HUE033209T2 - PI3 kináz inhibitor hatású benzimidazol-származékok - Google Patents

PI3 kináz inhibitor hatású benzimidazol-származékok Download PDF

Info

Publication number
HUE033209T2
HUE033209T2 HUE11831214A HUE11831214A HUE033209T2 HU E033209 T2 HUE033209 T2 HU E033209T2 HU E11831214 A HUE11831214 A HU E11831214A HU E11831214 A HUE11831214 A HU E11831214A HU E033209 T2 HUE033209 T2 HU E033209T2
Authority
HU
Hungary
Prior art keywords
cancer
leukemia
cell
methyl
vol
Prior art date
Application number
HUE11831214A
Other languages
English (en)
Inventor
Junya Qu
Ralph Rivero
Robert Sanchez
Rosanna Tedesco
Original Assignee
Glaxosmithkline Llc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Glaxosmithkline Llc filed Critical Glaxosmithkline Llc
Publication of HUE033209T2 publication Critical patent/HUE033209T2/hu

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/337Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having four-membered rings, e.g. taxol
    • 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
    • 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
    • A61P37/00Drugs for immunological or allergic disorders
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D235/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings
    • C07D235/02Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings condensed with carbocyclic rings or ring systems
    • C07D235/04Benzimidazoles; Hydrogenated benzimidazoles
    • C07D235/06Benzimidazoles; Hydrogenated benzimidazoles with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached in position 2
    • C07D235/08Radicals containing only hydrogen and carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D235/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings
    • C07D235/02Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings condensed with carbocyclic rings or ring systems
    • C07D235/04Benzimidazoles; Hydrogenated benzimidazoles
    • C07D235/06Benzimidazoles; Hydrogenated benzimidazoles with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached in position 2
    • C07D235/10Radicals substituted by halogen atoms or nitro radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D235/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings
    • C07D235/02Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, condensed with other rings condensed with carbocyclic rings or ring systems
    • C07D235/04Benzimidazoles; Hydrogenated benzimidazoles
    • C07D235/24Benzimidazoles; Hydrogenated benzimidazoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached in position 2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/10Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/10Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/04Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/10Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/10Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/10Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a carbon chain containing aromatic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
    • C07D417/10Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings linked by a carbon chain containing aromatic rings

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Hematology (AREA)
  • Oncology (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)

