WO2008027584A2 - 5- (2-furyl)-1, 3-thiazole derivatives useful as inhibitors of phosphatidylinositol 3-kinase - Google Patents

5- (2-furyl)-1, 3-thiazole derivatives useful as inhibitors of phosphatidylinositol 3-kinase Download PDF

Info

Publication number
WO2008027584A2
WO2008027584A2 PCT/US2007/019261 US2007019261W WO2008027584A2 WO 2008027584 A2 WO2008027584 A2 WO 2008027584A2 US 2007019261 W US2007019261 W US 2007019261W WO 2008027584 A2 WO2008027584 A2 WO 2008027584A2
Authority
WO
WIPO (PCT)
Prior art keywords
compound
optionally substituted
salt
aliphatic
nhc
Prior art date
Application number
PCT/US2007/019261
Other languages
French (fr)
Other versions
WO2008027584A3 (en
Inventor
Tiansheng Wang
Jeremy Green
Mark Cornebise
Brian Ledford
Jonathan Parsons
Adam Tanner
James Westcott
Original Assignee
Vertex Pharmaceuticals Incorporated
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 Vertex Pharmaceuticals Incorporated filed Critical Vertex Pharmaceuticals Incorporated
Priority to US12/438,995 priority Critical patent/US8268818B2/en
Priority to MX2009002239A priority patent/MX2009002239A/en
Priority to CA002662196A priority patent/CA2662196A1/en
Priority to JP2009526754A priority patent/JP2010502628A/en
Priority to AU2007290359A priority patent/AU2007290359B2/en
Priority to NZ575346A priority patent/NZ575346A/en
Priority to EP07811661A priority patent/EP2061787A2/en
Publication of WO2008027584A2 publication Critical patent/WO2008027584A2/en
Publication of WO2008027584A3 publication Critical patent/WO2008027584A3/en

Links

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/04Antipruritics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/16Otologicals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis

