WO2008068751A1 - Novel protein kinase modulators and therapeutic uses thereof - Google Patents

Novel protein kinase modulators and therapeutic uses thereof Download PDF

Info

Publication number
WO2008068751A1
WO2008068751A1 PCT/IL2007/001494 IL2007001494W WO2008068751A1 WO 2008068751 A1 WO2008068751 A1 WO 2008068751A1 IL 2007001494 W IL2007001494 W IL 2007001494W WO 2008068751 A1 WO2008068751 A1 WO 2008068751A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
halogen
compounds
ptk
receptor
Prior art date
Application number
PCT/IL2007/001494
Other languages
French (fr)
Inventor
Hadas Reuveni
Alexander Levitzki
Lilach Steiner
Revital Sasson
Iris Ben-David
Avi Weissberg
Original Assignee
Novotyr Therapeutics Ltd.
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 Novotyr Therapeutics Ltd. filed Critical Novotyr Therapeutics Ltd.
Priority to BRPI0719708-0A2A priority Critical patent/BRPI0719708A2/en
Priority to CA002671632A priority patent/CA2671632A1/en
Priority to AU2007330333A priority patent/AU2007330333A1/en
Priority to EP07827467.7A priority patent/EP2125712B1/en
Priority to JP2009539869A priority patent/JP2010511694A/en
Priority to US12/517,278 priority patent/US8058309B2/en
Publication of WO2008068751A1 publication Critical patent/WO2008068751A1/en
Priority to US13/293,546 priority patent/US20120083528A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C327/00Thiocarboxylic acids
    • C07C327/38Amides of thiocarboxylic acids
    • C07C327/40Amides of thiocarboxylic acids having carbon atoms of thiocarboxamide groups bound to hydrogen atoms or to acyclic carbon atoms
    • C07C327/44Amides of thiocarboxylic acids having carbon atoms of thiocarboxamide groups bound to hydrogen atoms or to acyclic carbon atoms to carbon atoms of an unsaturated carbon skeleton
    • 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
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis

Definitions

  • the present invention relates to novel tyrphostin derivatives, their preparation, pharmaceutical compositions comprising same, and their use in treatment of protein kinase related disorders.
  • PTKs Protein tyrosine kinases
  • PTKs are a family of enzymes, which transfer the ⁇ - phosphate of ATP to the side chain of tyrosine residues on substrate proteins.
  • PTKs are involved in a variety of cellular processes, including signal transduction and growth regulation. Phosphorylation of substrates by PTKs are key events in cellular signaling.
  • RTKs receptor tyrosine kinases
  • RTKs receptor tyrosine kinases
  • kinases belong to a family of transmembrane proteins and have been implicated in cellular signaling pathways. The predominant biological activity of some receptor kinases is the stimulation of cell growth and proliferation, while other receptor tyrosine kinases are involved in inhibiting growth and promoting differentiation.
  • a single tyrosine kinase can inhibit, or stimulate, cell proliferation depending on the cellular environment in which it is expressed (Schlessinger and Ullrich, Neuron (1992), 9(3): 383-391).
  • RTKs include receptors for platelet-derived growth factor (PDGF), fibroblast growth factor (FGF), hepatocyte growth factor (HGF), insulin, insulin-like growth factor 1 (IGF-I), nerve growth factor (NGF), vascular endothelial growth factor (VEGF), macrophage colony stimulating factor (M-CSF) and others.
  • PDGF platelet-derived growth factor
  • FGF fibroblast growth factor
  • HGF hepatocyte growth factor
  • IGF-I insulin-like growth factor 1
  • NEF nerve growth factor
  • VEGF vascular endothelial growth factor
  • M-CSF macrophage colony stimulating factor
  • Receptor tyrosine kinases are composed of at least three domains: an extracellular glycosylated ligand binding domain, a transmembrane domain and a cytoplasmic catalytic domain that can phosphorylate tyrosine residues. Binding of a ligand to membrane-bound receptors induces the formation of receptor dimers and allosteric changes thus activating the intercellular kinase domains which further results self-phosphorylation (autophosphorylation and/or transphosphorylation) of the receptor on tyrosine residues. Receptor phosphorylation stimulates physical association of the activated receptor with target molecules. Some of the target molecules are, in turn, phosphorylated, a process which transmits the signal to the cytoplasm.
  • the secondary signal transducer molecules generated by activated receptors result in a signal cascade that regulates cell functions such as cell division or differentiation. Intracellular signal transduction is reviewed in Aaronson, Science (1991), 254: 1146-1153; Schlessinger, J Trends Biochem. ScI (1988), 13: 443-447; Ullrich and Schlessinger, Cell (1990), 61: 203-212.
  • PTKs Various cell proliferative disorders have been associated with defects in pathways mediated by PTKs. Enhanced activities of PTKs resulting from overexpression of the normal kinase, upregulation of ligands of receptor tyrosine kinases or activating mutations, are a hallmark of many diseases which involve cellular proliferation, including cancer. Examples of specific receptor tyrosine kinases associated with cell proliferative disorders include platelet derived growth factor receptor (PDGFR), insulin-like growth factor 1 receptor (IGF-IR), epidermal growth factor receptor (EDFR), and the related HER2.
  • PDGFR platelet derived growth factor receptor
  • IGF-IR insulin-like growth factor 1 receptor
  • EDFR epidermal growth factor receptor
  • PTKs The involvement of PTKs in various diseases identifies them as targets for antiproliferative drugs. Indeed, numerous PTK blockers have been described in the literature including proposed mechanisms of action (Levitzki et al., Science (1995), 267:1782-88; Posner et al., MoI. Pharmacol. (1994), 45:673-683). Applicants have developed a family of PTK inhibitors, named tyrphostins, designed to mimic the tyrosine substrate (Levitzki et al., Science (1995), 267:1782-88; Levitzki et al., Biochem. Pharm. (1990), 40:913-920; Levitzki et al., FASEB J.
  • tyrphostins and in particular tyrphostins of the benzylidene malonitril type, are the hydrophilic catechol ring and the more lipophilic substituted cyano- vinyl radical.
  • Kinetic studies have shown that some tyrphostin compounds are pure competitive inhibitors vis-a-vis tyrosine substrates whereas for the ATP binding site they act as non-competitive inhibitors (Yaish et al., Science (1988), 242:933-935; Gazit et al., J Med. Chem.
  • the present invention relates to new tyrphostins compounds useful as inhibitors of protein tyrosine kinases (PTKs) in cells.
  • novel tyrphostin compounds show increased inhibitory properties of, but not limited to, insulin-like growth factor 1 receptor (IGFlR), platelet derived growth factor receptor (PDGFR), epidermal growth factor receptor (EGFR), and IGFlR-related insulin receptor (IR).
  • IGFlR insulin-like growth factor 1 receptor
  • PDGFR platelet derived growth factor receptor
  • EGFR epidermal growth factor receptor
  • IR IGFlR-related insulin receptor
  • the present invention is further directed to compounds that trigger Serine phosphorylation of the IGFlR direct substrate IRSl, thus providing long-lasting effects which enhance the inhibitory activity of these novel tyrphostins.
  • the present invention provides compounds represented by the structure of formula 1 :
  • R 1 , R 2 , R 5 and R 6 are independently selected from H, C 1 -C 4 alkyl, acyl and a functional group that gives rise to hydroxyl upon hydrolysis;
  • R 3 and R 7 are independently selected from H, halogen, haloalkyl and OR 8 wherein R is H, C 1 -C 4 alkyl, acyl or a functional group that gives rise to hydroxyl upon hydrolysis; R 4 is H or CN 5 including salts, hydrates, solvates, polymorphs, optical isomers, geometrical isomers, enantiomers, diastereomers, and mixtures thereof.
  • the present invention provides a compound represented by the structure of formula 1, wherein R 3 is halogen (e.g., F, Cl, Br or I) or haloalkyl (e.g. CF 3 ).
  • the present invention provides a compound represented by the structure of formula 1, wherein R 1 , R 2 , R 5 and R 6 are each H. In another embodiment, the present invention provides a compound represented by the structure of formula 1, wherein R 7 is hydrogen, halogen or OR 8 with R 8 being H or methyl. In one embodiment, the present invention further provides compounds of formula 1, wherein R 4 is H. In another embodiment, the present invention provides compounds of formula 1, wherein R 4 is CN.
  • the present invention further provides pharmaceutical compositions comprising any of the compounds represented by the structure of formula 1.
  • R 1 , R 2 , R 5 and R 6 are independently selected from H, Ci-C 4 alkyl, acyl and a functional group that gives rise to hydroxyl upon hydrolysis;
  • R 3 and R 7 are independently selected from H, halogen, haloalkyl and OR 8 wherein R 8 is H, C 1 -C 4 alkyl, acyl or a functional group that gives rise to hydroxyl upon hydrolysis;
  • R 4 is H or CN, including salts, hydrates, solvates, polymorphs, optical isomers, geometrical isomers, enantiomers, diastereomers, and mixtures thereof.
  • the present invention further provides a method of inhibiting a protein tyrosine kinase (PTK) comprising contacting the PTK with an effective inhibitory amount of a compound of formula 1.
  • PTK protein tyrosine kinase
  • the present invention further provides a method of inhibiting a protein tyrosine kinase (PTK) in a subject comprising the step of administering to the subject a therapeutically effective amount of a compound of formula 1, to inhibit a protein tyrosine kinase (PTK) in the subject, hi another embodiment, the method comprises administering to the subject a pharmaceutical composition comprising a therapeutically effective amount of any of the compound of formula 1, and a pharmaceutically acceptable excipient.
  • PTK protein tyrosine kinase
  • the present invention further provides a method of treating or preventing a protein tyrosine kinase (PTK) related disorder in a subject comprising the step of administering to the subject a therapeutically effective amount of any of the compounds of formula 1.
  • the method comprises administering to the subject a pharmaceutical composition comprising a therapeutically effective amount of a compound of formula 1 , and a pharmaceutically acceptable excipient.
  • the PTK related disorder is a cell proliferative disorder, a metabolic disorder or a fibrotic disorder.
  • the PTK related disorder is cancer
  • the PTK related disorder is diabetic nephropathy.
  • FIG.l Shows in schematic form a process for the synthesis of exemplary novel Tyrphostins of the invention.
  • X and Y are independently selected from hydrogen, halogen, haloalkyl and OR 8 wherein R 8 is H, C 1 -C 4 alkyl, acyl or a functional group that gives rise to hydroxyl upon hydrolysis.
  • FIG.2 Shows in schematic form a process for the synthesis of exemplary novel Tyrphostins of the invention.
  • X and Y are independently selected from hydrogen, halogen, haloalkyl and OR 8 wherein R 8 is H, C 1 -C 4 alkyl, acyl or a functional group that gives rise to hydroxy 1 upon hydrolysis.
  • FIG.3 Shows in schematic form a process for the synthesis of intermediates used in the synthesis of the novel Tyrphostins of the invention.
  • FIGS.4A and 4B show that compound 7 induces inhibition of IGFlR and its signaling in intact cancer cells.
  • Figure 4 A shows the inhibition of IGFlR activation and signaling in cancer cells following exposure to compound 7 in comparison to the control (compound # 20).
  • Compound 7 induces a decrease in the tyrosine-phosphorylation of IRSl, a direct substrate of IGFlR, and inhibition of IGFl-induced activation of the PKB and ERK pathways.
  • Figure 4B represents the dose-dependent inhibition of IGFl- induced signaling in cancer cells.
  • FIGS. 5 A 5B and 5C show that compound 7 induces Serine-phosphorylation on IRSl and a decrease in the cellular levels of IRSl in breast cancer MCF7 cells.
  • Figure 5 A shows a decrease in cellular levels of IRSl induced by 4 hours treatment with compound 7.
  • Figure 5B shows induced Serine-phosphorylation on IRSl following 4 hours treatment with compound 7.
  • Figure 5 C shows a long-lasting (24 hours following exposure) reduction in IRSl levels in cancer cells.
  • FIG. 6 shows Ser-phosphorylation on IRSl following treatment of breast cancer MCF7 cells with compounds 7, 8, 9, 10, 11, and 13. Inhibition of the activation of IGFlR downstream signaling pathways, namely ERK and PKB are demonstrated as well.
  • FIGS. 7A and 7B show treatment of ovarian cancer A2780 cells with compounds 7, 8 and 9 in comparison to compound 6.
  • Figure 7A shows that cells treated with compounds 6, 7, 8 and 9 undergo apoptosis following incubation (cleaved PARP detection) with compound 6 being slightly weaker.
  • Figure 7B shows Ser- phosphorylation on IRSl (detected by the shift of the IRSl band upward) as well as a decrease in IRSl levels induced by compounds 7, 8 and 9 in contrast to compound 6.
  • FIG. 8 shows that Ser-phosphorylation on IRSl as well as cleavage of PARP (apoptosis) are detected in metastatic melanoma YUMAC cells treated with compounds 7, 8, 9, 10, and 13 in comparison to compounds 11 and 19.
  • FIGS. 9A, 9B and 9C show that compound 7 inhibits the activation and downstream signaling of PDGFR, EGFR and IR tyrosine kinases in intact cells.
  • Figure 9A shows that compound 7 inhibits EGFR activation in hormone-refractory prostate cancer PC3 cells, and EGF-induced phosphorylation of PKB and STAT3, a direct substrate of EGFR.
  • Figure 9B shows that compound 7 inhibits ligand-induced PDGFR activation and downstream signaling in NIH3T3 cells over expressing PDGFR.
  • Figure 9C shows that compound 7 inhibits ligand-induced Insulin receptor (IR) activation and downstream regulation in NIH3T3 cells over expressing IR.
  • IR Insulin receptor
  • FIG. 10 shows that treatment of nude mice with compound 7 (IP, I/day, 50 mg/kg) resulted in 82% growth inhibition of hormone-refractory prostate cancer (HRPC) PC3 tumors.
  • HRPC hormone-refractory prostate cancer
  • FIG. 12 shows that treatment of nude mice with compounds 7 or 8 (IP, I/day, 20 mg/kg) resulted in 87% growth inhibition of ovarian cancer A2780 tumors.
  • FIG. 13 shows the high efficacy of compounds 7, 8, and 9 as compared to compound 6 in growth inhibition of ovarian cancer A2780 tumors in nude mice model.
  • FIG. 14 shows that treatment of nude mice, bearing ovarian A2780 tumors larger than 50mm 3 , with compound 8 (IP, I/day, 50 mg/kg) resulted in 82% growth inhibition of the tumors.
  • FIG. 15 shows that treatment of nude mice, bearing ovarian A2780 tumors larger than 470mm 3 , with compound 8 (IP, I/day, 50 mg/kg) resulted in approximately 85% growth inhibition of the tumors.
  • FIG. 16 shows that treatment of nude mice with compound 7 (intratumoral, I/day, 125 ⁇ g/day) resulted in 50% growth inhibition of pancreatic cancer Panel tumors.
  • FIG. 17 shows the efficacy of compound 7 as compared to compound 6 in growth inhibition of melanoma B16 tumors in nude mice model.
  • the inhibitors were administered IP at dose of 20 mg/kg, I/day.
  • FIG. 18 shows the efficacy of compound 8 in growth inhibition of human melanoma YUMAC tumors in nude mice model.
  • Compound 8 was administered IP at dose of 20 mg/kg, I/day.
  • FIGS. 19A and 19B show the effect of compound 6 on MCF7 cells.
  • Figure 19A shows that compound 6 inhibits IGFl -induced IGFlR activation and signaling (auto- phosphorylation of IGFlR 5 the IGFl -induced tyrosine phosphorylation of IRSl and the IGFl -induced activation of the Akt/PKB).
  • Figure 19B shows that compound 6 has no effect on the IRS 1 levels.
  • FIGS. 2OA and 2OB show the effect of compound 6 on EGFR activity in HRPC PC3 cells.
  • Figure 2OA shows that compound 6 inhibits EGF-induced phosphorylation of EGFR and its downstream elements, STAT3 and PKB.
  • Figure 2OB shows that protein levels are equal and no effect on STAT3, PBCB or ERK was detected following treatment of PC3 cells with compound 6.
  • the present invention relates to novel tyrphostin derivatives which are potent PTK-inhibitors.
  • the present invention further provides methods for inhibiting PTKs 5 for example IGF-I receptor (IGFlR).
  • IGFlR IGF-I receptor
  • the compounds are useful in treating or preventing PTK-related disease states, particularly PTK-related disorders which are associated with defects in signaling pathways mediated by PTKs. Examples of diseases involving cellular proliferation are cancer and psoriasis.
  • the compounds of the present invention are designed to have an enhanced inhibiting potency with respect to protein tyrosine kinases (PTKs), compared with previously disclosed tyrphostin derivatives (Blum et al., Biochem. (2000), 39:15705- 15712; US Pat. Nos.
  • PTKs protein tyrosine kinases
  • the compounds provided by the present invention are represented by the general formula 1 :
  • R 1 , R 2 , R 5 and R 6 are independently selected from H, C 1 -C 4 alkyl, acyl and a functional group that gives rise to hydroxyl upon hydrolysis;
  • R 3 and R 7 are independently selected from H, halogen, haloalkyl and OR 8 wherein R 8 is H, C 1 -C 4 alkyl, acyl or a functional group that gives rise to hydroxyl upon hydrolysis;
  • R 4 is H or CN, including salts, hydrates, solvates, polymorphs, optical isomers, geometrical isomers, enantiomers, diastereomers, and mixtures thereof.
  • the present invention provides a compound represented by the structure of formula 1, wherein R 1 , R 2 , R 5 and R 6 each independently H or methyl.
  • the present invention provides a compound represented by the structure of formula 1, wherein R 3 is halogen (e.g. F, Cl, Br, I), hydroxyl or haloalkyl (e.g. CF 3 ).
  • R 4 is CN.
  • the present invention provides a compound represented by the structure of formula 1, wherein R is H, halogen or OR with R being H or methyl.
  • the present invention further provides a compound of formula 1, wherein R 4 is H.
  • Representative and non-limiting examples of such structures are compounds selected from the group consisting of compounds 2-18:
  • alkyl group refers to a saturated aliphatic hydrocarbon, including straight-chain, branched-chain and cyclic alkyl groups.
  • the alkyl group has 1-12 carbons designated here as CrC ⁇ -alkyl.
  • the alkyl group has 1-6 carbons designated here as Ci-C ⁇ -alkyl.
  • the alkyl group has 1-4 carbons designated here as C 1 -C 4 - alkyl.
  • the alkyl group may be unsubstituted or substituted by one or more groups selected from halogen, hydroxy, alkoxy carbonyl, amido, alkylamido, dialkylamido, nitro, amino, alkylamino, dialkylamino, carboxyl, thio and thioalkyl.
  • a “hydroxy” group refers to an OH group.
  • An “alkoxy” group refers to an -O-alkyl group wherein R is alkyl as defined above.
  • a “thio” group refers to an - SH group.
  • An “alkylthio” group refers to an -SR group wherein R is alkyl as defined above.
  • amino group refers to an NH 2 group.
  • alkylamino group refers to an -NHR group wherein R is alkyl is as defined above.
  • a dialkylamino group refers to an -NRR' group wherein R and R' are alkyl as defined above.
  • An “amido” group refers to a -C(O)NH 2 group.
  • An alkylamido group refers to an -C(O)NHR group wherein R is alkyl is as defined above.
  • a dialkylamido group refers to an -C(O)NRR' group wherein R and R' are alkyl as defined above.
  • a “thioamide” group refers to a C(S)NHR, where R is either alkyl, aryl, alkylaryl or hydrogen.
  • halogen or "halo” as used herein alone or as part of another group refers to chlorine, bromine, fluorine, and iodine.
  • haloalkyl refers to an alkyl group having some or all of the hydrogens independently replaced by a halogen group including, but not limited to, trichloromethyl, tribromomethyl, trifluoromethyl, triiodomethyl, difluoromethyl, chlorodifluoromethyl, pentafluoroethyl, 1,1-difluoroethyl bromomethyl, chloromethyl, fluoromethyl, iodomethyl, and the like.
  • Examples of functional groups that give rise to hydroxyl upon hydrolysis include, but are not limited to, esters, anhydrides, carbamates, carbonates and the like.
  • R 1 , R 2 , R 5 or R 6 is an acyl group (COR)
  • the resulting function group is an ester (OCOR).
  • the resulting function group is a carbamate (OCONHR).
  • the resulting function group is a carbonate (OCOOR).
  • All stereoisomers of the compounds of the present invention are contemplated, either in admixture or in pure or substantially pure form. These compounds can have asymmetric centers at any of the atoms. Consequently, the compounds can exist in enantiomeric or diastereomeric forms or in mixtures thereof.
  • the present invention contemplates the use of any racemates (i.e. mixtures containing equal amounts of each enantiomers), enantiomerically enriched mixtures (i.e., mixtures enriched for one enantiomer), pure enantiomers or diastereomers, or any mixtures thereof.
  • the chiral centers can be designated as R or S or R 5 S or d,D, 1,L or d,l, D,L.
  • Compounds comprising amino acid residues include residues of D-amino acids, L-amino acids, or racemic derivatives of amino acids.
  • several of the compounds of the present invention contain one or more double bonds.
  • the present invention intends to encompass all structural and geometrical isomers including cis, trans, E and Z isomers, independently at each occurrence.
  • salt encompasses both basic and acid addition salts, including but not limited to carboxylate salts or salts with amine nitrogens, and include salts formed with the organic and inorganic anions and cations discussed below. Further encompassed by the term are salts formed by standard acid-base reactions with basic groups (such as amino groups) and organic or inorganic acids.
  • Such acids include hydrochloric, hydrofluoric, trifluoroacetic, sulfuric, phosphoric, acetic, succinic, citric, lactic, maleic, fumaric, palmitic, cholic, pamoic, mucic, D-glutamic, D-camphoric, glutaric, phthalic, tartaric, lauric, stearic, salicyclic, methanesulfonic, benzenesulfonic, sorbic, picric, benzoic, cinnamic, and the like.
  • organic or inorganic cation refers to counter-ions for the carboxylate anion of a carboxylate salt.
  • the counter-ions are chosen from the alkali and alkaline earth metals (such as lithium, sodium, potassium, barium, aluminum and calcium); ammonium and mono-, di- and tri-alkyl amines such as trimethylamine, cyclohexylamine; and the organic cations, such as dibenzylammonium, benzylammonium, 2-hydroxyethylarnmonium, bis(2-hydroxyethyl)ammonium, phenylethylbenzylammonium, dibenzylethylenediammonium, and like cations.
  • the present invention also includes solvates of compounds of formula 1 or any of compounds 2-19 and salts thereof.
  • “Solvate” means a physical association of a compound of the invention with one or more solvent molecules. This physical association involves varying degrees of ionic and covalent bonding, including hydrogen bonding. In certain instances the solvate will be capable of isolation.
  • “Solvate” encompasses both solution-phase and isolatable solvates. Non-limiting examples of suitable solvates include ethanolates, methanolates and the like.
  • “Hydrate” is a solvate wherein the solvent molecule is water.
  • the present invention also includes polymorphs of compounds of formula 1 or any of compounds 2-19 and salts thereof.
  • polymorph refers to a particular crystalline state of a substance, which can be characterized by particular physical properties such as X-ray diffraction, IR spectra, melting point, and the like.
  • the present invention provides compounds and compositions effective at inhibiting protein tyrosine kinases. These compounds and compositions are potentially useful in the treatment of a diseases associated with altered or abnormal activity of protein tyrosine kinases such as enhanced activity of protein tyrosine kinases.
  • the present invention provides a method of inhibiting a protein tyrosine kinase (PTK) comprising contacting the PTK with an effective inhibitory amount of a compound represented by any of formulas 1-19.
  • PTK protein tyrosine kinase
  • the present invention further provides a method of treating or preventing a protein tyrosine kinase (PTK) in a subject comprising the step of administering to the subject a therapeutically effective amount of any of the compounds represented by formulas 1-19.
  • the method comprises administering to the subject a pharmaceutical composition comprising a therapeutically effective amount of any of the compounds represented by formulas 1-19 and a pharmaceutically acceptable excipient.
  • the present invention further provides a method of inhibiting a protein tyrosine kinase (PTK) related disorder in a subject comprising the step of administering to the subject a therapeutically effective amount of any of the compounds represented by formulas 1-19.
  • the method comprises administering to the subject a pharmaceutical composition comprising a therapeutically effective amount of any of the compounds represented by formulas 1-19; and a pharmaceutically acceptable excipient.
  • a "protein tyrosine kinase” (PTK) is a protein belonging to a family of enzymes that transfer the ⁇ -phosphate of ATP to the side chain of tyrosine residues on substrate proteins. PTKs are involved in a variety of key cellular processes, including signal transduction and growth regulation.
  • a protein tyrosine kinase refers to a receptor tyrosine kinase (RTK) as well as a cellular tyrosine kinase (CTK or non-receptor tyrosine kinase).
  • RTK receptor tyrosine kinase
  • CTK cellular tyrosine kinase
  • the compounds of the present invention are effective at inhibiting both receptor and non-receptor protein tyrosine kinases.
  • a cellular tyrosine kinase (CTK or non-receptor tyrosine kinase) is an intracellular protein which takes part in signal transduction within the cell, including signal transduction to the nucleus.
  • CTKs are the Src family of oncoproteins.
  • a receptor tyrosine kinase (RTK) is a transmembrane protein that participates in transmembrane signaling pathways. The predominant biological activity of some receptor tyrosine kinases is the stimulation of cell growth and proliferation, while other receptor tyrosine kinases are involved in arresting growth and promoting differentiation.
  • RTKs include the receptors for platelet-derived growth factor (PDGF), fibroblast growth factor (FGF), hepatocyte growth factor (HGF), insulin, insulin-like growth factor- 1 (IGF-I), nerve growth factor (NGF), vascular endothelial growth factor (VEGF), and macrophage colony stimulating factor (M-CSF).
  • PDGF platelet-derived growth factor
  • FGF fibroblast growth factor
  • HGF hepatocyte growth factor
  • IGF-I insulin-like growth factor- 1
  • NEF nerve growth factor
  • VEGF vascular endothelial growth factor
  • M-CSF macrophage colony stimulating factor
  • protein tyrosine kinase related disorder refers to a disorder characterized by abnormal or altered PTK activity.
  • Abnormal or altered activity further refers to either (i) overexpression of PTK in cells that do not normally express PTKs; (ii) increased PTK expression leading to unwanted cell proliferation, differentiation and/or growth; or, (iii) decreased PTK expression leading to unwanted reductions in cell proliferation, differentiation and/or growth.
  • Over-activity of PTKs refers to either amplification of the gene encoding a particular PTK or production of a level of PTK activity which can correlate with cell proliferation, differentiation and/or growth.
  • Over- activity can also be the result of ligand independent or constitutive activation as a result of mutations such as deletions of a fragment of a PTK responsible for ligand binding.
  • the present invention is directed to preparations containing any of the compounds represented by formulas 1-19, which modulate PTK activity signal transduction by affecting the enzymatic activity of the protein tyrosine kinases thereby interfering with the signal transduction pathways mediated by such proteins.
  • protein tyrosine kinase related disorders are cell proliferative disorders, metabolic disorders or fibrotic disorders and inflammation.
  • cell proliferative disorders which are mediated by protein tyrosine kinases are cancer, psoriasis, diabetic nephropathy, blood vessel proliferative disorders, and mesangia cell proliferative disorders.