Description

(12) EUROPEAN PATENT SPECIFICATION (45) Date of publication and mention (51) Int Cl.: of the grant of the patent: C07D 235110 <2006·01> C07D 401110 <2006 01> 21.12.2016 Bulletin 2016/51 C07D 235124 <200e 01) C07D 4O3IO4<200601> C07D 403110 <2006·01) C07D 405104 <2006 01> (21) Application number: 11831214.9 C07D 405110^) 0070 409110^1) C07D 513104 (2006 01) C07D 417I04<2006 01> n , ,,.. C07D 417110(200601> A61K 31l4164<200e01> (22) Date of filing. 23.09.2011 A01N 43I52<2006 01> C07D 235108<2006 01> C07D 413104 <200601> C07D 413110 <2006·01) (86) International application number: PCT/US2011/052857 (87) International publication number: WO 2012/047538 (12.04.2012 Gazette 2012/15)
(54) BENZIMIDAZOLE DERIVATIVES AS PI3 KINASE INHIBITORS
BENZIMIDAZOLDERIVATE ALS PI3-KINASE-HEMMER
DERIVES DE BENZIMIDAZOLE UTILISES COMME INHIBITEURS DE PI3 KINASE (84) Designated Contracting States: · RIVERO, Ralph AL AT BE BG CH CY CZ DE DK EE ES FI FR GB Collegeville, Pennsylvania 19426 (US) GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO · SANCHEZ, Robert PL PT RO RS SE SI SK SM TR Collegeville, Pennsylvania 19426 (US)
Designated Extension States: · TEDESCO, Rosanna BA ME Collegeville, Pennsylvania 19426 (US) (30) Priority: 29.08.2011 US 201161528397 P (74) Representative: Goff, Dawn Caroline et al 06.10.2010 US 390314 P GlaxoSmithKline
Global Patents (CN925.1) (43) Date of publication of application: 980 Great West Road 14.08.2013 Bulletin 2013/33 Brentford, Middlesex TW8 9GS (GB) (73) Proprietor: GlaxoSmithKline LLC (56) References cited:
Wilmington DE 19808 (US) EP-A1-1 553 091 US-A1- 2007 066 606 US-B2- 7 223 757 (72) Inventors: * QU, Junya
Collegeville, Pennsylvania 19426 (US)
Description
Field of the invention [0001] This invention relates to the use of a benzimidazole derivative for the modulation, notably the inhibition of the activity or function of the phosphoinositide 3’ OH kinase family (hereinafter PI3 kinases), suitably, ΡΙ3Κα, PI3K6, ΡΙ3Κβ, and/or ΡΙ3Κγ. Suitably, the present invention relates to the use of a benzimidazole in the treatment of one or more disease states selected from: autoimmune disorders, inflammatory diseases, cardiovascular diseases, neurodegener-ative diseases, allergy, asthma, pancreatitis, multiorgan failure, kidney diseases, platelet aggregation, cancer, sperm motility, transplantation rejection, graft rejection and lung injuries. More suitably, the present invention relates to a ΡΙ3Κβ selective benzimidazole compound for treating cancer.
Background of the invention [0002] The phosphoinositide 3-kinase (PI3K) pathway is among the most commonly activated in human cancer and the importance in carcinogenesis is well established (Samuels Y and Ericson K. Oncogenic PI3K and its role in cancer. Current Opinion in Oncology, 2006;18:77-82). Initiation of signaling begins with the phosphorylation of phosphatidyli-nositol-4, 5-bisphosphate (PIP2) to produce phosphatidylinositol-3, 4, 5-P3 (PIP3). PIP3 is a critical second messenger which recruits proteins that contain pleckstrin homology domains to the cell membrane where they are activated. The most studied of these proteins is AKT which promotes cell survival, growth, and proliferation.
[0003] The PI3K family consists of 15 proteins that share sequence homology, particularly within their kinase domains, but have distinct substrate specificities and modes of regulation (Vivanco I and Sawyers CL. The phosphatidylinositol 3-kinase-AKT pathway in human cancer. Nature Reviews Cancer, 2002;2:489-501). Class I PI3Ks are heterodimers consisting of a p110 catalytic subunit complexed to one of several regulatory subunits collectively referred to as p85 and have been the most extensively studied in the context of tumorgenesis. The class 1A PI3K catalytic subunits comprise the p110a, p110β, and p1106 isoforms, which associate with one of five different regulatory subunits encoded by three separate genes. A single class 1B PI3K catalytic isoform ρ110γ interacts with one of two associated regulatory subunits (Crabbe T, Welham MJ, Ward SG, The PI3k inhibitor arsenal: choose your weapon Trends in Biochem Sci, 2007;32:450-456). Class 1 PI3Ks are primarily responsible for phosphorylating the critical PIP2 signaling molecule.
[0004] The link between the PI3K pathway and cancer was confirmed by a study which identified somatic mutations in the PIK3CA gene encoding the p110a protein. Subsequently, mutations in PIK3CA have been identified in numerous cancers including colorectal, breast, glioblastomas ovarian and lung. In contrast to PIK3CA, no somatic mutations in the β isoform have been identified. However, in overexpression studies, the ΡΙ3Κβ isoform has been implicated as necessary for transformation induced by the loss or inactivation of the PTEN tumor suppressor both in vitro and in vivo (Torbett NE, Luna A, Knight ZA, et al„ A chemical screen in diverse breast cancer cell lines reveals genetic enhancers and suppressors of sensitivity to PI3K isotype-selective inhibition. Biochem J 2008;415:97-110; Zhao JJ, Liu Z, Wang L, Shin E, Loda MF, Roberts TM, The oncogenic properties of mutant p110a and p110b phosphatidylinositol 3-kinases in human mammary epithelial cells. Proc Natl Acad Sci USA 2005;102:18443-8). Consistent with this finding, overexpression of the PIK3CB gene has been identified in some bladder, colon, glioblastomas and leukemias and siRNA mediated knockdown of p110β in glioblastoma cell lines results in suppression of tumor growth in vitro and in vivo (Pu P, Kang C, Zhang Z, et al., Downregulation of PIK3CB by siRNA suppresses malignant glioma cell growth in vitro and in vivo. Technolo Cancer Res Treat 2006;5:271-280). More recent data using shRNA demonstrated that downregulation of p110β and not p110a resulted in PI3K pathway inactivation and subsequent inactivation of tumor cell growth in PTEN deficient cancers cells both in vitro and in vivo (Wee S, Wiederschain, Maira S-M, Loo A, Miller C, et al., PTEN-deficient cancers depend on PIK3CB. Proc Natl Acad Sci 2008;105:13057-13062). Consistent with a role of PIK3CB signaling in PTEN null tumors, ρ110β was reported to be essential to the transformed phenotype in a PTEN-null prostate cancer model (Jia S, Liu Z, Zhang S, Liu P, Zhang L, etal., Essential roles of PI(3)K-p110b in cell growth, metabolism and tumorgenesis. Nature 2008;10:1038).
[0005] Further, it has been reported thatfibrogenesis, including systemic sclerosis (SSc), arthritis, nephropahty, liver cirrhosis, and some cancers, are related to PTEN deficiency and corresponding PI3K-Akt overexpression (Parapuram, S.K., etal., Loss of PTEN expression by dermal fibroblasts cuases skin fibrosis. J. of Investigative Dermatology, advance online publication 9 June 2011; doi: 10.1038/jid.2011.156). Taken together, these findings indicate PI3K p110β as a promising target for cancer and other syndromes related to PTEN loss (Hollander, M. Christine; Blumenthal, Gideon M.; Dennis, Phillip P.; PTEN loss in the continuum of common cancers, rare syndromes and mouse models. Nature Re-views/Cancer 2011; 11: 289-301). It is therefore desirable to create a potent, selective inhibitor of ΡΙ3Κ-β.
[0006] US20070066606A1 discloses benzyl-benzimidazolyl derivatives which are stated to be inhibitors of tyrosine kinases, useful for the treatment of tumours.
[0007] US7223757B2 discloses benzimidazole compounds which are stated to be inhibitors of tyrosine kinases, useful as anti-cancer agents.
Summary of the Invention [0008] Disclosed are novel compounds of formula (I):
wherein R1 is selected from H, C1_6alkyl, alkoxy, hydroxy, halogen, -CN, -NH2, -NHC(0)Ra, -NHS02Ra, -C02H, -C02Ra, -CONHRb, -CONH2, -CH2OH, and heteroaryl wherein the heteroaryl may be substituted by one or two C1.3alkyl groups; R2 is selected from H, -NHRa, alkoxy, halogen, -CF3, -CHF2, and C1_6alkyl; R3 is selected from aryl and heteroaryl, wherein said aryl or heteroaryl may be substituted by one to three Rc; R4 is selected from H or Ra; each R5 is independently selected from C1_6alkyl; each Ra is independently selected from C1_3alkyl;
Rb is selected from C^alkyl, and S02Me; each Rc is independently selected from C1.3alkyl, halogen, -CF3, and hydroxy; and n is 0-2, or a pharmaceutically acceptable salt thereof.
[0009] The present invention relates to the compound, 2-methyl-1-{[2-methyl-3-(trifluoromethyl)phenyl]methyl}-6-(4-morpholinyl)-1H-benzimidazole-4-carboxylic acid represented by the formula:
or a pharmaceutically acceptable salt thereof.
[0010] In another aspect of the present invention, there is provided a compound of formula (la) or a pharmaceutically acceptable salt thereof for use in therapy.
[0011] In another aspect, there is provided a compound of formula (la) or a pharmaceutically acceptable salt thereof for use in the treatment of a susceptible neoplasm in a mammal in need thereof.
[0012] In a another aspect of the present invention, there is provided the use of a compound of formula (la) or a pharmaceutically acceptable salt thereof, in the preparation of a medicament for use in the treatment of a susceptible neoplasm in a mammal in need thereof.
[0013] In another aspect of the present invention, there is provided a pharmaceutical composition comprising a compound of formula (la) or a pharmaceutically acceptable salt thereof for use in the treatment of a susceptible neoplasm in a mammal in need thereof.
Detailed Description of the Invention [0014] Disclosed are compounds of Formula (I).
The invention relates to the compound, 2-methyl-1-{[2-methyl-3-(trifluoromethyl)phenyl]methyl}-6-(4-morpholinyl)-1H-benzimidazole-4-carboxylic acid represented by the formula
or a pharmaceutically acceptable salt thereof.
Definitions [0015] By the term "aryl" as used herein, unless otherwise defined, is meant aromatic, hydrocarbon, ring system. The ring system may be monocyclic or fused polycyclic (e.g. bicyclic, tricyclic, etc.). In various embodiments, the monocyclic aryl ring is C5-C10, or C5-C7, or C5-C6, where these carbon numbers refer to the number of carbon atoms that form the ring system. A C6 ring system, i.e. a phenyl ring is a suitable aryl group. In various embodiments, the polycyclic ring is a bicyclic aryl group, where suitable bicyclic aryl groups are C8-C12, or C9-C10. A naphthyl ring, which has 10 carbon atoms, is a suitable polycyclic aryl group.
[0016] By the term "heteroaryl" as used herein, unless otherwise defined, is meant an aromatic ring system containing carbon(s) and at least one heteroatom. Heteroaryl may be monocyclic or polycyclic. A monocyclic heteroaryl group may have 1 to 4 heteroatoms in the ring, while a polycyclic heteroaryl may contain 1 to 10 hetero atoms. A polycyclic heteroaryl ring may contain fused, spiro or bridged ring junctions, for example, bicyclic heteroaryl is a polycyclic heteroaryl. Bicyclic heteroaryl rings may contain from 8 to 12 member atoms. Monocyclic heteroaryl rings may contain from 5 to 8 member atoms (carbons and heteroatoms). Exemplary heteroaryl groups include: benzofuran, benzothiene, benzothiophene, furan, imidazole, indole, isothiazole, oxazole, pyrazine, pyrazole, pyridazine, pyridine, pyrimidine, pyrrole, quinoline, isoquinoline, quinazoline, quinoxaline, thiazole, and thiophene. According to an alternative embodiment, heteroaryls may be substituted with one to three alkyl groups.
[0017] By the term "alkoxy" as used herein is meant -O(alkyl) including -OCH3,-OCH2CH3 and -OC(CH3)3 where alkyl is as described herein.
[0018] By the term "heteroatom" as used herein is meant oxygen, nitrogen or sulfur.
[0019] By the term "halogen" as used herein is meant a substituent selected from bromide, iodide, chloride and fluoride.
[0020] By the term "alkyl" and derivatives thereof and in all carbon chains as used herein, including alkyl chains defined by the term "-(CH2)n". "-(CH2)m“ and the like, is meant a linear or branched, saturated or unsaturated hydrocarbon chain, and unless otherwise defined, the carbon chain will contain from 1 to 12 carbon atoms.
[0021] By the term "co-administering" and derivatives thereof as used herein is meant either simultaneous administration or any manner of separate sequential administration of a PI3 kinase inhibiting compound, as described herein, and a further active ingredient or ingredients. The term further active ingredient or ingredients, as used herein, includes any compound or therapeutic agent known to or that demonstrates advantageous properties when administered to a patient in need of treatment. Suitably, if the administration is not simultaneous, the compounds are administered in a close time proximity to each other. Furthermore, it does not matter if the compounds are administered in the same dosage form, e.g. one compound may be administered topically and another compound may be administered orally.
[0022] The term "compound" as used herein includes all isomers of the compound. Examples of such isomers include: enantiomers, tautomers, rotamers.
[0023] Certain compounds described herein may contain one or more chiral atoms, or may otherwise be capable of existing as two enantiomers, or two or more diastereoisomers. Accordingly, the compounds of of Formula (I) disclosed include mixtures of enantiomers/diastereoisomers as well as purified enantiomers/diastereoisomers or enantiomerical-ly/diastereoisomerically enriched mixtures. Also disclosed are the individual isomers of the compounds represented by Formula (I) above as well as any wholly or partially equilibrated mixtures thereof. Also disclosed are the individual isomers of the compounds represented by Formula (I) above as mixtures with isomers thereof in which one or more chiral centers are inverted. The present invention also includes isotopomers of the compounds of Formula (la). Examples of such isotopomers include but not limited to compounds with one of more deuterium atoms.
[0024] A compound of Formula (la) is included in the pharmaceutical compositions of the invention. Where a -COOFI or-OFI group is present, pharmaceutically acceptable esters can be employed, for example methyl, ethyl, pivaloyloxyme-thyl, and the like for -COOFI, and acetate maleate and the like for -OFI, and those esters known in the art for modifying solubility or hydrolysis characteristics, for use as sustained release or prodrug formulations.
[0025] It will be appreciated by those skilled in the art that the compounds of formula (I) may be utilized as a pharmaceutically acceptable salt version thereof. The pharmaceutically acceptable salts of the compounds of formula (I) include conventional salts formed from pharmaceutically acceptable (i.e., non-toxic) inorganic or organic acids or bases as well as quaternary ammonium salts. Representative salts include the following: acetate, benzenesulfonate, benzoate, bicarbonate, bisulfate, bitartrate, borate, bromide, calcium edetate, camsylate, carbonate, chloride, clavulanate, citrate, dihydrochloride, edetate, edisylate, estolate, esylate, ethanol amine, fumarate, gluceptate, gluconate, glutamate, glycol-lylarsanilate, hexylresorcinate, hydrabamine, hydrobromide, hydrochloride, hydroxynaphthoate, iodide, isethionate, lactate, lactobionate, laurate, malate, maleate, mandelate, mesylate (methanesulfonate), methylbromide, methylnitrate, methylsulfate, monopotassium maleate, mucate, napsylate, nitrate, N-methylglucamine, oxalate, pamoate (embonate), palmitate, pantothenate, phosphate/diphosphate, polygalacturonate, potassium, salicylate, sodium, stearate, subacetate, succinate, tannate, tartrate, teoclate, tosylate (methylbenzenesulfonate), triethiodide, trimethylammonium and valerate. Other salts, such as oxalic and trifluoroacetic, which are not themselves pharmaceutically acceptable, may be useful in the preparation of salts useful as intermediates in obtaining compounds of this invention. In one embodiment, the compound of formula (I) is in the form of the free base. In one embodiment, the compound of formula (I) is in the form of the tris salt, i.e. tris(hydroxymethyl)aminomethane. In one embodiment, the compound of formula (I) is in the form of the sulfate salt. In one embodiment, the compound of formula (I) is in the form of the hydrochloride salt. In one embodiment, the compound of formula (I) is in the form of the sodium salt. Certain salt versions of the compounds may be solvates, particularly hydrates. In one embodiment, the compound of formula (I) or a pharmaceutically acceptable salt thereof is in the form of a mono-, di-, tri- or hemihydrate.
[0026] It has now been found that compounds of Formula (I) are inhibitors of the Phosphatoinositides 3-kinases (PI3Ks). When the phosphatoinositides 3-kinase (PI3K) enzyme is inhibited by a compound of Formula (I), PI3K is unable to exert its enzymatic, biological and/or pharmacological effects. The compound of the present invention is therefore useful in the treatment of autoimmune disorders, inflammatory diseases, cardiovascular diseases, neurodegenerative diseases, allergy, asthma, pancreatitis, multiorgan failure, kidney diseases, platelet aggregation, cancer, sperm motility, transplantation rejection, graft rejection and lung injuries.
[0027] Compounds according to Formula (I) are suitable for the modulation, notably the inhibition of the activity of phosphatoinositides 3-kinases (PI3K) and, more particularly, selective inhibitors of the beta isoform of phosphatoinositides 3-kinase (ΡΙ3Κβ). Therefore the compound of the present invention is also useful for the treatment of disorders which are mediated by PI3Ks. Said treatment involves the modulation - notably the inhibition or the down regulation -of the phosphatoinositides 3-kinases.
[0028] Because the pharmaceutically active compound of the present invention is active as a PI3 kinase inhibitor, particularly inhibiting ΡΙ3Κβ, either selectively or in conjunction with one or more of PI3K6, PI3Ka, and/or ΡΙ3Κγ, it exhibits therapeutic utility in treatment of susceptible neoplasms, particularly those neoplasms that exhibit a PTEN deficiency.
[0029] As used herein, the phrase "PTEN deficient" or "PTEN deficiency" shall describe tumors with deficiencies of the tumor suppressor function of PTEN (Phosphatase and Tensin Flomolog). Such deficiency includes mutation in the PTEN gene, reduction or absence of PTEN proteins when compared to PTEN wild-type, or mutation or absence of other genes that cause suppression of PTEN function.
[0030] As used herein, the term "treatment" or "treating" in the context of therapeutic methods, refers to alleviating the specified condition, eliminating or reducing the symptoms of the condition, slowing or eliminating the progression, invasion, or metastatic spread of the condition and preventing or delaying the reoccurrence of the condition in a previously afflicted subject. The present invention further provides use of the compound of the invention for the preparation of a medicament for the treatment of several conditions in a mammal (e.g., human) in need thereof.
[0031] "Susceptible neoplasm" as used herein refers to neoplasms which are susceptible to treatment by a kinase inhibitor and particularly neoplasms that are susceptible to treatment by a ΡΙ3Κβ inhibitor. Neoplasms which have been associated with inappropriate activity of the PTEN phosphatase and particularly neoplasms which exhibit mutation of PTEN, or mutation of an upstream activator of ΡΙ3Κβ kinase or overexpression of an upstream activator of ΡΙ3Κβ kinase, and are therefore susceptible to treatment with an ΡΙ3Κβ inhibitor are known in the art, and include both primary and metastatic tumors and cancers. According to one embodiment, description of the treatment of a susceptible neoplasm may be used interchangeably with description of the treatment of a cancer.
[0032] According to one embodiment, "susceptible neoplasms" includes, but are not limited to PTEN-defiecient neoplasms listed as follows: brain (gliomas), glioblastomas, leukemias,
Bannayan-Zonana syndrome, Cowden disease,
Lhermitte-Duclos disease, breast cancer, inflammatory breast cancer, colorectal cancer Wilm’s tumor,
Ewing’s sarcoma,
Rhabdomyosarcoma, ependymoma, medulloblastoma, colon cancer, head and neck cancer, kidney cancer, lung cancer, liver cancer, melanoma, squamous cell carcinoma, ovarian cancer, pancreatic cancer, prostate cancer, sarcoma cancer, osteosarcoma, giant cell tumor of bone, thyroid cancer, lymphoblastic T cell leukemia, chronic myelogenous leukemia, chronic lymphocytic leukemia, hairy-cell leukemia, acute lymphoblastic leukemia, acute myelogenous leukemia, chronic neutrophilic leukemia, acute lymphoblastic T cell leukemia, Plasmacytoma,
Immunoblastic large cell leukemia, Mantle cell leukemia,
Multiple myeloma,
Megakaryoblastic leukemia,
Acute megakaryocytic leukemia, promyelocytic leukemia,
Erythroleukemia, malignant lymphoma, hodgkins lymphoma, non-hodgkins lymphoma, lymphoblastic T cell lymphoma,
Burkitt’s lymphoma, follicular lymphoma, neuroblastoma, bladder cancer, urothelial cancer, vulval cancer, cervical cancer, endometrial cancer, renal cancer, mesothelioma, esophageal cancer, salivary gland cancer, hepatocellular cancer, gastric cancer, nasopharangeal cancer, buccal cancer, cancer of the mouth, GIST (gastrointestinal stromal tumor), and testicular cancer.
[0033] According to an alternative embodiment, the term "susceptible neoplasm" includes and is limited to hormone refractory prostate cancer, non-small-cell lung cancer, endometrial cancer, gastric cancer, melanoma, head and neck cancer, breast cancer, including trip-negative breast cancer, and glioma. PTEN deficiency has been correlated to such cancers as demonstrated in a number of published resources, e.g. Am J Clin Pathol. 2009 Feb;131(2):257-63 (glioblastoma), J Clin Neurosci. 2010 Dec; 17(12): 1543-7 (glioblastoma), Nat Genet. 2009 May;41 (5):619-24 (prostate cancer), Br J Cancer. 2008 Oct21 ;99(8): 1296-301 (prostate cancer), Int J Cancer. 2007 Mar 15; 120(6): 1284-92 (prostate cancer), J Invest Dermatol. 2006 Jan; 126(1):154-60 (melanoma), J Clin Oncol. 2006 Jan 10;24(2):288-95 (melanoma), Am J Clin Pathol. 2005 Oct; 124(4):528-36 (melanoma), Int J Oncol. 2009 Apr;34(4):983-93 (breast cancer), Epigenetics. 2011 May 1 ;6(5):638-49 (breast cancer), Gynecol Oncol. 2009 Feb;112(2):307-13 (ovarian cancer), Mod Pathol. 2010 Oct;23(10):1316-24 (ovarian cancer), J Pathol. 2010 Feb;220(3):392-400 (ovarian cancer), Lung. 2009 Mar-Apr;187(2):104-9 (lung cancer), Anticancer Res. 2007 Jan-Feb;27(1 B):575-81 (lung cancer), Am J Surg. 2008 Jun;195(6):719-25 (colon cancer), J Clin Oncol. 2009 Dec 10;27(35):5924-30 (colon cancer), Gynecol Oncol. 2004 Jun;93(3):621-7 (cervical cancer), and J Oral Pathol Med. 2002 Aug;31 (7):379-84 (head and neck cancer).
[0034] Disclosed herein is, a method of treating a susceptible neoplasm in a mammal in need thereof, comprising administering to the mammal a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof.
[0035] Disclosed herein is a method of treating fibrosis in a mammal in need thereof, comprising administering to the mammal a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof. Fibrosis includes, alternatively or collectively, systemic sclerosis (SSc), arthritis, nephropahty, and liver cirrhosis.
[0036] Disclosed herein is a method of treating hormone refractory prostate cancer in a mammal in need thereof, comprising administering to the mammal a therapeutically effective amount of a compound of formula (I) ora pharmaceutically acceptable salt thereof.
[0037] Disclosed herein is a method of treating non-small-cell lung cancer in a mammal in need thereof, comprising administering to the mammal a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof. Disclosed herein is a method of treating endometrial cancer in a mammal in need thereof, comprising administering to the mammal a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof.
[0038] Disclosed herein is a method of treating gastric cancer in a mammal in need thereof, comprising administering to the mammal a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof. Disclosed herein is a method of treating melanoma in a mammal in need thereof, comprising administering to the mammal a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof.
[0039] Disclosed herein is a method of treating head and neck cancer in a mammal in need thereof, comprising administering to the mammal a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof. Disclosed herein is a method of treating trip-negative breast cancer in a mammal in need thereof, comprising administering to the mammal a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof.
[0040] Disclosed herein is a method of treating glioma in a mammal in need thereof, comprising administering to the mammal a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof.
[0041] In another aspect of the present invention, there is provided a compound of formula (la), ora pharmaceutically acceptable salt thereof for use in therapy.
[0042] In another aspect, there is provided a compound of formula (la) or a pharmaceutically acceptable salt thereof for use in the treatment of a susceptible neoplasm in a mammal in need thereof.
[0043] In a another aspect of the present invention, there is provided the use of a compound of formula (la) or a pharmaceutically acceptable salt thereof, in the preparation of a medicament for use in the treatment of a susceptible neoplasm in a mammal in need thereof.
[0044] In another aspect of the present invention, there is provided a pharmaceutical composition comprising a compound of formula (la) or a pharmaceutically acceptable salt thereof for use in the treatment of a susceptible neoplasm in a mammal in need thereof.
[0045] When a compound of Formula (I) is administered for the treatment of cancer, the term "co-administering" and derivatives thereof as used herein is meant either simultaneous administration or any manner of separate sequential administration of a PI3 kinase inhibiting compound, as described herein, and a further active ingredient or ingredients, known to be useful in the treatment of cancer, including chemotherapy and radiation treatment. The term further active ingredient or ingredients, as used herein, includes any compound or therapeutic agent known to or that demonstrates advantageous properties when administered to a patient in need of treatment for cancer. Preferably, if the administration is not simultaneous, the compounds are administered in a close time proximity to each other. Furthermore, it does not matter if the compounds are administered in the same dosage form, e.g. one compound may be administered topically and another compound may be administered orally.
[0046] Typically, any anti-neoplastic agent that has activity versus a susceptible tumor being treated may be coadministered in the treatment of cancer in the present invention. Examples of such agents can be found in Cancer Principles and Practice f Oncology by V.T. Devita and S. Flellman (editors), 6th edition (February 15, 2001), Lippincott Williams &amp; Wilkins Publishers. A person of ordinary skill in the art would be able to discern which combinations of agents would be useful based on the particular characteristics of the drugs and the cancer involved. Typical anti-neoplastic agents useful in the present invention include, but are not limited to, anti-microtubule agents such as diterpenoids and vinca alkaloids; platinum coordination complexes; alkylating agents such as nitrogen mustards, oxazaphosphorines, alkylsulfonates, nitrosoureas, and triazenes; antibiotic agents such as anthracyclins, actinomycins and bleomycins; topoisomerase II inhibitors such as epipodophyllotoxins; antimetabolites such as purine and pyrimidine analogues and anti-folate compounds; topoisomerase I inhibitors such as camptothecins; hormones and hormonal analogues; signal transduction pathway inhibitors; non-receptor tyrosine kinase angiogenesis inhibitors; immunotherapeutic agents; proa-poptotic agents; and cell cycle signaling inhibitors.
[0047] Examples of a further active ingredient or ingredients for use in combination or co-administered with the present PI3 kinase inhibiting compound are chemotherapeutic agents.
[0048] Anti-microtubule or anti-mitotic agents are phase specific agents active against the microtubules of tumor cells during M or the mitosis phase of the cell cycle. Examples of anti-microtubule agents include, but are not limited to, diterpenoids and vinca alkaloids.
[0049] Diterpenoids, which are derived from natural sources, are phase specific anti - cancer agents that operate at the G2/M phases of the cell cycle. It is believed that the diterpenoids stabilize the β-tubulin subunit of the microtubules, by binding with this protein. Disassembly of the protein appears then to be inhibited with mitosis being arrested and cell death following. Examples of diterpenoids include, but are not limited to, paclitaxel and its analog docetaxel.
[0050] Paclitaxel, 5β,20-epoxy-1,2a,4^,1C^,13a,-hexa-hydroxytax-11-en-9-one 4,10-diacetate 2-benzoate 13-ester with (2R,3S)-N-benzoyl-3-phenylisoserine; is a natural diterpene product isolated from the Pacific yew tree Taxus brev-ifolia and is commercially available as an injectable solution TAXOL®. It is a member of the taxane family of terpenes. It was first isolated in 1971 by Wani et al. J. Am. Chem, Soc., 93:2325.1971), who characterized its structure by chemical and X-ray crystallographic methods. One mechanism for its activity relates to paclitaxel’s capacity to bind tubulin, thereby inhibiting cancer cell growth. Schiff et al., Proc. Natl. Acad, Sci. USA, 77:1561-1565 (1980); Schiff et al., Nature, 277:665-667 (1979); Kumar, J. Biol, Chem, 256: 10435-10441 (1981). For a review of synthesis and anticancer activity of some paclitaxel derivatives see: D. G. I. Kingston et al., Studies in Organic Chemistry vol. 26, entitled "New trends in Natural Products Chemistry 1986", Attaur-Rahman, P.W. Le Ouesne, Eds. (Elsevier, Amsterdam, 1986) pp 219-235.