Definitions

  • the present invention relates to compounds useful as inhibitors of phosphatidylinositol 3-kinase (PI3K).
  • PI3K phosphatidylinositol 3-kinase
  • the invention also provides pharmaceutically acceptable compositions comprising the compounds of the invention and methods of using the compositions in the treatment of various disorders.
  • P 13 Ks are a family of lipid kinases that catalyze the phosphorylation of the membrane lipid phosphatidylinositol (PI) on the 3'-OH of the inositol ring to produce PI 3-phosphate [PI(3)P, PIP], PI 3,4-bisphosphate [PI(3,4)P 2 , PIP2] and PI 3,4,5-trisphosphate [PI(3,4,5)P 3 , PIP3].
  • PI(3,4)P 2 and PI(3,4,5)P 3 act as recruitment sites for various intracellular signaling proteins, which in turn form signaling complexes to relay extracellular signals to the cytoplasmic face of the plasma membrane.
  • Class Ia includes PI3K ⁇ , PI3K ⁇ and PI3K ⁇ . All of the class Ia enzymes are heterodimeric complexes comprising a catalytic subunit (pi 10a, pi lO ⁇ or pi lO ⁇ ) associated with an SH2 domain-containing p85 adapter subunit. Class Ia PI3Ks are activated through tyrosine kinase signaling and are involved in cell proliferation and survival. PI3K ⁇ and PI3K ⁇ have also been implicated in tumorigenesis in a variety of human cancers. Thus, pharmacological inhibitors of PI3K ⁇ and PI3K ⁇ are useful for treating various types of cancer.
  • PI3K ⁇ the only member of the Class Ib PI3Ks, consists of a catalytic subunit pi 1Oy, which is associated with a pi 01 regulatory subunit.
  • PI3K ⁇ is regulated by G protein-coupled receptors (GPCRs) via association with ⁇ subunits of heterotrimeric G proteins.
  • GPCRs G protein-coupled receptors
  • PI3K ⁇ is expressed primarily in hematopoietic cells and cardiomyocytes and is involved in inflammation and mast cell function.
  • pharmacological inhibitors of PI3K ⁇ are useful for treating a variety of inflammatory diseases, allergies and cardiovascular diseases.
  • PBK inhibitors Although a number of PBK inhibitors have been developed, there is a need for additional compounds to inhibit PI3Ks for treating various disorders and diseases. Accordingly, it would be desirable to develop additional compounds that are useful as inhibitors of PBK.
  • R 1 , R 2 , R 3 , R 9 , Y, T, L and n are as defined herein.
  • These compounds, and pharmaceutically acceptable compositions thereof, are useful for treating or lessening the severity of a variety of disorders, including cancer, autoimmune diseases, inflammatory diseases, cardiovascular diseases, diabetes, allergic diseases, asthma or organ transplantation rejection in a patient.
  • the compounds and compositions provided by this invention are also useful for the study of PBK in biological and pathological phenomena; the study of intracellular signal transduction pathways mediated by such kinases; and the comparative evaluation of new kinase inhibitors.
  • the invention also provides assays for identifying inhibitors of PBK.
  • compounds of the invention may optionally be substituted with one or more substituents, such as are illustrated generally above, or as exemplified by particular classes, subclasses, and species of the invention.
  • substituents such as are illustrated generally above, or as exemplified by particular classes, subclasses, and species of the invention.
  • the phrase “optionally substituted” is used interchangeably with the phrase “substituted or unsubstituted.”
  • substituted refers to the replacement of one or more hydrogen radicals in a given structure with the radical of a specified substituent.
  • an optionally substituted group may have a substituent at each substitutable position of the group. When more than one position in a given structure can be substituted with more than one substituent selected from a specified group, the substituent may be either the same or different at each position.
  • stable refers to compounds that are not substantially altered when subjected to conditions to allow for their production, detection, and, preferably, their recovery, purification, and use for one or more of the purposes disclosed herein.
  • a stable compound or chemically feasible compound is one that is not substantially altered when kept at a temperature of 40 0 C or less, in the absence of moisture or other chemically reactive conditions, for at least a week.
  • aliphatic or "aliphatic group”, as used herein, means a straight-chain (i.e., unbranched) or branched, substituted or unsubstituted hydrocarbon chain that is completely saturated or that contains one or more units of unsaturation.
  • aliphatic groups contain 1-20 aliphatic carbon atoms. In some embodiments, aliphatic groups contain 1-10 aliphatic carbon atoms. In other embodiments, aliphatic groups contain 1-8 aliphatic carbon atoms. In still other embodiments, aliphatic groups contain 1-6 aliphatic carbon atoms, and In yet other embodiments, aliphatic groups contain 1-4 aliphatic carbon atoms.
  • Suitable aliphatic groups include, but are not limited to, linear or branched, substituted or unsubstituted alkyl, alkenyl, or alkynyl groups. Further examples of aliphatic groups include methyl, ethyl, propyl, butyl, isopropyl, isobutyl, vinyl, and sec-butyl.
  • cycloaliphatic refers to a hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic, that has a single point of attachment to the rest of the molecule, and wherein any individual ring in said bicyclic ring system has 3-7 members.
  • cycloaliphatic refers to a monocyclic C 3 -Cg hydrocarbon or bicyclic C 8 -Ci 2 hydrocarbon. Suitable cycloaliphatic groups include, but are not limited to, cycloalkyl, cycloalkenyl, and cycloalkynyl.
  • aliphatic groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cycloheptyl, and cycloheptenyl.
  • heterocycle refers to a monocyclic, bicyclic, or tricyclic ring system in which one or more ring members are an independently selected heteroatom and that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic, that has a single point of attachment to the rest of the molecule.
  • the "heterocycle", “heterocyclyl” or “heterocyclic” group has three to fourteen ring members in which one or more ring members is a heteroatom independently selected from oxygen, sulfur, nitrogen, or phosphorus, and each ring in the system contains 3 to 7 ring members.
  • heterocyclic rings include, but are not limited to, the following monocycles: 2-tetrahydrofuranyl, 3-tetrahydrofuranyl, 2-tetrahydrothiophenyl, 3-tetrahydrothiophenyl, 2-morpholino, 3-mo ⁇ holino, 4-morpholino, 2-thiomorpholino, 3-thiomorpholino, 4-thiomorpholino, 1-pyrrolidinyl, 2-pyrrolidinyl, 3-pyrrolidinyl, 1-tetrahydropiperazinyl, 2-tetrahydropiperazinyl, 3-tetrahydropiperazinyl, 1 -piperidinyl, 2-piperidinyl, 3-piperidinyl, 1-pyrazolinyl, 3-pyrazolinyl, 4-pyrazolinyl, 5-pyrazolinyl, 1 -piperidinyl, 2-piperidinyl, 3-piperidinyl, 4-piperidinyl, 2-thi
  • heteroatom means one or more of oxygen, sulfur, nitrogen, phosphorus, or silicon, including any oxidized form of nitrogen, sulfur, phosphorus, or silicon, the quaternized form of any basic nitrogen, or a substitutable nitrogen of a heterocyclic ring, for example N (as in 3,4-dihydro-2H-pyrrolyl), NH (as in pyrrolidinyl) or NR + (as in N-substituted pyrrolidinyl).
  • aralkyl refers to monocyclic, bicyclic, and tricyclic carbocyclic ring systems having a total of six to fourteen ring members, wherein at least one ring in the system is aromatic, wherein each ring in the system contains 3 to 7 ring members and that has a single point of attachment to the rest of the molecule.
  • aryl may be used interchangeably with the term “aryl ring”. Examples of aryl rings would include phenyl, naphthyl, and anthracene.
  • heteroaryl used alone or as part of a larger moiety as in
  • heteroarylkyl refers to monocyclic, bicyclic, and tricyclic ring systems having a total of five to fourteen ring members, wherein at least one ring in the system is aromatic, at least one ring in the system contains one or more heteroatoms, wherein each ring in the system contains 3 to 7 ring members and that has a single point of attachment to the rest of the molecule.
  • heteroaryl may be used interchangeably with the term “heteroaryl ring” or the term “heteroaromatic”.
  • heteroaryl rings include the following monocycles: 2-furanyl, 3-furanyl, N-imidazolyl, 2-imidazolyl, 4-imidazolyl, 5- imidazolyl, 3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, 2-oxazolyl, 4-oxazolyl, 5-oxazolyl, N-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl, pyridazinyl (e.g., 3-pyridazinyl), 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, tetrazolyl (e.g., 5-tetrazolyl), triazolyl (e.g., 2-triazolyl and 5-triazolyl), 2- thienyl, 3-
  • indolyl e.g., 2-indolyl
  • purinyl e.g., quinolinyl (e.g., 2- quinolinyl, 3-quinolinyl, 4-quinolinyl)
  • isoquinolinyl e.g., 1-isoquinolinyl, 3- isoquinolin
  • an aryl (including aralkyl, aralkoxy, aryloxyalkyl and the like) or heteroaryl (including heteroaralkyl and heteroarylalkoxy and the like) group may contain one or more substituents. Suitable substituents on the unsaturated carbon atom of an aryl or heteroaryl group are selected from those listed in the definitions of R 2 and R 4 below.
  • Optional substituents on the aliphatic group of R° are selected from NH 2 , NH(Ci -4 aliphatic), N(Ci.4aliphatic) 2 , halogen, Ci ⁇ aliphatic, OH, O(Ci ⁇ aliphatic), NO 2 , CN, CO 2 H, CO 2 (C ⁇ aliphatic), 00IaIoCi -4 aliphatic), or haloCi- 4 aliphatic, wherein each of the foregoing Ci ⁇ aliphatic groups of R° is unsubstituted.
  • an aliphatic or heteroaliphatic group, or a non- aromatic heterocyclic ring may contain one or more substituents.
  • Optional substituents on the aliphatic group of R are selected from NH 2 , NH(Ci -4 aliphatic), N(Ci -4 aliphatic) 2 , halogen, Ci -4 aliphatic, OH, 0(Ci -4 aliphatic), NO 2 , CN, CO 2 H, CO 2 (Ci -4 aliphatic), O(halo C1.4 aliphatic), or halo(Ci -4 aliphatic), wherein each of the foregoing Cj ⁇ aliphatic groups of R is unsubstituted.
  • Optional substituents on the aliphatic group or the phenyl ring of R + are selected from NH 2 , NH(Ci -4 aliphatic), N(Ci -4 aliphatic) 2 , halogen, Ci -4 aliphatic, OH, 0(Ci -4 aliphatic), NO 2 , CN, CO 2 H, CO 2 (Ci -4 aliphatic), O(halo C 1-4 aliphatic), orhalo(C t-4 aliphatic), wherein each of the foregoing Ci -4 aliphatic groups OfR + is unsubstituted.
  • two independent occurrences of R 0 may be taken together with the atom(s) to which each variable is bound to form a 5-8-membered heterocyclyl, aryl, or heteroaryl ring or a 3-8-membered cycloalkyl ring.
  • Exemplary rings that are formed when two independent occurrences of R 0 (or R + , or any other variable similarly defined herein) are taken together with the atom(s) to which each variable is bound include, but are not limited to the following: a) two independent occurrences of R° (or R + , or any other variable similarly defined herein) that are bound to the same atom and are taken together with that atom to form a ring, for example, N(R°)2, where both occurrences of R° are taken together with the nitrogen atom to form a piperidin-1-yl, piperazin-1-yl, or morpholin-4-yl group; and b) two independent occurrences of R° (or R + , or any other variable similarly defined herein) that are bound to different atoms and are taken together with both of those atoms to form a ring, for example where a phenyl group is substituted with two occurrences of OR 0
  • an alkyl or aliphatic chain can be optionally interrupted with another atom or group. This means that a methylene unit of the alkyl or aliphatic chain is optionally replaced with said other atom or group.
  • the optional replacements form a chemically stable compound.
  • Optional interruptions can occur both within the chain and at either end of the chain; i.e. both at the point of attachment and/or also at the terminal end.
  • Two optional replacements can also be adjacent to each other within a chain so long as it results in a chemically stable compound.
  • the replacement or interruption occurs at the terminal end, the replacement atom is bound to an H on the terminal end. For example, if -CH 2 CH 2 CH 3 were optionally interrupted with -O-, the resulting compound could be -OCH 2 CH 3 , -CH 2 OCH 3 , or -CH 2 CH 2 OH.
  • a bond drawn from a substituent to the center of one ring within a multiple-ring system represents substitution of the substituent at any substitutable position in any of the rings within the multiple ring system.
  • Figure a represents possible substitution in any of the positions shown in Figure b.
  • each substituent only represents substitution on the ring to which it is attached.
  • Y is an optionally substituent for ring A only
  • X is an optional substituent for ring B only.
  • structures depicted herein are also meant to include all isomeric (e.g., enantiomeric, diastereomeric, and geometric (or conformational)) forms of the structure; for example, the R and S configurations for each asymmetric center, (Z) and (E) double bond isomers, and (Z) and (E) conformational isomers. Therefore, single stereochemical isomers as well as enantiomeric, diastereomeric, and geometric (or conformational) mixtures of the present compounds are within the scope of the invention.
  • the present invention relates to a compound of formula I:
  • Y is -C(O)- or a bond
  • R 1 is a C
  • T is -SO 2 NR 4 -, -CONR 4 - or -C(O)O-;
  • R 3 is H or an optionally substituted group selected from a Cue aliphatic, a C 3- Io monocyclic orbicyclic cycloaliphatic, a C 6 -io monocyclic orbicyclic aryl, a 5-10 membered monocyclic or bicyclic heteroaryl, or a 5-10 membered monocyclic or bicyclic heterocyclyl, wherein R 3 is optionally substituted with 1-6 occurrences of J** 3 , or R 3 and R 4 , together with the nitrogen to which R 4 is attached, form a 5- 10 membered monocyclic or bicyclic heterocyclyl or heteroaryl; wherein n is 0 and said heterocyclyl or heteroaryl is optionally substituted with 1-6 occurrences ofJ 113 ; each J* 3 is independently selected from -(U) m -X;
  • U is a Ci- 4 aliphatic, wherein up to two methylene units are optionally and independently replaced by G u and wherein U is optionally substituted with 1-4 J u ;
  • X is H, halogen, OH, OR, NO 2 , NH 2 , NHR, NR 2 , SH, SR, CN, C(O)NH 2 , C(O)NHR, C(O)NR 2 , C(O)OH, C(O)OR, NHC(O)H, NHC(O)R, NRC(O)H, NRC(O)R, NHC(O)OH, NHC(O)OR, NRC(O)OH, NRC(O)OR, or a group selected from a Cue aliphatic, a C 3 .io monocyclic or bicyclic cycloaliphatic, a C 6- io monocyclic or bicyclic aryl, a 5-10 membered monocyclic or bicyclic heteroaryl, or a 5-10 membered monocyclic or bicyclic heterocyclyl, wherein said group is optionally substituted with 1-4 J x , or two X, together with the carbon(s) to which
  • R 9 is selected from H; halogen; OH; NO 2 ; NH 2 ; SH; CN; or a group selected from a Cj -6 aliphatic or a C3 -7 cycloaliphatic, wherein said group is optionally substituted with 1-6 occurrences of halogen, OH, NO 2 , NH 2 , SH or CN.
  • Y is C(O) and the compound is of formula II:
  • R 1 is a C 1-6 aliphatic optionally substituted with 1-6 occurrences of J .
  • R 1 is a Ci- 3 aliphatic optionally substituted with 1-3 occurrences of J .
  • R 1 is a Ci -3 aliphatic.
  • R 1 is CH 3 , CH 2 CH 3 or cyclopropyl.
  • R 2 is Ci -3 aliphatic, wherein R 2 is optionally substituted with 1-3 occurrences of J 1 * 2 .
  • R 2 is Ci -3 aliphatic.
  • R 2 is CH 3 .
  • said compound is of formula HIa, IHb or HIc:
  • said compound is of formula IHa', HIb' or IHc':
  • R and R together with the nitrogen to which R 4 is attached, form a 5-10 membered monocyclic or bicyclic heterocyclyl or heteroaryl; wherein said heterocyclyl or heteroaryl is optionally substituted with 1-6 occurrences of J 1 * 3 .
  • said heterocyclyl or heteroaryl formed by R 3 and R 4 is a 5-6 membered monocyclic heterocyclyl or 9-10 membered heterocyclyl or heteroaryl; wherein said monocyclic or bicyclic heterocyclyl or bicyclic heteroaryl contains 1-3 heteroatoms and is optionally substituted with 1-3 occurrences of J R3 .
  • said monocyclic or bicyclic heterocyclyl or bicyclic heteroaryl contains 1-2 heteroatoms and is optionally substituted with 1-2 occurrences of J R3 .
  • said monocyclic or bicyclic heterocyclyl or bicyclic heteroaryl is selected from the following:
  • each occurrence of J R3 is selected from J 113 is selected from halogen; OH; OR; CN; NH 2 ; NHR; NR 2 ; C(O)NH 2 ; C(O)NHR; C(O)NR 2 ; NHCOR; C(O)OR; C(O)OH; oxo; (CH 2 ) 0-3 phenyl optionally substituted with halogen, OH, OR, NO 2 , NH 2 , SH or CN; or Ci -4 aliphatic optionally substituted with halogen, OH, OR, NO 2 , NH 2 , SH or CN.
  • R is H or an optionally substituted group selected from a C ⁇ s aliphatic, a C 3 -io monocyclic or bicyclic cycloaliphatic, a C 6- 1 0 monocyclic or bicyclic aryl, a 5-10 membered monocyclic or bicyclic heteroaryl, or a 5-10 membered monocyclic or bicyclic heterocyclyl, wherein R 3 is optionally substituted with 1-6 occurrences of J m .
  • n is 1 and L is a Cu aliphatic optionally substituted with 1-2 occurrences of R'.
  • L is -CH 2 - or CH substituted with a Ci -2 aliphatic, which Cj -2 aliphatic is optionally substituted with 1-2 occurrences of halogen, OH, NO 2 , NH 2 , SH or CN.
  • L is -CH 2 -.
  • n is 0.
  • R 3 is a Ci -4 aliphatic, a C 3 - 6 monocyclic cycloaliphatic, phenyl, a 5-6 membered monocyclic heteroaryl, or a 5-10 membered monocyclic or bicyclic heterocyclyl, wherein R 3 is optionally substituted with 1-4 occurrences of J R3 .
  • R 3 is a Ci -4 aliphatic, a C 3 - 6 monocyclic cycloaliphatic, phenyl, a 5-6 membered monocyclic heteroaryl, or a 5-6 membered monocyclic heterocyclyl, wherein R 3 is optionally substituted with 1-4 occurrences of J 113 .
  • each occurrence of J R3 is selected from halogen; OH; OR; CN; NH 2 ; NHR; NR 2 ; C(O)NH 2 ; C(O)NHR; C(O)NR 2 ; NHCOR; C(O)OR; C(O)OH; oxo; (CH 2 )o- 3 ⁇ henyl optionally substituted with halogen, OH, OR, NO 2 , NH 2 , SH or CN; or Ci -4 aliphatic optionally substituted with halogen, OH, OR, NO 2 , NH 2 , SH or CN.
  • R 4 is
  • R 4 is H, CH 3 , CH 2 CH 3 , CH 2 CH 2 OH, CH 2 CHCH 2 , cyclopropyl or CH 2 phenyl. In yet a further embodiment, R 4 is H or CH 3 .
  • R 8 is selected from H, cyclopropyl, or a Ci -3 aliphatic optionally substituted with 1-3 occurrences of halogen or OH. In a further embodiment, R 8 is selected from H, halogen, methyl or ethyl. In yet a further embodiment, R 8 is H.
  • R 9 is selected from H, OH, halogen, cyclopropyl, or a C 1 - 3 aliphatic optionally substituted with 1-3 occurrences of halogen or OH.
  • R 9 is selected from H, OH, halogen, methyl or ethyl.
  • R 9 is H or methyl.
  • the invention provides a compound of Table I or Table 2: Table 1
  • the invention provides a pharmaceutical composition comprising a compound of any of the formulae or classes described herein.
  • the invention provides a pharmaceutical composition comprising a compound of Table 1.
  • the composition additionally comprises an additional therapeutic agent.
  • the invention provides a composition comprising a compound of this invention or a pharmaceutically acceptable derivative thereof and a pharmaceutically acceptable carrier, adjuvant, or vehicle.
  • the amount of compound in a composition of this invention is such that is effective to measurably inhibit a PBK, particularly PI3K ⁇ , in a biological sample or in a patient.
  • the amount of compound in the compositions of this invention is such that is effective to measurably inhibit PI3K ⁇ .
  • the composition of this invention is formulated for administration to a patient in need of such composition. Most preferably, the composition of this invention is formulated for oral administration to a patient.
  • patient means an animal, preferably a mammal, and most preferably a human.
  • compositions comprising any of the compounds or pharmaceutically acceptable salts thereof, as described herein, and further comprises a pharmaceutically acceptable carrier, adjuvant or vehicle.
  • these compositions optionally further comprise one or more additional therapeutic agents.
  • certain of the compounds of present invention can exist in free form for treatment, or where appropriate, as a pharmaceutically acceptable derivative thereof.
  • a pharmaceutically acceptable derivative includes, but is not limited to, pharmaceutically acceptable prodrugs, salts, esters, salts of such esters, or any other adduct or derivative which upon administration to a patient in need is capable of providing, directly or indirectly, a compound as otherwise described herein, or a metabolite or residue thereof.
  • the term "inhibitorily active metabolite or residue thereof means that a metabolite or residue thereof is also an inhibitor of PBK.
  • the term "pharmaceutically acceptable salt” refers to those salts which are, within the scope of sound medical judgement, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like.
  • compositions of this invention include those derived from suitable inorganic and organic acids and bases.
  • Examples of pharmaceutically acceptable, nontoxic acid addition salts are salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange.
  • inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid
  • organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange.
  • salts include adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionatc, digluconate, dodecyl sulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2- hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, olcate, oxalate, palmitate, pamoate, pect
  • Salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium and N + (Ci. 4 alkyl) 4 salts.
  • This invention also envisions the quaternization of any basic nitrogen-containing groups of the compounds disclosed herein. Water or oil-soluble or dispersable products may be obtained by such quaternization.
  • Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like.
  • Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, loweralkyl sulfonate and aryl sulfonate.
  • the pharmaceutically acceptable compositions of the present invention additionally comprise a pharmaceutically acceptable carrier, adjuvant, or vehicle, which, as used herein, includes any and all solvents, diluents, or other liquid vehicle, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, solid binders, lubricants and the like, as suited to the particular dosage form desired.
  • a pharmaceutically acceptable carrier, adjuvant, or vehicle which, as used herein, includes any and all solvents, diluents, or other liquid vehicle, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, solid binders, lubricants and the like, as suited to the particular dosage form desired.
  • Remington's Pharmaceutical Sciences, Sixteenth Edition, E. W. Martin (Mack Publishing Co., Easton, Pa., 1980) discloses various carriers used in formulating pharmaceutically acceptable compositions