  • Cancer is a disorder in which a population of cells has become, in varying degrees, unresponsive to the control mechanisms that normally govern proliferation and differentiation. Cancer refers to various types of malignant neoplasms and tumors, including metastasis to different sites.
  • Nonlimiting examples of cancers which can be treated by any of the compounds represented by formulas 1-19 are brain, ovarian, colon, prostate, kidney, bladder, breast, lung, oral and skin cancers which exhibit altered activity of PTK.
  • cancers which the compounds of the present invention are effective at treating or preventing are: adenocarcinoma, adrenal gland tumor, ameloblastoma, anaplastic tumor, anaplastic carcinoma of the thyroid cell, angiofibroma, angioma, angiosarcoma, apudoma, argentaffmoma, arrhenoblastoma, ascites tumor cell, ascitic tumor, astroblastoma, astrocytoma, ataxia-telangiectasia , atrial myxoma, basal cell carcinoma, benign tumor, bone cancer, bone tumor, brainstem glioma, brain tumor, breast cancer, Burkitt's lymphoma, carcinoma, cerebellar astrocytoma, cervical cancer, cherry angioma, cholangiocarcinoma, a cholangioma, chondroblastoma, chondroma, chondrosarcoma, chorioblastoma,
  • Blood vessel proliferative disorders refer to antiogenic and vasculogenic disorders generally resulting in abnormal proliferation of blood vessels.
  • Other examples of blood vessel proliferation disorders include arthritis and ocular diseases such as diabetic retinopathy, restenosis, retinopathies and atherosclerosis.
  • Mesangial cell proliferative disorders refer to disorders brought about by abnormal proliferation of mesangial cells.
  • Mesangial proliferative disorders include various human renal diseases such as glomerulonephritis, diabetic nephropathy, malignant nephrosclerosis, thrombic microangiopathy syndromes, transplant rejection and glomerulopathies, hi this regards, PDGFR has been implicated in the maintenance of mesangial cell proliferation.
  • Metabolic disorders that are implicated with abnormal PTK activity include psoriasis, diabetes mellitus, wound healing, inflammation and neurodegenerative diseases.
  • EGFR has been indicated in corneal and dermal wound healing.
  • Defects in the Insulin-R and IGF-IR receptor are indicated in type-II diabetes mellitus.
  • Fibrotic disorders refer to the abnormal formation of extracellular matrices.
  • fibrotic disorders include hepatic cirrhosis and mesangial cell proliferative disorders.
  • Hepatic cirrhosis is characterized by the increase in extracellular matrix constituents resulting in the formation of a hepatic scar.
  • the term "treating” as used herein refers to abrogating, inhibiting, slowing or reversing the progression of a disease, ameliorating clinical symptoms of a disease or preventing the appearance of clinical symptoms of a disease.
  • preventing is defined herein as barring a subject from acquiring a disorder or diseases.
  • administering refers to a method of bringing a compound of the present invention and a target protein tyrosine kinase together in such a manner that the tyrphostin can affect the catalytic activity of the tyrosine kinase directly; i.e. by interacting with the kinase itself, or indirectly; i.e. by interacting with another molecule on which the catalytic activity of the enzyme is dependent.
  • administration can be accomplished in vitro, i.e. in a test tube, or in vivo, i.e. in cells or tissues of living organisms, for example humans.
  • the present invention encompasses administering the compounds of the present invention to a subject.
  • contacting refers to bringing into contact the protein tyrosine kinase and the compounds defined herein, under in vivo conditions or in vitro conditions as defined above.
  • therapeutically effective amount refers to the amount of a compound being administered which relieves to some extent one or more of the symptoms of the disorder being treated.
  • Therapeutic effective doses for any compounds represented by formulas 1-19 described herein can be estimated initially from cell culture and/or an animal model. A dose can be formulated in an animal model, and this dose can be used to more precisely determine useful doses in humans.
  • an effective inhibitory amount refers to the amount of a compound being administered that inhibits to some extent the protein tyrosine kinase with which it is contacted.
  • the present invention further provides pharmaceutical compositions comprising any of the compounds represented by the structure of formulas 1-19, and a pharmaceutically acceptable carrier or excipient.
  • pharmaceutical composition means therapeutically effective amounts of the compounds of the present invention, together with suitable diluents, preservatives, solubilizers, emulsifiers, adjuvant and/or carriers.
  • a "therapeutically effective amount” as used herein refers to that amount which provides a therapeutic effect for a given condition and administration regimen.
  • compositions are liquids or Lyophilized or otherwise dried formulations and include diluents of various buffer content (e.g., Tris-HCL, acetate, phosphate), pH and ionic strength, additives such as albumin or gelatin to prevent absorption to surfaces, detergents (e.g., Tween 20, Tween 80, Pluronic F68, bile acid salts), solubilizing agents (e.g., glycerol, polyethylene glycerol), antioxidants (e.g., ascorbic acid, sodium metabisulfite), preservatives (e.g., Thimerosal, benzyl alcohol, parabens), bulking substances or tonicity modifiers (e.g., lactose, mannitol), covalent attachment of polymers such as polyethylene glycol to the protein, complexation with metal ions, or incorporation of the material into or onto particulate preparations of polymeric compounds such as polylactic acid, polglycolic acid, hydrogels
  • compositions coated with polymers e.g., poloxamers or poloxamines.
  • Other embodiments of the compositions of the invention incorporate particulate forms, protective coatings, protease inhibitors or permeation enhancers for various routes of administration, including parenteral, pulmonary, nasal and oral.
  • the pharmaceutical composition is administered parenterally, paracancerally, transmucosally, transdermally, intramuscularly, intravenously, intradermally, subcutaneously, intraperitonealy, intraventricularly, intracranially or intratumorally.
  • pharmaceutically acceptable carriers are well known to those skilled in the art and include, but are not limited to, 0.01-O.lM and preferably 0.05M phosphate buffer or 0.8% saline. Additionally, such pharmaceutically acceptable carriers may be aqueous or non-aqueous solutions, suspensions, and emulsions. Examples of nonaqueous solvents are propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable organic esters such as ethyl oleate. Aqueous carriers include water, alcoholic/aqueous solutions, emulsions or suspensions, including saline and buffered media.
  • Parenteral vehicles include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride, lactated Ringer's or fixed oils.
  • Intravenous vehicles include fluid and nutrient replenishers, electrolyte replenishers such as those based on Ringer's dextrose, and the like. Preservatives and other additives may also be present, such as, for example, antimicrobials, antioxidants, collating agents, inert gases and the like.
  • Controlled or sustained release compositions include formulation in lipophilic depots (e.g. fatty acids, waxes, oils). Also comprehended by the invention are particulate compositions coated with polymers (e.g. poloxamers or poloxamines) and the compound coupled to antibodies directed against tissue-specific receptors, ligands or antigens or coupled to ligands of tissue-specific receptors.
  • lipophilic depots e.g. fatty acids, waxes, oils.
  • particulate compositions coated with polymers e.g. poloxamers or poloxamines
  • compositions of the invention incorporate particulate forms, protective coatings, protease inhibitors or permeation enhancers for various routes of administration, including parenteral, pulmonary, nasal and oral.
  • the pharmaceutical composition can be delivered in a controlled release system.
  • the agent may be administered using intravenous infusion, an implantable osmotic pump, a transdermal patch, liposomes, or other modes of administration.
  • a pump may be used (see for example Saudek et al., JV. Engl. J. Med. (1989), 321:574-579.
  • polymeric materials can be used.
  • a controlled release system can be placed in proximity to the therapeutic target, i.e., the brain, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, Medical Applications of Controlled Release, supra (1984), 2:115-138.
  • a controlled release device is introduced into a subject in proximity to the site of inappropriate immune activation or a tumor.
  • the pharmaceutical preparation may comprise one or more of the compounds of formulas 1-19 alone, or may further include a pharmaceutically acceptable carrier, and can be in solid or liquid form such as tablets, powders, capsules, pellets, solutions, suspensions, elixirs, emulsions, gels, creams, or suppositories, including rectal and urethral suppositories.
  • Pharmaceutically acceptable carriers include gums, starches, sugars, cellulosic materials, and mixtures thereof.
  • the pharmaceutical preparation containing the selective androgen receptor modulator can be administered to a subject by, for example, subcutaneous implantation of a pellet; in a further embodiment, the pellet provides for controlled release of selective androgen receptor modulator over a period of time.
  • the preparation can also be administered by intravenous, intraarterial, or intramuscular injection of a liquid preparation, oral administration of a liquid or solid preparation, or by topical application. Administration can also be accomplished by use of a rectal suppository or a urethral suppository.
  • the pharmaceutical preparations of the invention can be prepared by known dissolving, mixing, granulating, or tablet-forming processes.
  • the selective androgen receptor modulators or their physiologically tolerated derivatives such as salts, esters, N-oxides, and the like are mixed with additives customary for this purpose, such as vehicles, stabilizers, or inert diluents, and converted by customary methods into a suitable form of administration, such as tablets, coated tablets, hard or soft gelatin capsules, aqueous, alcoholic or oily solutions.
  • suitable inert vehicles are conventional tablet bases such as lactose, sucrose, or cornstarch in combination with binders such as acacia, cornstarch, gelatin, or with disintegrating agents such as cornstarch, potato starch, alginic acid, or with a lubricant such as stearic acid or magnesium stearate.
  • suitable oily vehicles or solvents are vegetable or animal oils such as sunflower oil or fish-liver oil. Preparations can be effected both as dry and as wet granules.
  • the compounds of the present invention or their physiologically tolerated derivatives such as salts, hydrates and the like are converted into a solution, suspension, or emulsion, if desired with the substances customary and suitable for this purpose, for example, solubilizers or other auxiliaries.
  • sterile liquids such as water and oils, with or without the addition of a surfactant, and other pharmaceutically acceptable adjuvants.
  • Illustrative oils are those of petroleum, animal, vegetable, or synthetic origin, for example, peanut oil, soybean oil, or mineral oil.
  • water, saline, aqueous dextrose and related sugar solutions, and glycols such as propylene glycols or polyethylene glycols are preferred liquid carriers, particularly for injectable solutions.
  • compositions which contain an active component are well understood in the art.
  • such compositions are prepared as aerosols of the polypeptide delivered to the nasopharynx or as injectables, either as liquid solutions or suspensions, however, solid forms suitable for solution in, or suspension in, liquid prior to injection can also be prepared.
  • the preparation can also be emulsified.
  • the active therapeutic ingredient is often mixed with excipients that are pharmaceutically acceptable and compatible with the active ingredient. Suitable excipients are, for example, water, saline, dextrose, glycerol, ethanol, or the like and combinations thereof.
  • the composition can contain minor amounts of auxiliary substances such as wetting or emulsifying agents, pH buffering agents, which enhance the effectiveness of the active ingredient.
  • An active component can be formulated into the composition as neutralized pharmaceutically acceptable salt forms.
  • Pharmaceutically acceptable salts include the acid addition salts (formed with the free amino groups of the polypeptide or antibody molecule), which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like.
  • Salts formed from the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, 2-ethylamino ethanol, histidine, procaine, and the like.
  • inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, 2-ethylamino ethanol, histidine, procaine, and the like.
  • the compounds of the present invention or their physiologically tolerated derivatives such as salts, hydrates, and the like are prepared and applied as solutions, suspensions, or emulsions in a physiologically acceptable diluent with or without a pharmaceutical carrier.
  • the active compound can be delivered in a vesicle, in particular a liposome (see for example Langer, Science (1990), 249:1527-1533; Treat et al., Liposomes in the Therapy of Infectious Disease and Cancer (1989), Lopez- Berestein and Fidler (eds.), Liss, NY 5 353-365.
  • Anti-Aktl /2(PKB), anti-ERK2, and anti-IGFlR ⁇ antibodies were obtained from Santa Cruz Biotechnology.
  • Anti- phospho(T308)Akt, anti-phospho(Ser636/Ser639)IRSl and anti-PARP antibodies were obtained from Cell Signaling Technology.
  • Dulbecco's modified Eagle's medium (DMEM) and fetal calf serum (FCS) were obtained from Biological Industries, Bet- Haemek, Israel.
  • DMSO was obtained from BDH.
  • TBST 10 mM Tris-HCl, pH 7.5, 50 mM NaCl, and 0.1% Triton X-IOO
  • the receptor was incubated (10 ng/well) in 20 ⁇ M ATP, 10 mM MgCl 2 , 5 mM MnAc 2 , and 20 mM Tris-HCl, pH 7.4, with or without inhibitors, for 20 min at 30 0 C.
  • the plate was then washed with TBS with 0.2% Tween 20 (TBST) and blocked with 0.5% BSA in TBST.
  • Mouse monoclonal anti-phosphotyrosine antibody, conjugated to Horse radish peroxidase (PT-66, 1:50000) was added to the plate. Following incubation for 45 min at room temperature, the plate was washed repeatedly with TBST.
  • Detection was carried out with a color reagent, 2,2'- azido-bis-3-ethylbenzithiazoline-6-sulfonic acid, in citrate-phosphate buffer, pH 4.0, with 0.004% H 2 O 2 for 10 min and monitored at 405 nm, all at room temperature. IC 50 values of inhibitors were calculated using the REGRESSION program. The assay was optimized with respect to the amount of IGF-IR (partially purified from cells overexpressing IGFlR), reaction time, and ATP concentration. The signal was linear for 30 min in the range of IGFlR protein concentrations up to 35 ng/well.
  • IGFl-induced signaling and other signaling pathways e.g. the EGF, PDGF and insulin-induced signaling
  • Tyrosine autophosphorylation of the ⁇ -subunit of IGFlR as well as downstream signaling induced by IGFlR were assayed in breast cancer MCF7 cells.
  • Tyrosine autophosphorylation of the EGFR and the EGFR signaling was assayed in prostate cancer PC3 cells.
  • Tyrosine autophosphorylation of the PDGFR and its signaling was assayed in fibroblasts overexpressing PDGFR.
  • Tyrosine autophosphorylation of the IR and its signaling was assayed in fibroblasts overexpressing IR.
  • Cells were then stimulated for 5 min with 50 ng/ml IGF-I (MCF7 cells), 20 ng/ml EGF (PC3 cells), 50 ng/ml PDGF (fibroblasts overexpressing PDGFR) or 100 nM insulin (fibroblasts overexpressing IR), washed twice with PBS and lysed by boiling sample buffer (10% glycerol, 50 mM Tris-HCl, pH 6.8, 3% SDS, and 5% ⁇ -mercaptoethanol). Equal amounts of protein per lane were separated by 8% SDS-PAGE and transferred to a nitrocellulose membrane (Sartorius AG).
  • Phosphorylated proteins were immunoblotted with anti-phospho-IGFIR (phosphor-IGFIR), anti-phosphotyrosine-IRSl (phospho- IRSl), anti-phospho(T308)Akt (phospo-PKB), anti-phospho-Erk (phospo-ERK), anti- phosphotyrosine 4G10&PY20 (p Y-EGFR or pY-PDGFR), anti-phospho-Ser 636/639 -IRSl (pS 636/639 -IRSl) and anti-phospho-STAT3 ( ⁇ hospho-STAT3) antibodies. Detection was performed with horseradish peroxidase-conjugated secondary antibody using the ECL system.
  • Blots were then stripped of antibodies, blocked with TBST with 5% low fat milk and re-probed with antibodies detecting both the phosphorylated and the non- phosphorylated corresponding proteins e.g. IGF lR ⁇ , IRSl, PKB, ERK, IR ⁇ , PDGFR and Stat3.
  • lysates were prepared from cells exposed to inhibitors at various concentrations for 24 hours in the presence or the absence of FCS without stimulation. Lysate preparation and western blot were the same as described above. Apoptosis was detected by immunoblotting with rabbit anti-PARP antibodies.
  • Various cancer cell lines (listed in table 1) were plated at a density of 1000-5000 cells/well in 96-well plates in 90 ⁇ l growth medium containing 10% FCS, 100 units/ml penicillin and 100 ⁇ g/ml streptomycin. Inhibitors were added a day later in 10 ⁇ l of 1% DMSO in DDW to obtain final concentrations of 0, 0.1, 0.3, 1, 3, and 10 ⁇ M. The final concentration of DMSO (0.1% DMSO) was kept constant in all samples. Medium with inhibitors was refreshed a day and two days later.
  • the cells were fixed in 0.5% gluteraldehyde in medium for 10 min, washed three times with DDW 5 once with 0.1M sodium borate buffer pH 8.5 and stained with 1% methylene blue dissolved in 0.1 M borate buffer solution for 60 min. Excess dye was washed out and cell-bound dye was eluted with 200 ⁇ l/well of
  • Various cancer cells (listed in table 1) were seeded at a very low concentration (19 cells/well in 96-well plates or 63 cells/well in 24-well plates) in growth medium. A day later medium was replaced with growth medium containing various concentrations of inhibitors in a final concentration of 0.1% DMSO. The inhibitor-containing medium was refreshed three times a week. Following approximately two weeks cells were fixed by adding Gluteraldehide (0.5% final concentration) for 10 min, washed three time with DDW, once with borate buffer 0.1 M and stained for 1 hour with 1% Methylene Blue in borate buffer 0.1M. Access stain was washed with water, and following drying colonies were counted.
  • stain was extracted by 0.1N HCl for 1 hour and absorbance at 620 nm was measured by ELISA Reader.
  • the assays were performed in triplicates.
  • the values in table 1 represent IC 50 values derived from the dose-dependent growth curves obtained.
  • mice Human hormone-refractory prostate cancer PC3 cells (ATCC, 1.5x10 6 cells per mouse) were injected subcutaneously into the right leg of Nude:Hsd mice (purchased from Harlan). Ten days later, when palpable tumors had developed, mice were divided into 3 groups with similar average tumor size. The untreated group (UT) did not receive any treatment. The mice treated with compound 7 group were daily injected with compound 7 dissolved in the below described vehicle, and the vehicle-treated group (veh) was daily injected by the vehicle alone. Vehicle included 4.4% DMSO, 1.2% Ethanol and 50% PEG400 in DDW; Groups were composed of 3 mice per group. Dose administered IP was 50 mg/Kg (4 ml/Kg) once a day for one month.
  • ATCC 1.5x10 6 cells per mouse
  • Human ovary cancer A2780 cells (from ECACC, 2x10 6 cells per mouse) were injected subcutaneously into the right leg of female Nude:Hsd mice (purchased from Harlan). Inhibitors were injected IP daily at doses of 20mg/kg (Exp. 2&3) or 50 mg/kg (Exp. 4) dissolved in 4.4%DMSO, 0.12% EtOH, 50% PEG-400 in DDW at volume of 4 ml/kg. The Veh group received 4.4%DMSO, 0.12% EtOH, 50% PEG-400 in DDW at volume of 4 ml/kg. The mouse B16 melanoma cells (1.5xlO 6 cells per mouse) were injected subcutaneously into the right leg of male Nude:Hsd mice (purchased from Harlan). Inhibitors were injected IP daily at a dose of 20mg/kg dissolved in 4.4% % DMSO, 1.2% EtOH, 33% PEG-400 in DDW at volume of 4 ml/kg.
  • the human YUMAC metastatic melanoma cells (2x10 6 cells per mouse), provided by Dr. Ruth Halaban (Yale University), were injected subcutaneously into the right leg of male Nude. ⁇ sd mice (purchased from Harlan). Inhibitors were injected IP daily at a dose of 20mg/kg dissolved in 8% EtOH, 2% Tween-80, 20% PEG-400 and
  • Human pancreatic cancer Panel cells (2xlO 6 cells per mouse) were injected subcutaneously into the right leg of male Nude:Hsd mice (purchased from Harlan).
  • Compound 7 was injected intratumorally (IT) daily at doses of 125 ⁇ g/mouse in 0.06% DMSO and 2.16% PEG400 in saline.
  • Biochemical characterization of the compounds of the present invention Inhibition of IGFlR activity in cell-free assay As shown in table 1 and table 2, compounds 6-18 inhibited the kinase activity of partially purified IGFlR in a dose-dependent manner showing IC50 values of 30- 20OnM. Elimination of the hydroxyl groups increases IC50 values, as demonstrated by the comparison of compound 19 and 7 (table 1). Kinetic studies show that compound 7, as well as compounds 6 and 8 do not compete with ATP, as elevation of the ATP concentrations does not induce an increase in the IC50 values determined in a cellular- free kinase assay of IGF IR (table 2). Table 1: IC50 values of IGFlR activity in cellular free kinase assay
  • FIG. 4A shows that while the control molecule 20 (see structure hereinbelow) had no effect on the signaling, compound 7 dramatically inhibited the autophosphorylation of IGFlR 3 the IGFl -induced tyrosine phosphorylation of IRSl (a direct substrate of IGFlR) and the IGFl -induced activation of two main anti- apoptotic and proliferative pathways downstream IGFlR, the Akt/PKB and the MAPK/ERK pathways.
  • Figure 4B exemplifies a dose-dependent activity of compound 7 in MCF7 cells, showing an IC50 value of 1-2 ⁇ M in inhibition of the central anti-apoptotic pathway downstream IGFlR, the Akt/PKB pathway. Inhibition of the MAPK/ERK pathway activation is detected at even lower concentrations.
  • Compound 7 was unexpectedly found to induce a decrease in the cellular levels of IGFlR-direct substrate IRSl in breast cancer MCF7 cells (Figure 5). This effect was shown to be long-lasting ( Figure 5B). The decrease in IRSl levels was detected even at 24 hours following exposure of the cells to compound 7, accompanied by inhibition of its downstream signaling pathway — the antiapoptotic PKB/Akt pathway and subsequent cleavage of PARP known as a marker for cell apoptosis.
  • the decrease in IRSl levels induced by compound 7 is a result of the induction of a secondary negative regulation, in which IRSl undergoes inhibitory phosphorylation on serine residues and subsequent degradation. This effect is believed to be of high importance in anti-cancer activity stemming from inhibitory mechanisms of PTK signal transduction. It is further contemplated that, Compound 7 as well as other compounds of the present invention which consist of trihydroxy benzyl thioamide and halogen or halomethyl moieties in the second catecholic ring, induce Ser- phosphorylation of IRSl and/or a decrease in IRSl levels.
  • compound 7 inhibits other tyrosine kinases such as the EGFR, PDGFR and IR in intact cells (Figure 9). These kinases have a central role in mitogenesis. Following incubation with compound 7, cells expressing these kinase receptors (e.g. prostate cancer PC3 cells expressing EGFR, fibroblast over-expressing PDGFR and fibroblasts over-expressing IR) were stimulated with either EGF, PDGF or insulin, Figures 9A, 9B and 9C respectively, and lysates were immunoblotted with anti-phosphotyrosine antibodies to detect autophosphorylation of the receptors. Downstream signaling of these receptors was measured as well.
  • tyrosine kinases such as the EGFR, PDGFR and IR in intact cells ( Figure 9).
  • Ser/Thr-kinases like MEK or PDK are not inhibited by compound 7 as detected by the phosphorylation levels of their substrates (ERK and PKB(Thr308) respectively) in cells where compound 7 had no inhibitory effect on upstream regulators.
  • Table 3 The inhibitory activity (IC 5 0 values in ⁇ M) of compounds 6,7,8 and 9 in cell proliferation and in clonogenic assays of various cancer cell lines. Assays performed in triplicates.
  • the sensitivity of the human metastatic melanoma cells was tested against a wider set of compounds (table 4), and showed high sensitivity towards the molecules that induce IRSl Ser-phosphorylation, e.g. compounds 7, 8, 9, 10 and 13 (figures 6 & 8) as opposed to compounds 6, 17 and 19.
  • Table 4 Inhibition of human metastatic melanoma cells by a series of novel molecules. Results are presented in IC 50 values following 72 hours incubation of the cells with the molecules.
  • Compound 7 was tested for its inhibitory effect on tumor growth in animal model. Nude mice were subcutaneously injected with cancer cells in the flank of the mouse, and when tumors were measurable, administration of various compounds of the present invention started. Tumor dimensions presented in the graphs are those measured following the first administration. The model of human hormone-refractory prostate cancer (HRPC) PC3 was the first to be examined. Figure 10 shows that systemic administration (IP) of compound 7 at dosage of 50 mg/kg once a day, resulted in inhibition of the tumor growth by 82% as compared to controls (vehicle treated and untreated). Furthermore, no significant effect on body weight of the nude mice was detected following one month of administration (Fig. 11). Additionally, Compound 7 was found efficient when tested using intratumoral administration on both HRPC and pancreatic PANCl models (figure 16).
  • IP systemic administration
  • FIG. 19A shows that compound 6 inhibited the autophosphorylation of IGFlR, the IGFl -induced tyrosine phosphorylation of IRSl (a direct substrate of IGFlR), and the IGFl-induced activation of the Akt/PKB. As opposed to compound 7, compound 6 has no effect on either the IGFlR or the IRSl levels ( Figure 19B).
  • Compound 6 inhibited also EGFR and EGF-induced tyrosine phosphorylation of STAT3, a direct substrate of EGFR, and the EGF-induced activation of the PKB pathway ( Figures 20A&20B).

