US20070244113A1 - Compounds and therapeutical use thereof - Google Patents

Compounds and therapeutical use thereof Download PDF

Info

Publication number
US20070244113A1
US20070244113A1 US11/680,843 US68084307A US2007244113A1 US 20070244113 A1 US20070244113 A1 US 20070244113A1 US 68084307 A US68084307 A US 68084307A US 2007244113 A1 US2007244113 A1 US 2007244113A1
Authority
US
United States
Prior art keywords
alkyl
methyl
independently
halo
amine
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/680,843
Other languages
English (en)
Inventor
Sui Cai
Nilantha Sirisoma
Azra Pervin
John Drewe
Shailaja Kasibhatla
Songchun Jiang
Hong Zhang
Chris Pleiman
Vijay Baichwal
John Manfredi
Leena Bhoite
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Myrexis Inc
Cytovia Therapeutics LLC
Original Assignee
Myriad Genetics Inc
Cytovia Inc
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 Myriad Genetics Inc, Cytovia Inc filed Critical Myriad Genetics Inc
Priority to US11/680,843 priority Critical patent/US20070244113A1/en
Publication of US20070244113A1 publication Critical patent/US20070244113A1/en
Assigned to MYRIAD PHARMACEUTICALS, INC. reassignment MYRIAD PHARMACEUTICALS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MYRIAD GENETICS, INC.
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/70Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings condensed with carbocyclic rings or ring systems
    • C07D239/72Quinazolines; Hydrogenated quinazolines
    • C07D239/86Quinazolines; Hydrogenated quinazolines with hetero atoms directly attached in position 4
    • C07D239/94Nitrogen atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • 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
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • 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
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/70Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings condensed with carbocyclic rings or ring systems
    • C07D239/72Quinazolines; Hydrogenated quinazolines
    • C07D239/95Quinazolines; Hydrogenated quinazolines with hetero atoms directly attached in positions 2 and 4
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/12Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D473/00Heterocyclic compounds containing purine ring systems
    • C07D473/26Heterocyclic compounds containing purine ring systems with an oxygen, sulphur, or nitrogen atom directly attached in position 2 or 6, but not in both
    • C07D473/32Nitrogen atom
    • C07D473/34Nitrogen atom attached in position 6, e.g. adenine
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Definitions