  • any conventional carrier medium is incompatible with the compounds of the invention, such as by producing any undesirable biological effect or otherwise interacting in a deleterious manner with any other component(s) of the pharmaceutically acceptable composition, its use is contemplated to be within the scope of this invention.
  • Some examples of materials which can serve as pharmaceutically acceptable carriers include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, or potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, wool fat, sugars such as lactose, glucose and sucrose; starches such as corn starch and potato starch; cellulose and its derivatives such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; ge
  • the term "measurably inhibit”, as used herein means a measurable change in kinase activity, particularly POK activity, between a sample comprising a compound of this invention and a PI3K and an equivalent sample comprising PI3K in the absence of said compound.
  • compositions of the present invention may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir.
  • parenteral as used herein includes subcutaneous, intravenous, intramuscular, intra-articular, intra-synovial, intrasternal, intrathecal, intraocular, intrahepatic, intralesional and intracranial injection or infusion techniques.
  • the compositions are administered orally, intraperitoneally or intravenously.
  • Sterile injectable forms of the compositions of this invention may be aqueous or oleaginous suspension.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example as a solution in 1,3- butanediol.
  • a non-toxic parenterally-acceptable diluent or solvent for example as a solution in 1,3- butanediol.
  • acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium. [ 0158 ]
  • any bland fixed oil may be employed including synthetic mono- or di-glycerides.
  • Fatty acids such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically- acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions.
  • oils such as olive oil or castor oil
  • These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant, such as carboxymethyl cellulose or similar dispersing agents that are commonly used in the formulation of pharmaceutically acceptable dosage forms including emulsions and suspensions.
  • a long-chain alcohol diluent or dispersant such as carboxymethyl cellulose or similar dispersing agents that are commonly used in the formulation of pharmaceutically acceptable dosage forms including emulsions and suspensions.
  • Other commonly used surfactants such as Tweens, Spans and other emulsifying agents or bioavailability enhancers which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms may also be used for the purposes of formulation.
  • compositions of this invention may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, aqueous suspensions or solutions.
  • carriers commonly used include lactose and corn starch.
  • Lubricating agents such as magnesium stearate, are also typically added.
  • useful diluents include lactose and dried cornstarch.
  • aqueous suspensions are required for oral use, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening, flavoring or coloring agents may also be added.
  • compositions of this invention may be administered in the form of suppositories for rectal administration.
  • suppositories for rectal administration.
  • suppositories can be prepared by mixing the agent with a suitable non-irritating excipient that is solid at room temperature but liquid at rectal temperature and therefore will melt in the rectum to release the drug.
  • suitable non-irritating excipient include cocoa butter, beeswax and polyethylene glycols.
  • compositions of this invention may also be administered topically, especially when the target of treatment includes areas or organs readily accessible by topical application, including diseases of the eye, the skin, or the lower intestinal tract. Suitable topical formulations are readily prepared for each of these areas or organs.
  • Topical application for the lower intestinal tract can be effected in a rectal suppository formulation (see above) or in a suitable enema formulation. Topically-transdermal patches may also be used.
  • the pharmaceutically acceptable compositions may be formulated in a suitable ointment containing the active component suspended or dissolved in one or more carriers.
  • Carriers for topical administration of the compounds of this invention include, but are not limited to, mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene, polyoxypropylene compound, emulsifying wax and water.
  • the pharmaceutically acceptable compositions can be formulated in a suitable lotion or cream containing the active components suspended or dissolved in one or more pharmaceutically acceptable carriers.
  • Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
  • the pharmaceutically acceptable compositions may be formulated, e.g., as micronized suspensions in isotonic, pH adjusted sterile saline or other aqueous solution, or, preferably, as solutions in isotonic, pH adjusted sterile saline or other aqueous solution, either with or without a preservative such as benzylalkonium chloride.
  • the pharmaceutically acceptable compositions may be formulated in an ointment such as petrolatum.
  • the pharmaceutically acceptable compositions of this invention may also be administered by nasal aerosol or inhalation.
  • compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other conventional solubilizing or dispersing agents.
  • compositions of this invention are formulated for oral administration.
  • Liquid dosage forms for oral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs.
  • the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsiflers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethyl formamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
  • inert diluents commonly used in the art such as, for example, water or other solvents
  • the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
  • adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
  • injectable preparations for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol.
  • the acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S.P. and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil can be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid are used in the preparation of injectables.
  • the injectable formulations can be sterilized, for example, by filtration through a bacterial-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.
  • the rate of compound release can be controlled.
  • biodegradable polymers include poly(orthoesters) and poly(anhydrides).
  • Depot injectable formulations are also prepared by entrapping the compound in liposomes or microemulsions that are compatible with body tissues.
  • compositions for rectal or vaginal administration are preferably suppositories which can be prepared by mixing the compounds of this invention with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound.
  • suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound.
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules.
  • the active compound is mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar—agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such as quaternary ammonium compounds, g) wetting agents such as, for example, cetyl alcohol and glycerol monostearate, h) absorbents such as kaolin and bentonite clay
  • Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like.
  • the solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polethylene glycols and the like.
  • the active compounds can also be in micro-encapsulated form with one or more excipients as noted above.
  • the solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings, release controlling coatings and other coatings well known in the pharmaceutical formulating art.
  • the active compound may be admixed with at least one inert diluent such as sucrose, lactose or starch.
  • Such dosage forms may also comprise, as is normal practice, additional substances other than inert diluents, e.g., tableting lubricants and other tableting aids such a magnesium stearate and microcrystalline cellulose.
  • the dosage forms may also comprise buffering agents. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner.
  • buffering agents include polymeric substances and waxes.
  • Dosage forms for topical or transdermal administration of a compound of this invention include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches.
  • the active component is admixed under sterile conditions with a pharmaceutically acceptable carrier and any needed preservatives or buffers as may be required.
  • Ophthalmic formulation, ear drops, and eye drops are also contemplated as being within the scope of this invention.
  • the present invention contemplates the use of transdermal patches, which have the added advantage of providing controlled delivery of a compound to the body.
  • Such dosage forms can be made by dissolving or dispensing the compound in the proper medium.
  • Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate can be controlled by either providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel.
  • the compounds of the invention are preferably formulated in dosage unit form for ease of administration and uniformity of dosage.
  • dosage unit form refers to a physically discrete unit of agent appropriate for the patient to be treated. It will be understood, however, that the total daily usage of the compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment.
  • the specific effective dose level for any particular patient or organism will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed, and like factors well known in the medical arts.
  • compositions should be formulated so that a dosage of between 0.01 - 100 mg/kg body weight/day of the inhibitor can be administered to a patient receiving these compositions.
  • additional therapeutic agents which are normally administered to treat or prevent that condition, may also be present in the compositions of this invention.
  • additional therapeutic agents that are normally administered to treat or prevent a particular disease, or condition are known as "appropriate for the disease, or condition, being treated”.
  • chemotherapeutic agents or other antiproliferative agents may be combined with the compounds of this invention to treat proliferative diseases and cancer.
  • Chemotherapeutic drugs include but are not limited to alkylating drugs (mechlorethamine, chlorambucil, cyclophosphamide, Melphalan, Ifosfamide), antimetabolites (Methotrexate), purine antagonists and pyrimidine antagonists (6-Mercapto.purine, 5-Fluorouracil, Cytarabile, Gemcitabine), spindle poisons (Vinblastine, Vincristine, Vinorelbine, Paclitaxel), podophyllotoxins (Etoposide, Irinotecan, Topotecan), antibiotics (Doxorubicin, Bleomycin, Mitomycin), nitrosoureas (Carmustine, Lomustine), inorganic ions (Cisplatin, Carboplatin), enzymes (Asparaginase), biologic response modifiers (inter
  • additional chemotherapeutic agents include GleevecTM, adriamycin, dexamethasone, vincristine, cyclophosphamide, fluorouracil, topotecan, taxol, interferons, and platinum derivatives.
  • agents for treating Multiple Sclerosis such as beta interferon (e.g., Avonex ® and Rebif ® ), Copaxone ® , and mitoxantrone
  • treatments for asthma such as albuterol and Singulair ®
  • anti-inflammatory agents such as NSAIDS 5 corticosteroids, TNF blockers, IL-I RA, azathioprine, cyclophosphamide, and sulfasalazine
  • immunomodulatory and immunosuppressive agents such as cyclosporin, tacrolimus, rapamycin, mycophenolate mofetil, interferons, corticosteroids, cyclophophamide, azathioprine, and sulfasalazine
  • agents for treating cardiovascular disease such as beta-blockers, ACE inhibitors, diuretics, nitrates, calcium channel blockers, and statins;
  • the amount of additional therapeutic agent present in the compositions of this invention will be no more than the amount that would normally be administered in a composition comprising that therapeutic agent as the only active agent.
  • the amount of additional therapeutic agent in the presently disclosed compositions will range from about 50% to 100% of the amount normally present in a composition comprising that agent as the only therapeutically active agent.
  • the invention provides a method of inhibiting
  • PI3K activity in a patient comprising administering to said patient a compound or composition of the invention.
  • the invention comprises a method of treating or lessening the severity of a PI3K-mediated condition or disease in a patient.
  • PI3K-mediated disease means any disease or other deleterious condition in which PI3K, in particular PI3K ⁇ , is known to play a role.
  • a PI3K disease is one in which PI3K ⁇ is known to play a role.
  • the invention comprises a method of treating a PI3K-mediated disease.
  • Such conditions include, without limitation, autoimmune diseases, inflammatory diseases, thrombolytic diseases, cancer, cardiovascular diseases, diabetes, allergic diseases, asthma and respiratory diseases.
  • the invention provides a method of treating or lessening the severity of a disease of condition selected from cancer, autoimmune diseases, inflammatory diseases, cardiovascular diseases, diabetes, and allergic diseases or asthma, comprising administering to said patient a compound or composition of the invention.
  • the invention provides a method of treating or lessening the severity of cancer.
  • cancers that may be treated or ameliorated by a method of the invention include, without limitation, cancer of the breast, ovary, cervix, prostate, testis, genitourinary tract, esophagus, larynx, glioblastoma, neuroblastoma, stomach, skin, keratoacanthoma, lung, epidermoid carcinoma, large cell carcinoma, small cell carcinoma, lung adenocarcinoma, bone, colon, adenoma, pancreas, adenocarcinoma, thyroid, follicular carcinoma, undifferentiated carcinoma, papillary carcinoma, seminoma, melanoma, sarcoma, bladder carcinoma, liver carcinoma and biliary passages, kidney carcinoma, myeloid disorders, lymphoid disorders, Hodgkin's, hairy cells, buccal cavity and pharynx (oral), lip, tongue,
  • the invention also provides a method of treating or lessening the severity of leukemias, including, without limitation, acute lymphocytic leukemia (ALL), chronic myelogenous leukemia (CML), multiple myeloma and lymphomas.
  • ALL acute lymphocytic leukemia
  • CML chronic myelogenous leukemia
  • the invention provides a method of treating or lessening the severity of cancer selected from ovarian cancer, colon cancer, colorectal cancer, breast cancer, brain cancer and lung cancer. [0185 ]
  • the invention provides a method of treating or lessening the severity of an autoimmune disease or disorder.
  • Autoimmune diseases or disorders include, without limitation, rheumatoid arthritis, systemic lupus erythematosus (SLE), multiple sclerosis, glomerulonephritis, scleroderma, chronic thyroiditis, Graves' disease, autoimmune gastritis, type I diabetes, autoimmune hemolytic anemia, autoimmune neutropenia, thrombocytopenia, atopic dermatitis, myasthenia gravis, inflammatory bowel disease, ulcerative colitis, Crohn's disease, psoriasis, Sjogren's syndrome and graft vs. host disease.
  • the autoimmune disease or disorder is rheumatoid arthritis, SLE or mutiple sclerosis.
  • the invention provides a method of treating or lessening the severity of organ transplantation rejection.
  • the invention provides a method of treating or lessening the severity of an inflammatory disease.
  • Inflammatory diseases include, without limitation, chronic obstructive pulmonary disease (COPD), bronchitis, emphysema, farmer's lung and related diseases, eosinophilia, lung fibrosis, osteoarthritis, ankylosing spondylitis, sepsis, septic shock, inflammatory myopathies, meningitis, encephalitis, lacrimal parotid gland syndrome, acute respiratory distress syndrome and pancreatitis.
  • the inflammatory disease is acute respiratory distress syndrome or lacrimal parotid gland syndrome.
  • the invention provides a method of treating or lessening the severity of a cardiovascular disease.
  • Cardiovascular diseases include, without limitation, atherosclerosis, pulmonary hypertension, deep venous thrombosis, stroke, myocardial infarction, myocardial contractility disorders, ischemia, thromboembolism, pulmonary embolism, acute arterial ischemia, peripheral thrombotic occlusions, coronary artery disease and acute coronary syndrome (ACS).
  • the cardiovascular disease is atherosclerosis, a myocardial contractility disorder or acute coronary syndrome.
  • the invention provides a method of treating or lessening the severity of type II diabetes.
  • the invention provides a method of treating or lessening the severity of allergic diseases or asthma.
  • allergic diseases include, without limitation, perennial and seasonal allergic rhinitis, type I hypersensitivity reactions, atopic dermatitis, contact dermatitis, eczema,
  • the invention provides a method of treating or lessening the severity of rheumatoid arthritis, SLE, multiple sclerosis, lacrimal parotid gland syndrome, COPD, pancreatitis, atherosclerosis, myocardial infarction, diabetes or cancer.
  • the disease or disorder is rheumatoid arthritis, SLE, atherosclerosis, multiple sclerosis or cancer.
  • the disease or disorder is rheumatoid arthritis, multiple sclerosis or SLE.
  • the method comprises the additional step of administering to said patient one or more additional therapeutic agents.
  • Additional therapeutic agents include chemotherapeutic agents or other anti-proliferative agents, such as GleevecTM, adriamycin, dexamethasone, vincristine, cyclophosphamide, fluorouracil, topotecan, taxol, interferons, and platinum derivatives; agents for treating multiple sclerosis (MS) such as beta interferon (e.g., Avonex ® and Rebif ® ), Copaxone ® , and mitoxantrone; treatments for asthma such as albuterol and Singulair ® ; antiinflammatory agents such as NSAIDS, corticosteroids, TNF blockers, IL-I RA, azathioprine, cyclophosphamide, and sulfasalazine; immunomodulatory and immunosuppressive agents such as cyclosporin, tacroli
  • said additional therapeutic agent is appropriate for the disease being treated and said additional therapeutic agent is administered together with said composition as a single dosage form or separately from said composition as part of a multiple dosage form.
  • these additional therapeutic agents may be administered to the patient prior to, sequentially with or following administration of the compositions of this invention.
  • the invention provides a method of inhibiting
  • PI3K activity in a biological sample comprising contacting said biological sample with a compound or composition of the invention.
  • biological sample means an ex vivo sample, and includes, without limitation, cell cultures or extracts thereof; tissue or organ samples or extracts thereof, biopsied material obtained from a mammal or extracts thereof; and blood, saliva, urine, feces, semen, tears, or other body fluids or extracts thereof.
  • Inhibition of PBK activity, particularly PI3K ⁇ activity, in a biological sample is useful for a variety of purposes that arc known to one of skill in the art. Examples of such purposes include, but are not limited to, biological specimen storage and biological assays.
  • the compounds of this invention or pharmaceutical compositions thereof may also be used for coating an implantable medical device, such as prostheses, artificial valves, vascular grafts, stents and catheters.
  • an implantable medical device such as prostheses, artificial valves, vascular grafts, stents and catheters.
  • vascular stents for example, have been used to overcome restenosis (re-narrowing of the vessel wall after injury).
  • patients using stents or other implantable devices risk clot formation or platelet activation.
  • Suitable coatings and the general preparation of coated implantable devices are described in US Patents 6,099,562; 5,886,026; and 5,304,121.
  • the coatings are typically biocompatible polymeric materials such as a hydrogel polymer, polymethyldisiloxane, polycaprolactone, polyethylene glycol, polylactic acid, ethylene vinyl acetate, and mixtures thereof.
  • the coatings may optionally be further covered by a suitable topcoat of fluorosilicone, polysaccarides, polyethylene glycol, phospholipids or combinations thereof to impart controlled release characteristics in the composition.
  • Implantable devices coated with a compound of this invention are another embodiment of the present invention.
  • the compounds may also be coated on implantable medical devices, such as beads, or co-formulated with a polymer or other molecule, to provide a "drug depot", thus permitting the drug to be released over a longer time period than administration of an aqueous solution of the drug.
  • the invention also provides a method of screening a compound for the ability to inhibit phosphatidylinositol 3-kinase (PI3K) activity.
  • the method comprises: a) mixing the compound with PI3K, adenosine triphospate (ATP) and a phosphatidylininositol (PI) in a buffer in which PI3K is active to form a mixture; b) incubating the mixture at a temperature and for a time sufficient for said PI3K to phosphorylate said PI; c) measuring the amount of phosphorylated Pl that is produced comprising: i) adding a stop buffer to the mixture in step b) to form a stopped mixture; ii) transferring the quenched mixture to a glass fiber filter (GFF) or hydrophobic membrane; iii) washing the quenched mixture with a wash buffer; and iv) measuring the phosphorylated PI on the GFF or hydrophobic membrane; and d) determining
  • Table 3 depicts exemplary ⁇ -NMR data (NMR) and liquid chromatographic mass spectral data, reported as mass plus proton (M+H), as determined by electrospray, and retention time (RT) for certain compounds of the present invention, wherein compound numbers in Table 3 correspond to the compounds depicted in Tables 1 and 2 (empty cells in the NMR column indicate that results were not available): Table 3
  • kinase buffer 50 mM HEPES (pH 7.4), 5 mM DTT, 5 mM MgCl 2 and 50 mM NaCl] containing 137 ⁇ M phosphatidylinositol(4,5) bisphosphate diC8 [PI(4,5)P 2 ; Echelon Biosciences, Cat. No. P-4508] and 158 nM PDK ⁇ .
  • 30 ⁇ l of kinase buffer containing 80 ⁇ M ATP was added to start the reaction.
  • Table 4 depicts enzyme inhibition data (Ki) for certain exemplary compounds.
  • Compound numbers in Table 4 correspond to those compounds depicted in Tables 1 and 2.
  • "A” represents a Ki of less than 0.1 ⁇ M
  • "B” represents a K; of between 0.1 and ⁇ 1.0 ⁇ M
  • "C” represents a K; of > 1.0 ⁇ M and less than 5.0 ⁇ M. If there was more than one result for a given compound, the average Kj was provided.
  • Compound 1 was assessed by Protocol A; all other compounds were assessed by Protocol B. Table 4