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Diabetes (AREA)
  • Hematology (AREA)
  • Neurology (AREA)
  • Oncology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Obesity (AREA)
  • Urology & Nephrology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Dermatology (AREA)
  • Rheumatology (AREA)
  • Cardiology (AREA)
  • Vascular Medicine (AREA)
  • Ophthalmology & Optometry (AREA)
  • Neurosurgery (AREA)
  • Pain & Pain Management (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Emergency Medicine (AREA)
  • Biomedical Technology (AREA)
  • Endocrinology (AREA)
  • Immunology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Enzymes And Modification Thereof (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)

Abstract

The present invention provides new tyrphostin derivatives acting as substrate competitive protein tyrosine kinase (PTK) inhibitors and receptor tyrosine kinase (RTK) inhibitors, methods of their preparation, pharmaceutical compositions including such compounds, and methods of using these compounds and compositions, especially as chemotherapeutic agents for preventions and treatments of PTK and RTK related disorders such as metabolic, fibrotic, and cell proliferative disorders, in particular psoriasis and cancer.

Description

NOVEL PROTEIN KINASE MODULATORS AND THERAPEUTIC USES THEREOF
FIELD OF THE INVENTION
The present invention relates to novel tyrphostin derivatives, their preparation, pharmaceutical compositions comprising same, and their use in treatment of protein kinase related disorders.
BACKGROUND OF THE INVENTION
Protein tyrosine kinases (PTKs) are a family of enzymes, which transfer the γ- phosphate of ATP to the side chain of tyrosine residues on substrate proteins. PTKs are involved in a variety of cellular processes, including signal transduction and growth regulation. Phosphorylation of substrates by PTKs are key events in cellular signaling.
One class of PTKs are receptor tyrosine kinases (RTKs). These kinases belong to a family of transmembrane proteins and have been implicated in cellular signaling pathways. The predominant biological activity of some receptor kinases is the stimulation of cell growth and proliferation, while other receptor tyrosine kinases are involved in inhibiting growth and promoting differentiation. In some instances, a single tyrosine kinase can inhibit, or stimulate, cell proliferation depending on the cellular environment in which it is expressed (Schlessinger and Ullrich, Neuron (1992), 9(3): 383-391). RTKs include receptors for platelet-derived growth factor (PDGF), fibroblast growth factor (FGF), hepatocyte growth factor (HGF), insulin, insulin-like growth factor 1 (IGF-I), nerve growth factor (NGF), vascular endothelial growth factor (VEGF), macrophage colony stimulating factor (M-CSF) and others.
Receptor tyrosine kinases are composed of at least three domains: an extracellular glycosylated ligand binding domain, a transmembrane domain and a cytoplasmic catalytic domain that can phosphorylate tyrosine residues. Binding of a ligand to membrane-bound receptors induces the formation of receptor dimers and allosteric changes thus activating the intercellular kinase domains which further results self-phosphorylation (autophosphorylation and/or transphosphorylation) of the receptor on tyrosine residues. Receptor phosphorylation stimulates physical association of the activated receptor with target molecules. Some of the target molecules are, in turn, phosphorylated, a process which transmits the signal to the cytoplasm. The secondary signal transducer molecules generated by activated receptors, result in a signal cascade that regulates cell functions such as cell division or differentiation. Intracellular signal transduction is reviewed in Aaronson, Science (1991), 254: 1146-1153; Schlessinger, J Trends Biochem. ScI (1988), 13: 443-447; Ullrich and Schlessinger, Cell (1990), 61: 203-212.
Various cell proliferative disorders have been associated with defects in pathways mediated by PTKs. Enhanced activities of PTKs resulting from overexpression of the normal kinase, upregulation of ligands of receptor tyrosine kinases or activating mutations, are a hallmark of many diseases which involve cellular proliferation, including cancer. Examples of specific receptor tyrosine kinases associated with cell proliferative disorders include platelet derived growth factor receptor (PDGFR), insulin-like growth factor 1 receptor (IGF-IR), epidermal growth factor receptor (EDFR), and the related HER2.
The involvement of PTKs in various diseases identifies them as targets for antiproliferative drugs. Indeed, numerous PTK blockers have been described in the literature including proposed mechanisms of action (Levitzki et al., Science (1995), 267:1782-88; Posner et al., MoI. Pharmacol. (1994), 45:673-683). Applicants have developed a family of PTK inhibitors, named tyrphostins, designed to mimic the tyrosine substrate (Levitzki et al., Science (1995), 267:1782-88; Levitzki et al., Biochem. Pharm. (1990), 40:913-920; Levitzki et al., FASEB J. (1992), 6:3275-3282; US Pat. Nos. 5,217,999 and 5,773,476). The pharmacophores of these tyrphostins, and in particular tyrphostins of the benzylidene malonitril type, are the hydrophilic catechol ring and the more lipophilic substituted cyano- vinyl radical. Kinetic studies have shown that some tyrphostin compounds are pure competitive inhibitors vis-a-vis tyrosine substrates whereas for the ATP binding site they act as non-competitive inhibitors (Yaish et al., Science (1988), 242:933-935; Gazit et al., J Med. Chem. (1989), 32:2344- 2352). Nonetheless, many tyrphostins have shown competitive inhibition against both the substrate and ATP binding site (Posner et al., MoI. Pharmacol. (1994), 45:673-683). In a related group of tyrphostins, the hydrophilic catechol ring was exchanged by lipophilic dichloro- or dimethoxy-phenyl groups, to yield EGFR kinase inhibitors, effective in the low micromolar range. (Yoneda et al., Cancer Res. (1991), 51: 4430-
4435). However, there is an unmet need for tyrphostins with increased inhibitory properties.
SUMMARY OF THE INVENTION
The present invention relates to new tyrphostins compounds useful as inhibitors of protein tyrosine kinases (PTKs) in cells. These novel tyrphostin compounds show increased inhibitory properties of, but not limited to, insulin-like growth factor 1 receptor (IGFlR), platelet derived growth factor receptor (PDGFR), epidermal growth factor receptor (EGFR), and IGFlR-related insulin receptor (IR). The present invention is further directed to compounds that trigger Serine phosphorylation of the IGFlR direct substrate IRSl, thus providing long-lasting effects which enhance the inhibitory activity of these novel tyrphostins.
According to one aspect, the present invention provides compounds represented by the structure of formula 1 :
Figure imgf000004_0001
wherein
R1, R2, R5 and R6 are independently selected from H, C1-C4 alkyl, acyl and a functional group that gives rise to hydroxyl upon hydrolysis;
R3 and R7 are independently selected from H, halogen, haloalkyl and OR8 wherein R is H, C1-C4 alkyl, acyl or a functional group that gives rise to hydroxyl upon hydrolysis; R4 is H or CN5 including salts, hydrates, solvates, polymorphs, optical isomers, geometrical isomers, enantiomers, diastereomers, and mixtures thereof. In one embodiment, the present invention provides a compound represented by the structure of formula 1, wherein R3 is halogen (e.g., F, Cl, Br or I) or haloalkyl (e.g. CF3). In another embodiment, the present invention provides a compound represented by the structure of formula 1, wherein R1, R2, R5 and R6 are each H. In another embodiment, the present invention provides a compound represented by the structure of formula 1, wherein R7 is hydrogen, halogen or OR8 with R8 being H or methyl. In one embodiment, the present invention further provides compounds of formula 1, wherein R4 is H. In another embodiment, the present invention provides compounds of formula 1, wherein R4 is CN.
Representative and non-limiting examples of such structures are compounds selected from the group consisting of compounds 2-18:
Figure imgf000005_0001
Figure imgf000005_0002
Figure imgf000006_0001
Figure imgf000006_0002
Figure imgf000007_0001
Figure imgf000007_0002
Figure imgf000007_0003
Figure imgf000007_0004
Figure imgf000008_0001
Figure imgf000008_0002
Figure imgf000008_0003
15
Figure imgf000009_0001
16
Figure imgf000009_0002
17; and
Figure imgf000009_0003
18
Another similar derivative is compound no. 19:
Figure imgf000010_0001
19
The present invention further provides pharmaceutical compositions comprising any of the compounds represented by the structure of formula 1.
Figure imgf000010_0002
1 wherein
R1, R2, R5 and R6 are independently selected from H, Ci-C4 alkyl, acyl and a functional group that gives rise to hydroxyl upon hydrolysis;
R3 and R7 are independently selected from H, halogen, haloalkyl and OR8 wherein R8 is H, C1-C4 alkyl, acyl or a functional group that gives rise to hydroxyl upon hydrolysis;
R4 is H or CN, including salts, hydrates, solvates, polymorphs, optical isomers, geometrical isomers, enantiomers, diastereomers, and mixtures thereof. The present invention further provides a method of inhibiting a protein tyrosine kinase (PTK) comprising contacting the PTK with an effective inhibitory amount of a compound of formula 1. The present invention further provides a method of inhibiting a protein tyrosine kinase (PTK) in a subject comprising the step of administering to the subject a therapeutically effective amount of a compound of formula 1, to inhibit a protein tyrosine kinase (PTK) in the subject, hi another embodiment, the method comprises administering to the subject a pharmaceutical composition comprising a therapeutically effective amount of any of the compound of formula 1, and a pharmaceutically acceptable excipient.
The present invention further provides a method of treating or preventing a protein tyrosine kinase (PTK) related disorder in a subject comprising the step of administering to the subject a therapeutically effective amount of any of the compounds of formula 1. hi another embodiment, the method comprises administering to the subject a pharmaceutical composition comprising a therapeutically effective amount of a compound of formula 1 , and a pharmaceutically acceptable excipient. hi one embodiment, the PTK related disorder is a cell proliferative disorder, a metabolic disorder or a fibrotic disorder. In a preferred embodiment, the PTK related disorder is cancer, hi another embodiment the PTK related disorder is diabetic nephropathy.
Further embodiments and the full scope of applicability of the present invention will become apparent from the detailed description given hereinafter. However, it should be understood that the detailed description and specific examples, while indicating preferred embodiments of the invention, are given by way of illustration only, since various changes and modifications within the spirit and scope of the invention will become apparent to those skilled in the art from this detailed description.
BRIEF DESCRIPTION OF THE DRAWINGS
FIG.l Shows in schematic form a process for the synthesis of exemplary novel Tyrphostins of the invention. X and Y are independently selected from hydrogen, halogen, haloalkyl and OR8 wherein R8 is H, C1-C4 alkyl, acyl or a functional group that gives rise to hydroxyl upon hydrolysis.
FIG.2 Shows in schematic form a process for the synthesis of exemplary novel Tyrphostins of the invention. X and Y are independently selected from hydrogen, halogen, haloalkyl and OR8 wherein R8 is H, C1-C4 alkyl, acyl or a functional group that gives rise to hydroxy 1 upon hydrolysis.
FIG.3 Shows in schematic form a process for the synthesis of intermediates used in the synthesis of the novel Tyrphostins of the invention.
FIGS.4A and 4B show that compound 7 induces inhibition of IGFlR and its signaling in intact cancer cells. Figure 4 A shows the inhibition of IGFlR activation and signaling in cancer cells following exposure to compound 7 in comparison to the control (compound # 20). Compound 7 induces a decrease in the tyrosine-phosphorylation of IRSl, a direct substrate of IGFlR, and inhibition of IGFl-induced activation of the PKB and ERK pathways. Figure 4B represents the dose-dependent inhibition of IGFl- induced signaling in cancer cells.
FIGS. 5 A 5B and 5C show that compound 7 induces Serine-phosphorylation on IRSl and a decrease in the cellular levels of IRSl in breast cancer MCF7 cells. Figure 5 A shows a decrease in cellular levels of IRSl induced by 4 hours treatment with compound 7. Figure 5B shows induced Serine-phosphorylation on IRSl following 4 hours treatment with compound 7. Figure 5 C shows a long-lasting (24 hours following exposure) reduction in IRSl levels in cancer cells.
FIG. 6 shows Ser-phosphorylation on IRSl following treatment of breast cancer MCF7 cells with compounds 7, 8, 9, 10, 11, and 13. Inhibition of the activation of IGFlR downstream signaling pathways, namely ERK and PKB are demonstrated as well.
FIGS. 7A and 7B show treatment of ovarian cancer A2780 cells with compounds 7, 8 and 9 in comparison to compound 6. Figure 7A shows that cells treated with compounds 6, 7, 8 and 9 undergo apoptosis following incubation (cleaved PARP detection) with compound 6 being slightly weaker. Figure 7B shows Ser- phosphorylation on IRSl (detected by the shift of the IRSl band upward) as well as a decrease in IRSl levels induced by compounds 7, 8 and 9 in contrast to compound 6. FIG. 8 shows that Ser-phosphorylation on IRSl as well as cleavage of PARP (apoptosis) are detected in metastatic melanoma YUMAC cells treated with compounds 7, 8, 9, 10, and 13 in comparison to compounds 11 and 19.
FIGS. 9A, 9B and 9C show that compound 7 inhibits the activation and downstream signaling of PDGFR, EGFR and IR tyrosine kinases in intact cells. Figure 9A shows that compound 7 inhibits EGFR activation in hormone-refractory prostate cancer PC3 cells, and EGF-induced phosphorylation of PKB and STAT3, a direct substrate of EGFR. Figure 9B shows that compound 7 inhibits ligand-induced PDGFR activation and downstream signaling in NIH3T3 cells over expressing PDGFR. Figure 9C shows that compound 7 inhibits ligand-induced Insulin receptor (IR) activation and downstream regulation in NIH3T3 cells over expressing IR.
FIG. 10 shows that treatment of nude mice with compound 7 (IP, I/day, 50 mg/kg) resulted in 82% growth inhibition of hormone-refractory prostate cancer (HRPC) PC3 tumors.
FIG. 11 shows that treatment of nude mice with compound 7 (IP, I/day, 50 mg/kg) had no significant effect on body weight of the mice over 4 weeks (UT=untreated, Veh=vehicle).
FIG. 12 shows that treatment of nude mice with compounds 7 or 8 (IP, I/day, 20 mg/kg) resulted in 87% growth inhibition of ovarian cancer A2780 tumors.
FIG. 13 shows the high efficacy of compounds 7, 8, and 9 as compared to compound 6 in growth inhibition of ovarian cancer A2780 tumors in nude mice model.
FIG. 14 shows that treatment of nude mice, bearing ovarian A2780 tumors larger than 50mm3, with compound 8 (IP, I/day, 50 mg/kg) resulted in 82% growth inhibition of the tumors. FIG. 15 shows that treatment of nude mice, bearing ovarian A2780 tumors larger than 470mm3, with compound 8 (IP, I/day, 50 mg/kg) resulted in approximately 85% growth inhibition of the tumors.
FIG. 16 shows that treatment of nude mice with compound 7 (intratumoral, I/day, 125 μg/day) resulted in 50% growth inhibition of pancreatic cancer Panel tumors.
FIG. 17 shows the efficacy of compound 7 as compared to compound 6 in growth inhibition of melanoma B16 tumors in nude mice model. The inhibitors were administered IP at dose of 20 mg/kg, I/day.
FIG. 18 shows the efficacy of compound 8 in growth inhibition of human melanoma YUMAC tumors in nude mice model. Compound 8 was administered IP at dose of 20 mg/kg, I/day.
FIGS. 19A and 19B show the effect of compound 6 on MCF7 cells. Figure 19A shows that compound 6 inhibits IGFl -induced IGFlR activation and signaling (auto- phosphorylation of IGFlR5 the IGFl -induced tyrosine phosphorylation of IRSl and the IGFl -induced activation of the Akt/PKB). Figure 19B shows that compound 6 has no effect on the IRS 1 levels.
FIGS. 2OA and 2OB show the effect of compound 6 on EGFR activity in HRPC PC3 cells. Figure 2OA shows that compound 6 inhibits EGF-induced phosphorylation of EGFR and its downstream elements, STAT3 and PKB. Figure 2OB shows that protein levels are equal and no effect on STAT3, PBCB or ERK was detected following treatment of PC3 cells with compound 6.
FIG. 21 shows a dose-dependent inhibition of primary keratinocytes growth by compound 7 (IC50=2.3μM) in the presence of growth factor enriched medium. DETAILED DESCRIPTION OF THE INVENTION
The present invention relates to novel tyrphostin derivatives which are potent PTK-inhibitors. The present invention further provides methods for inhibiting PTKs5 for example IGF-I receptor (IGFlR). The compounds are useful in treating or preventing PTK-related disease states, particularly PTK-related disorders which are associated with defects in signaling pathways mediated by PTKs. Examples of diseases involving cellular proliferation are cancer and psoriasis. The compounds of the present invention are designed to have an enhanced inhibiting potency with respect to protein tyrosine kinases (PTKs), compared with previously disclosed tyrphostin derivatives (Blum et al., Biochem. (2000), 39:15705- 15712; US Pat. Nos. 5,773,476 and 5,217,999). The applicants have surprisingly found that the introduction of additional substituents on the catechol pharmacophore greatly enhances the inhibitory potency. Furthermore, introduction of additional substituents on the second aromatic ring was also found to significantly enhance the inhibiting potency of the new compounds. In addition, it is shown that a subfamily of the described molecules triggers Serine phosphorylation of IRSl, which serves as the direct substrate of IGF-IR. Without wishing to be bound by any theory or mechanism of action, this phosphorylation may lead to decoupling of IRSl with IGF-IR, thereby blocking IGF-IR signaling. Furthermore, this Ser-phosphorylation of IRSl is usually accompanied by a decrease in IRSl levels. These processes are shown to enhance the inhibitory potential of these tyrosine kinase inhibitors, thus introducing long-lasting effects.
The compounds provided by the present invention are represented by the general formula 1 :
Figure imgf000015_0001
wherein R1, R2, R5 and R6 are independently selected from H, C1-C4 alkyl, acyl and a functional group that gives rise to hydroxyl upon hydrolysis;
R3 and R7 are independently selected from H, halogen, haloalkyl and OR8 wherein R8 is H, C1-C4 alkyl, acyl or a functional group that gives rise to hydroxyl upon hydrolysis;
R4 is H or CN, including salts, hydrates, solvates, polymorphs, optical isomers, geometrical isomers, enantiomers, diastereomers, and mixtures thereof.
In one embodiment, the present invention provides a compound represented by the structure of formula 1, wherein R1, R2, R5 and R6 each independently H or methyl. In another embodiment, the present invention provides a compound represented by the structure of formula 1, wherein R3 is halogen (e.g. F, Cl, Br, I), hydroxyl or haloalkyl (e.g. CF3). In another embodiment, the present invention provides a compound represented by the structure of formula 1, wherein R4 is CN. In another embodiment, the present invention provides a compound represented by the structure of formula 1, wherein R is H, halogen or OR with R being H or methyl. In one embodiment, the present invention further provides a compound of formula 1, wherein R4 is H. Representative and non-limiting examples of such structures are compounds selected from the group consisting of compounds 2-18:
Figure imgf000016_0001
Figure imgf000017_0001
Figure imgf000017_0002
Figure imgf000018_0001
Figure imgf000019_0001
11
Figure imgf000019_0002
12
10
Figure imgf000019_0003
Figure imgf000020_0001
Figure imgf000020_0002
Another similar derivative is compound No. 19:
Figure imgf000021_0001
19
Without wishing to be bound by any theory or mechanism of action, when the hydroxyls are blocked by a methyl group, namely the compounds consist of methoxy groups in positions 1, 2, 5 & 6, the activity of the compounds is dramatically decreased in comparison to compounds having hydroxy groups at the equivalent positions. Compounds 2-5 are therefore less active in comparison to compounds 6-19 and the like.
Chemical Definitions
An "alkyl" group refers to a saturated aliphatic hydrocarbon, including straight-chain, branched-chain and cyclic alkyl groups. In one embodiment, the alkyl group has 1-12 carbons designated here as CrCπ-alkyl. In another embodiment, the alkyl group has 1-6 carbons designated here as Ci-Cβ-alkyl. In another embodiment, the alkyl group has 1-4 carbons designated here as C1-C4- alkyl. The alkyl group may be unsubstituted or substituted by one or more groups selected from halogen, hydroxy, alkoxy carbonyl, amido, alkylamido, dialkylamido, nitro, amino, alkylamino, dialkylamino, carboxyl, thio and thioalkyl.
A "hydroxy" group refers to an OH group. An "alkoxy" group refers to an -O-alkyl group wherein R is alkyl as defined above. A "thio" group refers to an - SH group. An "alkylthio" group refers to an -SR group wherein R is alkyl as defined above.
An "amino" group refers to an NH2 group. An alkylamino group refers to an -NHR group wherein R is alkyl is as defined above. A dialkylamino group refers to an -NRR' group wherein R and R' are alkyl as defined above.
An "amido" group refers to a -C(O)NH2 group. An alkylamido group refers to an -C(O)NHR group wherein R is alkyl is as defined above. A dialkylamido group refers to an -C(O)NRR' group wherein R and R' are alkyl as defined above.