  • This invention is in the field of medicinal chemistry.
  • the invention relates to compounds that are activators of caspases and inducers of apoptosis.
  • the invention also relates to the use of these compounds as therapeutically effective anti-cancer agents.
  • Organisms eliminate unwanted cells by a process variously known as regulated cell death, programmed cell death or apoptosis. Such cell death occurs as a normal aspect of animal development, as well as in tissue homeostasis and aging (Glucksmann, A., Biol. Rev. Cambridge Philos. Soc. 26:59-86 (1951); Glucksmann, A., Archives de Biologie 76:419-437 (1965); Ellis, et al., Dev. 112:591-603 (1991); Vaux, et al., Cell 76:777-779 (1994)).
  • Apoptosis regulates cell number, facilitates morphogenesis, removes harmful or otherwise abnormal cells and eliminates cells that have already performed their function. Additionally, apoptosis occurs in response to various physiological stresses, such as hypoxia or ischemia (PCT published application WO96/20721).
  • Apoptosis is achieved through an endogenous mechanism of cellular suicide (Wyllie, A. H., in Cell Death in Biology and Pathology , Bowen and Lockshin, eds., Chapman and Hall (1981), pp. 9-34).
  • a cell activates its internally encoded suicide program as a result of either internal or external signals.
  • the suicide program is executed through the activation of a carefully regulated genetic program (Wyllie, et al., Int. Rev. Cyt. 68:251 (1980); Ellis, et al., Ann. Rev. Cell Bio. 7:663 (1991)).
  • Apoptotic cells and bodies are usually recognized and cleared by neighboring cells or macrophages before lysis. Because of this clearance mechanism, inflammation is not induced despite the clearance of great numbers of cells (Orrenius, S., J. Internal Medicine 237:529-536 (1995)).
  • apoptotic cell death involves at least 14 genes, 2 of which are the pro-apoptotic (death-promoting) ced (for cell death abnormal) genes, ced-3 and ced-4.
  • CED-3 is homologous to interleukin 1 beta-converting enzyme, a cysteine protease, which is now called caspase-1.
  • caspase family of cysteine proteases comprises 14 different members, and more may be discovered in the future. All known caspases are synthesized as zymogens that require cleavage at an aspartyl residue prior to forming the active enzyme. Thus, caspases are capable of activating other caspases, in the manner of an amplifying cascade.
  • Apoptosis and caspases are thought to be crucial in the development of cancer ( Apoptosis and Cancer Chemotherapy , Hickman and Dive, eds., Humana Press (1999)).
  • cancer cells while containing caspases, lack parts of the molecular machinery that activates the caspase cascade. This makes the cancer cells lose their capacity to undergo cellular suicide and the cells become cancerous.
  • Control points are known to exist that represent points for intervention leading to activation.
  • CED-9-BCL-like and CED-3-ICE-like gene family products are intrinsic proteins regulating the decision of a cell to survive or die and executing part of the cell death process itself, respectively (see, Schmitt, et al., Biochem. Cell. Biol. 75:301-314 (1997)).
  • BCL-like proteins include BCL-xL and BAX-alpha, which appear to function upstream of caspase activation.
  • BCL-xL appears to prevent activation of the apoptotic protease cascade, whereas BAX-alpha accelerates activation of the apoptotic protease cascade.
  • chemotherapeutic drugs can trigger cancer cells to undergo suicide by activating the dormant caspase cascade. This may be a crucial aspect of the mode of action of most, if not all, known anticancer drugs (Los, et al., Blood 90:3118-3129 (1997); Friesen, et al., Nat. Med. 2:574 (1996)).
  • the mechanism of action of current antineoplastic drugs frequently involves an attack at specific phases of the cell cycle.
  • the cell cycle refers to the stages through which cells normally progress during their lifetime. Normally, cells exist in a resting phase termed Go. During multiplication, cells progress to a stage in which DNA synthesis occurs, termed S.
  • M phase specific antineoplastic drugs such as vincristine, vinblastine, and paclitaxel
  • S phase specific antineoplastic drugs such as vincristine, vinblastine, and paclitaxel
  • M phase specific antineoplastic drugs such as vinblastine and paclitaxel
  • EP520722 discloses derivatives of 4-anilino-quinazolines as inhibitors of the EGFR tyrosine kinase with antitumor activity: wherein, for example, R a is hydrogen, trifluoromethyl, or nitro, n is 1; and R b is halogen, trifluoromethyl or nitro.
  • EP602851 discloses quinazolines as inhibitors of the EGFR tyrosine kinase: wherein, for example R a is hydroxy, amino, ureido, or trifluoromethoxy, m is 1, 2 or 3; Q is a 9 or 10-membered bicyclic heterocyclic moiety.
  • EP635498 discloses 4-anilino-quinazolines as inhibitors of the EGFR tyrosine kinase: wherein, for example R 1 includes hydroxy, amino or C 1-4 alkoxy, R 2 is hydrogen, hydroxy, or halogen, R 3 is halogen, n is 1, 2 or 3.
  • EP635507 discloses tricyclic derivatives as inhibitors of the EGFR tyrosine kinase: wherein, R 1 and R 2 together form an optionally substituted 5 or 6 membered ring containing at least one heteroatom; R 3 includes hydrogen, hydroxy, or halogen, m is 1, 2 or 3.
  • WO9609294 discloses substituted heteroaromatic compounds as inhibitors of protein tyrosine kinase: wherein, for example X is N or CH; Y is O, S, or NR a wherein R a is H or C 1-8 alkyl; R 1 , R 2 , R 3 and R 3 includes amino, hydrogen, hydroxy, or halogen; R 4 includes amino, hydrogen, hydroxy, or halogen; n is 1, 2 or 3; R 5 is selected from the group comprising hydrogen, halogen, trifluoromethyl, C 1-4 alkyl and C 1-4 alkoxy; R 6 is a group ZR 7 wherein Z includes O, S or NH and R 7 is an optionally substituted C 3-6 cycloalkyl, or an optionally substituted 5,6,7,8,9,10-membered carbocyclic or heterocyclic moiety.
  • WO9713771 discloses substituted heteroaromatic compounds as inhibitors of protein tyrosine kinase: wherein, for example X is N or CH; U represents a fused 5,6,7-membered heterocyclic ring; Y is O, S, or NR a wherein R a is H or C 1-8 alkyl; R 1 included 5,6-membered heterocyclic ring, or amino, hydrogen, hydroxy, or halogen; n is 0, 1, 2 or 3.
  • R 2 is selected from the group comprising hydrogen, halogen, trifluoromethyl, C 1-4 alkyl and C 1-4 alkoxy;
  • R 3 is a group ZR 4 wherein Z includes O, S or NH and R 4 is an optionally substituted C 3-6 cycloalkyl, or an optionally substituted 5,6,7,8,9,10-membered carbocyclic or heterocyclic moiety.
  • R 5 includes hydrogen, hydroxy, or halogen; n is 1, 2 or 3.
  • WO9802438 discloses bicyclic heteroaromatic compounds as inhibitors of protein tyrosine kinase: wherein, for example X is N or CH; Y is O, S, or NR a wherein R a is H or C 1-8 alkyl; R 10 represents a phenyl group or a 5- or 6-membered heterocyclic ring, or amino, hydrogen, hydroxy, or halogen; n is 0 or 1. R 1 includes amino, hydrogen, hydroxy, or halogen; p is 0 to 3.
  • R 2 is selected from the group comprising hydrogen, halogen, trifluoromethyl, C 1-4 alkyl and C 1-4 alkoxy;
  • U represents a 5 to 10-membered mono or bicyclic ring system;
  • A represents a fused 5, 6, or 7-membered heterocyclic ring.
  • the present invention is related to the discovery that 4-arylamino-quinazolines and analogs, as represented in Formula I-VIb below, are potent tubulin inhibitors and active in inhibiting topoisomerase, particularly topoisomerase II. They are activators of the caspase cascade leading to the activation of caspase-3 and inducers or promoters of apoptosis. Thus, they are useful in treating or delaying the onset of diseases and disorders that are responsive to the inhibition of tubulin or topoisomerase, or to the induction of apoptosis.
  • one aspect of the present invention is directed to the use of compounds of the present invention in inhibiting tubulin, in inducing capase activities, particularly caspase-3 activities, in inhibiting topoisomerase I or II, and inducing or promoting apoptosis, by administering the compounds to cells in vitro or in vivo in warm-blood animals, particularly mammals.
  • Another aspect of the present invention is to provide a method for treating or delaying the onset of diseases and disorders that are responsive to inhibition of tubulin or topoisomerase II, including but not limited to neoplastic diseases (such as cancer), psoriasis, autoimmune diseases, and fungi infection.
  • the method comprises administering to a subject mammal in need of the treatment a therapeutically effective amount of a compound of the present invention.
  • Another aspect of the present invention is to provide novel compounds, and to also provide for the use of these novel compounds for treating, preventing or ameliorating neoplasia and cancer.
  • Yet another aspect of the present invention is to provide a pharmaceutical composition useful for treating disorders responsive to the inhibition of tubulin or topoisomerase II, and the induction of apoptosis, containing an effective amount of a compound of the present invention, preferably in admixture with one or more pharmaceutically acceptable carriers or diluents.
  • FIG. 1 depicts the results of a Topoisomerase II activity assay testing Example 1 compound
  • FIG. 2 shows the results of a Topoisomere I activity assay testing Example 1 compound
  • FIG. 3 depicts the binding of radiolabeled Example 102 compound to Topoisomerase II.
  • compounds of the present invention are potent inhibitors of tubulin. It is also discovered that the compounds can also inhibit topoisomerase activities, such as topoisomerase II-dependent conversion of supercoiled DNA to topoisomers.
  • the compounds are potent and highly efficacious activators of the caspase cascade particularly caspase-3, and inducers of apoptosis. Therefore, the compounds are useful for treating diseases and disorders responsive to induction of apoptosis, inhibition of tubulin and/or inhibition of topoisomerase II.
  • the present invention provides a method of inhibiting tubulin in cells in vitro or in warm-blood animals, particularly mammals, more particularly humans.
  • the term “inhibiting tubulin” means inhibiting the polymerization (or assembly) of tubulin monomers or promoting depolymerization of microtubules (i.e., tubulin disassembly). Inhibition of tubulin can be assayed, e.g., by the method described in Example 145 below.
  • the present invention also provides a method for inhibiting topoisomerase II in cells in vitro or in warm-blood animals, particularly mammals, more particularly humans.
  • the term “inhibiting topoisomerase II” means inhibiting the activities of the enzyme topoisomerase II in topoisomerase II-dependent conversion of supercoiled DNA to topoisomers. Inhibition of topoisomerase II activities can be assayed by, e.g., a method described in Example 151.
  • the present invention also provides a method of activating caspase, particularly caspase-3 and inducing apoptosis in cells in vitro or in warm-blood animals, particularly mammals, more particularly humans.
  • activating caspase means activating or enhancing the enzymatic (protease) activity of a caspase (e.g., caspase-3), which, if occurring inside cells, results in promoted apoptosis or cell death.
  • a caspase e.g., caspase-3
  • the ability of a compound in activating caspase, particularly caspase-3, can be assayed in a method as provided in Example 143 below.
  • inducing apoptosis means inducing apoptosis in cells so as to cause cell death. The ability of a compound to induce apoptosis can be tested in a method as described in Example 147 below.
  • the above various methods of the present invention can be practiced by or comprise treating cells in vitro or a warm-blood animal, particularly mammal, more particularly a human with an effective amount of a compound according to the present invention.
  • the phrase “treating . . . with . . . a compound” means either administering the compound to cells or an animal, or administering to cells or an animal the compound or another agent to cause the presence or formation of the compound inside the cells or the animal.
  • the methods of the present invention comprise administering to cells in vitro or to a warm-blood animal, particularly mammal, more particularly a human a pharmaceutical composition comprising an effective amount of a compound according to the present invention.
  • the methods of the present invention comprise treating cells in vitro or a warm-blood animal, particularly mammal, more particularly a human with an effective amount of a compound according to Formula I: or pharmaceutically acceptable salts or solvates thereof, wherein: Ar is aryl or heteroaryl; each of which is optionally substituted by one or more substituents wherein each substituent is independently halo, hydroxy, hydroxy-C 1-6 alkyl-, C 1-6 alkyl-C(O)O—, amino, nitro, cyano, C 1-6 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, C 1-6 acylamino, C 1-6 acyloxy, C 1-6 alkoxy, or C 1-6 alkylthiol-; R 1 is C 1-6 alkyl, preferably methyl or ethyl, more preferably methyl; A is an aromatic, heteroaromatic, heterocyclic, or carbocyclic ring; each of which is optionally substituted by one or more
  • Preferred compounds of Formula I include compounds wherein D is nitrogen, and
  • R 5 is hydrogen or flourine, preferably hydrogen.
  • ring A is benzo or fused cyclohexyl.
  • Another group of preferred compounds include compounds wherein Ar is phenyl, naphthyl, pyridyl, pyridazyl, pyrimidyl, pyrazyl, quinolyl, isoquinolyl, isoxazolyl, pyrazolyl, imidazolyl, thienyl, furyl or pyrrolyl; each of which is optionally substituted by one or more substituents wherein each substituent is independently halo, hydroxy, hydroxyC 1-6 alkyl-, C 1-6 alkyl-C(O)O—, amino, nitro, cyano, C 1 -C 6 alkyl, C 2 -C 4 alkenyl, C 2 -C 4 alkynyl, C 1 -C 6 acylamino, C 1 -C 6 acyloxy, C 1 -C 6 alkoxy, or C 1-6 alkylthi
  • Ar is phenyl, pyridyl, pyridazyl, pyrimidyl or pyrazyl, each of which is optionally substituted by one or more substituents wherein each substituent is as defined immediately above.
  • R 2 is H, halo, or a member of the group consisting of N 3 , C 1-4 alkoxy, C 1-4 alkylthiol, hydroxyC 1-4 alkyl, C 1-4 alkyl and —N(R a )(R b ) wherein R a and R b are independently H, OH (R a and R b are not both OH), C 2-4 hydroxyalkyl, or C 1-4 alkyl or R a and R b together with the nitrogen atom to which they are both linked form a 3, 4, 5 or 6-membered heterocycle; each of the member is optionally substituted by 1-4 substituents wherein each substituent is independently halo, OH, or C 1-4 alkyl.
  • one or two of Q, T, U and V are nitrogen, and both B and D are nitrogen.
  • R 1 is C 1-4 alkyl, preferably methyl or ethyl, more preferably methyl
  • R 2 -R 11 are independently H, halo, N 3 , OH, thiol, nitro, CN, NH 2 , C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 1-6 alkoxy, C 1-6 alkylthiol, halo-C 1-6 alkyl, C 2-6 alkenyl-O—, C 2-6 alkynyl-O—, hydroxy-C 1-6 alkyl, C 1-6 alkoxy-C 1-6 alkyl, C 1-6 acyl, C 1-6 acyloxy, —C 1-6 alkyl-C(O)O—C 1-6 alkyl, —C(O)O—C 1-6 alkyl, C 1-6 alkyl, C 1-6 al
  • R a and R b are not both OH
  • R 5 is H or F, more preferably H.
  • R 2 is R 2 is H, halo, or a member of the group consisting of: N 3 , C 1-4 alkoxy, C 1-4 alkylthiol, hydroxy-C 1-4 alkyl, C 1-4 alkyl and —N(R a )(R b ) wherein R a and R b are independently H, OH (R a and R b are not both OH), C 2-4 hydroxyalkyl, or C 1-4 alkyl or R a and R b together with the nitrogen atom to which they are both linked form a 3, 4, 5 or 6-membered heterocycle (e.g., morpholino); each of the member is optionally substituted by 1-4 substituents wherein each substituent is independently halo, OH, or C 1-4 alkyl.
  • R 1 is C 1-4 alkyl, preferably methyl or ethyl, more preferably methyl
  • R 2 -R 11 are independently H, halo, N 3 , OH, thiol, nitro, CN, NH 2 , C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 1-6 alkoxy, C 1-6 alkylthiol, halo-C 1-6 alkyl, C 2-6 alkenyl-O—, C 2-6 alkynyl-O—, hydroxy-C 1-6 alkyl, C 1-6 alkoxy-C 1-6 alkyl, C 1-6 acyl, C 1-6 acyloxy, —C 1-6 alkyl-C(O)O—C 1-6 alkyl, —C(O)O—C 1-6 alkyl, C 1-6 alkyl
  • R 5 is H or F, more preferably H.
  • R 2 is H, halo, or a member of the group consisting of: N 3 , C 1-4 alkoxy, C 1-4 alkylthiol, hydroxy-C 1-4 alkyl, C 1-4 alkyl and —N(R a )(R b ) wherein R a and R b are independently H, OH (R a and R b are not both OH), C 2-4 hydroxyalkyl, or C 1-4 alkyl or R a and R b together with the nitrogen atom to which they are both linked form a 3, 4, 5 or 6-membered heterocycle (e.g., morpholino); each of the member is optionally substituted by 1-4 substituents wherein each substituent is independently halo, OH, or C 1-4 alkyl.
  • Ar is phenyl, naphthyl, pyridyl, pyridazyl, pyrimidyl, pyrazyl, quinolyl, isoquinolyl, isoxazolyl, pyrazolyl, imidazolyl, thienyl, furyl or pyrrolyl; each of which is optionally substituted by one or more substituents wherein each substituent is as defined immediately above.
  • Ar is phenyl, pyridyl or pyridazyl, pyrimidyl, pyrazyl, each of which is optionally substituted by one or more substituents wherein each substituent is as defined immediately above.
  • R 2 is a member of the group consisting of H, halo, N 3 , C 1-4 alkoxy, C 1-4 alkylthiol, hydroxyC 1-4 alkyl, C 1-4 alkyl, and —N(R a )(R b ) wherein R a and R b are independently H, OH (R a and R b are not both OH), C 2-4 hydroxyalkyl, or C 1-4 alkyl or R a and R b together with the nitrogen atom to which they are both linked form a 3, 4, 5 or 6-membered heterocycle; each of the member being optionally substituted by 1-4 substituents wherein each substituent is independently halo, OH, or C 1-4 alkyl.
  • one or two of Q, T, U and V are N, and both B and D are N.
  • Other preferred compounds according to Formula III include compounds wherein Ar is phenyl, naphthyl, pyridyl, pyridazyl, pyrimidyl, pyrazyl, quinolyl, isoquinolyl, isoxazolyl, pyrazolyl, imidazolyl, thienyl, furyl or pyrrolyl; each of which is optionally substituted by one or more substituents wherein each substituent is as defined immediately above. More preferably, Ar is phenyl, pyridyl pyridazyl, pyrimidyl, or pyrazyl, each of which is optionally substituted by one or more substituents wherein each substituent is as defined immediately above.
  • Ar is pyridyl pyridazyl, pyrimidyl, or pyrazyl, each of which is optionally substituted by one or more substituents as defined above for Ar.
  • R 2 is a member of the group consisting of H, halo, N 3 , C 1-4 alkoxy, C 1-4 alkylthiol, hydroxyC 1-4 alkyl, C 1-4 alkyl, and —N(R a )(R b ) wherein R a and R b are independently H, OH (R a and R b are not both OH), C 2-4 hydroxyalkyl, or C 1-4 alkyl or R a and R b together with the nitrogen atom to which they are both linked form a 3, 4, 5 or 6-membered heterocycle; each of the member being optionally substituted by 1-4 substituents wherein each substituent is independently halo, OH, or C 1-4 alkyl.
  • R 2 is a member of the group consisting of
  • a compound according to Formula III is other than (5,6,7,8-tetrahydro-quinazolin-4-yl)-phenyl-ethyl-amine.
  • the methods of inhibiting tubulin, inhibiting topoisomerase II, activating caspase-3, inducing apoptosis and treating or delaying the onset of diseases and disorders responsive to the inhibition of tubulin or topoisomerase II or to the activation of caspase-3 or induction of apoptosis comprise administering an effective amount of a compound or a pharmaceutical composition containing an effective amount of the compound, which compound is represented by any one of Formulae IV, IVa, IVb, V, Va, Vb, Vc, VI, VIa and VIb, and each and all embodiments thereof and salts or solvates thereof, as provided below.
  • Additional compounds useful in such methods particularly the methods of inhibiting tubulin, inhibiting topoisomerase II, activating caspase-3 and inducing apoptosis, and treating diseases and disorders responsive to the inhibition of tubulin or topoisomerase II, or activating caspase-3 and inducing apoptosis include compounds according to Formula IV: and pharmaceutically acceptable salts and solvates thereof, wherein:
  • R 5 is not alkoxy. More preferably, when the A ring is aryl or heteroaryl and U is C, then R 5 is H or F, preferably H.
  • one of W, X, Y and Z is N. In other embodiments of the compounds of Formula IV, two of W, X, Y and Z are N. In any of the embodiments, preferably one or two of Q, T, U and V are N. In preferred embodiments, B and D both are N.
  • one or two of Q, T, U and V are N.
  • both B and D are N.
  • one or two of Q, T, U and V are N.
  • Still further additional compounds useful in such methods include compounds according to Formula VIb: or pharmaceutically acceptable salts or solvates thereof, wherein:
  • the compounds administered in the methods of the invention are able to induce caspase activation as determined by the method and under conditions (measurement at 24 hours) described in Example 143, preferably at an EC 50 no greater than 1,000 nM, more preferably at an EC 50 no greater than about 500 nM, more preferably at an EC 50 no greater than about 200 nM, more preferably at an EC 50 no greater than about 100 nM, even more preferably at an EC 50 no greater than about 50 nM, and most preferably at an EC 50 no greater than about 10 nM.
  • Also preferred in the above methods of the invention are compounds of Formula I-VIb, and pharmaceutically acceptable salts or solvates thereof, that are able to inhibit tubulin at an IC 50 of no greater than about 2,000 nM, more preferably no greater than about 1,000 nM, most preferably less than about 500 nM, as determined by the method and under conditions described in Example 145.
  • Exemplary compounds useful in the methods of the invention include, but are not limited to, compounds in Examples 1-142; and pharmaceutically acceptable salts or solvates thereof, and:
  • the present invention also provides novel compounds, which are potent tubulin inhibitors, topoisomerase II inhibitors, caspase-3 activators and/or apoptosis inducers/promoters.
  • novel compounds of the present invention are represented by Formula IV and pharmaceutically acceptable salts or solvates thereof: wherein
  • R 8 or R 10 or both are independently selected from the group OH; N 3 ; —XR 2a wherein X is S or O and R 2a is C 1-6 alkyl (preferably C 1-3 alkyl, more preferably CH 3 ) optionally substituted with OH or halo; —NH(R 2b ) or N(R 2b )(R 2c ) wherein R 2b and R 2c are independently C 1-6 alkyl (preferably C 1-3 alkyl, more preferably CH 3 ) optionally substituted with OH or halo (preferably F, and wherein optionally R 2b and R 2c may together form a 3-6 membered heterocycle; and —C(O)OR 2d wherein R 2d is C 1-6 alkyl (preferably C 1-3 alkyl); and more preferably R 9 is not H.
  • one of W, X, Y and Z is N. In other embodiments of the compounds of Formula IV, two of W, X, Y and Z are N. In any of the embodiments, preferably one or two of Q, T, U and V are N. In preferred embodiments, B and D both are N.
  • one of X, Y, W and Z is N. In other embodiments, two of X, Y, W and Z are N.
  • D is nitrogen, and more preferably both B and D are N.
  • compounds of the present invention have Formula IVa, or a pharmaceutically acceptable salt or solvate thereof, wherein:
  • only one of W, X, Y and Z is N. In other specific embodiments, two of W, X, Y and Z are N.
  • R 9 is selected from the group:
  • R 9a is methyl, ethyl, fluoromethyl (CH 2 F, CHF 2 , CF 3 ), or fluoroethyl;
  • R 9b is H or C 1-2 alkyl.
  • D is N, and more preferably both B and D are N.
  • R 1 when R 1 is ethyl then at least one of R 8 , R 9 , and R 10 is not H; preferably R 9 is not H.
  • R 8 or R 10 or both are independently selected from the group OH; N 3 ; amido; N-dimethylamido; —XR 9a wherein X is S or O and R 9a is C 1-6 alkyl (preferably C 1-3 alkyl, more preferably CH 3 ) optionally substituted with OH or halo; C 1-3 alkyl optionally substituted with halo (preferably F); —N(R a )(R b ) wherein R a and R b are independently H, OH (R a and R b are not both OH), or C 1-6 alkyl (preferably C 1-3 alkyl, more preferably CH 3 ) optionally substituted with OH or halo (preferably F, and wherein optionally R 2b and R 2c may together form a
  • R 9 is selected from the group:
  • R 9a is methyl, ethyl, fluoromethyl (e.g., CH 2 F, CHF 2 , CF 3 ), fluoroethyl;
  • R 9b is H or C 1-2 alkyl.
  • novel compounds of the present invention are those represented by Formula V: or a pharmaceutically acceptable salt or solvate thereof, wherein:
