGE sodium dodecyl sulphate-polyacrylamide gel electrophoresis
  • the protein components separated by SDS-PAGE are transferred to a nitrocellulose membrane, the presence of pTyr is detected using an antiphosphotyrosine (anti-pTyr) antibody.
  • anti-pTyr antiphosphotyrosine
  • the substrate of interest be first isolated by incubating the cell lysate with a substrate-specific anchoring antibody bound to a solid support, and thereafter, washing away non-bound cellular components, and assessing the presence or absence of pTyr on the solid support by an anti-pTyr antibody.
  • This preferred method can readily be performed in a microtitre plate format by an automated robotic system, allowing for testing of large numbers of samples within a reasonably short time frame. Compounds of the present invention were identified and evaluated by this preferred method as described in sections infra.
  • the anti-pTyr antibody can be detected by labelling it with a radioactive substance which facilitates its detection by autoradiography.
  • the anti-pTyr antibody can be conjugated with an enzyme, such as horseradish peroxidase, and detected by subsequent addition of a calorimetric substrate for the enzyme.
  • a further alternative involves detecting the anti-pTyr antibody by reacting with a second antibody which recognizes the anti-pTyr antibody, this second antibody being labelled with either a radioactive substance or an enzyme as previously described. Any other methods for the detection of an antibody known in the art may be used.
  • the above methods may also be used in a cell-free system wherein cell lysate containing the signal-transducing substrate molecule and phosphatase is mixed with a compound of the invention and a kinase.
  • the substrate is phosphorylated by initiating the kinase reaction by the addition of adenosine triphosphate (ATP) .
  • ATP adenosine triphosphate
  • the reaction mixture may be analyzed by the SDS-PAGE technique or it may be added to substrate-specific anchoring antibody bound to a solid support, and a detection procedure as described above is performed on the separated or captured substrate to assess the presence or absence of pTyr. The results are compared to those obtained with reaction mixtures to which the compound is not added.
  • the cell-free system does not require the natural ligand or knowledge of its identity.
  • Posner et al . U.S. Patent No. 5,155,031
  • Posner et al . describes the use of insulin receptor as a substrate and rat adipocytes as target cells to demonstrate the ability of pervanadate to inhibit PTP activity.
  • Burke et al . (1994, Biochem Biophys Res Comm 204:129-134) describes the use of autophosphorylated insulin receptor and recombinant PTP1B in assessing the inhibitory activity of a phosphotyrosyl mimetic.
  • the ability of the compounds of the invention to modulate the activity of PTPs, which controls signal transduction, may also be measured by scoring for morphological or functional changes associated with ligand binding. Any qualitative or quantitative techniques known in the art may be applied for observing and measuring cellular processes which comes under the control of phosphatases in a signaling pathway. Such cellular processes may include, but are not limited to, anabolic and catabolic processes, cell proliferation, cell differentiation, cell adhesion, cell migration and cell death.
  • vanadate has been shown to activate an insulin-sensitive facilitated transport system for glucose and glucose analogs in rat adipocytes (Dubyak et al . , 1980, J Biol Chem 256:5306-5312).
  • the activity of the compounds of the invention may be assessed by measuring the increase in the rate of transport of glucose analog, such as 2-deoxy- 3 H- glucose, in rat adipocytes that have been exposed to the compounds.
  • Vanadate also mimic the effect of insulin on glucose oxidation in rat adipocytes (Shechter et al .
  • the compounds of the invention may be tested for stimulation of glucose oxidation by measuring the conversion of 14 C-glucose to 14 C0 2 .
  • the effect of sodium orthovanadate on erythropoietin-mediated cell proliferation has been measured by cell cycle analysis based on DNA content as estimated by incorporation of tritiated thymidine during DNA synthesis (Spivak et al . , 1992, Exp Hematol, 20:500-504).
  • the activity of the compounds of the invention toward phosphatases that play a role in cell proliferation may be assessed by cell cycle analysis.
  • the activity of the compounds of the invention can also be assessed in animals using experimental models of disorders caused by or related to dysfunctional signal transduction.
  • the activity of the compounds may be tested for its effect on insulin receptor signal transduction in non- obese diabetic mice (Lund et al . , 1990, Nature 345:727-729), BB Wistar rats and streptozotocin-induced diabetic rats (Solomon et al . , 1989, Am J Med Sci 297:372-376).
  • the activity of the compounds may also be assessed in animal carcinogenesis experiments since phosphatases can play an important role in dysfunctional signal transduction leading to cellular transformation.
  • okadaic acid a phosphatase inhibitor, has been shown to promote tumor formation on mouse skin (Suganuma et al . , 1988, Proc Natl Acad Sci 85:1768-1771).
  • the data obtained from these cell culture assays and animal studies can be used in formulating a range of dosages for use in humans.
  • the dosage of the compounds of the invention should lie within a range of circulating concentrations with little or no toxicity.
  • the dosage may vary within this range depending on the dosage form employed and the route of administration.