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Engineering & Computer Science (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Immunology (AREA)
  • Neurology (AREA)
  • Diabetes (AREA)
  • Biomedical Technology (AREA)
  • Neurosurgery (AREA)
  • Dermatology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Pulmonology (AREA)
  • Cardiology (AREA)
  • Hematology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Rheumatology (AREA)
  • Obesity (AREA)
  • Ophthalmology & Optometry (AREA)
  • Vascular Medicine (AREA)
  • Hospice & Palliative Care (AREA)
  • Urology & Nephrology (AREA)
  • Communicable Diseases (AREA)
  • Psychiatry (AREA)
  • Oncology (AREA)
  • Pain & Pain Management (AREA)
  • Endocrinology (AREA)
  • Emergency Medicine (AREA)
  • Transplantation (AREA)

Abstract

The present invention relates to compounds of formula (I) useful as inhibitors of P13K particularly of P13Kγ. The invention also provides pharmaceutically acceptable compositions comprising said compounds and methods of using the compositions in the treatment of various disease, conditions, or disorders.

Description

INHIBITORS OF PHOSPHATIDYLTNOSITOL 3-KINASE
TECHNICAL FIELD OF THE INVENTION
[ 0100] The present invention relates to compounds useful as inhibitors of phosphatidylinositol 3-kinase (PI3K). The invention also provides pharmaceutically acceptable compositions comprising the compounds of the invention and methods of using the compositions in the treatment of various disorders.
BACKGROUND OF THE INVENTION
[0101] P 13 Ks are a family of lipid kinases that catalyze the phosphorylation of the membrane lipid phosphatidylinositol (PI) on the 3'-OH of the inositol ring to produce PI 3-phosphate [PI(3)P, PIP], PI 3,4-bisphosphate [PI(3,4)P2, PIP2] and PI 3,4,5-trisphosphate [PI(3,4,5)P3, PIP3]. PI(3,4)P2 and PI(3,4,5)P3 act as recruitment sites for various intracellular signaling proteins, which in turn form signaling complexes to relay extracellular signals to the cytoplasmic face of the plasma membrane. [ 0102 ] Eight mammalian PBKs have been identified so far, including four class I PI3Ks. Class Ia includes PI3Kα, PI3Kβ and PI3Kδ. All of the class Ia enzymes are heterodimeric complexes comprising a catalytic subunit (pi 10a, pi lOβ or pi lOδ) associated with an SH2 domain-containing p85 adapter subunit. Class Ia PI3Ks are activated through tyrosine kinase signaling and are involved in cell proliferation and survival. PI3Kα and PI3Kβ have also been implicated in tumorigenesis in a variety of human cancers. Thus, pharmacological inhibitors of PI3Kα and PI3Kβ are useful for treating various types of cancer.
[ 0103 ] PI3Kγ, the only member of the Class Ib PI3Ks, consists of a catalytic subunit pi 1Oy, which is associated with a pi 01 regulatory subunit. PI3Kγ is regulated by G protein-coupled receptors (GPCRs) via association with βγ subunits of heterotrimeric G proteins. PI3Kγ is expressed primarily in hematopoietic cells and cardiomyocytes and is involved in inflammation and mast cell function. Thus, pharmacological inhibitors of PI3Kγ are useful for treating a variety of inflammatory diseases, allergies and cardiovascular diseases. [ 0104 ] Although a number of PBK inhibitors have been developed, there is a need for additional compounds to inhibit PI3Ks for treating various disorders and diseases. Accordingly, it would be desirable to develop additional compounds that are useful as inhibitors of PBK.
SUMMARY OF THE INVENTION
[0105] It has now been found that compounds of this invention, and pharmaceutically acceptable compositions thereof, are effective as inhibitors of PBK, particularly PBKγ. These compounds have the general formula I:
Figure imgf000003_0001
or a pharmaceutically acceptable salt thereof, wherein R1, R2, R3, R9, Y, T, L and n are as defined herein.
[0106] These compounds, and pharmaceutically acceptable compositions thereof, are useful for treating or lessening the severity of a variety of disorders, including cancer, autoimmune diseases, inflammatory diseases, cardiovascular diseases, diabetes, allergic diseases, asthma or organ transplantation rejection in a patient.
[0107 ] The compounds and compositions provided by this invention are also useful for the study of PBK in biological and pathological phenomena; the study of intracellular signal transduction pathways mediated by such kinases; and the comparative evaluation of new kinase inhibitors.
[ 0108 ] The invention also provides assays for identifying inhibitors of PBK.
DETAILED DESCRIPTION OF THE INVENTION Definitions and General Terminology
[ 0109 ] As used herein, the following definitions shall apply unless otherwise indicated. For purposes of this invention, the chemical elements are identified in accordance witlvthe Periodic Table of the Elements, CAS version, and the Handbook of Chemistry and Physics, 75th Ed. 1994. Additionally, general principles of organic chemistry are described in "Organic Chemistry", Thomas Sorrell, University Science Books, Sausalito: 1999, and "March's Advanced Organic Chemistry", 5th Ed., Smith, M.B. and March, J., eds. John Wiley & Sons, New York: 2001, the entire contents of which are hereby incorporated by reference.
[ 0110 ] As described herein, compounds of the invention may optionally be substituted with one or more substituents, such as are illustrated generally above, or as exemplified by particular classes, subclasses, and species of the invention. It will be appreciated that the phrase "optionally substituted" is used interchangeably with the phrase "substituted or unsubstituted." In general, the term "substituted", whether preceded by the term "optionally" or not, refers to the replacement of one or more hydrogen radicals in a given structure with the radical of a specified substituent. Unless otherwise indicated, an optionally substituted group may have a substituent at each substitutable position of the group. When more than one position in a given structure can be substituted with more than one substituent selected from a specified group, the substituent may be either the same or different at each position.
[ 0111 ] As described herein, when the term "optionally substituted" precedes a list, said term refers to all of the subsequent substitutable groups in that list. If a substituent radical or structure is not identified or defined as "optionally substituted", the substituent radical or structure is unsubstituted. For example, if X is halogen; optionally substituted Qoalkyl or phenyl; X may be either optionally substituted alkyl or optionally substituted phenyl. Likewise, if the term "optionally substituted" follows a list, said term also refers to all of the substitutable groups in the prior list unless otherwise indicated. For example: if X is halogen, C|-3alkyl or phenyl wherein X is optionally substituted by Jx, then both C|_3alkyl and phenyl may be optionally substituted by Jx. As is apparent to one having ordinary skill in the art, groups such as H, halogen, NO2, CN, NH2, OH, or OCF3 would not be included because they are not substitutable groups. [0112 ] Combinations of substituents envisioned by this invention are preferably those that result in the formation of stable or chemically feasible compounds. The term "stable", as used herein, refers to compounds that are not substantially altered when subjected to conditions to allow for their production, detection, and, preferably, their recovery, purification, and use for one or more of the purposes disclosed herein. In some embodiments, a stable compound or chemically feasible compound is one that is not substantially altered when kept at a temperature of 400C or less, in the absence of moisture or other chemically reactive conditions, for at least a week. [ 0113 ] The term "aliphatic" or "aliphatic group", as used herein, means a straight-chain (i.e., unbranched) or branched, substituted or unsubstituted hydrocarbon chain that is completely saturated or that contains one or more units of unsaturation. Unless otherwise specified, aliphatic groups contain 1-20 aliphatic carbon atoms. In some embodiments, aliphatic groups contain 1-10 aliphatic carbon atoms. In other embodiments, aliphatic groups contain 1-8 aliphatic carbon atoms. In still other embodiments, aliphatic groups contain 1-6 aliphatic carbon atoms, and In yet other embodiments, aliphatic groups contain 1-4 aliphatic carbon atoms. Suitable aliphatic groups include, but are not limited to, linear or branched, substituted or unsubstituted alkyl, alkenyl, or alkynyl groups. Further examples of aliphatic groups include methyl, ethyl, propyl, butyl, isopropyl, isobutyl, vinyl, and sec-butyl.
[ 0114 ] The term "cycloaliphatic" (or "carbocycle" or "carbocyclyl") refers to a hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic, that has a single point of attachment to the rest of the molecule, and wherein any individual ring in said bicyclic ring system has 3-7 members. Unless otherwise specified, the term "cycloaliphatic" refers to a monocyclic C3-Cg hydrocarbon or bicyclic C8-Ci2 hydrocarbon. Suitable cycloaliphatic groups include, but are not limited to, cycloalkyl, cycloalkenyl, and cycloalkynyl. Further examples of aliphatic groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cycloheptyl, and cycloheptenyl. [0115] The term "heterocycle", "heterocyclyl" or "heterocyclic" as used herein refers to a monocyclic, bicyclic, or tricyclic ring system in which one or more ring members are an independently selected heteroatom and that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic, that has a single point of attachment to the rest of the molecule. In some embodiments, the "heterocycle", "heterocyclyl" or "heterocyclic" group has three to fourteen ring members in which one or more ring members is a heteroatom independently selected from oxygen, sulfur, nitrogen, or phosphorus, and each ring in the system contains 3 to 7 ring members. [ 0116 ] Examples of heterocyclic rings include, but are not limited to, the following monocycles: 2-tetrahydrofuranyl, 3-tetrahydrofuranyl, 2-tetrahydrothiophenyl, 3-tetrahydrothiophenyl, 2-morpholino, 3-moφholino, 4-morpholino, 2-thiomorpholino, 3-thiomorpholino, 4-thiomorpholino, 1-pyrrolidinyl, 2-pyrrolidinyl, 3-pyrrolidinyl, 1-tetrahydropiperazinyl, 2-tetrahydropiperazinyl, 3-tetrahydropiperazinyl, 1 -piperidinyl, 2-piperidinyl, 3-piperidinyl, 1-pyrazolinyl, 3-pyrazolinyl, 4-pyrazolinyl, 5-pyrazolinyl, 1 -piperidinyl, 2-piperidinyl, 3-piperidinyl, 4-piperidinyl, 2-thiazolidinyl, 3-thiazolidinyl, 4-thiazolidinyl, 1 -imidazolidinyl, 2-imidazolidinyl, 4-imidazolidinyl, 5-imidazolidinyl; and the following bicycles: 3-lH-benzimidazol-2-one, 3-(l-alkyl)-benzimidazol-2-one, indolinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, benzothiolane, benzodithiane, and 1 ,3-dihydro-imidazol-2-one.
[0117 ] The term "heteroatom" means one or more of oxygen, sulfur, nitrogen, phosphorus, or silicon, including any oxidized form of nitrogen, sulfur, phosphorus, or silicon, the quaternized form of any basic nitrogen, or a substitutable nitrogen of a heterocyclic ring, for example N (as in 3,4-dihydro-2H-pyrrolyl), NH (as in pyrrolidinyl) or NR+ (as in N-substituted pyrrolidinyl).
[ 0118 ] The term "unsaturated", as used herein, means that a moiety has one or more units of unsaturation.
[0119] The term "aryl" used alone or as part of a larger moiety as in
"aralkyl", "aralkoxy", or "aryloxyalkyl", refers to monocyclic, bicyclic, and tricyclic carbocyclic ring systems having a total of six to fourteen ring members, wherein at least one ring in the system is aromatic, wherein each ring in the system contains 3 to 7 ring members and that has a single point of attachment to the rest of the molecule. The term "aryl" may be used interchangeably with the term "aryl ring". Examples of aryl rings would include phenyl, naphthyl, and anthracene.
[0120] The term "heteroaryl", used alone or as part of a larger moiety as in
"heteroaralkyl" or "heteroarylalkoxy", refers to monocyclic, bicyclic, and tricyclic ring systems having a total of five to fourteen ring members, wherein at least one ring in the system is aromatic, at least one ring in the system contains one or more heteroatoms, wherein each ring in the system contains 3 to 7 ring members and that has a single point of attachment to the rest of the molecule. The term "heteroaryl" may be used interchangeably with the term "heteroaryl ring" or the term "heteroaromatic". [ 0121] Further examples of heteroaryl rings include the following monocycles: 2-furanyl, 3-furanyl, N-imidazolyl, 2-imidazolyl, 4-imidazolyl, 5- imidazolyl, 3-isoxazolyl, 4-isoxazolyl, 5-isoxazolyl, 2-oxazolyl, 4-oxazolyl, 5-oxazolyl, N-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl, pyridazinyl (e.g., 3-pyridazinyl), 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, tetrazolyl (e.g., 5-tetrazolyl), triazolyl (e.g., 2-triazolyl and 5-triazolyl), 2- thienyl, 3-thienyl, pyrazolyl (e.g., 2-pyrazolyl), isothiazolyl, 1 ,2,3-oxadiazolyl, 1,2,5- oxadiazolyl, 1,2,4-oxadiazolyl, 1,2,3 -triazolyl, 1.2,3-thiadiazolyl. 1,3,4-thiadiazolyl, 1,2,5-thiadiazolyl, pyrazinyl, 1,3,5-triazinyl, and the following bicycles: benzimidazolyl, benzofuryl, benzothiophenyl, indolyl (e.g., 2-indolyl), purinyl, quinolinyl (e.g., 2- quinolinyl, 3-quinolinyl, 4-quinolinyl), and isoquinolinyl (e.g., 1-isoquinolinyl, 3- isoquinolinyl, or 4-isoquinolinyl).
[ 0122] In some embodiments, an aryl (including aralkyl, aralkoxy, aryloxyalkyl and the like) or heteroaryl (including heteroaralkyl and heteroarylalkoxy and the like) group may contain one or more substituents. Suitable substituents on the unsaturated carbon atom of an aryl or heteroaryl group are selected from those listed in the definitions of R2 and R4 below. Other suitable substituents include: halogen; -R°; -OR°; -SR°; 1,2-methylenedioxy; 1 ,2-ethylenedioxy; phenyl (Ph) optionally substituted with R°; -O(Ph) optionally substituted with R°; -(CH2) I-2(Ph), optionally substituted with R0; -CH=CH(Ph), optionally substituted with R°; -NO2; -CN; -N(R°)2; -NR0C(O)R0; -NR0C(S)R0; -NR°C(O)N(R°)2; -NR°C(S)N(R°)2; -NR0CO2R0; -NR0NR0C(O)R0; -NRONR°C(O)N(RO)2; -NR0NR0CO2R0; -C(O)C(O)R0; -C(O)CH2C(O)R0; -CO2R0; -C(O)R0; -C(S)R0; -C(O)N(R°)2; -C(S)N(R°)2; -OC(O)N(R°)2; -OC(O)R0; -C(O)N(OR0) R°; -C(NOR0) R°; -S(O)2R0; -S(O)3R0; -SO2N(R°)2; -S(O)R0; -NR°SO2N(R°)2; -NR0SO2R0; -N(OR°)R°; -C(=NH)-N(R°)2; or -(CH2)0-2NHC(O)Ro; wherein each independent occurrence of R° is selected from hydrogen, optionally substituted Ci-6 aliphatic, an unsubstituted 5-6 membered heteroaryl or heterocyclic ring, phenyl, -O(Ph), or -CH2(Ph), or, two independent occurrences of R°, on the same substituent or different substituents, taken together with the atom(s) to which each R° group is bound, form a 5- 8-membered heterocyclyl, aryl, or heteroaryl ring or a 3-8-membered cycloalkyl ring, wherein said heteroaryl or heterocyclyl ring has 1-3 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Optional substituents on the aliphatic group of R° are selected from NH2, NH(Ci-4aliphatic), N(Ci.4aliphatic)2, halogen, Ci ^aliphatic, OH, O(Ci ^aliphatic), NO2, CN, CO2H, CO2(C ^aliphatic), 00IaIoCi-4 aliphatic), or haloCi- 4aliphatic, wherein each of the foregoing Ci^aliphatic groups of R° is unsubstituted. [ 0123 ] In some embodiments, an aliphatic or heteroaliphatic group, or a non- aromatic heterocyclic ring may contain one or more substituents. Suitable substituents on the saturated carbon of an aliphatic or heteroaliphatic group, or of a non-aromatic heterocyclic ring are selected from those listed above for the unsaturated carbon of an aryl or heteroaryl group and additionally include the following: =O, =S, =NNHR , =NN(R*)2, =NNHC(O)R*, =NNHCO2(alkyl), =NNHSO2(alkyl), or =NR*, where each R* is independently selected from hydrogen or an optionally substituted Ci .6 aliphatic. Optional substituents on the aliphatic group of R are selected from NH2, NH(Ci-4 aliphatic), N(Ci-4 aliphatic)2, halogen, Ci-4 aliphatic, OH, 0(Ci-4 aliphatic), NO2, CN, CO2H, CO2(Ci-4 aliphatic), O(halo C1.4 aliphatic), or halo(Ci-4 aliphatic), wherein each of the foregoing Cj ^aliphatic groups of R is unsubstituted.
[ 0124 ] In some embodiments, optional substituents on the nitrogen of a non- aromatic heterocyclic ring include -R+, -N(R+)2, -C(O)R+, -CO2R+, -C(O)C(O)R+, - C(O)CH2C(O)R+, -SO2R+, -SO2N(R+)2, -C(=S)N(R+)2, -C(=NH)-N(R+)2, or -NR+SO2R+; wherein R+ is hydrogen, an optionally substituted C\.& aliphatic, optionally substituted phenyl, optionally substituted -O(Ph), optionally substituted -CH2(Ph), optionally substituted -(CH2) I-2(Ph); optionally substituted -CH=CH(Ph); or an unsubstituted 5-6 membered heteroaryl or heterocyclic ring having one to four heteroatoms independently selected from oxygen, nitrogen, or sulfur, or,. two independent occurrences of R+, on the same substituent or different substituents, taken together with the atom(s) to which each R+ group is bound, form a 5-8-membered heterocyclyl, aryl, or heteroaryl ring or a 3-8- membered cycloalkyl ring, wherein said heteroaryl or heterocyclyl ring has 1 -3 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Optional substituents on the aliphatic group or the phenyl ring of R+ are selected from NH2, NH(Ci-4 aliphatic), N(Ci-4 aliphatic)2, halogen, Ci-4 aliphatic, OH, 0(Ci-4 aliphatic), NO2, CN, CO2H, CO2(Ci-4 aliphatic), O(halo C1-4 aliphatic), orhalo(Ct-4 aliphatic), wherein each of the foregoing Ci-4aliphatic groups OfR+ is unsubstituted. [0125] As detailed above, in some embodiments, two independent occurrences of R0 (or R+, or any other variable similarly defined herein), may be taken together with the atom(s) to which each variable is bound to form a 5-8-membered heterocyclyl, aryl, or heteroaryl ring or a 3-8-membered cycloalkyl ring. Exemplary rings that are formed when two independent occurrences of R0 (or R+, or any other variable similarly defined herein) are taken together with the atom(s) to which each variable is bound include, but are not limited to the following: a) two independent occurrences of R° (or R+, or any other variable similarly defined herein) that are bound to the same atom and are taken together with that atom to form a ring, for example, N(R°)2, where both occurrences of R° are taken together with the nitrogen atom to form a piperidin-1-yl, piperazin-1-yl, or morpholin-4-yl group; and b) two independent occurrences of R° (or R+, or any other variable similarly defined herein) that are bound to different atoms and are taken together with both of those atoms to form a ring, for example where a phenyl group is substituted with two occurrences of OR0
Figure imgf000009_0001
these two occurrences of R° are taken together with the oxygen atoms to which they are bound to form a fused 6-membered oxygen containing ring:
Figure imgf000009_0002
It will be appreciated that a variety of other rings can be formed when two independent occurrences of R0 (or R+, or any other variable similarly defined herein) are taken together with the atom(s) to which each variable is bound and that the examples detailed above are not intended to be limiting.
[0126] In some embodiments, an alkyl or aliphatic chain can be optionally interrupted with another atom or group. This means that a methylene unit of the alkyl or aliphatic chain is optionally replaced with said other atom or group. Examples of such atoms or groups would include, but are not limited to, -NR-, -O-, -S-, -CQ2-, -OC(O)-, -C(O)CO-, -C(O)-, -C(O)NR-, -C(=N-CN), -NRCO-, -NRC(O)O-, -SO2NR-, -NRSO2-, -NRC(O)NR-, -OC(O)NR-, -NRSO2NR-, -SO-, or -SO2-, wherein R is defined herein. Unless otherwise specified, the optional replacements form a chemically stable compound. Optional interruptions can occur both within the chain and at either end of the chain; i.e. both at the point of attachment and/or also at the terminal end. Two optional replacements can also be adjacent to each other within a chain so long as it results in a chemically stable compound. Unless otherwise specified, if the replacement or interruption occurs at the terminal end, the replacement atom is bound to an H on the terminal end. For example, if -CH2CH2CH3 were optionally interrupted with -O-, the resulting compound could be -OCH2CH3, -CH2OCH3, or -CH2CH2OH. [ 0127 ] As described herein, a bond drawn from a substituent to the center of one ring within a multiple-ring system (as shown below), represents substitution of the substituent at any substitutable position in any of the rings within the multiple ring system. For example, Figure a represents possible substitution in any of the positions shown in Figure b.
Figure imgf000010_0001
Figure a
Figure imgf000010_0002
[0128 ] This also applies to multiple ring systems fused to optional ring systems (which would be represented by dotted lines). For example, in Figure c, X is an optional substituent both for ring A and ring B.
ft \^ AJ'> B .÷' X
Figure c
[0129] If, however, two rings in a multiple ring system each have different substituents drawn from the center of each ring, then, unless otherwise specified, each substituent only represents substitution on the ring to which it is attached. For example, in Figure d, Y is an optionally substituent for ring A only, and X is an optional substituent for ring B only.
Figure imgf000010_0003
Figure d
[0130] Unless otherwise stated, structures depicted herein are also meant to include all isomeric (e.g., enantiomeric, diastereomeric, and geometric (or conformational)) forms of the structure; for example, the R and S configurations for each asymmetric center, (Z) and (E) double bond isomers, and (Z) and (E) conformational isomers. Therefore, single stereochemical isomers as well as enantiomeric, diastereomeric, and geometric (or conformational) mixtures of the present compounds are within the scope of the invention.
[0131] Unless otherwise stated, all tautomeric forms of the compounds of the invention are within the scope of the invention. Additionally, unless otherwise stated, structures depicted herein are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms. For example, compounds having the present structures except for the replacement of hydrogen by deuterium or tritium, or the replacement of a carbon by a 13C- or l4C-enriched carbon are within the scope of this invention. Such compounds are useful, for example, as analytical tools or probes in biological assays. Description of Compounds of the Invention
10132 ] The present invention relates to a compound of formula I:
Figure imgf000011_0001
I or a pharmaceutically acceptable salt thereof, wherein:
Y is -C(O)- or a bond;
R1 is a C|-6 aliphatic, a C3-]O monocyclic or bicyclic cycloaliphatic, a C6-Io monocyclic or bicyclic aryl, a 5-10 membered monocyclic or bicyclic heteroaryl, or a 5-10 membered monocyclic or bicyclic heterocyclyl, wherein R1 is optionally substituted with 1-6 occurrences of JRI; each JR1 is independently selected from halogen, OH, OR, NO2, NH2, NHR, NR2, SH, SR, CN, C(O)NH2, C(O)NHR, C(O)NR2, C(O)OH, C(O)OR, NHC(O)H, NHC(O)R, NRC(O)H, NRC(O)R, NHC(O)OH, NHC(O)OR, NRC(O)OH, NRC(O)OR, or R; or two JR1, together with the carbon(s) to which they are attached, form a cyclopropyl ring or C=O; each R is independently selected from a C1-4 aliphatic or C3-4 cycloaliphatic optionally substituted with 1-4 occurrences of halogen, OH, NO2, NH2, SH or CN;
R2 is H or a C)-6 aliphatic or C3-6 cycloaliphatic, wherein R2 is optionally substituted with 1-4 occurrences of J1*2; each J*2 is independently selected from halogen, OH, OR, NO2, NH2, NHR, NR2, SH, SR, CN, C(O)NH2, C(O)NHR, C(O)NR2, C(O)OH, C(O)OR, NHC(O)H, NHC(O)R, NRC(O)H, NRC(O)R, NHC(O)OH, NHC(O)OR, NRC(O)OH, NRC(O)OR, or R; or two J112, together with the carbon(s) to which they are attached, form a cyclopropyl ring or C=O;
T is -SO2NR4-, -CONR4- or -C(O)O-;
R is H or a Cj-6 aliphatic or C3-7 cycloaliphatic optionally substituted with 1-6 occurrences of JR4; each JR4 is independently selected from halogen, OH, OR', NO2, NH2, NHR', NR' 2, SH, SR', CN, 5-6 membered aryl or heteroaryl, or R'; or two JR4, together with the carbon(s) to which they are attached, form a cyclopropyl ring or C=O; each R' is independently selected from a Ci-6 aliphatic or C3-7 cycloaliphatic optionally substituted with 1-6 occurrences of halogen, OH, NO2, NH2, SH or CN;
L is a Ci-6 aliphatic wherein up to three methylene units are replaced by -NH-, -NR5-, -O- , -S-, -CO2-, -OC(O)-, -C(O)CO-, -C(O)-, -C(O)NH-, -C(O)NR5-, -NC(=N- CN)N, -NHCO-, -NR5CO-, -NHC(O)O-, -NR5C(O)O-, -SO2NH-, -SO2NR5-, -NHSO2-, -NR5SO2-, -NHC(O)NH-, -NR5C(O)NH-, -NHC(O)NR5-, -NR5C(O)NR5, -OC(O)NH-, -OC(O)NR5-, -NHSO2NH-, -NR5SO2NH-, -NHSO2NR5-, -NR5SO2NR5-, -SO-, or -SO2-, wherein L is optionally substituted with 1-4 occurrences of R6; each R5 is independently selected from C]-6 aliphatic, C3.ιo monocyclic or bicyclic cycloaliphatic, Cβ-io monocyclic or bicyclic aryl, 5-10 membered monocyclic or bicyclic heteroaryl, or 5-10 membered monocyclic or bicyclic heterocyclyl; or two R5 groups, on the same substituent or different substituents, together with the atom(s) to which each R5 group is bound, form a 3-8 membered heterocyclyl; each R6 is independently selected from halogen, OH, OR', NO2, NH2, NHR', NR'2, SH, SR', CN, or R'; or two R6, together with the carbon(s) to which they are attached, form a cyclopropyl ring or C=O; n is 0 or 1 ;