  • R 9 is H
  • at least one of R 8 and R 10 is not H, more preferably R 9 is other than H.
  • B is C and D is N. In other specific embodiments, B is N and D is C. In preferred embodiments, both B and D are N.
  • R 2 is H; halo; N 3 ;
  • C 1-6 alkyl preferably C 1-3 alkyl, more preferably CH 3 ) optionally substituted with OH or halo (preferably F, e.g., monofluoro, difluoro, or trifluoro);
  • R 2a wherein X is S or O, and R 2a is C 1-6 alkyl (preferably C 1-3 alkyl, more preferably CH 3 ) optionally substituted with OH or halo (preferably F, e.g., monofluoro-, difluoro-, or trifluoro-substituted);
  • R d is C 1-3 alkyl, preferably methyl or ethyl;
  • R a and R b are independently H, OH (R a and R b are not both OH), C 1-3 alkyl (preferably CH 3 ), C 1-6 hydroxyalkyl (preferably C 2-3 hydroxyalkyl, more preferably —CH 2 CH 2 OH), or C 1-6 alkyl (preferably C 1-3 alkyl, more preferably CH 3 ) that is optionally substituted with —N(R e )(R f ) wherein R e and R f are independently H, OH (R e and R f are not both OH), C 1-3 alkyl (preferably CH 3 ), or C 2-3 hydroxyalkyl (preferably —CH 2 CH2OH), and wherein optionally R a and R b together may form a 3, 4, 5 or 6-membered heterocycle (e.g., piperidinyl, pyrrolidinyl, and morpholinyl).
  • R a and R b together may form a 3, 4, 5 or 6-membered heterocycle (e.
  • R 2 is H, methyl, ethyl, Cl, F, fluoromethyl (CH 2 F, CHF 2 , CF 3 ), C 1-3 hydroxyalkyl (preferably CH 2 OH or CH 2 CH 2 OH), NH 2 , NH 2 OH, —NHCH 2 CH 2 OH, NHCH 3 , N(CH 3 ) 2 , N 3 , morpholino, OCH 3 , OC 2 H 5 , or SCH 3 .
  • R 2 is H, methyl, Cl, —CH 2 OH, —NH 2 , —NHCH 3 , —NHCH 2 CH 2 OH, —OCH 3 , —SCH 3 , or —CH 2 F.
  • R 9 is selected from the group consisting of H; OH; Cl; N 3 ; C 1-3 alkyl (preferably methyl or ethyl) or C 1-3 haloalkyl (preferably monofluoromethyl, difluoromethyl, trifluoromethyl); —OR 9a , wherein R 9a is C 1-3 alkyl (i.e., methyl, ethyl, propyl, isopropyl) or C 1-3 haloalkyl (e.g., fluoroalkyl, preferably fluoromethyl, i.e., CH 2 F, CHF 2 , CF 3 ); —N(R a )(R b ) wherein R a and R b are independently H or C 1-3 alkyl; or —COOR 9b , wherein R 9b is C 1-3 alkyl (preferably methyl or ethyl); and optionally R 9 and one of R 8 and R 10 together form a 3, 4, 5, or 6-
  • R 9 is —OCH 3 , —OC 2 H 5 , —N(CH 3 ) 2 , —CO 2 CH 3 , —OCHF 2 , or N 3 .
  • R 9 when R 9 is H, at least one of R 8 and R 10 is not H. In another embodiment, when R 9 is alkyl, R 2 is not H.
  • compounds of the invention include compounds of Formula V or pharmaceutically acceptable salts or solvates thereof, wherein:
  • C 1-6 alkyl preferably C 1-3 alkyl, more preferably CH 3 ) optionally substituted with OH or halo (preferably F, e.g., monofluoro, difluoro, or trifluoro);
  • R 2a wherein X is S or O, and R 2a is C 1-6 alkyl (preferably C 1-3 alkyl, more preferably CH 3 ) optionally substituted with OH or halo (preferably F, e.g., monofluoro-, difluoro-, or trifluoro-substituted);
  • R d is C 1-3 alkyl, preferably methyl or ethyl;
  • R a and R b are independently H, OH (R a and R b are not both OH), C 1-3 alkyl (preferably CH 3 ), C 1-6 hydroxyalkyl (preferably C 2-3 hydroxyalkyl, more preferably —CH 2 CH 2 OH), or C 1-6 alkyl (preferably C 1-3 alkyl, more preferably CH 3 ) that is optionally substituted with —N(R e )(R f ) wherein R e and R f are independently H, OH (R e and R f are not both OH), C 1-3 alkyl (preferably CH 3 ), or C 2-3 hydroxyalkyl (preferably —CH 2 CH2OH), and wherein optionally R a and R b with N together may form a 3, 4, 5 or 6-membered heterocycle (e.g., piperidinyl, pyrrolidinyl, and morpholinyl);
  • compounds of the invention include compounds of Formula V or pharmaceutically acceptable salts or solvates thereof, wherein:
  • compounds of the invention include compounds of Formula V or pharmaceutically acceptable salts or solvates thereof, wherein R 1 is methyl; R 2 is H, methyl, Cl, —CH 2 OH, —NH 2 , —NHCH 3 , —NHCH 2 CH 2 OH, —OCH 3 , —SCH 3 , or —CH 2 F; R 3 and R 12 are independently H, methyl, —OCH 3 , or Cl; R 4 is H, methyl, or NH 2 ; R 5 is H; R 6 and R 13 are independently H or methyl; R 7 and R 11 are independently H or F; R 8 and R 10 are independently H, or F or OCH 3 ; and R 9 is —OCH 3 , —OC 2 H 5 , —N(CH 3 ) 2 , —CO 2 CH 3 , —OCHF 2 , or N 3 ; and B, D, Q, T, U, V, W, X, Y, and Z are as defined above
  • one of W, X, Y and Z is N, or two of W, X, Y and Z are N.
  • one or two of Q, T, U and V are N.
  • Q and V can be both N and T and U are C.
  • R 9 is H
  • at least one of R 8 and R 10 is not H, more preferably R 9 is other than H.
  • B is C and D is N. In other specific embodiments,
  • B is N and D is C. In preferred embodiments, both B and D are N.
  • C 1-6 alkyl preferably C 1-3 alkyl, more preferably CH 3 ) optionally substituted with OH or halo (preferably F, e.g., monofluoro, difluoro, or trifluoro);
  • R 2a wherein X is S or O, and R 2a is C 1-6 alkyl (preferably C 1-3 alkyl, more preferably CH 3 ) optionally substituted with OH or halo (preferably F, e.g., monofluoro-, difluoro-, or trifluoro-substituted);
  • R d is C 1-3 alkyl, preferably methyl or ethyl;
  • R a and R b are independently H, OH (R a and R b are not both OH), C 1-3 alkyl (preferably CH 3 ), C 1-6 hydroxyalkyl (preferably C 2-3 hydroxyalkyl, more preferably —CH 2 CH 2 OH), or C 1-6 alkyl (preferably C 1-3 alkyl, more preferably CH 3 ) that is optionally substituted with —N(R e )(R f ) wherein R e and R f are independently H, OH (R e and R f are not both OH), C 1-3 alkyl (preferably CH 3 ), or C 2-3 hydroxyalkyl (preferably —CH 2 CH2OH), and wherein optionally R a and R b together may form a 3, 4, 5 or 6-membered heterocycle (e.g., piperidinyl, pyrrolidinyl, and morpholinyl).
  • R a and R b together may form a 3, 4, 5 or 6-membered heterocycle (e.
  • R 2 is H, methyl, ethyl, Cl, F, fluoromethyl (CH 2 F, CHF 2 , CF 3 ), C 1-3 hydroxyalkyl (preferably CH 2 OH or CH 2 CH 2 OH), NH 2 , NH 2 OH, —NHCH 2 CH 2 OH, NHCH 3 , N(CH 3 ) 2 , N 3 , morpholino, OCH 3 , OC 2 H 5 , or SCH 3 .
  • R 2 is H, methyl, Cl, —CH 2 OH, —NH 2 , —NHCH 3 , —NHCH 2 CH 2 OH, —OCH 3 , —SCH 3 , or —CH 2 F.
  • R 9 is selected from the group consisting of H; OH; Cl; N 3 ; C 1-3 alkyl (preferably methyl or ethyl) or C 1-3 haloalkyl (preferably monofluoromethyl, difluoromethyl, trifluoromethyl); —OR 9a , wherein R 9a is C 1-3 alkyl (i.e., methyl, ethyl, propyl, isopropyl) or C 1-3 haloalkyl (e.g., fluoroalkyl, preferably fluoromethyl, i.e., CH 2 F, CHF 2 , CF 3 ); —N(R a )(R b ) wherein R a and R b are independently H or C 1-3 alkyl; or —COOR 9b , wherein R 9b is C 1-3 alkyl (preferably methyl or ethyl); and optionally R 9 and one of R 8 and R 10 together form a 3, 4, 5, or 6-
  • R 9 is —OCH 3 , —OC 2 H 5 , —N(CH 3 ) 2 , —CO 2 CH 3 , —OCHF 2 , or N 3 .
  • R 9 is alkyl
  • R 2 is not H
  • compounds of the invention include compounds of Formula Va or pharmaceutically acceptable salts or solvates thereof, wherein:
  • C 1-6 alkyl preferably C 1-3 alkyl, more preferably CH 3 ) optionally substituted with OH or halo (preferably F, e.g., monofluoro, difluoro, or trifluoro);
  • R 2a wherein X is S or O, and R 2a is C 1-6 alkyl (preferably C 1-3 alkyl, more preferably CH 3 ) optionally substituted with OH or halo (preferably F, e.g., monofluoro-, difluoro-, or trifluoro-substituted);
  • R d is C 1-3 alkyl, preferably methyl or ethyl;
  • R a and R b are independently H, OH (R a and R b are not both OH), C 1-3 alkyl (preferably CH 3 ), C 1-6 hydroxyalkyl (preferably C 2-3 hydroxyalkyl, more preferably —CH 2 CH 2 OH), or C 1-6 alkyl (preferably C 1-3 alkyl, more preferably CH 3 ) that is optionally substituted with —N(R e )(R f ) wherein R e and R f are independently H, OH (R e and R f are not both OH), C 1-3 alkyl (preferably CH 3 ), or C 2-3 hydroxyalkyl (preferably —CH 2 CH2OH), and wherein optionally R a and R b together may form a 3, 4, 5 or 6-membered heterocycle (e.g., piperidinyl, pyrrolidinyl, and morpholinyl);
  • compounds of the invention include compounds of Formula Va or pharmaceutically acceptable salts or solvates thereof, wherein:
  • compounds of the invention include compounds of Formula Va or pharmaceutically acceptable salts or solvates thereof, wherein R 1 is methyl; R 2 is H, methyl, Cl, —CH 2 OH, —NH 2 , —NHCH 3 , —NHCH 2 CH 2 OH, —OCH 3 , —SCH 3 , or —CH 2 F; R 3 and R 12 are independently H, methyl, —OCH 3 , or Cl; R 4 is H, methyl, or NH 2 ; R 5 is H; R 6 and R 13 are independently H or methyl; R 7 and R 11 are independently H or F; R 8 and R 10 are independently H, or F or OCH 3 ; and R 9 is —OCH 3 , —OC 2 H 5 , —N(CH 3 ) 2 , —CO 2 CH 3 , —OCHF 2 , or N 3 ; and B, D, W, X, Y, and Z are as defined above, provided that when B, D,
  • one of W, X, Y and Z is N, or two of W, X, Y and Z are N.
  • Another group of preferred compounds of the present invention are those represented by Formula V with the proviso that both B and D are nitrogen.
  • Specifically such compounds are represented by Formula Vb: or pharmaceutically acceptable salts or solvates thereof, wherein:
  • R 9 is H
  • at least one of R 8 and R 10 is not H, more preferably R 9 is other than H.
  • R 2 is H; halo; N 3 ;
  • C 1-6 alkyl preferably C 1-3 alkyl, more preferably CH 3 ) optionally substituted with OH or halo (preferably F, e.g., monofluoro, difluoro, or trifluoro);
  • R 2a wherein X is S or O, and R 2a is C 1-6 alkyl (preferably C 1-3 alkyl, more preferably CH 3 ) optionally substituted with OH or halo (preferably F, e.g., monofluoro-, difluoro-, or trifluoro-substituted);
  • R d is C 1-3 alkyl, preferably methyl or ethyl
  • R a and R b are independently H, OH (R a and R b are not both OH), C 1-3 alkyl (preferably CH 3 ), C 1-6 hydroxyalkyl (preferably C 2-3 hydroxyalkyl, more preferably —CH 2 CH 2 OH), or C 1-6 alkyl (preferably C 1-3 alkyl, more preferably CH 3 ) that is optionally substituted with —N(R e )(R f ) wherein R e and R f are independently H, OH (R e and R f are not both OH), C 1-3 alkyl (preferably CH 3 ), or C 2-3 hydroxyalkyl (preferably —CH 2 CH2OH), and wherein optionally R a and R b together may form a 3, 4, 5 or 6-membered heterocycle (e.g., piperidinyl, pyrrolidinyl, and morpholinyl).
  • R a and R b together may form a 3, 4, 5 or 6-membered heterocycle (e.
  • R 2 is H, methyl, ethyl, Cl, F, fluoromethyl (CH 2 F, CHF 2 , CF 3 ), C 1-3 hydroxyalkyl (preferably CH 2 OH or CH 2 CH 2 OH), NH 2 , NH 2 OH, —NHCH 2 CH 2 OH, NHCH 3 , N(CH 3 ) 2 , N 3 , morpholino, OCH 3 , OC 2 H 5 , or SCH 3 .
  • R 2 is H, methyl, Cl, —CH 2 OH, —NH 2 , —NHCH 3 , —NHCH 2 CH 2 OH, —OCH 3 , —SCH 3 , or —CH 2 F.
  • R 9 is selected from the group consisting of H; OH; Cl; N 3 ; C 1-3 alkyl (preferably methyl or ethyl) or C 1-3 haloalkyl (preferably monofluoromethyl, difluoromethyl, trifluoromethyl); —OR 9a , wherein R 9a is C 1-3 alkyl (i.e., methyl, ethyl, propyl, isopropyl) or C 1-3 haloalkyl (e.g., fluoroalkyl, preferably fluoromethyl, i.e., CH 2 F, CHF 2 , CF 3 ); —N(R a )(R b ) wherein R a and R b are independently H or C 1-3 alkyl; or —COOR 9b , wherein R 9b is C 1-3 alkyl (preferably methyl or ethyl); and optionally R 9 and one of R 8 and R 10 together form a 3, 4, 5, or 6-
  • R 9 is —OCH 3 , —OC 2 H 5 , —N(CH 3 ) 2 , —CO 2 CH 3 , —OCHF 2 , or N 3 .
  • R 9 is alkyl
  • R 2 is not H
  • compounds of the invention include compounds of Formula Vb or pharmaceutically acceptable salts or solvates thereof, wherein:
  • C 1-6 alkyl preferably C 1-3 alkyl, more preferably CH 3 ) optionally substituted with OH or halo (preferably F, e.g., monofluoro, difluoro, or trifluoro);
  • R 2a wherein X is S or O, and R 2a is C 1-6 alkyl (preferably C 1-3 alkyl, more preferably CH 3 ) optionally substituted with OH or halo (preferably F, e.g., monofluoro-, difluoro-, or trifluoro-substituted);
  • R d is C 1-3 alkyl, preferably methyl or ethyl;
  • R a and R b are independently H, OH (R a and R b are not both OH), C 1-3 alkyl (preferably CH 3 ), C 1-6 hydroxyalkyl (preferably C 2-3 hydroxyalkyl, more preferably —CH 2 CH 2 OH), or C 1-6 alkyl (preferably C 1-3 alkyl, more preferably CH 3 ) that is optionally substituted with —N(R e )(R f ) wherein R e and R f are independently H, OH (R e and R f are not both OH), C 1-3 alkyl (preferably CH 3 ), or C 2-3 hydroxyalkyl (preferably —CH 2 CH2OH), and wherein optionally R a and R b together may form a 3, 4, 5 or 6-membered heterocycle (e.g., piperidinyl, pyrrolidinyl, and morpholinyl);
  • compounds of the invention include compounds of Formula Vb or pharmaceutically acceptable salts or solvates thereof, wherein:
  • compounds of the invention include compounds of Formula Vb or pharmaceutically acceptable salts or solvates thereof, wherein R 1 is methyl; R 2 is H, methyl, Cl, —CH 2 OH, —NH 2 , —NHCH 3 , —NHCH 2 CH 2 OH, —OCH 3 , —SCH 3 , or —CH 2 F; R 3 is H, methyl, —OCH 3 , or Cl; R 4 is H, methyl, or NH 2 ; R 5 is H; R 6 is H or methyl; R 7 and R 11 are independently H or F; R 8 and R 10 are independently H, or F or OCH 3 ; and R 9 is —OCH 3 , —OC 2 H 5 , —N(CH 3 ) 2 , —CO 2 CH 3 , —OCHF 2 , or N 3 ; and Q, T, U, V, W, X, Y, and Z are as defined above, provided that when Q, T, U, V, W, X,
  • one of W, X, Y and Z is N, or two of W, X, Y and Z are N.
  • one or two of Q, T, U and V are N.
  • Q and V can be both N, and T and U are C.
  • R 9 is H
  • at least one of R 8 and R 10 is not H, more preferably R 9 is other than H.
  • R 2 is H; halo; N 3 ;
  • C 1-6 alkyl preferably C 1-3 alkyl, more preferably CH 3 ) optionally substituted with OH or halo (preferably F, e.g., monofluoro, difluoro, or trifluoro);
  • R 2a wherein X is S or O, and R 2a is C 1-6 alkyl (preferably C 1-3 alkyl, more preferably CH 3 ) optionally substituted with OH or halo (preferably F, e.g., monofluoro-, difluoro-, or trifluoro-substituted);
  • R d is C 1-3 alkyl, preferably methyl or ethyl;
  • R a and R b are independently H, OH (R a and R b are not both OH), C 1-3 alkyl (preferably CH 3 ), C 1-6 hydroxyalkyl (preferably C 2-3 hydroxyalkyl, more preferably —CH 2 CH 2 OH), or C 1-6 alkyl (preferably C 1-3 alkyl, more preferably CH 3 ) that is optionally substituted with —N(R e )(R f ) wherein R e and R f are independently H, OH (R e and R f are not both OH), C 1-3 alkyl (preferably CH 3 ), or C 2-3 hydroxyalkyl (preferably —CH 2 CH2OH), and wherein optionally R a and R b together may form a 3, 4, 5 or 6-membered heterocycle (e.g., piperidinyl, pyrrolidinyl, and morpholinyl).
  • R a and R b together may form a 3, 4, 5 or 6-membered heterocycle (e.
  • R 2 is H, methyl, ethyl, Cl, F, fluoromethyl (CH 2 F, CHF 2 , CF 3 ), C 1-3 hydroxyalkyl (preferably CH 2 OH or CH 2 CH 2 OH), NH 2 , NH 2 OH, —NHCH 2 CH 2 OH, NHCH 3 , N(CH 3 ) 2 , N 3 , morpholino, OCH 3 , OC 2 H 5 , or SCH 3 .
  • R 2 is H, methyl, Cl, —CH 2 OH, —NH 2 , —NHCH 3 , —NHCH 2 CH 2 OH, —OCH 3 , —SCH 3 , or —CH 2 F.
  • R 9 is selected from the group consisting of H; OH; Cl; N 3 ; C 1-3 alkyl (preferably methyl or ethyl) or C 1-3 haloalkyl (preferably monofluoromethyl, difluoromethyl, trifluoromethyl); —OR 9a , wherein R 9a is C 1-3 alkyl (i.e., methyl, ethyl, propyl, isopropyl) or C 1-3 haloalkyl (e.g., fluoroalkyl, preferably fluoromethyl, i.e., CH 2 F, CHF 2 , CF 3 ); —N(R a )(R b ) wherein R a and R b are independently H or C 1-3 alkyl; or —COOR 9b , wherein R 9b is C 1-3 alkyl (preferably methyl or ethyl); and optionally R 9 and one of R 8 and R 10 together form a 3, 4, 5, or 6-
  • R 9 is —OCH 3 , —OC 2 H 5 , —N(CH 3 ) 2 , —CO 2 CH 3 , —OCHF 2 , or N 3 .
  • R 9 when R 9 is H, then at least one of R 8 and R 10 is not H. In another embodiment, when R 9 is alkyl, then R 2 is not H.
  • compounds of the invention include compounds of Formula Vc or pharmaceutically acceptable salts or solvates thereof, wherein:
  • compounds of the invention include compounds of Formula Vc or pharmaceutically acceptable salts or solvates thereof, wherein:
  • compounds of the invention include compounds of Formula Vc or pharmaceutically acceptable salts or solvates thereof, wherein R 1 is methyl; R 2 is H, methyl, Cl, —CH 2 OH, —NH 2 , —NHCH 3 , —NHCH 2 CH 2 OH, —OCH 3 , —SCH 3 , or —CH 2 F; R 3 and R 12 are independently H, methyl, —OCH 3 , or Cl; R 4 is H, methyl, or NH 2 ; R 5 is H; R 6 and R 13 are independently H or methyl; R 7 and R 11 are independently H or F; R 8 and R 10 are independently H, or F or OCH 3 ; and R 9 is —OCH 3 , —OC 2 H 5 , —N(CH 3 ) 2 , —CO 2 CH 3 , —OCHF 2 , or N 3 ; and W, X, Y, and Z are as defined above, provided that when W, X, Y, and Z are as defined above
  • one of W, X, Y and Z is N, or two of W, X, Y and Z are N.
  • compounds of the invention include compounds of Formula VI or pharmaceutically acceptable salts or solvates thereof, wherein:
  • R 9 is selected from the group consisting of H, OH, C 1 , N 3 ;
  • C 1-6 alkyl (preferably C 1-3 alkyl, more preferably CH 3 ) optionally substituted with 1, 2 or 3 substituents, each substituent being independently OH, halo, C 1-3 alkoxy, (halo)C 1-3 alkoxy, —N(R a )(R b ) where R a and R b are independently H, OH (R a and R b are not both OH), C 2-4 hydroxyalkyl, or C 1-3 alkyl or R a and R b together with the nitrogen atom to which they are both linked to form a 3, 4, 5 or 6-membered heterocycle;
  • R c is C 1-6 alkyl (preferably C 1-3 alkyl, more preferably CH 3 ) optionally substituted with 1, 2 or 3 substituents, each substituent being independently OH, halo, C 1-3 alkoxy, or (halo)C 1-3 alkoxy;
  • R d is an C 1-6 alkyl (preferably C 1-3 alkyl, more preferably methyl or ethyl) optionally substituted with 1, 2 or 3 substituents, each substituent being independently OH, halo, C 1-3 alkoxy (e.g., fluoroalkoxy), —N(R a )(R b ) where R a and R b are independently H, OH (R a and R b are not both OH), C 2-4 hydroxyalkyl, or C 1-3 alkyl or R a and R b together with the nitrogen atom to which they both are linked form a 3, 4, 5 or 6-membered heterocycle;
  • R a and R b are independently H, OH (R a and R b are not both OH), C 2-4 hydroxyalkyl, C 1-3 alkyl, or C 1-3 alkyl substituted with —N(R e )(R f ) where R e and R f are independently H, OH (R e and R f are not both OH), or C 1-3 alkyl; wherein optionally R a and R b together with the N form a 3, 4, 5 or 6-membered heterocycle, and optionally R e and R f together with the nitrogen atom to which they both are linked form a 3, 4, 5 or 6-membered heterocycle; or
  • R a and R b are independently H or C 1-3 alkyl; and optionally R 9 and one of R 8 and R 10 together form a 3, 4, 5, or 6-membered heterocycle; preferably R 9 is selected from the group outlined above except R 9 is not H and C 1-6 alkyl.
  • R 9 is H; OH; Cl; N 3 ; C 1-3 alkyl (preferably methyl; C 1-3 haloalkyl (preferably monofluoromethyl, difluoromethyl, trifluoromethyl); —OR 9a , wherein R 9a is C 1-4 alkyl or C 1-3 haloalkyl (e.g., fluoroalkyl, preferably fluoromethyl, i.e., CH 2 F, CHF 2 , CF 3 ); —NH(R a ); —N(R a )(R b ) where R a and R b are independently C 1-3 alkyl; or —COOR 9b , wherein R 9b is C 1-3 alkyl (preferably methyl or ethyl); and optionally R 9 and one of R 8 and R 10 together form a 3, 4, 5, or 6-membered heterocycle; preferably R 9 is selected from the group outlined above except R 9 is not H or C 1-3 alkyl.
  • R 9 is N 3 , —OR 9a wherein R 9a is C 1-3 alkyl optionally substituted with 1-7 F, —N(R a )(R b ) where R a and R b are independently C 1-3 alkyl, —COOR 9b where R 9b is C 1-3 alkyl.
  • R 9 is —OCH 3 , —OC 2 H 5 , —N(CH 3 ) 2 , —CO 2 CH 3 , —OCHF 2 , or N 3 .
  • R 9 is H, R 8 or R 10 or both are independently OH; Cl; N 3 ; —XR 9a , where X is O or S, and R 9a is C 1-4 alkyl or C 1-3 haloalkyl (e.g., fluoroalkyl, preferably fluoromethyl, i.e., CH 2 F, CHF 2 , CF 3 ); —NH(R a ) or —N(R a )(R b ) where R a and R b are independently C 1-3 alkyl; or —COOR 9b , wherein R 9b is C 1-3 alkyl (preferably methyl or ethyl).
  • R 9 is H, R 8 or R 10 or both are independently N 3 , —OR 9a , wherein R 9a is C 1-4 alkyl or C 1-3 haloalkyl; —N(R a )(R b ) where R a and R b are independently C 1-3 alkyl; or —COOR 9b , wherein R 9b is C 1-3 alkyl (preferably methyl or ethyl). Even more preferably when R 9 is H, R 8 or R 10 or both are C 1-3 alkoxy or C 1-3 haloalkoxy.
  • R 2 when R 9 is H then R 2 is not H, and preferably R 2 is halo; C 1-3 alkyl optionally substituted with OH or halo (preferably F, e.g., monofluoro, difluoro, or trifluoro); —XR 2a wherein X is S or O and R 2a is C 1-3 alkyl (preferably C 1-3 alkyl, more preferably CH 3 ) optionally substituted with halo (preferably F, e.g., monofluoro-, difluoro-, or trifluoro-substituted); or —N(R a )(R b ) wherein R a and R b are independently H, OH (R a and R b are not both OH), C 1-3 alkyl (preferably CH 3 ), C 2-3 hydroxyalkyl.
  • R a and R b are independently H, OH (R a and R b are not both OH), C 1-3 alkyl (preferably CH 3
  • R 2 is methyl, ethyl, Cl, F, fluoromethyl (CH 2 F, CHF 2 , CF 3 ), C 1-3 hydroxyalkyl (preferably CH 2 OH or CH 2 CH 2 OH), NH 2 , NH 2 OH, —NHCH 2 CH 2 OH, NHCH 3 , N(CH 3 ) 2 , N 3 , morpholino, OCH 3 , OC 2 H 5 , or SCH 3 .
  • R 9 is C 1-6 alkyl or C 1-6 haloalkyl
  • R 2 is not H, and preferably is methyl, ethyl, Cl, F, fluoromethyl (CH 2 F, CHF 2 , CF 3 ), C 1-3 hydroxyalkyl (preferably CH 2 OH or CH 2 CH 2 OH), NH 2 , NH 2 OH, —NHCH 2 CH 2 OH, NHCH 3 , N(CH 3 ) 2 , N 3 , morpholino, OCH 3 , OC 2 H 5 , or SCH 3 .
  • R 2 is H; halo; N 3 ; C 1-6 alkyl (preferably C 1-3 alkyl, more preferably CH 3 ) optionally substituted with OH or halo (preferably F, e.g., monofluoro, difluoro, or trifluoro); —XR 2a wherein X is S or O, and R 2a is C 1-6 alkyl (preferably C 1-3 alkyl, more preferably CH 3 ) optionally substituted with OH or halo (preferably F, e.g., monofluoro-, difluoro-, or trifluoro-substituted); —CO 2 R d , wherein R d is C 1-3 alkyl, preferably methyl or ethyl; or —N(R a )(R b ) wherein R a and R b are independently H, OH (R a and R b are not both OH), C 1-3 alkyl (preferably CH 3 ), C
  • R 2 is H; halo; C 1-3 alkyl optionally substituted with OH or halo (preferably F, e.g., monofluoro, difluoro, or trifluoro); —XR 2a wherein X is S or O and R 2a is C 1-3 alkyl (preferably C 1-3 alkyl, more preferably CH 3 ) optionally substituted with halo (preferably F, e.g., monofluoro-, difluoro-, or trifluoro-substituted); or —N(R a )(R b ) wherein R a and R b are independently H, OH (R a and R b are not both OH), C 1-3 alkyl (preferably CH 3 ), C 2-3 hydroxyalkyl.
  • R 2 is H, methyl, ethyl, Cl, F, fluoromethyl (CH 2 F, CHF 2 , CF 3 ), C 1-3 hydroxyalkyl (preferably CH 2 OH or CH 2 CH 2 OH), NH 2 , NH 2 OH, —NHCH 2 CH 2 OH, NHCH 3 , N(CH 3 ) 2 , N 3 , morpholino, OCH 3 , OC 2 H 5 , or SCH 3 .
  • R 2 is H, methyl, Cl, —CH 2 OH, —NH 2 , —NHCH 3 , —NHCH 2 CH 2 OH, —OCH 3 , —SCH 3 , or —CH 2 F.
  • one or two of Q, T, U and V are N.
  • Q and V are N and T and U are C.
  • compounds of the invention include compounds of Formula VI or pharmaceutically acceptable salts or solvates thereof, wherein:
  • C 1-6 alkyl (preferably C 1-3 alkyl, more preferably CH 3 ) optionally substituted with 1, 2 or 3 substituents, each substituent being independently OH, halo, C 1-3 alkoxy, (halo)C 1-3 alkoxy, —N(R a )(R b ) where R a and R b are independently H, OH (R a and R b are not both OH), C 2-4 hydroxyalkyl, or C 1-3 alkyl or R a and R b together with the nitrogen atom to which they are both linked form a 3, 4, 5 or 6-membered heterocycle;
  • R c is C 1-6 alkyl (preferably C 1-3 alkyl, more preferably CH 3 ) optionally substituted with 1, 2 or 3 substituents, each substituent being independently OH, halo, C 1-3 alkoxy, or (halo)C 1-3 alkoxy;
  • R d is an C 1-6 alkyl (preferably C 1-3 alkyl, more preferably methyl or ethyl) optionally substituted with 1, 2 or 3 substituents, each substituent being independently OH, halo, C 1-3 alkoxy (e.g., fluoroalkoxy), —N(R a )(R b ) where R a and R b are independently H, OH (R a and R b are not both OH), C 2-4 hydroxyalkyl, or C 1-3 alkyl or R a and R b together with the nitrogen atom to which they both are linked form a 3, 4, 5 or 6-membered heterocycle;
  • R a and R b are independently H, OH (R a and R b are not both OH), C 2-4 hydroxyalkyl, C 1-3 alkyl, or C 1-3 alkyl substituted with —N(R e )(R f ) where R e and R f are independently H, OH (R e and R f are not both OH), or C 1-3 alkyl; wherein optionally R a and R b together with the N form a 3, 4, 5 or 6-membered heterocycle, and optionally R e and R f together with the nitrogen atom to which they both are linked form a 3, 4, 5 or 6-membered heterocycle; or