  • the present invention encompasses compounds capable of regulating and/or modulating signal transduction by inhibiting the activity of protein tyrosine phosphatases. More specifically, the present invention encompasses compounds capable of inhibiting protein tyrosine phosphatase activity. These compounds will be referred to herein generically as "phosphatase inhibitors", even though these compounds either upregulate or downregulate cellular processes that are controlled by signal transduction. Generally, the compounds of the present invention may be heterocyclic nitrogen containing compounds, for example; nitrothiazole compounds or derivatives thereof, as described by the following general formula I:
  • the nitrogen containing heterocyclic nucleus identified by the term "Q" is preferably nitropyridine or nitrothiazole.
  • the present invention encompasses pharmaceutically acceptable salts or analogs of the above compounds.
  • Preferred compounds of the present invention include those of formula II:
  • A represents (i) a substituted or unsubstituted monocyclic five or six membered ring having 1-4 hetero ring atoms, at least one of which is nitrogen, the remainder of which are selected from nitrogen, oxygen or sulfur, e .g.
  • R is hydrogen, halogen, cyano, nitro, amino, amido, carboxy, acylamino, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyi, substituted cycloalkyi, aryl, substituted aryl, arylalkyi, e .g. , benzyl; aryloxy, e .g. , phenoxy; a five or six membered heterocyclic ring containing 0 to 3 hetero atoms which are either sulfur, nitrogen or oxygen, which heterocyclic ring may be substituted or unsubstituted;
  • R 2 is hydrogen, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyi, substituted cycloalkyi, aryl, substituted aryl; arylalkyi, substituted arylalkyi; and n is
  • R 2 is hydrogen, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyi, substituted cycloalkyi, aryl, substituted aryl; arylalkyi; substituted arylalkyi;
  • R 3 is hydrogen, halogen, cyano, nitro, hydroxy, alkyl, substituted alkyl, alkoxy, substituted alkoxy, cycloalkyi, substituted cycloalkyi, aryl, substituted aryl, arylalkyi; substituted arylalkyi, carboxy, amido, amino, acylamino, sulfonyl, sulfonamido, aminosulfone, a five or six membered heterocyclic ring containing 1 to 2 hetero atoms which are either sulfur, nitrogen or oxygen, which heterocyclic ring may be substituted or unsubstituted.
  • Exemplary structures falling within group (iii) above are cyclopentyl, cyclohexyl, adamantyl, tetrahydroquinoline, tetrahydropyrazole, as well as substituted derivatives thereof.
  • the nitrothiazole compounds are election- accepting compounds, some of which have been reported as light sensitive agents for photographic materials.
  • Compounds within the scope of the present invention are described in U.S. Patent Nos. 5,198,333, 3,870,725 and 3,850,939, and applicants' commonly assigned and copending U.S. patent applications "Pharmaceutical Compositions and Methods for Modulating Signal Transduction” (Pennie & Edmonds Docket No. 007683-0123-999, filed June 7, 1996) and “Methods of Inhibiting Phosphatase Activity and Treatment of Disorders Associated Therewith” (Serial No. 08/481,954, Pennie & Edmonds Docket No. 007683-0073-999, filed June 7, 1995), which are incorporated by reference herein in their entirety.
  • Compounds within the scope of the present inventions also include: CH,.
  • A is as defined above , and R' is hydrogen, C 1 -C 4 alkyl and substituted C x -C 4 alkyl .
  • R' is hydrogen, C 1 -C 4 alkyl and substituted C x -C 4 alkyl .
  • Such compounds also possess potent activity in inhibiting or promoting phosphatase activity.
  • the invention encompasses the above described compounds (see formulas I, II, III and IV) wherein the thio linkage is replaced by an amino linkage.
  • the compounds of the present invention may also include naphthopyrones and derivatives thereof which is described by the formula (VI) :
  • A is a single or double bond
  • Rl, R2, which are the same or different, are Cl to C4 alkyl group or an aryl group;
  • R4, R5, R6, which are the same or different, are H, OH, Cl to C4 alkoxy, aryl, Cl to C6 alkyl, alkylaryl, arylalkoxy, ester, carboxylic acid, carboxamide, carbamate, or cyano;
  • R3 is a substituent identical to that of formula VI providing a compound of formula VII:
  • R 3 is an aryl group, e .g. , mono, bi or tricyclic aryl group; which links to formula VI directly or by amide, carbon or ketone groups.
  • COMPOUND 30 (CD 04728)
  • the compounds of the present invention can be synthesized in accordance with standard organic chemistry techniques using readily available starting materials.
  • the present invention is further directed to pharmaceutical compositions comprising a pharmaceutically effective amount of the above-described compounds and a pharmaceutically acceptable carrier or excipient.
  • a composition is believed to inhibit the activity of protein tyrosine phosphatases which may be useful in treatment of diseases related to dysfunctional signal transduction, including diabetes and cancer.
  • such composition may act directly on the cells responsible for the disease (e.g., tumor cells).
  • compositions of the present invention may be included in methods for treating, among other diseases, diabetic retinopathy, glioma, melanoma, Kaposi's sarcoma, hemangioma and ovarian, breast, lung, pancreatic, prostate, colon and epidermoid cancer.