[0051] Paclitaxel has been approved for clinical use in the treatment of refractory ovarian cancer in the United States (Markman et al., Yale Journal of Biology and Medicine, 64:583, 1991; McGuire et al., Ann. Intern, Med., 111:273,1989) and for the treatment of breast cancer (Holmes et al., J. Nat. Cancer Inst., 83:1797,1991.) It is a potential candidate for treatment of neoplasms in the skin (Einzig et. al., Proc. Am. Soc. Clin. Oncol., 20:46) and head and neck carcinomas (Forastire et. al., Sem. Oncol., 20:56, 1990). The compound also shows potential for the treatment of polycystic kidney disease (Woo et. al., Nature, 368:750. 1994), lung cancer and malaria. Treatment of patients with paclitaxel results in bone marrow suppression (multiple cell lineages, Ignoff, R.J. et. al, Cancer Chemotherapy Pocket Guide, 1998) related to the duration of dosing above a threshold concentration (50nM) (Kearns, C.M. et. al., Seminars in Oncology, 3(6) p.16-23, 1995).
[0052] Docetaxel, (2R,3S)-N-carboxy-3-phenylisoserine,N-ferf-butyl ester, 13-ester with 5β-20-epoxy-1,2a,4^,1C^,13a-hexahydroxytax-11-en-9-one 4-acetate 2-benzoate, trihydrate; is commercially available as an injectable solution asTAXOTERE®. Docetaxel is indicated for the treatment of breast cancer. Docetaxel is a semisynthetic derivative of paclitaxel q.v., prepared using a natural precursor, 10-deacetyl-baccatin III, extracted from the needle of the European Yew tree. The dose limiting toxicity of docetaxel is neutropenia.
[0053] Vinca alkaloids are phase specific anti-neoplastic agents derived from the periwinkle plant. Vinca alkaloids act at the M phase (mitosis) of the cell cycle by binding specifically to tubulin. Consequently, the bound tubulin molecule is unable to polymerize into microtubules. Mitosis is believed to be arrested in metaphase with cell death following. Examples of vinca alkaloids include, but are not limited to, vinblastine, vincristine, and vinorelbine.
[0054] Vinblastine, vincaleukoblastine sulfate, is commercially available as VELBAN® as an injectable solution. Al- though, it has possible indication as a second line therapy of various solid tumors, it is primarily indicated in the treatment of testicular cancer and various lymphomas including Hodgkin’s Disease; and lymphocytic and histiocytic lymphomas. Myelosuppression is the dose limiting side effect of vinblastine.
[0055] Vincristine, vincaleukoblastine, 22-oxo-, sulfate, is commercially available as ONCOVIN® as an injectable solution. Vincristine is indicated for the treatment of acute leukemias and has also found use in treatment regimens for Hodgkin’s and non-Hodgkin’s malignant lymphomas. Alopecia and neurologic efFects are the most common side effect of vincristine and to a lesser extent myelosupression and gastrointestinal mucositis effects occur.
[0056] Vinorelbine, 3’,4’-didehydro -4’-deoxy-C’-norvincaleukoblastine [R-(R*,R*)-2,3-dihydroxybutanedioate (1:2)(salt)], commercially available as an injectable solution of vinorelbine tartrate (NAVELBINE®), is a semisynthetic vinca alkaloid. Vinorelbine is indicated as a single agent or in combination with other chemotherapeutic agents, such as cisplatin, in the treatment of various solid tumors, particularly non-small cell lung, advanced breast, and hormone refractory prostate cancers. Myelosuppression is the most common dose limiting side effect of vinorelbine.
[0057] Platinum coordination complexes are non-phase specific anti-cancer agents, which are interactive with DNA. The platinum complexes entertumor cells, undergo aquation and form intra-and interstrand crosslinks with DNA causing adverse biological effects to the tumor. Examples of platinum coordination complexes include, but are not limited to, cisplatin and carboplatin.
[0058] Cisplatin, cis-diamminedichloroplatinum, is commercially available as PLATINOL® as an injectable solution. Cisplatin is primarily indicated in the treatment of metastatic testicular and ovarian cancer and advanced bladder cancer. The primary dose limiting side effects of cisplatin are nephrotoxicity, which may be controlled by hydration and diuresis, and ototoxicity.
[0059] Carboplatin, platinum, diammine [1,1-cyclobutane-dicarboxylate(2-)-0,0’], is commercially available as PAR-APLATIN® as an injectable solution. Carboplatin is primarily indicated in the first and second line treatment of advanced ovarian carcinoma. Bone marrow suppression is the dose limiting toxicity of carboplatin.
[0060] Alkylating agents are non-phase anti-cancer specific agents and strong electrophiles. Typically, alkylating agents form covalent linkages, by alkylation, to DNA through nucleophilic moieties of the DNA molecule such as phosphate, amino, sulfhydryl, hydroxy, carboxyl, and imidazole groups. Such alkylation disrupts nucleic acid function leading to cell death. Examples of alkylating agents include, but are not limited to, nitrogen mustards such as cyclophosphamide, melphalan, and chlorambucil; alkyl sulfonates such as busulfan; nitrosoureas such as carmustine; and triazenes such as dacarbazine.
[0061] Cyclophosphamide, 2-[bis(2-chloroethyl)amino]tetrahydro-2H-1,3,2-oxazaphosphorine 2-oxide monohydrate, is commercially available as an injectable solution or tablets as CYTOXAN®. Cyclophosphamide is indicated as a single agent or in combination with other chemotherapeutic agents, in the treatment of malignant lymphomas, multiple myeloma, and leukemias. Alopecia, nausea, vomiting and leukopenia are the most common dose limiting side effects of cyclophosphamide.
[0062] Melphalan, 4-[bis(2-chloroethyl)amino]-L-phenylalanine, is commercially available as an injectable solution or tablets as ALKERAN®. Melphalan is indicated for the palliative treatment of multiple myeloma and non-resectable epithelial carcinoma of the ovary. Bone marrow suppression is the most common dose limiting side effect of melphalan.
[0063] Chlorambucil, 4-[bis(2-chloroethyl)amino]benzenebutanoic acid, is commercially available as LEUKERAN® tablets. Chlorambucil is indicated for the palliative treatment of chronic lymphatic leukemia, and malignant lymphomas such as lymphosarcoma, giant follicular lymphoma, and Hodgkin’s disease. Bone marrow suppression is the most common dose limiting side effect of chlorambucil.
[0064] Busulfan, 1,4-butanediol dimethanesulfonate, is commercially available as MYLERAN® TABLETS. Busulfan is indicated for the palliative treatment of chronic myelogenous leukemia. Bone marrow suppression is the most common dose limiting side effects of busulfan.
[0065] Carmustine, 1,3-[bis(2-chloroethyl)-1 -nitrosourea, is commercially available as single vials of lyophilized material as BiCNU®. Carmustine is indicated for the palliative treatment as a single agent or in combination with other agents for brain tumors, multiple myeloma, Hodgkin’s disease, and non-Hodgkin’s lymphomas. Delayed myelosuppression is the most common dose limiting side effects of carmustine.
[0066] Dacarbazine, 5-(3,3-dimethyl-1-triazeno)-imidazole-4-carboxamide, is commercially available as single vials of material as DTIC-Dome®. Dacarbazine is indicated for the treatment of metastatic malignant melanoma and in combination with other agents for the second line treatment of Hodgkin’s Disease. Nausea, vomiting, and anorexia are the most common dose limiting side effects of dacarbazine.
[0067] Antibiotic anti-neoplastics are non-phase specific agents, which bind or intercalate with DNA. Typically, such action results in stable DNA complexes or strand breakage, which disrupts ordinary function of the nucleic acids leading to cell death. Examples of antibiotic anti-neoplastic agents include, but are not limited to, actinomycins such as dactin-omycin, anthrocyclins such as daunorubicin and doxorubicin; and bleomycins.
[0068] Dactinomycin, also know as Actinomycin D, is commercially available in injectable form as COSMEGEN®. Dactinomycin is indicated for the treatment of Wilm’s tumor and rhabdomyosarcoma. Nausea, vomiting, and anorexia are the most common dose limiting side effects of dactinomycin.
[0069] Daunorubicin, (8S-cis-)-8-acetyl-10-[(3-amino-2,3,6-trideoxy-a-L-lyxo-hexopyranosyl)oxy]-7,8,9,10-tetrahy-dro-6,8,11-trihydroxy-1-methoxy-5,12 naphthacenedione hydrochloride, is commercially available as a liposomal injectable form as DAUNOXOME® or as an injectable as CERUBIDINE®. Daunorubicin is indicated for remission induction in the treatment of acute nonlymphocytic leukemia and advanced HIV associated Kaposi’s sarcoma. Myelosuppression is the most common dose limiting side effect of daunorubicin.
[0070] Doxorubicin, (8S, 10S)-10-[(3-amino-2,3,6-trideoxy-a-L-lyxo-hexopyranosyl)oxy]-8-glycoloyl, 7,8,9,10-tetrahydro-6, 8,11-trihydroxy-1-methoxy-5,12 naphthacenedione hydrochloride, is commercially available as an injectable form as RUBEX® or ADRIAMYCIN RDF®. Doxorubicin is primarily indicated for the treatment of acute lymphoblastic leukemia and acute myeloblastic leukemia, but is also a useful component in the treatment of some solid tumors and lymphomas. Myelosuppression is the most common dose limiting side effect of doxorubicin.
[0071] Bleomycin, a mixture of cytotoxic glycopeptide antibiotics isolated from a strain of Streptomyces verticillus, is commercially available as BLENOXANE®. Bleomycin is indicated as a palliative treatment, as a single agent or in combination with other agents, of squamous cell carcinoma, lymphomas, and testicular carcinomas. Pulmonary and cutaneous toxicities are the most common dose limiting side effects of bleomycin.
[0072] Topoisomerase II inhibitors include, but are not limited to, epipodophyllotoxins.
[0073] Epipodophyllotoxins are phase specific anti-neoplastic agents derived from the mandrake plant. Epipodophyllotoxins typically affect cells in the S and G2 phases of the cell cycle by forming a ternary complex with topoisomerase II and DNA causing DNA strand breaks. The strand breaks accumulate and cell death follows. Examples of epipodophyllotoxins include, but are not limited to, etoposide and teniposide.
[0074] Etoposide, 4’-demethyl-epipodophyllotoxin 9[4,6-0-(R)-ethylidene-p-D-glucopyranoside], is commercially available as an injectable solution or capsules as VePESID® and is commonly known as VP-16. Etoposide is indicated as a single agent or in combination with other chemotherapy agents in the treatment of testicular and non-small cell lung cancers. Myelosuppression is the most common side effect of etoposide. The incidence of leucopenia tends to be more severe than thrombocytopenia.
[0075] Teniposide, 4’-demethyl-epipodophyllotoxin 9[4,6-0-(R)-thenylidene-p-D-glucopyranoside], is commercially available as an injectable solution as VUMON® and is commonly known as VM-26. Teniposide is indicated as a single agent or in combination with other chemotherapy agents in the treatment of acute leukemia in children. Myelosuppression is the most common dose limiting side effect of teniposide. Teniposide can induce both leucopenia and thrombocytopenia.
[0076] Antimetabolite neoplastic agents are phase specific anti-neoplastic agents that act at S phase (DNA synthesis) of the cell cycle by inhibiting DNA synthesis or by inhibiting purine or pyrimidine base synthesis and thereby limiting DNA synthesis. Consequently, S phase does not proceed and cell death follows. Examples of antimetabolite anti-neoplastic agents include, but are not limited to, fluorouracil, methotrexate, cytarabine, mecaptopurine, thioguanine, and gemcitabine.
[0077] 5-Fluorouracil, 5-fluoro-2,4- (1H,3H) pyrimidinedione, is commercially available as fluorouracil. Administration of 5-fluorouracil leads to inhibition of thymidylate synthesis and is also incorporated into both RNA and DNA. The result typically is cell death. 5-Fluorouracil is indicated as a single agent or in combination with other chemotherapy agents in the treatment of carcinomas of the breast, colon, rectum, stomach and pancreas. Myelosuppression and mucositis are dose limiting side effects of 5-fluorouracil. Other fluoropyrimidine analogs include 5-fluoro deoxyuridine (floxuridine) and 5-fluorodeoxyuridine monophosphate.
[0078] Cytarabine, 4-amino-1-p-D-arabinofuranosyl-2(1 H)-pyrimidinone, is commercially available as CYTOSAR-U® and is commonly known as Ara-C. It is believed that cytarabine exhibits cell phase specificity at S-phase by inhibiting DNA chain elongation by terminal incorporation of cytarabine into the growing DNA chain. Cytarabine is indicated as a single agent or in combination with other chemotherapy agents in the treatment of acute leukemia. Other cytidine analogs include 5-azacytidine and 2’,2’-difluorodeoxycytidine (gemcitabine). Cytarabine induces leucopenia, thrombocytopenia, and mucositis.
[0079] Mercaptopurine, 1,7-dihydro-6H-purine-6-thione monohydrate, is commercially available as PURINETHOL®. Mercaptopurine exhibits cell phase specificity at S-phase by inhibiting DNA synthesis by an as of yet unspecified mechanism. Mercaptopurine is indicated as a single agent or in combination with other chemotherapy agents in the treatment of acute leukemia. Myelosuppression and gastrointestinal mucositis are expected side effects of mercaptopurine at high doses. A useful mercaptopurine analog is azathioprine.
[0080] Thioguanine, 2-amino-1,7-dihydro-6H-purine-6-thione, is commercially available as TABLOID®. Thioguanine exhibits cell phase specificity at S-phase by inhibiting DNA synthesis by an as ofyet unspecified mechanism. Thioguanine is indicated as a single agent or in combination with other chemotherapy agents in the treatment of acute leukemia. Myelosuppression, including leucopenia, thrombocytopenia, and anemia, is the most common dose limiting side effect of thioguanine administration. However, gastrointestinal side effects occur and can be dose limiting. Other purine analogs include pentostatin, erythrohydroxynonyladenine, fludarabine phosphate, and cladribine.
[0081] Gemcitabine, 2’-deoxy-2’,2’-difluorocytidine monohydrochloride (β-isomer), is commercially available as GEM- ZAR®. Gemcitabine exhibits cell phase specificity at S-phase and by blocking progression of cells through the G1/S boundary. Gemcitabine is indicated in combination with cisplatin in the treatment of locally advanced non-small cell lung cancer and alone in the treatment of locally advanced pancreatic cancer. Myelosuppression, including leucopenia, thrombocytopenia, and anemia, is the most common dose limiting side effect of gemcitabine administration.
[0082] Methotrexate, N-[4[[(2,4-diamino-6-pteridinyl) methyl]methylamino] benzoyl]-L-glutamic acid, is commercially available as methotrexate sodium. Methotrexate exhibits cell phase effects specifically at S-phase by inhibiting DNA synthesis, repair and/or replication through the inhibition of dyhydrofolic acid reductase which is required for synthesis of purine nucleotides and thymidylate. Methotrexate is indicated as a single agent or in combination with other chemotherapy agents in the treatment of choriocarcinoma, meningeal leukemia, non-Hodgkin’s lymphoma, and carcinomas of the breast, head, neck, ovary and bladder. Myelosuppression (leucopenia, thrombocytopenia, and anemia) and mucositis are expected side effect of methotrexate administration.
[0083] Camptothecins, including, camptothecin and camptothecin derivatives are available or under development as Topoisomerase I inhibitors. Camptothecins cytotoxic activity is believed to be related to its Topoisomerase I inhibitory activity. Examples of camptothecins include, but are not limited to irinotecan, topotecan, and the various optical forms of 7-(4-methylpiperazino-methylene)-10,11-ethylenedioxy-20-camptothecin described below.
[0084] Irinotecan HCI, (4S)-4,11-diethyl-4-hydroxy-9-[(4-piperidinopiperidino) carbonyloxy]-1H-pyrano[3’,4’,6,7]in-dolizino[1,2-b]quinoline-3,14(4H,12H)-dione hydrochloride, is commercially available as the injectable solution CAMP- TOSAR®.
[0085] Irinotecan is a derivative of camptothecin which binds, along with its active metabolite SN-38, to the topoisomerase I - DNA complex. It is believed that cytotoxicity occurs as a result of irreparable double strand breaks caused by interaction of the topoisomerase I : DNA : irintecan or SN-38 ternary complex with replication enzymes. Irinotecan is indicated for treatment of metastatic cancer of the colon or rectum. The dose limiting side effects of irinotecan HCI are myelosuppression, including neutropenia, and Gl effects, including diarrhea.
[0086] Topotecan HCI, (S)-10-[(dimethylamino)methyl]-4-ethyl-4,9-dihydroxy-1H-pyrano[3’,4’,6,7]indolizino[1,2-b]quinoline-3,14-(4H,12H)-dione monohydrochloride, is commercially available as the injectable solution HYCAMTIN®. Topotecan is a derivative of camptothecin which binds to the topoisomerase I - DNA complex and prevents religation of singles strand breaks caused by Topoisomerase I in response to torsional strain of the DNA molecule. Topotecan is indicated for second line treatment of metastatic carcinoma of the ovary and small cell lung cancer. The dose limiting side effect of topotecan HCI is myelosuppression, primarily neutropenia.
[0087] Also of interest, is the camptothecin derivative of formula A following, currently under development, including the racemic mixture (R,S) form as well as the R and S enantiomers:
A known by the chemical name "7-(4-methylpiperazino-methylene)-10,11-ethylenedioxy-20(R,S)-camptothecin (racemic mixture) or "7-(4-methylpiperazino-methylene)-10,11-ethylenedioxy-20(R)-camptothecin (R enantiomer) or "7-(4-meth-ylpiperazino-methylene)-10,11-ethylenedioxy-20(S)-camptothecin (S enantiomer). Such compound as well as related compounds are described, including methods of making, in U.S. Patent Nos. 6,063,923; 5,342,947; 5,559,235; 5,491,237 and pending U.S. patent Application No. 08/977,217 filed November 24, 1997.
[0088] Hormones and hormonal analogues are useful compounds for treating cancers in which there is a relationship between the hormone(s) and growth and/or lack of growth of the cancer. Examples of hormones and hormonal analogues useful in cancer treatment include, but are not limited to, adrenocorticosteroids such as prednisone and prednisolone which are useful in the treatment of malignant lymphoma and acute leukemia in children ; aminoglutethimide and other aromatase inhibitors such as anastrozole, letrazole, vorazole, and exemestane useful in the treatment of adrenocortical carcinoma and hormone dependent breast carcinoma containing estrogen receptors; progestrins such as megestrol acetate useful in the treatment of hormone dependent breast cancer and endometrial carcinoma; estrogens, androgens, and anti-androgens such as flutamide, nilutamide, bicalutamide, cyproterone acetate and 5a-reductases such as finas teride and dutasteride, useful in the treatment of prostatic carcinoma and benign prostatic hypertrophy; antiestrogens such as tamoxifen, toremifene, raloxifene, droloxifene, iodoxyfene, as well as selective estrogen receptor modulators (SERMS) such those described in U.S. Patent Nos. 5,681,835, 5,877,219, and 6,207,716, useful in the treatment of hormone dependent breast carcinoma and other susceptible cancers; and gonadotropin-releasing hormone (GnRH) and analogues thereof which stimulate the release of leutinizing hormone (LH) and/or follicle stimulating hormone (FSH) for the treatment of prostatic carcinoma, for instance, LHRH agonists and antagagonists such as goserelin acetate and luprolide.
[0089] Signal transduction pathway inhibitors are those inhibitors, which block or inhibit a chemical process which evokes an intracellular change. As used herein this change is cell proliferation or differentiation. Signal tranduction inhibitors useful in the present invention include inhibitors of receptor tyrosine kinases, non-receptor tyrosine kinases, SH2/SH3domain blockers, serine/threonine kinases, phosphotidyl inositol-3 kinases, myoinositol signaling, and Ras oncogenes.
[0090] Several protein tyrosine kinases catalyse the phosphorylation of specific tyrosyl residues in various proteins involved in the regulation of cell growth. Such protein tyrosine kinases can be broadly classified as receptor or nonreceptor kinases.
[0091] Receptor tyrosine kinases are transmembrane proteins having an extracellular ligand binding domain, a transmembrane domain, and a tyrosine kinase domain. Receptor tyrosine kinases are involved in the regulation of cell growth and are generally termed growth factor receptors. Inappropriate or uncontrolled activation of many of these kinases, i.e. aberrant kinase growth factor receptor activity, for example by overexpression or mutation, has been shown to result in uncontrolled cell growth. Accordingly, the aberrant activity of such kinases has been linked to malignant tissue growth. Consequently, inhibitors of such kinases could provide cancer treatment methods. Growth factor receptors include, for example, epidermal growth factor receptor (EGFr), platelet derived growth factor receptor (PDGFr), erbB2, erbB4, vascular endothelial growth factor receptor (VEGFr), tyrosine kinase with immunoglobulin-like and epidermal growth factor homology domains (TIE-2), insulin growth factor -I (IGFI) receptor, macrophage colony stimulating factor (cfms), BTK, ckit, cmet, fibroblast growth factor (FGF) receptors, Trk receptors (TrkA, TrkB, and TrkC), ephrin (eph) receptors, and the RET protooncogene. Several inhibitors of growth receptors are underdevelopment and include ligand antagonists, antibodies, tyrosine kinase inhibitors and anti-sense oligonucleotides. Growth factor receptors and agents that inhibit growth factor receptor function are described, for instance, in Kath, John C., Exp. Opin. Ther. Patents (2000) 10(6):803-818; Shawver et al DDT Vol 2, No. 2 February 1997; and Lofts, F. J. et al, "Growth factor receptors as targets", New Molecular Targets for Cancer Chemotherapy, ed. Workman, Paul and Kerr, David, CRC press 1994, London.
[0092] Tyrosine kinases, which are not growth factor receptor kinases are termed non-receptor tyrosine kinases. Nonreceptor tyrosine kinases useful in the present invention, which are targets or potential targets of anti-cancer drugs, include cSrc, Lck, Fyn, Yes, Jak, cAbl, FAK (Focal adhesion kinase), Brutons tyrosine kinase, and Bcr-Abl. Such nonreceptor kinases and agents which inhibit non-receptor tyrosine kinase function are described in Sinh, S. and Corey, S.J., (1999) Journal of Hematotherapy and Stem Cell Research 8 (5): 465 - 80; and Bolen, J.B., Brugge, J.S., (1997) Annual review of Immunology. 15: 371-404.
[0093] SH2/SH3 domain blockers are agents that disrupt SH2 or SH3 domain binding in a variety of enzymes or adaptor proteins including, PI3-K p85 subunit, Src family kinases, adaptor molecules (She, Crk, Nek, Grb2) and Ras-GAP. SH2/SH3 domains as targets for anti-cancer drugs are discussed in Smithgall, T.E. (1995), Journal of Pharmacological and Toxicological Methods. 34(3) 125-32.
[0094] Inhibitors of Serine/Threonine Kinases including MAP kinase cascade blockers which include blockers of Raf kinases (rafk), Mitogen or Extracellular Regulated Kinase (MEKs), and Extracellular Regulated Kinases (ERKs); and Protein kinase C family member blockers including blockers of PKCs (alpha, beta, gamma, epsilon, mu, lambda, iota, zeta). IkB kinase family (IKKa, IKKb), PKB family kinases, AKT kinase family members, and TGF beta receptor kinases. Such Serine/Threonine kinases and inhibitors thereof are described in Yamamoto, T., Taya, S., Kaibuchi, K., (1999), Journal of Biochemistry. 126 (5) 799-803; Brodt, P, Samani, A., and Navab, R. (2000), Biochemical Pharmacology, 60. 1101-1107; Massague, J., Weis-Garcia, F. (1996) Cancer Surveys. 27:41-64; Philip, P.A., and Harris, A.L. (1995), Cancer Treatment and Research. 78: 3-27, Lackey, K. et al Bioorganic and Medicinal Chemistry Letters, (10), 2000, 223-226; U.S. Patent No. 6,268,391; and Martinez-lacaci, L., et al, Int. J. Cancer (2000), 88(1), 44-52.
[0095] Inhibitors of Phosphotidyl inositol-3 Kinase family members including blockers of PI3-kinase, ATM, DNA-PK, and Ku are also useful in the present invention. Such kinases are discussed in Abraham, R.T. (1996), Current Opinion in Immunology. 8 (3) 412-8; Canman, C.E., Lim, D.S. (1998), Oncogene 17 (25) 3301-3308; Jackson, S.P. (1997), International Journal of Biochemistry and Cell Biology. 29 (7):935-8; and Zhong, H. et al, Cancer res, (2000) 60(6), 1541-1545.
[0096] Also useful in the present invention are Myo-inositol signaling inhibitors such as phospholipase C blockers and Myoinositol analogues. Such signal inhibitors are described in Powis, G., and Kozikowski A., (1994) New Molecular Targets for Cancer Chemotherapy ed., Paul Workman and David Kerr, CRC press 1994, London.
[0097] Another group of signal transduction pathway inhibitors are inhibitors of Ras Oncogene. Such inhibitors include inhibitors of farnesyltransferase, geranyl-geranyl transferase, and CAAX proteases as well as anti-sense oligonucleotides, ribozymes and immunotherapy. Such inhibitors have been shown to block ras activation in cells containing wild type mutant ras , thereby acting as antiproliferation agents. Ras oncogene inhibition is discussed in Scharovsky, O.G., Rozados, V.R., Gervasoni, S.l. Matar, P. (2000), Journal of Biomedical Science. 7(4) 292-8; Ashby, M.N. (1998), Current Opinion in Lipidology. 9 (2) 99 - 102; and BioChem. Biophys. Acta, (19899) 1423(3):19-30.
[0098] As mentioned above, antibody antagonists to receptor kinase ligand binding may also serve as signal transduction inhibitors. This group of signal transduction pathway inhibitors includes the use of humanized antibodies to the extracellular ligand binding domain of receptor tyrosine kinases. For example Imclone C225 EGFR specific antibody (see Green, M.C. et al, Monoclonal Antibody Therapy for Solid Tumors, Cancer Treat. Rev., (2000), 26(4), 269-286); Flerceptin ® erbB2 antibody (see Tyrosine Kinase Signalling in Breast cancenerbB Family Receptor Tyrosine Kinases, Breast cancer Res., 2000, 2(3), 176-183); and 2CB VEGFR2 specific antibody (see Brekken, R.A. et al, Selective Inhibition of VEGFR2 Activity by a monoclonal Anti-VEGF antibody blocks tumor growth in mice, Cancer Res. (2000) 60, 5117-5124).
[0099] Non-receptor kinase angiogenesis inhibitors may also find use in the present invention. Inhibitors of angiogenesis related VEGFR and TIE2 are discussed above in regard to signal transduction inhibitors (both receptors are receptor tyrosine kinases). Accordingly, non-receptor tyrosine kinase inhibitors may be used in combination with the inhibitor of the present invention. For example, anti-VEGF antibodies, which do not recognize VEGFR (the receptor tyrosine kinase), but bind to the ligand; small molecule inhibitors of integrin (alphav beta3) that will inhibit angiogenesis; endostatin and angiostatin (non-RTK) may also prove useful in combination with the disclosed family inhibitors. (See Bruns CJ et al (2000), Cancer Res., 60: 2926-2935; Schreiber AB, Winkler ME, and Derynck R. (1986), Science, 232: 1250-1253; Yen L et al. (2000), Oncogene 19: 3460-3469).
[0100] Agents used in immunotherapeutic regimens may also be useful in combination with the compounds of formula (I). There are a number of immunologic strategies to generate an immune response against erbB2 or EGFR. These strategies are generally in the realm of tumor vaccinations. The efficacy of immunologic approaches may be greatly enhanced through combined inhibition of erbB2/EGFR signaling pathways using a small molecule inhibitor. Discussion of the immunologic/tumor vaccine approach against erbB2/EGFR are found in Reilly RT et al. (2000), Cancer Res. 60: 3569-3576; and Chen Y, Flu D, Eling DJ, Robbins J, and Kipps TJ. (1998), Cancer Res. 58: 1965-1971.
[0101] Agents used in proapoptotic regimens (e.g., bcl-2 antisense oligonucleotides) may also be used in the combination of the present invention. Members of the Bcl-2 family of proteins block apoptosis. Upregulation of bcl-2 has therefore been linked to chemoresistance. Studies have shown that the epidermal growth factor (EGF) stimulates anti-apoptotic members of the bcl-2 family (i.e., mcl-1). Therefore, strategies designed to downregulate the expression of bcl-2 in tumors have demonstrated clinical benefit and are now in Phase ll/lll trials, namely Genta’s G3139 bcl-2 antisense oligonucleotide. Such proapoptotic strategies using the antisense oligonucleotide strategy for bcl-2 are discussed in Water JS et al. (2000), J. Clin. Oncol. 18: 1812-1823; and Kitada S et al. (1994), Antisense Res. Dev. 4: 71-79.
[0102] Cell cycle signalling inhibitors inhibit molecules involved in the control of the cell cycle. A family of protein kinases called cyclin dependent kinases (CDKs) and their interaction with a family of proteins termed cyclins controls progression through the eukaryotic cell cycle. The coordinate activation and inactivation of different cyclin/CDK complexes is necessary for normal progression through the cell cycle. Several inhibitors of cell cycle signalling are underdevelopment. For instance, examples of cyclin dependent kinases, including CDK2, CDK4, and CDK6 and inhibitors for the same are described in, for instance, Rosania et al, Exp. Opin. Ther. Patents (2000) 10(2):215-230.
[0103] In one embodiment, the cancer treatment method of the claimed invention includes the co-administration a compound of formula (la) and/or a pharmaceutically acceptable salt, hydrate, solvate or pro-drug thereof and at least one anti-neoplastic agent, such as one selected from the group consisting of anti-microtubule agents, platinum coordination complexes, alkylating agents, antibiotic agents, topoisomerase II inhibitors, antimetabolites, topoisomerase I inhibitors, hormones and hormonal analogues, signal transduction pathway inhibitors, non-receptor tyrosine kinase angiogenesis inhibitors, immunotherapeutic agents, proapoptotic agents, and cell cycle signaling inhibitors.