  • R a and R b are independently H or C 1-3 alkyl; and optionally R 9 and one of R 8 and R 10 together form a 3, 4, 5, or 6-membered heterocycle; provided that when R 9 is H, at least one of R 8 and R 10 is not hydrogen or alkyl;
  • Q, T, U and V are independently C or N, provided that at least one of Q, T, U and V is N, wherein when Q, T, U or V is N, then there is no substituent at the N. In some specific embodiments, one or two of Q, T, U and V are N.
  • R 9 is H, R 8 or R 10 or both are independently OH; Cl; N 3 ; —XR 9a , where X is O or S, and R 9a is C 1-4 alkyl or C 1-3 haloalkyl (e.g., fluoroalkyl, preferably fluoromethyl, i.e., CH 2 F, CHF 2 , CF 3 ); —NH(R a ) or —N(R a )(R b ) where R a and R b are independently C 1-3 alkyl; or —COOR 9b , wherein R 9b is C 1-3 alkyl (preferably methyl or ethyl).
  • R 9 is H, R 8 or R 10 or both are independently N 3 , —OR 9a , wherein R 9a is C 1-4 alkyl or C 1-3 haloalkyl, or —N(R a )(R b ) where R a and R b are independently C 1-3 alkyl; or —COOR 9b , wherein R 9b is C 1-3 alkyl (preferably methyl or ethyl). Even more preferably when R 9 is H, R 8 or R 10 or both are C 1-3 alkoxy or C 1-3 haloalkoxy.
  • R 9 is C 1-6 alkyl or C 1-6 haloalkyl
  • R 2 is methyl, ethyl, Cl, F, fluoromethyl (CH 2 F, CHF 2 , CF 3 ), C 1-3 hydroxyalkyl (preferably CH 2 OH or CH 2 CH 2 OH), NH 2 , NH 2 OH, —NHCH 2 CH 2 OH, NHCH 3 , N(CH 3 ) 2 , N 3 , morpholino, OCH 3 , OC 2 H 5 , or SCH 3 .
  • compounds of the invention include compounds of Formula VI or pharmaceutically acceptable salts or solvates thereof, wherein:
  • one or two of Q, T, U and V are N.
  • R 9 is H
  • R 8 or R 10 or both are independently OH; Cl; N 3 ; C 1-3 haloalkyl (preferably monofluoromethyl, difluoromethyl, trifluoromethyl); —XR 9a , where X is O or S, and R 9a is C 1-4 alkyl or C 1-3 haloalkyl (e.g., fluoroalkyl, preferably fluoromethyl, i.e., CH 2 F, CHF 2 , CF 3 ); —NH(R a ) or —N(R a )(R b ) where R a and R b are independently C 1-3 alkyl; or —COOR 9b , wherein R 9b is C 1-3 alkyl (preferably methyl or ethyl).
  • R 9 is H, R 8 or R 10 or both are independently N 3 , —OR 9a , wherein R 9a is C 1-4 alkyl or C 1-3 haloalkyl, or —N(R a )(R b ) where R a and R b are independently C 1-3 alkyl; or —COOR 9b , wherein R 9b is C 1-3 alkyl (preferably methyl or ethyl). Even more preferably when R 9 is H, R 8 or R 10 or both are C 1-3 alkoxy or C 1-3 halo alkoxy.
  • R 9 is C 1-6 alkyl or C 1-6 haloalkyl
  • R 2 is not H and preferably R 2 is halo; C 1-3 alkyl optionally substituted with OH or halo (preferably F, e.g., monofluoro, difluoro, or trifluoro); —XR 2a wherein X is S or O and R 2a is C 1-3 alkyl (preferably C 1-3 alkyl, more preferably CH 3 ) optionally substituted with halo (preferably F, e.g., monofluoro-, difluoro-, or trifluoro-substituted); or —N(R a )(R b ) wherein R a and R b are independently H, OH (R a and R b are not both OH), C 1-3 alkyl (preferably CH 3 ), C 2-3 hydroxyalkyl.
  • R 2 is methyl, ethyl, Cl, F, fluoromethyl (CH 2 F, CHF 2 , CF 3 ), C 1-3 hydroxyalkyl (preferably CH 2 OH or CH 2 CH 2 OH), NH 2 , NH 2 OH, —NHCH 2 CH 2 OH, NHCH 3 , N(CH 3 ) 2 , N 3 , morpholino, OCH 3 , OC 2 H 5 , or SCH 3 .
  • R 9 is H
  • R 8 or R 10 or both are OCH 3 and preferably R 7 and R 11 are H
  • R 2 is not hydrogen and preferably R 2 is methyl or chloro.
  • R 9 is alkyl or haloalkyl or chloro
  • R 2 is not hydrogen and preferably R 2 is methyl or chloro.
  • compounds of the invention include compounds of Formula VI or pharmaceutically acceptable salts or solvates thereof, wherein:
  • R 9 is H
  • R 8 or R 10 or both are independently OH; Cl; N 3 ; C 1-3 haloalkyl (preferably monofluoromethyl, difluoromethyl, trifluoromethyl); —XR 9a , where X is O or S, and R 9a is C 1-4 alkyl or C 1-3 haloalkyl (e.g., fluoroalkyl, preferably fluoromethyl, i.e., CH 2 F, CHF 2 , CF 3 ); —NH(R a ) or —N(R a )(R b ) where R a and R b are independently C 1-3 alkyl; or —COOR 9b , wherein R 9b is C 1-3 alkyl (preferably methyl or ethyl).
  • R 9 is H, R 8 or R 10 or both are independently N 3 , —OR 9a , wherein R 9a is C 1-4 alkyl or C 1-3 haloalkyl, or —N(R a )(R b ) where R a and R b are independently C 1-3 alkyl; or —COOR 9b , wherein R 9b is C 1-3 alkyl (preferably methyl or ethyl). Even more preferably when R 9 is H, R 8 or R 10 or both are C 1-3 alkoxy or C 1-3 halo alkoxy.
  • R 2 is not H and preferably R 2 is methyl, ethyl, Cl, F, fluoromethyl (CH 2 F, CHF 2 , CF 3 ), C 1-3 hydroxyalkyl (preferably CH 2 OH or CH 2 CH 2 OH), NH 2 , NH 2 OH, —NHCH 2 CH 2 OH, NHCH 3 , N(CH 3 ) 2 , N 3 , morpholino, OCH 3 , OC 2 H 5 , or SCH 3 .
  • R 9 is H
  • R 8 or R 10 or both are OCH 3 and preferably R 7 and R 11 are H
  • R 2 is not hydrogen and preferably R 2 is methyl or chloro.
  • R 9 is alkyl or haloalkyl or chloro
  • R 2 is not hydrogen and preferably R 2 is methyl or chloro.
  • compounds of the invention include compounds of Formula VI or pharmaceutically acceptable salts or solvates thereof, wherein:
  • R 1 is methyl
  • R 2 is H, methyl, ethyl, Cl, F, fluoromethyl (CH 2 F, CHF 2 , CF 3 ), CH 2 OH, NH 2 , NHCH 3 , N(CH 3 ) 2 , —NHCH 2 CH 2 OH, OCH 3 , or SCH 3 ;
  • R 3 is H, CH 3 , OCH 3 , F, or Cl;
  • R 4 and R 6 are independently H, CH 3 , NH 2 , F, or Cl;
  • R 5 is H;
  • R 7 and R 11 are independently H, F, or OCH 3 ;
  • R 8 and R 10 are independently H, F, Cl, or OCH 3 ; and
  • R 9 is selected from the group:
  • R 9a is selected from the group of methyl, ethyl, fluoromethyl (e.g., CH 2 F, CHF 2 , CF 3 ), and fluoroethyl;
  • Q, T, U and V are independently C or N, and at least one of Q, T, U and V is N, wherein when Q, T, U or V is N, then there is no substituent at the N.
  • one or two of Q, T, U and V are nitrogen.
  • compounds of the invention include compounds of Formula VI or pharmaceutically acceptable salts or solvates thereof, wherein:
  • R 1 is CH 3 ;
  • R 2 is H, methyl, Cl, —CH 2 OH, —NH 2 , —NHCH 3 , —NHCH 2 CH 2 OH, —OCH 3 , —SCH 3 , or —CH 2 F;
  • R 3 is H, —CH 3 , —OCH 3 , or Cl;
  • R 4 is H, CH 3 , or NH 2 ;
  • R 5 is H
  • R 6 is H, or CH 3 ;
  • R 7 and R 11 are independently H, or F;
  • R 8 and R 10 are independently H, or F or OCH 3 ;
  • R 9 is —OCH 3 or —OC 2 H 5 , —N(CH 3 ) 2 , —CO 2 CH 3 , —OCHF 2 , or N 3 ; exactly one of B and D is N; and
  • Q, T, U and V are independently C or N, and at least one of Q, T, U and V is N, wherein when Q, T, U or V is N, then there is no substituent at the N.
  • one or two of Q, T, U and V are nitrogen.
  • the present invention provides compounds of Formula VI or pharmaceutically acceptable salts or solvates thereof, wherein R 1 is CH 3 ; R 2 is Cl, methyl, or CH 2 F; R 3 is H, CH 3 , F, or Cl; R 4 , R 5 and R 6 are H; R 7 , R 8 , R 10 and R 11 are independently H or F; R 9 is —OCH 3 or —N(CH 3 ) 2 ; and Q, T, U and V are independently C or N, and at least one of Q, T, U and V is N, wherein when Q, T, U or V is N, then there is no substituent at the N. In some specific embodiments, one or two of Q, T, U and V are nitrogen.
  • compounds of the invention include compounds of Formula VI or pharmaceutically acceptable salts or solvates thereof, wherein:
  • R 9 is selected from the group consisting of H, C 1 , N 3 ;
  • C 1-6 alkyl (preferably C 1-3 alkyl, more preferably CH 3 ) optionally substituted with 1, 2 or 3 substituents, each substituent being independently OH, halo, C 1-3 alkoxy, (halo)C 1-3 alkoxy, —N(R a )(R b ) where R a and R b are independently H, OH (R a and R b are not both OH), C 2-4 hydroxyalkyl, or C 1-3 alkyl or R a and R b together with the nitrogen atom to which they are both linked form a 3, 4, 5 or 6-membered heterocycle;
  • R c is C 1-6 alkyl (preferably C 1-3 alkyl, more preferably CH 3 ) optionally substituted with 1, 2 or 3 substituents, each substituent being independently OH, halo, C 1-3 alkoxy, or (halo)C 1-3 alkoxy;
  • R d is an C 1-6 alkyl, preferably C 1-3 alkyl;
  • R a and R b are independently H, OH (R a and R b are not both OH), C 2-4 hydroxyalkyl, C 1-3 alkyl, or —N(R e )(R f ) where R e and R f are independently H, OH (R e and R f are not both OH), or C 1-3 alkyl; wherein optionally R a and R b together with the N form a 3, 4, 5 or 6-membered heterocycle, and optionally R e and R f together with the nitrogen atom to which they both are linked form a 3, 4, 5 or 6-membered heterocycle; or
  • R 9 is selected from such groups except R 9 is not H or chloro.
  • R 9 is H; OH; Cl; N 3 ; C 1-3 alkyl (preferably methyl; C 1-3 haloalkyl (preferably monofluoromethyl, difluoromethyl, trifluoromethyl); —OR 9a , wherein R 9a is C 1-4 alkyl or C 1-3 haloalkyl (e.g., fluoroalkyl, preferably fluoromethyl, i.e., CH 2 F, CHF 2 , CF 3 ); —NH(R a ) or —N(R a )(R b ) where R a and R b are independently C 1-3 alkyl; or —COOR 9b , wherein R 9b is C 1-3 alkyl (preferably methyl or ethyl); and optionally R 9 and one of R 8 and R 10 together form a 3, 4, 5, or 6-membered heterocycle.
  • R 9 is selected from such groups except R 9 is not H or chloro.
  • R 9 is N 3 ; —OR 9a , wherein R 9a is C 1-3 alkyl optionally substituted with 1-7 F; —N(R a )(R b ) where R a and R b are independently C 1-3 alkyl; or —COOR 9b where R 9b is C 1-3 alkyl.
  • R 9 is —OCH 3 , —OC 2 H 5 , —N(CH 3 ) 2 , —CO 2 CH 3 , —OCHF 2 , or N 3 .
  • R 9 is H, R 8 or R 10 or both are independently OH; N 3 ; —XR 9a , where X is O or S, and R 9a is C 1-4 alkyl or C 1-3 haloalkyl (e.g., fluoroalkyl, preferably fluoromethyl, i.e., CH 2 F, CHF 2 , CF 3 ); —NH(R a ) or —N(R a )(R b ) where R a and R b are independently C 1-3 alkyl; or —COOR 9b , wherein R 9b is C 1-3 alkyl (preferably methyl or ethyl).
  • R 9 is H, R 8 or R 10 or both are independently N 3 , —OR 9a , wherein R 9a is C 1-4 alkyl or C 1-3 haloalkyl, or —N(R a )(R b ) where R a and R b are independently C 1-3 alkyl; or —COOR 9b , wherein R 9b is C 1-3 alkyl (preferably methyl or ethyl). Even more preferably when R 9 is H, R 8 or R 10 or both are C 1-3 alkoxy or C 1-3 halo alkoxy.
  • R 9 is H
  • R 8 and R 10 are not H or one H and the other halo
  • R 2 is not H, and preferably R 2 is halo
  • N 3 C 1-3 alkyl optionally substituted with OH or halo (preferably F, e.g., monofluoro, difluoro, or trifluoro)
  • —XR 2a wherein X is S or O and R 2a is C 1-3 alkyl (more preferably CH 3 ) optionally substituted with halo (preferably F, e.g., monofluoro-, difluoro-, or trifluoro-substituted
  • R a and R b are independently H, OH (R a and R b are not both OH), C 1-3 alkyl (preferably CH 3 ), C 2-3 hydroxyalkyl.
  • R 2 is methyl, ethyl, Cl, F, fluoromethyl (CH 2 F, CHF 2 , CF 3 ), C 1-3 hydroxyalkyl (preferably CH 2 OH or CH 2 CH 2 OH), NH 2 , NH 2 OH, —NHCH 2 CH 2 OH, NHCH 3 , N(CH 3 ) 2 , N 3 , morpholino, OCH 3 , OC 2 H 5 , or SCH 3 .
  • R 9 is C 1-6 alkyl, halo, or C 1-6 haloalkyl
  • R 2 is not H, and preferably R 2 is halo; N 3 , C 1-3 alkyl optionally substituted with OH or halo (preferably F, e.g., monofluoro, difluoro, or trifluoro); —XR 2a wherein X is S or O and R 2a is C 1-3 alkyl (more preferably CH 3 ) optionally substituted with halo (preferably F, e.g., monofluoro-, difluoro-, or trifluoro-substituted); or —N(R a )(R b ) wherein R a and R b are independently H, OH (R a and R b are not both OH), C 1-3 alkyl (preferably CH 3 ), C 2-3 hydroxyalkyl.
  • R 2 is methyl, ethyl, Cl, F, fluoromethyl (CH 2 F, CHF 2 , CF 3 ), C 1-3 hydroxyalkyl (preferably CH 2 OH or CH 2 CH 2 OH), NH 2 , NH 2 OH, —NHCH 2 CH 2 OH, NHCH 3 , N(CH 3 ) 2 , N 3 , morpholino, OCH 3 , OC 2 H 5 , or SCH 3 .
  • R 2 is H; halo; N 3 ; C 1-6 alkyl (preferably C 1-3 alkyl, more preferably CH 3 ) optionally substituted with OH or halo (preferably F, e.g., monofluoro, difluoro, or trifluoro); —XR 2a wherein X is S or O, and R 2a is C 1-6 alkyl (preferably C 1-3 alkyl, more preferably CH 3 ) optionally substituted with OH or halo (preferably F, e.g., monofluoro-, difluoro-, or trifluoro-substituted); —CO 2 R d , wherein R d is C 1-3 alkyl, preferably methyl or ethyl; or —N(R a )(R b ) wherein R a and R b are independently H, OH (R a and R b are not both OH), C 1-3 alkyl (preferably CH 3 ), C
  • R 2 is H; halo; C 1-3 alkyl optionally substituted with OH or halo (preferably F, e.g., monofluoro, difluoro, or trifluoro); —XR 2a wherein X is S or O and R 2a is C 1-3 alkyl (preferably C 1-3 alkyl, more preferably CH 3 ) optionally substituted with halo (preferably F, e.g., monofluoro-, difluoro-, or trifluoro-substituted); or —N(R a )(R b ) wherein R a and R b are independently H, OH (R a and R b are not both OH), C 1-3 alkyl (preferably CH 3 ), C 2-3 hydroxyalkyl.
  • R 2 is H, methyl, ethyl, Cl, F, fluoromethyl (CH 2 F, CHF 2 , CF 3 ), C 1-3 hydroxyalkyl (preferably CH 2 OH or CH 2 CH 2 OH), NH 2 , NH 2 OH, —NHCH 2 CH 2 OH, NHCH 3 , N(CH 3 ) 2 , N 3 , morpholino, OCH 3 , OC 2 H 5 , or SCH 3 .
  • R 2 is H, methyl, Cl, —CH 2 OH, —NH 2 , —NHCH 3 , —NHCH 2 CH 2 OH, —OCH 3 , —SCH 3 , or —CH 2 F.
  • compounds of the invention include compounds of Formula VI or pharmaceutically acceptable salts or solvates thereof, wherein:
  • R 9 is H, R 8 or R 10 or both are independently OH; N 3 ; —XR 9a , where X is O or S, and R 9a is C 1-4 alkyl or C 1-3 haloalkyl (e.g., fluoroalkyl, preferably fluoromethyl, i.e., CH 2 F, CHF 2 , CF 3 ); —NH(R 3 ) or —N(R a )(R b ) where R a and R b are independently C 1-3 alkyl; or —COOR 9b , wherein R 9b is C 1-3 alkyl (preferably methyl or ethyl).
  • R 9a is C 1-4 alkyl or C 1-3 haloalkyl
  • R 9a is C 1-4 alkyl or C 1-3 haloalkyl (e.g., fluoroalkyl, preferably fluoromethyl, i.e., CH 2 F, CHF 2 , CF 3
  • R 9 is H
  • R 8 or R 10 or both are independently N 3
  • —OR 9a wherein R 9a is C 1-4 alkyl or C 1-3 haloalkyl, or —N(R 3 )(R b ) where R a and R b are independently C 1-3 alkyl; or —COOR 9b , wherein R 9b is C 1-3 alkyl (preferably methyl or ethyl).
  • R 9 is H, R 8 or R 10 or both are C 1-3 alkoxy or C 1-3 halo alkoxy.
  • R 2 is not H and preferably R 2 is methyl, ethyl, Cl, F, fluoromethyl (CH 2 F, CHF 2 , CF 3 ), C 1-3 hydroxyalkyl (preferably CH 2 OH or CH 2 CH 2 OH), NH 2 , NH 2 OH, —NHCH 2 CH 2 OH, NHCH 3 , N(CH 3 ) 2 , N 3 , morpholino, OCH 3 , OC 2 H 5 , or SCH 3 .
  • compounds of the invention include compounds of Formula VI or pharmaceutically acceptable salts or solvates thereof, wherein:
  • R 9 is H
  • R 8 or R 10 or both are independently —XR 9a , where X is O or S
  • R 9a is C 1-4 alkyl or C 1-3 haloalkyl (e.g., fluoroalkyl, preferably fluoromethyl, i.e., CH 2 F, CHF 2 , CF 3 ); —N(R 3 )(R b ) where R a and R b are independently C 1-3 alkyl; or —COOR 9b , wherein R 9b is C 1-3 alkyl (preferably methyl or ethyl).
  • R 9 is H, R 8 or R 10 or both are independently N 3 , —OR 9a , wherein R 9a is C 1-4 alkyl or C 1-3 haloalkyl, or —N(R a )(R b ) where R a and R b are independently C 1-3 alkyl; or —COOR 9b , wherein R 9b is C 1-3 alkyl (preferably methyl or ethyl). Even more preferably when R 9 is H, R 8 or R 10 or both are C 1-3 alkoxy or C 1-3 halo alkoxy.
  • R 9 is H then R 8 and R 10 are not both H or one H and the other halo, and R 2 is not H.
  • R 2 is not H and preferably R 2 is methyl, ethyl, Cl, F, fluoromethyl (CH 2 F, CHF 2 , CF 3 ), C 1-3 hydroxyalkyl (preferably CH 2 OH or CH 2 CH 2 OH), NH 2 , NH 2 OH, —NHCH 2 CH 2 OH, NHCH 3 , N(CH 3 ) 2 , N 3 , morpholino, OCH 3 , OC 2 H 5 , or SCH 3 .
  • R 9 is H
  • R 8 or R 10 or both are OCH 3 and preferably R 7 and R 11 are H
  • R 2 is not hydrogen and preferably R 2 is methyl or chloro.
  • R 9 is alkyl or haloalkyl or chloro
  • R 2 is not hydrogen and preferably R 2 is methyl or chloro.
  • compounds of the invention include compounds of Formula VI or pharmaceutically acceptable salts or solvates thereof, wherein:
  • R 9 is H
  • R 8 or R 10 or both are independently C 1-3 alkoxy (preferably OCH 3 ) or C 1-3 alkylthiol, each being optionally substituted with 1-4 F. Even more preferably when R 9 is H, R 8 or R 10 or both are methoxy or ethoxy. Also preferably R 2 is not H.
  • R 2 is not H, preferably R 2 is methyl, ethyl, Cl, F, fluoromethyl (CH 2 F, CHF 2 , CF 3 ), C 1-3 hydroxyalkyl (preferably CH 2 OH or CH 2 CH 2 OH), NH 2 , NH 2 OH, —NHCH 2 CH 2 OH, NHCH 3 , N(CH 3 ) 2 , N 3 , morpholino, OCH 3 , OC 2 H 5 , or SCH 3 .
  • R 9 is H
  • R 8 or R 10 or both are OCH 3 and preferably R 7 and R 11 are H
  • R 2 is not hydrogen and preferably R 2 is methyl or chloro.
  • R 9 is alkyl or haloalkyl or chloro
  • R 2 is not hydrogen and preferably R 2 is methyl or chloro.
  • compounds of the invention include compounds of Formula VI or pharmaceutically acceptable salts or solvates thereof, wherein:
  • R 9 is H
  • R 8 or R 10 or both are independently C 1-3 alkoxy (preferably OCH 3 ) or C 1-3 alkylthiol, each being optionally substituted with 1-4 F. Even more preferably when R 9 is H, R 8 or R 10 or both are methoxy or ethoxy. Also preferably R 2 is not H.
  • R 2 is not H, preferably R 2 is methyl, ethyl, Cl, F, fluoromethyl (CH 2 F, CHF 2 , CF 3 ), C 1-3 hydroxyalkyl (preferably CH 2 OH or CH 2 CH 2 OH), NH 2 , NH 2 OH, —NHCH 2 CH 2 OH, NHCH 3 , N(CH 3 ) 2 , N 3 , morpholino, OCH 3 , OC 2 H 5 , or SCH 3 .
  • R 9 is H
  • R 8 or R 10 or both are OCH 3 and preferably R 7 and R 11 are H
  • R 2 is not hydrogen and preferably R 2 is methyl or chloro.
  • R 9 is alkyl or haloalkyl or chloro
  • R 2 is not hydrogen and preferably R 2 is methyl or chloro.
  • compounds of the invention include compounds of Formula VI or pharmaceutically acceptable salts or solvates thereof, wherein:
  • R 12 is selected from the group of methyl, ethyl, fluoromethyl (e.g., CH 2 F, CHF 2 , CF 3 ), and fluoroethyl;
  • compounds of the invention include compounds of Formula VI or pharmaceutically acceptable salts or solvates thereof, wherein:
  • the present invention provides compounds of Formula VI or pharmaceutically acceptable salts or solvates thereof, wherein R 1 is CH 3 ; R 2 is Cl, methyl, or CH 2 F; R 3 is H, CH 3 , F, or Cl; R 4 , R 5 and R 6 are H; R 7 , R 8 , R 10 and R 11 are independently H or F; and R 9 is —OCH 3 or —N(CH 3 ) 2 ; exactly one of B and D is N provided that when B or D is N there is no substituent at the N; and all of Q, T, U, and V are C.
  • one group of the compounds of Formula VI are those represented by Formula VIa: or pharmaceutically acceptable salts or solvates thereof, wherein:
  • R 9 is selected from the group consisting of H, OH, C 1 , N 3 ;
  • C 1-6 alkyl (preferably C 1-3 alkyl, more preferably CH 3 ) optionally substituted with 1, 2 or 3 substituents, each substituent being independently OH, halo, C 1-3 alkoxy, (halo)C 1-3 alkoxy, —N(R a )(R b ) where R a and R b are independently H, OH (R a and R b are not both OH), C 2-4 hydroxyalkyl, or C 1-3 alkyl or R a and R b together with the nitrogen atom to which they are both linked to form a 3, 4, 5 or 6-membered heterocycle;
  • R c is C 1-6 alkyl (preferably C 1-3 alkyl, more preferably CH 3 ) optionally substituted with 1, 2 or 3 substituents, each substituent being independently OH, halo, C 1-3 alkoxy, or (halo)C 1-3 alkoxy;
  • R d is an C 1-6 alkyl (preferably C 1-3 alkyl, more preferably methyl or ethyl) optionally substituted with 1, 2 or 3 substituents, each substituent being independently OH, halo, C 1-3 alkoxy (e.g., fluoroalkoxy), —N(R a )(R b ) where R a and R b are independently H, OH (R a and R b are not both OH), C 2-4 hydroxyalkyl, or C 1-3 alkyl or R a and R b together with the nitrogen atom to which they both are linked form a 3, 4, 5 or 6-membered heterocycle;
  • R a and R b are independently H, OH (R a and R b are not both OH), C 2-4 hydroxyalkyl, C 1-3 alkyl, or C 1-3 alkyl substituted with —N(R e )(R f ) where R e and R f are independently H, OH (R e and R f are not both OH), or C 1-3 alkyl; wherein optionally R a and R b together with the N form a 3, 4, 5 or 6-membered heterocycle, and optionally R e and R f together with the nitrogen atom to which they both are linked form a 3, 4, 5 or 6-membered heterocycle; or
  • R 9 is selected from the group outlined above except R 9 is not H and C 1-6 alkyl.
  • R 9 is H; OH; Cl; N 3 ; C 1-3 alkyl (preferably methyl; C 1-3 haloalkyl (preferably monofluoromethyl, difluoromethyl, trifluoromethyl); —OR 9a , wherein R 9a is C 1-4 alkyl or C 1-3 haloalkyl (e.g., fluoroalkyl, preferably fluoromethyl, i.e., CH 2 F, CHF 2 , CF 3 ); —NH(R a ); —N(R a )(R b ) where R a and R b are independently C 1-3 alkyl; or —COOR 9b , wherein R 9b is C 1-3 alkyl (preferably methyl or ethyl); and optionally R 9 and one of R 8 and R 10 together form a 3, 4, 5, or 6-membered heterocycle; preferably R 9 is selected from the group outlined above except R 9 is not H or C 1-3 alkyl.
  • R 9 is N 3 , —OR 9a wherein R 9a is C 1-3 alkyl optionally substituted with 1-7 F, —N(R a )(R b ) where R a and R b are independently C 1-3 alkyl, —COOR 9b where R 9b is C 1-3 alkyl.
  • R 9 is —OCH 3 , —OC 2 H 5 , —N(CH 3 ) 2 , —CO 2 CH 3 , —OCHF 2 , or N 3 .
  • R 9 is H, R 8 or R 10 or both are independently OH; Cl; N 3 ; —XR 9a , where X is O or S, and R 9a is C 1-4 alkyl or C 1-3 haloalkyl (e.g., fluoroalkyl, preferably fluoromethyl, i.e., CH 2 F, CHF 2 , CF 3 ); —NH(R a ) or —N(R a )(R b ) where R a and R b are independently C 1-3 alkyl; or —COOR 9b , wherein R 9b is C 1-3 alkyl (preferably methyl or ethyl).
  • R 9 is H, R 8 or R 10 or both are independently N 3 , —OR 9a , wherein R 9a is C 1-4 alkyl or C 1-3 haloalkyl; —N(R a )(R b ) where R a and R b are independently C 1-3 alkyl; or —COOR 9b , wherein R 9b is C 1-3 alkyl (preferably methyl or ethyl). Even more preferably when R 9 is H, R 8 or R 10 or both are C 1-3 alkoxy or C 1-3 haloalkoxy.
  • R 2 when R 9 is H then R 2 is not H, and preferably R 2 is halo; C 1-3 alkyl optionally substituted with OH or halo (preferably F, e.g., monofluoro, difluoro, or trifluoro); —XR 2a wherein X is S or O and R 2a is C 1-3 alkyl (preferably C 1-3 alkyl, more preferably CH 3 ) optionally substituted with halo (preferably F, e.g., monofluoro-, difluoro-, or trifluoro-substituted); or —N(R a )(R b ) wherein R a and R b are independently H, OH (R a and R b are not both OH), C 1-3 alkyl (preferably CH 3 ), C 2-3 hydroxyalkyl.
  • R a and R b are independently H, OH (R a and R b are not both OH), C 1-3 alkyl (preferably CH 3
  • R 2 is methyl, ethyl, Cl, F, fluoromethyl (CH 2 F, CHF 2 , CF 3 ), C 1-3 hydroxyalkyl (preferably CH 2 OH or CH 2 CH 2 OH), NH 2 , NH 2 OH, —NHCH 2 CH 2 OH, NHCH 3 , N(CH 3 ) 2 , N 3 , morpholino, OCH 3 , OC 2 H 5 , or SCH 3 .
  • R 9 is C 1-6 alkyl or C 1-6 haloalkyl
  • R 2 is not H, and preferably is methyl, ethyl, Cl, F, fluoromethyl (CH 2 F, CHF 2 , CF 3 ), C 1-3 hydroxyalkyl (preferably CH 2 OH or CH 2 CH 2 OH), NH 2 , NH 2 OH, —NHCH 2 CH 2 OH, NHCH 3 , N(CH 3 ) 2 , N 3 , morpholino, OCH 3 , OC 2 H 5 , or SCH 3 .
  • R 2 is H; halo; N 3 ; C 1-6 alkyl (preferably C 1-3 alkyl, more preferably CH 3 ) optionally substituted with OH or halo (preferably F, e.g., monofluoro, difluoro, or trifluoro); —XR 2a wherein X is S or O, and R 2a is C 1-6 alkyl (preferably C 1-3 alkyl, more preferably CH 3 ) optionally substituted with OH or halo (preferably F, e.g., monofluoro-, difluoro-, or trifluoro-substituted); —CO 2 R d , wherein R d is C 1-3 alkyl, preferably methyl or ethyl; or —N(R a )(R b ) wherein R a and R b are independently H, OH (R a and R b are not both OH), C 1-3 alkyl (preferably CH 3 ), C
  • R 2 is H; halo; C 1-3 alkyl optionally substituted with OH or halo (preferably F, e.g., monofluoro, difluoro, or trifluoro); —XR 2a wherein X is S or O and R 2a is C 1-3 alkyl (preferably C 1-3 alkyl, more preferably CH 3 ) optionally substituted with halo (preferably F, e.g., monofluoro-, difluoro-, or trifluoro-substituted); or —N(R a )(R b ) wherein R a and R b are independently H, OH (R a and R b are not both OH), C 1-3 alkyl (preferably CH 3 ), C 2-3 hydroxyalkyl.
  • R 2 is H, methyl, ethyl, Cl, F, fluoromethyl (CH 2 F, CHF 2 , CF 3 ), C 1-3 hydroxyalkyl (preferably CH 2 OH or CH 2 CH 2 OH), NH 2 , NH 2 OH, —NHCH 2 CH 2 OH, NHCH 3 , N(CH 3 ) 2 , N 3 , morpholino, OCH 3 , OC 2 H 5 , or SCH 3 .
  • R 2 is H, methyl, Cl, —CH 2 OH, —NH 2 , —NHCH 3 , —NHCH 2 CH 2 OH, —OCH 3 , —SCH 3 , or —CH 2 F.
  • one or two of Q, T, U and V are N.
  • Q and V are N and T and U are C.
  • compounds of the invention include compounds of Formula VIa or pharmaceutically acceptable salts or solvates thereof, wherein:
  • C 1-6 alkyl (preferably C 1-3 alkyl, more preferably CH 3 ) optionally substituted with 1, 2 or 3 substituents, each substituent being independently OH, halo, C 1-3 alkoxy, (halo)C 1-3 alkoxy, —N(R a )(R b ) where R a and R b are independently H, OH (R a and R b are not both OH), C 2-4 hydroxyalkyl, or C 1-3 alkyl or R a and R b together with the nitrogen atom to which they are both linked form a 3, 4, 5 or 6-membered heterocycle;