  • the present invention is also directed to methods for treating diseases, including but not limited to metabolic disorders, diabetes, diabetic retinopathy, rheumatoid arthritis, anemia, immunodeficiency, inflammation, neurodegenerative diseases, and cancer and more particularly cancer related to solid cell tumor growth (e .g. , glioblastoma, melanoma and Kaposi's sarcoma and ovarian, lung, mammary, prostate, pancreatic, colon and epidermoid carcinoma) .
  • diseases including but not limited to metabolic disorders, diabetes, diabetic retinopathy, rheumatoid arthritis, anemia, immunodeficiency, inflammation, neurodegenerative diseases, and cancer and more particularly cancer related to solid cell tumor growth (e .g. , glioblastoma, melanoma and Kaposi's sarcoma and ovarian, lung, mammary, prostate, pancreatic, colon and epidermoid carcinoma) .
  • salts refers to those salts which retain the biological effectiveness and properties of the compound and which are obtained by reaction with inorganic acids or bases such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid and the like.
  • the invention is further directed, where applicable, to solvated as well as unsolvated forms of the compounds (e . g. , hydrated forms) having the ability to regulate and/or modulate phosphatase activity.
  • the compounds described above may be prepared by any process known to be applicable to the preparation of chemically-related compounds. Suitable processes are illustrated by the representative examples provided. Infra . Necessary starting materials may be obtained by standard procedures of organic chemistry.
  • the identified compounds can be administered to a human patient, by itself, or in pharmaceutical compositions where it is mixed with suitable carriers or excipient(s) at doses to treat or ameliorate a variety of disorders, including metabolic disorders, anemia, immunodeficiency, inflammation, neurodegenerative disorders, solid cell tumor growth, including Kaposi's sarcoma, glioblastoma, and melanoma and ovarian, lung, mammary, prostate, pancreatic, colon and epidermoid carcinoma, diabetes, diabetic retinopathy, hemangioma and rheumatoid arthritis.
  • a therapeutically effective dose further refers to that amount of the compound sufficient to result in amelioration of symptoms of uncontrolled vasculogenesis and angiogenesis.
  • the formulations of the present invention normally will consist of at least one compound of the invention mixed with a carrier, or diluted by a carrier, or enclosed or encapsulated by an ingestible carrier in the form of a capsule, sachet, cachet, paper or other container or by a disposable container such as an ampoule.
  • a carrier or diluent may be a solid, semi-solid or liquid material, which serves as a vehicle, excipient or medium for the active therapeutic substance.
  • diluents or carriers which may be employed in the pharmaceutical compositions of the present invention are lactose, dextrose, sucrose, sorbitol, mannitol, propylene glycol, liquid paraffin, white soft paraffin, kaolin, microcrystalline cellulose, calcium silicate, silica polyvinylpyrrolidone, cetostearyl alcohol, starch, gum acacia, calcium phosphate, cocoa butter, oil of theobroma, arachis oil, alginates, tragacanth, gelatin, syrup B.P., methyl cellulose, polyoxyethylene sorbitan monolaurate, ethyl lactate and propylhydroxybenzoate, sorbitan trioleate, sorbitan sesquioleate and oleyl alcohol.
  • Suitable routes of administration may, for example, include oral, rectal, transmucosal, or intestinal administration; parenteral delivery, including intramuscular, subcutaneous, intramedullary injections, as well as intrathecal, direct intraventricular, intravenous, intraperitoneal, intranasal, or intraocular injections; transdermal, topical, vaginal and the like.
  • Dosage forms include but are not limited to tablets, troches, dispersions, suspensions, suppositories, solutions, capsules, creams, patches, minipumps and the like.
  • compositions of the present invention may be manufactured in a manner that is itself known, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or lyophilizing processes.
  • compositions for use in accordance with the present invention thus may be formulated in conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries which facilitate processing of the active compounds into preparations which can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen.
  • the agents of the invention may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hanks's solution. Ringer's solution, or physiological saline buffer.
  • physiologically compatible buffers such as Hanks's solution. Ringer's solution, or physiological saline buffer.
  • penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.
  • the compounds can be formulated readily by combining the active compounds with pharmaceutically acceptable carriers well known in the art.
  • Such carriers enable the compounds of the invention to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a patient to be treated.
  • Pharmaceutical preparations for oral use can be obtained solid excipient, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores.
  • Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropyl ethyl-cellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone (PVP) .
  • disintegrating agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
  • Dragee cores are provided with suitable coatings.
  • suitable coatings For this purpose, concentrated sugar solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures.
  • Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.
  • compositions which can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol.
  • the push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols.
  • stabilizers may be added. All formulations for oral administration should be in dosages suitable for such administration.