[0104] The pharmaceutically active compound of the present invention is incorporated into convenient dosage forms such as capsules, tablets, or injectable preparations. Solid or liquid pharmaceutical carriers are employed. Solid carriers include, starch, lactose, calcium sulfate dihydrate, terra alba, sucrose, talc, gelatin, agar, pectin, acacia, magnesium stearate, and stearic acid. Liquid carriers include syrup, peanut oil, olive oil, saline, and water. Similarly, the carrier or diluent may include any prolonged release material, such as glyceryl monostearate or glyceryl distearate, alone or with a wax. The amount of solid carrier varies widely but, preferably, will be from about 25 mg to about 1 g per dosage unit. When a liquid carrier is used, the preparation will be in the form of a syrup, elixir, emulsion, soft gelatin capsule, sterile injectable liquid such as an ampoule, or an aqueous or nonaqueous liquid suspension.
[0105] The pharmaceutical preparations are made following conventional techniques of a pharmaceutical chemist involving mixing, granulating, and compressing, when necessary, for tablet forms, or mixing, filling and dissolving the ingredients, as appropriate, to give the desired oral or parenteral products.
[0106] Doses of the presently invented pharmaceutically active compound in a pharmaceutical dosage unit as described above will be an efficacious, nontoxic quantity preferably selected from the range of 0.001 -100 mg/kg of active compound, preferably 0.001 - 50 mg/kg. When treating a human patient in need of a PI3K inhibitor, the selected dose is administered preferably from 1-6 times daily, orally or parenterally. Preferred forms of parenteral administration include topically, rectally, transdermally, by injection and continuously by infusion. Oral dosage units for human administration preferably contain from 0.05 to 3500 mg of active compound. According to one embodiment, the oral dosagefor human administration contains 100 to 1000 mg per day. Oral administration, which uses lower dosages is preferred. Parenteral administration, at high dosages, however, also can be used when safe and convenient for the patient.
[0107] Optimal dosages to be administered may be readily determined by those skilled in the art, and will vary with the particular PI3 kinase inhibitor in use, the strength of the preparation, the mode of administration, and the advancement of the disease condition. Additional factors depending on the particular patient being treated will result in a need to adjust dosages, including patient age, weight, diet, and time of administration. Exemplary dosages include oral formulations equivalent to 10 mg, 25 mg, and 100 mg of the compound of formula (I), to be administered alone, in multiples, or in combination. Another exemplary dosage includes oral formulations of the tris(hydroxymethyl)aminomethane salt of 2-methyl-1-{[2-methyl-3-(trifluoromethyl)phenyl]methyl}-6-(4-morpholinyl)-1H-benzimidazole-4-carboxylic acid equivalent to 10 mg, 25 mg, or 100 mg of the free base of 2-methyl-1-{[2-methyl-3-(trifluoromethyl)phenyl]methyl}-6-(4-morpholinyl)-1 H-benzimidazole-4-carboxylic acid.
[0108] The method of this invention of inducing PI3 kinase inhibitory activity in mammals, including humans, comprises administering to a subject in need of such activity an effective PI3 kinase modulating/inhibiting amount of a pharmaceutically active compound of the present invention.
[0109] The invention also provides for the use of a compound of Formula (la) in the manufacture of a medicament for use in therapy.
[0110] The invention also provides for the use of a compound of Formula (la) in the manufacture of a medicament for use in treating autoimmune disorders, inflammatory diseases, cardiovascular diseases, neurodegenerative diseases, allergy, asthma, pancreatitis, multiorgan failure, kidney diseases, platelet aggregation, cancer, sperm motility, transplantation rejection, graft rejection and lung injuries.
[0111] The invention also provides for a pharmaceutical composition for use as a PI3 inhibitor which comprises a compound of Formula (la) and a pharmaceutically acceptable carrier.
[0112] The invention also provides for a pharmaceutical composition for use in the treatment of autoimmune disorders, inflammatory diseases, cardiovascular diseases, neurodegenerative diseases, allergy, asthma, pancreatitis, multiorgan failure, kidney diseases, platelet aggregation, cancer, sperm motility, transplantation rejection, graft rejection and lung injuries, which comprises a compound of Formula (la) and a pharmaceutically acceptable carrier.
[0113] In addition, the pharmaceutically active compounds of the present invention can be co-administered with further active ingredients, including compounds known to have utility when used in combination with a PI3 kinase inhibitor.
[0114] Without further elaboration, it is believed that one skilled in the art can, using the preceding description, utilize the present invention to its fullest extent. The following examples are, therefore, to be construed as merely illustrative and not a limitation of the scope of the present invention in any way.
Experimental Procedures [0115] Compounds of Formula (I) may be prepared using the general schemes I -VII, as described below.
Scheme I (R2 = Me)
[0116] 2,6-Dinitroaniline 1 can be brominated with bromine in acetic acid to provide 4-bromo-2,6-dinitroaniline 2 that can be reduced to the di-amino nitro benzene 3 with (NH4+)2S. Subsequent reaction of 3 with 2,4-pentanedione in the presence of strong acid at reflux temperatures, in an alcoholic solvent, affords nitrobenzimidazole 4. Alkylation to afford substituted benzimidazole 5 can be accomplished with a suitably substituted alkyl halide with a base, such as K2C03, in a polar aprotic solvent, such as DMF. Palladium-catalyzed displacement of the aromatic bromine with morpholine can then afford substituted nitrobenzimidazole 6 which can then be reduced to the amino benzimidazole 7. Amino benzimidazole 7 can then be converted into hydroxyl analog 8, sulfonamide 9, amide 10, and halo analog 11, using standard organic manipulations.
Scheme II
[0117] 2,6-Dinitroaniline 1 can be brominated with bromine in acetic acid to provide 4-bromo-2,6-dinitroaniline 2 that can be reduced to the di-amino nitro benzene 3 with (NH4+)2S. Subsequent reaction of 3 with a carboxylic acid in the presence of strong acid at elevated temperatures affords nitrobenzimidazole 4. Alkylation to afford substituted benzimidazole 5 can be accomplished with a suitably substituted alkyl halide with a base, such as K2COs, in a polar aprotic solvent, such as DMF. Palladium-catalyzed displacement of the aromatic bromine with morpholine can then afford substituted nitrobenzimidazole 6 which can then be reduced to the amino benzimidazole 7. Amino benzimidazole 7 can then be converted into hydroxyl analog 8, sulfonamide 9, amide 10, and halo analog 11, using standard organic manipulations.
Scheme III (R1 = OMe; R2 = Me)
[0118] 2-Amino-3-nitrophenol 1 can be methylated with Mel and K2C03 in DMF to afford methoxy nitro aniline 2. Bromination, with bromine in acetic acid, followed by acetylation with acetic anhydride in acetic acid and sulfuric acid, can provide intermediate 4. Palladium-catalyzed displacement of the aromatic bromide with morpholine can then afford intermediate 5. Iron-induced nitro reduction followed by ring closure can then provide benzimidazole 6 that can be alkylated with a suitably substituted alkyl bromide using a base, such as K2C03, in a polar aprotic solvent such as DMF, to afford final products 7.
Scheme IV
[0119] Aminobenzimidazole 1 can be converted to bromobenzimidazole 2 using sodium nitrite with NaBr in aqueous HBr. Palladium catalyzed coupling with an aryl boronic acid in the presence of a suitable phosphine with an inorganic base in a polar non-protic solvent can then provide final substituted benzimidazoles 3. Het includes 2-, 3- furanyls, and 1,3-thiozols.
Scheme V
[0120] Palladium catalyzed carbonylaton of bromo-benzimidazole 1 can be accomplished by bubbling carbon-monoxide gas in methanol with triethylamine to provide methyl ester 2. Ester hydrolysis can then be accomplished with lithium hydroxide in THF/water to provide final product benzimidazole acid 3.
Scheme VI
[0121] Palladium catalyzed cyanation of bromo-benzimidazole 1 can be accomplished with zinc cyanide in DMF to provide benzimidazole nitrile 2. The nitrile can be converted to the primary carboxamide with KOH and peroxide in THF to provide amide 3. Treatment of the carboxamide 3 with DMF-DMA can provide intermediate 4 that can then be cyclized to triazole analogs 5 with hydrazine in acetic acid.
Scheme VII
[0122] Amination of 5-chloro-2-nitrobenzoic acid with O-methyl hydroxyl amine and t-butoxide in the presence of copper acetate can provide 3-amino-5-chloro-2-nitrobenzoic acid 2. Esterification can be accomplished with methanol and sulfuric acid to provide methyl ester 3 that can be reacted with morpholine in DMF with K2C03 to provide phenyl morpholine analog 4. Nitro reduction can be accomplished using a variety of metal reductions to provide diamine 5. Condensation of 5 with a variety of carboxylic acids can provide benzimidazole methylester 6 that can be further converted to final products 7 (R1 = C02Me, C02H, CONH2, CN, triazole, tetrazole) after alkylation with an alkyl halide, followed by standard organic manipulations as previously described.
Reference Example A
[0123]
Preparation of methyl 2-methyl-6-(4-morpholinyl)-1-(1-naphthalenylmethyl)-1 H-benzimidazole-4-carboxylate a) 3-amino-5-chloro-2-nitrobenzoic acid [0124]
[0125] Under nitrogen, to a solution of t-BuOK (156.8 g) and Cu(OAc)2 (3.6 g) in DMF (1.2 L) was added a solution of 5-chloro-2-nitrobenzoic acid (40.0 g) and MeONH2HCI (33.2 g) in DMF (300 mL) at 0° C. After 3h the reaction was quenched by addition of H20 (2.5 L) and acidified with 10% HCI solution to pH= I.The mixture was extracted with EA (2 L X 2) and the combined organic layers were then washed with brine, dried over anhydrous Na2S04, filtered and concentrated in-vacuo to afford the crude product as a yellow solid (43.2g, yield 100%). 1H NMR (300 MHz, CDCI3): δ ppm 6.88 (s, 1H, J= 2.4Hz), 6.91 (d, 1H, J= 2.4Hz), 8.08 (br s, 2H); LC-MS: m/e = 217 [M+1]+. b) methyl 3-amino-5-chloro-2-nitrobenzoate [0126]
[0127] A mixture of 3-amino-5-chloro-2-nitrobenzoic acid (43.2 g) and HATU (2-(1 H-7-Azabenzotriazol-1-yl)-1,1,3,3-tetramethyl uranium hexafluorophosphate Methanaminium, commercially available) (76 g) in MeOH (81 mL), Et3N (83 mL) and THF (300 mL) was stirred at room temperature for 3h. When TLC showed no starting material, the solvent was removed in-vacuo and the residue was then diluted with EtOAc (2 L). It was then washed with brine (1 Lx3) and dried over anhydrous Na2S04, filtered and concentrated in-vacuo. The residue was then purified by silica gel chromatography eluted with EtOAc : petroleum ether = 1 : 8 to afford the desired product as a yellow solid (29.5 g, yield 64%). 1H NMR (300 MHz, CDCI3): δ ppm 3.90 (s, 3H,s), 5.85 (brs, 2H), 6.80 (d, 1H, J =2.4 Hz), 6.90 (d, 1H, J = 2.4 Hz); LC-MS: m/e = 231 [M+1]+. c) methyl 3-amino-5-(4-morpholinyl)-2-nitrobenzoate [0128]
[0129] A mixture of combined batches of methyl 3-amino-5-chloro-2-nitrobenzoate (39 g), morpholine (29.5 g) and K2COs (47 g) was stirred in DMF (200ml) at 110 °C for 5 h. The mixture was cooled to room temperature and poured into water (1 L). It was extracted with EtOAc (500 mL x 3). The combined organic layers were washed with brine, dried over anhydrous Na2S04, filtered and concentrated in-vacuo to afford the desired product as a yellow solid (22 g, yield 46%). 1H NMR (300 MHz, CDCI3): δ ppm 3.31 (t, 4H, J= 4.8 Hz), 3.82 (t, 4H, J= 4.8 Hz), 3.89 (s, 3H), 6.03 (d, 1H, J= 2.4 Hz), 6.34 (d, 1H, J= 2.4 Hz); LC-MS: m/e = 282 [M+1]+. d) methyl 2-methyl-5-(4-morpholinyl)-1 H-benzimidazole-7-carboxylate [0130]
[0131] To a solution of methyl 3-amino-5-(4-morpholinyl)-2-nitrobenzoate (22 g) stirring at reflux in HOAc (400 mL) was added iron powder in portions (13 g). After the addition, the mixture was stirred at reflux for 5 h. It was cooled to room temperature and the solvent was removed in-vacuo. The residue was neutralized with aqueous Na2COs solution (1 L). It was extracted with EtOAc (500 mL X3). The combined organic layers were then concentrated in-vacuo and the residue was purified by silica gel chromatography eluted with MeOH : DCM = 1 : 30 to afford the desired product as a solid (16.6 g, yield 77%). 1H NMR (300 MHz, CDCI3): δ ppm 2.67 (s, 3H), 3.17 (t, 4H, J= 4.8 Hz), 3.90 (t, 4H, J= 4.8 Hz), 3.98 (s, 3H), 7.44 (d, 1H, J= 1.8 Hz), 7.54 (d, 1H, J= 1.8 Hz); LC-MS: m/e = 276 [M+1]+. e) methyl 2-methyl-6-(4-morpholinyl)-1-(1-naphthalenylmethyl)-1H-benzimidazole-4-carboxylate [0132] A mixture of methyl 2-methyl-5-(4-morpholinyl)-1 H-benzimidazole-7-carboxylate (4.125 g), 1-(bromome-thyl)naphthalene (5 g) and K2C03 (6.2 g) was stirred at 80° C for 3 h. When TLC showed no starting material remaining, the mixture was cooled to room temperature and then poured into water (500 mL). It was extracted with EtOAc (500 mL X 3) and the combined organic layers were washed with brine (500 mL X 3) and then concentrated in-vacuo. The residue was purified by silica gel chromatography eluted with MeOH : DCM= 1 : 100 to afford the desired product as a yellow solid (4.6 g, 74%). 1H NMR (300 MHz, DMSO-d6): δ ppm 2.42 (s, 3H), 3.04 (t, 4H, J= 4.8 Hz), 3.68 (t, 4H, J= 4.8 Hz), 3.90 (s, 3H,), 6.02 (s, 1H), 6.28 (d, 1H, J= 7.5 Hz), 7.29 (d, 1H, J=2.4 Hz), 7.32 (d, 1H, J= 7.5 Hz), 7.39 (d, 1H, J=2.4), 7.60-7.71 (m, 2H), 7.84 (d, 1H, J= 8.4 Hz), 8.01 (d, 1H, J= 7.5 Hz), 8.24 (d, 1H, J= 7.5 Hz); LC-MS: m/e = 416 [M+1]+.
Reference Example B
[0133]
Preparation of methyl 2-methyl-1-{[2-methyl-3-(trifluoromethyl)phenvl1methyl}-6-(4-morpholinyl-1 H-benzimidazole-4- carboxylate [0134] A solution of methyl 2-methyl-5-(4-morpholinyl)-1H-benzimidazole-7-carboxylate prepared as described in Reference Example A step d (500mg, 1.8 mmol), 1-(bromomethyl)-2-methyl-3-(trifluoromethyl)benzene (483 mg, 1.9 mmol) and K2COs (497 mg, 3.6 mmol) in DMF (50 mL) was stirred at 80° C for 3 h. The reaction mixture was cooled to rt and poured into water (50 mL), extracted with EtOAc (30 mL x 3). The combined organic layers were washed with brine, dried over Na2S04 and concentrated. The resulting residue was purified by silica gel chromatography eluted with DCM : MeOH = 50 : 1 to give the crude product (230 mg, yield 29%), as a white solid. 1H NMR (300 MHz, DMSO-d6): δ ppm 2.39 (s, 3H), 2.54 (s, 3H), 3.08 (t, 4H, J=4.8 Hz), 3.72 (t, 4H, J=4.8 Hz), 3.89 (s, 3H), 5.57 (s, 2H), 6.27 (d, 1H, J=7.5 Hz), 7.22 (t, 1H, J=7.5 Hz), 7.27(d, 1H, J=2.4 Hz), 7.38 (d, 1H, J=2.4 Hz) 7.60 (d, 1H, J=7.5 Hz); LC-MS: m/e = 448 [M+1]+
Example 1 [0135]
Preparation of 2-methyl-1-{[2-methyl-3-(trifluoromethyl)phenyl]methyl}-6-(4-morpholinvn-1H-benzimidazole-4-carboxy- lie acid [0136] An aqueous solution of 2 N LiOH (1.2 mL) was added to a solution of methyl 2-methyl-1-{[2-methyl-3-(trifluor-omethyl)phenyl]methyl}-6-(4-morpholinyl)-1H-benzimidazole-4-carboxylate, prepared as described in Reference Example B (180 mg, 0.4 mmol) in THF (10 mL) and stirred at 50° C for 1 h. When TLC showed no starting material remaining, the mixture was cooled to rt and THF was removed under reduced pressure. The pH of the mixture was acidified to pH 3. The suspension was filtered and the filtrate was collected, and washed with water (10mL) to give the product as a white solid (152 mg, yield 88%). 1H NMR (300 MHz, DMSO-d6): δ ppm 2.46 (s, 3H), 2.54 (s, 3H), 3.10 (t, 4H, J=4.8 Hz), 3.73 (t, 4H, J=4.8 Hz), 5.63 (s, 2H), 6.37 (d, 1H, J=7.8 Hz), 7.26 (t, 1H, J=7.8 Hz), 7.35 (d, 1H, J=2.4 Hz), 7.44 (d, 1H, J=2.4 Hz), 7.62 (d, 1H, J=7.8 Hz); LC-MS: m/e = 434 [M+1]+.
Example 2 [0137]
Preparation of 2-methyl-1-{[2-methyl-3-(trifluoromethyl)phenyl]methyl}-6-(4-morpholinyl-1H-benzimidazole-4-carboxylic acid 2-amino-2-(hydroxymethyl)-1,3-propanediol salt [0138] Seed crystal preparation - Batch 1: To the 2-methyl-1-{[2-methyl-3-(trifluoromethyl)phenyl]methyl}-6-(4-mor-pholinyl)-1H-benzimidazole-4-carboxylic acid (52.9 mg, 0.122 mmol), methanol (2.0 mL) was added. To the slurry, tromethamine (2-amino-2-(hydroxymethyl)-1,3-propanediol) (3.0 M solution in water, 1.0 equivalent) was added. The slurry was heated to 60C and kept stirring at 60C for 3 hours. The slurry was then cooled slowly (0.1 C/min) to20C. Once the temperature of the slurry reached 20C, the slurry was kept stirring at 20C for 8 hours. The crystalline solids were isolated by vacuum filtration. The yield of the desired salt was 57.2 mg (85% yield).
[0139] Seed crystal preparation - Batch 2: To the 2-methyl-1-{[2-methyl-3-(trifluoromethyl)phenyl]methyl}-6-(4-mor-pholinyl)-1 H-benzimidazole-4-carboxylic acid (353.0 mg), methanol (14.0 mL) was added. The slurry was heated to 60C and tromethamine (3.0 M solution in water, 1.0 equivalent) was added in four aliquots over 15 minutes followed by the addition of crystalline seeds of crystalline tromethamine salt from batch 1. The slurry was stirred at 60C for 3 hours, cooled (1 C/min) to 20C, and stirred at 20C for 8 hours. The solids were isolated by vacuum filtration, dried at 60C under vacuum for 5 hours. The yield of the tromethamine salt was 401.5 mg (-88.9% yield).
[0140] Batch 3: 2-methyl-1-{[2-methyl-3-(trifluoromethyl)phenyl]methyl}-6-(4-morpholinyl)-1H-benzimidazole-4-car-boxylic acid (40.0 g, 92 mmol) was suspended in Methanol (1.6 L) in a 3L rounded-bottom flask. The resulting slurry was heated to 60°C mixing on a buchii rotary evaporator water bath and tris(hydroxymethyl)aminomethane (3M solution in water) (0.031 L, 92 mmol) was added in four aliquots over 15 minutes followed by the addition of seed crystals as produced by method analogous to Example 86, Batch 2, above (108 mg). This slurry was stirred (flask rotated on buchii rotovap) at 60 °C for 3 hours, then cooled (-1 °C/min) to 20 °C (room temperature), then finally magnetically stirred at 20 °C (room temperature) for 8 hours. The resulting white solid was isolated by vacuum filtration, dried under vacuum at 60 °C for 8 hours to provide 2-methyl-1-{[2-methyl-3-(trifluoromethyl)phenyl]methyl}-6-(4-morpholinyl)-1 H-benzimida-zole-4-carboxylic acid - 2-amino-2-(hydroxymethyl)-1,3-propanediol (1:1) (47.76 g, 86 mmol, 93 % yield) as a white solid. Both proton NMR and LCMS are consistent with the proposed structure. 1H NMR (400 MHz, DMSO-d6) δ ppm 7.61 (d, J=7.83 Hz, 1 H) 7.37 (d, J=2.27 Hz, 1 H) 7.17 - 7.33 (m, 2 H) 6.33 (d, J=7.83 Hz, 1 H) 5.59 (s, 2 H) 3.66 - 3.80 (m, 4 H) 2.98 - 3.15 (m, 4 H) 2.50 - 2.58 (m, 10 H) 2.43 (s, 3 H); LCMS m/z MH+ =434.3.
Biological Assays [0141] The compound of the present invention was tested according to the following assays and found to be an inhibitor of PI3 kinases, particularly ΡΙ3Κβ. The activities (IC50) of exemplified compoundswere under 10 nM. The IC50 value can be converted and presented as plC50 value. HTRF In vitro Profiling Assays for PI3K Inhibition [0142] The PI3-Kinase profiling assays were developed to measure the compound-dependent inhibition of the alpha, beta, delta, and gamma isoforms of PI3Kin an In vitro catalytic assay. This assay was developed and optimized from a kit produced by Upstate (Millipore catalog # 33-017). Briefly, this procedure utilizes a pre-formed HTRF (Homogeneous Time-Resolved Fluorescence energy transfer) complex between four binding partners: 1) biotinylated PIP3, 2) GST tagged pleckstrin homology (PH) domain, 3) Europium labeled anti-GST monoclonal antibody, and 4) Streptavidin-Allophycocyanin (APC). The native PIP3 produced by PI 3-Kinase activity displaces biotin-PIP3 from the PH domain, resulting in the dissociation of the HTRF complex and a decrease in the fluorescence signal. The format of this assay is the same for all 4 isoforms of PI3K; the differences lie in the concentration of enzyme used to achieve the most robust signal. The alpha and delta assays are run at 400pM enzyme; the beta assay is at 200pM enzyme and the gamma assay is run at InM enzyme. In addition, the alpha, beta and delta assays are run with 150mM NaCI while the gamma assay is run in the absence of NaCI. The ATP concentration is 100uM in the alpha, beta, and delta assays and 15uM ATP in the gamma assay. All reactions are run at 10uM PIP2 [0143] Compounds were serially diluted (3-fold in 100% DMSO) across a 384-well polypropylene mother plate from column 1 to column 12 and column 13 to column 24, to yield 11 concentrations for each test compound. Columns 6 and 18 contain only DMSO. Once titrations were made, Ο.Οδμί. was transferred to a 384-well low-volume assay plate (Greiner 784076). This assay plate contained three pharmacological controls (known PI3K inhibitors) and 3 assay controls: (1) Enzyme without inhibitor; (2) Buffer minus enzyme, and (3) Buffer minus enzyme plus native PIP3. DMSO was stamped into all wells of columns 6 and 18. PIP3 was added at 40 μΜ in 1X Reaction buffer (1μΙ_ of 200 μΜ PIP3) to alternating rows of column 18 (wells 18 B, D, F, H, J, L, N, P). The no-enzyme control reactions were run in wells 18 A, C, E, G, I, K, M, O(0.VLof 100% DMSO).
[0144] The PI3-Kinase profiling assay was optimized using the HTRF kit provided by Upstate (Millipore). The assay kit contained seven reagents: 1) 4X Reaction Buffer; 2) native PIP2 (substrate); 3) Stop A (EDTA); 4) Stop B (Biotin-PIP3); 5) Detection Mix A (Streptavidin-APC); 6) Detection Mix B (Eu-labeled Anti-GST plus GST-tagged PH-domain); 7) Detection Mix C (KF). In addition, the following items were obtained or purchased: PI3Kinase (prepared by GSK BR&amp;AD), dithiothreitol (Sigma, D-5545), Adenosine-5’-triphosphate (ATP, Teknova cat. # A0220), native PIP3 (1,2-dioctanoyl-sn-glycero-3-[phosphoinositil-3,4,5-triphosphate] tetraammonium salt (Avanti polar lipids, 850186P), DMSO (Sigma, 472301).
[0145] PI3Kinase Reaction Buffer was prepared by diluting the stock 1:4 with de-ionized water. Freshly prepared DTT was added at a final concentration of 5 mM on the day of use. Enzyme addition and compound pre-incubation were initiated by the addition of 2.5μΙ_ of PI3K (at twice its final concentration) in 1X reaction buffer to all wells using a Multidrop Combi. Plates were incubated at room temperature for 15 minutes. Reactions were initiated by addition of 2.5μί_ of 2X substrate solution (PIP2 and ATP in 1X reaction buffer) using a Multidrop Combi. Plates were incubated at room temperature for one hour. Reactions were quenched by the addition of 2.5μί_ of stop solution (Stop A and Stop B pre-mixed at a ratio of 5:1, respectively) to all wells using the Multidrop Combi. The quenched reactions were then processed to detect product formation by adding 2.5μΙ_ of Detection Solution to all wells using the Mulitdrop Combi (Detection mix C, Detection mix A, and Detection mix B combined together in an 18:1:1 ratio, i.e.: for a 6000 μΙ_ total volume, mix 5400 μΙ_ Detection mix C, 300μΙ_ Detection mix A, and 300 μι. Detection mix B. Note: this solution should be prepared 2 hours prior to use). Following a one hour incubation in the dark, the FITRF signal was measured on the Envision plate reader set for 330nm excitation and dual emission detection at 620nm (Eu) and 665nm (APC).
[0146] The loss of the FITRF signal is due to the displacement of biotinylated-PIP3 from the PFI domain by the PI3K-dependent conversion of PIP2 to PIP3. This loss of signal is nonlinear with respect to both increasing product and time. This non-linear detection will impact accuracy of IC50 calculations; therefore, there is a need for a correction factor to obtain more accurate IC50 values. This correction is derived from the assay standards in the wells of column 6 and 18 of the assay plate.
[0147] All data were calculated using the ratio of acceptor (APC) to donor (Europium) fluorescence in each well of the assay plate. The percent inhibition for each compound concentration was calculated as follows: %inhibition = 100*(flu-orescence ratio - CtrlB)/(CtrlA - CtrIB) where CtrlA= (-) PI3Kinase reaction and CrtlB= PI3Kinase + DMSO.
[0148] An IC50 was then calculated fitting the %inhibition data to the equation: %inhibition = min + (max-min)/(1 + ([inhibitor]/IC50)An) where min is the %inhibition with no inhibitor (typically 0%), max is the signal in the (-) Enzyme control, and n is the Hill slope (typically 1). Finally, the IC50 was converted to plC50 (plC50 = -log(IC50)), and the plC50 value was corrected by using plate controls and the equation below: plC50 (corrected) = plC50 (observed) + log10((CtrlA-CtrlB)/(CtrlB-CtrlC)), where CtrlA and CtrIB are as defined above and CrtlC= 10μΜ PI(3,4,5)P3, 100% displacement of biotinylated PI(3,4,5)P3.
[0149] The compound listed in Table 1 was tested generally according to the assays described herein. Table 1 lists the plC50 values for either an experimental run or an average of two or more experimental runs with the examples shown.
Table 1
Cellular Assays - Cell Growth Inhibition Assay in PTEN wild-type or PTEN deficient tumor cell lines [0150] Twenty-two Phosphatase and Tensin Flomolog (PTEN) wild-type or PTEN deficient tumor cell lines were cultured generally according to instructions supplied by cell culture supplier American Type Culture Collection, Manassas, VA, with 10% fetal bovine serum at 5% C02 and 37°C. Cells were seeded into either a T-75 or a T-175 flask 3-4 days prior to 96-well assay plating such that the flasks were approximately 70-80% confluent of the time of harvest. Cells were harvested using 0.25% trypsin-EDTA (Invitrogen #25200056). Trypan Blue exclusion staining was used to determine cell number.
[0151] Viable cells were plated in clear, flat bottom 96-well plates (BD #353075) under anchorage independent conditions at 2,000-10,000 cells per well depending on the cell line. To generate anchorage independent growth conditions, a 5% agar stock solution in water was made and autoclaved to melt and sterilize. From the 5% agar solution, a 0.6% agar/media + 10% fetal bovine serum (FBS) solution was made to generate a bottom agar layer in the plates to prevent cell attachment. Seventy five microliters per well of the 0.6% agar-media solution was added to the plates. After solidification, a cell solution of 266,870 to 1,334,022 cells (depending on the cell line) in 10 ml of 0.3% agar/ media + 10% FBS was made and 75 μΙ of the cell/media/agar suspension was added to the plates. After the cell layer solidified, 50 μΙ of media + 10% FBS was added to the top of the cells. A 0.3% Brij 35 (Sigma B4184) solution in media + 10% FBS was added to column 12 as a background subtraction control. The cells were incubated overnight at 5% C02 and 37°C. The next day one plate of cells was processed at the time of compound addition to quantify the starting number of cells (T = 0 or TO).
[0152] To generate the compound titration plates, 15 μΙ of a 2 mM or 20 μΙ of a 20 mM solution of the compound of example 1 was diluted in clear bottom polypropylene 96-well plate (BD #351190) using a 10 point, 3-fold titration or a 20 point 2-fold titration, respectively. Three hundred microliters of media was added to the compound dilutions. Ten microliters per well of the serial dilutions was added to the cells and the plates incubated for 6 days at 5% C02 and 37°C. The final concentration of DMSO in all wells was 0.15% and the highest final concentration of the compound of example 1 was 3.7 μΜ or 30.7 μΜ.
[0153] Following the 6-day incubation, 20 μΙ of Alamar Blue (Invitrogen #DAL1100) was added to the cells, incubated at 5% C02 and 37°C for 6 hours and the plates read on a Spectramax (Gemini EM) at 530nm (excitation) and 590nm (emission) with the auto cutoff disabled. For analysis of cell growth inhibition dose response curves, the data was plotted as the percent of the DMSO-treated control samples (DMSO samples set to 100%). The cellular response was determined for the compound of example 1 and control compounds by fitting the concentration response with a 4 parameter curve fit using XLfit software and determining the concentration that inhibits 50% of the Ymax-Ymin window (EC50). The EC50 is the mid point of active compound effect window (between Ymax plateau and Ymin plateau of compound) and represents the concentration of the compound of example 1 where 50% of its maximal effect is observed. Values from wells containing 0.3% Brij 35 (under anchorage independent conditions) were subtracted from all samples for background correction.
[0154] The results shown in Table 2 demonstrate that multiple cell lines with loss of the tumor supressor PTEN were sensitive, while relatively few wild-type PTEN tumor cell lines were sensitive.
Table 2
In Vivo Experiments
Dose dependent tumor inhibition [0155] The activity of the compound of example 1 was evaluated in vivo against PC-3 (prostate carcinoma cell line encoding a deficient PTEN protein) xenograft mouse model. The PC-3 tumor bearing mice were generated by injecting 2.5 x 106 PC-3 cells suspended 1:1 in Matrigel subcutaneously in the flank of female nude mice (Charles River -Wilmington; strain Crl: CD-1-Foxn1). One set of mice, each approximately 19 weeks of age, were implanted with the cells for the 100,30, and 10 mg/kg doses and another set of mice, each approximately 11 weeks of age, were implanted with the cells for the 10, 3, and 1 mg/kg doses.
[0156] Mice bearing PC-3 xenografts were randomized into dosing groups of n=8 based on tumor volume 29 (100, 30, and 10 mg/kg) or 28 (1,3,10 mg/kg) days after tumor cells were implanted. Treatment of mice commenced the next day and continued for 21 days. Mice received once daily oral gavage with compound or vehicle at 10 mL/kg.
[0157] Tumor growth was measured twice weekly in two dimensions with vernier callipers; the longest dimension was defined as the length (1), and the width (w) was measured perpendicular to the length. Tumor volumes (V) were calculated using the following equation: V = (Vfc)1 w2. Means of the tumor volumes were used to compare treatment groups. Stable disease for this study is defined as a tumor volume which during the course of compound treatment does not substantially increase or decrease but stays similar to the volume prior to drug treatment compared to vehicle treated in which the tumor volume continues to increase during the course of the study. Tumor growth delay is defined as tumor volume that is reduced during the course of the compound treatment relative to vehicle treated tumor volume.