  • R c is C 1-6 alkyl (preferably C 1-3 alkyl, more preferably CH 3 ) optionally substituted with 1, 2 or 3 substituents, each substituent being independently OH, halo, C 1-3 alkoxy, or (halo)C 1-3 alkoxy;
  • R d is an C 1-6 alkyl (preferably C 1-3 alkyl, more preferably methyl or ethyl) optionally substituted with 1, 2 or 3 substituents, each substituent being independently OH, halo, C 1-3 alkoxy (e.g., fluoroalkoxy), —N(R a )(R b ) where R a and R b are independently H, OH (R a and R b are not both OH), C 2-4 hydroxyalkyl, or C 1-3 alkyl or R a and R b together with the nitrogen atom to which they both are linked form a 3, 4, 5 or 6-membered heterocycle;
  • R a and R b are independently H, OH (R a and R b are not both OH), C 2-4 hydroxyalkyl, C 1-3 alkyl, or C 1-3 alkyl substituted with —N(R e )(R f ) where R e and R f are independently H, OH (R e and R f are not both OH), or C 1-3 alkyl; wherein optionally R a and R b together with the N form a 3, 4, 5 or 6-membered heterocycle, and optionally R e and R f together with the nitrogen atom to which they both are linked form a 3, 4, 5 or 6-membered heterocycle; or
  • R a and R b are independently H or C 1-3 alkyl; and optionally R 9 and one of R 8 and R 10 together form a 3, 4, 5, or 6-membered heterocycle; provided that when R 9 is H, at least one of R 8 and R 10 is not hydrogen or alkyl; and
  • Q, T, U and V are independently C or N, provided that at least one of Q, T, U and V is N, wherein when Q, T, U or V is N, then there is no substituent at the N. In some specific embodiments, one or two of Q, T, U and V are N.
  • R 9 is H, R 8 or R 10 or both are independently OH; Cl; N 3 ; —XR 9a , where X is O or S, and R 9a is C 1-4 alkyl or C 1-3 haloalkyl (e.g., fluoroalkyl, preferably fluoromethyl, i.e., CH 2 F, CHF 2 , CF 3 ); —NH(R a ) or —N(R a )(R b ) where R a and R b are independently C 1-3 alkyl; or —COOR 9b , wherein R 9b is C 1-3 alkyl (preferably methyl or ethyl).
  • R 9 is H, R 8 or R 10 or both are independently N 3 , —OR 9a , wherein R 9a is C 1-4 alkyl or C 1-3 haloalkyl, or —N(R a )(R b ) where R a and R b are independently C 1-3 alkyl; or —COOR 9b , wherein R 9b is C 1-3 alkyl (preferably methyl or ethyl). Even more preferably when R 9 is H, R 8 or R 10 or both are C 1-3 alkoxy or C 1-3 haloalkoxy.
  • R 9 is C 1-6 alkyl or C 1-6 haloalkyl
  • R 2 is methyl, ethyl, Cl, F, fluoromethyl (CH 2 F, CHF 2 , CF 3 ), C 1-3 hydroxyalkyl (preferably CH 2 OH or CH 2 CH 2 OH), NH 2 , NH 2 OH, —NHCH 2 CH 2 OH, NHCH 3 , N(CH 3 ) 2 , N 3 , morpholino, OCH 3 , OC 2 H 5 , or SCH 3 .
  • compounds of the invention include compounds of Formula VIa or pharmaceutically acceptable salts or solvates thereof, wherein:
  • one or two of Q, T, U and V are N.
  • R 9 is H
  • R 8 or R 10 or both are independently OH; Cl; N 3 ; C 1-3 haloalkyl (preferably monofluoromethyl, difluoromethyl, trifluoromethyl); —XR 93 , where X is O or S, and R 9a is C 1-4 alkyl or C 1-3 haloalkyl (e.g., fluoroalkyl, preferably fluoromethyl, i.e., CH 2 F, CHF 2 , CF 3 ); —NH(R a ) or —N(R a )(R b ) where R a and R b are independently C 1-3 alkyl; or —COOR 9b , wherein R 9b is C 1-3 alkyl (preferably methyl or ethyl).
  • R 9 is H, R 8 or R 10 or both are independently N 3 , —OR 9a , wherein R 9a is C 1-4 alkyl or C 1-3 haloalkyl, or —N(R a )(R b ) where R a and R b are independently C 1-3 alkyl; or —COOR 9b , wherein R 9b is C 1-3 alkyl (preferably methyl or ethyl). Even more preferably when R 9 is H, R 8 or R 10 or both are C 1-3 alkoxy or C 1-3 halo alkoxy.
  • R 9 is C 1-6 alkyl or C 1-6 haloalkyl
  • R 2 is not H and preferably R 2 is halo; C 1-3 alkyl optionally substituted with OH or halo (preferably F, e.g., monofluoro, difluoro, or trifluoro); —XR 2a wherein X is S or O and R 2a is C 1-3 alkyl (preferably C 1-3 alkyl, more preferably CH 3 ) optionally substituted with halo (preferably F, e.g., monofluoro-, difluoro-, or trifluoro-substituted); or —N(R a )(R b ) wherein R a and R b are independently H, OH (R a and R b are not both OH), C 1-3 alkyl (preferably CH 3 ), C 2-3 hydroxyalkyl.
  • R 2 is methyl, ethyl, Cl, F, fluoromethyl (CH 2 F, CHF 2 , CF 3 ), C 1-3 hydroxyalkyl (preferably CH 2 OH or CH 2 CH 2 OH), NH 2 , NH 2 OH, —NHCH 2 CH 2 OH, NHCH 3 , N(CH 3 ) 2 , N 3 , morpholino, OCH 3 , OC 2 H 5 , or SCH 3 .
  • R 9 is H
  • R 8 or R 10 or both are OCH 3 and preferably R 7 and R 11 are H
  • R 2 is not hydrogen and preferably R 2 is methyl or chloro.
  • R 9 is alkyl or haloalkyl or chloro
  • R 2 is not hydrogen and preferably R 2 is methyl or chloro.
  • compounds of the invention include compounds of Formula VIa or pharmaceutically acceptable salts or solvates thereof, wherein:
  • R 9b is C 1-3 alkyl (preferably methyl or ethyl), and optionally R 8 and R 9 together form a 3, 4, 5, or 6-membered carbocycle or heterocycle; and
  • Q, T, U and V are independently C or N, and at least one of Q, T, U and V is N, wherein when Q, T, U or V is N, then there is no substituent at the N.
  • one or two of Q, T, U and V are nitrogen.
  • R 9 is H
  • R 8 or R 10 or both are independently OH; Cl; N 3 ; C 1-3 haloalkyl (preferably monofluoromethyl, difluoromethyl, trifluoromethyl); —XR 9a , where X is O or S, and R 9a is C 1-4 alkyl or C 1-3 haloalkyl (e.g., fluoroalkyl, preferably fluoromethyl, i.e., CH 2 F, CHF 2 , CF 3 ); —NH(R a ) or —N(R a )(R b ) where R a and R b are independently C 1-3 alkyl; or —COOR 9b , wherein R 9b is C 1-3 alkyl (preferably methyl or ethyl).
  • R 9 is H, R 8 or R 10 or both are independently N 3 , —OR 9a , wherein R 9a is C 1-4 alkyl or C 1-3 haloalkyl, or —N(R a )(R b ) where R a and R b are independently C 1-3 alkyl; or —COOR 9b , wherein R 9b is C 1-3 alkyl (preferably methyl or ethyl). Even more preferably when R 9 is H, R 8 or R 10 or both are C 1-3 alkoxy or C 1-3 halo alkoxy.
  • R 2 is not H and preferably R 2 is methyl, ethyl, Cl, F, fluoromethyl (CH 2 F, CHF 2 , CF 3 ), C 1-3 hydroxyalkyl (preferably CH 2 OH or CH 2 CH 2 OH), NH 2 , NH 2 OH, —NHCH 2 CH 2 OH, NHCH 3 , N(CH 3 ) 2 , N 3 , morpholino, OCH 3 , OC 2 H 5 , or SCH 3 .
  • R 9 is H
  • R 8 or R 10 or both are OCH 3 and preferably R 7 and R 11 are H
  • R 2 is not hydrogen and preferably R 2 is methyl or chloro.
  • R 9 is alkyl or haloalkyl or chloro
  • R 2 is not hydrogen and preferably R 2 is methyl or chloro.
  • compounds of the invention include compounds of Formula VIa or pharmaceutically acceptable salts or solvates thereof, wherein:
  • compounds of the invention include compounds of Formula VIa or pharmaceutically acceptable salts or solvates thereof, wherein:
  • the present invention provides compounds of Formula VIa or pharmaceutically acceptable salts or solvates thereof, wherein R 1 is CH 3 ; R 2 is Cl, methyl, or CH 2 F; R 3 is H, CH 3 , F, or Cl; R 4 , R 5 and R 6 are H; R 7 , R 5 , R 10 and R 11 are independently H or F; R 9 is —OCH 3 or —N(CH 3 ) 2 ; and Q, T, U and V are independently C or N, and at least one of Q, T, U and V is N, wherein when Q, T, U or V is N, then there is no substituent at the N. In some specific embodiments, one or two of Q, T, U and V are nitrogen.
  • R 9 is selected from the group consisting of H, C 1 , N 3 ;
  • C 1-6 alkyl (preferably C 1-3 alkyl, more preferably CH 3 ) optionally substituted with 1, 2 or 3 substituents, each substituent being independently OH, halo, C 1-3 alkoxy, (halo)C 1-3 alkoxy, —N(R a )(R b ) where R a and R b are independently H, OH (R a and R b are not both OH), C 2-4 hydroxyalkyl, or C 1-3 alkyl or R a and R b together with the nitrogen atom to which they are both linked form a 3, 4, 5 or 6-membered heterocycle;
  • R c is C 1-6 alkyl (preferably C 1-3 alkyl, more preferably CH 3 ) optionally substituted with 1, 2 or 3 substituents, each substituent being independently OH, halo, C 1-3 alkoxy, or (halo)C 1-3 alkoxy;
  • R d is an C 1-6 alkyl, preferably C 1-3 alkyl;
  • R a and R b are independently H, OH (R a and R b are not both OH), C 2-4 hydroxyalkyl, C 1-3 alkyl, or —N(R e )(R f ) where R e and R f are independently H, OH (R e and R f are not both OH), or C 1-3 alkyl; wherein optionally R a and R b together with the N form a 3, 4, 5 or 6-membered heterocycle, and optionally R e and R f together with the nitrogen atom to which they both are linked form a 3, 4, 5 or 6-membered heterocycle; or
  • R 9 is selected from such groups except R 9 is not H or chloro.
  • R 9 is H; OH; Cl; N 3 ; C 1-3 alkyl (preferably methyl; C 1-3 haloalkyl (preferably monofluoromethyl, difluoromethyl, trifluoromethyl); —OR 9a , wherein R 9a is C 1-4 alkyl or C 1-3 haloalkyl (e.g., fluoroalkyl, preferably fluoromethyl, i.e., CH 2 F, CHF 2 , CF 3 ); —NH(R a ) or —N(R a )(R b ) where R a and R b are independently C 1-3 alkyl; or —COOR 9b , wherein R 9b is C 1-3 alkyl (preferably methyl or ethyl); and optionally R 9 and one of R 8 and R 10 together form a 3, 4, 5, or 6-membered heterocycle.
  • R 9 is selected from such groups except R 9 is not H or chloro.
  • R 9 is N 3 ; —OR 9a , wherein R 9a is C 1-3 alkyl optionally substituted with 1-7 F; —N(R a )(R b ) where R a and R b are independently C 1-3 alkyl; or —COOR 9b where R 9b is C 1-3 alkyl.
  • R 9 is —OCH 3 , —OC 2 H 5 , —N(CH 3 ) 2 , —CO 2 CH 3 , —OCHF 2 , or N 3 .
  • R 9 is H, R 8 or R 10 or both are independently OH; N 3 ; —XR 9a , where X is O or S, and R 9a is C 1-4 alkyl or C 1-3 haloalkyl (e.g., fluoroalkyl, preferably fluoromethyl, i.e., CH 2 F, CHF 2 , CF 3 ); —NH(R a ) or —N(R a )(R b ) where R a and R b are independently C 1-3 alkyl; or —COOR 9b , wherein R 9b is C 1-3 alkyl (preferably methyl or ethyl).
  • R 9 is H, R 8 or R 10 or both are independently N 3 , —OR 9a , wherein R 9a is C 1-4 alkyl or C 1-3 haloalkyl, or —N(R a )(R b ) where R a and R b are independently C 1-3 alkyl; or —COOR 9b , wherein R 9b is C 1-3 alkyl (preferably methyl or ethyl). Even more preferably when R 9 is H, R 8 or R 10 or both are C 1-3 alkoxy or C 1-3 halo alkoxy.
  • R 9 is H
  • R 8 and R 10 are not H or one H and the other halo
  • R 2 is not H, and preferably R 2 is halo
  • N 3 C 1-3 alkyl optionally substituted with OH or halo (preferably F, e.g., monofluoro, difluoro, or trifluoro)
  • —XR 2a wherein X is S or O and R 2a is C 1-3 alkyl (more preferably CH 3 ) optionally substituted with halo (preferably F, e.g., monofluoro-, difluoro-, or trifluoro-substituted
  • R a and R b are independently H, OH (R a and R b are not both OH), C 1-3 alkyl (preferably CH 3 ), C 2-3 hydroxyalkyl.
  • R 2 is methyl, ethyl, Cl, F, fluoromethyl (CH 2 F, CHF 2 , CF 3 ), C 1-3 hydroxyalkyl (preferably CH 2 OH or CH 2 CH 2 OH), NH 2 , NH 2 OH, —NHCH 2 CH 2 OH, NHCH 3 , N(CH 3 ) 2 , N 3 , morpholino, OCH 3 , OC 2 H 5 , or SCH 3 .
  • R 9 is C 1-6 alkyl, halo, or C 1-6 haloalkyl
  • R 2 is not H, and preferably R 2 is halo; N 3 , C 1-3 alkyl optionally substituted with OH or halo (preferably F, e.g., monofluoro, difluoro, or trifluoro); —XR 2a wherein X is S or O and R 2a is C 1-3 alkyl (more preferably CH 3 ) optionally substituted with halo (preferably F, e.g., monofluoro-, difluoro-, or trifluoro-substituted); or —N(R a )(R b ) wherein R a and R b are independently H, OH (R a and R b are not both OH), C 1-3 alkyl (preferably CH 3 ), C 2-3 hydroxyalkyl.
  • R 2 is methyl, ethyl, Cl, F, fluoromethyl (CH 2 F, CHF 2 , CF 3 ), C 1-3 hydroxyalkyl (preferably CH 2 OH or CH 2 CH 2 OH), NH 2 , NH 2 OH, —NHCH 2 CH 2 OH, NHCH 3 , N(CH 3 ) 2 , N 3 , morpholino, OCH 3 , OC 2 H 5 , or SCH 3 .
  • R 2 is H; halo; N 3 ; C 1-6 alkyl (preferably C 1-3 alkyl, more preferably CH 3 ) optionally substituted with OH or halo (preferably F, e.g., monofluoro, difluoro, or trifluoro); —XR 2a wherein X is S or O, and R 2a is C 1-6 alkyl (preferably C 1-3 alkyl, more preferably CH 3 ) optionally substituted with OH or halo (preferably F, e.g., monofluoro-, difluoro-, or trifluoro-substituted); —CO 2 R d , wherein R d is C 1-3 alkyl, preferably methyl or ethyl; or —N(R a )(R b ) wherein R a and R b are independently H, OH (R a and R b are not both OH), C 1-3 alkyl (preferably CH 3 ), C
  • R 2 is H; halo; C 1-3 alkyl optionally substituted with OH or halo (preferably F, e.g., monofluoro, difluoro, or trifluoro); —XR 2a wherein X is S or O and R 2a is C 1-3 alkyl (preferably C 1-3 alkyl, more preferably CH 3 ) optionally substituted with halo (preferably F, e.g., monofluoro-, difluoro-, or trifluoro-substituted); or —N(R a )(R b ) wherein R a and R b are independently H, OH (R a and R b are not both OH), C 1-3 alkyl (preferably CH 3 ), C 2-3 hydroxyalkyl.
  • R 2 is H, methyl, ethyl, Cl, F, fluoromethyl (CH 2 F, CHF 2 , CF 3 ), C 1-3 hydroxyalkyl (preferably CH 2 OH or CH 2 CH 2 OH), NH 2 , NH 2 OH, —NHCH 2 CH 2 OH, NHCH 3 , N(CH 3 ) 2 , N 3 , morpholino, OCH 3 , OC 2 H 5 , or SCH 3 .
  • R 2 is H, methyl, Cl, —CH 2 OH, —NH 2 , —NHCH 3 , —NHCH 2 CH 2 OH, —OCH 3 , —SCH 3 , or —CH 2 F.
  • compounds of the invention include compounds of Formula VIb or pharmaceutically acceptable salts or solvates thereof, wherein:
  • C 1-3 alkyl preferably CH 3
  • C 1-3 alkoxy preferably OCH 3
  • C 1-3 haloalkyl preferably monofluoromethyl, difluoromethyl, trifluoromethyl
  • —XR 9a where X is O or S, and R 9a is C 1-4 alkyl or C 1-3 haloalkyl (e.g., fluoroalkyl, preferably fluoromethyl, i.e., CH 2 F, CHF 2 , CF 3 ); —NH(R a ) or —N(R 3 )(R a ) where R a and R b are independently C 1-3 alkyl; or —COOR 9b , wherein R 9b is C 1-3 alkyl (preferably methyl or ethyl). and
  • R 9 is H, R 8 or R 10 or both are independently OH; N 3 ; —XR 9a , where X is O or S, and R 9a is C 1-4 alkyl or C 1-3 haloalkyl (e.g., fluoroalkyl, preferably fluoromethyl, i.e., CH 2 F, CHF 2 , CF 3 ); —NH(R a ) or —N(R a )(R b ) where R a and R b are independently C 1-3 alkyl; or —COOR 9b , wherein R 9b is C 1-3 alkyl (preferably methyl or ethyl).
  • R 9 is H, R 8 or R 10 or both are independently N 3 , —OR 9a , wherein R 9a is C 1-4 alkyl or C 1-3 haloalkyl, or —N(R a )(R b ) where R a and R b are independently C 1-3 alkyl; or —COOR 9b , wherein R 9b is C 1-3 alkyl (preferably methyl or ethyl). Even more preferably when R 9 is H, R 8 or R 10 or both are C 1-3 alkoxy or C 1-3 halo alkoxy.
  • R 2 is not H and preferably R 2 is methyl, ethyl, Cl, F, fluoromethyl (CH 2 F, CHF 2 , CF 3 ), C 1-3 hydroxyalkyl (preferably CH 2 OH or CH 2 CH 2 OH), NH 2 , NH 2 OH, —NHCH 2 CH 2 OH, NHCH 3 , N(CH 3 ) 2 , N 3 , morpholino, OCH 3 , OC 2 H 5 , or SCH 3 .
  • compounds of the invention include compounds of Formula VIb or pharmaceutically acceptable salts or solvates thereof, wherein:
  • R 9 is H
  • R 8 or R 10 or both are independently —XR 9a , where X is O or S
  • R 9a is C 1-4 alkyl or C 1-3 haloalkyl (e.g., fluoroalkyl, preferably fluoromethyl, i.e., CH 2 F, CHF 2 , CF 3 ); —N(R a )(R b ) where R a and R b are independently C 1-3 alkyl; or —COOR 9b , wherein R 9b is C 1-3 alkyl (preferably methyl or ethyl).
  • R 9 is H, R 8 or R 10 or both are independently N 3 , —OR 9a , wherein R 9a is C 1-4 alkyl or C 1-3 haloalkyl, or —N(R a )(R b ) where R a and R b are independently C 1-3 alkyl; or —COOR 9b , wherein R 9b is C 1-3 alkyl (preferably methyl or ethyl). Even more preferably when R 9 is H, R 8 or R 10 or both are C 1-3 alkoxy or C 1-3 halo alkoxy.
  • R 9 is H then R 8 and R 10 are not both H or one H and the other halo, and R 2 is not H.
  • R 2 is not H and preferably R 2 is methyl, ethyl, Cl, F, fluoromethyl (CH 2 F, CHF 2 , CF 3 ), C 1-3 hydroxyalkyl (preferably CH 2 OH or CH 2 CH 2 OH), NH 2 , NH 2 OH, —NHCH 2 CH 2 OH, NHCH 3 , N(CH 3 ) 2 , N 3 , morpholino, OCH 3 , OC 2 H 5 , or SCH 3 .
  • R 9 is H
  • R 8 or R 10 or both are OCH 3 and preferably R 7 and R 11 are H
  • R 2 is not hydrogen and preferably R 2 is methyl or chloro.
  • R 9 is alkyl or haloalkyl or chloro
  • R 2 is not hydrogen and preferably R 2 is methyl or chloro.
  • compounds of the invention include compounds of Formula VIb or pharmaceutically acceptable salts or solvates thereof, wherein:
  • R 9 is H
  • R 8 or R 10 or both are independently C 1-3 alkoxy (preferably OCH 3 ) or C 1-3 alkylthiol, each being optionally substituted with 1-4 F. Even more preferably when R 9 is H, R 8 or R 10 or both are methoxy or ethoxy. Also preferably R 2 is not H.
  • R 2 is not H, preferably R 2 is methyl, ethyl, Cl, F, fluoromethyl (CH 2 F, CHF 2 , CF 3 ), C 1-3 hydroxyalkyl (preferably CH 2 OH or CH 2 CH 2 OH), NH 2 , NH 2 OH, —NHCH 2 CH 2 OH, NHCH 3 , N(CH 3 ) 2 , N 3 , morpholino, OCH 3 , OC 2 H 5 , or SCH 3
  • R 2 is methyl, ethyl, Cl, F, fluoromethyl (CH 2 F, CHF 2 , CF 3 ), C 1-3 hydroxyalkyl (preferably CH 2 OH or CH 2 CH 2 OH), NH 2 , NH 2 OH, —NHCH 2 CH 2 OH, NHCH 3 , N(CH 3 ) 2 , N 3 , morpholino, OCH 3 , OC 2 H 5 , or SCH 3
  • R 9 is H
  • R 8 or R 10 or both are OCH 3 and preferably R 7 and R 11 are H
  • R 2 is not hydrogen and preferably R 2 is methyl or chloro.
  • R 9 is alkyl or haloalkyl or chloro
  • R 2 is not hydrogen and preferably R 2 is methyl or chloro.
  • compounds of the invention include compounds of Formula VIb or pharmaceutically acceptable salts or solvates thereof, wherein:
  • R 9 is H
  • R 8 or R 10 or both are independently C 1-3 alkoxy (preferably OCH 3 ) or C 1-3 alkylthiol, each being optionally substituted with 1-4 F. Even more preferably when R 9 is H, R 8 or R 10 or both are methoxy or ethoxy. Also preferably R 2 is not H.
  • R 2 is not H, preferably R 2 is methyl, ethyl, Cl, F, fluoromethyl (CH 2 F, CHF 2 , CF 3 ), C 1-3 hydroxyalkyl (preferably CH 2 OH or CH 2 CH 2 OH), NH 2 , NH 2 OH, —NHCH 2 CH 2 OH, NHCH 3 , N(CH 3 ) 2 , N 3 , morpholino, OCH 3 , OC 2 H 5 , or SCH 3 .
  • R 9 is H
  • R 8 or R 10 or both are OCH 3 and preferably R 7 and R 11 are H
  • R 2 is not hydrogen and preferably R 2 is methyl or chloro.
  • R 9 is alkyl or haloalkyl or chloro
  • R 2 is not hydrogen and preferably R 2 is methyl or chloro.
  • compounds of the invention include compounds of Formula VIb or pharmaceutically acceptable salts or solvates thereof, wherein:
  • R 12 is selected from the group of methyl, ethyl, fluoromethyl (e.g., CH 2 F, CHF 2 , CF 3 ), and fluoroethyl;
  • compounds of the invention include compounds of Formula VIb or pharmaceutically acceptable salts or solvates thereof, wherein:
  • the present invention provides compounds of Formula VIb or pharmaceutically acceptable salts or solvates thereof, wherein R 1 is CH 3 ; R 2 is Cl, methyl, or CH 2 F; R 3 is H, CH 3 , F, or Cl; R 4 , R 5 and R 6 are H; R 7 , R 8 , R 10 and R 11 are independently H or F; and R 9 is —OCH 3 or —N(CH 3 ) 2 .
  • compounds of Formula VIb include compounds wherein:
  • the compounds of this embodiment are not selected form the group consisting of:
  • compounds of Formula VIb include compounds wherein:
  • the compounds of the present invention as disclosed above, preferred are those that can induce caspase activation as determined by the method and under conditions (measurement at 24 hours) described in Example 143, preferably at an EC 50 of no greater than about 1,000 nM, more preferably at an EC 50 of no greater than about 500 nM, more preferably at an EC 50 of no greater than about 200 nM, even more preferably at an EC 50 of no greater than about 100 nM, and most preferably at an EC 50 of no greater than about 10 nM.
  • Also preferred compounds are those of Formula I-VIb, and pharmaceutically acceptable salts or solvates thereof, that are able to inhibit tubulin at an IC 50 of no greater than about 2,000 nM, preferably no greater than about 1,000 nM, more preferably less than about 500 nM, as determined by the method and under conditions described in Example 145.
  • Exemplary compounds of the present invention are compounds provided in Examples 1-142; and pharmaceutically acceptable salts or prodrugs thereof, including but not limited to:
  • exemplary compounds of the invention include: (2-Chloro-quinazolin-4-yl)-(6-methoxy-pyridin-3-yl)-methyl-amine;
  • exemplary compounds of the invention include: (4-Methoxy-phenyl)-(2-methyl-pyrido[2,3-d]pyrimidin-4-yl)-methyl-amine; and
  • exemplary compounds of the invention include:
  • exemplary compounds of the invention include:
  • exemplary compounds of the invention include: (2-Chloro-quinazolin-4-yl)-(4-methoxy-phenyl)-methyl-amine;
  • exemplary compounds of the invention include: (4-Methoxy-phenyl)-methyl-quinazolin-4-yl-amine; and
  • exemplary compounds useful in the methods of the invention include:
  • alkyl as employed herein by itself or as part of another group refers to both straight and branched chain radicals of up to ten carbons.
  • Useful alkyl groups include straight-chained and branched C 1-10 alkyl groups, more preferably C 1-6 alkyl groups.
  • Typical C 1-10 alkyl groups include methyl, ethyl, propyl, isopropyl, butyl, sec-butyl, tert-butyl, 3-pentyl, hexyl and octyl groups, which may be optionally substituted.
  • alkenyl as employed herein by itself or as part of another group means a straight or branched chain radical of 2-10 carbon atoms, unless the chain length is limited thereto, including at least one double bond between two of the carbon atoms in the chain.
  • Typical alkenyl groups include ethenyl, 1-propenyl, 2-propenyl, 2-methyl-1-propenyl, 1-butenyl and 2-butenyl.
  • alkynyl is used herein to mean a straight or branched chain radical of 2-10 carbon atoms, unless the chain length is limited thereto, wherein there is at least one triple bond between two of the carbon atoms in the chain.
  • Typical alkynyl groups include ethynyl, 1-propynyl, 1-methyl-2-propynyl, 2-propynyl, 1-butynyl and 2-butynyl.
  • Useful alkoxy groups include oxygen substituted by one of the C 1-10 alkyl groups mentioned above, which may be optionally substituted.
  • Alkoxy substituents include, without limitation, halo, morpholino, amino including alkylamino and dialkylamino, and carboxy including esters thereof.
  • Useful alkylthio groups include sulfur substituted by one of the C 1-10 alkyl groups mentioned above, which may be optionally substituted. Also included are the sulfoxides and sulfones of such alkylthio groups.
  • Useful amino groups include —NH 2 , —NHR 18 and —NR 18 R 19 , wherein R 18 and R 18 are C 1-10 alkyl or cycloalkyl groups, or R 18 and R 19 are combined with the N to form a ring structure, such as a piperidine, or R 18 and R 19 are combined with the N and other group to form a ring, such as a piperazine.
  • the alkyl group may be optionally substituted.
  • Optional substituents on the alkyl, alkenyl, alkynyl, cycloalkyl, carbocyclic and heterocyclic groups include one or more halo, hydroxy, carboxyl, amino, nitro, cyano, C 1 -C 6 acylamino, C 1 -C 6 acyloxy, C 1 -C 6 alkoxy, aryloxy, alkylthio, C 6 -C 10 aryl, C 4 -C 7 cycloalkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 6 -C 10 aryl(C 2 -C 6 )alkenyl, C 6 -C 10 aryl(C 2 -C 6 )alkynyl, saturated and unsaturated heterocyclic or heteroaryl.
  • Optional substituents on the aryl, arylalkyl, arylalkenyl, arylalkynyl and heteroaryl and heteroarylalkyl groups include one or more halo, C 1 -C 6 haloalkyl, C 6 -C 10 aryl, C 4 -C 7 cycloalkyl, C 1 -C 6 alkyl, C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 6 -C 10 aryl(C 1 -C 6 )alkyl, C 6 -C 10 aryl(C 2 -C 6 )alkenyl, C 6 -C 10 aryl(C 2 -C 6 )alkynyl, C 1 -C 6 hydroxyalkyl, nitro, amino, ureido, cyano, C 1 -C 6 acylamino, hydroxy, thiol, C 1 -C 6 acyloxy, azido,
  • aryl as employed herein by itself or as part of another group refers to monocyclic, bicyclic or tricyclic aromatic groups containing from 6 to 14 carbons in the ring portion.
  • Useful aryl groups include C 6-14 aryl, preferably C 6-10 aryl.
  • Typical C 6-14 aryl groups include phenyl, naphthyl, phenanthrenyl, anthracenyl, indenyl, azulenyl, biphenyl, biphenylenyl and fluorenyl groups.
  • Carbocycle as employed herein include cycloalkyl and partially saturated carbocyclic groups.
  • Useful cycloalkyl groups are C 3-8 cycloalkyl.
  • Typical cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl.
  • Useful saturated or partially saturated carbocyclic groups are cycloalkyl groups as described above, as well as cycloalkenyl groups, such as cyclopentenyl, cycloheptenyl and cyclooctenyl.
  • Useful halo or halogen groups include fluorine, chlorine, bromine and iodine.
  • arylalkyl is used herein to mean any of the above-mentioned C 1-10 alkyl groups substituted by any of the above-mentioned C 6-14 aryl groups.
  • the arylalkyl group is benzyl, phenethyl or naphthylmethyl.