R3 is H or an optionally substituted group selected from a Cue aliphatic, a C3-Io monocyclic orbicyclic cycloaliphatic, a C6-io monocyclic orbicyclic aryl, a 5-10 membered monocyclic or bicyclic heteroaryl, or a 5-10 membered monocyclic or bicyclic heterocyclyl, wherein R3 is optionally substituted with 1-6 occurrences of J**3, or R3 and R4, together with the nitrogen to which R4 is attached, form a 5- 10 membered monocyclic or bicyclic heterocyclyl or heteroaryl; wherein n is 0 and said heterocyclyl or heteroaryl is optionally substituted with 1-6 occurrences ofJ113; each J*3 is independently selected from -(U)m-X;
U is a Ci-4 aliphatic, wherein up to two methylene units are optionally and independently replaced by Gu and wherein U is optionally substituted with 1-4 Ju;
Gu is -NH-, -NR7-, -O-, -S-, -CO2-, -OC(O)-, -C(O)CO-, -C(O)-, -C(O)NH-, -C(O)NR7-, -NC(=N-CN)N-, -NHCO-, -NR7CO-, -NHC(O)O-, -NR7C(O)O-, -SO2NH-, - SO2NR7-, -NHSO2-, -NR7SO2-, -NHC(O)NH-, -NR7C(O)NH-, -NHC(O)NR7-, -NR7C(O)NR7, -OC(O)NH-, -OC(O)NR7-, -NHSO2NH-, -NR7SO2NH-, -NHSO2NR7-, -NR7SO2NR7-, -SO-, or -SO2-;
R7 is C1-^ aliphatic or C3-7 cycloaliphatic optionally substituted with 1-6 occurrences of halogen, OH, NO2, NH2, SH or CN; m is O or 1 ; each Ju is independently selected from halogen, OH, OR', NO2, NH2, NHR', NR'2, SH, SR', CN, or R'; or two Ju, together with the carbon(s) to which they are attached, form a cyclopropyl ring or C=O;
X is H, halogen, OH, OR, NO2, NH2, NHR, NR2, SH, SR, CN, C(O)NH2, C(O)NHR, C(O)NR2, C(O)OH, C(O)OR, NHC(O)H, NHC(O)R, NRC(O)H, NRC(O)R, NHC(O)OH, NHC(O)OR, NRC(O)OH, NRC(O)OR, or a group selected from a Cue aliphatic, a C3.io monocyclic or bicyclic cycloaliphatic, a C6-io monocyclic or bicyclic aryl, a 5-10 membered monocyclic or bicyclic heteroaryl, or a 5-10 membered monocyclic or bicyclic heterocyclyl, wherein said group is optionally substituted with 1-4 Jx, or two X, together with the carbon(s) to which they are attached, form a cyclopropyl ring or C=O; wherein X is not H when m is O; each Jx is independently selected from halogen, OH, OR, NO2, NH2, NHR, NR2, SH, SR, CN, C(O)NH2, C(O)NHR, C(O)NR2, C(O)OH, C(O)OR, NHC(O)H, NHC(O)R, NRC(O)H, NRC(O)R, NHC(O)OH, NHC(O)OR, NRC(O)OH, NRC(O)OR; and
R9 is selected from H; halogen; OH; NO2; NH2; SH; CN; or a group selected from a Cj-6 aliphatic or a C3-7 cycloaliphatic, wherein said group is optionally substituted with 1-6 occurrences of halogen, OH, NO2, NH2, SH or CN.
[0133 ] In one embodiment, Y is C(O) and the compound is of formula II:
Figure imgf000014_0001
II.
[0134] In one embodiment of either of the above formulae, R1 is a C1-6 aliphatic optionally substituted with 1-6 occurrences of J . In a further embodiment, R1 is a Ci-3 aliphatic optionally substituted with 1-3 occurrences of J . In yet a further embodiment, R1 is a Ci-3 aliphatic. In still a further embodiment, R1 is CH3, CH2CH3 or cyclopropyl.
[0135] In one embodiment of either of the above formulae or compounds, R2 is Ci-3 aliphatic, wherein R2 is optionally substituted with 1-3 occurrences of J1*2. hi a further embodiment, R2 is Ci-3 aliphatic. In yet a further embodiment, R2 is CH3. [0136] In one embodiment of any of the above formulae or compounds, said compound is of formula HIa, IHb or HIc:
Figure imgf000014_0002
[ 0137 ] In one embodiment of any of the above formulae or compounds, said compound is of formula IHa', HIb' or IHc':
Figure imgf000015_0001
HIa' HIb' IHc'.
[ 0138 ] " In one embodiment of the above formulae or compounds, R and R , together with the nitrogen to which R4 is attached, form a 5-10 membered monocyclic or bicyclic heterocyclyl or heteroaryl; wherein said heterocyclyl or heteroaryl is optionally substituted with 1-6 occurrences of J1*3. In a further embodiment, said heterocyclyl or heteroaryl formed by R3 and R4 is a 5-6 membered monocyclic heterocyclyl or 9-10 membered heterocyclyl or heteroaryl; wherein said monocyclic or bicyclic heterocyclyl or bicyclic heteroaryl contains 1-3 heteroatoms and is optionally substituted with 1-3 occurrences of JR3. In yet a further embodiment, said monocyclic or bicyclic heterocyclyl or bicyclic heteroaryl contains 1-2 heteroatoms and is optionally substituted with 1-2 occurrences of JR3. In a still further embodient, said monocyclic or bicyclic heterocyclyl or bicyclic heteroaryl is selected from the following:
Figure imgf000015_0002
wherein Z is CH2, NH, O or S, and wherein the hydrogens of said CH2 of Z is optionally substituted by 1-2 occurrences of JR3 and the hydrogen of said NH of Z is optionally substituted by JR3. [ 0139 ] In a further embodiment, each occurrence of JR3 is selected from J113 is selected from halogen; OH; OR; CN; NH2; NHR; NR2; C(O)NH2; C(O)NHR; C(O)NR2; NHCOR; C(O)OR; C(O)OH; oxo; (CH2)0-3phenyl optionally substituted with halogen, OH, OR, NO2, NH2, SH or CN; or Ci-4 aliphatic optionally substituted with halogen, OH, OR, NO2, NH2, SH or CN.
[ 0140 ] In another embodiment of the above formulae or compounds, R is H or an optionally substituted group selected from a C\s aliphatic, a C3-io monocyclic or bicyclic cycloaliphatic, a C6- 10 monocyclic or bicyclic aryl, a 5-10 membered monocyclic or bicyclic heteroaryl, or a 5-10 membered monocyclic or bicyclic heterocyclyl, wherein R3 is optionally substituted with 1-6 occurrences of Jm. In a further embodiment, n is 1 and L is a Cu aliphatic optionally substituted with 1-2 occurrences of R'. In yet a further embodiment, L is -CH2- or CH substituted with a Ci-2 aliphatic, which Cj-2 aliphatic is optionally substituted with 1-2 occurrences of halogen, OH, NO2, NH2, SH or CN. In a still further embodiment, L is -CH2-. In another embodiment, n is 0. [0141] In embodiment, R3 is a Ci-4 aliphatic, a C3-6 monocyclic cycloaliphatic, phenyl, a 5-6 membered monocyclic heteroaryl, or a 5-10 membered monocyclic or bicyclic heterocyclyl, wherein R3 is optionally substituted with 1-4 occurrences of JR3. In a further embodiment, R3 is a Ci-4 aliphatic, a C3-6 monocyclic cycloaliphatic, phenyl, a 5-6 membered monocyclic heteroaryl, or a 5-6 membered monocyclic heterocyclyl, wherein R3 is optionally substituted with 1-4 occurrences of J113. [ 0142 ] In a further embodiment, each occurrence of JR3 is selected from halogen; OH; OR; CN; NH2; NHR; NR2; C(O)NH2; C(O)NHR; C(O)NR2; NHCOR; C(O)OR; C(O)OH; oxo; (CH2)o-3ρhenyl optionally substituted with halogen, OH, OR, NO2, NH2, SH or CN; or Ci-4 aliphatic optionally substituted with halogen, OH, OR, NO2, NH2, SH or CN.
[ 01433 In one embodiment of any of the above formulae or compounds, R4 is
H or Ci-3 aliphatic optionally substituted with one occurrence of JR . In a further embodiment, R4 is H, CH3, CH2CH3, CH2CH2OH, CH2CHCH2, cyclopropyl or CH2phenyl. In yet a further embodiment, R4 is H or CH3.
[0144 ] In one embodiment of any of the above formulae or compounds, R8 is selected from H, cyclopropyl, or a Ci-3 aliphatic optionally substituted with 1-3 occurrences of halogen or OH. In a further embodiment, R8 is selected from H, halogen, methyl or ethyl. In yet a further embodiment, R8 is H.
[0145] In one embodiment of any of the above formulae or compounds, R9 is selected from H, OH, halogen, cyclopropyl, or a C1-3 aliphatic optionally substituted with 1-3 occurrences of halogen or OH. In a further embodiment, R9 is selected from H, OH, halogen, methyl or ethyl. In yet a further embodiment, R9 is H or methyl. [0146] In another embodiment, the invention provides a compound of Table I or Table 2: Table 1
Figure imgf000017_0001
Figure imgf000018_0001
Figure imgf000019_0001
Figure imgf000020_0001
Table 2
Figure imgf000021_0001
Figure imgf000022_0001
Figure imgf000023_0001
Uses, Formulation and Administration Pharmaceutically acceptable compositions
[ 0147 ] In another embodiment, the invention provides a pharmaceutical composition comprising a compound of any of the formulae or classes described herein. In a further embodiment, the invention provides a pharmaceutical composition comprising a compound of Table 1. In a further embodiment, the composition additionally comprises an additional therapeutic agent.
[0148] According to another embodiment, the invention provides a composition comprising a compound of this invention or a pharmaceutically acceptable derivative thereof and a pharmaceutically acceptable carrier, adjuvant, or vehicle. In one embodiment, the amount of compound in a composition of this invention is such that is effective to measurably inhibit a PBK, particularly PI3Kγ, in a biological sample or in a patient. In another embodiment, the amount of compound in the compositions of this invention is such that is effective to measurably inhibit PI3Kα. Preferably the composition of this invention is formulated for administration to a patient in need of such composition. Most preferably, the composition of this invention is formulated for oral administration to a patient.
[ 0149 ] The term "patient", as used herein, means an animal, preferably a mammal, and most preferably a human.
[0150] Accordingly, in another aspect of the present invention, pharmaceutically acceptable compositions are provided, wherein these compositions comprise any of the compounds or pharmaceutically acceptable salts thereof, as described herein, and further comprises a pharmaceutically acceptable carrier, adjuvant or vehicle. In certain embodiments, these compositions optionally further comprise one or more additional therapeutic agents. [ 0151] It will also be appreciated that certain of the compounds of present invention can exist in free form for treatment, or where appropriate, as a pharmaceutically acceptable derivative thereof. According to the present invention, a pharmaceutically acceptable derivative includes, but is not limited to, pharmaceutically acceptable prodrugs, salts, esters, salts of such esters, or any other adduct or derivative which upon administration to a patient in need is capable of providing, directly or indirectly, a compound as otherwise described herein, or a metabolite or residue thereof. As used herein, the term "inhibitorily active metabolite or residue thereof means that a metabolite or residue thereof is also an inhibitor of PBK.
[ 0152 ] As used herein, the term "pharmaceutically acceptable salt" refers to those salts which are, within the scope of sound medical judgement, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like.
[ 0153 ] Pharmaceutically acceptable salts are well known in the art. For example, S. M. Berge et al., describe pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 1977, 66, 1-19, incorporated herein by reference. Pharmaceutically acceptable salts of the compounds of this invention include those derived from suitable inorganic and organic acids and bases. Examples of pharmaceutically acceptable, nontoxic acid addition salts are salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange. Other pharmaceutically acceptable salts include adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionatc, digluconate, dodecyl sulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2- hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, olcate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, p-toluenesulfonate, undecanoate, valerate salts, and the like. Salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium and N+(Ci.4alkyl)4 salts. This invention also envisions the quaternization of any basic nitrogen-containing groups of the compounds disclosed herein. Water or oil-soluble or dispersable products may be obtained by such quaternization. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, loweralkyl sulfonate and aryl sulfonate.
[0154 ] As described above, the pharmaceutically acceptable compositions of the present invention additionally comprise a pharmaceutically acceptable carrier, adjuvant, or vehicle, which, as used herein, includes any and all solvents, diluents, or other liquid vehicle, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, solid binders, lubricants and the like, as suited to the particular dosage form desired. Remington's Pharmaceutical Sciences, Sixteenth Edition, E. W. Martin (Mack Publishing Co., Easton, Pa., 1980) discloses various carriers used in formulating pharmaceutically acceptable compositions and known techniques for the preparation thereof. Except insofar as any conventional carrier medium is incompatible with the compounds of the invention, such as by producing any undesirable biological effect or otherwise interacting in a deleterious manner with any other component(s) of the pharmaceutically acceptable composition, its use is contemplated to be within the scope of this invention.
[ 0155 ] Some examples of materials which can serve as pharmaceutically acceptable carriers include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, or potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, polyacrylates, waxes, polyethylene-polyoxypropylene-block polymers, wool fat, sugars such as lactose, glucose and sucrose; starches such as corn starch and potato starch; cellulose and its derivatives such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients such' as cocoa butter and suppository waxes; oils such as peanut oil, cottonseed oil; safflower oil; sesame oil; olive oil; corn oil and soybean oil; glycols; such a propylene glycol or polyethylene glycol; esters such as ethyl oleate and ethyl laurate; agar; buffering agents such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water-; isotonic saline; Ringer's solution; ethyl alcohol, and phosphate buffer solutions, as well as other nontoxic compatible lubricants such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, releasing agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the composition, according to the judgment of the formulator.
[ 0156 ] The term "measurably inhibit", as used herein means a measurable change in kinase activity, particularly POK activity, between a sample comprising a compound of this invention and a PI3K and an equivalent sample comprising PI3K in the absence of said compound.
[0157 ] The compositions of the present invention may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir. The term "parenteral" as used herein includes subcutaneous, intravenous, intramuscular, intra-articular, intra-synovial, intrasternal, intrathecal, intraocular, intrahepatic, intralesional and intracranial injection or infusion techniques. Preferably, the compositions are administered orally, intraperitoneally or intravenously. Sterile injectable forms of the compositions of this invention may be aqueous or oleaginous suspension. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally-acceptable diluent or solvent, for example as a solution in 1,3- butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. [ 0158 ] For this purpose, any bland fixed oil may be employed including synthetic mono- or di-glycerides. Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically- acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions. These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant, such as carboxymethyl cellulose or similar dispersing agents that are commonly used in the formulation of pharmaceutically acceptable dosage forms including emulsions and suspensions. Other commonly used surfactants, such as Tweens, Spans and other emulsifying agents or bioavailability enhancers which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms may also be used for the purposes of formulation.
[0159 ] The pharmaceutically acceptable compositions of this invention may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, aqueous suspensions or solutions. In the case of tablets for oral use, carriers commonly used include lactose and corn starch. Lubricating agents, such as magnesium stearate, are also typically added. For oral administration in a capsule form, useful diluents include lactose and dried cornstarch. When aqueous suspensions are required for oral use, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening, flavoring or coloring agents may also be added. [0160] Alternatively, the pharmaceutically acceptable compositions of this invention may be administered in the form of suppositories for rectal administration. These can be prepared by mixing the agent with a suitable non-irritating excipient that is solid at room temperature but liquid at rectal temperature and therefore will melt in the rectum to release the drug. Such materials include cocoa butter, beeswax and polyethylene glycols.
[0161] The pharmaceutically acceptable compositions of this invention may also be administered topically, especially when the target of treatment includes areas or organs readily accessible by topical application, including diseases of the eye, the skin, or the lower intestinal tract. Suitable topical formulations are readily prepared for each of these areas or organs.
[ 0162 ] Topical application for the lower intestinal tract can be effected in a rectal suppository formulation (see above) or in a suitable enema formulation. Topically-transdermal patches may also be used.
[ 0163 ] For topical applications, the pharmaceutically acceptable compositions may be formulated in a suitable ointment containing the active component suspended or dissolved in one or more carriers. Carriers for topical administration of the compounds of this invention include, but are not limited to, mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene, polyoxypropylene compound, emulsifying wax and water. Alternatively, the pharmaceutically acceptable compositions can be formulated in a suitable lotion or cream containing the active components suspended or dissolved in one or more pharmaceutically acceptable carriers. Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
[ 0164 ] For ophthalmic use, the pharmaceutically acceptable compositions may be formulated, e.g., as micronized suspensions in isotonic, pH adjusted sterile saline or other aqueous solution, or, preferably, as solutions in isotonic, pH adjusted sterile saline or other aqueous solution, either with or without a preservative such as benzylalkonium chloride. Alternatively, for ophthalmic uses, the pharmaceutically acceptable compositions may be formulated in an ointment such as petrolatum. The pharmaceutically acceptable compositions of this invention may also be administered by nasal aerosol or inhalation. Such compositions are prepared according to techniques well-known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other conventional solubilizing or dispersing agents.
[0165] Most preferably, the pharmaceutically acceptable compositions of this invention are formulated for oral administration.
[0166] Liquid dosage forms for oral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsiflers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethyl formamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. Besides inert diluents, the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents. [ 0167 ] Injectable preparations, for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S.P. and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil can be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid are used in the preparation of injectables. [0168] The injectable formulations can be sterilized, for example, by filtration through a bacterial-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.
[ 0169 ] In order to prolong the effect of a compound of the present invention, it is often desirable to slow the absorption of the compound from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material with poor water solubility. The rate of absorption of the compound then depends upon its rate of dissolution that, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered compound form is accomplished by dissolving or suspending the compound in an oil vehicle. Injectable depot forms are made by forming microencapsule matrices of the compound in biodegradable polymers such as polylactide-polyglycolide. Depending upon the ratio of compound to polymer and the nature of the particular polymer employed, the rate of compound release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the compound in liposomes or microemulsions that are compatible with body tissues.
[ 0170 ] Compositions for rectal or vaginal administration are preferably suppositories which can be prepared by mixing the compounds of this invention with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound. [ 0171 ] Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the active compound is mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar—agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such as quaternary ammonium compounds, g) wetting agents such as, for example, cetyl alcohol and glycerol monostearate, h) absorbents such as kaolin and bentonite clay, and i) lubricants such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof. In the case of capsules, tablets and pills, the dosage form may also comprise buffering agents.
[0172 ] Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like. The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polethylene glycols and the like.
[0173 ] The active compounds can also be in micro-encapsulated form with one or more excipients as noted above. The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings, release controlling coatings and other coatings well known in the pharmaceutical formulating art. In such solid dosage forms the active compound may be admixed with at least one inert diluent such as sucrose, lactose or starch. Such dosage forms may also comprise, as is normal practice, additional substances other than inert diluents, e.g., tableting lubricants and other tableting aids such a magnesium stearate and microcrystalline cellulose. In the case of capsules, tablets and pills, the dosage forms may also comprise buffering agents. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes.
[0174] Dosage forms for topical or transdermal administration of a compound of this invention include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches. The active component is admixed under sterile conditions with a pharmaceutically acceptable carrier and any needed preservatives or buffers as may be required. Ophthalmic formulation, ear drops, and eye drops are also contemplated as being within the scope of this invention. Additionally, the present invention contemplates the use of transdermal patches, which have the added advantage of providing controlled delivery of a compound to the body. Such dosage forms can be made by dissolving or dispensing the compound in the proper medium. Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate can be controlled by either providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel.
[ 0175 ] The compounds of the invention are preferably formulated in dosage unit form for ease of administration and uniformity of dosage. The expression "dosage unit form" as used herein refers to a physically discrete unit of agent appropriate for the patient to be treated. It will be understood, however, that the total daily usage of the compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment. The specific effective dose level for any particular patient or organism will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed, and like factors well known in the medical arts.
[0176] The amount of the compounds of the present invention that may be combined with the carrier materials to produce a composition in a single dosage form will vary depending upon the host treated, the particular mode of administration. Preferably, the compositions should be formulated so that a dosage of between 0.01 - 100 mg/kg body weight/day of the inhibitor can be administered to a patient receiving these compositions.
[ 0177 ] Depending upon the parti cular condition, or disease, to be treated or prevented, additional therapeutic agents, which are normally administered to treat or prevent that condition, may also be present in the compositions of this invention. As used herein, additional therapeutic agents that are normally administered to treat or prevent a particular disease, or condition, are known as "appropriate for the disease, or condition, being treated".