  • the compositions may take the form of tablets or lozenges formulated in conventional manner.
  • the compounds for use according to the present invention are conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebulizer, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or
  • the compounds may be formulated for parenteral administration by injection, e . g. , by bolus injection or continuous infusion.
  • Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi- dose containers, with an added preservative.
  • the compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • compositions for parenteral administration include aqueous solutions of the active compounds in water-soluble form. Additionally, suspensions of the active compounds may be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
  • the active ingredient may be in powder form for constitution with a suitable vehicle, e .g. , sterile pyrogen-free water, before use.
  • a suitable vehicle e .g. , sterile pyrogen-free water
  • the compounds may also be formulated in rectal compositions such as suppositories or retention enemas, e .g. , containing conventional suppository bases such as cocoa butter or other glycerides.
  • the compounds may also be formulated as a depot preparation. Such long acting formulations may be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection.
  • the compounds may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
  • a pharmaceutical carrier for the hydrophobic compounds is a cosolvent system comprising benzyl alcohol, a nonpolar surfactant, a water-miscible organic polymer, and an aqueous phase.
  • the cosolvent system may be the VPD co-solvent system.
  • VPD is a solution of 3% w/v benzyl alcohol, 8% w/v of the nonpolar surfactant polysorbate 80, and 65% w/v polyethylene glycol 300, made up to volume in absolute ethanol.
  • the VPD co-solvent system (VPD:5W) consists of VPD diluted 1:1 with a 5% dextrose in water solution. This co- solvent system dissolves hydrophobic compounds well, and itself produces low toxicity upon systemic administration.
  • co-solvent system may be varied considerably without destroying its solubility and toxicity characteristics.
  • identity of the co-solvent components may be varied: for example, other low- toxicity nonpolar surfactants may be used instead of polysorbate 80; the fraction size of polyethylene glycol may be varied; other biocompatible polymers may replace polyethylene glycol, e .g. , polyvinyl pyrrolidone; and other sugars or polysaccharides may substitute for dextrose.
  • hydrophobic pharmaceutical compounds may be employed.
  • Liposomes and emulsions are well known examples of delivery vehicles or carriers for hydrophobic drugs.
  • Certain organic solvents such as dimethylsulfoxide also may be employed, although usually at the cost of greater toxicity.
  • the compounds may be delivered using a sustained-release system, such as semipermeable matrices of solid hydrophobic polymers containing the therapeutic agent.
  • sustained-release materials have been established and are well known by those skilled in the art. Sustained-release capsules may, depending on their chemical nature, release the compounds for a few weeks up to over 100 days. Depending on the chemical nature and the biological stability of the therapeutic reagent, additional strategies for protein stabilization may be employed.
  • compositions also may comprise suitable solid or gel phase carriers or excipients.
  • suitable solid or gel phase carriers or excipients include but are not limited to calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin, and polymers such as polyethylene glycols.
  • the compounds of the present invention may also be administered by controlled release means and/or delivery devices including Alzet ® osmotic pumps which are available from Alza Corporation. Suitable delivery devices are described in U.S. Patent Nos. 3,845,770; 3,916,899; 3,536,809; 3,598,123; 3,944,064 and 4,008,719, the disclosures of which are incorporated in their entirety by reference herein.
  • Many of the phosphatase modulating compounds of the invention may be provided as salts with pharmaceutically compatible counterions.
  • Pharmaceutically compatible salts may be formed with many acids, including but not limited to hydrochloric, sulfuric, acetic, lactic, tartaric, malic, succinic, etc. Salts tend to be more soluble in aqueous or other protonic solvents that are the corresponding free base forms.
  • compositions suitable for use in the present invention include compositions wherein the active ingredients are contained in an effective amount to achieve its intended purpose. More specifically, a therapeutically effective amount means an amount effective to prevent development of or to alleviate the existing symptoms of the subject being treated. Determination of the effective amounts is well within the capability of those skilled in the art, especially in light of the detailed disclosure provided herein.
  • the therapeutically effective dose can be estimated initially from cell culture assays.
  • a dose can be formulated in animal models to achieve a circulating concentration range that includes the IC50 as determined in cell culture (i.e., the concentration of the test compound which achieves a half-maximal inhibition of the PTP activity) .
  • Such information can be used to more accurately determine useful doses in humans.
  • a therapeutically effective dose refers to that amount of the compound that results in amelioration of symptoms or a prolongation of survival in a patient.