  • arylalkenyl is used herein to mean any of the above-mentioned C 2-10 alkenyl groups substituted by any of the above-mentioned C 6-14 aryl groups.
  • arylalkynyl is used herein to mean any of the above-mentioned C 2-10 alkynyl groups substituted by any of the above-mentioned C 6-14 aryl groups.
  • aryloxy is used herein to mean oxygen substituted by one of the above-mentioned C 6-14 aryl groups, which may be optionally substituted.
  • Useful aryloxy groups include phenoxy and 4-methylphenoxy.
  • arylalkoxy is used herein to mean any of the above mentioned C 1-10 alkoxy groups substituted by any of the above-mentioned aryl groups, which may be optionally substituted.
  • Useful arylalkoxy groups include benzyloxy and phenethyloxy.
  • Useful haloalkyl groups include C 1-10 alkyl groups substituted by one or more fluorine, chlorine, bromine or iodine atoms, e.g., fluoromethyl, difluoromethyl, trifluoromethyl, pentafluoroethyl, 1,1-difluoroethyl, chloromethyl, chlorofluoromethyl and trichloromethyl groups.
  • acylamino (acylamido) groups are any C 1-6 acyl (alkanoyl) attached to an amino nitrogen, e.g., acetamido, chloroacetamido, propionamido, butanoylamido, pentanoylamido and hexanoylamido, as well as aryl-substituted C 1-6 acylamino groups, e.g., benzoylamido, and pentafluorobenzoylamido.
  • acyloxy groups are any C 1-6 acyl (alkanoyl) attached to an oxy (—O—) group, e.g., formyloxy, acetoxy, propionoyloxy, butanoyloxy, pentanoyloxy and hexanoyloxy.
  • heterocycle is used herein to mean a saturated or partially saturated 3-7 membered monocyclic, or 7-10 membered bicyclic ring system, which consists of carbon atoms and from one to four heteroatoms independently selected from the group consisting of O, N, and S, wherein the nitrogen and sulfur heteroatoms can be optionally oxidized, the nitrogen can be optionally quaternized, and including any bicyclic group in which any of the above-defined heterocyclic rings is fused to a benzene ring, and wherein the heterocyclic ring can be substituted on carbon or on a nitrogen atom if the resulting compound is stable.
  • Useful saturated or partially saturated heterocyclic groups include tetrahydrofuranyl, pyranyl, piperidinyl, piperazinyl, pyrrolidinyl, imidazolidinyl, imidazolinyl, indolinyl, isoindolinyl, quinuclidinyl, morpholinyl, isochromanyl, chromanyl, pyrazolidinyl, pyrazolinyl, tetronoyl and tetramoyl groups.
  • heteroaryl refers to groups having 5 to 14 ring atoms; 6, 10 or 14 ⁇ electrons shared in a cyclic array; and containing carbon atoms and 1, 2 or 3 oxygen, nitrogen or sulfur heteroatoms.
  • Useful heteroaryl groups include thienyl (thiophenyl), benzo[b]thienyl, naphtho[2,3-b]thienyl, thianthrenyl, furyl (furanyl), isobenzofuranyl, chromenyl, xanthenyl, phenoxanthiinyl, pyrrolyl, including without limitation 2H-pyrrolyl, imidazolyl, pyrazolyl, pyridyl (pyridinyl), including without limitation 2-pyridyl, 3-pyridyl, and 4-pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, indolizinyl, isoindolyl, 3H-indolyl, indolyl, indazolyl, purinyl, 4H-quinolizinyl, isoquinolyl, quinolyl, phthalzinyl, naphthyridinyl,
  • heteroaryl group contains a nitrogen atom in a ring
  • nitrogen atom may be in the form of an N-oxide, e.g., a pyridyl N-oxide, pyrazinyl N-oxide and pyrimidinyl N-oxide.
  • heteroaryloxy is used herein to mean oxygen substituted by one of the above-mentioned heteroaryl groups, which may be optionally substituted.
  • Useful heteroaryloxy groups include pyridyloxy, pyrazinyloxy, pyrrolyloxy, pyrazolyloxy, imidazolyloxy and thiophenyloxy.
  • heteroarylalkoxy is used herein to mean any of the above-mentioned C 1-10 alkoxy groups substituted by any of the above-mentioned heteroaryl groups, which may be optionally substituted.
  • stereoisomers including optical isomers.
  • the invention includes all stereoisomers and both the racemic mixtures of such stereoisomers as well as the individual enantiomers that may be separated according to methods that are well known to those of ordinary skill in the art.
  • addition salts include inorganic and organic acid addition salts, such as hydrochloride, hydrobromide, phosphate, sulphate, citrate, lactate, tartrate, maleate, fumarate, mandelate and oxalate; and inorganic and organic base addition salts with bases, such as sodium hydroxy, Tris(hydroxymethyl)aminomethane (TRIS, tromethane) and N-methyl-glucamine.
  • inorganic and organic acid addition salts such as hydrochloride, hydrobromide, phosphate, sulphate, citrate, lactate, tartrate, maleate, fumarate, mandelate and oxalate
  • bases such as sodium hydroxy, Tris(hydroxymethyl)aminomethane (TRIS, tromethane) and N-methyl-glucamine.
  • prodrugs of the compounds of the invention include the simple esters of carboxylic acid containing compounds (e.g., those obtained by condensation with a C 1-4 alcohol according to methods known in the art); esters of hydroxy containing compounds (e.g., those obtained by condensation with a C 1-4 carboxylic acid, C 3-6 dioic acid or anhydride thereof, such as succinic and fumaric anhydrides according to methods known in the art); imines of amino containing compounds (e.g., those obtained by condensation with a C 1-4 aldehyde or ketone according to methods known in the art); carbamate of amino containing compounds, such as those described by Leu, et. al., ( J. Med. Chem.
  • the compounds of this invention may be prepared using methods known to those skilled in the art, or the novel methods of this invention.
  • the compounds of this invention with Formulae I-VIb can be prepared as illustrated by the exemplary reaction in Scheme 1. Reaction of optionally substituted quinazoline-2,4-dione with phosphorylchloride produces the corresponding 2,4-dichloroquinazoline, which is reacted with an optionally substituted aniline, such as N-methyl-4-methoxy-aniline, to produce the substituted 2-chloro-4-anilino-quinazoline.
  • an optionally substituted aniline such as N-methyl-4-methoxy-aniline
  • N-alkyl-arylamine or N-alkyl-heteroarylamine could be prepared by reaction of the arylamine or heteroarylamine with a ketone or aldehyde, such as acetone, in the presence of a reducing agent, such as NaCNBH 3 .
  • a reducing agent such as NaCNBH 3 .
  • the N-alkyl-arylamine or N-alkyl-heteroarylamine is then reacted with optionally substituted 2,4-dichloroquinazoline to produce the corresponding 4-substituted 2-chloro-quinazoline.
  • 2-substituted 4-chloro-quinazoline such as 4-chloro-2-fluoromethyl-quinazoline
  • a substituted aniline such as N-methyl-4-methoxy-aniline
  • 2-substituted 4-anilino-quinazoline such as 2-fluoromethyl-4-anilino-quinazoline
  • substituted acetonitriles that can be used for the reaction include chloro-acetonitrile and bromo-acetonitrile, as well as acetonitrile and propionitrile.
  • the compound is then converted to the corresponding 4-chloro-quinazoline, such as 4-chloro-2-methyl-6-nitro-quinazoline by reaction with phosphorylchloride.
  • Reaction of the 4-chloro-quinazoline, such as 4-chloro-2-methyl-6-nitro-quinazoline with a substituted arylamine or heteroarylamine, such as N-methyl-4-methoxy-aniline produces the corresponding 4-(arylamino or heteroarylamino)-quinazoline, such as substituted 2-methyl-6-nitro-4-anilino-quinazoline.
  • Other substituted 2-amino-benzoic acid that can be used for the reaction include 2-amino-4-nitro-benzoic acid, 2-amino-5-chloro-benzoic acid.
  • the resulting compound is then converted to the corresponding 4-chloro-2-methyl-pyrido[2,3-d](heteroaryl or heterocycle), such as 4-chloro-2-methyl-pyrido[2,3-d]pyrimidine by reaction with phosphorylchloride, which is treated with an optionally substituted arylamino or heteroarylamino, such as N-methyl-4-methoxy-aniline to produce the corresponding optionally substituted 4-(arylamino or heteroarylamino)-2-methyl-pyrido[2,3-d](heteroaryl or heterocycle), such as substituted 4-anilino-2-methyl-pyrido[2,3-d]pyrimidine.
  • An important aspect of the present invention is the discovery that compounds having Formulae I-VIb are activators of caspases and inducers of apoptosis.
  • Another important aspect of the invention is the discovery that compounds having Formulae I-Vc are inhibitors of tubulin polymerization. Therefore, these compounds are useful in treating diseases that are responsive to activating caspases, inducing apoptosis, or inhibiting tubulin. For example, these compounds are useful in a variety of clinical conditions in which there is uncontrolled cell growth and spread of abnormal cells, such as in the case of cancer.
  • the present invention also includes a therapeutic method comprising administering to an animal an effective amount of a compound, or a pharmaceutically acceptable salt or prodrug of said compound of Formulae I-Vc, wherein said therapeutic method is useful to treat cancer, which is a group of diseases characterized by the uncontrolled growth and spread of abnormal cells.
  • Such diseases include, but are not limited to, Hodgkin's disease, non-Hodgkin's lymphoma, acute lymphocytic leukemia, chronic lymphocytic leukemia, multiple myeloma, neuroblastoma, breast carcinoma, ovarian carcinoma, lung carcinoma, Wilms' tumor, cervical carcinoma, testicular carcinoma, soft-tissue sarcoma, primary macroglobulinemia, bladder carcinoma, chronic granulocytic leukemia, primary brain carcinoma, malignant melanoma, small-cell lung carcinoma, stomach carcinoma, colon carcinoma, malignant pancreatic insulinoma, malignant carcinoid carcinoma, choriocarcinoma, mycosis fungoides, head or neck carcinoma, osteogenic sarcoma, pancreatic carcinoma, acute granulocytic leukemia, hairy cell leukemia, neuroblastoma, rhabdomyosarcoma, Kaposi's sarcoma, genitourinary carcinoma, thyroid carcinoma,
  • compositions containing therapeutically effective concentrations of the compounds formulated for oral, intravenous, local and topical application, for the treatment of neoplastic diseases and other diseases are administered to an individual exhibiting the symptoms of one or more of these disorders.
  • the amounts are effective to ameliorate or eliminate one or more symptoms of the disorders.
  • An effective amount of a compound for treating a particular disease is an amount that is sufficient to ameliorate, or in some manner reduce, the symptoms associated with the disease.
  • Such amount may be administered as a single dosage or may be administered according to a regimen, whereby it is effective.
  • the amount may cure the disease but, typically, is administered in order to ameliorate the symptoms of the disease. Typically, repeated administration is required to achieve the desired amelioration of symptoms.
  • Another aspect of the present invention is to provide a pharmaceutical composition, containing an effective amount of a compound of Formulae I-VIb, or a pharmaceutically acceptable salt of said compound, in admixture with one or more pharmaceutically acceptable carriers or diluents.
  • a pharmaceutical composition comprising a compound of Formulae I-Vc disclosed herein, or a pharmaceutically acceptable salt of said compound, in combination with a pharmaceutically acceptable vehicle is provided.
  • Preferred pharmaceutical compositions comprise compounds of Formulae I-VIb, and pharmaceutically acceptable salts, esters, or prodrugs thereof, that are able to induce caspase activation as determined by the method described in Example 145, preferably at an EC 50 no greater than 1,000 nM, more preferably at an EC 50 no greater than 500 nM, more preferably at an EC 50 no greater than 200 nM, more preferably at an EC 50 no greater than 100, and most preferably at an EC 50 no greater than 10 nM.
  • Other preferred compositions comprise compounds of Formula I-VIb, and pharmaceutically acceptable salts, esters, or prodrugs thereof, that are able to inhibit tubulin polymerization as determined by the method described in Example 147.
  • Another embodiment of the present invention is directed to a composition effective to inhibit neoplasia comprising a compound, or a pharmaceutically acceptable salt or prodrug of said compound of Formulae I-VIb, which functions as a caspase cascade activator and inducer of apoptosis or inhibitor of tubulin polymerization, in combination with at least one known cancer chemotherapeutic agent, or a pharmaceutically acceptable salt of said agent.
  • cancer chemotherapeutic agents which may be used for combination therapy include, but not are limited to alkylating agents, such as busulfan, cis-platin, mitomycin C, and carboplatin; antimitotic agents, such as colchicine, vinblastine, paclitaxel, and docetaxel; topo I inhibitors, such as camptothecin and topotecan; topo II inhibitors, such as doxorubicin and etoposide; RNA/DNA antimetabolites, such as 5-azacytidine, 5-fluorouracil and methotrexate; DNA antimetabolites, such as 5-fluoro-2′-deoxy-uridine, ara-C, hydroxyurea and thioguanine; EGFR inhibitors, such as Iressa® (gefitinib) and Tarceva® (erlotinib); proteosome inhibitors; antibodies, such as campath, Herceptin® (trastuzumab), Avastin® (bevacizum), anti
  • cancer chemotherapeutic agents which may be used for combination therapy include melphalan, chlorambucil, cyclophosamide, ifosfamide, vincristine, mitoguazone, epirubicin, aclarubicin, bleomycin, mitoxantrone, elliptinium, fludarabine, octreotide, retinoic acid, tamoxifen, Gleevec® (imatinib mesylate) and alanosine.
  • the compound of the invention may be administered together with at least one known chemotherapeutic agent as part of a unitary pharmaceutical composition.
  • the compound of the invention may be administered apart from at least one known cancer chemotherapeutic agent.
  • the compound of the invention and at least one known cancer chemotherapeutic agent are administered substantially simultaneously, i.e. the compounds are administered at the same time or one after the other, so long as the compounds reach therapeutic levels in the blood at the same time.
  • the compound of the invention and at least one known cancer chemotherapeutic agent are administered according to their individual dose schedule, so long as the compounds reach therapeutic levels in the blood.
  • alpha-1-adrenoceptor antagonists such as doxazosin, terazosin, and tamsulosin can inhibit the growth of prostate cancer cell via induction of apoptosis (Kyprianou, N., et al., Cancer Res 60:4550-4555, (2000)).
  • another embodiment of the present invention is directed to a composition effective to inhibit neoplasia comprising a compound, or a pharmaceutically acceptable salt or prodrug of a compound described herein, which functions as a caspase cascade activator and inducer of apoptosis or inhibitor of tubulin polymerization, in combination with at least one known alpha-1-adrenoceptor antagonists, or a pharmaceutically acceptable salt of said agent.
  • known alpha-1-adrenoceptor antagonists which can be used for combination therapy include, but are not limited to, doxazosin, terazosin, and tamsulosin.
  • sigma-2 receptors are expressed in high densities in a variety of tumor cell types (Vilner, B. J., et al., Cancer Res. 55: 408-413 (1995)) and that sigma-2 receptor agonists, such as CB-64D, CB-184 and haloperidol activate a novel apoptotic pathway and potentiate antineoplastic drugs in breast tumor cell lines. (Kyprianou, N., et al., Cancer Res. 62:313-322 (2002)).
  • another embodiment of the present invention is directed to a composition effective to inhibit neoplasia comprising a compound, or a pharmaceutically acceptable salt or prodrug of a compound described herein, which functions as a caspase cascade activator and inducer of apoptosis or inhibitor of tubulin polymerization, in combination with at least one known sigma-2 receptor agonist, or a pharmaceutically acceptable salt of said agonist.
  • known sigma-2 receptor agonists which can be used for combination therapy include, but are not limited to, CB-64D, CB-184 and haloperidol.
  • another embodiment of the present invention is directed to a composition effective to inhibit neoplasia comprising a compound, or a pharmaceutically acceptable salt or prodrug of a compound described herein, which functions as a caspase cascade activator and inducer of apoptosis or inhibitor of tubulin polymerization, in combination with at least one known HMG-CoA reductase inhibitor, or a pharmaceutically acceptable salt of said agent.
  • known HMG-CoA reductase inhibitors which can be used for combination therapy include, but are not limited to, lovastatin, simvastatin, pravastatin, fluvastatin, atorvastatin and cerivastatin.
  • another embodiment of the present invention is directed to a composition effective to inhibit neoplasia comprising a compound, or a pharmaceutically acceptable salt or prodrug of a compound described herein, which functions as a caspase cascade activator and inducer of apoptosis or inhibitor of tubulin polymerization, in combination with at least one known HIV protease inhibitor, or a pharmaceutically acceptable salt of said agent.
  • HIV protease inhibitors which can be used for combination therapy include, but are not limited to, amprenavir, abacavir, CGP-73547, CGP-61755, DMP-450, indinavir, nelfinavir, tipranavir, ritonavir, saquinavir, ABT-378, AG 1776, and BMS-232,632.
  • retinoids such as fenretinide (N-(4-hydroxyphenyl)retinamide, 4HPR)
  • fenretinide N-(4-hydroxyphenyl)retinamide
  • 4HPR also was reported to have good activity in combination with gamma-radiation on bladder cancer cell lines (Zou, C., et al, Int. J. Oncol. 13:1037-1041 (1998)).
  • another embodiment of the present invention is directed to a composition effective to inhibit neoplasia comprising a compound, or a pharmaceutically acceptable salt or prodrug of a compound described herein, which functions as a caspase cascade activator and inducer of apoptosis or inhibitor of tubulin polymerization, in combination with at least one known retinoid and synthetic retinoid, or a pharmaceutically acceptable salt of said agent.
  • retinoids and synthetic retinoids which can be used for combination therapy include, but are not limited to, bexarotene, tretinoin, 13-cis-retinoic acid, 9-cis-retinoic acid, ⁇ -difluoromethylornithine, ILX23-7553, fenretinide, and N-4-carboxyphenyl retinamide.
  • proteasome inhibitors such as lactacystin
  • lactacystin exert anti-tumor activity in vivo and in tumor cells in vitro, including those resistant to conventional chemotherapeutic agents.
  • proteasome inhibitors may also prevent angiogenesis and metastasis in vivo and further increase the sensitivity of cancer cells to apoptosis (Almond, J. B., et al., Leukemia 16:433-443 (2002)).
  • another embodiment of the present invention is directed to a composition effective to inhibit neoplasia comprising a compound, or a pharmaceutically acceptable salt or prodrug of a compound described herein, which functions as a caspase cascade activator and inducer of apoptosis or inhibitor of tubulin polymerization, in combination with at least one known proteasome inhibitor, or a pharmaceutically acceptable salt of said agent.
  • known proteasome inhibitors which can be used for combination therapy include, but are not limited to, lactacystin, MG-132, and PS-341.
  • tyrosine kinase inhibitors such as STI571 (Gleevec® (imatinib mesylate)
  • STI571 Galeevec® (imatinib mesylate)
  • etoposide Liu, W. M., et al. Br. J. Cancer 86:1472-1478 (2002).
  • another embodiment of the present invention is directed to a composition effective to inhibit neoplasia comprising a compound, or a pharmaceutically acceptable salt or prodrug of a compound described herein, which functions as a caspase cascade activator and inducer of apoptosis or inhibitor of tubulin polymerization, in combination with at least one known tyrosine kinase inhibitor, or a pharmaceutically acceptable salt of said agent.
  • tyrosine kinase inhibitors which can be used for combination therapy include, but are not limited to, Gleevec® (imatinib mesylate), ZD1839 Iressa® (gefitinib), SH268, genistein, CEP2563, SU6668, SU11248, and EMD121974.
  • prenyl-protein transferase inhibitors such as farnesyl protein transferase inhibitor R15777
  • preclinical antitumor activity against human breast cancer Kelland, L. R., et. al., Clin. Cancer Res. 7:3544-3550 (2001)
  • Synergy of the protein farnesyltransferase inhibitor SCH66336 and cisplatin in human cancer cell lines also has been reported (Adjei, A. A., et al., Clin. Cancer. Res. 7:1438-1445 (2001)).
  • another embodiment of the present invention is directed to a composition effective to inhibit neoplasia comprising a compound, or a pharmaceutically acceptable salt or prodrug of a compound described herein, which functions as a caspase cascade activator and inducer of apoptosis, in combination with at least one known prenyl-protein transferase inhibitor, including farnesyl protein transferase inhibitor, inhibitors of geranylgeranyl-protein transferase type I (GGPTase-I) and geranylgeranyl-protein transferase type-II, or a pharmaceutically acceptable salt of said agent.
  • known prenyl-protein transferase inhibitors which can be used for combination therapy include, but are not limited to, R15777, SCH66336, L-778,123, BAL9611 and TAN-1813.
  • CDK cyclin-dependent kinase
  • another embodiment of the present invention is directed to a composition effective to inhibit neoplasia comprising a compound, or a pharmaceutically acceptable salt or prodrug of a compound described herein, which functions as a caspase cascade activator and inducer of apoptosis or inhibitor of tubulin polymerization, in combination with at least one known cyclin-dependent kinase inhibitor, or a pharmaceutically acceptable salt of said agent.
  • known cyclin-dependent kinase inhibitor which can be used for combination therapy include, but are not limited to, flavopiridol, UCN-01, roscovitine and olomoucine.
  • another embodiment of the present invention is directed to a composition effective to inhibit neoplasia comprising a compound, or a pharmaceutically acceptable salt or prodrug of a compound described herein, which functions as a caspase cascade activator and inducer of apoptosis or inhibitor of tubulin polymerization, in combination with at least one known COX-2 inhibitor, or a pharmaceutically acceptable salt of said inhibitor.
  • known COX-2 inhibitors which can be used for combination therapy include, but are not limited to, celecoxib, valecoxib, and rofecoxib.
  • Another embodiment of the present invention is directed to a composition effective to inhibit neoplasia comprising a bioconjugate of a compound described herein, which functions as a caspase cascade activator and inducer of apoptosis or inhibitor of tubulin polymerization, in bioconjugation with at least one known therapeutically useful antibody, such as Herceptin® (trastuzumab) or Rituxan® (rituximab), growth factors, such as DGF, NGF; cytokines, such as IL-2, IL-4, or any molecule that binds to the cell surface.
  • the antibodies and other molecules will deliver a compound described herein to its targets and make it an effective anticancer agent.
  • the bioconjugates could also enhance the anticancer effect of therapeutically useful antibodies, such as Herceptin® (trastuzumab) or Rituxan® (rituximab).
  • another embodiment of the present invention is directed to a composition effective to inhibit neoplasia comprising a compound, or a pharmaceutically acceptable salt or prodrug of a compound described herein, which functions as a caspase cascade activator and inducer of apoptosis or inhibitor of tubulin polymerization, in combination with radiation therapy.
  • the compound of the invention may be administered at the same time as the radiation therapy is administered or at a different time.
  • Yet another embodiment of the present invention is directed to a composition effective for post-surgical treatment of cancer, comprising a compound, or a pharmaceutically acceptable salt or prodrug of a compound described herein, which functions as a caspase cascade activator and inducer of apoptosis or inhibitor of tubulin polymerization.
  • the invention also relates to a method of treating cancer by surgically removing the cancer and then treating the animal with one of the pharmaceutical compositions described herein.
  • a wide range of immune mechanisms operate rapidly following exposure to an infectious agent. Depending on the type of infection, rapid clonal expansion of the T and B lymphocytes occurs to combat the infection.
  • the elimination of the effector cells following an infection is one of the major mechanisms for maintaining immune homeostasis.