[0178] In one embodiment, chemotherapeutic agents or other antiproliferative agents may be combined with the compounds of this invention to treat proliferative diseases and cancer. Chemotherapeutic drugs include but are not limited to alkylating drugs (mechlorethamine, chlorambucil, cyclophosphamide, Melphalan, Ifosfamide), antimetabolites (Methotrexate), purine antagonists and pyrimidine antagonists (6-Mercapto.purine, 5-Fluorouracil, Cytarabile, Gemcitabine), spindle poisons (Vinblastine, Vincristine, Vinorelbine, Paclitaxel), podophyllotoxins (Etoposide, Irinotecan, Topotecan), antibiotics (Doxorubicin, Bleomycin, Mitomycin), nitrosoureas (Carmustine, Lomustine), inorganic ions (Cisplatin, Carboplatin), enzymes (Asparaginase), biologic response modifiers (interferons, interleukins, and tumor necrosis factor (TNF), hormones (Tamoxifen, Leuprolide, Flutamide, and Megestrol), Gleevec™, adriamycin, dexamethasone, and cyclophosphamide. In a further embodiment, additional chemotherapeutic agents include Gleevec™, adriamycin, dexamethasone, vincristine, cyclophosphamide, fluorouracil, topotecan, taxol, interferons, and platinum derivatives. [ 0179 ] Other examples of agents the inhibitors of this invention may also be combined with include, without limitation: agents for treating Multiple Sclerosis (MS) such as beta interferon (e.g., Avonex® and Rebif®), Copaxone®, and mitoxantrone; treatments for asthma such as albuterol and Singulair®; anti-inflammatory agents such as NSAIDS5 corticosteroids, TNF blockers, IL-I RA, azathioprine, cyclophosphamide, and sulfasalazine; immunomodulatory and immunosuppressive agents such as cyclosporin, tacrolimus, rapamycin, mycophenolate mofetil, interferons, corticosteroids, cyclophophamide, azathioprine, and sulfasalazine; agents for treating cardiovascular disease such as beta-blockers, ACE inhibitors, diuretics, nitrates, calcium channel blockers, and statins; agents for treating diabetes, including insulin, glitazones and sulfonyl ureas; and agents for treating blood disorders such as corticosteroids and antileukemic agents.
[0180] The amount of additional therapeutic agent present in the compositions of this invention will be no more than the amount that would normally be administered in a composition comprising that therapeutic agent as the only active agent. Preferably the amount of additional therapeutic agent in the presently disclosed compositions will range from about 50% to 100% of the amount normally present in a composition comprising that agent as the only therapeutically active agent.
Uses of the Compounds and Compositions
[0181] In one embodiment, the invention provides a method of inhibiting
PI3K activity in a patient, comprising administering to said patient a compound or composition of the invention.
[0182] In another embodiment, the invention comprises a method of treating or lessening the severity of a PI3K-mediated condition or disease in a patient. The term "PI3K-mediated disease", as used herein means any disease or other deleterious condition in which PI3K, in particular PI3Kγ, is known to play a role. In another embodiment, a PI3K disease is one in which PI3Kα is known to play a role. In a further embodiment, the invention comprises a method of treating a PI3K-mediated disease. Such conditions include, without limitation, autoimmune diseases, inflammatory diseases, thrombolytic diseases, cancer, cardiovascular diseases, diabetes, allergic diseases, asthma and respiratory diseases. [ 0183 ] In another embodiment, the invention provides a method of treating or lessening the severity of a disease of condition selected from cancer, autoimmune diseases, inflammatory diseases, cardiovascular diseases, diabetes, and allergic diseases or asthma, comprising administering to said patient a compound or composition of the invention.
[ 0184 ] In one embodiment, the invention provides a method of treating or lessening the severity of cancer. Examples of cancers that may be treated or ameliorated by a method of the invention include, without limitation, cancer of the breast, ovary, cervix, prostate, testis, genitourinary tract, esophagus, larynx, glioblastoma, neuroblastoma, stomach, skin, keratoacanthoma, lung, epidermoid carcinoma, large cell carcinoma, small cell carcinoma, lung adenocarcinoma, bone, colon, adenoma, pancreas, adenocarcinoma, thyroid, follicular carcinoma, undifferentiated carcinoma, papillary carcinoma, seminoma, melanoma, sarcoma, bladder carcinoma, liver carcinoma and biliary passages, kidney carcinoma, myeloid disorders, lymphoid disorders, Hodgkin's, hairy cells, buccal cavity and pharynx (oral), lip, tongue, mouth, pharynx, small intestine, colon-rectum, large intestine, rectum, brain and central nervous system. The invention also provides a method of treating or lessening the severity of leukemias, including, without limitation, acute lymphocytic leukemia (ALL), chronic myelogenous leukemia (CML), multiple myeloma and lymphomas. In one embodiment, the invention provides a method of treating or lessening the severity of cancer selected from ovarian cancer, colon cancer, colorectal cancer, breast cancer, brain cancer and lung cancer. [0185 ] , In another embodiment, the invention provides a method of treating or lessening the severity of an autoimmune disease or disorder. Autoimmune diseases or disorders include, without limitation, rheumatoid arthritis, systemic lupus erythematosus (SLE), multiple sclerosis, glomerulonephritis, scleroderma, chronic thyroiditis, Graves' disease, autoimmune gastritis, type I diabetes, autoimmune hemolytic anemia, autoimmune neutropenia, thrombocytopenia, atopic dermatitis, myasthenia gravis, inflammatory bowel disease, ulcerative colitis, Crohn's disease, psoriasis, Sjogren's syndrome and graft vs. host disease. In one embodiment, the autoimmune disease or disorder is rheumatoid arthritis, SLE or mutiple sclerosis.
[ 0186 ] In another embodiment, the invention provides a method of treating or lessening the severity of organ transplantation rejection. [0187 ] In another embodiment, the invention provides a method of treating or lessening the severity of an inflammatory disease. Inflammatory diseases include, without limitation, chronic obstructive pulmonary disease (COPD), bronchitis, emphysema, farmer's lung and related diseases, eosinophilia, lung fibrosis, osteoarthritis, ankylosing spondylitis, sepsis, septic shock, inflammatory myopathies, meningitis, encephalitis, lacrimal parotid gland syndrome, acute respiratory distress syndrome and pancreatitis. In one embodiment, the inflammatory disease is acute respiratory distress syndrome or lacrimal parotid gland syndrome.
[ 0188 ] In another embodiment, the invention provides a method of treating or lessening the severity of a cardiovascular disease. Cardiovascular diseases include, without limitation, atherosclerosis, pulmonary hypertension, deep venous thrombosis, stroke, myocardial infarction, myocardial contractility disorders, ischemia, thromboembolism, pulmonary embolism, acute arterial ischemia, peripheral thrombotic occlusions, coronary artery disease and acute coronary syndrome (ACS). In one embodiment, the cardiovascular disease is atherosclerosis, a myocardial contractility disorder or acute coronary syndrome.
[0189] In another embodiment, the invention provides a method of treating or lessening the severity of type II diabetes.
[0190] In another embodiment, the invention provides a method of treating or lessening the severity of allergic diseases or asthma. Examples of allergic diseases include, without limitation, perennial and seasonal allergic rhinitis, type I hypersensitivity reactions, atopic dermatitis, contact dermatitis, eczema, [0191] In a further embodiment, the invention provides a method of treating or lessening the severity of rheumatoid arthritis, SLE, multiple sclerosis, lacrimal parotid gland syndrome, COPD, pancreatitis, atherosclerosis, myocardial infarction, diabetes or cancer. In another embodiment, the disease or disorder is rheumatoid arthritis, SLE, atherosclerosis, multiple sclerosis or cancer. In another embodiment, the disease or disorder is rheumatoid arthritis, multiple sclerosis or SLE.
[0192 ] In a further embodiment, the method comprises the additional step of administering to said patient one or more additional therapeutic agents. Additional therapeutic agents include chemotherapeutic agents or other anti-proliferative agents, such as Gleevec™, adriamycin, dexamethasone, vincristine, cyclophosphamide, fluorouracil, topotecan, taxol, interferons, and platinum derivatives; agents for treating multiple sclerosis (MS) such as beta interferon (e.g., Avonex® and Rebif®), Copaxone®, and mitoxantrone; treatments for asthma such as albuterol and Singulair®; antiinflammatory agents such as NSAIDS, corticosteroids, TNF blockers, IL-I RA, azathioprine, cyclophosphamide, and sulfasalazine; immunomodulatory and immunosuppressive agents such as cyclosporin, tacrolimus, rapamycin, mycophenolate mofetil, interferons, corticosteroids, cyclophophamide, azathioprine, and sulfasalazine; agents for treating cardiovascular disease such as beta-blockers, ACE inhibitors, diuretics, nitrates, calcium channel blockers, and statins; agents for treating diabetes, including insulin, glitazones and sulfonyl ureas; and agents for treating blood disorders such as corticosteroids and anti-leukemic agents. In one embodiment, said additional therapeutic agent is appropriate for the disease being treated and said additional therapeutic agent is administered together with said composition as a single dosage form or separately from said composition as part of a multiple dosage form. When these additional therapeutic agents are administered separately they may be administered to the patient prior to, sequentially with or following administration of the compositions of this invention.
[0193] In another embodiment, the invention provides a method of inhibiting
PI3K activity in a biological sample, comprising contacting said biological sample with a compound or composition of the invention.
[0194 ] The term "biological sample", as used herein, means an ex vivo sample, and includes, without limitation, cell cultures or extracts thereof; tissue or organ samples or extracts thereof, biopsied material obtained from a mammal or extracts thereof; and blood, saliva, urine, feces, semen, tears, or other body fluids or extracts thereof.
[0195] Inhibition of PBK activity, particularly PI3Kγ activity, in a biological sample is useful for a variety of purposes that arc known to one of skill in the art. Examples of such purposes include, but are not limited to, biological specimen storage and biological assays.
[0196] The compounds of this invention or pharmaceutical compositions thereof may also be used for coating an implantable medical device, such as prostheses, artificial valves, vascular grafts, stents and catheters. Vascular stents, for example, have been used to overcome restenosis (re-narrowing of the vessel wall after injury). However, patients using stents or other implantable devices risk clot formation or platelet activation. These unwanted effects may be prevented or mitigated by pre-coating the device with a pharmaceutically acceptable composition comprising a compound of this invention because, without wishing to be bound to a theory, a compound of the invention could prevent or decrease inflammation or unwanted cellular proliferation, [ 0197 ] Suitable coatings and the general preparation of coated implantable devices are described in US Patents 6,099,562; 5,886,026; and 5,304,121. The coatings are typically biocompatible polymeric materials such as a hydrogel polymer, polymethyldisiloxane, polycaprolactone, polyethylene glycol, polylactic acid, ethylene vinyl acetate, and mixtures thereof. The coatings may optionally be further covered by a suitable topcoat of fluorosilicone, polysaccarides, polyethylene glycol, phospholipids or combinations thereof to impart controlled release characteristics in the composition. Implantable devices coated with a compound of this invention are another embodiment of the present invention. The compounds may also be coated on implantable medical devices, such as beads, or co-formulated with a polymer or other molecule, to provide a "drug depot", thus permitting the drug to be released over a longer time period than administration of an aqueous solution of the drug.
[ 0198 ] The invention also provides a method of screening a compound for the ability to inhibit phosphatidylinositol 3-kinase (PI3K) activity. The method comprises: a) mixing the compound with PI3K, adenosine triphospate (ATP) and a phosphatidylininositol (PI) in a buffer in which PI3K is active to form a mixture; b) incubating the mixture at a temperature and for a time sufficient for said PI3K to phosphorylate said PI; c) measuring the amount of phosphorylated Pl that is produced comprising: i) adding a stop buffer to the mixture in step b) to form a stopped mixture; ii) transferring the quenched mixture to a glass fiber filter (GFF) or hydrophobic membrane; iii) washing the quenched mixture with a wash buffer; and iv) measuring the phosphorylated PI on the GFF or hydrophobic membrane; and d) determining whether said compound inhibits said P13K. Methodology for Synthesis and Characterization of Compounds
[ 0199 ] All references provided in the Examples are herein incorporated by reference. As used herein, all abbreviations, symbols and conventions are consistent with those used in the contemporary scientific literature. See, e.g., Janet S. Dodd, ed., The ACS Style Guide: A Manual for Authors and Editors, 2nd Ed., Washington, D.C.: American Chemical Society, 1997, herein incorporated in its entirety by reference.
EXAMPLES
Example 1: Preparation of Compounds of the Invention [ 0200 ] N-(5-bromo-4-methylthiazol-2-yl)cyclopropanecarboxamide
Figure imgf000038_0001
[ 0201] To a solution of 4-methylthiazol-2-amine (20.4 g) in pyridine (80 ml) under an ice bath was added dropwise cyclopropanecarbonyl chloride (19.7 ml) and the resulting reaction was stirred at room temperature (RT) overnight. A half volume of pyridine was removed using a rotary evaporator to afford a residue, to which water (100 ml) was added. Filtration, washing with water (3xl00ml) and drying under vacuum afforded N-(4-methylthiazol-2-yl)cyclopropanecarboxamide as a white solid (29.92 g).
[0202 ] To N-(4-methylthiazol-2-yl)cyclopropanecarboxamide (10.05 g) in acetic acid (100 ml) under an ice- water bath was added bromine (3.2 ml). After 30 min, water (100ml) was added. Filtration, washing with water (3X) and drying under vacuum afforded N-(5-bromo-4-methylthiazol-2-yl)cyclopropanecarboxamide as a white solid (12.47 g). H NMR (500 MHz, CDC13) 2.39 (s, 3 H), 1.64 - 1.60 (m, 1 H), 1.27 - 1.24 (m, 2 H), 1.00 - 0.96 (m. 2 H) ppm; LC/MS: 261 (M+l) /2.91 min.
[0203 ] Ethyl 5-(2-(cyclopropanecarboxamido)-4-methylthiazol-5- yl)furan-2-carboxy!ate (Compound 1)
Figure imgf000038_0002
Compound 1 [ 0204 ] A solution of JV-(5-bromo-4-methylthiazol-2- yl)cyclopropanecarboxamide (1.0 g), Pd2dba3 (70 mg), and tri-orthotolyl phosphine (300 mg) in 40 mL of THF was degassed via nitrogen bubbling for 2 hrs. To this solution was added 15 mL of a 2.0 M solution of (5-(ethoxycarbonyl)furan-2-yl)(ethyl)zinc (2 equiv) as a single portion. The reaction was warmed to 60° overnight under nitrogen. The reaction was quenched with 10 mL of methanol, concentrated to dryness and the residue suspended in methylene chloride, which was filtered. The filtrate was concentrated and he residue was purified via silica gel chromatography (CH2CI2 to EtOAc) to yield 100 mg of compound 1 as a white solid.
[0205] N-(5-bromo-4-methylthiazol-2-yl) acetamide
Figure imgf000039_0001
[0206] To a solution of 4-methy]thiazol-2-amine (36.28 g) in pyridine (130 ml) under an ice bath was added dropwise acetyl chloride (30 ml) in 30 min. The resulting suspension was left at RT overnight. Most of the pyridine was removed under vacuum. To the residue was added water (500 ml) and the pH was adjusted to 7 by adding aqueous HCl. Extraction with ethyl acetate (3 X 400 ml), washing with brine, drying and concentration under a rotary evaporator gave a residue, which was washed with ether to generate pure N-(4-methylthiazol-2-yl)acetamide as a white solid (41.48 g). [0207 ] To N-(4-methylthiazol-2-yl)acetamide (39.48 g) in acetic acid (200 ml) under a water-ice bath was added dropwise bromine (16 ml). After 2h, water (500 ml) was added. Filtration, washing with water (3X) and drying gave N-(5-bromo-4- methylthiazol-2-yl) acetamide as off-white solid (52.13 g). Hl-NMR (300MHz, d6- DMSO) 12.27 (s, 1 H), 2.22 (s, 3 H), 2.13 (s, 3 H) ppm. [ 0208] N-(5-(5-formylfuran-2-yl)-4-methylthiazol-2-yI)acetamide
Figure imgf000039_0002
[0209] To a solution of 3.0 g of N-(5-bromo-4-methylthiazol-2-yl)acetamide in 50 mL of ΝMP was added 3.0 g of 5-formylfuran-2-ylboronic acid. To the stirred solution was then added 300 mg of l,l'bis-(diphenylphosphino)-ferrocene) palladium dichloride followed by 10 mL of saturated sodium bicabonate. The resulting biphasic mixture was irradiated at 1500C in a microwave for 20 min. The resulting mixture was poured into 200 mL of water and filtered. The filtrate was extracted with ether and the ether layers were combined, dried over sodium sulfate and concentrated. The resulting oil was precipitated with methylene chloride and filtered, yielding 1.2 g of N-(S-(S- formylfuran-2-yl)-4-methylthiazol-2-yl)acetamide as an orange solid which was used without further purification. LC/MS: 251.07 (M+H)/ 2.18 min. [0210] 5-(2-acetamido-4-methylthiazol-5-yI)furan-2-carboxylic acid
Figure imgf000040_0001
[ 0211 J To a solution of 1.2 g of of 7V-(5-(5-formylfuran-2-yl)-4- methylthiazol-2-yl)acetamide in 100 mL of tert-butanol was added 5 mL of water and 20 mL of 2N monobasic sodium phosphate. The resulting suspension was cooled to 00C while being stirred. To the chilled suspension was added 20 mL of a 2M solution of 2- methyl-2-butene in THF followed by 3 g of sodium chlorite in 10 mL of water. The reaction was allowed to warm to room temperature overnight and the upper organic layer was separated and dried over sodium sulfate and used as is in the subsequent reactions. LC/MS: 267.04 (M+H)/2.04 min.
[0212] (R)-5-(2-acetamido-4-methylthiazol-5-yl)-N-(2-hydroxy-l- phenylethyl)furan-2-carboxamide (Compound 10)
Figure imgf000040_0002
[0213] To 5 mL of the crude 4.49 millimolar solution of 5-(2-acetamido-4- methylthiazol-5-yl)furan-2-carboxylic acid was added 200 mg of TBTU and 1 mL of Hunig's base. To the stirred reaction mixture was then added 200 mg of S- phenylglycinol. The reaction was allowed to stir overnight and was then concentrated to dryness and dissolved in 1 mL of DMSO and filtered. The filtrate was purified via reverse phase chromatography to yield 4.1 mg of (R)-5-(2-acetamido-4-methylthiazol-5- yl)-N-(2-hydroxy-l-phenylethyl)furan-2-carboxamide as a white solid. LC/MS: 386.2 (M+H)/2.0 min.
[0214] Compounds 2-9 and 11-18 were prepared in an analogous fashion.
[0215 ] N-(5-(furan-2-yl)-4-methylthiazol-2-yl)acetamide
Figure imgf000041_0001
[0216] A mixture of N-(5-bromo-4-methylthiazol-2-yl) acetamide (4 g), furan-2-yl-2-boronic acid (4.22 g), tetrakis(triphenylphosphine)palladium (1.8 g) and 2 N sodium carbonate (36 ml) in DME (60 ml) was stirred at 95°C for 18 h. After cooling, water (100 ml) was added. Extraction with ethyl acetate (3 X 80 ml), drying, concentration and purification (SiO2, 0 to 100% ethyl acetate in hexane) gave pureN-(5- (furan-2-yl)-4-methylthiazol-2-yl)acetamide (2.95 g).
[ 0217 ] 5-(2-Acetamido-4-methylthiazol-5-yl)furan-2-sulfonyl chloride
Figure imgf000041_0002
[ 0218 ] To N-(5-(furan-2-yl)-4-methylthiazol-2-yl)acetamide (0.443 g) under an ice-salt bath was added sulfurochloridoic acid (ClSO3H, 5 ml). After 30 min, the reaction was poured into crushed ice (150 g). Extraction with dichloromethane (3 X 30 ml), drying and concentration gave 5-(2-acetamido-4-methylthiazol-5-yl)furan-2- sulfonyl chloride (0.359 g) that was used directly for next step. [0219] Preparation of Compounds 19-74
Figure imgf000041_0003
VRTs
[0220] To a solution of 20 mg (0.66 mmol) 5-(2-acetamido-4-methylthiazol-
5-yl)furan-2-sulfonyl chloride in 1 mL dry chloroform was added 0.28 mL (2 mmol, 3 equivalents) triethylamine with stirring to give a colorless solution. To this was added was the appropriate amine (NR1R2; 1.5 equivalent). The resulting reaction was stirred at RT over night. Evaporation gave a residue that was purified by HPLC to give the sulfonamides. Example 2: Analytical Results
[ 0221] Table 3 below depicts exemplaryΗ-NMR data (NMR) and liquid chromatographic mass spectral data, reported as mass plus proton (M+H), as determined by electrospray, and retention time (RT) for certain compounds of the present invention, wherein compound numbers in Table 3 correspond to the compounds depicted in Tables 1 and 2 (empty cells in the NMR column indicate that results were not available): Table 3
Figure imgf000042_0001
Figure imgf000044_0001
Example 3: PΪ3K Inhibition Assay
[ 0222 ] Compounds were screened for their ability to inhibit PDKγ using one of the assays below. Protocol A
[0223 ] To each well of a 96 well polystyrene plate [Corning, Costar Item No.
3696], was added 1.5 μl of a compound of the invention (at various concentrations) along with 28.5 μl of kinase buffer [50 mM HEPES (pH 7.4), 5 mM DTT, 5 mM MgCl2 and 50 mM NaCl] containing 137 μM phosphatidylinositol(4,5) bisphosphate diC8 [PI(4,5)P2; Echelon Biosciences, Cat. No. P-4508] and 158 nM PDKγ. After mixing, 30 μl of kinase buffer containing 80 μM ATP was added to start the reaction. Final substrate concentrations in the assay were 50 mM HEPES (pH 7.4), 5 mM DTT, 5 mM MgCl2, 50 mM NaCl, 40 μM ATP (200 μCi/μmole ATP), 65 μM PI(4,5)P2 and 75 nM PDKγ. After 20 minutes at room temperature (25°C), the reaction was stopped with 30 μl of kinase buffer containing 150 mM EDTA and 1.5 M ammonium sulfate. [ 0224] Each well of a 96 well hydrophobic membrane filter plate [Millipore,
0.45 μm hydrophobic membrane, Cat. No. MSIP N4B] was washed with 100 μL wash buffer [50 mM HEPES (pH 7.4), 5 mM DTT, 5 mM MgCl2, 50 mM NaCl, 5 mM EDTA and 0.5 M ammonium sulfate]. 60 μL of the stopped assay was transferred to each well of the filter plate and soaked for 30 minutes. After washing each well three times with 200 μL wash buffer, 50 μl of scintillation fluid was added. The filter plate was allowed to stand overnight prior to determining 33P incorporation on a Perkin-Elmer Microbeta Trilux™ liquid scintillation counter and luminescence counter. Protocol B
[0225] To each well of a 96 well polystyrene plate [Corning Costar Item No.
3795], 61 μL of Reaction Mix [55 mM HEPES pH 7.5, 11 mM MgCl2, 27 mM NaCl, 5.5 mM DTT, 0.11 mg/mL BSA, 51 μM phosphatidylinositol(4,5)bisphosphate diC16 (PI(4,5)P2; Avanti Polar Lipids, Cat. No. 840046P) and 18 nM PI3Kγ] was added. Separately, ten 2.5-fold serial dilutions of compounds of the invention were prepared in 100% DMSO with the last two wells reserved for no inhibitor controls (enzyme + no compound and no enzyme control + no compound). 1 μL of compound or DMSO alone was added by 12-channel hand pipet and incubated for 10 minutes at room temperature (25°C). To initiate the reaction, 5 μL of ATP Mix [49 mM HEPES pH 7.5 and 1340 μM 33P-ATP was added and allowed to incubate for 30 min. at 25°C. Final concentrations of the reaction were 50 mM HEPES 7.5, 10 mM MgCl2, 25 mM NaCl, 5 mM DTT, 0.1 mg/mL BSA, 46 μM PI(4.5)P2, 16.6 nM PI3Kγ, and 100 μM ATP. Final inhibitor concentrations ranged from 10 μM to 1 nM.
[ 0226] After incubation, 60 μL of the reaction was quenched by transferring to 50 μL of 20% TCA/Water in each well of a 96 well glass fiber filter plate [Millipore Cat. No. MAFBN0B50]. The plate was dried by vacuum and then washed five times in a Bio-Tek Instruments ELX-405 Auto Plate Washer with 150 μL of 5% TCA/Water. 50 μL of scintillation fluid was added to each well and the plate was read immediately on a Perkin-Elmer TopCount™ NXT liquid scintillation counter.
[ 0227 ] Inhibition data was background subtracted (no enzyme control + no inhibitor control) and fit to the competitive tight binding Kj equation described by Morrison and Stone. [J.F. Morrison and S.R. Stone, Comments MoI. Cell Biophys. 2 (1985) 347-368]. [0228 ] Table 4 depicts enzyme inhibition data (Ki) for certain exemplary compounds. Compound numbers in Table 4 correspond to those compounds depicted in Tables 1 and 2. In Table 4, "A" represents a Ki of less than 0.1 μM, "B" represents a K; of between 0.1 and < 1.0 μM, and "C" represents a K; of > 1.0 μM and less than 5.0 μM. If there was more than one result for a given compound, the average Kj was provided. Compound 1 was assessed by Protocol A; all other compounds were assessed by Protocol B. Table 4
Figure imgf000046_0003
Figure imgf000046_0001
Figure imgf000046_0002
[ 0229 ] While we have described a number of embodiments of this invention, it is apparent that our basic examples may be altered to provide other embodiments which utilize the compounds and methods of this invention. Therefore, it will be appreciated that the scope of this invention is to be defined by the appended claims rather than by the specific embodiments which have been represented by way of example.