[0158] The results demonstrated that treatment of female nude mice bearing PC-3 prostate xenografts with 10, 30, and 100 mg/kg the compound of example 1 for 21 days resulted in stable disease with the 1 and 3 mg/kg doses resulting in tumor growth delay relative to vehicle during the dosing period. B) Pharmacodynamic effects [0159] The activity of the compound of example 1 was evaluated in vivo against PC-3 (prostate carcinoma cell line encoding a deficient PTEN protein) xenograft mouse model. Female nude mice (Charles River Laboratories, Wilmington, DE; strain CD-1-Foxn1, ~6 weeks of age) were injected subcutaneously with 2 million PC-3 (human prostate carcinoma) cells mixed 1:1 with Matrigel in the flank. Tumors were allowed to grow for approximately 5 weeks.
[0160] Mice bearing PC-3 xenografts were administered 3 mg/kg of the compound of example 1 or 10 mg/kg of the compound of example 1 and euthanized using carbon dioxide after 1, 2, 4, 6, 8, 10, and 24 hours (n=3 mice/treat-ment/timepoint); an additional 3 mice bearing PC-3 xenografts were administered vehicle and euthanized after 2 hours. The tumor was excised. Half of each tumor was immediately processed by Medicon (BD Catalog # 340592) in 1 mL Meso-Scale Discovery (MSD) lysis bufFerwith protease inhibitors (Roche complete protease cocktail, cat# 04 693 116 001) and phosphatase inhibitors (Sigma, cat# P2850 and P-5726) for 30-60 seconds and transferred to 1.5 mL Eppendorf tubes. Tubes remained on wet ice until they were centrifuged for 10 minutes at 4°C at maximum speed in a tabletop refrigerated centrifuge.
[0161] Tumor lysates were serially diluted in 96-well polypropylene plates on wet ice. Lysates (150 μί) were loaded in row 1; rows 2-12 were loaded with 75 μί of complete Meso Scale Discovery (MSD) lysis buffer (supplied in MSD kit; # K15100D-3). Samples were serially diluted 2-fold across the plate by sequential transfer of 75 μί through well 11; row 12 contained lysis buffer only. MSD Multi-Spot assay plates (whole cell lysate kit: Phospho(ser473),Total AKT Assay, catalog # K15100D-3) were blocked with 150 μί of 3% Blocker A overnight at 4°C with shaking before being washed 4Xwith 200 μί MSD Triswash buffer. Fifty microliters of the serially diluted lysates were pipetted onto the blocked MSD plates, covered, and incubated overnight at 4°C with shaking. Plates were washed with Tris buffer as before. Detection antibody was added (25 μίΛ/νβΙΙ) at a final concentration of 10 nM in 1 mL Blocker A and 2 mLTris wash buffer and incubated for 1 hour at room temperature with shaking. Plates were washed as described above, before the addition of 150 μί of MSD read buffer and read immediately on a 6000 MSD plate reader. All work was performed in accordance with Institutional Animal Care and Use Committee (IACUC) protocols PA0079 and PA0271.
[0162] The non-lysate controls in column 12 were averaged and used as background to subtract from all wells. P/T AKT was calculated as shown: (phospho AKT(Ser473) signal)/[(phospho AKT(Ser473) signal) + (total AKT signal)]. Values from three points in each row of diluted samples identified as being in the linear range of detection were averaged to represent each tumor sample’s P/T AKT value. Averages and standard deviations of the P/T AKT value for each group of 3 mice were determined. Percent inhibition was calculated for each group as follows: 100-[(sample P/T AKT value)/(vehicle P/T AKT value)]*100.
[0163] The the compound of example 1 exhibited dose dependent inhibition of the pharmacodynamic marker pAKT (pAKT/tAKT).
Additional references: [0164] The compounds of the present invention can also be tested to determine their inhibitory activity at ΡΙ3Κα, PI3K6 ΡΙ3Κβ and ΡΙ3Κγ according to international patent publication No. W02009/039140.
[0165] The pharmaceutically active compounds within the scope of this invention are useful as PI3 Kinase inhibitors in mammals, particularly humans, in need thereof.
[0166] Disclosed is a method of treating diseases associated with PI3 kinase inhibition, particularly: autoimmune disorders, inflammatory diseases, cardiovascular diseases, neurodegenerative diseases, allergy, asthma, pancreatitis, multiorgan failure, kidney diseases, platelet aggregation, cancer, sperm motility, transplantation rejection, graft rejection and lung injuries and other conditions requiring PI3 kinase modulation/inhibition, which comprises administering an effective compound of Formula (I) or a pharmaceutically acceptable salt, hydrate, solvate or pro-drug thereof. The compounds of Formula (I) also provide for a method of treating the above indicated disease states because of their ability to act as PI3 inhibitors. The drug may be administered to a patient in need thereof by any conventional route of administration, including, but not limited to, intravenous, intramuscular, oral, subcutaneous, intradermal, and parenteral.
Claims 1. A compound of Formula (la)
which is 2-methyl-1-{[2-methyl-3-(trifluoromethyl)phenyl]methyl}-6-(4-morpholinyl) 1 H-benzimidazole-4-carboxylic acid, or a pharmaceutically acceptable salt thereof. 2. A compound according to claim 1 of Formula (la)
which is 2-methyl-1-{[2-methyl-3-(trifluoromethyl)phenyl]methyl}-6-(4-morpholinyl) 1 H-benzimidazole-4-carboxylic acid. 3. A pharmaceutically acceptable salt of the compound of formula (la) according to claim 1. 4. A pharmaceutically acceptable salt of the compound of formula (la) according to claim 3, wherein the salt is the 1:1 Tris salt. 5. A pharmaceutical composition which comprises a compound of Formula (la) or a pharmaceutically acceptable salt thereof according to any of claims 1-4 and a pharmaceutically acceptable carrier. 6. A pharmaceutical composition according to claim 5 for use in the treatment of a susceptible neoplasm in a mammal in need thereof. 7. A compound of Formula (la) or a pharmaceutically acceptable salt thereof according to any of claims 1-4 for use in therapy. 8. A combination of (a) 2-methyl-1-{[2-methyl-3-(trifluoromethyl)phenyl]methyl}-6-(4-morpholinyl)-1 H-benzimidazole-4-carboxylic acid or a pharmaceutically acceptable salt thereof and (b) at least one anti-neoplastic agent; for use in the treatment of cancer. 9. A compound of Formula (la) or a pharmaceutically acceptable salt thereof according to any of claims 1-4, or a pharmaceutical composition according to claim 5, or a combination according to claim 8, for use in the treatment of a PTEN-deficient susceptible neoplasm selected from brain (gliomas), glioblastomas, leukemias, Bannayan-Zonana syndrome, Cowden disease, Lhermitte-Duclos disease, breast cancer, inflammatory breast cancer, colorectal cancer, Wilm’s tumor, Ewing’s sarcoma, Rhabdomyosarcoma, ependymoma, medulloblastoma, colon cancer, head and neck cancer, kidney cancer, lung cancer, liver cancer, melanoma, squamous cell carcinoma, ovarian cancer, pancreatic cancer, prostate cancer, sarcoma, cancer, osteosarcoma, giant cell tumor of bone, thyroid cancer, lym-phoblasticT cell, leukemia, chronic myelogenous leukemia, chronic lymphocytic leukemia, hairy-cell leukemia, acute lymphoblastic leukemia, acute myelogenous leukemia, chronic neutrophilic leukemia, acute lymphoblastic T cell leukemia, Plasmacytoma, Immunoblastic large cell leukemia, Mantle cell leukemia, Multiple myeloma, Megakary-oblastic leukemia, Acute megakaryocytic leukemia, promyelocytic leukemia, Erythroleukemia, malignant lymphoma, hodgkins lymphoma, non-hodgkins lymphoma, lymphoblastic T cell lymphoma, Burkitt’s lymphoma, follicular lymphoma, neuroblastoma, bladder cancer, urothelial cancer, vulval cancer, cervical cancer, endometrial cancer, renal cancer, mesothelioma, esophageal cancer, salivary gland cancer, hepatocellular cancer, gastric cancer, nasopha-rangeal cancer, buccal cancer, cancer of the mouth, GIST (gastrointestinal stromal tumor), and testicular cancer. 10. A compound of Formula (la) ora pharmaceutically acceptable salt thereof for use, ora pharmaceutical composition for use, or a combination for use, according to claim 9 wherein said PTEN-deficient neoplasm is selected from prostate cancer, non-small-cell lung cancer, endometrial cancer, gastric cancer, melanoma, head and neck cancer, breast cancer, including triple-negative breast cancer, and glioma. 11. A compound of Formula (la) ora pharmaceutically acceptable salt thereof for use, ora pharmaceutical composition for use, ora combination for use, according to claim 10 wherein said prostate cancer is hormone refractory prostate cancer. 12. Use of a compound of Formula (la), or a pharmaceutically acceptable salt thereof, according to any of claims 1-4 in the manufacture of a medicament for use in the treatment of a PTEN-deficient susceptible neoplasm which is selected from brain (gliomas), glioblastomas, leukemias, Bannayan-Zonana syndrome, Cowden disease, Lhermitte-Duclos disease, breast cancer, inflammatory breast cancer, colorectal cancer, Wilm’s tumor, Ewing’s sarcoma, Rhabdomyosarcoma, ependymoma, medulloblastoma, colon cancer, head and neck cancer, kidney cancer, lung cancer, liver cancer, melanoma, squamous cell carcinoma, ovarian cancer, pancreatic cancer, prostate cancer, sarcoma, cancer, osteosarcoma, giant cell tumor of bone, thyroid cancer, lymphoblastic T cell, leukemia, chronic myelogenous leukemia, chronic lymphocytic leukemia, hairy-cell leukemia, acute lymphoblastic leukemia, acute myelogenous leukemia, chronic neutrophilic leukemia, acute lymphoblastic T cell leukemia, Plasmacytoma, Immunoblastic large cell leukemia, Mantle cell leukemia, Multiple myeloma, Megakaryoblastic leukemia, Acute megakaryocytic leukemia, promyelocytic leukemia, Erythroleukemia, malignant lymphoma, hodgkins lymphoma, non-hodgkins lymphoma, lymphoblasticT cell lymphoma, Burkitt’s lymphoma, follicular lymphoma, neuroblastoma, bladder cancer, urothelial cancer, vulval cancer, cervical cancer, endometrial cancer, renal cancer, mesothelioma, esophageal cancer, salivary gland cancer, hepatocellular cancer, gastric cancer, nasopharangeal cancer, buccal cancer, cancer of the mouth, GIST (gastrointestinal stromal tumor), and testicular cancer. 13. Use according to claim 12, wherein said PTEN-deficient susceptible neoplasm is selected from prostate cancer, non-small-cell lung cancer, endometrial cancer, gastric cancer, melanoma, head and neck cancer, breast cancer, including triple-negative breast cancer, and glioma. 14. Use according to claim 13, wherein said prostate cancer is hormone refractory prostate cancer.
Patentansprüche 1. Eine Verbindung der Formel (la),
wobei es sich um 2-Methyl-1-{[2-methyl-3-(trifluormethyl)phenyl]methyl}-6-(4-morpholinyl)-1H-benzimidazol-4-car-bonsäure oder ein pharmazeutisch verträgliches Salz davon handelt. 2. Eine Verbindung gemäß Anspruch 1 der Formel (la),
wobei es sich um 2-Methyl-1-{[2-methyl-3-(trifluormethyl)phenyl]methyl}-6-(4-morpholinyl)-1H-benzimidazol-4-car-bonsäure handelt. 3. Ein pharmazeutisch verträgliches Salz der Verbindung der Formel (la) gemäß Anspruch 1. 4. Ein pharmazeutisch verträgliches Salz der Verbindung der Formel (la) gemäß Anspruch 3, wobei das Salz das 1:1 Trissalz ist. 5. Ein Arzneimittel, umfassend eine Verbindung der Formel (la) oder ein pharmazeutisch verträgliches Salz davon gemäß einem der Ansprüche 1-4 und einen pharmazeutisch verträglichen Träger. 6. Ein Arzneimittel gemäß Anspruch 5 zur Verwendung bei der Behandlung eines empfindlichen Neoplasmas in einem Säuge, der dieses benötigt. 7. Eine Verbindung der Formel (la) oder ein pharmazeutisch verträgliches Salz davon gemäß einem der Ansprüche 1-4 zur Verwendung in der Therapie. 8. Eine Kombination aus (a) 2-Methyl-1-{[2-methyl-3-(trifluormethyl)phenyl]methyl}-6-(4-morpholinyl)-1H-benzimida-zol-4-carbonsäure oderein pharmazeutisch verträgliches Salz davon und (b) mindestens einem anti-neoplastischem Mittel; zur Verwendung bei der Behandlung von Krebs. 9. Eine Verbindung der Formel (la) oder ein pharmazeutisch verträgliches Salz davon gemäß einem der Ansprüche 1-4, oder ein Arzneimittel gemäß Anspruch 5, oder eine Kombination gemäß Anspruch 8, zur Verwendung bei der Behandlung eines PTEN-defizienten empfindlichen Neoplasmas ausgewählt aus Gehirn (Gliome), Glioblastomen, Leukämien, Bannayan-Zonana-Syndrom, Cowden-Syndrom, Lhermitte-Duclos-Syndrom, Brustkrebs, entzündlichem Brustkrebs, Kolorektalkrebs, Wilms-Tumor, Ewing-Sarkom, Rhabdomyosarkom, Ependymom, Medulloblastom, Darmkrebs, Kopf-und Halskrebs, Nierenkrebs, Lungenkrebs, Leberkrebs, Melanom, schuppenartigem Zellenkarzinom, Eierstockkrebs, Bauchspeicheldrüsenkrebs, Prostata krebs, Sarkom, Krebs, Osteosarkom, Riesenzelltumor des Knochen, Schilddrüsenkrebs, Lymphoblast-T-Zelle, Leukämie, chronischer myeloischer Leukämie, chro- nischer lymphozytischer Leukämie, Haarzellleukämie, akuter lymphoblastischer Leukämie, akuter myeloischer Leukämie, chronischer Neutrophilenleukämie, akuter lymphoblastischer T-Zell-Leukämie, Plasmozytom, immunoblas-tischer Großzellleukämie, Mantelzellleukämie, multiples Myelom, megakaryoblastischer Leukämie, akuter mega-karyocytischer Leukämie, Promyelozytenleukämie, Erythroleukämie, bösartigem Lymphom, Hodgkin-Lymphom, Non-Hodgkin-Lymphom, lymphoblastischem T-Zell-Lymphom, Burkitt-Lymphom, folikulärem Lymphom, Neuroblastom, Blasenkrebs, urothelialem Krebs, Scheidenkrebs, Gebärmutterhalskrebs, Endometriumkrebs, Nierenkrebs, Mesotheliom, Speiseröhrenkrebs, Speicheldrüsenkrebs, hepatozellulärem Krebs, Magenkrebs, Nasenrachenkrebs, Mundhöhlenkrebs, Mundkrebs, GIST (gastrointestinaler Stromatumor) und Hodenkrebs. 10. Eine Verbindung der Formel (la) oder ein pharmazeutisch verträgliches Salz davon zur Verwendung, oder ein Arzneitmittel zur Verwendung, oder eine Kombination zur Verwendung, gemäß Anspruch 9, wobei das PTEN-defiziente Neoplasma ausgewählt ist aus Prostata krebs, Nicht-Kleinzell-Lungenkrebs, Endometriumkrebs, Magenkrebs, Melanom, Kopf- und Halskrebs, Brustkrebs, einschließlich triple-negativem Brustkrebs und Gliom. 11. Eine Verbindung der Formel (la) oder ein pharmazeutisch verträgliches Salz davon zur Verwendung, oder ein Arzneitmittel zur Verwendung, oder eine Kombination zur Verwendung, gemäß Anspruch 10, wobei der Prostatakrebs ho rmon refraktärer Prostatakrebs ist. 12. Verwendung einer Verbindung der Formel (la), oder eines pharmazeutisch verträglichen Salzes davon, gemäß einem der Ansprüche 1 - 4 bei der Herstellung eines Medikaments zur Verwendung bei der Behandlung eines PTEN-defizienten empfindlichen Neoplasmas, das ausgewählt ist aus Gehirn (Gliome), Glioblastomen, Leukämien, Bannayan-Zonana-Syndrom, Cowden-Syndrom, Lhermitte-Duclos-Syndrom, Brustkrebs, entzündlichem Brustkrebs, Kolorektalkrebs, Wilms-Tumor, Ewing-Sarkom, Rhabdomyosarkom, Ependymom, Medulloblastom, Darmkrebs, Kopf- und Halskrebs, Nierenkrebs, Lungenkrebs, Leberkrebs, Melanom, schuppenartigem Zellenkarzinom, Eierstockkrebs, Bauchspeicheldrüsenkrebs, Prostatakrebs, Sarkom, Krebs, Osteosarkom, Riesenzelltumor des Knochen, Schilddrüsenkrebs, Lymphoblast-T-Zelle, Leukämie, chronischer myeloischer Leukämie, chronischer lymphozytischer Leukämie, Haarzellleukämie, akuter lymphoblastischer Leukämie, akuter myeloischer Leukämie, chronischer Neutrophilenleukämie, akuter lymphoblastischer T-Zell-Leukämie, Plasmozytom, immunoblastischer Großzellleukämie, Mantelzellleukämie, multiples Myelom, megakaryoblastischer Leukämie, akuter megakaryocy-tischer Leukämie, Promyelozytenleukämie, Erythroleukämie, bösartigem Lymphom, Hodgkin-Lymphom, Non-Ho-dgkin-Lymphom, lymphoblastischem T-Zell-Lymphom, Burkitt-Lymphom, folikulärem Lymphom, Neuroblastom, Blasenkrebs, urothelialem Krebs, Scheidenkrebs, Gebärmutterhalskrebs, Endometriumkrebs, Nierenkrebs, Mesotheliom, Speiseröhrenkrebs, Speicheldrüsenkrebs, hepatozellulärem Krebs, Magenkrebs, Nasenrachenkrebs, Mundhöhlenkrebs, Mundkrebs, GIST (gastrointestinaler Stromatumor) und Hodenkrebs. 13. Verwendung gemäß Anspruch 12, wobei das PTEN-defiziente empfindliche Neoplasma ausgewählt ist aus Prostatakrebs, Nicht-Kleinzell-Lungenkrebs, Endometriumkrebs, Magenkrebs, Melanom, Kopf- und Halskrebs, Brustkrebs, einschließlich triple-negativem Brustkrebs und Gliom. 14. Verwendung gemäß Anspruch 13, wobei der Prostatakrebs hormonrefraktärer Prostatakrebs ist.
Revendications 1. Composé de formule (la)
qui est l’acide 2-méthyl-1-{[2-méthyl-3-(trifluoro-méthyl)phényl]méthyl}-6-(4-morpholynil)-1 H-benzimidazole-4-car-boxylique, ou un de ses sels pharmaceutiquement acceptables. 2. Composé suivant la revendication 1, de formule (la)
qui est l’acide 2-méthyl-1-{[2-méthyl-3-(trifluoro-méthyl)phényl]méthyl}-6-(4-morpholinyl)-1 H-benzimidazole-4-car-boxylique. 3. Sel pharmaceutiquement acceptable du composé de formule (la) suivant la revendication 1. 4. Sel pharmaceutiquement acceptable du composé de formule (la) suivant la revendication 3, le sel étant le sel Tris 1:1. 5. Composition pharmaceutique qui comprend un composé de formule (la) ou un de ses sels pharmaceutiquement acceptables suivant l’une quelconque des revendications 1 à 4, et un support pharmaceutiquement acceptable. 6. Composition pharmaceutique suivant la revendication 5, pour une utilisation dans le traitement d’un néoplasme sensible chez un mammifère en ayant besoin. 7. Composé de formule (la) ou un de ses sels pharmaceutiquement acceptables suivant l’une quelconque des revendications 1 à 4, pour une utilisation en thérapie. 8. Association (a) d’acide 2-méthyl-1-{[2-méthyl-3-trifluorométhyl)phényl]méthyl}-6-(4-morpholinyl)-1 H-benzimidazo-le-4-carboxylique ou d’un de ses sels pharmaceutiquement acceptables et (b) d’au moins un agent anti-néoplasique pour une utilisation dans le traitement du cancer. 9. Composé de formule (la) ou un de ses sels pharmaceutiquement acceptables suivant l’une quelconque des revendications 1 à 4, ou composition pharmaceutique suivant la revendication 5, ou association suivant la revendication 8, pour une utilisation dans le traitement d’un néoplasme sensible déficient en PTEN choisi entre des néoplasmes du cerveau (gliomes), des glioblastomes, des leucémies, le syndrome de Bannayan-Zonana, la maladie de Cowden, la maladie de Lhermitte-Duclos, le cancer du sein, le cancer mammaire inflammatoire, le cancer colorectal, la tumeur deWilm, le sarcome d’Ewing, un rhabdomyosarcome, un épendymome, un médulloblastome, le cancer du côlon, le cancer de la tête et du cou, le cancer des reins, le cancer du poumon, le cancer du foie, un mélanome, un carcinome à cellules squameuses, le cancer des ovaires, le cancer du pancréas, le cancer de la prostate, un sarcome, un cancer, un ostéosarcome, une tumeur à cellules géantes des os, le cancer de la thyroïde, la leucémie lymphoblastique à lymphocytes T, la leucémie myélogène chronique, la leucémie lymphocytaire chronique, la leucémie à tricholeucocytes, la leucémie lymphoblastique aigüe, la leucémie myélogène aigüe, la leucémie neutrophile chronique, la leucémie lymphoblastique aigüe à lymphocytes T, un plasmacytome, la leucémie immunoblastique à grandes cellules, la leucémie des cellules du manteau, un myélome multiple, la leucémie mégakaryoblastique, la leucémie mégakaryocytaire aigüe, la leucémie promyélocytaire, l’érythroleucémie, un lymphome malin, un lymphome de Hodgkin, un lymphone non-hodgkinien, un lymphome lymphoblastique à lymphocytes T, le lymphome de Burkitt, le lymphome folliculaire, un neuroblastome, le cancer de vessie, le cancer urothélial, le cancer de la vulve, le cancerdu col de l’utérus, le cancerde l’endomètre, le cancerdes reins, un mésothéliome, le cancerde l’oesophage, le cancerdes glandes salivaires, le cancer hépato-cellulaire, le cancer gastrique, le cancer nasopharyngé, le cancer buccal, le cancerde la bouche, une GIST (tumeur stromale gastrointestinale) et le cancer testiculaire. 10. Composé de Formule (la) ou un de ses sels pharmaceutiquement acceptables pour une utilisation, ou composition pharmaceutique pour une utilisation, ou association pour une utilisation suivant la revendication 9, ledit néoplasme déficient en PTEN étant choisi entre le cancer de la prostate, le cancer pulmonaire non à petites cellules, le cancer de l’endomètre, le cancer gastrique, un mélanome, le cancer de la tête et du cou, le cancer du sein, y compris le cancerdu sein triplement négatif, et un gliome. 11. Composé de Formule (la) ou un de ses sels pharmaceutiquement acceptables pour une utilisation, ou composition pharmaceutique pour une utilisation, ou une association pour une utilisation suivant la revendication 10, ledit cancer de la prostate étant un cancerde la prostate réfractaire au traitement hormonal. 12. Utilisation d’un composé de formule (la), ou d’un de ses sels pharmaceutiquement acceptables, suivant l’une quelconque des revendications 1 à 4, dans la production d’un médicament pour une utilisation dans le traitement d’un néoplasme sensible déficient en PTEN qui est choisi entre des néoplasmes du cerveau (gliomes), des glioblastomes, des leucémies, le syndrome de Bannayan-Zonana, la maladie de Cowden, la maladie de Lhermitte-Duclos, le cancer du sein, le cancer mammaire inflammatoire, le cancer colorectal, la tumeur de Wilm, le sarcome d’Ewing, un rhabdomyosarcome, un épendymome, un médulloblastome, le cancerdu côlon, le cancerde la tête et du cou, le cancer des reins, le cancerdu poumon, le cancer du foie, un mélanome, un carcinome à cellules squameuses, le cancer des ovaires, le cancer du pancréas, le cancer de la prostate, un sarcome, un cancer, un ostéosarcome, une tumeur à cellules géantes des os, le cancer de la thyroïde, la leucémie lymphoblastique à lymphocytes T, la leucémie myélogène chronique, la leucémie lymphocytaire chronique, la leucémie à tricholeucocytes, la leucémie lymphoblastique aigüe, la leucémie myélogène aigüe, la leucémie neutrophile chronique, la leucémie lymphoblastique aigüe à lymphocytes T, un plasmacytome, la leucémie immunoblastique à grandes cellules, la leucémie de cellules du manteau, un myélome multiple, la leucémie mégakaryoblastique, la leucémie mégakaryocytaire aigüe, la leucémie promyélocytaire, l’érythroleucémie, un lymphome malin, le lymphome de Hodgkin, un lymphome non-hodg-kinien, un lymphome lymphoblastique à lymphocytes T, le lymphome de Burkitt, le lymphome folliculaire, un neuroblastome, le cancerde la vessie, le cancer urothélial, le cancerde la vulve, le cancerdu col de l’utérus, le cancer de l’endomètre, le cancer rénal, un mésothéliome, le cancer de l’oesophage, le cancer des glandes salivaires, le cancer hépatocellulaire, le cancer gastrique, le cancer nasopharyngé, le cancer buccal, le cancer de la bouche, une GIST (tumeur stromale gastrointestinale) et le cancer testiculaire. 13. Utilisation suivant la revendication 12, ledit néoplasme sensible déficient en PTEN étant choisi entre le cancerde la prostate, le cancer pulmonaire non à petites cellules, le cancerde l’endomètre, le cancer gastrique, un mélanome, le cancer de la tête et du cou, le cancer du sein, y compris un cancer du sein triplement négatif, et un gliome. 14. Utilisation suivant la revendication 13, ledit cancer de la prostate étant un cancer de la prostate réfractaire au traitement hormonal.
REFERENCES CITED IN THE DESCRIPTION
This list of references cited by the applicant is for the reader's convenience only. It does not form part of the European patent document. Even though great care has been taken in compiling the references, errors or omissions cannot be excluded and the EPO disclaims all liability in this regard.
Patent documents cited in the description • US 20070066606A1 [0006] · US 97721797 A[0087] • US 7223757 B2 [0007] · US 5681835 A [0088] • US 6063923 A [0087] · US 5877219 A [0088] • US 5342947 A [0087] · US 6207716 B [0088] • US 5559235 A [0087] · US 6268391 B [0094] • US 5491237 A [0087] · WO 2009039140 A [0164]
Non-patent literature cited in the description • SAMUELS Y ; ERICSON K. Oncogenic PI3K and its · JClinNeurosci., December2010, vol. 17(12), 1543-7 role in cancer. Current Opinion in Oncology, 2006, [0033] vol. 18, 77-82 [0002] · Nat Genet., May 2009, vol. 41 (5), 619-24 [0033] • VIVANCO I; SAWYERS CL. The phosphatidylinosi- · BrJ Cancer, 21 October 2008, vol. 99 (8), 1296-301 tol 3-kinase-AKT pathway in human cancer. Nature [0033]
Reviews Cancer, 2002, vol. 2, 489-501 [0003] · Int J Cancer, 15 March 2007, vol. 120 (6), 1284-92 • Trends in Biochem Sci, 2007, vol. 32,450-456 [0003] [0033] • TORBETTNE ; LUNAA ; KNIGHTZAetal.Achem- · J Invest Dermatol., January 2006, vol. 126(1), 154-60 ical screen in diverse breast cancer cell lines reveals [0033] genetic enhancers and suppressors of sensitivity to · J Clin Oncol., 10 January 2006, vol. 24 (2), 288-95 PI3K isotype-selective inhibition. Biochem J, 2008, [0033] vol. 415, 97-110 [0004] · Am J Clin Pathol., October 2005, vol. 124(4),528-36 • ZHAO JJ ; LIU Z ; WANG L ; SHIN E ; LODA MF ; [0033] ROBERTS TM. The oncogenic properties of mutant · Int J Oncol., April 2009, vol. 34 (4), 983-93 [0033] p110a and p110b phosphatidylinositol 3-kinases in · Epigenetics, 01 May 2011, vol. 6 (5), 638-49 [0033] human mammary epithelial cells. Proc Natl Acad Sci · Gynecol Oncol., February 2009, vol. 112 (2), 307-13 USA, 2005, vol. 102, 18443-8 [0004] [0033] • PU P ; KANGC ; ZHANG Z etal. Downregulation of · Mod Pathol., October 2010, vol. 23 (10), 1316-24 PIK3CB by siRNA suppresses malignant glioma cell [0033] growth in vitro and in vivo. Technolo Cancer Res · J Pathol., February 2010, vol. 220(3), 382-400 [0033]
Treat, 2006, vol. 5, 271-280 [0004] · Lung, March 2009, vol. 187 (2), 104-9 [0033] • WEE S ; WIEDERSCHAIN, MAIRA S-M ; LOO A ; · Anticancer Res., January 2007, vol. 27 (1B), 575-81 MILLER C et al. PTEN-deficient cancers depend on [0033] PIK3CB. Proc Natl Acad Sci, 2008, vol. 105, · Am J Surg., June 2008, vol. 195 (6), 719-25 [0033] 13057-13062 [0004] · J Clin Oncol., 10 December 2009, vol. 27 (35), • JIAS ; LIU Z; ZHANG S ; LIU P ; ZHANG L etal. 5924-30(0033]
Essential roles of PI(3)K-p110b in cell growth, me- · Gynecol Oncol., June 2004, vol. 93 (3), 621-7 [0033] tabolism and tumorgenesis. Nature, 2008, vol. 10, · J Oral Pathol Med., August 2002, vol. 31 (7),379-84 1038 [0004] [0033] • PARAPURAM, S.K. etal. Loss of PTEN expression · Cancer Principles and Practice f Oncology. Lippincott by dermal fibroblasts cuases skin fibrosis. J. of Inves- Williams &amp; Wilkins Publishers, 15 February 2001 tigative Dermatology, 09 June 2011 [0005] [0046] • HOLLANDER, M. CHRISTINE; BLUMENTHAL, · WANI et al. J. Am. Chem, Soc., 1971, vol. 93, 2325 GIDEON M.; DENNIS, PHILLIP P. PTEN loss in the [0050] continuum of common cancers, rare syndromes and · SCHIFF et al. Proc. Natl. Acad, Sci., 1980, vol. 77, mouse models. Nature Reviews!Cancer, 2011, vol. 1561-1565 [0050] 11,289-301 [0005] · SCHIFF etal. Nature, 1979, vol. 277,665-667 [0050] • Am J Clin Pathol., February 2009, vol. 131 (2), 257-63 · KUMAR. J. Biol, Chem, 1981, vol. 256,10435-10441 [0033] [0050] • New trends in Natural Products Chemistry 1986. D. · ABRAHAM, R.T. Current Opinion in Immunology, G. I. KINGSTON etal. Studies in Organic Chemistry. 1996, vol. 8 (3), 412-8 [0095]
Elsevier, 1986, vol. 26, 219-235 [0050] · CANMAN, C.E. ; LIM, D.S. Oncogene, 1998, vol. 17 • MARKMAN et al. Yale Journal of Biology and Med- (25), 3301-3308 [0095] icine, 1991, vol. 64, 583 [0051] · JACKSON, S.P. International Journal of Biochemis- • MCGUIRE et al. Ann. Intern, Med., 1989, vol. 111, try and Cell Biology, 1997, vol. 29 (7), 935-8 [0095] 273 [0051] · ZHONG, H. et al. Cancer res, 2000, vol. 60 (6), • HOLMES et al. J. Nat. Cancer Inst., 1991, vol. 83, 1541-1545 [0095] 1797 [0051] · POWIS, G. ; KOZIKOWSKI A. New Molecular Tar- • EINZIG. Proc. Am. Soc. Clin. Oncol., vol. 20, 46 gets for Cancer Chemotherapy. CRC press, 1994 [0051] [0096] • FORASTIRE. Sem. Oncol., 1990, vol. 20, 56 [0051] · SCHAROVSKY, O.G. ; ROZADOS, V.R. ; GERVA- • WOO. Nature, 1994, vol. 368, 750 [0051] SONI, S.l.; MATAR, P. Journal of Biomedical Sci- • IGNOFF, R.J. Cancer Chemotherapy Pocket Guide, ence, 2000, vol. 7 (4), 292-8 [0097] 1998 [0051] · ASHBY, M.N. Current Opinion in Lipidology, 1998, • KEARNS, C.M. Seminars in Oncology, 1995, vol. 3 vol. 9 (2), 99-102 [0097] (6), 16-23 [0051] · BioChem. Biophys. Acta, vol. 1423 (3), 19-30 [0097] • KATH, JOHN C. Exp. Opin. Ther. Patents, 2000, vol. · GREEN, M.C. et al. Monoclonal Antibody Therapy 10 (6), 803-818 [0091] for Solid Tumors. Cancer Treat. Rev., 2000, vol. 26 • SHAWVER et al. DDT, 02 February 1997, vol. 2 (4), 269-286 [0098]
[0091] · Tyrosine Kinase Signalling in Breast cancenerbB • Growth factor receptors as targets. LOFTS, F. J. et Family Receptor Tyrosine Kinases, Breast cancer al. New Molecular Targets for Cancer Chemothera- Res., 2000, vol. 2 (3), 176-183 [0098] py. CRC press, 1994 [0091] · BREKKEN, R.A. et al. Selective Inhibition of • SINH, S. ; COREY, S.J. Journal of Hematotherapy VEGFR2 Activity by a monoclonal Anti-VEGF anti- and Stem Cell Research, 1999, vol. 8 (5), 465-80 body blocks tumor growth in mice. Cancer Res., [0092] 2000, vol. 60, 5117-5124 [0098] • BOLEN, J.B. ; BRUGGE, J.S. Annual review of Im- · BRUNS CJ et al. Cancer Res., 2000, vol. 60, munology, 1997, vol. 15, 371-404 [0092] 2926-2935 [0099] • SMITHGALL, T.E. Journal of Pharmacological and · SCHREIBER AB ; WINKLER ME ; DERYNCK R.
Toxicological Methods, 1995, vol. 34 (3), 125-32 Science, 1986, vol. 232, 1250-1253 [0099] [0093] · YEN L et al. Oncogene, 2000, vol. 19, 3460-3469 • YAMAMOTO, T. ; TAYA, S. ; KAIBUCHI, K. Journal [0099] of Biochemistry, 1999, vol. 126(5), 799-803 [0094] · REILLY RT et al. Cancer Res., 2000, vol. 60, • BRODT, P ; SAMANI, A.; NAVAB, R. Biochemical 3569-3576 [0100]
Pharmacology, 2000, vol. 60, 1101-1107 [0094] · CHEN Y ; HU D ; ELING DJ ; ROBBINS J ; KIPPS • MASSAGUE, J.; WEIS-GARCIA, F. Cancer Sur- TJ. Cancer Res., 1998, vol. 58, 1965-1971 [0100] veys, 1996, vol. 27, 41-64 [0094] · WATER JS et al. J. Clin. Oncol., 2000, vol. 18, • PHILIP, P.A.; HARRIS, A.L. Cancer Treatment and 1812-1823 [0101]
Research, 1995, vol. 78, 3-27 [0094] · KITADA S et al. Antisense Res. Dev., 1994, vol. 4, • LACKEY, K. et al. Bioorganic and Medicinal Chem- 71-79 [0101] istry Letters, 2000, (10), 223-226 [0094] · ROSANIA etal. Exp. Opin. Ther. Patents, 2000, vol. • MARTINEZ-IACACI, L. et al. Int. J. Cancer, 2000, 10 (2), 215-230 [0102] vol. 88 (1), 44-52 [0094]