  • Toxicity and therapeutic efficacy of such compounds can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population) .
  • the dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio between LD50 and ED50.
  • Compounds which exhibit high therapeutic indices are preferred. The data obtained from these cell culture assays and animal studies can be used in formulating a range of dosage for use in human.
  • the dosage of such compounds lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity.
  • the dosage may vary within this range depending upon the dosage form employed and the route of administration utilized.
  • the exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition. (See e . g. , Fingl et al . , 1975, in "The Pharmacological Basis of Therapeutics", Ch. 1 pl) .
  • Dosage amount and interval may be adjusted individually to provide plasma levels of the active moiety which are sufficient to maintain the phosphatase modulating effects, or minimal effective concentration (MEC) .
  • MEC minimal effective concentration
  • the MEC will vary for each compound but can be estimated from in vitro data; e.g., the concentration necessary to achieve a 50-90% inhibition of the phosphatase using the assays described herein. Dosages necessary to achieve the MEC will depend on individual characteristics and route of administration. However, HPLC assays or bioassays can be used to determine plasma concentrations.
  • Dosage intervals can also be determined using the MEC value.
  • Compounds should be administered using a regimen which maintains plasma levels above the MEC for 10-90% of the time, preferably between 30-90% and most preferably between 50-90%.
  • Usual patient dosages for systemic administration range from 1 to 2000 mg/day, commonly from 1 to 250 mg/day, and typically from 10 to 150 mg/day. Stated in terms of patient body weight, usual dosages range from 0.02 to 25 mg/kg/day, commonly from 0.02 to 3 mg/kg/day, typically from 0.2 to 1.5 mg/kg/day. Stated in terms of patient body surface areas, usual dosages range from 0.5 to 1200 mg/m 2 /day, commonly from 0.5 to 150 mg/m 2 /day, typically from 5 to 100 mg/m 2 /day. Usual average plasma levels should be maintained within 50 to 5000 ⁇ g/ml, commonly 50 to 1000 ⁇ g/ml, and typically 100 to 500 ⁇ g/ml. In cases of local administration or selective uptake, the effective local concentration of the drug may not be related to plasma concentration.
  • composition administered will, of course, be dependent on the subject being treated, on the subject's weight, the severity of the affliction, the manner of administration and the judgment of the prescribing physician. Desirable blood levels may be maintained by a continuous infusion of the compound as ascertained by plasma levels measured by HPLC. It should be noted that the attending physician would know how to and when to terminate, interrupt or adjust therapy to lower dosage due to toxicity, or bone marrow, liver or kidney dysfunctions. Conversely, the attending physician would also know to adjust treatment to higher levels if the clinical response is not adequate (precluding toxicity) .
  • a prophylactic or therapeutic dose of the compound in the acute or chronic management of disease will vary with the severity of the condition to be treated and the route of administration. Again, it should be noted that the clinician or physician would know when to interrupt and/or adjust the treatment dose due to toxicity or bone marrow, liver or kidney dysfunctions.
  • the dose, and perhaps the dosage frequency will also vary according to the age, body weight, and response of the individual patient. In general, as discussed above, the total daily dose ranges for the compounds for the majority of the disorders described herein, is from about 0.02 to about 25 mg/kg patient.
  • a daily dose range should be between about 0.02 to about 3 mg/kg, while most preferably a daily dose range should be between about 0.2 to about 1.5 mg/kg per day. It is further recommended that infants, children, and patients over 65 years, and those with impaired renal, or hepatic function, initially receive low doses, and that they be titrated based on individual clinical response(s) and blood level(s). It may be necessary to use dosages outside these ranges in some cases as will be apparent to those of ordinary skill in the art.
  • compositions may, if desired, be presented in a pack or dispenser device which may contain one or more unit dosage forms containing the active ingredient.
  • the pack may for example comprise metal or plastic foil, such as a blister pack.
  • the pack or dispenser device may be accompanied by instructions for administration.
  • Compositions comprising a compound of the invention formulated in a compatible pharmaceutical carrier may also be prepared, placed in an appropriate container, and labelled for treatment of an indicated condition. Suitable conditions indicated on the label may include treatment of a tumor, such as a glioma or glioblastoma and inhibition of angiogenesis.
  • Any compound of the invention which inhibits or diminishes the PTP activity in a signaling pathway may be used in the therapeutic methods of the invention.
  • the activity of the compound is sufficiently specific for the PTPs in the pathway so that the compound does not interfere with the function of other phosphatases in the cell.
  • the compounds of the invention may be identified and evaluated by, for example; methods described in section 7 infra.
  • the compounds and pharmaceutical compositions of the invention can be used for treating diabetes mellitus.
  • the pathogenesis of diabetes generally involves insufficient. or a total lack of insulin signal transduction.