  • the elimination of the effector cells has been shown to be regulated by apoptosis.
  • Autoimmune diseases have lately been determined to occur as a consequence of deregulated cell death.
  • the immune system directs its powerful cytotoxic effector mechanisms against specialized cells, such as oligodendrocytes in multiple sclerosis, the beta cells of the pancreas in diabetes mellitus, and thyrocytes in Hashimoto's thyroiditis (Ohsako, S.
  • lymphocyte apoptosis receptor Fas/APO-1/CD95 Mutations of the gene encoding the lymphocyte apoptosis receptor Fas/APO-1/CD95 are reported to be associated with defective lymphocyte apoptosis and autoimmune lymphoproliferative syndrome (ALPS), which is characterized by chronic, histologically benign splenomegaly, generalized lymphadenopathy, hypergammaglobulinemia, and autoantibody formation.
  • APS autoimmune lymphoproliferative syndrome
  • Fas-Fas ligand (FasL) interaction is known to be required for the maintenance of immune homeostasis.
  • Experimental autoimmune thyroiditis (EAT) characterized by autoreactive T and B cell responses and a marked lymphocytic infiltration of the thyroid, is a good model to study the therapeutic effects of FasL. Batteux, F., et al., ( J. Immunol. 162:603-608 (1999)) reported that by direct injection of DNA expression vectors encoding FasL into the inflamed thyroid, the development of lymphocytic infiltration of the thyroid was inhibited and induction of infiltrating T cells death was observed. These results show that FasL expression on thyrocytes may have a curative effect on ongoing EAT by inducing death of pathogenic autoreactive infiltrating T lymphocytes.
  • Bisindolylmaleimide VIII is known to potentiate Fas-mediated apoptosis in human astrocytoma 1321N1 cells and in Molt-4T cells; both of which were resistant to apoptosis induced by anti-Fas antibody in the absence of bisindolylmaleimide VIII. Potentiation of Fas-mediated apoptosis by bisindolylmaleimide VIII was reported to be selective for activated, rather than non-activated, T cells, and was Fas-dependent. Zhou T., et al., ( Nat. Med.
  • Psoriasis is a chronic skin disease that is characterized by scaly red patches.
  • Psoralen plus ultraviolet A (PUVA) is a widely used and effective treatment for psoriasis vulgarism
  • Coven, et al., Photodermatol. Photoimmunol. Photomed. 15:22-27 (1999) reported that lymphocytes treated with psoralen 8-MOP or TMP and UVA, displayed DNA degradation patterns typical of apoptotic cell death.
  • Ozawa, et al., J. Exp. Med. 189:711-718 (1999) reported that induction of T cell apoptosis could be the main mechanism by which 312-nm UVB resolves psoriasis skin lesions.
  • methotrexate Low doses of methotrexate may be used to treat psoriasis to restore a clinically normal skin. Heenen, et al., Arch. Dermatol. Res. 290:240-245 (1998), reported that low doses of methotrexate may induce apoptosis and that this mode of action could explain the reduction in epidermal hyperplasia during treatment of psoriasis with methotrexate. Therefore, an effective amount of a compound, or a pharmaceutically acceptable salt or prodrug of the compound of Formulae I-Vc, which functions as a caspase cascade activator and inducer of apoptosis, is an effective treatment for hyperproliferative skin diseases, such as psoriasis.
  • Synovial cell hyperplasia is a characteristic of patients with rheumatoid arthritis (RA). It is believed that excessive proliferation of RA synovial cells, as well as defects in synovial cell death, may be responsible for synovial cell hyperplasia. Wakisaka, et al., Clin. Exp. Immunol. 114:119-128 (1998), found that although RA synovial cells could die via apoptosis through a Fas/FasL pathway, apoptosis of synovial cells was inhibited by proinflammatory cytokines present within the synovium. Wakisaka, et al.
  • an effective amount of a compound, or a pharmaceutically acceptable salt or prodrug of the compound of Formulae I-Vc, which functions as a caspase cascade activator and inducer of apoptosis is an effective treatment for rheumatoid arthritis.
  • an effective amount of a compound, or a pharmaceutically acceptable salt or prodrug of the compound of Formulae I-Vc, which functions as a caspase cascade activator and inducer of apoptosis, is an effective treatment for inflammation.
  • Caspase cascade activators and inducers of apoptosis may also be a desirable therapy in the elimination of pathogens, such as HIV, Hepatitis C and other viral pathogens.
  • pathogens such as HIV, Hepatitis C and other viral pathogens.
  • the long lasting quiecence, followed by disease progression, may be explained by an anti-apoptotic mechanism of these pathogens leading to persistent cellular reservoirs of the virions. It has been reported that HIV-1 infected T leukemia cells or peripheral blood mononuclear cells (PBMCs) underwent enhanced viral replication in the presence of the caspase inhibitor Z-VAD-fink.
  • PBMCs peripheral blood mononuclear cells
  • Z-VAD-fmk also stimulated endogenous virus production in activated PBMCs derived from HIV-1-infected asymptomatic individuals (Chinnaiyan, A., et al., Nat. Med. 3:333 (1997)). Therefore, apoptosis serves as a beneficial host mechanism to limit the spread of HIV and new therapeutics using caspase/apoptosis activators are useful to clear viral reservoirs from the infected individuals.
  • HCV infection also triggers anti-apoptotic mechanisms to evade the host's immune surveillance leading to viral persistence and hepatocarcinogenesis (Tai, D. I., et al. Hepatology 3:656-64 (2000)). Therefore, apoptosis inducers are useful as therapeutics for HIV, HCV, HBV, and other infectious disease.
  • Stent implantation has become the new standard angioplasty procedure.
  • in-stent restenosis remains the major limitation of coronary stenting.
  • New approaches have been developed to target pharmacological modulation of local vascular biology by local administration of drugs. This allows for drug applications at the precise site and time of vessel injury.
  • Numerous pharmacological agents with antiproliferative properties are currently under clinical investigation, including actinomycin D, rapamycin or paclitaxel coated stents (Regar E., et al., Br. Med. Bull. 59:227-248 (2001)). Therefore, apoptosis inducers, which are antiproliferative, are useful as therapeutics for the prevention or reduction of in-stent restenosis.
  • Another important aspect of the present invention is the surprising discovery that compounds of the present invention are potent and highly efficacious activators of caspase-3, inhibitors of tubulin polymerization, and inhibitors of topoisomerase even in drug resistant cancer cells, which enables these compounds to inhibit the growth and proliferation of drug resistant cancer cells, and to cause apoptosis and cell death in the drug resistant cancer cells.
  • the compounds of the present invention are not substrates for the MDR transporters such as Pgp-1 (MDR-1), MRP-1 and BCRP. This is particularly surprising in view of the fact that almost all of the commercially available tubulin-interacting chemotherapeutics are substrates for multidrug resistance transporters (MDRs).
  • Multidrug resistance is the major cause of chemotherapy failure.
  • Drug resistance is typically caused by ATP-dependent efflux of drug from cells by ATP-binding cassette (ABC) transporters.
  • ABC transporters ABCB1 MDR-1, P glycoprotein
  • ABCC1 MRP1
  • ABCG2 BCRP, MXR
  • the compounds of the present invention are effective in killing drug resistant cancer cells. Therefore, compounds of this invention are useful for the treatment of drug resistant cancer.
  • another aspect of the present invention is the application of the methods and compounds of the present invention as described above to tumors that have acquired resistance to other anticancer drugs.
  • a compound of the present invention is administered to a cancer patient who has been treated with another anti-cancer drug.
  • a compound of the present invention is administered to a patient who has been treated with and is not responsive to another anti-cancer drug or developed resistance to such other anti-cancer compound.
  • a compound of the present invention is administered to a patient who has been treated with another anti-cancer drug and is refractory to said other anti-cancer drug.
  • the compounds of the present invention can be used in treating cancer in a patient who is not responsive or is resistant to any other anti-cancer agent.
  • Examples of such other anti-cancer agent may include alkylating agents, antimitotic agents, topo I inhibitors, topo II inhibitors, RNA/DNA antimetabolites, EGFR inhibitors, angiogenesis inhibitors, tubulin inhibitors (e.g., vinblastine, taxol® (paclitaxel), and analogues thereof), proteosome inhibitors, etc., some of the exemplary compounds of which are provided above and are general known in the art, e.g., melphalan, chlorambucil, cyclophosamide, ifosfamide, vincristine, mitoguazone, epirubicin, aclarubicin, bleomycin, mitoxantrone, elliptinium, fludarabine, octreotide, retinoic acid, tamoxifen, Gleevec® (imatinib mesylate) and alanosine.
  • tubulin inhibitors e.g., vinblastine, tax
  • the compounds can be used in treating patients having any type of diseases responsive to the inhibition of tubulin or inhibition of topoisomerase (including but not limited to the types of cancer described above) who are not responsive or become resistant to another therapeutic agent, e.g., another anti-cancer agent.
  • another therapeutic agent e.g., another anti-cancer agent.
  • compositions within the scope of this invention include all compositions wherein the compounds of the present invention are contained in an amount that is effective to achieve its intended purpose. While individual needs vary, determination of optimal ranges of effective amounts of each component is within the skill of the art.
  • the compounds may be administered to animals, e.g., mammals, orally at a dose of 0.0025 to 50 mg/kg of body weight, per day, or an equivalent amount of the pharmaceutically acceptable salt thereof, to a mammal being treated. Preferably, approximately 0.01 to approximately 10 mg/kg of body weight is orally administered.
  • the dose is generally approximately one-half of the oral dose.
  • a suitable intramuscular dose would be approximately 0.0025 to approximately 25 mg/kg of body weight, and most preferably, from approximately 0.01 to approximately 5 mg/kg of body weight.
  • a known cancer chemotherapeutic agent is also administered, it is administered in an amount that is effective to achieve its intended purpose.
  • the amounts of such known cancer chemotherapeutic agents effective for cancer are well known to those skilled in the art.
  • the unit oral dose may comprise from approximately 0.01 to approximately 50 mg, preferably approximately 0.1 to approximately 10 mg of the compound of the invention.
  • the unit dose may be administered one or more times daily, as one or more tablets, each containing from approximately 0.1 to approximately 10 mg, conveniently approximately 0.25 to 50 mg of the compound or its solvates.
  • the compound may be present at a concentration of approximately 0.01 to 100 mg per gram of carrier.
  • the compounds of the invention may be administered as part of a pharmaceutical preparation containing suitable pharmaceutically acceptable carriers comprising excipients and auxiliaries, which facilitate processing of the compounds into preparations that may be used pharmaceutically.
  • suitable pharmaceutically acceptable carriers comprising excipients and auxiliaries, which facilitate processing of the compounds into preparations that may be used pharmaceutically.
  • the preparations particularly those preparations which may be administered orally and that may be used for the preferred type of administration, such as tablets, dragees, and capsules, and also preparations that may be administered rectally, such as suppositories, as well as suitable solutions for administration by injection or orally, contain from approximately 0.01 to 99 percent, preferably from approximately 0.25 to 75 percent of active compound(s), together with the excipient.
  • non-toxic pharmaceutically acceptable salts of the compounds of the present invention are also included within the scope of the present invention.
  • Acid addition salts are formed by mixing a solution of the compounds of the present invention with a solution of a pharmaceutically acceptable non-toxic acid, such as hydrochloric acid, fumaric acid, maleic acid, succinic acid, acetic acid, citric acid, tartaric acid, carbonic acid, phosphoric acid, oxalic acid, and the like.
  • Basic salts are formed by mixing a solution of the compounds of the present invention with a solution of a pharmaceutically acceptable non-toxic base, such as sodium hydroxide, potassium hydroxide, choline hydroxide, sodium carbonate, Tris, N-methyl-glucamine and the like.
  • compositions of the invention may be administered to any animal, which may experience the beneficial effects of the compounds of the invention.
  • animals are mammals, e.g., humans and veterinary animals, although the invention is not intended to be so limited.
  • compositions of the present invention may be administered by any means that achieve their intended purpose.
  • administration may be by parenteral, subcutaneous, intravenous, intramuscular, intraperitoneal, transdermal, buccal, intrathecal, intracranial, intranasal or topical routes.
  • administration may be by the oral route.
  • the dosage administered will be dependent upon the age, health, and weight of the recipient, kind of concurrent treatment, if any, frequency of treatment, and the nature of the effect desired.
  • compositions of the present invention are manufactured in a manner, which is itself known, e.g., by means of conventional mixing, granulating, dragee-making, dissolving, or lyophilizing processes.
  • pharmaceutical preparations for oral use may be obtained by combining the active compounds with solid excipients, optionally grinding the resulting mixture and processing the mixture of granules, after adding suitable auxiliaries, if desired or necessary, to obtain tablets or dragee cores.
  • Suitable excipients are, in particular: fillers, such as saccharides, e.g. lactose or sucrose, mannitol or sorbitol; cellulose preparations and/or calcium phosphates, e.g. tricalcium phosphate or calcium hydrogen phosphate; as well as binders, such as starch paste, using, e.g., maize starch, wheat starch, rice starch, potato starch, gelatin, tragacanth, methyl cellulose, hydroxypropylmethylcellulose, sodium carboxymethylcellulose, and/or polyvinyl pyrrolidone.
  • fillers such as saccharides, e.g. lactose or sucrose, mannitol or sorbitol
  • cellulose preparations and/or calcium phosphates e.g. tricalcium phosphate or calcium hydrogen phosphate
  • binders such as starch paste, using, e.g., maize starch, wheat starch, rice starch, potato
  • disintegrating agents may be added, such as the above-mentioned starches and also carboxymethyl-starch, cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof, such as sodium alginate.
  • Auxiliaries are, above all, flow-regulating agents and lubricants, e.g., silica, talc, stearic acid or salts thereof, such as magnesium stearate or calcium stearate, and/or polyethylene glycol.
  • Dragee cores are provided with suitable coatings which, if desired, are resistant to gastric juices.
  • concentrated saccharide solutions may be used, which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, polyethylene glycol and/or titanium dioxide, lacquer solutions and suitable organic solvents or solvent mixtures.
  • suitable cellulose preparations such as acetylcellulose phthalate or hydroxypropymethyl-cellulose phthalate, are used.
  • Dye stuffs or pigments may be added to the tablets or dragee coatings, e.g., for identification or in order to characterize combinations of active compound doses.
  • Other pharmaceutical preparations which may be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol.
  • the push-fit capsules may contain the active compounds in the form of: granules, which may be mixed with fillers, such as lactose; binders, such as starches; and/or lubricants, such as talc or magnesium stearate and, optionally, stabilizers.
  • the active compounds are preferably dissolved or suspended in suitable liquids, such as fatty oils, or liquid paraffin.
  • suitable liquids such as fatty oils, or liquid paraffin.
  • stabilizers may be added.
  • Possible pharmaceutical preparations which may be used rectally include, e.g., suppositories, which consist of a combination of one or more of the active compounds with a suppository base.
  • Suitable suppository bases are, e.g., natural or synthetic triglycerides, or paraffin hydrocarbons.
  • gelatin rectal capsules which consist of a combination of the active compounds with a base.
  • Possible base materials include, e.g., liquid triglycerides, polyethylene glycols, or paraffin hydrocarbons.
  • Suitable formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form, e.g., water-soluble salts and alkaline solutions.
  • suspensions of the active compounds as appropriate oily injection suspensions may be administered.
  • Suitable lipophilic solvents or vehicles include fatty oils, e.g., sesame oil, or synthetic fatty acid esters, e.g., ethyl oleate or triglycerides or polyethylene glycol-400 (the compounds are soluble in PEG-400), or cremophor, or cyclodextrins.
  • Aqueous injection suspensions may contain substances which increase the viscosity of the suspension include, e.g., sodium carboxymethyl cellulose, sorbitol, and/or dextran.
  • the suspension may also contain stabilizers.
  • compounds of the invention are employed in topical and parenteral formulations and are used for the treatment of skin cancer.
  • the topical compositions of this invention are formulated preferably as oils, creams, lotions, ointments and the like by choice of appropriate carriers.
  • Suitable carriers include vegetable or mineral oils, white petrolatum (white soft paraffin), branched chain fats or oils, animal fats and high molecular weight alcohol (greater than C 12 ).
  • the preferred carriers are those in which the active ingredient is soluble.
  • Emulsifiers, stabilizers, humectants and antioxidants may also be included, as well as agents imparting color or fragrance, if desired.
  • transdermal penetration enhancers may be employed in these topical formulations. Examples of such enhancers are found in U.S. Pat. Nos. 3,989,816 and 4,444,762.
  • Creams are preferably formulated from a mixture of mineral oil, self-emulsifying beeswax and water in which mixture of the active ingredient, dissolved in a small amount of an oil, such as almond oil, is admixed.
  • An oil such as almond oil
  • a typical example of such a cream is one which includes approximately 40 parts water, approximately 20 parts beeswax, approximately 40 parts mineral oil and approximately 1 part almond oil.
  • Ointments may be formulated by mixing a solution of the active ingredient in a vegetable oil, such as almond oil, with warm soft paraffin and allowing the mixture to cool.
  • a vegetable oil such as almond oil
  • a typical example of such an ointment is one which includes approximately 30% almond oil and approximately 70% white soft paraffin by weight.
  • 2,4-Dichloroquinazoline A suspension of 2,4-quinazolinedione (5.0 g, 30.8 mmol) in neat phosphorylchloride (50 mL) was heated under reflux for 18 h. The reaction mixture was concentrated under vacuum. The crude product was purified by chromatography (Silica gel) using ethyl acetate and hexane (1:4) to give 2,4-dichloroquinazoline as white solid (4.8 g, 96%).
  • the title compound was prepared from 2,4-dichloroquinazoline (250 mg, 1.25 mmol) and 4-methyl-N-methylaniline (196 mg, 1.43 mmol) by a procedure similar to example 1b and was isolated as white powder (210 mg, 84%).
  • the title compound was prepared from 2,4-dichloroquinazoline (60 mg, 0.302 mmol) and 4-chloro-N-methylaniline (50 mg, 0.332 mmol) by a procedure similar to example 1b and was isolated as white powder (30 mg, 50%).
  • the title compound was prepared from 2,4-dichloroquinazoline (50 mg, 0.251 mmol) and 4-nitro-N-methylaniline (46 mg, 0.302 mmol) by a procedure similar to example 1b and was isolated as yellow powder (6 mg, 12%).
  • the title compound was prepared from 2,4-dichloroquinazoline (50 mg, 0.251 mmol) and 5-methoxyindole (40 mg, 0.302 mmol) similar to example 1b and was isolated as white powder (14 mg, 28%).
  • the title compound was prepared from (2-chloro-quinazolin-4-yl)-(4-methoxy-phenyl)-methyl-amine (15 mg, 0.050 mmol) and 2-hydroxylethylamine (20 ⁇ L) by a procedure similar to example 10 and was isolated as white solid (12 mg, 80%).
  • the title compound was prepared from (2-chloro-quinazolin-4-yl)-(4-methoxy-phenyl)-methyl-amine (45 mg, 0.151 mmol) and 3,7-dimethyl-2,6-diene-octamine (60 ⁇ L, 0.301 mmol) by a procedure similar to example 10 and was isolated as white powder (15 mg, 33%).
  • the title compound was prepared from (2-chloro-quinazolin-4-yl)-(4-methoxy-phenyl)-methyl-amine (10 mg, 0.033 mmol) and 2-morpholin-4-yl-ethylamine (30 ⁇ L) by a procedure similar to example 10 and was isolated as white powder (10 mg, 100%).
  • the title compound was prepared from (2-chloro-quinazolin-4-yl)-(4-methoxy-phenyl)-methyl-amine (15 mg, 0.050 mmol) and morpholine (30 ⁇ L) by a procedure similar to example 10 and was isolated as white powder (10 mg, 66%).
  • the title compound was prepared from (2-chloro-quinazolin-4-yl)-(4-methyl-phenyl)-methyl-amine (50 mg, 0.177 mmol) and 3,7-dimethyl-2,6-diene-octamine (50 ⁇ L, 0.265 mmol) by a procedure similar to example 10 and was isolated as white powder (7 mg, 14%).
  • the title compound was prepared from 2,4-dichloroquinazoline (60 mg, 0.3.02 mmol) and 4-chloro-N-methylaniline (72 mg, 0.513 mmol) by a procedure similar to example 1b and was isolated as white powder (50 mg, 83%).
  • the title compound was prepared from (2-chloro-6,7-dimethoxyquinazolin-4-yl)-(4-methoxy-phenyl)-methyl-amine (50 mg, 0.139 mmol) and 2-morpholin-4-yl-ethylamine (25 ⁇ L, 0.167 mmol) by a procedure similar to example 10 and was isolated as white powder (27 mg, 54%).
  • the title compound was prepared from (2-chloro-6,7-dimethoxyquinazolin-4-yl)-(4-methoxy-phenyl)-methyl-amine (60 mg, 0.167 mmol) and 3,7-dimethyl-2,6-diene-octamine (50 mg, 0.424 mmol) by a procedure similar to example 10 and was isolated as white powder (68 mg, 85%).
  • reaction mixture was quenched by adding 50 uL of water, diluted with 25 mL of ethyl acetate, washed with water (25 mL ⁇ 3), saturated NaCl, dried over anhydrous MgSO 4 , filtered and concentrated. The residue was purified by chromatography (20% ethyl acetate/hexanes) to give the title compound (14.3 mg, 0.048 mmol, 70%).
  • the title compound was prepared from (2-chloro-quinazolin-4-yl)-(4-methoxy-phenyl)-methylamine (10 mg, 0.033 mmol) and histamine hydrochloride (16 mg, 0.10) by a procedure similar to example 10 and was isolated as white solid (7 mg, 70%).
  • the title compound was prepared from (2-chloro-quinazolin-4-yl)-(4-methoxy-phenyl)-methyl-amine (15 mg, 0.050 mmol) and N 1 ,N 1 -dimethyl-propane-1,3-diamine (16 ⁇ l, 0.070 mmol) by a procedure similar to example 10 and was isolated as white powder (11 mg, 73%).
  • the title compound was prepared from (2-chloro-quinazolin-4-yl)-(6-methoxy-pyridin-3-yl)-methyl-amine (12 mg, 0.040 mmol) and ethanolamine (28 ⁇ l) by a procedure similar to example 10 and was isolated as off white solid (8 mg, 66%).
  • the title compound was prepared from (2-Chloro-6,7-dimethoxyquinazolin-4-yl)-(4-methoxy-phenyl)-amine (26 mg, 0.079 mmol) and ethanolamine (30 ⁇ l) by a procedure similar to example 10 and was isolated as white powder (14 mg, 54%).
  • the title compound was prepared from 2,4-dichloroquinazoline (30 mg, 0.152 mmol) and 4-methylamino-benzoic acid methyl ester (27 mg, 0.167 mmol) by a procedure similar to example 1b and was isolated as off white powder (25 mg, 83%).
  • Example 89-90 Compounds of Example 89-90 were prepared by a procedure similar to Example 56.
US11/680,843 2003-07-03 2007-03-01 Compounds and therapeutical use thereof Abandoned US20070244113A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/680,843 US20070244113A1 (en) 2003-07-03 2007-03-01 Compounds and therapeutical use thereof