A "thioamide" group refers to a C(S)NHR, where R is either alkyl, aryl, alkylaryl or hydrogen.
The term "halogen" or "halo" as used herein alone or as part of another group refers to chlorine, bromine, fluorine, and iodine. The term "haloalkyl" refers to an alkyl group having some or all of the hydrogens independently replaced by a halogen group including, but not limited to, trichloromethyl, tribromomethyl, trifluoromethyl, triiodomethyl, difluoromethyl, chlorodifluoromethyl, pentafluoroethyl, 1,1-difluoroethyl bromomethyl, chloromethyl, fluoromethyl, iodomethyl, and the like.
Examples of functional groups that give rise to hydroxyl upon hydrolysis include, but are not limited to, esters, anhydrides, carbamates, carbonates and the like. For example, when any of R1, R2, R5 or R6 is an acyl group (COR), the resulting function group is an ester (OCOR). When any of R1, R2, R5 or R6 is an amide group (CONHR), the resulting function group is a carbamate (OCONHR). When any of R1, R2, R5 or R6 is a carboxylate group (COOR), the resulting function group is a carbonate (OCOOR). All stereoisomers of the compounds of the present invention are contemplated, either in admixture or in pure or substantially pure form. These compounds can have asymmetric centers at any of the atoms. Consequently, the compounds can exist in enantiomeric or diastereomeric forms or in mixtures thereof. The present invention contemplates the use of any racemates (i.e. mixtures containing equal amounts of each enantiomers), enantiomerically enriched mixtures (i.e., mixtures enriched for one enantiomer), pure enantiomers or diastereomers, or any mixtures thereof. The chiral centers can be designated as R or S or R5S or d,D, 1,L or d,l, D,L. Compounds comprising amino acid residues include residues of D-amino acids, L-amino acids, or racemic derivatives of amino acids. In addition, several of the compounds of the present invention contain one or more double bonds. The present invention intends to encompass all structural and geometrical isomers including cis, trans, E and Z isomers, independently at each occurrence.
One or more of the compounds of the invention, may be present as a salt. The term "salt" encompasses both basic and acid addition salts, including but not limited to carboxylate salts or salts with amine nitrogens, and include salts formed with the organic and inorganic anions and cations discussed below. Further encompassed by the term are salts formed by standard acid-base reactions with basic groups (such as amino groups) and organic or inorganic acids. Such acids include hydrochloric, hydrofluoric, trifluoroacetic, sulfuric, phosphoric, acetic, succinic, citric, lactic, maleic, fumaric, palmitic, cholic, pamoic, mucic, D-glutamic, D-camphoric, glutaric, phthalic, tartaric, lauric, stearic, salicyclic, methanesulfonic, benzenesulfonic, sorbic, picric, benzoic, cinnamic, and the like.
The term "organic or inorganic cation" refers to counter-ions for the carboxylate anion of a carboxylate salt. The counter-ions are chosen from the alkali and alkaline earth metals (such as lithium, sodium, potassium, barium, aluminum and calcium); ammonium and mono-, di- and tri-alkyl amines such as trimethylamine, cyclohexylamine; and the organic cations, such as dibenzylammonium, benzylammonium, 2-hydroxyethylarnmonium, bis(2-hydroxyethyl)ammonium, phenylethylbenzylammonium, dibenzylethylenediammonium, and like cations. See, for example, Berge et al, J Pharm. ScL (1977), 66:1-19, which is incorporated herein by reference. Other cations encompassed by the above term include the protonated form of procaine, quinine and N-methylgmcosamine, and the protonated forms of basic amino acids such as glycine, ornithine, histidine, phenylglycine, lysine and arginine. Furthermore, any zwitterionic form of the instant compounds formed by a carboxylic acid and an amino group are also contemplated.
The present invention also includes solvates of compounds of formula 1 or any of compounds 2-19 and salts thereof. "Solvate" means a physical association of a compound of the invention with one or more solvent molecules. This physical association involves varying degrees of ionic and covalent bonding, including hydrogen bonding. In certain instances the solvate will be capable of isolation. "Solvate" encompasses both solution-phase and isolatable solvates. Non-limiting examples of suitable solvates include ethanolates, methanolates and the like. "Hydrate" is a solvate wherein the solvent molecule is water. The present invention also includes polymorphs of compounds of formula 1 or any of compounds 2-19 and salts thereof. The term "polymorph" refers to a particular crystalline state of a substance, which can be characterized by particular physical properties such as X-ray diffraction, IR spectra, melting point, and the like.
Therapeutic Use
The present invention provides compounds and compositions effective at inhibiting protein tyrosine kinases. These compounds and compositions are potentially useful in the treatment of a diseases associated with altered or abnormal activity of protein tyrosine kinases such as enhanced activity of protein tyrosine kinases. Thus, in one embodiment, the present invention provides a method of inhibiting a protein tyrosine kinase (PTK) comprising contacting the PTK with an effective inhibitory amount of a compound represented by any of formulas 1-19.
The present invention further provides a method of treating or preventing a protein tyrosine kinase (PTK) in a subject comprising the step of administering to the subject a therapeutically effective amount of any of the compounds represented by formulas 1-19. In another embodiment, the method comprises administering to the subject a pharmaceutical composition comprising a therapeutically effective amount of any of the compounds represented by formulas 1-19 and a pharmaceutically acceptable excipient.
The present invention further provides a method of inhibiting a protein tyrosine kinase (PTK) related disorder in a subject comprising the step of administering to the subject a therapeutically effective amount of any of the compounds represented by formulas 1-19. In another embodiment, the method comprises administering to the subject a pharmaceutical composition comprising a therapeutically effective amount of any of the compounds represented by formulas 1-19; and a pharmaceutically acceptable excipient. A "protein tyrosine kinase" (PTK) is a protein belonging to a family of enzymes that transfer the γ-phosphate of ATP to the side chain of tyrosine residues on substrate proteins. PTKs are involved in a variety of key cellular processes, including signal transduction and growth regulation. A protein tyrosine kinase, as used herein, refers to a receptor tyrosine kinase (RTK) as well as a cellular tyrosine kinase (CTK or non-receptor tyrosine kinase). Thus the compounds of the present invention are effective at inhibiting both receptor and non-receptor protein tyrosine kinases.
A cellular tyrosine kinase (CTK or non-receptor tyrosine kinase) is an intracellular protein which takes part in signal transduction within the cell, including signal transduction to the nucleus. Examples of CTKs are the Src family of oncoproteins. A receptor tyrosine kinase (RTK) is a transmembrane protein that participates in transmembrane signaling pathways. The predominant biological activity of some receptor tyrosine kinases is the stimulation of cell growth and proliferation, while other receptor tyrosine kinases are involved in arresting growth and promoting differentiation. RTKs include the receptors for platelet-derived growth factor (PDGF), fibroblast growth factor (FGF), hepatocyte growth factor (HGF), insulin, insulin-like growth factor- 1 (IGF-I), nerve growth factor (NGF), vascular endothelial growth factor (VEGF), and macrophage colony stimulating factor (M-CSF).
The term "protein tyrosine kinase related disorder" as used herein refers to a disorder characterized by abnormal or altered PTK activity. Abnormal or altered activity further refers to either (i) overexpression of PTK in cells that do not normally express PTKs; (ii) increased PTK expression leading to unwanted cell proliferation, differentiation and/or growth; or, (iii) decreased PTK expression leading to unwanted reductions in cell proliferation, differentiation and/or growth. Over-activity of PTKs refers to either amplification of the gene encoding a particular PTK or production of a level of PTK activity which can correlate with cell proliferation, differentiation and/or growth. Over- activity can also be the result of ligand independent or constitutive activation as a result of mutations such as deletions of a fragment of a PTK responsible for ligand binding.
Thus, in one embodiment, the present invention is directed to preparations containing any of the compounds represented by formulas 1-19, which modulate PTK activity signal transduction by affecting the enzymatic activity of the protein tyrosine kinases thereby interfering with the signal transduction pathways mediated by such proteins.
Examples of protein tyrosine kinase related disorders are cell proliferative disorders, metabolic disorders or fibrotic disorders and inflammation. Examples of cell proliferative disorders which are mediated by protein tyrosine kinases are cancer, psoriasis, diabetic nephropathy, blood vessel proliferative disorders, and mesangia cell proliferative disorders.
Cancer is a disorder in which a population of cells has become, in varying degrees, unresponsive to the control mechanisms that normally govern proliferation and differentiation. Cancer refers to various types of malignant neoplasms and tumors, including metastasis to different sites. Nonlimiting examples of cancers which can be treated by any of the compounds represented by formulas 1-19 are brain, ovarian, colon, prostate, kidney, bladder, breast, lung, oral and skin cancers which exhibit altered activity of PTK. Specific examples of cancers which the compounds of the present invention are effective at treating or preventing are: adenocarcinoma, adrenal gland tumor, ameloblastoma, anaplastic tumor, anaplastic carcinoma of the thyroid cell, angiofibroma, angioma, angiosarcoma, apudoma, argentaffmoma, arrhenoblastoma, ascites tumor cell, ascitic tumor, astroblastoma, astrocytoma, ataxia-telangiectasia , atrial myxoma, basal cell carcinoma, benign tumor, bone cancer, bone tumor, brainstem glioma, brain tumor, breast cancer, Burkitt's lymphoma, carcinoma, cerebellar astrocytoma, cervical cancer, cherry angioma, cholangiocarcinoma, a cholangioma, chondroblastoma, chondroma, chondrosarcoma, chorioblastoma, choriocarcinoma, colon cancer, common acute lymphoblastic leukaemia, craniopharyngioma, cystocarcinoma, cystofibroma, cystoma, cytoma, ductal carcinoma in situ, ductal papilloma, dysgerminoma, encephaloma, endometrial carcinoma, endothelioma, ependymoma, epithelioma, erythroleukaemia, Ewing's sarcoma, extra nodal lymphoma, feline sarcoma, fibroadenoma, fibrosarcoma, follicular cancer of the thyroid, ganglioglioma, gastrinoma, glioblastoma multiforme, glioma, gonadoblastoma, haemangioblastoma, haemangioendothelioblastoma, haemangioendothelioma, haemangiopericytoma, haematolymphangioma, haemocytoblastoma, haemocytoma, hairy cell leukaemia, hamartoma, hepatocarcinoma, hepatocellular carcinoma, hepatoma, histoma, Hodgkin's disease, hypernephroma, infiltrating cancer, infiltrating ductal cell carcinoma, insulinoma, juvenile angiofibroma, Kaposi sarcoma, kidney tumour, large cell lymphoma, leukemia, chronic leukemia, acute leukemia, lipoma, liver cancer, liver metastases, Lucke carcinoma, lymphadenoma, lymphangioma, lymphocytic leukaemia, lymphocytic lymphoma, lymphocytoma, lymphoedema, lymphoma, lung cancer, malignant mesothelioma, malignant teratoma, mastocytoma, medulloblastoma, melanoma, meningioma, mesothelioma, metastatic cancer, Morton's neuroma, multiple myeloma, myeloblastoma, myeloid leukemia, myelolipoma, myeloma, myoblastoma, myxoma, nasopharyngeal carcinoma, nephroblastoma, neuroblastoma, neurofibroma, neurofibromatosis, neuroglioma, neuroma, non-Hodgkin's lymphoma, oligodendroglioma, optic glioma, osteochondroma, osteogenic sarcoma, osteosarcoma, ovarian cancer, Paget's disease of the nipple, pancoast tumor, pancreatic cancer, phaeochromocytoma, pheochromocytoma, plasmacytoma, primary brain tumor, progonoma, prolactinoma, renal cell carcinoma, retinoblastoma, rhabdomyosarcoma, rhabdosarcoma, solid tumor, sarcoma, secondary tumor, seminoma, skin cancer, small cell carcinoma, squamous cell carcinoma, strawberry haemangioma, T- cell lymphoma, teratoma, testicular cancer, thymoma, trophoblastic tumor, rumourigenic, vestibular schwannoma, Wilm's tumor, or a combination thereof.
Blood vessel proliferative disorders refer to antiogenic and vasculogenic disorders generally resulting in abnormal proliferation of blood vessels. The formation and spreading of blood vessels, or vasculogenesis and angiogenesis, respectively, play important roles in a variety of physiological processes such as embryonic development, corpus luteum formation, wound healing and organ regeneration, as well as a pivotal role in cancer development. Other examples of blood vessel proliferation disorders include arthritis and ocular diseases such as diabetic retinopathy, restenosis, retinopathies and atherosclerosis. Mesangial cell proliferative disorders refer to disorders brought about by abnormal proliferation of mesangial cells. Mesangial proliferative disorders include various human renal diseases such as glomerulonephritis, diabetic nephropathy, malignant nephrosclerosis, thrombic microangiopathy syndromes, transplant rejection and glomerulopathies, hi this regards, PDGFR has been implicated in the maintenance of mesangial cell proliferation.
Metabolic disorders that are implicated with abnormal PTK activity include psoriasis, diabetes mellitus, wound healing, inflammation and neurodegenerative diseases. For example, EGFR has been indicated in corneal and dermal wound healing. Defects in the Insulin-R and IGF-IR receptor are indicated in type-II diabetes mellitus. Fibrotic disorders refer to the abnormal formation of extracellular matrices.
Examples of fibrotic disorders include hepatic cirrhosis and mesangial cell proliferative disorders. Hepatic cirrhosis is characterized by the increase in extracellular matrix constituents resulting in the formation of a hepatic scar. The term "treating" as used herein refers to abrogating, inhibiting, slowing or reversing the progression of a disease, ameliorating clinical symptoms of a disease or preventing the appearance of clinical symptoms of a disease. The term "preventing" is defined herein as barring a subject from acquiring a disorder or diseases. The term "administering" as used herein refers to a method of bringing a compound of the present invention and a target protein tyrosine kinase together in such a manner that the tyrphostin can affect the catalytic activity of the tyrosine kinase directly; i.e. by interacting with the kinase itself, or indirectly; i.e. by interacting with another molecule on which the catalytic activity of the enzyme is dependent. As used herein, administration can be accomplished in vitro, i.e. in a test tube, or in vivo, i.e. in cells or tissues of living organisms, for example humans. In one embodiment, the present invention encompasses administering the compounds of the present invention to a subject.
The term "contacting" as used herein refers to bringing into contact the protein tyrosine kinase and the compounds defined herein, under in vivo conditions or in vitro conditions as defined above.
The term "therapeutically effective amount" refers to the amount of a compound being administered which relieves to some extent one or more of the symptoms of the disorder being treated. Therapeutic effective doses for any compounds represented by formulas 1-19 described herein can be estimated initially from cell culture and/or an animal model. A dose can be formulated in an animal model, and this dose can be used to more precisely determine useful doses in humans.
The term "effective inhibitory amount" refers to the amount of a compound being administered that inhibits to some extent the protein tyrosine kinase with which it is contacted.
Pharmaceutical Compositions:
The present invention further provides pharmaceutical compositions comprising any of the compounds represented by the structure of formulas 1-19, and a pharmaceutically acceptable carrier or excipient. As used herein, "pharmaceutical composition" means therapeutically effective amounts of the compounds of the present invention, together with suitable diluents, preservatives, solubilizers, emulsifiers, adjuvant and/or carriers. A "therapeutically effective amount" as used herein refers to that amount which provides a therapeutic effect for a given condition and administration regimen. Such compositions are liquids or Lyophilized or otherwise dried formulations and include diluents of various buffer content (e.g., Tris-HCL, acetate, phosphate), pH and ionic strength, additives such as albumin or gelatin to prevent absorption to surfaces, detergents (e.g., Tween 20, Tween 80, Pluronic F68, bile acid salts), solubilizing agents (e.g., glycerol, polyethylene glycerol), antioxidants (e.g., ascorbic acid, sodium metabisulfite), preservatives (e.g., Thimerosal, benzyl alcohol, parabens), bulking substances or tonicity modifiers (e.g., lactose, mannitol), covalent attachment of polymers such as polyethylene glycol to the protein, complexation with metal ions, or incorporation of the material into or onto particulate preparations of polymeric compounds such as polylactic acid, polglycolic acid, hydrogels, etc, or onto liposomes, microemulsions, micelles, unilamellar or multilamellar vesicles, erythrocyte ghosts, or spheroplasts. Such compositions will influence the physical state, solubility, stability, rate of in vivo release, and rate of in vivo clearance. Controlled or sustained release compositions include formulation in lipophilic depots (e.g., fatty acids, waxes, oils).
Further comprehended by the invention are particulate compositions coated with polymers (e.g., poloxamers or poloxamines). Other embodiments of the compositions of the invention incorporate particulate forms, protective coatings, protease inhibitors or permeation enhancers for various routes of administration, including parenteral, pulmonary, nasal and oral. In one embodiment the pharmaceutical composition is administered parenterally, paracancerally, transmucosally, transdermally, intramuscularly, intravenously, intradermally, subcutaneously, intraperitonealy, intraventricularly, intracranially or intratumorally.
Moreover, as used herein "pharmaceutically acceptable carriers" are well known to those skilled in the art and include, but are not limited to, 0.01-O.lM and preferably 0.05M phosphate buffer or 0.8% saline. Additionally, such pharmaceutically acceptable carriers may be aqueous or non-aqueous solutions, suspensions, and emulsions. Examples of nonaqueous solvents are propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable organic esters such as ethyl oleate. Aqueous carriers include water, alcoholic/aqueous solutions, emulsions or suspensions, including saline and buffered media.
Parenteral vehicles include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride, lactated Ringer's or fixed oils. Intravenous vehicles include fluid and nutrient replenishers, electrolyte replenishers such as those based on Ringer's dextrose, and the like. Preservatives and other additives may also be present, such as, for example, antimicrobials, antioxidants, collating agents, inert gases and the like.
Controlled or sustained release compositions include formulation in lipophilic depots (e.g. fatty acids, waxes, oils). Also comprehended by the invention are particulate compositions coated with polymers (e.g. poloxamers or poloxamines) and the compound coupled to antibodies directed against tissue-specific receptors, ligands or antigens or coupled to ligands of tissue-specific receptors.
Other embodiments of the compositions of the invention incorporate particulate forms, protective coatings, protease inhibitors or permeation enhancers for various routes of administration, including parenteral, pulmonary, nasal and oral.
Compounds modified by the covalent attachment of water-soluble polymers such as polyethylene glycol, copolymers of polyethylene glycol and polypropylene glycol, carboxymethyl cellulose, dextran, polyvinyl alcohol, polyvinylpyrrolidone or polyproline are known to exhibit substantially longer half-lives in blood following intravenous injection than do the corresponding unmodified compounds. Such modifications may also increase the compound's solubility in aqueous solution, eliminate aggregation, enhance the physical and chemical stability of the compound, and greatly reduce the immunogenicity and reactivity of the compound. As a result, the desired in vivo biological activity may be achieved by the administration of such polymer-compound abducts less frequently or in lower doses than with the unmodified compound.