Claims (8)

  1. FI $nfe inhibtof tpfcű SZABADALMI IGÉNYPONTOIC ' ' ' 11P11 ^SZTNhJo0031721 ' L Az (fa) kepleiü vegyület
    s^ely a AmeBM - {12-m«tl;18^aB^m«3a»sF ^karbonsav, vagy annak gyógyásüilagi slfogaáhaté-séja, 2. Α«·1 igénypont szerinti (fa) képkfn vegyslft
    4-karbonsav, %,Jm I, Iflnypom szerinti (la) yplslü vrsgplNf gyógyászatiig elfogaftalAséia,
  2. 4. Az (fa) képiem vegyidet 3. i|l#ypÉ:i:P#1Íl gyógyáaaatslag dfbgpfbÉi|^^::ÉÉI a só a/. 1; s ?ris?.-M>.
  3. 5. Gyógyszeikészkmény, mely az 1-4, igénypontok bármelyike szerinti (la) képiéin vegyiktől vagy annak: gyögyáazatifag élfbisAato séfáí &amp;i$a? gyögyászaülag elfogadható bratloróanyag' n tana ha az,
  4. 6. Az 5. igénypont szerlpi fy^gfSl^íÉ^^tekiíó^osy feezelislk ygli alkalmaznia, egy etre s/oralo emtdsbefH. ?. Az 1-4, igénypontok bármelyike szerinti (la) képletü vegyidet vagy ||pg|ieíMg elfogadható sója, a |p|páéafoan való aikaknazásra. t,:Az:p|2'®tMiyi#[2'rnéril^ IH- 'tetójB.#a?.ol-4>karboi?s8Vi vagy annak gyógyászaidag elfogadható sóp Is ψ) legalább e|y daganAóltenes szer kofabiüácio|a; iák kezelésében való aikátruazásra.
  5. 9. Az 1-4, igénypontok bármelyike szerinti (la) képletü vegvület vagy annak fobgyászatilag elfogadható sója, vagy az 5. igénypont szeriMi gyógyszvtkéxzgatóay, vagy a 8. igénypont szerinti kombináció, az agy, (gliomák), glioblasíomák, leukémiák, Bannayan-Zonana szindróma, Cowder? betegség, IPemdtte-líadoá betegség, vnelMk, gyulladásos mellrák, koloreualis rák, WdsmtaisuÉyiwöig'dMtfaóö* harásícsíkölí lkom szárítóra» ídmbdomyosareouu), eprudydmnÉg medudoblasforna, vastagbél rák. fej és ayak rák, veserák, tudorik. májrák, melanoma pikkelysejtes kareütoras, petefészek rák, ImsnyálraMgy A, prosztata rák, szárítom,», mk< osteosarcerua, csontok óriássejtes tumora, pajzsmmgy rák, íimfoblasztos T-sejt, leukémia, krónikus m decid leukémia, krómkus lirn.focltás leukémia, ,n s -e o i, akut hu <rádas o ránk Crna, akut mJtu ni leukémia, krónikus neuüoíd leukémia, akut lindoblasztos Ί'-sejt leukémia, plosnmcyfoma. immivnoblasztos nagy-sejtes leukémia, köpeny-sejtes leukémia, myeloma multiplex, megakaricÉláS&amp;Má Ii«É#mÍ%;ltet utegaknriocfokus leukémia, promielocitás leukémia, erítKHeukéima, malígnss:ftmMma, hodgkins limloma, nem-hpdgklns Hnifóma, "f-sejt limfohlaaztos fcfpma, Burknt-Hmfómav ml mnlar 4- i s m nenroblasíoow. hobngrák, uouurásúh', tok, vulva tűk, nuhnyaktal méintyálkahátlya rák, vesesejt.es rák, mesotheboma, nyelőcső rák, nyáhuirigy rák, májsejt rák, gyomorráig orr-garat rák, szájüregi rák, szilfák, ©3IT (gaazéröintesztirtális szfofoa tumort és hererák közül kezelésében való alkMmazásra.
  6. 10. Az fial vegyidet vagy annak gyógyászat ilag elfogadliaíó apának alkaimazlsis vagy egy gyógyszerkészümény alkalmazása vagy egy kombmlció aiksjmázása a §, jgépfgiur szerinti alkalmazásra, ahol az említett PTEN'-hiányos fogékony neopiaznut a prosztatarák, némAMséjtes tüdőrák, rnélmyilkabárfya rák, gyomorrák, rneióioma, fej és nyak rák. Mellrák,: beleértve a tripla-negatívmellrákok és glfóma közül van kiválasztva,
  7. 11. Az ΐ la) képkyü vegyidet, vagy annak gydgyászatiing elfogadható sójának alkalmazása, vagy gyógyszerkészítmény alkalmazása, vagy kombináció alkalmazása a lÖ. igéin pont szerinti alkalmazásra, ahol az vmhfou ntos/tatanif hmmm>-reftaku-i pio-zmtarak. 1 VI 1 gé' \ponm\ ha i\ ke orm la k>.p tú u^töm\,,>. n at \ gvőgyávaíilag elfogadható sójának alkalmazása az agy, (gÜontll|h g||^Í8§t«ák, leukemlfe Bánnáyan-Zonuna szmdrörna i msden belefog. 1 Ueor.ete Oneíos boa gucg. mellrák, ;,:yn!b<.U">\ melbák. koiorokuü^ rák. Wdrts tum-n, komé s/arkonut, harántcsíkolt tzom szarként·» <t1talxlömyo$arcomah ependymoma, methrlloblastoma, vastagbél rák, fej és nyak iák, vese-rak, uléSrák, náájí^;,:.^iiaft&amp;írtó, pikkelyaejtes hasnyálmirigy rák* j^^isÉsa^i^Mcoma, rák, osieostiéómá.. ésontok óriássojtes ítémora, iwomirigy rák, hrnbdtlas/tosT-sejí, leukémia;; krómkus mtkxtid leukémia* krónikus-inntbcuas leukémia, hutás sejtes leukémia, akut timíohta.viON teukenua, akta tnthmid jguMmia. krónikus ocutroíh leukémia*. aktit. limfoftlasztos T-ssejt leukémia, plaauuteyioata, imrotmobkt.szíos nagy-sejtes leukémia, köpeny-sejtes lettkémia, mveloma multiplex, megakariohlasztos leukémia, akut. .ntegakariocttikus leukémia, pronikdoeitás leukémia, ^W&amp;MlMkéBusu makgnns limféma. bodgkios limloma, Ttem-bfxlgkins limloma, T-sejt iimisbláa^éos iktibma, Burkitt-hmfőim foiheulális limloma, ueumbl axióma, hólyagrák, nrotheliálís rák, vuiva rák, méhoyakrák, ialfetyálkaháttya rák, vesesejmszák, mesoiheiiomá, nyelőcső iák, nyáonhigy ták, májsejt rák, gyomorrilg orr-garat rák, szájüregi rák, száj-rak, CI1ST rgaNroomtcN/imabs sztonu tnntm\ es hetei tk kívül kn ako/tou PTFNdnámos ti yekors mepiuzma keo'lösex', x< to , - ab" ν,Ό- t < zoleaio esogsszetk^riUT'Cnx ígloÉllÉláiri,
  8. 13,. A J,2, igénygotiíszerintf ulkálmazás. ahol az említett P'TEN-hiányos fogékony: neoplazma a prosztatarák, nem-kissejtes tüdőrák, rnéhnyálkaháttya rák, gyomorrák, mehmonvt, tej es nyak mk, oKi'.tuk belerróe a tnplmieme:-·. mellrákot, es _ betua kozni v.at kiválasztva. 1-1 \H teerxpoot <· eneít a'-m ,a o, ahol a o maca pttv utak Vreon re fraktér prosztatarák. Á tne ghatállpgoit t
HUE11831214A 2010-10-06 2011-09-23 PI3 kináz inhibitor hatású benzimidazol-származékok HUE033209T2 (hu)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US39031410P 2010-10-06 2010-10-06
US201161528397P 2011-08-29 2011-08-29