  • the paucity or absence of the insulin signal may be caused by a variety of reasons such as a lack of insulin, loss of binding affinity, defective receptor or underexpression of receptor.
  • Insulin receptor activity can be modulated by inhibiting the phosphatases in the signaling using the compounds of the invention.
  • the insulin signal may be restored or stimulated in cells through the inhibition of dephosphorylating activity, even in the absence of insulin.
  • the example of diabetes mellitus illustrates the principles of therapeutic applications of the invention which may be applied to other disorders that implicate signal transduction by phosphotyrosine phosphatases.
  • the compounds and pharmaceutical compositions of the invention may be used to treat immune disorders in which cytokine signal transduction is deficient.
  • Cytokines plays a crucial role in he opoiesis as well as coordinating immune and inflammatory responses.
  • the compounds may be used to replace or enhance the activity of a cytokine in signaling the differentiation and proliferation of hemopoietic cells, as well as B and T cells in response to antigenic stimulation, and thus be useful for treating anemia and immunodeficiency.
  • the compounds may also be used as an anti ⁇ inflammatory agent to treat disorders such as rheumatoid arthritis.
  • the compounds may also be therapeutically useful in treating neurodegenerative diseases by stimulating the growth and differentiation of neuronal cells which is regulated by neurotrophin-mediated signal transduction.
  • the compounds and pharmaceutical compositions of the invention may be used to treat cancer, such as glioma, melanoma, Kaposi's sarcoma, hemangioma and ovarian, breast, lung, pancreatic, liver, prostate, colon and epidermoid cancer, in which the malignant cells proliferate and/or metastasize as a result of uncontrolled signal transduction mediated by growth factors.
  • cancer such as glioma, melanoma, Kaposi's sarcoma, hemangioma and ovarian, breast, lung, pancreatic, liver, prostate, colon and epidermoid cancer
  • overexpression of a PTK such as HER2 has been shown to correlate with the aberrant growth characteristics of tumor cells.
  • Vasculogenesis and/or angiogenesis that facilitates tumor growth may also be inhibited by the compounds.
  • the compounds may modulate signal transduction in these tumor cells so that normal growth characteristics are restored.
  • the compounds may also be useful in treating psoriasis which is caused by excessive epidermal growth factor mediated signal transduction.
  • the ability of the compounds of the invention to inhibit dephosphorylation of phosphotyrosine (pTyr) residues on insulin receptor (IR) is described.
  • the assay may be used with any compounds of the invention.
  • substrate molecules such as platelet derived growth factor receptor
  • the activities of the compounds of the invention toward different protein tyrosine phosphatases may be assessed.
  • IR an endogenous kinase activity is active at low level even in the absence of insulin binding. Thus, no insulin is needed to stimulate phosphorylation of IR.
  • cell lysates were prepared and added to microtitre plates coated with anti-insulin receptor antibody. The level of phosphorylation of the captured insulin receptor was detected using an anti-pTyr antibody and an enzyme-linked secondary antibody.
  • the cell line used for the IR assay was NIH3T3 (ATCC# CRL 1658) engineered to over-express the human IR (H25 cells) .
  • Growth media for these cells is DMEM (Gibco) containing 10% fetal bovine serum, 1% L-glutamine, and 20mM Hepes.
  • the anchoring antibody used was BBE which recognizes the extracellular domain of human IR, and was purified by the Enzymology Laboratories, Sugen Inc.
  • PBS Gibco: KH2P04 0.20 g/1, K2HP04 2.16 g/1, KCl 0.20 g/1, NaCI 8.00 g/1, pH7.2.
  • Anti-pTyr Rabbit polyclonal antiphosphotyrosine antibody
  • Goat anti-rabbit IgG POD conjugate (Tago, Burlingame, CA, Cat.No. 6430) was used as the secondary antibody.
  • TBST buffer 50 mM Tris-HCl, 150 mM NaCI, 0.1% Triton X-100, adjusted to pH7.2 with ION HCl.
  • Blocking buffer PBS plus 5% milk (Carnation instant non-fat dry milk) .
  • 5X HNTG buffer 100 mM HEPES, 750 mM NaCI, 50% glycerol, 0.5% Triton X-100, pH 7.5.
  • ABTS solution 100 mM citric acid, 250 mM Na2HP04 , 0.5 mg/ml ABTS (2 ,2'-azinobis(3-ethylbenzthiazlinesulfonic acid), adjusted to pH 4.0 with IN HCl.
  • Cell lysis buffer HNTG containing ImM Na3V04 (0.5M solution kept as a 100X stock at -80°C in aliquots), 5mM NaP207 and 5mM EDTA prepared fresh nad keep on ice until ready for use.
  • Corning 25805-96 were coated with the anchoring antibody at 0.5 ⁇ g per well, in lOO ⁇ l PBS for at least two hours at room temperature or overnight at 4°C. Before use, the coating buffer was replaced with 100 ⁇ l blocking buffer, and the precoated assay plate was shaken at room temperature for 30 minutes. The wells were washed 3 times with water and once with TBST buffer before adding lysate.
  • the assay was set up in the 96-well tissue culture plate. Before adding the compounds to the cells, media in the wells was replaced by serum free DMEM medium, 90 ⁇ l per well. Positive control wells receive 80 ⁇ l DMEM. Negative controls received 90 ⁇ l DMEM.
  • the compounds of the invention were diluted 1:10 with DMEM and lO ⁇ l/well of the diluted test substances were transferred to the cells in the wells to achieve a final dilution of 1:100.
  • Positive and negative control wells received lO ⁇ l/well of dimethyl sulphoxide (DMSO) to achieve a final concentration of 1%.