In yet another embodiment, the pharmaceutical composition can be delivered in a controlled release system. For example, the agent may be administered using intravenous infusion, an implantable osmotic pump, a transdermal patch, liposomes, or other modes of administration. In one embodiment, a pump may be used (see for example Saudek et al., JV. Engl. J. Med. (1989), 321:574-579. hi another embodiment, polymeric materials can be used. In yet another embodiment, a controlled release system can be placed in proximity to the therapeutic target, i.e., the brain, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, Medical Applications of Controlled Release, supra (1984), 2:115-138. Preferably, a controlled release device is introduced into a subject in proximity to the site of inappropriate immune activation or a tumor. Other controlled release systems are discussed in the review by Langzτ, Science (1990), 249: 1527-1533). The pharmaceutical preparation may comprise one or more of the compounds of formulas 1-19 alone, or may further include a pharmaceutically acceptable carrier, and can be in solid or liquid form such as tablets, powders, capsules, pellets, solutions, suspensions, elixirs, emulsions, gels, creams, or suppositories, including rectal and urethral suppositories. Pharmaceutically acceptable carriers include gums, starches, sugars, cellulosic materials, and mixtures thereof. The pharmaceutical preparation containing the selective androgen receptor modulator can be administered to a subject by, for example, subcutaneous implantation of a pellet; in a further embodiment, the pellet provides for controlled release of selective androgen receptor modulator over a period of time. The preparation can also be administered by intravenous, intraarterial, or intramuscular injection of a liquid preparation, oral administration of a liquid or solid preparation, or by topical application. Administration can also be accomplished by use of a rectal suppository or a urethral suppository.
The pharmaceutical preparations of the invention can be prepared by known dissolving, mixing, granulating, or tablet-forming processes. For oral administration, the selective androgen receptor modulators or their physiologically tolerated derivatives such as salts, esters, N-oxides, and the like are mixed with additives customary for this purpose, such as vehicles, stabilizers, or inert diluents, and converted by customary methods into a suitable form of administration, such as tablets, coated tablets, hard or soft gelatin capsules, aqueous, alcoholic or oily solutions. Examples of suitable inert vehicles are conventional tablet bases such as lactose, sucrose, or cornstarch in combination with binders such as acacia, cornstarch, gelatin, or with disintegrating agents such as cornstarch, potato starch, alginic acid, or with a lubricant such as stearic acid or magnesium stearate. Examples of suitable oily vehicles or solvents are vegetable or animal oils such as sunflower oil or fish-liver oil. Preparations can be effected both as dry and as wet granules. For parenteral administration (subcutaneous, intravenous, intraarterial, or intramuscular injection), the compounds of the present invention or their physiologically tolerated derivatives such as salts, hydrates and the like are converted into a solution, suspension, or emulsion, if desired with the substances customary and suitable for this purpose, for example, solubilizers or other auxiliaries. Examples are sterile liquids such as water and oils, with or without the addition of a surfactant, and other pharmaceutically acceptable adjuvants. Illustrative oils are those of petroleum, animal, vegetable, or synthetic origin, for example, peanut oil, soybean oil, or mineral oil. In general, water, saline, aqueous dextrose and related sugar solutions, and glycols such as propylene glycols or polyethylene glycols are preferred liquid carriers, particularly for injectable solutions.
The preparation of pharmaceutical compositions which contain an active component is well understood in the art. Typically, such compositions are prepared as aerosols of the polypeptide delivered to the nasopharynx or as injectables, either as liquid solutions or suspensions, however, solid forms suitable for solution in, or suspension in, liquid prior to injection can also be prepared. The preparation can also be emulsified. The active therapeutic ingredient is often mixed with excipients that are pharmaceutically acceptable and compatible with the active ingredient. Suitable excipients are, for example, water, saline, dextrose, glycerol, ethanol, or the like and combinations thereof. hi addition, if desired, the composition can contain minor amounts of auxiliary substances such as wetting or emulsifying agents, pH buffering agents, which enhance the effectiveness of the active ingredient. An active component can be formulated into the composition as neutralized pharmaceutically acceptable salt forms. Pharmaceutically acceptable salts include the acid addition salts (formed with the free amino groups of the polypeptide or antibody molecule), which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed from the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, 2-ethylamino ethanol, histidine, procaine, and the like.
For topical administration to body surfaces using, for example, creams, gels, drops, and the like, the compounds of the present invention or their physiologically tolerated derivatives such as salts, hydrates, and the like are prepared and applied as solutions, suspensions, or emulsions in a physiologically acceptable diluent with or without a pharmaceutical carrier. hi another embodiment, the active compound can be delivered in a vesicle, in particular a liposome (see for example Langer, Science (1990), 249:1527-1533; Treat et al., Liposomes in the Therapy of Infectious Disease and Cancer (1989), Lopez- Berestein and Fidler (eds.), Liss, NY5 353-365. The following examples are presented in order to more folly illustrate certain embodiments of the invention. They should in no way, however, be construed as limiting the broad scope of the invention. One skilled in the art can readily devise many variations and modifications of the principles disclosed herein without departing from the scope of the invention.
Example 1: Synthesis
The general procedure for the synthesis of compounds 6-8, 11-13, and 15-17 is drawn schematically in figure 1, and disclosed hereinbelow:
General procedure for the synthesis of the following intermediate compounds: denoted (i) wherein Y=H, (ii) wherein Y=OMe and (iii) wherein Y=Br:
Figure imgf000033_0001
An amine (1.2 equiv) and methyl cyanoacetate (1 equiv) were stirred at room temperature until the precipitation of the product was observed. The product was collected by filtration, washed twice with ethanol, and dried under reduced pressure.
The product was obtained as a white solid in 70 - 80 % yield. For compound (i): 1H NMR (300 MHz, in CDCl3): δ 6.85 (m, 3H), 6.3 (bs,
IH), 4.41 (d, J= 6.0 Hz, 2H), 3.89 (s, 3H), 3.87 (s, 3H), 3.40 (s, 2H). MS (ESI): found
(m/z) 235.67; calculated [please confirm] for C12H14N2O3 (MH+) 235.25.
For compound (ii): 1H NMR (300 MHz, in CDCl3): δ 6.49 (s, 2H), 6.37 (bs,
IH)5 4.40 (d, J= 4.4 Hz, 2H), 3.86 (s, 6H), 3.84 (s, 3H), 3.43 (s, 2H). MS (ESI): found (m/z) 265.60; calculated for C13H17N2O4 (MH+) 265.11.
For compound (iii): 1H NMR (300 MHz, in CDCl3): δ 7.04 (s, IH), 6.79 (s,
IH), 4.39 (d, J= 6.0 Hz, 2H)5 3.89 (s, 3H)5 3.87 (s, 3H)5 3.45 (s, 2H). General procedure for the synthesis of the following intermediate compounds denoted (iv) wherein Y=H, (v) wherein Y=OMe and (vi) wherein Y=Br:
Figure imgf000034_0001
An amide (1 equiv) and Lawesson's reagent (0.55 equiv) were heated in dry toluene (ca. 2 mL/mmol of compounds (i-iii)) under reflux for 3 hours (until TLC indicated the disappearance of the amide). The reaction mixture was cooled and evaporated under reduced pressure. The residue was purified by flash chromatography to yield a pale yellow solid in 50 - 60 % yield.
For compound (iv): 1H NMR (400 MHz, in CDCl3+ Acetone-d6): δ 9.20 (bs, IH), 6.84 (m, 2H)5 6.78 (d, J= 8 Hz, IH) 4.71 (d, J= 5.1 Hz, 2H)5 3.89 (s, 2H)5 3.81 (s, 3H), 3.80 (s, 3H). MS (CI): found (m/z) 251.43; calculated for C12H14N2O2S (MH+) 250.32.
For compound (v): 1H NMR (300 MHz5 in Acetone-d6): δ 9.20 (bs, IH)5 6.72 (S5 2H)5 4.77 (d, J= 5.2 Hz5 2H)5 4.06 (s, 2H), 3.80 (s, 6H)5 3.71 (s, 3H). MS (CI): found (m/z) 281.51 ; calculated for C13H17N2O3S (MH+) 281.34.
For compound (vi): 1H NMR (400 MHz5 in CDCl3): δ 7.08 (s, IH)5 6.82 (s, IH)5 4.78 (d5 J= 5.2 Hz5 2H)5 3.98 (s, 2H)5 3.88 (s, 3H)5 3.87 (s, 3H).
General procedure for the synthesis of the following intermediate compounds denoted (vii) wherein X=Br and Y=H, (viii) wherein X=I and Y=H, (ix) wherein X=F and Y=OMe, (x) wherein X=Cl and Y=OMe, (xi) wherein X=Br and Y=OMe, (xii) wherein X=I and Y=OMe, (xiii) wherein X=CF3 and Y=OMe, (xiv) wherein X=Br and Y=Br, and (xv) wherein X=OMe and Y=Br:
Figure imgf000034_0002
A catalytic amount of ^-alanine (0.2 equiv) was added to a solution of β- cyanothioamide (1 equiv) and an aldehyde ((1.2 equiv) commercially available except for 3,4-dimethoxy-5-(trifluoromethyl)benzaldehyde which was prepared according to Backstrom et al, J Med. Chem. (1989), 32:841-846) in ethanol (ca. 20 mL/mmol of compounds (iv-vi)). The solution was heated to 600C for 0.5 hour to overnight. The product was precipitated, collected by filtration, washed with H2O, EtOH, and ether and then dried under reduced pressure to yield a pure yellow solid in 70 % to quantitative yield.
For compound (vii): 1H NMR (400 MHz, in CDCl3): δ 8.69 (s, IH), 7.95 (bt, IH), 7.70 (d, J= 2.0 Hz, IH), 7.67 (d, J= 2.0 Hz, IH), 6.91 (m, 3H), 4.94 (d, J = 5.0 Hz, 2H), 3.98 (s, 3H), 3.90 (s, 6H). For compound (viii): 1H NMR (400 MHz, in CDCl3): δ 8.67 (s, IH), 7.95 (bt,
IH), 7.86 (d, J= 2.0 Hz, IH), 7.72 (d, J = 2.0 Hz, IH), 6.92 (m, 3H), 4.94 (d, J= 5.2 Hz, 2H), 3.98 (s, 3H), 3.90 (s, 6H).
For compound (ix): 1H NMR (400 MHz, in Acetone-d6): δ 9.60 (bs, IH), 8.24 (s, IH), 7.55 (m, 3H), 6.81 (s, IH), 4.98 (s, 2H), 3.96 (s, 3H), 3.83 (s, 6H), 3.73 (s, 3H). For compound (x): 1H NMR (400 MHz, in CDCl3): δ 8.69 (s, IH), 7.99 (bt,
IH), 7.58 (d, J= 2.0 Hz, IH), 7.55 (d, J= 2.0 Hz, IH), 6.60 (s, IH), 4.92 (d, J= 5.2 Hz, 2H), 3.96 (s, 3H), 3.87 (s, 6H), 3.84 (s, 3H).
For compound (xi): 1H NMR (300 MHz, in Acetone-d6): δ 9.62 (bt, IH), 8.21 (s, IH), 7.81 (s, IH), 7.76 (s, IH), 6.79 (s, 2H), 4.96 (m, 2H), 3.94 (s, 3H), 3.81 (s, 6H), 3.71 (s, 3H). MS (CI): found (m/z) 494.73; calculated for C21H22BrN2O5S (MH+) 494.37.
For compound (xii): 1H NMR (400 MHz, in CDCl3): δ 8.66 (s, IH), 7.99 (bt, IH), 7.86 (d, J= 2.0 Hz, IH), 7.72 (d, J= 2.0 Hz, IH), 6.60 (s, 2H), 4.93 (d, J= 5.0 Hz, 2H), 3.97 (s, 3H), 3.88 (s, 6H), 3.86 (s, 3H). MS (CI): found (m/z) 540.67; calculated for C21H21IN2O5S (M+) 540.37.
For compound (xiii): 1H NMR (200 MHz, in CDCl3): 8.75 (s, IH), δ 8.22 (bt, IH), 7.91 (d, J= 2.0 Hz, IH), 7.69 (d, J= 2.0 Hz, IH), 6.61 (s, 2H), 4.94 (d, J= 5.0 Hz, 2H), 4.03 (s, 3H), 3.88 (s, 6H), 3.86 (s, 3H).
For compound (xiv): 1H NMR (400 MHz, in Acetone-d6+CDCl3): δ 9.30 (bt, IH), 8.43 (s, IH), 7.72 (d, J= 2.0 Hz, IH), 7.17 (d, J= 2.0 Hz, IH), 7.07 (d, J= 2.0 Hz, IH), 5.01 (d, J= 4.4 Hz, 2H), 3.95 (s, 3H), 3.87 (s, 3H), 3.81 (s, 3H). For compound (xv): 1H NMR (400 MHz, in CDCl3): δ 8.74 (s, IH), 8.00 (bt, IH), 7.34 (s, 2H)5 7.14 (s, IH), 6.90 (s, IH), 4.96 (d, J= 4.5 Hz, 2H), 3.96 (s, 6H), 3.88 (s, 3H), 3.86 (s, 3H).
General procedure for the synthesis of compounds 6-8, 11-13, 15-17:
Boron tribromide (1.5 equiv excess for each hydroxyl group) was added to a cold solution of the protected product in anhydrous CH2Cl2 (ca. 20 mL/mmol of compounds (vii-xv)). The reaction mixture was allowed to warm to room temperature and stirred for 2- 4 hours (until HPLC indicated the formation of the desired deprotected product). The solution was cooled and then treated with dilute hydrochloric acid. The solution was extracted three times with ethyl acetate, the organic layer was dried over Na2SO4, filtered and the solvent was evaporated. The crude product was recrystallized from water/ethanol to give yellow solid in 60-70% yield.
For compound 6: 1H NMR (400 MHz, in Acetone-d6): δ 9.45 (bs, IH), 8.13 (s, IH), 7.69 (d, J= 2.1 Hz, IH), 7.66 (d, J= 2.1 Hz, IH), 7.11 (d, J= 2.0 Hz, IH), 6.97 (d, J= 2.0 Hz, IH), 4.90 (d, J= 5.4 Hz, 2H). MS (ESI): found (m/z) 498.73; calculated for C17H13Br2N2O4S (MH+) 498.88.
For compound 7: 1H NMR (300 MHz, in Acetone-d6): 9.48 (bs, IH), 8.08 (s, IH), 7.66 (d, J= 2.0 Hz, IH), 7.63 (d, J= 2.0 Hz, IH), 6.46 (s, 2H), 4.79 (d, J= 5.7 Hz, 2H). MS (ESI): found (m/z) 438.40; calculated for C17H14BrN2O5S (MH+) 438.26.
For compound 8: 1H NMR (200 MHz, in Acetone-d6): δ 9.42 (bs, IH), 8.24 (s, IH), 7.91 (d, J= 2.1 Hz, IH), 7.64 (d, J= 2.1 Hz, IH), 6.47 (s, 2H), 4.79 (d, J= 5.5 Hz, 2H). MS (ESI): found (m/z) 484.80; calculated for C17H14IN2O5S (M+) 484.96.
For compound 11: 1H NMR (300 MHz, in Acetone-d6): δ 9.42 (bs, IH), 8.08 (s, IH), 7.61 (d, J= 2.0 Hz, IH), 7.49 (d, J= 2.0 Hz, IH), 6.47 (s, 2H), 4.80 (d, J- 4.6 Hz, 2H). MS (ESI): found (m/z) 393.07; calculated for C17H14ClN2O5S (MH+) 393.81.
For compound 12: 1H NMR (300 MHz, in Acetone-d6): δ 9.47 (bs, IH), 8.10 (s, IH), 7.88 (d, J= 2.0 Hz, IH), 7.45 (d, J= 2.0 Hz5 IH), 6.47 (s, 2H), 4.81 (d, J= 5.4 Hz5 2H). For compound 13: 1H NMR (300 MHz, in Acetone-d6): δ 9.47 (bs, IH), 8.06
(s, IH), 7.80 (d, J= 2.1 Hz5 IH)5 7.71 (d, H = 2.1 Hz, IH), 6.48 (s, 2H), 4.79 (d, J= 5.4 Hz, 2H). MS (ESI): found (m/z) 427.33; calculated for C18Hi4F3N2O5S (MH+) 427.37. For compound 15: 1H NMR (400 MHz, in Acetone-d6): δ 9.40 (bs, IH)5 8.08 (s, IH), 7.82 (d, J = 2.0 Hz, IH), 7.72 (d, J= 2.0 Hz, IH), 6.93 (m, IH), 6.79 (m, 2H), 4.86 (d, J = 5.4 Hz, 2H). MS (ESI): found (m/z) 422.80; calculated for C17H14BrN2O4S (MH+) 422.27. For compound 16: 1H NMR (300 MHz, in Acetone-d6): δ 9.40 (bs, IH), 8.13
(s, IH), 7.82 (d, J= 2.0 Hz, IH), 7.68 (d, J = 2.0 Hz, IH), 6.93 (s, IH), 6.78 (s, 2H), 4.86 (d, J = 5.7 Hz, 2H). MS (ESI): found (m/z) 468.67; calculated for C17H13IN2O4S (M+) 468.27.
For compound 17: 1H NMR (400 MHz, in Acetone-d6): δ 9.45 (bs, IH), 8.13 (s, IH), 7.15 (s, 2H), 7.08 (d, J= 2.0 Hz, IH), 6.94 (d, H = 2.0 Hz, IH), 4.90 (d, J= 5.0 Hz, 2H). MS (ESI): found (m/z) 437.67; calculated for C17H13BrN2O5S (M+) 437.26.
The general procedure for the synthesis of compounds 5, 9-10,14 and 18 is drawn schematically in figure 2, and disclosed hereinbelow:
General procedure for the synthesis of the following intermediate compounds denoted (xvi) wherein X=Br, (xvii) wherein X=I, and (xviii) wherein X=CFs:
Figure imgf000037_0001
A catalytic amount of piperidine (0.2 equiv) was added to a solution of aldehyde
((I equiv) commercially available except for 3,4-dimethoxy-5-(trifluoromethyl) benzaldehyde which was prepared according to Backstrom et al., J. Med. Chem. (1989), 32:841-846) and malonic acid (1.5 equiv) in pyridine. The reaction mixture was heated to 1200C for 6 h. The solution was cooled to room temperature and concentrated HCl was added dropwise to pH <3. The white solid was collected by filtration, washed with water and dried under reduced pressure.
For compound (xvi): 1H NMR (300 MHz, in CDCl3): δ 7.65 (d, J= 15.9 Hz), 7.35 (d, J = 2.1 Hz, IH), 7.01 (d, J = 2.1 Hz, IH), 6.35 (d, J = 15.9 Hz, IH), 3.90 (s, 3H), 3.88 (s, 3H). For compound (xvii): 1H NMR (400 MHz, in CDCl3): δ 7.64 (d, J = 2.0 Hz, IH), 7.56 (d, J= 2.0 Hz), 7.04 (d, J= 2.0 Hz, IH), 6.35 (d, J= 16.0 Hz, IH), 3.92 (s, 3H), 3.88 (s, 3H).
For compound (xviii): 1H NMR (400 MHz, in CDCl3): δ 7.63 (d, J = 16 Hz), 7.61 (s, IH), 7.43 (s, IH), 6.50 (d, J= 16 Hz, IH), 3.90 (s, 3H), 3.88 (s, 3H).
General procedure for the synthesis of the following intermediate compounds denoted (xix) wherein X=Br and Y=OMe, (xx) wherein X=I and Y=OMe, (xxi) wherein X=CF3 and Y=OMe and (xxii) wherein X=Br and Y=Br:
Figure imgf000038_0001
The solution of compounds (xvi-xviii, 1 equiv) in oxalyl chloride (4 equiv) was stirred for 1-2 hours at room temperature. The excess of oxalyl chloride was distilled off and the mixture was evaporated to dryness. The residue was dissolved in CH2Cl2 and added drop wise to a solution of an amine (0.85 equiv) and Et3N (4 equiv) in CH2Cl2. the reaction mixture was stirred at room temperature for 0.5-1 hour (until TLC indicated the disappearance of the amine). The solvent was evaporated under reduced pressure and the residual oil was purified by flash chromatography.
For compound (xix): 1H NMR (300 MHz, in CDCl3): δ 7.52 (d, J= 15.8 Hz, IH), 7.29 (s, IH), 6.92 (s, IH), 6.50 (s, 2H), 6.37 (d, J = 15.8 Hz, IH), 6.23 (bt, IH), 4.46 (d, J= 5.7 Hz, 2H), 3.81-3.85 (s, 15H). MS (ESI): found (m/z) 467.87; calculated for C21H25BrNO6 (MH+) 466.32.
For compound (xx): 1H NMR (400 MHz, in CDCl3): δ 7.51 (d, J= 2.0 Hz, IH), 7.50 (d, J= 15.6 Hz), 6.96 (d, J= 2.0 Hz, IH), 6.51 (s, 2H), 6.35 (d, J= 15.6 Hz, IH), 6.10 (bt, J = 5.2 Hz, IH), 4.47 (d, J = 5.2 Hz, 2H), 3.85 (s, 3H), 3.84 (s, 3H) 3.82 (s, 6H), 3.81 (s, 3H).
For compound (xxi): 1H NMR (300 MHz, in CDCl3): δ 7.52 (d, J= 15.8 Hz, IH), 7.29 (s, IH), 6.92 (s, IH), 6.50 (s, 2H), 6.37 (d, J = 15.8 Hz, IH), 6.23 (bt, IH), 4.46 (d, J= 5.7 Hz, 2H), 3.81-3.85 (s, 15H). MS (ESI): found (m/z) 467.87; calculated for C21H25BrNO6 (MH+) 466.32. For compound (xxii): 1H NMR (400 MHz, in CDCl3 + Acetone-d6): 8.56 (bt, J = 6.0 Hz ,1H), 7.39 (d, J= 2.0 Hz, IH), δ 7.38 (d, J= 15.6 Hz, IH)3 7.28 (d, J= 2.0 Hz, IH), 7.09 (d, J= 2.0 Hz, IH), 7.05 (d, J= 2.0 Hz, IH), 6.68 (d, J= 15.6 Hz, IH), 4.32 (d, J = 6.0 Hz, 2H), 3.85 (s, 3H), 3.80 (s, 3H), 3.74 (s, 3H), 3.67 (s, 3H). MS (ESI): found (m/z) 467.87; calculated for C21H25BrNO6 (MH+) 466.32.
General procedure for the synthesis of the following compounds denoted 5 wherein X=Br and Y=OMe, (xxiii) wherein X=I and Y=OMe, (xxiv) wherein X=CF3 and Y=OMe, and (xxv) wherein X=Br and Y=Br:
Figure imgf000039_0001
An amide (1 equiv) and Lawesson's reagent (0.55 equiv) were refluxed in toluene for 3 hours (until TLC indicated the disappearance of the amide). The reaction mixture was cooled and evaporated under reduced pressure. The residue was purified by flash chromatography to yield a pale yellow solid in 50 - 60 % yield.
For compound 5: 1H NMR (300 MHz, in CDCl3): δ 7.75 (d, J= 15.3 Hz, IH), 7.41 (d, J = 2.1 Hz, IH), 7.29 (d, J= 2.1 Hz, IH), 7.16 (d, J = 15.3 Hz, IH), 6.76 (s, 2H), 4.90 (m, 2H), 3.94 (s, 3H), 3.83 (s, 3H), 3.77 (s, 6H), 3.70 (s, 3H). MS (ESI): found (m/z) 483.87; calculated for C21H25BrNO5S (MH+) 483.38. For compound (xxiii): 1H NMR (400 MHz, in CDCl3): δ 7.71 (d, J= 15.2 Hz,
IH), 7.6 (bt, IH), 7.56 (d, J= 1.8 Hz, IH), 7.01 (d, J= 1.8 Hz, IH), 6.78 (d, J= 15.2 Hz, IH), 6.55 (s, 2H), 4.86 (d, J= 5.0 Hz, 2H), 3.86 (s, 3H), 3.84 (s, 3H), 3.83 (s, 6H), 3.82 (s, 3H).
For compound (xxiv): 1H NMR (300 MHz, in CDCl3): δ 7.75 (d, J= 15.3 Hz, IH), 7.41 (d, J= 2.1 Hz, IH), 7.29 (d, J= 2.1 Hz, IH), 7.16 (d, J= 15.3 Hz, IH), 6.76 (s, 2H), 4.90 (m, 2H), 3.94 (s, 3H), 3.83 (s, 3H), 3.77 (s, 6H), 3.70 (s, 3H). MS (ESI): found (m/z) 483.87; calculated for C21H25BrNO5S (MH+) 483.38.
For compound (xxv): 1H NMR (400 MHz, in CDCl3): δ 7.74 (d, J= 15.2 Hz, IH), 7.52 (bt, 1H),7.37 (d, J= 1.9 Hz, IH), 7.15 (d, J= 1.9 Hz, IH), 7.01 (d, J= 1.9 Hz, IH), 6.89 (d, J= 1.9 Hz, IH), 6.78 (d, J= 15.2 Hz, IH), 4.92 (d, J= 5.3, 2H), 3.91 (s, 3H), 3.90 (s, 3H), 3.88 (s, 3H), 3.87 (s, 3H).
General procedure for the synthesis of compounds 9-10, 14 and 18: Boron tribromide (1.5 equiv excess for each hydroxyl group) was added to a cold solution of the protected product in anhydrous CH2Cl2 (ca. 20 mL/mmol of compounds 5 and (xxiii-xxv)). The reaction mixture was allowed to warm to room temperature and stirred for 2- 4 hours (until HPLC indicated the formation of the desired deprotected product). The solution was cooled and then treated with dilute hydrochloric acid. The solution was extracted three times with ethyl acetate and the combined organic layer was dried over Na2SO4 and evaporated under reduced pressure. The crude product was recrystallized from water/ethanol to give the desired product in 60-70% yield.
For compound 9: 1H NMR (400 MHz, in Acetone-d6): δ 9.16 (bs, IH), 7.69 (d, J = 15.4 Hz, IH), 7.31 (d, J= 1.9 Hz, IH), 7.09 (d, J= 1.9 Hz, IH), 7.06 (d, J = 15.4 Hz, IH), 6.44 (s, 2H), 4.76 (d, J = 5.7 Hz, 2H). MS (ESI): found (m/z) 411.93; calculated for C16H15BrNO5S (MH+) 411.97.
For compound 10: 1H NMR (400 MHz, in Acetone-d6): δ 9.2 (bs, IH), 7.67 (d, J= 15.2 Hz, IH), 7.51 (d, J= 2.0 Hz, IH), 7.12 (d, J= 2.0 Hz, IH), 7.02 (d, J= 15.2 Hz, IH), 6.44 (s, 2H), 4.76 (d, J = 5.2 Hz, 2H). MS (ESI): found (m/z) 460.13; calculated for C16H15INO5S (MH+) 460.26.
For compound 14: 1H NMR (400 MHz, in Acetone-d6): δ 9.16 (bd, IH), 7.69 (d, J= 15.4 Hz, IH), 7.31 (d, J= 1.9 Hz, IH), 7.09 (d, J= 1.9 Hz, IH), 7.06 (d, J= 15.4 Hz, IH), 6.44 (s, 2H), 4.76 (d, J = 5.7 Hz, 2H). MS (ESI): found (m/z) 411.93; calculated for C16H15BrNO5S (MH+) 411.97.
For compound 18: 1H NMR (300 MHz, in Acetone-d6): δ 9.23 (bt, IH), 7.68 (d, J= 15.2 Hz, IH), 7.31 (d, J= 2.0 Hz, IH), 7.09 (d, J= 2.0 Hz, IH), 7.09 (d, J= 2.0 Hz, IH), 6.99 (d, J= 15.2 Hz, IH), 6.91 (d, J= 2.0 Hz, IH), 4.85 (d, J= 5.6 Hz, 2H). MS (ESI): found (m/z) 476.27; calculated for C16H14Br2NO4S (MH+) 476.15. The general procedure for the synthesis of intermediates used in the synthesis of the novel tyrphostins of the present invention is shown in figure 3 and described hereinbelow: General procedure for the synthesis of the following compound denoted herein (xxvi):
Figure imgf000041_0001
5-Bromoveratraldehyde (5.00 g, 20.5 mmol, 1 equiv) was dissolved in minimum amount of warm ethanol, and a solution of hydroxylamine hydrochloride (1.71 g, 24.6 mmol, 1.2 equiv) in water (30 mL) was added. Then aqueous solution of 10 % sodium hydroxide (1.09 g, 27.3 mmol, 1.33 equiv) was added and the mixture was stirred at room temperature (until TLC indicated the disappearance of the aldehyde). After evaporation of the ethanol, the product was precipitated, collected by filtration, washed with water and dried under reduced pressure to yield a pure white solid in quantitative yield. 1U NMR (300 MHz, in CDCl3): δ 8.01 (s, IH), 7.71 (s, IH), 7.27 (d, J= 1.8 Hz, IH), 7.15 (OL9 J= 1.8 Hz, IH), 3.89 (s, 3H), 3.88 (s, 3H).
General procedure for the synthesis of the following compound denoted herein (xxvii):