Publications (1)

Publication Number Publication Date
HUE033209T2 true HUE033209T2 (hu) 2017-11-28

Family

ID=45925614

Family Applications (1)

Application Number Title Priority Date Filing Date
HUE11831214A HUE033209T2 (hu) 2010-10-06 2011-09-23 PI3 kináz inhibitor hatású benzimidazol-származékok

Country Status (37)

Country Link
US (10) US20130196990A1 (hu)
EP (2) EP3170813B1 (hu)
JP (1) JP5719028B2 (hu)
KR (1) KR101594002B1 (hu)
CN (1) CN103124496B (hu)
AR (1) AR083296A1 (hu)
AU (1) AU2011312594B2 (hu)
BR (1) BR112013008259A2 (hu)
CA (1) CA2812608C (hu)
CL (1) CL2013000935A1 (hu)
CO (1) CO6700852A2 (hu)
CR (1) CR20130192A (hu)
CY (1) CY1118792T1 (hu)
DK (1) DK2624696T3 (hu)
DO (1) DOP2013000059A (hu)
EA (1) EA022623B1 (hu)
ES (2) ES2616238T3 (hu)
HR (1) HRP20170279T1 (hu)
HU (1) HUE033209T2 (hu)
IL (1) IL225140A (hu)
JO (1) JO3194B1 (hu)
LT (1) LT2624696T (hu)
MA (1) MA34591B1 (hu)
ME (1) ME02663B (hu)
MX (1) MX337662B (hu)
MY (1) MY170236A (hu)
NZ (1) NZ608069A (hu)
PE (1) PE20140192A1 (hu)
PL (1) PL2624696T3 (hu)
PT (1) PT2624696T (hu)
RS (1) RS55662B1 (hu)
SG (1) SG188974A1 (hu)
SI (1) SI2624696T1 (hu)
TW (1) TWI513690B (hu)
UY (1) UY33648A (hu)
WO (1) WO2012047538A1 (hu)
ZA (1) ZA201301951B (hu)

Families Citing this family (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20130196990A1 (en) 2010-10-06 2013-08-01 Junya Qu Benzimidazole Derivatives As PI3 Kinase Inhibitors
US8778937B2 (en) * 2011-12-20 2014-07-15 Glaxosmithkline Llc Benzimidazole boronic acid derivatives as PI3 kinase inhibitors
KR20150103735A (ko) * 2013-01-09 2015-09-11 글락소스미스클라인 인털렉츄얼 프로퍼티 (넘버 2) 리미티드 조합물
WO2015042027A1 (en) * 2013-09-19 2015-03-26 Glaxosmithkline Llc Combination drug therapy
AU2014321456B2 (en) * 2013-09-19 2018-05-24 Glaxosmithkline Llc Combination drug therapy
NZ718487A (en) 2013-10-16 2020-01-31 Shanghai Yingli Pharm Co Ltd Fused heterocyclic compound, preparation method therefor, pharmaceutical composition, and uses thereof
CN103910682B (zh) * 2013-11-28 2016-03-02 大连理工大学 一种基于邻苯二胺环化的苯并咪唑类化合物制备方法
JP2017502016A (ja) * 2013-12-20 2017-01-19 バイオメッド バレー ディスカバリーズ,インコーポレイティド 1型mek阻害剤およびerk阻害剤の組み合わせを使用するがんの処置
EA201692267A1 (ru) 2014-06-13 2017-06-30 Джилид Сайэнс, Инк. Ингибиторы фосфатидилинозитол-3-киназы
BR112016028876A2 (pt) 2014-06-13 2017-08-22 Gilead Sciences Inc composto, composição farmacêutica, método para tratar uma doença ou condição em um ser humano, kit, e, uso de um composto, um sal, isômero ou uma mistura farmaceuticamente aceitável dos mesmos.
WO2015191754A2 (en) 2014-06-13 2015-12-17 Gilead Sciences, Inc. Phosphatidylinositol 3-kinase inhibitors
EA201692249A1 (ru) 2014-06-13 2017-05-31 Джилид Сайэнс, Инк. Ингибиторы фосфатидилинозитол-3-киназы
AU2015274696B2 (en) 2014-06-13 2018-09-27 Gilead Sciences, Inc. Phosphatidylinositol 3-kinase inhibitors
JP6586463B2 (ja) 2014-12-19 2019-10-02 ヤンセン ファーマシューティカ エヌ.ベー. PI3Kβ阻害剤としての複素環連結イミダゾピリダジン誘導体
BR112017012930A2 (pt) * 2014-12-19 2018-01-09 Janssen Pharmaceutica Nv derivados de imidazopiridazina como inibidores de pi3kbeta.
GB201504689D0 (en) 2015-03-19 2015-05-06 Glaxosmithkline Ip Dev Ltd Chemical compounds
US10526316B2 (en) 2015-10-09 2020-01-07 Janssen Pharmaceutica Nv Quinoxaline and pyridopyrazine derivatives as PI3Kβ inhibitors
CA3011189C (en) * 2016-01-14 2024-02-20 Bayer Pharma Aktiengesellschaft 5-substituted 2-(morpholin-4-yl)-1,7-naphthyridines
JP7158286B2 (ja) * 2016-06-16 2022-10-21 ヤンセン ファーマシューティカ エヌ.ベー. PI3Kβ阻害剤としてのアザベンゾイミダゾール誘導体
CA3025594C (en) 2016-06-16 2024-06-18 Janssen Pharmaceutica Nv Bicyclic pyridine, pyrazine, and pyrimidine derivatives as pi3k beta inhibitors
CN109689062B (zh) * 2016-07-11 2023-02-17 丹娜法伯癌症研究院 使用抗PI3Kβ和抗免疫检查点药剂的组合治疗PTEN缺陷型上皮癌的方法
WO2018057808A1 (en) 2016-09-23 2018-03-29 Gilead Sciences, Inc. Benzimidazole derivatives and their use as phosphatidylinositol 3-kinase inhibitors
TW201825465A (zh) 2016-09-23 2018-07-16 美商基利科學股份有限公司 磷脂醯肌醇3-激酶抑制劑
TW201813963A (zh) 2016-09-23 2018-04-16 美商基利科學股份有限公司 磷脂醯肌醇3-激酶抑制劑
TW201815787A (zh) 2016-09-23 2018-05-01 美商基利科學股份有限公司 磷脂醯肌醇3-激酶抑制劑
EP3548034A4 (en) * 2016-12-05 2020-07-08 Microbiotix, Inc. BROADBAND SPECTRUM INHIBITORS OF FILOVIRUS
BR112019020309A2 (pt) 2017-03-29 2020-04-28 Janssen Pharmaceutica Nv derivados de quinoxalina e piridopirazina como inibidores de pi3k-beta
MD3658552T2 (ro) 2017-07-28 2024-02-29 Yuhan Corp Procedeu de obținere a N-(5-((4-(4-((dimetilamino)metil)-3-fenil-1H-pirazol-1-il)pirimidin-2-il)amino)-4-metoxi-2-morfolinofenil)acrilamidei prin prin reacţia aminei corespunzătoare cu clorură de 3-halo-propionil
WO2019106605A1 (en) 2017-12-01 2019-06-06 Board Of Regents, The University Of Texas System Combination treatment for cancer
US11253189B2 (en) 2018-01-24 2022-02-22 Medtronic Ardian Luxembourg S.A.R.L. Systems, devices, and methods for evaluating neuromodulation therapy via detection of magnetic fields
CN111000847B (zh) * 2020-01-02 2021-03-19 黑龙江中医药大学 一种治疗肺纤维化的药物制剂及其用途
CN111346095B (zh) * 2020-03-14 2021-06-08 温州医科大学附属第二医院、温州医科大学附属育英儿童医院 用于治疗神经外科术后头痛的药物制剂