Claims

CLAIMS We claim: •
1. A compound of formula I
Figure imgf000047_0001
I or a pharmaceutically acceptable salt thereof, wherein:
Y is -C(O)- or a bond;
R1 is a C|-6 aliphatic, wherein up to three methylene units are replaced by -NH-, -NR1 '-, -O-, -S-, -CO2-, -OC(O)-, -C(O)CO-, -C(O)-, -C(O)O-, -C(O)NH-, -C(O)NR11-, -NC(=N-CN)N, -NHCO-, -NR11CO-, -NHC(O)O-, -NR1 1C(O)O-, -SO2NH-, -SO2NR11-, -NHSO2-, -NR11SO2-, -NHC(O)NH-, -NR1 1C(O)NH-, -NHC(O)NR1 1-, -NR1 1C(O)NR1 1, -OC(O)NH-, -OC(O)NR1 1-, -NHSO2NH-, -NR1 1SO2NH-, -NHSO2NR11-, -NR11SO2NR11-, -SO-, or -SO2-; a C3-10 monocyclic or bicyclic cycloaliphatic; a C6-io monocyclic or bicyclic aryl; a 5-10 membered monocyclic or bicyclic heteroaryl; or a 5-10 membered monocyclic or bicyclic heterocyclyl; wherein R1 is optionally substituted with 1-6 occurrences ofJR1; each R1 ' is independently selected from Ci-6 aliphatic, C3-10 monocyclic or bicyclic cycloaliphatic, Ce-io monocyclic or bicyclic aryl, 5-10 membered monocyclic or bicyclic heteroaryl, or 5-10 membered monocyclic or bicyclic heterocyclyl; or two R ' groups, on the same substiruent or different substituents, together with the atom(s) to which each R11 group is bound, form a 3-8 membered heterocyclyl; each JR1 is independently selected from halogen, OH, OR, NO2, NH2, NHR, NR2, SH, SR, CN, C(O)NH2, C(O)NHR, C(O)NR2, C(O)OH, C(O)OR, NHC(O)H, NHC(O)R, NRC(O)H, NRC(O)R, NHC(O)OH, NHC(O)OR, NRC(O)OH, NRC(O)OR, or R; or two JR1, together with the carbon(s) to which they are attached, form a cyclopropyl ring or C=O; each R is independently selected from a Ci-4 aliphatic or C3_4 cycloaliphatic optionally substituted with 1-4 occurrences of halogen, OH, NO2, NH2, SH or CN;
R2 is H, a Ci.6 aliphatic, C3-6 cycloaliphatic, halogen, OH, OR, NO2, NH2, NHR, NR2, SH, SR, CN, C(O)NH2, C(O)NHR, C(O)NR2, C(O)OH, C(O)OR, NHC(O)H, NHC(O)R, NRC(O)H, NRC(O)R, NHC(O)OH, NHC(O)OR, NRC(O)OH, NRC(O)OR, or R; wherein said Ci-6 aliphatic or C3-6 cycloaliphatic is optionally substituted with 1-4 occurrences of J1*2; each J*2 is independently selected from halogen, OH, OR, NO2, NH2, NHR, NR2, SH, SR, CN, C(O)NH2, C(O)NHR, C(O)NR2, C(O)OH, C(O)OR, NHC(O)H, NHC(O)R, NRC(O)H, NRC(O)R, NHC(O)OH, NHC(O)OR, NRC(O)OH, NRC(O)OR, or R; or two J*2, together with the carbon(s) to which they are attached, form a cyclopropyl ring or C=O;
T is -SO2NR4-, -CONR4- or -C(O)O-;
R4 is H or a Ci-6 aliphatic or C3-7 cycloaliphatic optionally substituted with 1-6 occurrences of JR4; each JR4 is independently selected from halogen, OH, OR', NO2, NH2, NHR', NR'2, SH, SR', CN, 5-6 membered aryl or heteroaryl, or R'; or two JR4, together with the carbon(s) to which they are attached, form a cyclopropyl ring or C=O; each R' is independently selected from a Ci-6 aliphatic or C3-7 cycloaliphatic optionally substituted with 1-6 occurrences of halogen, OH, NO2, NH2, SH or CN; b is O or 1 ;
L is a C i-6 aliphatic wherein up to three methylene units are replaced by -NH-, -NR5-, -O- , -S-, -CO2-, -OC(O)-, -C(O)CO-, -C(O)-, -C(O)NH-, -C(O)NR5-, -NC(=N- CN)N, -NHCO-, -NR5CO-, -NHC(O)O-, -NR5C(O)O-, -SO2NH-, -SO2NR5-, -NHSO2-, -NR5SO2-, -NHC(O)NH-, -NR5C(O)NH-, -NHC(O)NR5-, -NR5C(O)NR5, -OC(O)NH-, -OC(O)NR5-, -NHSO2NH-, -NR5SO2NH-, -NHSO2NR5-, -NR5SO2NR5-, -SO-, or -SO2-, wherein L is optionally substituted with 1 -4 occurrences of R ; each R5 is independently selected from Ci-6 aliphatic, C3.10 monocyclic or bicyclic cycloaliphatic, C6-1Q monocyclic or bicyclic aryl, 5-10 membered monocyclic or bicyclic heteroaryl, or 5-10 membered monocyclic or bicyclic heterocyclyl; or two R5 groups, on the same substituent or different substituents, together with the atom(s) to which each R5 group is bound, form a 3-8 membered heterocyclyl; each R6 is independently selected from halogen, OH, OR', NO2, NH2, NHR', NR'2, SH, SR', CN, or R'; or two R6, together with the carbon(s) to which they are attached, form a cyclopropyl ring or C=O; n is 0 or 1 ;
R3 is H or an optionally substituted group selected from a Ci-6 aliphatic, a C3-Io monocyclic or bicyclic cycloaliphatic, a Cβ-io monocyclic or bicyclic aryl, a 5-10 membered monocyclic or bicyclic heteroaryl, or a 5-10 membered monocyclic or bicyclic heterocyclyl, wherein R3 is optionally substituted with 1-6 occurrences of J1*3; or R3 and R4, together with the nitrogen to which R4 is attached, form a 5- 10 membered monocyclic or bicyclic heterocyclyl or heteroaryl, wherein n is 0 and said heterocyclyl or heteroaryl is optionally substituted with 1-6 occurrences ofJ113; each J^ is independently selected from -(U)m-X;
U is a Ci-4 aliphatic, wherein up to two methylene units are optionally and independently replaced by Gu and wherein U is optionally substituted with 1-4 Ju;
Gu is -NH-, -NR7-, -O-, -S-, -CO2-, -OC(O)-, -C(O)CO-, -C(O)-, -C(O)NH-, -C(O)NR7-, -NC(=N-CN)N-, -NHCO-, -NR7CO-, -NHC(O)O-, -NR7C(O)O-, -SO2NH-, - SO2NR7-, -NHSO2-, -NR7SO2-, -NHC(O)NH-, -NR7C(O)NH-, -NHC(O)NR7-, -NR7C(O)NR7, -OC(O)NH-, -OC(O)NR7-, -NHSO2NH-, -NR7SO2NH-, -NHSO2NR7-, -NR7SO2NR7-, -SO-, or -SO2-;
R7 is Ci-6 aliphatic or C3-7 cycloaliphatic optionally substituted with 1-6 occurrences of halogen, OH, NO2, NH2, SH or CN; m is O or 1 ; each Ju is independently selected from halogen, OH, OR', NO2, NH2, NHR', NR'2, SH, SR', CN, or R'; or two Ju, together with the carbon(s) to which they are attached, form a cyclopropyl ring or C=O;
X is H, halogen, OH, OR, NO2, NH2, NHR, NR2, SH, SR, CN, C(O)NH2, C(O)NHR, C(O)NR2, C(O)OH, C(O)OR, NHC(O)H, NHC(O)R, NRC(O)H, NRC(O)R, NHC(O)OH, NHC(O)OR, NRC(O)OH, NRC(O)OR, or a group selected from a Ci-6 aliphatic, a C3-Io monocyclic or bicyclic cycloaliphatic, a C6-Io monocyclic or bicyclicaryl, a 5-10 membered monocyclic or bicyclic heteroaryl, or a 5-10 membered monocyclic or bicyclic heterocyclyl, wherein said group is optionally substituted with 1 -4 Jx, or two X, together with the carbon(s) to which they are attached, form a cyclopropyl ring or C=O; wherein X is not H when m is 0; each Jx is independently selected from halogen, OH, OR5 NO2, NH2, NHR3 NR2, SH, SR, CN, C(O)NH2, C(O)NHR, C(O)NR2, C(O)OH, C(O)OR, NHC(O)H, NHC(O)R, NRC(O)H, NRC(O)R, NHC(O)OH, NHC(O)OR, NRC(O)OH, NRC(O)OR; and
R9 is selected from H; halogen; OH; NO2; NH2; SH; CN; or a group selected from a Ci-6 aliphatic or a C3-7 cycloaliphatic, wherein said group is optionally substituted with 1-6 occurrences of halogen, OH, NO2, NH2, SH or CN.
2. The compound or salt thereof according to claim 2, wherein Y is C(O) and the compound is of formula II:
Figure imgf000050_0001
3. The compound or salt thereof according to either of claims 1 or 2, wherein R1 is a Ci-6 aliphatic optionally substituted with 1-6 occurrences of JR1.
4. The compound or salt thereof according to claim 3, wherein R1 is a Ci-3 aliphatic optionally substituted with 1-3 occurrences of JR1.
5. The compound or salt thereof according to claim 4, wherein R1 is a Ci-3 aliphatic.
6. The compound or salt thereof according to claim 5, wherein R1 is CH3, CH2CH3 or cyclopropyl.
7. The compound or salt thereof according to any one of claims 1-6, wherein R2 is Ci-3 aliphatic, wherein R2 is optionally substituted with 1-3 occurrences of j"2.
8. The compound or salt thereof according to claim 7, wherein R2 is C1-3 aliphatic.
9. The compound or salt thereof according to claim 8, wherein R 2 i -s CH3
10. The compound or salt thereof according to any one of claims 1-9, wherein said compound is of formula HIa, HIb or HIc:
Figure imgf000051_0001
HIa nib IIIc.
11. The compound or salt thereof according to claim 10, wherein said compound is of formula HIa', IHb' or IIIc':
Figure imgf000051_0002
Ilia' HIb' IIIc'.
12. The compound or salt thereof according to any one of claims 1-11, wherein R4 is H or Ci-3 aliphatic optionally substituted with one occurrence of JR4.
13. The compound or salt thereof according to claim 12, wherein R4 is H, CH3, CH2CH3, CH2CH2OH, CH2CHCH2, cyclopropyl or CH2phenyl.
14. The compound or salt thereof according to claim 13, wherein R4 is H or CH3.
15. The compound or salt thereof according to any one of claims 1-12, wherein R3 and R4, together with the nitrogen to which R4 is attached, form a 5-10 membered monocyclic or bicyclic heterocyclyl or heteroaryl; wherein said heterocyclyl or heteroaryl is optionally substituted with 1-6 occurrences of J1*3.
16. The compound or salt thereof according to claim 15, wherein said heterocyclyl or heteroaryl formed by R and R is a 5-6 membered monocyclic heterocyclyl or 9-10 membered heterocyclyl or heteroaryl; wherein said monocyclic or bicyclic heterocyclyl or bicyclic heteroaryl contains 1-3 heteroatoms and is optionally substituted with 1-3 occurrences of JR3.
17. The compound or salt thereof according to claim 16, wherein said monocyclic or bicyclic heterocyclyl or bicyclic heteroaryl contains 1-2 heteroatoms and is optionally substituted with 1-2 occurrences of 3*°.
18. The compound or salt thereof according to claim 16, wherein said monocyclic or bicyclic heterocyclyl or bicyclic heteroaryl is selected from the following:
Figure imgf000052_0001
wherein Z is CH2, NH, O or S5 and wherein the hydrogens of said CH2 of Z is optionally substituted by 1-2 occurrences of J1*3 and the hydrogen of said NH of Z is optionally substituted by J1*3.
19. The compound or salt thereof according to any one of claims 15-18, wherein each occurrence of J*3 is selected from J^3 is selected from halogen; OH; OR; CN; NH2; NHR; NR2; C(O)NH2; C(O)NHR; C(O)NR2; NHCOR; C(O)OR; C(O)OH; oxo; (CH2)0- 3phenyl optionally substituted with halogen, OH, OR, NO2, NH2, SH or CN; or Ci-4 aliphatic optionally substituted with halogen, OH, OR, NO2, NH2, SH or CN.
20. The compound or salt thereof according to any one of claims 1-11, wherein R3 is H or an optionally substituted group selected from a C^ aliphatic, a C3. io monocyclic or bicyclic cyclo aliphatic, a Cβ-io monocyclic or bicyclic aryl, a 5-10 membered monocyclic or bicyclic heteroaryl, or a 5-10 membered monocyclic or bicyclic heterocyclyl, wherein R3 is optionally substituted with 1-6 occurrences of J113.
21. The compound or salt thereof according to claim 20, wherein n is 1 and L is a C i-3 aliphatic optionally substituted with 1-2 occurrences of R'.
22. The compound or salt thereof according to claim 21, wherein L is -CH2- or CH substituted with a C1-2 aliphatic, which Ci-2 aliphatic is optionally substituted with 1- 2 occurrences of halogen, OH3 NO2, NH2, SH or CN.
23. The compound or salt thereof according to claim 22, wherein L is -CH2-.
24. The compound or salt thereof according to claim 20, wherein n is O.
25. The compound or salt thereof according to any one of claims 20-24, wherein R3 is a Ci-4 aliphatic, a C3-6 monocyclic cycloaliphatic, phenyl, a 5-6 membered monocyclic heteroaryl, or a 5-10 membered monocyclic or bicyclic heterocyclyl, wherein R3 is optionally substituted with 1-4 occurrences of JR3.
26. The compound or salt thereof according to claim 25, wherein R3 is a Ci-4 aliphatic, a C3-6 monocyclic cycloaliphatic, phenyl, a 5-6 membered monocyclic heteroaryl, or a 5-6 membered monocyclic heterocyclyl, wherein R is optionally substituted with 1-4 occurrences of JR3.
27. The compound or salt thereof according to any one of claims 20-26, wherein each occurrence of JR3 is selected from halogen; OH; OR; CN; NH2; NHR; NR2; C(O)NH2; C(O)NHR; C(O)NR2; NHCOR; C(O)OR; C(O)OH; oxo; (CH2)0-3phenyl optionally substituted with halogen, OH, OR, NO2, NH2, SH or CN; or C, ^ aliphatic optionally substituted with halogen, OH, OR, NO2, NH2, SH or CN.
28. The compound or salt thereof according to any one of claims 1-27, wherein R8 is selected from H, cyclopropyl, or a C 1.3 aliphatic optionally substituted with 1-3 occurrences of halogen or OH.
29. The compound or salt thereof according to claim 28, wherein R8 is selected from H, halogen, methyl or ethyl.
30. The compound or salt thereof according to claim 29, wherein R8 is H.
31. The compound or salt thereof according to any one of claims 1-30, wherein R9 is selected from H, OH, halogen, cyclopropyl, or a Ci-3 aliphatic optionally substituted with 1-3 occurrences of halogen or OH.
32. The compound or salt thereof according to claim 31, wherein R9 is selected from H, OH, halogen, methyl or ethyl.
33. The compound or salt thereof according to claim 32, wherein R9 is H or methyl.
34. The compound or salt thereof according to claim 33, wherein R9 is H.
35. The compound according to claim 1, which is selected from Table 1 or a pharmaceutically acceptable salt thereof.
36. The compound according to claim 1, which is selected from Table 2 or a pharmaceutically acceptable salt thereof.
37. A pharmaceutical composition comprising a compound according to any one of claims 1-36 and a pharmaceutically acceptable carrier, adjuvant, or vehicle.
38. The composition according to claim 37, additionally comprising a therapeutic agent selected from a chemotherapeutic agent, an anti-proliferative agent, an agent for treating asthma, an anti -inflammatory agent, an immunomodulatory agent, an immunosuppressive agent, an agent for treating cardiovascular disease, an agent for treating diabetes, or an agent for treating blood disorders.
39. A method of inhibiting phosphatidylinositol 3-kinase (PI3K) activity in a biological sample, comprising contacting said biological sample with a compound or salt thereof according to any one of claims 1-36 or with a composition according to either of claims 37-38.
40. A method of inhibiting PI3K kinase activity in a patient, comprising administering to said patient a compound or salt thereof according to any one of claims 1-36 or with a composition according to either of claims 37-38.
41. A method of treating or lessening the severity of a disease or condition selected from cancer, an autoimmune disease, an inflammatory disease, a cardiovascular disease, diabetes, an allergic disease, asthma or organ transplantation rejection in a patient in need thereof, comprising the step of administering to said patient a compound or salt thereof according to any one of claims 1-36 or with a composition according to either of claims 37-38.
42. The method according to claim 41, wherein said disease or condition is selected from rheumatoid arthritis, systemic lupus erythematosus, atherosclerosis, lacrimal parotid gland syndrome, chronic obstructive pulmonary disease, pancreatitis, myocardial infarction, diabetes, multiple sclerosis or cancer.
43. The method of claim 41, comprising the additional step of administering to said patient an additional therapeutic agent selected from a chemotherapeutic agent, an anti-proliferative agent, an agent for treating asthma, an anti-inflammatory agent, an immunomodulatory agent, an immunosuppressive agent, an agent for treating cardiovascular disease, an agent for treating diabetes, an agent for treating blood disorders, or any combination thereof, wherein said additional therapeutic agent is appropriate for the disease being treated.
44. A method of screening a compound for the ability to inhibit phosphatidylinositol 3-kinase (PI3K) activity comprising: a) mixing the compound with P13K, adenosine triphospate (ATP) and a phosphatidylininositol (PI) in a buffer in which PI3K is active to form a mixture; b) incubating the mixture at a temperature and for a time sufficient for said PI3K to phosphorylate said PI; c) measuring the amount of phosphorylated PI that is produced comprising: i) adding a stop buffer to the mixture in step b) to form a stopped mixture; ii) transferring the quenched mixture to a glass fiber filter (GFF) or hydrophobic membrane; iii) washing the quenched mixture with a wash buffer; and iv) measuring the phosphorylated PI on the GFF or hydrophobic membrane; and d) determining whether said compound inhibits said PI3K.
45. The method according to claim 44, wherein said method further comprises a negative control comprising: a') mixing PI3K, adenosine triphospate (ATP) and a phosphatidylininositol (PI) in a buffer in which PI3K is active to form a negative control mixture; b') incubating the mixture at a temperature and for a time sufficient for said PBK to phosphorylate said PI; and c') measuring the amount of phosphorylatcd PI that is produced comprising: i) adding a stop buffer to the mixture in step b') to form a stopped mixture; ii) transferring the quenched mixture to a glass fiber filter (GFF) or hydrophobic membrane; iii) washing the quenched mixture with a wash buffer; and iv) measuring the phosphorylated PI on the GFF or hydrophobic membrane.
46. The method according to claim 45, wherein step d) is performed by comparing the amount of phosphorylated PI produced in step c) with the amount of phosphorylated PI produced in step c'); wherein a decrease in the amount of phosphorylated PI that is produced in the presence of said compound compared to the amount of phosphorylated PI that is produced in the absence of said compound indicates that said compound is an inhibitor of PDK.
47. The method according to claim 44, wherein said method further comprises a positive control comprising: a") mixing a known PI3K inhibitor with PI3K, adenosine triphospate (ATP) and a phosphatidylininositol (PI) in a buffer in which PI3K is active to form a positive control mixture; b") incubating the mixture at a temperature and for a time sufficient for said PI3K to phosphorylate said PI in the absence of said known PI3K inhibitor; and c") measuring the amount of phosphorylated PI that is produced comprising: comprising: i) adding a stop buffer to the mixture in step b") to form a stopped mixture; ii) transferring the quenched mixture to a glass fiber filter (GFF) or hydrophobic membrane; iii) washing the quenched mixture with a wash buffer; and iv) measuring the phosphorylated PI on the GFF or hydrophobic membrane.
48. The method according to claim 47, wherein the inhibition of PI3K by said compound is compared to the inhibition of PI3K by said known inhibitor.
49. The method according to any one of claims 44-48, wherein said PI3K is PBKγ. '
50. The method according to claim 49, wherein said PBKγ is at a final concentration of 10-100 nM.
51. The method according to any one of claims 44-50, wherein said PI is unphosphorylated PI, PI 4-phosphate [PI(4)P] or PI 4,5-bisphosphate [PI(4,5)P2].
52. The method according to claim 51, wherein said PI is PI(4,5)P2.
53. The method according to claim 52, wherein said PI(4,5)P2 is PI(4,5)P2diC8- C16.
54. The method according to claim 53, wherein said PI(4,5)P2diC8-C16 is PI(4,5)P2diC8 or PI(4,5)P2diC16.
55. The method according to claim 54, wherein said PI(4,5)P2diC8 or PI(4,5)P2diC16 is at a final concentration of 20-100 μM.
56. The method according to any one of claim 44-55, wherein said buffer comprises a magnesium salt, NaCl and a reducing agent at a pH of between 7.0 and 8.0.
57. The method according to claim 56, wherein said magnesium salt is MgCl2.
58. The method according to claim 57, wherein said reducing agent is dithiothreitol (DTT) or 2-mercaρtoethanol.
59. The method according to claim 58, wherein said reducing agent is DTT.
60. The method according to claim 59, wherein said buffer is at a pH of between 7.3 and 7.7.
61. The method according to claim 60, wherein said buffer is at a pH of 7.4 to 7.6.
62. The method according to either of claims 59 or 60, wherein said buffer comprises HEPES.
63. The method according to claim 62, wherein said buffer comprises 40-100 mM HEPES pH 7.4-7.6, 2-7 mM DTT, 2-20 mM MgCl2, and 20-100 mM NaCl.
64. The method according to claim 63, wherein said buffer comprises 40-60 mM HEPES pH 7.4-7.5, 5-6 mM DTT and 25-60 mM NaCl.
65. The method according to any one of claims 44-64, wherein said ATP is at a final concentration of 20-200 μM.
66. The method according to claim 65, wherein said ATP is at a final concentration of 40-100 μM.
67. The method according to any one of claims 44-66, wherein step b) is performed at between 18-370C for 10 minutes to 6 hours.
68. The method according to claim 67, wherein step b) is performed at between 18-3O°C for 10 minutes to 1 hour.
69. The method according to claim 68, wherein step b) is performed at between 20-270C for 15-30 minutes.
70. The method according to any one of claims 44-69, wherein step c) is performed by measuring a labeled phosphate on the PI, wherein said labeled phosphate is transferred from a γ-labeled ATP.
71. The method according to claim 70, wherein said labeled phosphate is a radioactively labeled phosphate transferred from a radioactively γ-labeled ATP.
72. The method according to claim 71 , wherein said ATP is [γ- 33* P]-ATP or [γ-
32P]-ATP.
73. The method according to any one of claims 44-72, wherein said stop buffer is a trichloroacetic acid (TCA)Λvater mixture when said GFF is used.
74. The method according to claim 73, wherein said stop buffer is at a final concentration of 5-20% TCA/water and said wash buffer is at a final concentration of 5- 10% TC A/water.
75. The method according to claim 74, wherein said stop buffer comprises ammonium sulfate when said hydrophobic membrane is used.
76. The method according to claim 75, wherein said stop buffer and said wash buffer comprises a final concentration of 0.3-0.7 M ammonium sulfate and said hydrophobic membrane is a 0.45 μm polyvinylidine fluoride (PVDF) membrane.
77. The method according to claim 76, wherein said stop buffer and said wash buffer comprises a final concentration of 0.5 M ammonium sulfate.
78. The method according to any one of claims 44-77, wherein said method is performed in a six-well, 12-well, 24-well, 48-well, 96-well or 384-well plate.
PCT/US2007/019261 2006-09-01 2007-09-04 5- (2-furyl)-1, 3-thiazole derivatives useful as inhibitors of phosphatidylinositol 3-kinase WO2008027584A2 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
US12/438,995 US8268818B2 (en) 2006-09-01 2007-09-04 Inhibitors of C-MET and uses thereof
MX2009002239A MX2009002239A (en) 2006-09-01 2007-09-04 5- (2-furyl)-1, 3-thiazole derivatives useful as inhibitors of phosphatidylinositol 3-kinase.
CA002662196A CA2662196A1 (en) 2006-09-01 2007-09-04 Inhibitors of phosphatidylinositol 3-kinase
JP2009526754A JP2010502628A (en) 2006-09-01 2007-09-04 5- (2-Furyl) -1,3-thiazole derivatives useful as inhibitors of phosphatidylinositol 3-kinase
AU2007290359A AU2007290359B2 (en) 2006-09-01 2007-09-04 5- (2-furyl)-1, 3-thiazole derivatives useful as inhibitors of phosphatidylinositol 3-kinase
NZ575346A NZ575346A (en) 2006-09-01 2007-09-04 5- (2-furyl)-1, 3-thiazole derivatives useful as inhibitors of phosphatidylinositol 3-kinase
EP07811661A EP2061787A2 (en) 2006-09-01 2007-09-04 5-(2-furyl)-1,3-thiazole derivatives useful as inhibitors of phosphatidylinositol 3-kinase