Figure imgf000041_0002
To a solution of (xxvi) (5.00 g, 19.2 mmol, 1 equiv) in 20 mL of acetic acid was added zinc (3.77 gr, 57.6 mmol, 3 equiv). The solution was refluxed until TLC showed the disappearance of the oxime. The zinc salts were filtered and washed with ethyl acetate. The filtrate was evaporated and aqueous sodium hydroxide was added. The aqueous layer was extracted three times with ethyl acetate. The organic layer was washed with brine, dried over Na2SO4 and evaporated under reduced pressure to yield yellowish oil in 55 % yield. 1H NMR (300 MHz, in CDCl3): δ 7.09 (d, J= 2.1 Hz, IH), 6.85 (d, J= 2.1 Hz, IH)5 3.90 (s, 3H), 3.86 (s, 3H), 3.82 (s, 2H).
Example 2: Biological Activity
Reagents and Antibodies
All chemicals used for chemical synthesis, namely bovine serum albumin, poly(Glu,Tyr) 4:1 (pGT), 2,2'-azido-bis-3-ethylbenzithiazoline-6-sulfonic acid, IGFl, methylene blue, 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide (MTT), HRP-conjugated anti-phosphotyrosine PT-66 and diphosphorylated mitogen- activated protein kinase antibodies (pERK) were purchased from Sigma. Anti-phospho- IRSl antibody was obtained from Oncogene Research Products, Germany; anti-IRSl was obtained from Upstate Biotechnology, Inc. Anti-Aktl /2(PKB), anti-ERK2, and anti-IGFlRβ antibodies were obtained from Santa Cruz Biotechnology. Anti- phospho(T308)Akt, anti-phospho(Ser636/Ser639)IRSl and anti-PARP antibodies were obtained from Cell Signaling Technology. Dulbecco's modified Eagle's medium (DMEM) and fetal calf serum (FCS) were obtained from Biological Industries, Bet- Haemek, Israel. DMSO was obtained from BDH.
Inhibition of IGFlR-catalyzed Substrate Phosphorylation
The general protein-tyrosine kinase substrate, poly(Glu,Tyr) 4:1 (pGT), was coated onto a 96-well Maxisorp plates (Nunc) by adding 125 μl of 0.1 mg/ml pGT in PBS to each well. Plates were sealed and incubated for 16 hours at 370C, washed once with TBST (10 mM Tris-HCl, pH 7.5, 50 mM NaCl, and 0.1% Triton X-IOO) and once with DDW, dried for 2-3 hours, and stored at 40C. The receptor was incubated (10 ng/well) in 20 μM ATP, 10 mM MgCl2, 5 mM MnAc2, and 20 mM Tris-HCl, pH 7.4, with or without inhibitors, for 20 min at 300C. The plate was then washed with TBS with 0.2% Tween 20 (TBST) and blocked with 0.5% BSA in TBST. Mouse monoclonal anti-phosphotyrosine antibody, conjugated to Horse radish peroxidase (PT-66, 1:50000) was added to the plate. Following incubation for 45 min at room temperature, the plate was washed repeatedly with TBST. Detection was carried out with a color reagent, 2,2'- azido-bis-3-ethylbenzithiazoline-6-sulfonic acid, in citrate-phosphate buffer, pH 4.0, with 0.004% H2O2 for 10 min and monitored at 405 nm, all at room temperature. IC50 values of inhibitors were calculated using the REGRESSION program. The assay was optimized with respect to the amount of IGF-IR (partially purified from cells overexpressing IGFlR), reaction time, and ATP concentration. The signal was linear for 30 min in the range of IGFlR protein concentrations up to 35 ng/well.
Inhibition of IGFl-induced signaling and other signaling pathways (e.g. the EGF, PDGF and insulin-induced signaling) in intact cells
Tyrosine autophosphorylation of the β-subunit of IGFlR as well as downstream signaling induced by IGFlR were assayed in breast cancer MCF7 cells. Tyrosine autophosphorylation of the EGFR and the EGFR signaling was assayed in prostate cancer PC3 cells. Tyrosine autophosphorylation of the PDGFR and its signaling was assayed in fibroblasts overexpressing PDGFR. Tyrosine autophosphorylation of the IR and its signaling was assayed in fibroblasts overexpressing IR. Cells were seeded in 6- well plates (MCF7-250,000 cells/well, PC3-150,000 cells/well, fibroblasts- 140,000 cells/well) and 24 hours later medium was replaced by serum-free medium (DMEM supplemented with 100 units/ml penicillin and 100 μg/ml streptomycin). Following 20 hours of serum-starvation medium was replaced with medium containing various concentrations of the inhibitors in 0.1% DMSO for additional 4 hours. Cells were then stimulated for 5 min with 50 ng/ml IGF-I (MCF7 cells), 20 ng/ml EGF (PC3 cells), 50 ng/ml PDGF (fibroblasts overexpressing PDGFR) or 100 nM insulin (fibroblasts overexpressing IR), washed twice with PBS and lysed by boiling sample buffer (10% glycerol, 50 mM Tris-HCl, pH 6.8, 3% SDS, and 5% β-mercaptoethanol). Equal amounts of protein per lane were separated by 8% SDS-PAGE and transferred to a nitrocellulose membrane (Sartorius AG). Phosphorylated proteins were immunoblotted with anti-phospho-IGFIR (phosphor-IGFIR), anti-phosphotyrosine-IRSl (phospho- IRSl), anti-phospho(T308)Akt (phospo-PKB), anti-phospho-Erk (phospo-ERK), anti- phosphotyrosine 4G10&PY20 (p Y-EGFR or pY-PDGFR), anti-phospho-Ser636/639-IRSl (pS636/639-IRSl) and anti-phospho-STAT3 (ρhospho-STAT3) antibodies. Detection was performed with horseradish peroxidase-conjugated secondary antibody using the ECL system. Blots were then stripped of antibodies, blocked with TBST with 5% low fat milk and re-probed with antibodies detecting both the phosphorylated and the non- phosphorylated corresponding proteins e.g. IGF lRβ, IRSl, PKB, ERK, IRβ, PDGFR and Stat3. In addition, lysates were prepared from cells exposed to inhibitors at various concentrations for 24 hours in the presence or the absence of FCS without stimulation. Lysate preparation and western blot were the same as described above. Apoptosis was detected by immunoblotting with rabbit anti-PARP antibodies.
Proliferation assay
Various cancer cell lines (listed in table 1) were plated at a density of 1000-5000 cells/well in 96-well plates in 90 μl growth medium containing 10% FCS, 100 units/ml penicillin and 100 μg/ml streptomycin. Inhibitors were added a day later in 10 μl of 1% DMSO in DDW to obtain final concentrations of 0, 0.1, 0.3, 1, 3, and 10 μM. The final concentration of DMSO (0.1% DMSO) was kept constant in all samples. Medium with inhibitors was refreshed a day and two days later. Following exposure of the cells to the inhibitors for 72 hours at 370C, the cells were fixed in 0.5% gluteraldehyde in medium for 10 min, washed three times with DDW5 once with 0.1M sodium borate buffer pH 8.5 and stained with 1% methylene blue dissolved in 0.1 M borate buffer solution for 60 min. Excess dye was washed out and cell-bound dye was eluted with 200 μl/well of
0.1M HCl. The optical density value was read at 620nm in ELISA plate reader. The data was analyzed in Microsoft Excel, using the vehicle control as 100% proliferation. The assays were performed in triplicates. The values in table 1 represent ICs0 values derived from the dose-dependent growth curves obtained.
Clonogenic assay
Various cancer cells (listed in table 1) were seeded at a very low concentration (19 cells/well in 96-well plates or 63 cells/well in 24-well plates) in growth medium. A day later medium was replaced with growth medium containing various concentrations of inhibitors in a final concentration of 0.1% DMSO. The inhibitor-containing medium was refreshed three times a week. Following approximately two weeks cells were fixed by adding Gluteraldehide (0.5% final concentration) for 10 min, washed three time with DDW, once with borate buffer 0.1 M and stained for 1 hour with 1% Methylene Blue in borate buffer 0.1M. Access stain was washed with water, and following drying colonies were counted. Alternatively, stain was extracted by 0.1N HCl for 1 hour and absorbance at 620 nm was measured by ELISA Reader. The assays were performed in triplicates. The values in table 1 represent IC50 values derived from the dose-dependent growth curves obtained.
In-vivo effects on prostate, ovarian, melanoma and pancreatic tumor growth in xenograft models
Human hormone-refractory prostate cancer PC3 cells (ATCC, 1.5x106 cells per mouse) were injected subcutaneously into the right leg of Nude:Hsd mice (purchased from Harlan). Ten days later, when palpable tumors had developed, mice were divided into 3 groups with similar average tumor size. The untreated group (UT) did not receive any treatment. The mice treated with compound 7 group were daily injected with compound 7 dissolved in the below described vehicle, and the vehicle-treated group (veh) was daily injected by the vehicle alone. Vehicle included 4.4% DMSO, 1.2% Ethanol and 50% PEG400 in DDW; Groups were composed of 3 mice per group. Dose administered IP was 50 mg/Kg (4 ml/Kg) once a day for one month. The length (1) and the width (w) of the tumors were measured every day and the volumes of the tumors were calculated as follows: V=Iw2^. Graphs present average volumes of the tumors versus time in days. The procedure was followed for the other described in-vivo studies with the following modifications:
Human ovary cancer A2780 cells (from ECACC, 2x106 cells per mouse) were injected subcutaneously into the right leg of female Nude:Hsd mice (purchased from Harlan). Inhibitors were injected IP daily at doses of 20mg/kg (Exp. 2&3) or 50 mg/kg (Exp. 4) dissolved in 4.4%DMSO, 0.12% EtOH, 50% PEG-400 in DDW at volume of 4 ml/kg. The Veh group received 4.4%DMSO, 0.12% EtOH, 50% PEG-400 in DDW at volume of 4 ml/kg. The mouse B16 melanoma cells (1.5xlO6 cells per mouse) were injected subcutaneously into the right leg of male Nude:Hsd mice (purchased from Harlan). Inhibitors were injected IP daily at a dose of 20mg/kg dissolved in 4.4% % DMSO, 1.2% EtOH, 33% PEG-400 in DDW at volume of 4 ml/kg.
The human YUMAC metastatic melanoma cells (2x106 cells per mouse), provided by Dr. Ruth Halaban (Yale University), were injected subcutaneously into the right leg of male Nude.Ηsd mice (purchased from Harlan). Inhibitors were injected IP daily at a dose of 20mg/kg dissolved in 8% EtOH, 2% Tween-80, 20% PEG-400 and
20% solutol in DDW at volume of 4 ml/kg. Human pancreatic cancer Panel cells (2xlO6 cells per mouse) were injected subcutaneously into the right leg of male Nude:Hsd mice (purchased from Harlan). Compound 7 was injected intratumorally (IT) daily at doses of 125 μg/mouse in 0.06% DMSO and 2.16% PEG400 in saline.
In-vitro inhibition of keratinocyte growth
Primary keratinocytes were seeded in 96-well plates in growth factor enriched medium. After 48 hours medium was refreshed and compound 7 was added at the indicated concentrations (0 is 0.1% DMSO which was kept constant in all wells). Every 24 hours medium and compound 7 were refreshed, and 5 days following seeding cells were fixed with 0.5% gluteraldehide for 10 min, stained with Methylene-Blue. Access stain was washed away and the bound stain was extracted by 0.1N HCl. Absorbance at 620 nm wavelength was read in ELISA reader.
RESULTS
Biochemical characterization of the compounds of the present invention: Inhibition of IGFlR activity in cell-free assay As shown in table 1 and table 2, compounds 6-18 inhibited the kinase activity of partially purified IGFlR in a dose-dependent manner showing IC50 values of 30- 20OnM. Elimination of the hydroxyl groups increases IC50 values, as demonstrated by the comparison of compound 19 and 7 (table 1). Kinetic studies show that compound 7, as well as compounds 6 and 8 do not compete with ATP, as elevation of the ATP concentrations does not induce an increase in the IC50 values determined in a cellular- free kinase assay of IGF IR (table 2). Table 1: IC50 values of IGFlR activity in cellular free kinase assay
Figure imgf000047_0001
Table 2: Kinetic studies testing the competition of the inhibitors with ATP in cellular free kinase assay of IGFlR
Figure imgf000047_0002
Inhibition of IGFlR in cells
Exposure of breast cancer MCF7 cells to compound 7 yielded a significant inhibition of IGFl -induced signaling. Figure 4A shows that while the control molecule 20 (see structure hereinbelow) had no effect on the signaling, compound 7 dramatically inhibited the autophosphorylation of IGFlR3 the IGFl -induced tyrosine phosphorylation of IRSl (a direct substrate of IGFlR) and the IGFl -induced activation of two main anti- apoptotic and proliferative pathways downstream IGFlR, the Akt/PKB and the MAPK/ERK pathways.
Figure imgf000048_0001
Compound 20
Figure 4B exemplifies a dose-dependent activity of compound 7 in MCF7 cells, showing an IC50 value of 1-2 μM in inhibition of the central anti-apoptotic pathway downstream IGFlR, the Akt/PKB pathway. Inhibition of the MAPK/ERK pathway activation is detected at even lower concentrations.
Compound 7 was unexpectedly found to induce a decrease in the cellular levels of IGFlR-direct substrate IRSl in breast cancer MCF7 cells (Figure 5). This effect was shown to be long-lasting (Figure 5B). The decrease in IRSl levels was detected even at 24 hours following exposure of the cells to compound 7, accompanied by inhibition of its downstream signaling pathway — the antiapoptotic PKB/Akt pathway and subsequent cleavage of PARP known as a marker for cell apoptosis.
Without wishing to be bound by any particular mechanism or theory, it is contemplated that the decrease in IRSl levels induced by compound 7 is a result of the induction of a secondary negative regulation, in which IRSl undergoes inhibitory phosphorylation on serine residues and subsequent degradation. This effect is believed to be of high importance in anti-cancer activity stemming from inhibitory mechanisms of PTK signal transduction. It is further contemplated that, Compound 7 as well as other compounds of the present invention which consist of trihydroxy benzyl thioamide and halogen or halomethyl moieties in the second catecholic ring, induce Ser- phosphorylation of IRSl and/or a decrease in IRSl levels. These structures show higher activity both in- vitro (inhibition of cancer cell proliferation) and in- vivo (inhibition of tumor growth), as compared to related molecules (eg. compound 6) which show no effect on IRSl levels or Ser-phosphorylation in cells. The increase in IRS 1 Ser-phosphorylation induced by the treatment of the cells with compound 7 is demonstrated by immunoblotting with specific antibodies against IRSl phosphorylated on Ser residues 636 & 639 (Figure 5B) and by the shift in the IRSl bands in figure 5 A. Ser-phosphorylation of IRSl is known to induce decoupling of IRSl from IGFlR, and thereby inhibit IGFlR signaling. It is further shown that other related inhibitors in this sub-family (e.g. compounds 8, 9,10, 11 and 13) also trigger IRSl-ser phosphorylation in breast cancer MCF7 cells (Figure 6) as well as in metastatic melanoma YUMAC cells (Figure 8). In ovarian cancer, A2780 cells, compound 7 as well as compounds 8 and 9 induced Ser-phosphorylation (shift up of IRSl band in SDS-PAGE) and a decrease in cellular IRSl levels (Figure 7). This long- term effect on IRSl is accompanied by apoptosis of the cancer cells as demonstrated for the ovarian cancer cells following 21 hours of exposure (Figure 7) and for the melanoma cells following 24 hours of exposure (Figure 8; 3μM) to the novel inhibitors. In addition to the inhibitory effects of compound 7 on IGFlR pathway, compound 7 inhibits other tyrosine kinases such as the EGFR, PDGFR and IR in intact cells (Figure 9). These kinases have a central role in mitogenesis. Following incubation with compound 7, cells expressing these kinase receptors (e.g. prostate cancer PC3 cells expressing EGFR, fibroblast over-expressing PDGFR and fibroblasts over-expressing IR) were stimulated with either EGF, PDGF or insulin, Figures 9A, 9B and 9C respectively, and lysates were immunoblotted with anti-phosphotyrosine antibodies to detect autophosphorylation of the receptors. Downstream signaling of these receptors was measured as well.
Ser/Thr-kinases like MEK or PDK are not inhibited by compound 7 as detected by the phosphorylation levels of their substrates (ERK and PKB(Thr308) respectively) in cells where compound 7 had no inhibitory effect on upstream regulators.
Growth inhibition
Compounds 6, 7, 8, and 9 were tested for their inhibitory potential in cell proliferation assay. In this assay cells were exposed to increasing concentrations of the molecules (at 0.1% DMSO in all wells) a day after seeding. Medium and inhibitors were refreshed every day, and following three days of treatment, cells were fixed and stained with methylene-Blue. IC50 values were determined from the curves of the optical density against compound concentration. The assay was performed in triplicates.
A panel of cancer cell lines from various indications was tested for its sensitivity to the molecules (table 3). Compounds 7, 8 and 9 showed anti-cancer activity while compound 6 had lower activity. The activity correlated to the ability of these molecules to induce Ser-phosphorylation & reduction of IRSl levels in cells. As opposed to compound 6 (Figure 19A&B), compounds 7, 8, and 9 induce an increase in IRSl-ser- phosphorylation (Figures 5-8) and a decrease in IRSl levels (Figure 5 and 7). 24 hours treatment triggers apoptosis of these cells as detected by the cleavage of PARP (figure 5C, 7A and 8).
Table 3: The inhibitory activity (IC50 values in μM) of compounds 6,7,8 and 9 in cell proliferation and in clonogenic assays of various cancer cell lines. Assays performed in triplicates.
Figure imgf000051_0001
The sensitivity of the human metastatic melanoma cells was tested against a wider set of compounds (table 4), and showed high sensitivity towards the molecules that induce IRSl Ser-phosphorylation, e.g. compounds 7, 8, 9, 10 and 13 (figures 6 & 8) as opposed to compounds 6, 17 and 19.
Table 4: Inhibition of human metastatic melanoma cells by a series of novel molecules. Results are presented in IC50 values following 72 hours incubation of the cells with the molecules.
Figure imgf000052_0001
In-vivo studies
Compound 7 was tested for its inhibitory effect on tumor growth in animal model. Nude mice were subcutaneously injected with cancer cells in the flank of the mouse, and when tumors were measurable, administration of various compounds of the present invention started. Tumor dimensions presented in the graphs are those measured following the first administration. The model of human hormone-refractory prostate cancer (HRPC) PC3 was the first to be examined. Figure 10 shows that systemic administration (IP) of compound 7 at dosage of 50 mg/kg once a day, resulted in inhibition of the tumor growth by 82% as compared to controls (vehicle treated and untreated). Furthermore, no significant effect on body weight of the nude mice was detected following one month of administration (Fig. 11). Additionally, Compound 7 was found efficient when tested using intratumoral administration on both HRPC and pancreatic PANCl models (figure 16).
Since IGFlR is reported to be involved in many types of cancers, the sensitivity of a panel of cancer cell lines as well as several other analogs to compound 7, was screened in-vitro (table 3). Based on this screen, the efficacy of compounds 7 & 8 in ovarian cancer A2780 model was examined. Figure 12 shows that IP administration of 20 mg/kg of either compound 7 or 8 once a day resulted in 87% inhibition of tumor growth. A comparative study of compounds 6, 7, 8 and 9 (administered IP once a day, 20 mg/kg) shows that compounds 7, 8 and 9 inhibit ovarian tumor growth by approximately 90% and even induce the regression of small tumors, whereas compound 6 administration (under the same conditions) resulted in 33% inhibition of tumor growth solely (Fig. 13).
The efficacy of compounds 6 and 7 was tested on a melanoma B16 in- vivo model as well. Both molecules had a significant inhibitory effect with compound 7 being more efficient (figure 17). The efficacy of the compounds of the present invention on melanoma tumors was demonstrated also by the inhibition of the human metasatic melanoma YUMAC tumors in nude mice by compound 8 (figure 18).
Subsequently, the efficacy of compound 8 on inhibition of ovarian cancer A2780 tumor growth was examined. The administration began when the tumor size was larger than 50 mm or alternatively when the tumor size was larger than 470 mm . Figures 14 & 15 show that IP administration of 50 mg/kg of compound 8 once a day resulted in a significant and dramatic inhibition of tumor growth. The compounds of the present invention were thus demonstrated as efficient anti-cancer agents for various cancers both in-vitro and in- vivo.
In-vitro studies for Psoriasis
Primary keratinocytes grown in enriched medium were demonstrated useful as an in-vitro model for psoriasis. Compound 7 inhibited the in vitro growth (IC50=2.3 μM) of primary keratinocytes, and is therefore a candidate as an anti-psoriatic agent (figure 21). Biochemical characterization of compound 6
Inhibition of the tyrosine kinase activity of IGFlR and EGFR in cells Exposure of breast cancer MCF7 cells to compound 6 yielded a significant inhibition of IGFl -induced signaling. Figure 19A shows that compound 6 inhibited the autophosphorylation of IGFlR, the IGFl -induced tyrosine phosphorylation of IRSl (a direct substrate of IGFlR), and the IGFl-induced activation of the Akt/PKB. As opposed to compound 7, compound 6 has no effect on either the IGFlR or the IRSl levels (Figure 19B). Compound 6 inhibited also EGFR and EGF-induced tyrosine phosphorylation of STAT3, a direct substrate of EGFR, and the EGF-induced activation of the PKB pathway (Figures 20A&20B).
Growth inhibition Exposure of various cancer cells to compound 6 caused a significant inhibition of colony formation both in soft agar and in plates (table 3, clonogenic assay).
While certain embodiments of the invention have been illustrated and described, it is to be clear that the invention is not limited to the embodiments described herein. Numerous modifications, changes, variations, substitutions and equivalents will be apparent to those skilled in the art without departing from the spirit and scope of the present invention as described by the claims, which follow.