  • Positive control wells additionally received lO ⁇ l/well of 0.1M Na 3 V0 4 so that the final concentration is lOmM.
  • the tissue culture plate was shaken for 1 minute before incubation at 37°C, 5% C0 2 . After 90 minutes of incubation, the media was removed by inversion of the plate, and lOO ⁇ l/well of lysis buffer was added to the cells. The tissue culture plate was shaken for 5 minutes and then placed on ice for 10 minutes. The cells were homogenized by repeated aspirating and dispensing, and the lysate was transferred to the corresponding wells of a precoated assay plate.
  • the substrate in the cell lysates was allowed to bind to the anchoring antibody for 1 hour shaking at room temperature. The lysate was then removed, and the assay plate was washed. All ELISA plate washings were done by rinsing in water 3 times followed by one rinse with TBST. The plate was dried by tapping it on paper towels. Phosphotyrosine was detected by adding lOO ⁇ l/well anti-pTyr antiserum diluted 1:3000 with TBST to the wells and incubating for 30 minutes shaking at room temperature. The unbound excess anti-pTyr antiserum was then removed, and the assay plate was washed as described above.
  • Results of several nitrothiazole compounds of the invention are presented in Table I above.
  • the activity of the compounds are represented by the concentration of the compound which produces the indicated percentage increase in the content of phosphotyrosine over the vanadate control (see Table I) .
  • the assay may also be used for testing compounds of the invention for their ability to inhibit the dephosphorylation of other substrate molecules, such as insulin-like growth factor 1 receptor (IGF-1R) and epidermal growth factor receptor (EGFR) .
  • IGF-1R insulin-like growth factor 1 receptor
  • EGFR epidermal growth factor receptor
  • NIH3T3/IGF-1R cells expressing IGF-1R starved in serum free media were seeded in the wells of tissue culture plates at a density 20,000 cells/well. The wells of ELISA plate were coated with anti- IGF-1R antibodies.
  • NIH3T3/EGFR cells expressing EGFR grown in media containing 0.5% for 40 hours were seeded in the wells of 96-well tissue culture plates at a density 10,000 cells/well.
  • the wells of ELISA plate were coated with anti-EGFR antibodies.
  • TrkA-HA TrkA receptor
  • ELISA enzyme-linked immunosorbent assay
  • fluorescent assay format 10 overexpressed in PC12 cells in an enzyme-linked immunosorbent assay (ELISA) or fluorescent assay format.
  • the assay may be used with any compounds of the invention.
  • substrate molecules such as platelet derived growth factor receptor, may be used in
  • the assay by using a different target cell and anchoring antibody.
  • the activities of the compounds of the invention toward different protein tyrosine phosphatases may be assessed.
  • FBS fetal bovine serum
  • Detection antibody Biotinylated anti- phosphotyrosine monoclonal antibody, 4G10-Biotin. (Upstate Biotechnology Cat. # 16-103) 12. Fluorescent probe: Europium-labeled Streptavidin. (Wallac Cat. # 1244-360) 13. Assay buffer, used to dilute Eu-Streptavidin. (Wallac Cat. # 1244-106) This can also be home made using the following formulation: 1 X TBS 0.005% Tween-40, Sigma Cat. # P-1504
  • HA under the CMV promoter (vector pcDNA3, Invitrogen) .
  • the cells need to be passed when they are 60-70% confluent (about 1 10 7 cells/plate) , usually 1/6 once every four days. 2.
  • Viral infection (day 1).
  • the infection was performed on 40-50% confluent 10- cm plates (3-4 days after splitting) which usually contain 6-8 10 6 cells per plate.
  • the 96-well plates were precoated with collagen I.
  • the collagen stock was diluted to
  • - Detection antibody was removed and washed as above. - 100 ⁇ l Eu-Steptavidin at 50 ng/ml in Assay Buffer (dilution 1/2000 of stock sol. at 100 ng/ ⁇ l) was added. Samples were incubated for 3045 min. at R.T. with agitation.
  • Enhancement Solution 100 ⁇ l Enhancement Solution was added and samples were shaken for 5 min. at R.T.
  • This assay was used to assess the ability of the compounds of the invention to inhibit phosphatase activity that is involved in the signaling pathway that regulates the insulin-induced facilitated transport of glucose into adipocytes. It has been shown that incubation of isolated adipocytes with vanadate resulted in a dose-dependent increase in the rate of glucose uptake. Any compounds of the invention may be tested in this assay.
  • the cell line used for the glucose transport assay was 3T3-L1, a preadipocyte cell line (American Type Culture Collection CL173) which overexpress the insulin receptor.
  • the 3T3-L1 cells were first differentiated by treating cells under confluent growth in DMEM containing 10% fetal bovine serum (FBS) with 0.5mM 3-isobutyl-l-methyl-xanthine, 5 ⁇ g/ml porcine insulin, 250mM dexamethasone for 2 days. The cells were then grown in DMEM containing 10% FBS and 5 ⁇ g/ml porcine insulin for two days, after which the cells were cultured in DMEM containing only 10% FBS.
  • FBS fetal bovine serum
  • Cells for use in the assay were first grown overnight in DMEM media and 1% FBS at 37°C at 5% C0 2 . Two hours before use, the overnight media was replaced with serum free DMEM containing 5mM glucose. After washing the cells twice with phosphate buffered saline (PBS) , serial dilutions of the compounds of the invention diluted 1:100 into DMEM were added to the wells for a final concentration range of 0.1 ⁇ M to 500 ⁇ M. Negative control wells received DMEM only. The cells were incubated with the test compound for 1-4 hours at 37°C at 5% C0 2 .
  • PBS phosphate buffered saline