Family Cites Families (180)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0400835A1 (en) 1989-05-15 1990-12-05 Merck & Co. Inc. Substituted benzimidazoles as angiotensin II antagonists
IL95975A (en) 1989-10-24 1997-06-10 Takeda Chemical Industries Ltd N-benzyl- 2-alkylbenzimidazole derivatives, their production and pharmaceutical compositions containing them
US5559235A (en) 1991-10-29 1996-09-24 Glaxo Wellcome Inc. Water soluble camptothecin derivatives
TW284688B (hu) 1991-11-20 1996-09-01 Takeda Pharm Industry Co Ltd
ES2150941T3 (es) 1992-04-03 2000-12-16 Upjohn Co Aminas biciclicas-heterociclicas eficaces farmaceuticamente.
US5502187A (en) 1992-04-03 1996-03-26 The Upjohn Company Pharmaceutically active bicyclic-heterocyclic amines
GB9210400D0 (en) 1992-05-15 1992-07-01 Merck Sharp & Dohme Therapeutic agents
US5342947A (en) 1992-10-09 1994-08-30 Glaxo Inc. Preparation of water soluble camptothecin derivatives
JPH0867674A (ja) 1993-07-02 1996-03-12 Senju Pharmaceut Co Ltd 眼圧降下剤
CZ154994A3 (en) 1993-07-02 1995-09-13 Senju Pharma Co Visual hypotensive agent
US5681835A (en) 1994-04-25 1997-10-28 Glaxo Wellcome Inc. Non-steroidal ligands for the estrogen receptor
PE27997A1 (es) 1994-04-29 1997-09-20 Lilly Co Eli Antagonistas de receptores de taquicininas
US5491237A (en) 1994-05-03 1996-02-13 Glaxo Wellcome Inc. Intermediates in pharmaceutical camptothecin preparation
US5563143A (en) 1994-09-21 1996-10-08 Pfizer Inc. Catechol diether compounds as inhibitors of TNF release
DE19514579A1 (de) 1995-04-20 1996-10-24 Boehringer Ingelheim Kg Verwendung von alpha¶1¶¶L¶-Agonisten zur Behandlung der Harninkontinenz
JP2000501107A (ja) 1996-01-09 2000-02-02 イーライ・リリー・アンド・カンパニー ベンズイミダゾリル神経ペプチドy受容体アンタゴニスト
AU2139097A (en) 1996-03-01 1997-09-16 Eli Lilly And Company Methods of treating or preventing sleep apnea
AU2207897A (en) 1996-03-11 1997-10-01 Eli Lilly And Company Methods of treating or preventing interstitial cystitis
JP2001515482A (ja) 1997-03-07 2001-09-18 メタバシス・セラピューティクス・インコーポレイテッド フルクトース−1,6−ビスホスファターゼの新規なベンズイミダゾールインヒビター
GB9716557D0 (en) 1997-08-06 1997-10-08 Glaxo Group Ltd Benzylidene-1,3-dihydro-indol-2-one derivatives having anti-cancer activity
WO2000059905A1 (en) 1999-04-02 2000-10-12 Neurogen Corporation ARYL AND HETEROARYL FUSED AMINOALKYL-IMIDAZOLE DERIVATIVES: SELECTIVE MODULATORS OF GABAa RECEPTORS
US6380235B1 (en) 1999-05-04 2002-04-30 American Home Products Corporation Benzimidazolones and analogues
JP2000323278A (ja) 1999-05-14 2000-11-24 Toray Ind Inc 発光素子
CA2378047A1 (en) 1999-07-15 2001-01-25 Methvin Isaac Heterocyclic compounds for the treatment of migraine
WO2001012600A1 (en) 1999-08-12 2001-02-22 Cor Therapeutics, Inc. INHIBITORS OF FACTOR Xa
WO2001021634A1 (en) 1999-09-21 2001-03-29 Lion Bioscience Ag Benzimidazole derivatives and combinatorial libraries thereof
JP4721087B2 (ja) 1999-12-03 2011-07-13 小野薬品工業株式会社 トリアザスピロ[5.5]ウンデカン誘導体およびそれらを有効成分とする薬剤
AU2001236605A1 (en) 2000-02-01 2001-08-14 Cor Therapeutics, Inc. Indole and benzimidazole inhibitors of factor xa
PA8535601A1 (es) 2000-12-21 2002-11-28 Pfizer Derivados benzimidazol y piridilimidazol como ligandos para gabaa
US6472095B2 (en) 2000-12-29 2002-10-29 Utc Fuel Cells, Llc Hybrid fuel cell reactant flow fields
TW593278B (en) 2001-01-23 2004-06-21 Wyeth Corp 1-aryl-or 1-alkylsulfonylbenzazole derivatives as 5-hydroxytryptamine-6 ligands
WO2002072549A1 (en) 2001-03-12 2002-09-19 Millennium Pharmaceuticals, Inc. Functionalized heterocycles as modulators of chemokine receptor function and methods of use therefor
WO2002074769A1 (fr) 2001-03-19 2002-09-26 Ono Pharmaceutical Co., Ltd. Medicaments contenant comme principe actif des derives du triazaspiro [5.5] undecane
FR2822463B1 (fr) 2001-03-21 2004-07-30 Lipha Derives bicycliques de guanidines et leurs applications en therapeutique
US20030009034A1 (en) 2001-03-22 2003-01-09 Neil Wishart Transition metal mediated process
MXPA03008690A (es) * 2001-03-28 2003-12-12 Bristol Myers Squibb Co Inhibidores novedosos de tirosina cinasa.
US7081454B2 (en) * 2001-03-28 2006-07-25 Bristol-Myers Squibb Co. Tyrosine kinase inhibitors
US6677365B2 (en) 2001-04-03 2004-01-13 Telik, Inc. Antagonists of MCP-1 function and methods of use thereof
PL366639A1 (en) 2001-04-20 2005-02-07 Wyeth Heterocyclylalkoxy-, -alkylthio- and -alkylaminobenzazole derivatives as 5-hydroxytryptamine-6 ligands
CN1293072C (zh) 2001-04-20 2007-01-03 惠氏公司 作为5-羟色胺-6配体的杂环基氧基-、-硫代-和-氨基吲哚衍生物
US7030150B2 (en) 2001-05-11 2006-04-18 Trimeris, Inc. Benzimidazole compounds and antiviral uses thereof
US20030096852A1 (en) 2001-06-15 2003-05-22 Genentech, Inc. Human growth hormone antagonists
US6841566B2 (en) 2001-07-20 2005-01-11 Boehringer Ingelheim, Ltd. Viral polymerase inhibitors
FR2827862A1 (fr) 2001-07-27 2003-01-31 Lipha Derives imidazolylalkylarylalcanoiques et leurs applications en therapeutique
DE10139416A1 (de) 2001-08-17 2003-03-06 Aventis Pharma Gmbh Aminoalkyl substituierte aromatische Bicyclen, Verfahren zu ihrer Herstellung und ihre Verwendung als Arzneimittel
FR2829765A1 (fr) 2001-09-14 2003-03-21 Lipha Derives imidazolylalkoxylarylalcanoiques leurs applications en therapeutique
WO2004089942A2 (en) 2001-10-02 2004-10-21 Acadia Pharmaceuticals Inc. Benzimidazolidinone derivatives as muscarinic agents
MXPA04003103A (es) 2001-10-02 2004-07-27 Acadia Pharm Inc Derivados de bencimidazolidinona como agentes muscarinicos.
WO2003030902A1 (en) 2001-10-09 2003-04-17 Tularik Inc. Imidazole derivates as anti-inflammatory agents
EP1442028A4 (en) 2001-11-06 2009-11-04 Bristol Myers Squibb Co SUBSTITUTED ACID DERIVATIVES, WHICH APPRECIATE AS ANTIDIBILICS AND AGENTS AGAINST OBESITAS, AND METHODS
US7622479B2 (en) 2001-11-26 2009-11-24 Takeda Pharmaceutical Company Limited Bicyclic derivative, its production and use
CA2473740A1 (en) 2002-01-18 2003-07-31 David Solow-Cordero Methods of treating conditions associated with an edg receptor
DE60336633D1 (de) 2002-02-15 2011-05-19 Bridgestone Corp Kautschukzusammensetzung und daraus hergestellter pneumatischer reifen
BR0306016A (pt) 2002-03-13 2005-01-04 Array Biopharma Inc Derivados de benzimidazol n3 alquilado como inibidores da mek
NZ535158A (en) 2002-03-13 2007-06-29 Array Biopharma Inc N3 alkylated benzimidazole derivatives as MEK inhibitors
US7235537B2 (en) 2002-03-13 2007-06-26 Array Biopharma, Inc. N3 alkylated benzimidazole derivatives as MEK inhibitors
AU2003220202A1 (en) 2002-03-13 2003-09-29 Array Biopharma, Inc N3 alkylated benzimidazole derivatives as mek inhibitors
SE0200843D0 (sv) 2002-03-19 2002-03-19 Astrazeneca Ab Chemical compounds
DE10228103A1 (de) 2002-06-24 2004-01-15 Bayer Cropscience Ag Fungizide Wirkstoffkombinationen
DE10229777A1 (de) 2002-07-03 2004-01-29 Bayer Ag Indolin-Phenylsulfonamid-Derivate
US7205412B2 (en) 2002-07-03 2007-04-17 Samsung Electronics Co., Ltd. Antibiotic additive and ink composition comprising the same
WO2004014905A1 (en) 2002-08-08 2004-02-19 Boehringer Ingelheim Pharmaceuticals, Inc. Substituted benzimidazole compounds
US20040063938A1 (en) 2002-09-30 2004-04-01 Pfizer Inc Process for preparing haloalkyl pyrimidines
AU2003294249A1 (en) 2002-11-08 2004-06-03 Trimeris, Inc. Hetero-substituted benzimidazole compounds and antiviral uses thereof
EP1571146A4 (en) 2002-12-10 2010-09-01 Ono Pharmaceutical Co NITROGENIC HETEROCYCLIC COMPOUNDS AND THEIR MEDICAL USE
US7135493B2 (en) 2003-01-13 2006-11-14 Astellas Pharma Inc. HDAC inhibitor
WO2004082621A2 (en) 2003-03-15 2004-09-30 Bethesda Pharmaceuticals, Inc. Novel ppar agonists, pharmaceutical compositions and uses thereof
KR20050119201A (ko) * 2003-04-09 2005-12-20 니뽄 다바코 산교 가부시키가이샤 5원 헤테로방향족 고리 화합물 및 이의 의약적 용도
JPWO2004093912A1 (ja) 2003-04-23 2006-07-13 協和醗酵工業株式会社 好中球性炎症疾患の予防および/または治療剤
EP1620413A2 (en) 2003-04-30 2006-02-01 Cytokinetics, Inc. Compounds, compositions, and methods
SE0301699D0 (sv) * 2003-06-10 2003-06-10 Astrazeneca Ab Benzimidazole derivatives, compositions containing them, preparation thereof and uses thereof
US7563748B2 (en) 2003-06-23 2009-07-21 Cognis Ip Management Gmbh Alcohol alkoxylate carriers for pesticide active ingredients
CA2530589A1 (en) 2003-07-02 2005-01-20 Sugen Inc. Arylmethyl triazolo and imidazopyrazines as c-met inhibitors
US7312215B2 (en) * 2003-07-29 2007-12-25 Bristol-Myers Squibb Company Benzimidazole C-2 heterocycles as kinase inhibitors
DE10342503A1 (de) * 2003-09-12 2005-04-14 Merck Patent Gmbh Benzyl-Benzimidazolylderivate
ATE472534T1 (de) 2003-09-22 2010-07-15 S Benzimidazolderivate: herstellung und pharmazeutische anwendungen
WO2005051929A1 (en) 2003-11-28 2005-06-09 Ranbaxy Laboratories Limited Conversion of aromatic nitriles into tetrazoles
WO2005051928A1 (en) 2003-11-28 2005-06-09 Ranbaxy Laboratories Limited Process for production of tetrazolyl compounds
WO2005066151A2 (en) 2003-12-19 2005-07-21 Takeda San Diego, Inc. Histone deacetylase inhibitors
WO2005079791A1 (en) 2004-02-12 2005-09-01 Boehringer Ingelheim Pharmaceuticals, Inc. Thiophene -2- carboxylic acid - (1h - benzimidazol - 2 yl) - amide derivatives and related compounds as inhibitors of the tec kinase itk (interleukin -2- inducible t cell kinase) for the treatment of inflammation, immunological and allergic disorders
WO2005082904A1 (ja) 2004-02-26 2005-09-09 Kyowa Hakko Kogyo Co., Ltd. 好中球性炎症疾患の予防及び/または治療剤
CA2558030A1 (en) 2004-03-15 2005-09-29 Eli Lilly And Company 4- (5- (aminomethyl) -indole-1-ylmethyl) -benzamide derivatives and related compounds as opioid receptor antagonists for the treatment of obesity
AU2005224079A1 (en) 2004-03-15 2005-09-29 Anormed, Inc. Process for the synthesis of a CXCR4 antagonist
JP4604086B2 (ja) 2004-05-07 2010-12-22 エグゼリクシス, インコーポレイテッド Rafモジュレーターおよびその使用方法
EP1753730A1 (en) 2004-06-04 2007-02-21 Takeda Pharmaceutical Company Limited Dipeptidyl peptidase inhibitors
EP1769092A4 (en) 2004-06-29 2008-08-06 Europ Nickel Plc IMPROVED LIXIVIATION OF BASE METALS
EP1789045A2 (en) 2004-08-16 2007-05-30 Smithkline Beecham Corporation Chemical compounds
AU2005287137B2 (en) 2004-09-17 2012-03-22 Foldrx Pharmaceuticals, Inc. Compounds, compositions and methods of inhibiting a-synuclein toxicity
CA2585766A1 (en) 2004-11-01 2006-05-11 Nuada, Llc Compounds and methods of use thereof
WO2007134169A2 (en) 2006-05-10 2007-11-22 Nuada, Llc Indole, benzimidazole, and benzolactam boronic acid compounds, analogs thereof and methods of use thereof
US20090264384A1 (en) 2004-11-01 2009-10-22 Nuada, Inc. Indole, benzimidazole, and benzolactam boronic acid compounds, analogs thereof and methods of use thereof
US8003806B2 (en) 2004-11-12 2011-08-23 OSI Pharmaceuticals, LLC Integrin antagonists useful as anticancer agents
WO2006060737A2 (en) 2004-12-03 2006-06-08 Takeda San Diego, Inc. Mitotic kinesin inhibitors
EP1856096B1 (en) 2005-01-10 2010-09-01 Bristol-Myers Squibb Company Phenylglycinamide derivatives useful as anticoagulants
WO2006078907A1 (en) 2005-01-20 2006-07-27 Amgen Inc. 2-substituted benzimidazole derivatives as vanilloid receptor ligands and their use in treatments
CN1834090B (zh) 2005-03-18 2011-06-29 中国科学院上海药物研究所 苯并咪唑类化合物、其制备方法以及用途
US7777040B2 (en) 2005-05-03 2010-08-17 Cgi Pharmaceuticals, Inc. Certain substituted ureas, as modulators of kinase activity
US7470827B2 (en) 2005-06-03 2008-12-30 Ppg Industries Ohio, Inc. Composition for the vapor phase dehydrohalogenation of 1,1,2-trihaloethane to 1,1-dihaloethylene and methods for preparing and using such composition
DE602006020138D1 (de) 2005-06-29 2011-03-31 Compumedics Ltd Sensoranordnung mit leitfähiger brücke
EP1904501A2 (en) 2005-07-11 2008-04-02 Smithkline Beecham Corporation Chemical compounds
JP2007063261A (ja) 2005-08-01 2007-03-15 Kyowa Hakko Kogyo Co Ltd X線照射による肺障害の予防及び/または治療剤
WO2007023880A1 (ja) 2005-08-24 2007-03-01 Kyowa Hakko Kogyo Co., Ltd. ケモカイン産生阻害剤
US7691885B2 (en) 2005-08-29 2010-04-06 Vertex Pharmaceuticals Incorporated Pyridones useful as inhibitors of kinases
TW200720261A (en) 2005-08-31 2007-06-01 Sankyo Co Phenylene derivatives
EP1937650B1 (en) 2005-09-08 2011-06-15 S*BIO Pte Ltd Heterocyclic compounds
WO2007054965A2 (en) 2005-09-23 2007-05-18 Alembic Limited Process for preparation of tetrazoles from aromatic cyano derivatives
GB0520164D0 (en) 2005-10-04 2005-11-09 Novartis Ag Organic compounds
US7465795B2 (en) 2005-12-20 2008-12-16 Astrazeneca Ab Compounds and uses thereof
ES2391472T3 (es) 2005-12-20 2012-11-27 Astrazeneca Ab Derivados de cinnolina sustituidos como moduladores del receptor de GABAA y método para su síntesis
KR20080080211A (ko) 2005-12-23 2008-09-02 아스트라제네카 아베 항박테리아성 피롤로피리딘, 피롤로피리미딘 및피롤로아제핀
JP5165586B2 (ja) 2005-12-28 2013-03-21 バーテックス ファーマシューティカルズ インコーポレイテッド 嚢胞性線維症の処置のためのATP結合カセットトランスポーターのモジュレーターとしての、1−(ベンゾ[d][1,3]ジオキソール−5−イル)−N−(フェニル)シクロプロパン−カルボキサミド誘導体および関連化合物
JP2009525955A (ja) 2006-01-13 2009-07-16 タケダ サン ディエゴ インコーポレイテッド ヒストンデアセチラーゼ阻害剤
WO2007089548A2 (en) 2006-01-26 2007-08-09 Foldrx Pharmaceuticals, Inc. Compounds and methods for modulating protein trafficking
WO2007091950A1 (en) 2006-02-07 2007-08-16 Astrazeneca Ab Benzimidazoles and imidazopyridines useful in the treatment of diseases or disorders associated with cannabinoid receptor 2 (cb2) such as pain
WO2007101347A1 (en) 2006-03-07 2007-09-13 Aegera Therapeutics Inc. Bir domain binding compounds
AU2007233179A1 (en) 2006-03-31 2007-10-11 Astrazeneca Ab Bicyclic benzimidazole compounds and their use as metabotropic glutamate receptor potentiators
DE102006025777A1 (de) 2006-05-31 2007-12-06 Merck Patent Gmbh Neue Materialien für organische Elektrolumineszenzvorrichtungen
EP1873157A1 (en) 2006-06-21 2008-01-02 Bayer Schering Pharma Aktiengesellschaft Pyrazolopyrimidines and salts thereof, pharmaceutical compositions comprising same, methods of preparing same and uses of same
TW200801513A (en) 2006-06-29 2008-01-01 Fermiscan Australia Pty Ltd Improved process
EP1878724A1 (en) 2006-07-15 2008-01-16 sanofi-aventis A regioselective palladium catalyzed synthesis of benzimidazoles and azabenzimidazoles
WO2008012623A1 (en) 2006-07-25 2008-01-31 Pfizer Products Inc. Benzimidazolyl compounds as potentiators of mglur2 subtype of glutamate receptor
WO2008019309A1 (en) 2006-08-04 2008-02-14 Metabasis Therapeutics, Inc. Novel inhibitors of fructose 1,6-bisphosphatase
US8236783B2 (en) 2006-08-15 2012-08-07 Duke University ROS-sensitive iron chelators and methods of using the same
JP2010503651A (ja) * 2006-09-14 2010-02-04 アストラゼネカ アクチボラグ 増殖性障害の治療用のPI3K及びmTOR阻害剤としての2−ベンゾイミダゾリル−6−モルホリノ−フェニルピリミジン誘導体
JO3598B1 (ar) 2006-10-10 2020-07-05 Infinity Discovery Inc الاحماض والاسترات البورونية كمثبطات اميد هيدروليز الحامض الدهني
BRPI0717773A2 (pt) 2006-10-19 2013-11-05 Signal Pharm Llc Composto, composição farmacêutica, e, métodos para tratar ou prevenir câncer, uma condição inflamatória, uma condição imunológica ou uma condição metabólica, e para inibir uma quinase em uma célula que expressa a dita quinase
US20080249081A1 (en) 2006-10-24 2008-10-09 Roger Olsson Compounds for the treatment of pain and screening methods therefor
CL2008000192A1 (es) 2007-01-23 2008-07-25 Palau Pharma Sa Compuestos derivados de purina sustituidos; proceso de preparacion; composicion farmaceutica; y uso en el tratamiento de enfermedades tales como rechazo a transplantes, leucemias, diabetes, entre otras.
US8242151B2 (en) 2007-02-07 2012-08-14 Kyowa Hakko Kirin Co., Ltd. Tricyclic compounds
ES2703592T3 (es) 2007-03-07 2019-03-11 Mei Pharma Inc Combinación de agente anticancerígeno de bencimidazol y un segundo agente anticancerígeno
DK2134691T3 (da) 2007-03-08 2012-05-07 Janssen Pharmaceutica Nv Quinolinonderivativer som parp- og tank-inhibitorer
CN101778838A (zh) 2007-05-24 2010-07-14 惠氏有限责任公司 作为组胺-3拮抗剂的氮杂环基苯甲酰胺衍生物
TW200906825A (en) 2007-05-30 2009-02-16 Scripps Research Inst Inhibitors of protein kinases
ATE539063T1 (de) 2007-06-26 2012-01-15 Sanofi Sa Regioselektive, kupferkatalysierte synthese von benzimidazolen und azabenzimidazolen
EP2014663A1 (de) 2007-07-12 2009-01-14 Bayer Schering Pharma AG Thienopyrimidylamine als Modulatoren des EP2-Rezeptors
EP2014662A1 (de) 2007-07-12 2009-01-14 Bayer Schering Pharma Aktiengesellschaft Indolylalkylthienopyrimidylamine als Modulatoren des EP2-Rezeptors
WO2009025793A2 (en) 2007-08-21 2009-02-26 Senomyx, Inc. Human t2r bitterness receptors and uses thereof
WO2009027736A2 (en) 2007-08-27 2009-03-05 Astrazeneca Ab 2,4 diaminopyrimid'lnes for the treatment of myeloproliferative disorders and cancer
WO2009039140A1 (en) 2007-09-17 2009-03-26 Smithkline Beecham Corporation Pyridopyrimidine derivatives as pi3 kinase inhibitors
US20090118301A1 (en) 2007-11-02 2009-05-07 Arbor Vita Corporation Compositions and Methods for Treating Cancer
UA102828C2 (en) 2007-11-27 2013-08-27 Целльзом Лимитед Amino triazoles as p13k inhibitors
JP2011507851A (ja) 2007-12-19 2011-03-10 ザ スクリプス リサーチ インスティチュート Rhoキナーゼインヒビターとしてのベンゾイミダゾールおよびアナログ
AU2009203693B2 (en) 2008-01-11 2012-06-07 Novartis Ag Pyrimidines as kinase inhibitors
AR070127A1 (es) 2008-01-11 2010-03-17 Novartis Ag Pirrolo - pirimidinas y pirrolo -piridinas
JP2011510010A (ja) 2008-01-15 2011-03-31 ワイス・エルエルシー 3H−[1,2,3]トリアゾロ[4,5−d]ピリミジン化合物、mTORキナーゼおよびPI3キナーゼ阻害剤としてのそれらの使用、ならびにそれらの合成
JP2011510030A (ja) 2008-01-22 2011-03-31 ベーリンガー インゲルハイム インターナショナル ゲゼルシャフト ミット ベシュレンクテル ハフツング 置換アミノ−ベンゾイミダゾール類、該化合物を含む医薬、これらの使用及びこれらの製造方法
FR2928924B1 (fr) 2008-03-21 2010-04-23 Sanofi Aventis Derives polysubstitues de 6-heteroaryle-imidazo°1,2-a! pyridines, leur preparation et leur application en therapeutique
ATE513818T1 (de) 2008-03-27 2011-07-15 Janssen Pharmaceutica Nv Chinazolinonderivate als tubulinpolymerisationshemmer
EP2271626B1 (en) 2008-03-27 2014-11-26 Janssen Pharmaceutica, N.V. Tetrahydrophenanthridinones and tetrahydrocyclopentaquinolinones as parp and tubulin polymerization inhibitors
US20090253161A1 (en) 2008-04-03 2009-10-08 Duke University Fluorescent prochelators for cellular iron detection
TW201000107A (en) 2008-04-09 2010-01-01 Infinity Pharmaceuticals Inc Inhibitors of fatty acid amide hydrolase
CN102056932A (zh) 2008-06-04 2011-05-11 阿斯利康(瑞典)有限公司 作为抗菌药的噻唑并[5,4-b]吡啶和唑[5,4-b]吡啶衍生物
JP2010031250A (ja) 2008-06-23 2010-02-12 Sumitomo Chemical Co Ltd 組成物及び該組成物を用いてなる発光素子
WO2010000020A1 (en) 2008-06-30 2010-01-07 Cathrx Ltd A catheter
AU2009266889B2 (en) 2008-07-03 2013-05-02 Glaxosmithkline Llc Benzimidazoles and related analogs as sirtuin modulators
AU2009269087A1 (en) * 2008-07-07 2010-01-14 Xcovery Holding Company Llc PI3K isoform selective inhibitors
WO2010006096A1 (en) 2008-07-11 2010-01-14 Smithkline Beecham Corporation Processes for the preparation of anti-viral compounds and compositions containing them
US8501957B2 (en) 2008-12-10 2013-08-06 China Medical University Benzimidazole compounds and their use as anticancer agents
PE20110924A1 (es) 2009-01-23 2011-12-24 Rigel Pharmaceuticals Inc Derivados de 2, 4-diamino-pirimidina n2, n4-disustituidos como inhibidores de jak3
MY160130A (en) 2009-02-27 2017-02-28 Enanta Pharm Inc Hepatitis c virus inhibitors
US20120006688A1 (en) 2009-03-18 2012-01-12 Basf Se Electrolyte and surface-active additives for the electrochemical deposition of smooth, dense aluminum layers from ionic liquids
EP2408449A4 (en) 2009-03-18 2012-08-08 Univ Leland Stanford Junior METHODS AND COMPOSITIONS FOR TREATING INFECTION WITH A FLAVIVIRIDAE FAMILY VIRUS
WO2010108187A2 (en) 2009-03-20 2010-09-23 Brandeis University Compounds and methods for treating mammalian gastrointestinal microbial infections
WO2010114726A1 (en) 2009-03-31 2010-10-07 Merck Sharp & Dohme Corp. Aminobenzotriazole derivatives
BRPI1015367B8 (pt) 2009-04-02 2021-05-25 Centro Nac De Investigaciones Oncologicas Cnio derivados imidazo[2,1-b][1,3,4]tiadiazol
WO2010118208A1 (en) 2009-04-09 2010-10-14 Exelixis, Inc. Benzoxazepin-4- (5h) -yl derivatives and their use to treat cancer
JP2012525390A (ja) 2009-04-27 2012-10-22 ハイ ポイント ファーマシューティカルズ,リミティド ライアビリティ カンパニー 置換イミダゾ[1,2−a]ピリジン誘導体、医薬組成物、及びβ−セクレターゼ阻害剤としての使用方法
WO2010126922A1 (en) 2009-04-30 2010-11-04 Glaxosmithkline Llc Benzimidazolecarboxamides as inhibitors of fak
KR20120034666A (ko) 2009-05-22 2012-04-12 엑셀리시스, 인코포레이티드 증식성 질환에 대한 벤족사제핀 기반 p13k/mt0r의 억제제
AU2010254200A1 (en) 2009-05-26 2011-12-08 Exelixis, Inc. Benzoxazepines as inhibitors of PI3K/m TOR and methods of their use and manufacture
WO2010141360A1 (en) 2009-06-05 2010-12-09 Merck Sharp & Dohme Corp. Biaryl benzotriazole derivatives
WO2010144686A1 (en) 2009-06-10 2010-12-16 North Carolina State University Inhibition and dispersion of bacterial biofilms with benzimidazole derivatives
WO2011000020A1 (en) 2009-06-12 2011-01-06 Sbc Research Pty Ltd Enhanced method of detection
JP2011003793A (ja) 2009-06-19 2011-01-06 Idemitsu Kosan Co Ltd 有機el素子
US20120178769A1 (en) 2009-07-15 2012-07-12 The University Of Medicine And Dentistry Novel compounds for inhibiting eef-2 kinase activity
CA2779177C (en) 2009-10-29 2016-08-30 Bristol-Myers Squibb Company Quinuclidine compounds as alpha-7 nicotinic acetylcholine receptor ligands
KR20120123325A (ko) 2009-12-15 2012-11-08 시오노기세야쿠 가부시키가이샤 혈관 내피 리파아제 저해 활성을 갖는 옥사디아졸 유도체
ES2362337B1 (es) 2009-12-17 2012-05-16 Consejo Superior De Investigaciones Cient�?Ficas (Csic) Derivados de aminociclitoles, procedimiento de obtencion y usos.
US20130196990A1 (en) * 2010-10-06 2013-08-01 Junya Qu Benzimidazole Derivatives As PI3 Kinase Inhibitors
EP3056092A4 (en) 2013-10-11 2016-08-17 Unicharm Corp PET FOOD

Also Published As

Publication number Publication date
US20140018534A1 (en) 2014-01-16
MX337662B (es) 2016-03-14
ES2616238T3 (es) 2017-06-12
CN103124496B (zh) 2016-03-30
AU2011312594B2 (en) 2014-10-02
WO2012047538A1 (en) 2012-04-12
IL225140A (en) 2016-09-29
TWI513690B (zh) 2015-12-21
US8435988B2 (en) 2013-05-07
UY33648A (es) 2012-04-30
US9062003B2 (en) 2015-06-23
US8865912B2 (en) 2014-10-21
MX2013003918A (es) 2013-06-05
CA2812608A1 (en) 2012-04-12
PE20140192A1 (es) 2014-02-24
CY1118792T1 (el) 2017-07-12
MY170236A (en) 2019-07-11
CO6700852A2 (es) 2013-06-28
MA34591B1 (fr) 2013-10-02
EP2624696A4 (en) 2014-03-26
EP3170813B1 (en) 2018-12-12
TW201307297A (zh) 2013-02-16
ZA201301951B (en) 2014-08-27
US8541411B2 (en) 2013-09-24
EP2624696B1 (en) 2016-12-21
SG188974A1 (en) 2013-06-28
EP3170813A1 (en) 2017-05-24
US10314845B2 (en) 2019-06-11
US9872860B2 (en) 2018-01-23
EA022623B1 (ru) 2016-02-29
HRP20170279T1 (hr) 2017-04-07
NZ608069A (en) 2014-06-27
AU2011312594A1 (en) 2013-04-11
JP2013539753A (ja) 2013-10-28
AR083296A1 (es) 2013-02-13
EA201390302A1 (ru) 2013-09-30
US20130197221A1 (en) 2013-08-01
CA2812608C (en) 2020-07-14
US20170112844A1 (en) 2017-04-27
EP2624696A1 (en) 2013-08-14
US20150246889A1 (en) 2015-09-03
PL2624696T3 (pl) 2017-07-31
US20120088767A1 (en) 2012-04-12
JO3194B1 (ar) 2018-03-08
CL2013000935A1 (es) 2013-07-05
RS55662B1 (sr) 2017-06-30
CN103124496A (zh) 2013-05-29
PT2624696T (pt) 2017-03-21
DK2624696T3 (en) 2017-03-13
DOP2013000059A (es) 2013-05-31
US8674090B2 (en) 2014-03-18
CR20130192A (es) 2013-10-03
ES2714384T3 (es) 2019-05-28
US9156797B2 (en) 2015-10-13
US20140142321A1 (en) 2014-05-22
BR112013008259A2 (pt) 2017-12-12
US20130196990A1 (en) 2013-08-01
US20140378456A1 (en) 2014-12-25
US20180263994A1 (en) 2018-09-20
KR20130099142A (ko) 2013-09-05
US20190255064A1 (en) 2019-08-22
KR101594002B1 (ko) 2016-02-15
JP5719028B2 (ja) 2015-05-13
LT2624696T (lt) 2017-03-10
ME02663B (me) 2017-06-20
US10660898B2 (en) 2020-05-26
SI2624696T1 (sl) 2017-04-26

Similar Documents

Publication Publication Date Title
DK2624696T3 (en) BENZIMIDAZOLD DERIVATIVES AS PI3-KINASE INHIBITORS
US8906910B2 (en) Imidazopyridine derivatives as PI3 kinase
US8952018B2 (en) Pharmaceutical combination of MEK inhibitor and B-Raf inhibitors
US8778937B2 (en) Benzimidazole boronic acid derivatives as PI3 kinase inhibitors
US20160367662A1 (en) Combinations of trametinib, panitumumab and dabrafenib for the treatment of cancer
JP2016503399A (ja) 組合せ