Claims

THE CLAIMSWhat we claim is:
1. A compound represented by the structure of formula 1 :
Figure imgf000055_0001
wherein
R , R , R and R are independently selected from H, C1-C4 alkyl, acyl and a functional group that gives rise to hydroxyl upon hydrolysis;
R3 and R7 are independently selected from H, halogen, haloalkyl and OR8 wherein R8 is H, C1-C4 alkyl, acyl or a functional group that gives rise to hydroxyl upon hydrolysis; R4 is H or CN, including salts, hydrates, solvates, polymorphs, optical isomers, geometrical isomers, enantiomers, diastereomers, and mixtures thereof.
2. A compound according to claim 1, wherein R4 is CN.
3. A compound according to claim 2, wherein R1, R2, R5 and R6 are each hydrogen.
4. A compound according to claim 2, wherein R 1 , τ R>25 r R> 5 and R are each CH3.
5. A compound according to claim 2, wherein R3 and R7 are each a hydrogen, halogen, halomethyl, OH or OCH3.
6. A compound of claim 2, wherein R1, R2, R5 and R6 are each H, R3 is halogen and R7 is OH.
7. A compound of claim 2, wherein R1, R2, R5 and R6 are each H, and R3 and
R7 are each halogen.
8. A compound of claim 2, wherein R1, R2, R5 and R6 are each H, R3 is halomethyl and R7 is OH.
9. A compound of claim 2, wherein R1, R2, R5 and R6 are each H, R3 is halogen and R7 is H.
10. A compound of claim 2, wherein R1, R2, R5 and R6 are each H5 R3 is OH and R7 is halogen.
11. A compound of claim 2, wherein R1, R2, R5 and R6 are each CH3, R3 is halogen and R7 is OCH3.
12. A compound of claim 2, wherein R1, R2, R5 and R6 are each CH3j and R3 and
R7 are each halogen.
13. A compound according to claim 1, wherein R4 is hydrogen.
14. A compound according to claim 13, wherein R1, R2, R5 and R6 are each hydrogen.
15. A compound according to claim 13, wherein R1, R2, R5 and R6 are each CH3.
16. A compound according to claim 13, wherein R and R are each hydrogen, halogen, halomethyl, OH or OCH3.
17. A compound of claim 13, wherein R1, R2, R5 and R6 are each H, R3 is halogen and R7 is OH.
18. A compound of claim 13, wherein R1, R2, R5 and R6 are each H, and R3 and
R7 are each halogen.
19. A compound of claim 13, wherein R1, R2, R5 and R6 are each H, R3 is halomethyl and R7 is OH.
20. A compound of claim 13, wherein R1, R2, R5 and R6 are each H, R3 is halogen and R7 is H.
21. A compound of claim 13, wherein R1, R2, R5 and R6 are each H, R3 is OH and R7 is halogen.
22. A compound of claim 13, wherein R1, R2, R5 and R6 are each CH3, R3 is halogen and R7 is OCH3.
23. A compound of claim 13, wherein R1, R2, R5 and R6 are each CH3, and R3 and R are each halogen.
24. A compound selected from the group consisting of:
Figure imgf000057_0001
Figure imgf000058_0001
Figure imgf000058_0002
Figure imgf000059_0001
Figure imgf000059_0002
Figure imgf000059_0003
Figure imgf000060_0001
Figure imgf000060_0002
Figure imgf000061_0001
15
Figure imgf000061_0002
16
Figure imgf000061_0003
17
Figure imgf000061_0004
Figure imgf000062_0001
19
25. A pharmaceutical composition, comprising a therapeutically effective amount of compound of claim 1 or 24, and a pharmaceutically acceptable carrier or excipient.
26. A method of inhibiting a protein tyrosine kinase (PTK), comprising the step of contacting said PTK with an effective inhibitory amount of a compound according to claim 1 or 24.
27. A method for treating or preventing a protein tyrosine kinase (PTK) related disorder in a subject comprising the step of administering to said subject a therapeutically effective amount of a compound according to claim 1 or 24.
28. The method according to claim 27, wherein the PTK related disorder is a cell proliferative disorder, a fibrotic disorder, or a metabolic disorder.
29. The method according to claim 27, wherein the PTK related disorder is cancer.
30. The method according to any of claims 26-29, wherein said protein kinase is a receptor protein tyrosine kinase (RTK).
31. The method according to claim 30, wherein said receptor protein kinase is selected from the group consisting of: a platelet-derived growth factor receptor (PDGFR), a fibroblast growth factor receptor (FGFR)5 a hepatocyte growth factor receptor (HGFR), an insulin receptor, an insulin-like growth factor- 1 receptor (IGF-IR), an epidermal growth factor receptor (EDFR), a nerve growth factor receptor (NGFR), a vascular endothelial growth factor receptor (VEGFR), and a macrophage colony stimulating factor (M-CSFR).
32. A method according to any of claims 26-31, wherein said compound is selected from the group consisting of:
Figure imgf000063_0001
Figure imgf000063_0002
Figure imgf000063_0003
4
Figure imgf000064_0001
Figure imgf000065_0001
Figure imgf000066_0001
Figure imgf000066_0002
IS
Figure imgf000066_0003
16
Figure imgf000067_0001
17
Figure imgf000067_0002
18; and
Figure imgf000067_0003
19
33. A method according to any of claims 26-32, further comprising administering a pharmaceutical composition comprising said compound, and a pharmaceutically acceptable carrier.
34. Use of a compound according to claim 1 or 24 for the preparation of a medicament for treating or preventing a protein tyrosine kinase (PTK) related disorder.
35. Use of claim 34, wherein the compound is selected from the group consisting of compounds 6, 7, 8, 9, 10 and 13.
PCT/IL2007/001494 2006-12-04 2007-12-04 Novel protein kinase modulators and therapeutic uses thereof WO2008068751A1 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
BRPI0719708-0A2A BRPI0719708A2 (en) 2006-12-04 2007-12-04 NEW PROTEIN KINASE MODULATORS AND THEIR THERAPEUTIC USES.
CA002671632A CA2671632A1 (en) 2006-12-04 2007-12-04 Novel protein kinase modulators and therapeutic uses thereof
AU2007330333A AU2007330333A1 (en) 2006-12-04 2007-12-04 Novel protein kinase modulators and therapeutic uses thereof
EP07827467.7A EP2125712B1 (en) 2006-12-04 2007-12-04 Novel protein kinase modulators and therapeutic uses thereof
JP2009539869A JP2010511694A (en) 2006-12-04 2007-12-04 Novel protein kinase modifiers and their use in therapy
US12/517,278 US8058309B2 (en) 2006-12-04 2007-12-04 Protein kinase modulators and therapeutic uses thereof
US13/293,546 US20120083528A1 (en) 2006-12-04 2011-11-10 Novel protein kinase modulators and therapeutic uses thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US87251106P 2006-12-04 2006-12-04
US60/872,511 2006-12-04

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/293,546 Continuation-In-Part US20120083528A1 (en) 2006-12-04 2011-11-10 Novel protein kinase modulators and therapeutic uses thereof

Publications (1)

Publication Number Publication Date
WO2008068751A1 true WO2008068751A1 (en) 2008-06-12

Family

ID=39111493

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IL2007/001494 WO2008068751A1 (en) 2006-12-04 2007-12-04 Novel protein kinase modulators and therapeutic uses thereof

Country Status (8)

Country Link
US (1) US8058309B2 (en)
EP (1) EP2125712B1 (en)
JP (1) JP2010511694A (en)
CN (1) CN101652345A (en)
AU (1) AU2007330333A1 (en)
BR (1) BRPI0719708A2 (en)
CA (1) CA2671632A1 (en)
WO (1) WO2008068751A1 (en)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012090204A1 (en) * 2010-12-27 2012-07-05 Novotyr Therapeutics Ltd. 2 - (2 - phenylethenyl) - 1, 3 -benzothiazine derivatives useful for the treatment cancer
WO2012117396A1 (en) 2011-03-01 2012-09-07 Novotyr Therapeutics Ltd Tyrphostin derivative in combination with cytotoxic compounds for treating cancer
US8637575B2 (en) 2008-06-05 2014-01-28 Novotyr Therapeutics Ltd. Modulators of protein kinase signaling
WO2015008206A1 (en) 2013-07-14 2015-01-22 Yissum Research Development Company Of The Hebrew University Of Jerusalem, Ltd. Igf-1r signaling pathway inhibitors useful in the treatment of neurodegenerative diseases
WO2016125169A1 (en) * 2015-02-05 2016-08-11 Tyrnovo Ltd. Combinations of irs/stat3 dual modulators and anti-cancer agents for treating cancer
WO2019097503A1 (en) * 2017-11-16 2019-05-23 Tyrnovo Ltd. Combinations of irs/stat3 dual modulators and anti pd-1/pd-l1 antibodies for treating cancer
WO2019246273A1 (en) 2018-06-20 2019-12-26 Progenity, Inc. Treatment of a disease of the gastrointestinal tract with a jak or other kinase inhibitor
CN115466205A (en) * 2021-11-04 2022-12-13 特尔诺沃有限公司 Isolated trans isomer of 3- (2-bromo-3, 4-dihydroxy-phenyl) -N- (3, 4, 5-trihydroxy-benzyl) -thioacrylamide
RU2796275C2 (en) * 2017-11-16 2023-05-22 Тырново Лтд. Combinations of dual modulators irs/stat3 and antibodies to pd-1/pd-l1 for cancer treatment

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995024190A2 (en) * 1994-03-07 1995-09-14 Sugen, Inc. Receptor tyrosine kinase inhibitors for inhibiting cell proliferative disorders and compositions thereof

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5217999A (en) 1987-12-24 1993-06-08 Yissum Research Development Company Of The Hebrew University Of Jerusalem Styryl compounds which inhibit EGF receptor protein tyrosine kinase
US20040197335A1 (en) * 2001-06-14 2004-10-07 Shimon Slavin Non-myeloablative tolerogenic treatment with tyrphostins
JP4511183B2 (en) * 2001-12-10 2010-07-28 デュ,ヤンシェン Treatment of neurodegenerative and cardiovascular diseases
SI1511710T1 (en) * 2002-05-31 2014-04-30 Proteotech, Inc. Compounds, compositions and methods for the treatment of amyloid diseases and synucleinopathies such as alzheimer s disease, type 2 diabetes, and parkinson s disease

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1995024190A2 (en) * 1994-03-07 1995-09-14 Sugen, Inc. Receptor tyrosine kinase inhibitors for inhibiting cell proliferative disorders and compositions thereof

Cited By (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8637575B2 (en) 2008-06-05 2014-01-28 Novotyr Therapeutics Ltd. Modulators of protein kinase signaling
WO2012090204A1 (en) * 2010-12-27 2012-07-05 Novotyr Therapeutics Ltd. 2 - (2 - phenylethenyl) - 1, 3 -benzothiazine derivatives useful for the treatment cancer
US9073880B2 (en) 2010-12-27 2015-07-07 Yissum Research Development Company of the Hebrew University of Jeruselem Ltd. 2-(2-phenylethenyl) 1,3-benzodiazepine derivatives useful for the treatment of cancer
WO2012117396A1 (en) 2011-03-01 2012-09-07 Novotyr Therapeutics Ltd Tyrphostin derivative in combination with cytotoxic compounds for treating cancer
WO2015008206A1 (en) 2013-07-14 2015-01-22 Yissum Research Development Company Of The Hebrew University Of Jerusalem, Ltd. Igf-1r signaling pathway inhibitors useful in the treatment of neurodegenerative diseases
US10188659B2 (en) 2013-07-14 2019-01-29 Yissum Research Development Company Of The Hebrew University Of Jerusalem, Ltd. IGF-1R signaling pathway inhibitors useful in the treatment of neurodegenerative diseases
EP3021944A4 (en) * 2013-07-14 2017-02-22 Yissum Research Development Company of The Hebrew University of Jerusalem Ltd. Igf-1r signaling pathway inhibitors useful in the treatment of neurodegenerative diseases
US9770454B2 (en) 2013-07-14 2017-09-26 Yissum Research Development Company Of The Hebrew University Of Jerusalem, Ltd. IGF-1R signaling pathway inhibitors useful in the treatment of neurodegenerative diseases
CN107250108A (en) * 2015-02-05 2017-10-13 特尔诺沃有限公司 IRS/STAT3 dual modulators and the combination of anticancer for treating cancer
WO2016125169A1 (en) * 2015-02-05 2016-08-11 Tyrnovo Ltd. Combinations of irs/stat3 dual modulators and anti-cancer agents for treating cancer
EP3750530A1 (en) 2015-02-05 2020-12-16 TyrNovo Ltd. Combinations of irs/stat3 dual modulators and anti-cancer agents for treating cancer
US10912745B2 (en) 2015-02-05 2021-02-09 Tyrnovo Ltd. Combinations of IRS/Stat3 dual modulators and anti-cancer agents for treating cancer
WO2019097503A1 (en) * 2017-11-16 2019-05-23 Tyrnovo Ltd. Combinations of irs/stat3 dual modulators and anti pd-1/pd-l1 antibodies for treating cancer
RU2796275C2 (en) * 2017-11-16 2023-05-22 Тырново Лтд. Combinations of dual modulators irs/stat3 and antibodies to pd-1/pd-l1 for cancer treatment
WO2019246273A1 (en) 2018-06-20 2019-12-26 Progenity, Inc. Treatment of a disease of the gastrointestinal tract with a jak or other kinase inhibitor
CN115466205A (en) * 2021-11-04 2022-12-13 特尔诺沃有限公司 Isolated trans isomer of 3- (2-bromo-3, 4-dihydroxy-phenyl) -N- (3, 4, 5-trihydroxy-benzyl) -thioacrylamide
CN115466205B (en) * 2021-11-04 2023-10-24 特尔诺沃有限公司 Isolated trans isomer of 3- (2-bromo-3, 4-dihydroxy-phenyl) -N- (3, 4, 5-trihydroxy-benzyl) -thioacrylamide

Also Published As

Publication number Publication date
CN101652345A (en) 2010-02-17
JP2010511694A (en) 2010-04-15
CA2671632A1 (en) 2008-06-12
EP2125712B1 (en) 2014-07-23
EP2125712A1 (en) 2009-12-02
BRPI0719708A2 (en) 2014-02-18
US20100056635A1 (en) 2010-03-04
US8058309B2 (en) 2011-11-15
AU2007330333A1 (en) 2008-06-12

Similar Documents

Publication Publication Date Title
US8058309B2 (en) Protein kinase modulators and therapeutic uses thereof
KR102618773B1 (en) Combination therapy involving diaryl macrocyclic compounds
US5700823A (en) Treatment of platelet derived growth factor related disorders such as cancers
US8236956B2 (en) Cyclopamine analogues and methods of use thereof
US8637575B2 (en) Modulators of protein kinase signaling
TW201728583A (en) Aminothiazole compounds and use thereof
US20050137172A1 (en) Haloacetamide and azide substituted compounds and methods of use thereof
KR20040089606A (en) Treating benign prostate hyperplasia with sarms
SK4152000A3 (en) Azabenzimidazole-based compounds for modulating serine/threonine protein kinase function
WO2003075828A2 (en) Compounds useful in the treatment of cancer
US20120083528A1 (en) Novel protein kinase modulators and therapeutic uses thereof
EP1631540A2 (en) Prodrugs of selective androgen receptor modulators and methods of use thereof
CN112979510A (en) Sulfonamide compounds and their use as STAT5 inhibitors
KR20180100373A (en) Uses of Ascochlorin Derivatives and AMPK Activators
US9073880B2 (en) 2-(2-phenylethenyl) 1,3-benzodiazepine derivatives useful for the treatment of cancer
US20050143430A1 (en) Catechol bioisosteres
US11298358B2 (en) Combination therapy using ascochlorin derivative
EP3021944B1 (en) Igf-1r signaling pathway inhibitors useful in the treatment of neurodegenerative diseases
US20170342047A1 (en) Cancerous Cell Growth Inhibiting Compounds
US20050080055A1 (en) Method of treating breast cancer with androgen receptor antagonists
WO2023079545A1 (en) Isolated trans isomer of 3-(2-bromo-3,4-dihydroxy-phenyl)-n-(3,4,5-trihydroxy-benzyl)-thioacrylamide
US20050085449A1 (en) Haloacetamide and azide substituted compounds and methods of use thereof

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200780050587.5

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 07827467

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2007330333

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: 2007827467

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2009539869

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2671632

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 3739/DELNP/2009

Country of ref document: IN

ENP Entry into the national phase

Ref document number: 2007330333

Country of ref document: AU

Date of ref document: 20071204

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 12517278

Country of ref document: US

ENP Entry into the national phase

Ref document number: PI0719708

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20090603