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Emergency Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
EP96918441A 1995-06-07 1996-06-07 Phosphatase inhibitoren Withdrawn EP0831795A2 (de)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US47613795A 1995-06-07 1995-06-07
US476137 1995-06-07
PCT/US1996/009960 WO1996040113A2 (en) 1995-06-07 1996-06-07 Phosphatase inhibitors

Publications (1)

Publication Number Publication Date
EP0831795A2 true EP0831795A2 (de) 1998-04-01

Family

ID=23890656

Family Applications (1)

Application Number Title Priority Date Filing Date
EP96918441A Withdrawn EP0831795A2 (de) 1995-06-07 1996-06-07 Phosphatase inhibitoren

Country Status (3)

Country Link
EP (1) EP0831795A2 (de)
AU (1) AU6110496A (de)
WO (1) WO1996040113A2 (de)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TWI493205B (zh) * 2011-06-08 2015-07-21 Linear Techn Inc 用於增進以半導體為基礎之取樣系統之效能的系統與方法(二)

Families Citing this family (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5958957A (en) * 1996-04-19 1999-09-28 Novo Nordisk A/S Modulators of molecules with phosphotyrosine recognition units
JP2000511883A (ja) * 1996-04-19 2000-09-12 ノボ ノルディスク アクティーゼルスカブ ホスホチロシン認識ユニットを有する分子のモジュレーター
ES2384551T3 (es) * 1997-03-05 2012-07-06 Sugen, Inc. Formulaciones para agentes farmacéuticos hidrófobos
AU749132B2 (en) 1997-08-28 2002-06-20 Biovitrum Ab Inhibitors of protein tyrosine phosphatase
US6410585B1 (en) * 1997-08-28 2002-06-25 Scott D. Larsen Inhibitors of protein tyrosine phosphatase
JP2004500308A (ja) * 1998-03-12 2004-01-08 ノボ ノルディスク アクティーゼルスカブ プロテインチロシンホスアターゼのモジュレーター
US6355786B1 (en) 1998-10-30 2002-03-12 Vanderbilt University Purified and isolated protein zero related (PZR) and therapeutic and screening methods using same
US7064114B2 (en) 1999-03-19 2006-06-20 Parker Hughes Institute Gel-microemulsion formulations
AU7049401A (en) * 2000-05-03 2001-11-12 Hoffmann La Roche Alkynyl phenyl heteroaromatic glucokinase activators
US6498182B2 (en) 2000-09-26 2002-12-24 Biovitrum Ab Compounds
US7521473B2 (en) 2004-02-25 2009-04-21 Wyeth Inhibitors of protein tyrosine phosphatase 1B
WO2013020024A2 (en) * 2011-08-03 2013-02-07 Karyopharm Therapeutics, Inc. Maleimide compounds and methods of treatment

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3870725A (en) * 1971-03-30 1975-03-11 Lilly Industries Ltd Nitrothiazole derivatives
US3850939A (en) * 1973-02-20 1974-11-26 Parke Davis & Co 5-nitro-2-thiazolylthiopyridine,1-oxides
DE69001809T2 (de) * 1990-03-19 1994-01-05 Agfa Gevaert Nv Elektronenacceptoren enthaltende Photomaterialien.

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO9640113A3 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TWI493205B (zh) * 2011-06-08 2015-07-21 Linear Techn Inc 用於增進以半導體為基礎之取樣系統之效能的系統與方法(二)

Also Published As

Publication number Publication date
WO1996040113A2 (en) 1996-12-19
AU6110496A (en) 1996-12-30
WO1996040113A3 (en) 1997-01-23

Similar Documents

Publication Publication Date Title
US5798374A (en) Methods of inhibiting phosphatase activity and treatment of disorders associated therewith
US5602171A (en) Methods of inhibiting phosphatase activity and treatment of disorders associated therewith using naphthopyrones and derivatives thereof
US6080772A (en) Thiazole compounds and methods of modulating signal transduction
MXPA96006404A (es) Composiciones farmaceuticas y metodos para formular trasnduccion de señales
Chen et al. Metformin suppresses adipogenesis through both AMP-activated protein kinase (AMPK)-dependent and AMPK-independent mechanisms
EP1007042A1 (de) Neue heterozyklische verbindungen zur modulation der protein-tyrosin-enzym-verwandten zellulären signaltransduktion
US6235769B1 (en) Methods of preventing and treating neurological disorders with compounds that modulate the function of the C-RET receptor protein tyrosine kinase
US6596772B1 (en) Phosphate mimics and methods of treatment using phosphatase inhibitors
AU760964B2 (en) 3-heteroarylidenyl-2-indolinone compounds for modulating protein kinase activityand for use in cancer chemotherapy
EP0831795A2 (de) Phosphatase inhibitoren
US20090291857A1 (en) Methods to identify inhibitors of the unfolded protein response
US20020068687A1 (en) Methods and compositions for inhibiting cell proliferative disorders
EP0788358A2 (de) Pharmazeutische pyrazolzusammensetzungen zur verwendung als proteinkinasehemmern
Gorojankina et al. Discovery, molecular and pharmacological characterization of GSA-10, a novel small-molecule positive modulator of Smoothened
Chiusa et al. Cancer therapy modulates VEGF signaling and viability in adult rat cardiac microvascular endothelial cells and cardiomyocytes
US6316479B1 (en) Isoxazole-4-carboxamide compounds active against protein tryosine kinase related disorders
Zhang et al. An underlying mechanism of dual Wnt inhibition and AMPK activation: mitochondrial uncouplers masquerading as Wnt inhibitors
US20220257571A1 (en) Inhibitor of map kinase interacting serine/threonine kinase 1 (mnk1) and map kinase interacting serine/threonine kinase 2 (mnk2), cancer therapy and therapeutic combinations
Little et al. Suramin inhibits PDGF-stimulated receptor phosphorylation, proteoglycan synthesis and glycosaminoglycan hyperelongation in human vascular smooth muscle cells
Caligiuri et al. FT-6876, a potent and selective inhibitor of CBP/p300, is active in preclinical models of androgen receptor-positive breast cancer
CA3078247A1 (en) Methods and compositions for treating urea cycle disorders, in particular otc deficiency
Parameswaran et al. SB203580 reverses adrenomedullin's effect on proliferation and apoptosis in cultured mesangial cells
CN113194942A (zh) 用于抑制和/或治疗生长相关疾病和/或其临床病症的组合物和方法
Kansra et al. Src family kinase inhibitors block amphiregulin-mediated autocrine ErbB signaling in normal human keratinocytes
Yu et al. Anti-proliferation effect of 3-amino-2-imino-3, 4-dihydro-2H-1, 3-benzothiazin-4-one (BJ-601) on human vascular endothelial cells: G0/G1 p21-associated cell cycle arrest

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 19971217

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): CH DE FR GB IT LI

17Q First examination report despatched

Effective date: 20000824

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20010306