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US84193506P 2006-09-01 2006-09-01
US60/841,935 2006-09-01

Publications (2)

Publication Number Publication Date
WO2008027584A2 true WO2008027584A2 (en) 2008-03-06
WO2008027584A3 WO2008027584A3 (en) 2008-06-19

Family

ID=38984052

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2007/019261 WO2008027584A2 (en) 2006-09-01 2007-09-04 5- (2-furyl)-1, 3-thiazole derivatives useful as inhibitors of phosphatidylinositol 3-kinase

Country Status (9)

Country Link
US (1) US8268818B2 (en)
EP (1) EP2061787A2 (en)
JP (1) JP2010502628A (en)
CN (1) CN101528738A (en)
AU (1) AU2007290359B2 (en)
CA (1) CA2662196A1 (en)
MX (1) MX2009002239A (en)
NZ (1) NZ575346A (en)
WO (1) WO2008027584A2 (en)

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009021740A2 (en) 2007-08-15 2009-02-19 Sanofis-Aventis Substituted tetrahydronaphthalenes, process for the preparation thereof and the use thereof as medicaments
WO2010144513A1 (en) * 2009-06-10 2010-12-16 Vertex Pharmaceuticals Incorporated Inhibitors of phosphatidylinositol 3-kinase
WO2011048936A1 (en) 2009-10-19 2011-04-28 大正製薬株式会社 Aminothiazole derivative
WO2011107494A1 (en) 2010-03-03 2011-09-09 Sanofi Novel aromatic glycoside derivatives, medicaments containing said compounds, and the use thereof
WO2011157827A1 (en) 2010-06-18 2011-12-22 Sanofi Azolopyridin-3-one derivatives as inhibitors of lipases and phospholipases
WO2011161030A1 (en) 2010-06-21 2011-12-29 Sanofi Heterocyclic substituted methoxyphenyl derivatives having an oxo group, method for producing same, and use thereof as gpr40 receptor modulators
WO2012004270A1 (en) 2010-07-05 2012-01-12 Sanofi Spirocyclically substituted 1,3-propane dioxide derivatives, methods for the production thereof and use of the same as medicament
WO2012004269A1 (en) 2010-07-05 2012-01-12 Sanofi (2-aryloxy-acetylamino)-phenyl-propionic acid derivatives, method for producing same and use thereof as pharmaceuticals
WO2012010413A1 (en) 2010-07-05 2012-01-26 Sanofi Aryloxy-alkylene substituted hydroxyphenyl hexynoic acids, methods for the production thereof and use of the same as medicament
EP2567959A1 (en) 2011-09-12 2013-03-13 Sanofi 6-(4-Hydroxy-phenyl)-3-styryl-1H-pyrazolo[3,4-b]pyridine-4-carboxylic acid amide derivatives as kinase inhibitors
WO2013037390A1 (en) 2011-09-12 2013-03-21 Sanofi 6-(4-hydroxy-phenyl)-3-styryl-1h-pyrazolo[3,4-b]pyridine-4-carboxylic acid amide derivatives as kinase inhibitors
WO2012172438A3 (en) * 2011-06-07 2013-03-28 Dynamix Pharmaceuticals Ltd. Compositions and methods for modulating a kinase
WO2013045413A1 (en) 2011-09-27 2013-04-04 Sanofi 6-(4-hydroxy-phenyl)-3-alkyl-1h-pyrazolo[3,4-b]pyridine-4-carboxylic acid amide derivatives as kinase inhibitors
WO2014068070A1 (en) 2012-10-31 2014-05-08 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for preventing antiphospholipid syndrome (aps)
WO2016059220A1 (en) 2014-10-16 2016-04-21 INSERM (Institut National de la Santé et de la Recherche Médicale) Tcr-activating agents for use in the treatment of t-all
US10004730B2 (en) 2011-10-12 2018-06-26 University of Pittsburgh—of the Commonwealth System of Higher Education Small molecules targeting androgen receptor nuclear localization and/or level in prostate cancer
US10544110B2 (en) 2013-09-20 2020-01-28 University of Pittsburgh—of the Commonwealth System of Higher Education Small molecule inhibitors of the nuclear translocation of androgen receptor for the treatment of castration-resistant prostate cancer
US10882834B2 (en) 2013-09-20 2021-01-05 University of Pittsburgh—of the Commonwealth System of Higher Education Compounds for treating prostate cancer
US10980806B2 (en) 2016-03-24 2021-04-20 University of Pittsburgh—of the Commonwealth System of Higher Education Small molecule inhibitors of the nuclear translocation of androgen receptor for the treatment of castration-resistant prostate cancer

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2603216A4 (en) * 2010-08-11 2013-12-18 Millennium Pharm Inc Heteroaryls and uses thereof

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003072557A1 (en) * 2002-02-28 2003-09-04 Novartis Ag 5-phenylthiazole derivatives and use as pi3 kinase inhibitors
WO2004078754A1 (en) * 2003-03-06 2004-09-16 Novartis Ag 5-phenylthiazole derivatives and their use as p13 kinase inhibitors
WO2004096797A1 (en) * 2003-05-02 2004-11-11 Novartis Ag Inhibitors of phosphatidylinositol 3-kinase
WO2006051270A1 (en) * 2004-11-09 2006-05-18 Astrazeneca Ab 5-heteroaryl thiazoles and their use as p13k inhibitors
EP1700856A1 (en) * 2003-12-26 2006-09-13 Kyowa Hakko Kogyo Co., Ltd. Thiazole derivative

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5308857A (en) * 1988-08-15 1994-05-03 Fujisawa Pharmaceutical Co., Ltd. Furylthiazole derivatives, processes for the preparation thereof and pharmaceutical composition comprising the same
EP1795781B1 (en) * 2004-09-03 2009-11-11 Aichi Steel Corporation Vibration damping member for machine part and method of manufacturing the same

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003072557A1 (en) * 2002-02-28 2003-09-04 Novartis Ag 5-phenylthiazole derivatives and use as pi3 kinase inhibitors
WO2004078754A1 (en) * 2003-03-06 2004-09-16 Novartis Ag 5-phenylthiazole derivatives and their use as p13 kinase inhibitors
WO2004096797A1 (en) * 2003-05-02 2004-11-11 Novartis Ag Inhibitors of phosphatidylinositol 3-kinase
EP1700856A1 (en) * 2003-12-26 2006-09-13 Kyowa Hakko Kogyo Co., Ltd. Thiazole derivative
WO2006051270A1 (en) * 2004-11-09 2006-05-18 Astrazeneca Ab 5-heteroaryl thiazoles and their use as p13k inhibitors

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
USOVA, E. B.; KRAPIVIN, G. D.; KUL'NEVICH, V. G.: "Synthesis of 5-(5-substituted 2-furyl)thiazole derivatives by reaction of 2-(5-substituted furfuryl)thiuronium salts and acetic anhydride" KHIMIYA GETEROTSIKLICHESKIKH SOEDINENII, vol. 4, 1990, pages 557-562, XP009095544 *

Cited By (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009021740A2 (en) 2007-08-15 2009-02-19 Sanofis-Aventis Substituted tetrahydronaphthalenes, process for the preparation thereof and the use thereof as medicaments
WO2010144513A1 (en) * 2009-06-10 2010-12-16 Vertex Pharmaceuticals Incorporated Inhibitors of phosphatidylinositol 3-kinase
CN102548990A (en) * 2009-06-10 2012-07-04 沃泰克斯药物股份有限公司 Inhibitors of phosphatidylinositol 3-kinase
US8344153B2 (en) 2009-06-10 2013-01-01 Vertex Pharmaceuticals Incorporated Inhibitors of phosphatidylinositol 3-kinase
US8637557B2 (en) 2009-10-19 2014-01-28 Taisho Pharmaceutical Co., Ltd Aminothiazole derivative
WO2011048936A1 (en) 2009-10-19 2011-04-28 大正製薬株式会社 Aminothiazole derivative
WO2011107494A1 (en) 2010-03-03 2011-09-09 Sanofi Novel aromatic glycoside derivatives, medicaments containing said compounds, and the use thereof
WO2011157827A1 (en) 2010-06-18 2011-12-22 Sanofi Azolopyridin-3-one derivatives as inhibitors of lipases and phospholipases
WO2011161030A1 (en) 2010-06-21 2011-12-29 Sanofi Heterocyclic substituted methoxyphenyl derivatives having an oxo group, method for producing same, and use thereof as gpr40 receptor modulators
WO2012004270A1 (en) 2010-07-05 2012-01-12 Sanofi Spirocyclically substituted 1,3-propane dioxide derivatives, methods for the production thereof and use of the same as medicament
WO2012004269A1 (en) 2010-07-05 2012-01-12 Sanofi (2-aryloxy-acetylamino)-phenyl-propionic acid derivatives, method for producing same and use thereof as pharmaceuticals
WO2012010413A1 (en) 2010-07-05 2012-01-26 Sanofi Aryloxy-alkylene substituted hydroxyphenyl hexynoic acids, methods for the production thereof and use of the same as medicament
AU2012270029B2 (en) * 2011-06-07 2017-08-10 Clevexel Pharma Compositions and methods for modulating a kinase
WO2012172438A3 (en) * 2011-06-07 2013-03-28 Dynamix Pharmaceuticals Ltd. Compositions and methods for modulating a kinase
WO2013037390A1 (en) 2011-09-12 2013-03-21 Sanofi 6-(4-hydroxy-phenyl)-3-styryl-1h-pyrazolo[3,4-b]pyridine-4-carboxylic acid amide derivatives as kinase inhibitors
EP2567959A1 (en) 2011-09-12 2013-03-13 Sanofi 6-(4-Hydroxy-phenyl)-3-styryl-1H-pyrazolo[3,4-b]pyridine-4-carboxylic acid amide derivatives as kinase inhibitors
WO2013045413A1 (en) 2011-09-27 2013-04-04 Sanofi 6-(4-hydroxy-phenyl)-3-alkyl-1h-pyrazolo[3,4-b]pyridine-4-carboxylic acid amide derivatives as kinase inhibitors
US10004730B2 (en) 2011-10-12 2018-06-26 University of Pittsburgh—of the Commonwealth System of Higher Education Small molecules targeting androgen receptor nuclear localization and/or level in prostate cancer
WO2014068070A1 (en) 2012-10-31 2014-05-08 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for preventing antiphospholipid syndrome (aps)
US10544110B2 (en) 2013-09-20 2020-01-28 University of Pittsburgh—of the Commonwealth System of Higher Education Small molecule inhibitors of the nuclear translocation of androgen receptor for the treatment of castration-resistant prostate cancer
US10882834B2 (en) 2013-09-20 2021-01-05 University of Pittsburgh—of the Commonwealth System of Higher Education Compounds for treating prostate cancer
WO2016059220A1 (en) 2014-10-16 2016-04-21 INSERM (Institut National de la Santé et de la Recherche Médicale) Tcr-activating agents for use in the treatment of t-all
US10980806B2 (en) 2016-03-24 2021-04-20 University of Pittsburgh—of the Commonwealth System of Higher Education Small molecule inhibitors of the nuclear translocation of androgen receptor for the treatment of castration-resistant prostate cancer
US11766433B2 (en) 2016-03-24 2023-09-26 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Small molecule inhibitors of the nuclear translocation of androgen receptor for the treatment of castration-resistant prostate cancer

Also Published As

Publication number Publication date
WO2008027584A3 (en) 2008-06-19
JP2010502628A (en) 2010-01-28
NZ575346A (en) 2012-02-24
AU2007290359A1 (en) 2008-03-06
CA2662196A1 (en) 2008-03-06
MX2009002239A (en) 2009-04-27
AU2007290359B2 (en) 2012-09-20
US8268818B2 (en) 2012-09-18
US20100144721A1 (en) 2010-06-10
CN101528738A (en) 2009-09-09
EP2061787A2 (en) 2009-05-27

Similar Documents

Publication Publication Date Title
AU2007290359B2 (en) 5- (2-furyl)-1, 3-thiazole derivatives useful as inhibitors of phosphatidylinositol 3-kinase
JP5389785B2 (en) Thiazoles and pyrazoles useful as kinase inhibitors
WO2007117494A1 (en) Deazapurines useful as inhibitors of janus kinases
US8686143B2 (en) Compounds useful as inhibitors of Janus kinases
JP2010528021A (en) Thiazoles and pyrazoles useful as kinase inhibitors
NZ582349A (en) Heterocyclic compounds useful as raf kinase inhibitors
EP1973911A2 (en) Azaindoles useful as inhibitors of janus kinases
AU2005309566A1 (en) Bicyclic inhibitors or Rho kinase
WO2007064797A2 (en) Inhibitors of c-met and uses thereof
EP1660487B1 (en) Pyrrole compositions useful as inhibitors of c-met
JP2012529525A (en) Inhibitors of phosphatidylinositol 3-kinase
AU2010250055A1 (en) Tri-cyclic pyrazolopyridine kinase inhibitors
JP2010519174A (en) Heterocyclic inhibitors of c-Met and methods of use thereof
JP2009515984A (en) Azaindazole useful as a kinase inhibitor
JP2011518168A (en) Inhibitors of phosphatidylinositol 3-kinase
JP2013506691A (en) Pyrazole inhibitors of phosphatidylinositol 3-kinase

Legal Events

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

Ref document number: 200780039434.0

Country of ref document: CN

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

Ref document number: 07811661

Country of ref document: EP

Kind code of ref document: A2

WWE Wipo information: entry into national phase

Ref document number: 2009526754

Country of ref document: JP

Ref document number: 2662196

Country of ref document: CA

Ref document number: MX/A/2009/002239

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 826/KOLNP/2009

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2007290359

Country of ref document: AU

Ref document number: 575346

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 2007811661

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2007290359

Country of ref document: AU

Date of ref document: 20070904

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 12438995

Country of ref document: US