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US48432503P 2003-07-03 2003-07-03
US49300603P 2003-08-07 2003-08-07
US55755604P 2004-03-29 2004-03-29
US57128804P 2004-05-14 2004-05-14
US10/885,903 US7618975B2 (en) 2003-07-03 2004-07-06 4-arylamino-quinazolines and analogs as activators of caspases and inducers of apoptosis and the use thereof
US11/680,843 US20070244113A1 (en) 2003-07-03 2007-03-01 Compounds and therapeutical use thereof

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/885,903 Continuation US7618975B2 (en) 2003-07-03 2004-07-06 4-arylamino-quinazolines and analogs as activators of caspases and inducers of apoptosis and the use thereof

Publications (1)

Publication Number Publication Date
US20070244113A1 true US20070244113A1 (en) 2007-10-18

Family

ID=33568832

Family Applications (7)

Application Number Title Priority Date Filing Date
US10/885,903 Expired - Fee Related US7618975B2 (en) 2003-07-03 2004-07-06 4-arylamino-quinazolines and analogs as activators of caspases and inducers of apoptosis and the use thereof
US11/620,498 Abandoned US20070208044A1 (en) 2003-07-03 2007-01-05 Compounds and therapeutical use thereof
US11/680,843 Abandoned US20070244113A1 (en) 2003-07-03 2007-03-01 Compounds and therapeutical use thereof
US11/693,432 Abandoned US20070249601A1 (en) 2003-07-03 2007-03-29 Compounds and therapeutical use thereof
US11/779,086 Abandoned US20080032974A1 (en) 2003-07-03 2007-07-17 Compounds and therapeutical use thereof
US11/781,660 Abandoned US20080039479A1 (en) 2003-07-03 2007-07-23 Compounds and therapeutical use thereof
US15/081,530 Abandoned US20170050937A1 (en) 2003-07-03 2016-03-25 Compounds and therapeutical use thereof

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US10/885,903 Expired - Fee Related US7618975B2 (en) 2003-07-03 2004-07-06 4-arylamino-quinazolines and analogs as activators of caspases and inducers of apoptosis and the use thereof
US11/620,498 Abandoned US20070208044A1 (en) 2003-07-03 2007-01-05 Compounds and therapeutical use thereof

Family Applications After (4)

Application Number Title Priority Date Filing Date
US11/693,432 Abandoned US20070249601A1 (en) 2003-07-03 2007-03-29 Compounds and therapeutical use thereof
US11/779,086 Abandoned US20080032974A1 (en) 2003-07-03 2007-07-17 Compounds and therapeutical use thereof
US11/781,660 Abandoned US20080039479A1 (en) 2003-07-03 2007-07-23 Compounds and therapeutical use thereof
US15/081,530 Abandoned US20170050937A1 (en) 2003-07-03 2016-03-25 Compounds and therapeutical use thereof

Country Status (9)

Country Link
US (7) US7618975B2 (US20070244113A1-20071018-C00053.png)
EP (1) EP1660092A2 (US20070244113A1-20071018-C00053.png)
JP (2) JP5129957B2 (US20070244113A1-20071018-C00053.png)
KR (1) KR101218213B1 (US20070244113A1-20071018-C00053.png)
CN (1) CN1984660B (US20070244113A1-20071018-C00053.png)
AU (1) AU2004253967B2 (US20070244113A1-20071018-C00053.png)
CA (1) CA2531327A1 (US20070244113A1-20071018-C00053.png)
NZ (1) NZ544472A (US20070244113A1-20071018-C00053.png)
WO (1) WO2005003100A2 (US20070244113A1-20071018-C00053.png)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20070244114A1 (en) * 2004-07-06 2007-10-18 Myriad Genetics, Incorporated Compounds and therapeutical use thereof
US20080004297A1 (en) * 2003-07-03 2008-01-03 Myriad Genetics, Inc. Compounds and therapeutical use thereof
US20080032974A1 (en) * 2003-07-03 2008-02-07 Myriad Genetics, Incorporated Compounds and therapeutical use thereof
US20080051398A1 (en) * 2005-01-03 2008-02-28 Myriad Genetics, Inc. Method of treating brain cancer
US20080319048A1 (en) * 2007-06-22 2008-12-25 Scidose Llc Solubilized formulation of docetaxel without tween 80
US7772274B1 (en) 2009-10-19 2010-08-10 Scidose, Llc Docetaxel formulations with lipoic acid
US20110092580A1 (en) * 2009-10-19 2011-04-21 Scidose Llc Docetaxel formulations with lipoic acid and/or dihydrolipoic acid
US20110092579A1 (en) * 2009-10-19 2011-04-21 Scidose Llc Solubilized formulation of docetaxel
US8309562B2 (en) 2003-07-03 2012-11-13 Myrexis, Inc. Compounds and therapeutical use thereof
US8912228B2 (en) 2009-10-19 2014-12-16 Scidose Llc Docetaxel formulations with lipoic acid

Families Citing this family (70)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7344702B2 (en) * 2004-02-13 2008-03-18 Bristol-Myers Squibb Pharma Company Contrast agents for myocardial perfusion imaging
WO2005048953A2 (en) 2003-11-13 2005-06-02 Ambit Biosciences Corporation Amide derivatives as kinase modulators
EP1750689A1 (en) 2004-05-15 2007-02-14 Vertex Pharmaceuticals Incorporated Treating seizures using ice inhibitors
EP2295054A1 (en) 2004-05-27 2011-03-16 Vertex Pharmaceuticals Incorporated Ice inhibitors for the treatment of autoinflammatory diseases
JP2008526734A (ja) * 2004-12-31 2008-07-24 エスケー ケミカルズ カンパニー リミテッド 糖尿及び肥満治療予防に有効なキナゾリン誘導体
EP1833511A4 (en) * 2005-01-03 2011-01-19 Myriad Genetics Inc METHOD OF TREATING BRAIN CANCER
JP5354775B2 (ja) * 2005-06-14 2013-11-27 ベイラー ユニバーシティ チューブリン結合活性を有するコンブレタスタチンアナログ
CA2609251A1 (en) * 2005-06-16 2006-12-28 Myriad Genetics, Inc. Pharmaceutical compositions and use thereof
JP2009501232A (ja) * 2005-07-15 2009-01-15 シェーリング コーポレイション 癌の処置に有用なキナゾリン誘導体
JP2009507229A (ja) * 2005-09-02 2009-02-19 ザ・ユニバーシティ・オブ・トレド 生物学的状態の診断及び/又は治療に有用なバイオマーカーを同定するための方法及び組成物
NZ569817A (en) * 2005-12-21 2011-10-28 Abbott Lab Anti-viral compounds
CN101384592A (zh) * 2005-12-21 2009-03-11 艾博特公司 抗病毒化合物
WO2007081517A2 (en) 2005-12-21 2007-07-19 Abbott Laboratories Anti-viral compounds
KR20080112380A (ko) * 2006-04-14 2008-12-24 아스트라제네카 아베 Csf-1r 키나제 억제제로서의 4-아닐리노퀴놀린-3-카르복스아미드
KR101129868B1 (ko) * 2006-10-04 2012-04-12 화이자 프로덕츠 인코포레이티드 칼슘 수용체 길항제로서의 피리도[4,3-d]피리미딘-4(3H)-온 유도체
WO2008133753A2 (en) 2006-12-20 2008-11-06 Abbott Laboratories Anti-viral compounds
EP2144888A4 (en) * 2007-04-10 2012-10-03 Myrexis Inc METHODS OF TREATING CANCER
EP2144886A4 (en) * 2007-04-10 2012-10-03 Myrexis Inc METHOD OF TREATING MELANOMA
WO2008124828A1 (en) * 2007-04-10 2008-10-16 Myriad Genetics, Inc. Methods for treating vascular disruption disorders
NZ580866A (en) * 2007-04-10 2011-02-25 Myriad Pharmaceuticals Inc Method of treating brain cancer
EP2144887A4 (en) * 2007-04-10 2012-10-03 Myrexis Inc PHARMACEUTICAL FORMS AND METHOD FOR THE TREATMENT OF CANCER
US20090209536A1 (en) * 2007-06-17 2009-08-20 Kalypsys, Inc. Aminoquinazoline cannabinoid receptor modulators for treatment of disease
WO2010006115A1 (en) * 2008-07-11 2010-01-14 Myriad Pharmaceuticals, Inc. Pharmaceutical compounds as cytotoxic agents and the use thereof
US20100068197A1 (en) * 2008-07-11 2010-03-18 Myriad Pharmaceuticals, Inc. Pharmaceutical compounds as inhibitors of cell proliferation and the use thereof
CN101463014B (zh) * 2008-12-26 2013-07-10 复旦大学 二芳基苯并嘧啶类衍生物及其药物组合物和用途
US8329907B2 (en) 2009-04-02 2012-12-11 Merck Patent Gmbh Autotaxin inhibitors
BR112012008330B1 (pt) 2009-09-03 2022-03-22 Bristol-Myers Squibb Company Compostos quinazolinas, suas composições farmacêuticas e seus usos
CN101716351B (zh) * 2009-12-08 2012-04-11 中国人民解放军军事医学科学院野战输血研究所 一类吡啶化合物在制备rna干涉增强剂中的应用
US20110224240A1 (en) * 2010-01-11 2011-09-15 Myrexis, Inc. Methods of treating cancer and related diseases
MX348958B (es) 2010-02-08 2017-07-04 Lantheus Medical Imaging Inc Métodos y aparatos para sintetizar agentes formadores de imagen e intermedios de estos.
CA2799653A1 (en) 2010-06-04 2011-12-08 Bertrand Leblond Substituted isoquinolines and their use as tubulin polymerization inhibitors
WO2012061785A2 (en) 2010-11-05 2012-05-10 Brandeis University Ice inhibiting compounds and uses thereof
WO2012088266A2 (en) 2010-12-22 2012-06-28 Incyte Corporation Substituted imidazopyridazines and benzimidazoles as inhibitors of fgfr3
JP5813785B2 (ja) * 2011-02-04 2015-11-17 デュケイン ユニバーシティー オブ ザ ホリー スピリット 抗チューブリン活性を有する二環式および三環式のピリミジンチロシンキナーゼ阻害剤ならびに患者の処置方法
KR101940340B1 (ko) 2011-03-04 2019-01-18 글락소스미스클라인 인털렉츄얼 프로퍼티 디벨로프먼트 리미티드 키나제 억제제로서의 아미노-퀴놀린
TWI547494B (zh) 2011-08-18 2016-09-01 葛蘭素史克智慧財產發展有限公司 作為激酶抑制劑之胺基喹唑啉類
NZ730134A (en) 2012-06-13 2018-07-27 Incyte Holdings Corp Substituted tricyclic compounds as fgfr inhibitors
AU2013203000B9 (en) 2012-08-10 2017-02-02 Lantheus Medical Imaging, Inc. Compositions, methods, and systems for the synthesis and use of imaging agents
AR092529A1 (es) 2012-09-13 2015-04-22 Glaxosmithkline Llc Compuesto de aminoquinazolina, composicion farmaceutica que lo comprende y uso de dicho compuesto para la preparacion de un medicamento
AR092530A1 (es) 2012-09-13 2015-04-22 Glaxosmithkline Llc Compuesto de amino-quinolina, composicion farmaceutica que lo comprende y uso de dicho compuesto para la preparacion de un medicamento
CA2902132C (en) 2013-02-21 2020-09-22 Glaxosmithkline Intellectual Property Development Limited Quinazolines as kinase inhibitors
EP2769723A1 (en) 2013-02-22 2014-08-27 Ruprecht-Karls-Universität Heidelberg Compounds for use in inhibiting HIV capsid assembly
EP2769722A1 (en) 2013-02-22 2014-08-27 Ruprecht-Karls-Universität Heidelberg Compounds for use in inhibiting HIV capsid assembly
CN109912594A (zh) 2013-04-19 2019-06-21 因赛特控股公司 作为fgfr抑制剂的双环杂环
CN104130200B (zh) * 2014-07-01 2016-04-20 中山大学 一种2-取代苯基-4-芳胺基喹唑啉衍生物及其制备方法和应用
US10851105B2 (en) 2014-10-22 2020-12-01 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
MA41551A (fr) 2015-02-20 2017-12-26 Incyte Corp Hétérocycles bicycliques utilisés en tant qu'inhibiteurs de fgfr4
TWI712601B (zh) 2015-02-20 2020-12-11 美商英塞特公司 作為fgfr抑制劑之雙環雜環
EP3283466A4 (en) * 2015-04-16 2018-09-12 Icahn School of Medicine at Mount Sinai Ksr antagonists
CA3158951A1 (en) * 2016-08-15 2018-02-22 Purdue Research Foundation 4-substituted aminoisoquinoline derivatives
CN106380465B (zh) * 2016-09-05 2019-03-12 郑州大学 含1,2,3-三氮唑结构单元的2,4-二取代喹唑啉类化合物及其制备方法和用途
CN108239071B (zh) * 2016-12-27 2020-12-04 沈阳药科大学 酰胺及硫代酰胺类衍生物及其制备方法和应用
AR111960A1 (es) 2017-05-26 2019-09-04 Incyte Corp Formas cristalinas de un inhibidor de fgfr y procesos para su preparación
US11254654B2 (en) * 2017-06-30 2022-02-22 The University Of North Carolina At Chapel Hill Heterochromatin gene repression inhibitors
MA52494A (fr) 2018-05-04 2021-03-10 Incyte Corp Formes solides d'un inhibiteur de fgfr et leurs procédés de préparation
CR20200591A (es) 2018-05-04 2021-03-31 Incyte Corp Sales de un inhibidor de fgfr
WO2020185532A1 (en) 2019-03-08 2020-09-17 Incyte Corporation Methods of treating cancer with an fgfr inhibitor
WO2021007269A1 (en) 2019-07-09 2021-01-14 Incyte Corporation Bicyclic heterocycles as fgfr inhibitors
BR112022007163A2 (pt) 2019-10-14 2022-08-23 Incyte Corp Heterociclos bicíclicos como inibidores de fgfr
US11566028B2 (en) 2019-10-16 2023-01-31 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
CA3162010A1 (en) 2019-12-04 2021-06-10 Incyte Corporation Derivatives of an fgfr inhibitor
WO2021113479A1 (en) 2019-12-04 2021-06-10 Incyte Corporation Tricyclic heterocycles as fgfr inhibitors
US11691971B2 (en) 2020-06-19 2023-07-04 Incyte Corporation Naphthyridinone compounds as JAK2 V617F inhibitors
US11753413B2 (en) 2020-06-19 2023-09-12 Incyte Corporation Substituted pyrrolo[2,1-f][1,2,4]triazine compounds as JAK2 V617F inhibitors
TW202216713A (zh) 2020-07-02 2022-05-01 美商英塞特公司 作為jak2 v617f抑制劑之三環脲化合物
WO2022006456A1 (en) 2020-07-02 2022-01-06 Incyte Corporation Tricyclic pyridone compounds as jak2 v617f inhibitors
WO2022046989A1 (en) 2020-08-27 2022-03-03 Incyte Corporation Tricyclic urea compounds as jak2 v617f inhibitors
WO2022140231A1 (en) 2020-12-21 2022-06-30 Incyte Corporation Deazaguaine compounds as jak2 v617f inhibitors
TW202313611A (zh) 2021-06-09 2023-04-01 美商英塞特公司 作為fgfr抑制劑之三環雜環
CN114436975B (zh) * 2022-01-26 2023-10-31 贵州省中国科学院天然产物化学重点实验室(贵州医科大学天然产物化学重点实验室) 2-三氟甲基-4-氨基喹唑啉类化合物及其应用

Citations (79)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2883382A (en) * 1957-08-29 1959-04-21 Parke Davis & Co Benzo-quinolylamino-2-[di(beta-chloroethyl)aminomethyl]-phenols
US3031450A (en) * 1959-04-30 1962-04-24 Thomae Gmbh Dr K Substituted pyrimido-[5, 4-d]-pyrimidines
US3502681A (en) * 1968-04-10 1970-03-24 Roussel Uclaf 7- or 8-chloro-4-phenylamino-chloroquinolines
US3632761A (en) * 1969-08-08 1972-01-04 Upjohn Co Method of obtaining antihypertensive and antianxiety effects
US3971783A (en) * 1973-03-07 1976-07-27 Pfizer Inc. 4-Aminoquinazoline derivatives as cardiac stimulants
US4025629A (en) * 1974-01-07 1977-05-24 The Upjohn Company P-(trifluoromethylquinolylamino)benzamides, pharmaceutical dosage forms and method of treatment
US4322420A (en) * 1978-09-11 1982-03-30 Sankyo Company Limited Method of using 4-anilinoquinazoline derivatives as analgesic and anti-inflammatory agents
US4435003A (en) * 1980-01-31 1984-03-06 Ciba-Geigy Corporation Chromogenic quinazolines
US4510307A (en) * 1980-08-20 1985-04-09 Asahi Kasei Kogyo Kabushiki Kaisha 6-Quinazolinesulfonyl derivatives and process for preparation thereof
US4675047A (en) * 1980-07-01 1987-06-23 Alexander Serban Substituted phenylamine- and phenyloxy-quinazolines as herbicides
US5114939A (en) * 1988-01-29 1992-05-19 Dowelanco Substituted quinolines and cinnolines as fungicides
US5145843A (en) * 1988-01-29 1992-09-08 Dowelanco Quinoline and cinnoline fungicides
US5187168A (en) * 1991-10-24 1993-02-16 American Home Products Corporation Substituted quinazolines as angiotensin II antagonists
US5223505A (en) * 1989-04-21 1993-06-29 Imperial Chemical Industries Plc Pyrimidine derivatives
US5236925A (en) * 1991-10-24 1993-08-17 American Home Products Corporation Fused pyrimidines as angiotensin II antagonists
US5276148A (en) * 1991-05-23 1994-01-04 Basf Aktiengesellschaft Phenylazobenzenes or naphthylazobenzenes having a plurality of fiber-reactive groups
US5294622A (en) * 1988-01-29 1994-03-15 Dowelanco Substituted quinolines and cinnolines
US5330989A (en) * 1991-10-24 1994-07-19 American Home Products Corporation Heterocycles substituted with biphenyl-3-cyclobutene-1,2-dione derivatives
US5409930A (en) * 1991-05-10 1995-04-25 Rhone-Poulenc Rorer Pharmaceuticals Inc. Bis mono- and bicyclic aryl and heteroaryl compounds which inhibit EGF and/or PDGF receptor tyrosine kinase
US5436233A (en) * 1992-07-15 1995-07-25 Ono Pharmaceutical Co., Ltd. 4-aminoquinazoline derivatives
US5480883A (en) * 1991-05-10 1996-01-02 Rhone-Poulenc Rorer Pharmaceuticals Inc. Bis mono- and bicyclic aryl and heteroaryl compounds which inhibit EGF and/or PDGF receptor tyrosine kinase
US5604251A (en) * 1992-03-07 1997-02-18 Hoechst Aktiengesellschaft Imidazole derivatives with a biphenylsulfonylurea or biphenylsulfonylurethane side chain and their use as angitensin II receptors
US5618814A (en) * 1993-08-02 1997-04-08 Dr. Karl Thomae Gmbh Trisubstituted pyrimido [5,4-d] pyrimidines for modulating multi-drug resistance and pharmaceutical compositions containing these compounds
US5618829A (en) * 1993-01-28 1997-04-08 Mitsubishi Chemical Corporation Tyrosine kinase inhibitors and benzoylacrylamide derivatives
US5654298A (en) * 1990-04-19 1997-08-05 Imperial Chemical Industries Amine derivatives
US5654307A (en) * 1994-01-25 1997-08-05 Warner-Lambert Company Bicyclic compounds capable of inhibiting tyrosine kinases of the epidermal growth factor receptor family
US5656643A (en) * 1993-11-08 1997-08-12 Rhone-Poulenc Rorer Pharmaceuticals Inc. Bis mono-and bicyclic aryl and heteroaryl compounds which inhibit EGF and/or PDGF receptor tyrosine kinase
US5707989A (en) * 1994-09-07 1998-01-13 Dr. Karl Thomae Gmbh Pyrimido 5,4-D!pyrimidines, medicaments comprising these compounds, their use and processes for their preparation
US5710158A (en) * 1991-05-10 1998-01-20 Rhone-Poulenc Rorer Pharmaceuticals Inc. Aryl and heteroaryl quinazoline compounds which inhibit EGF and/or PDGF receptor tyrosine kinase
US5714493A (en) * 1991-05-10 1998-02-03 Rhone-Poulenc Rorer Pharmaceuticals, Inc. Aryl and heteroaryl quinazoline compounds which inhibit CSF-1R receptor tyrosine kinase
US5721237A (en) * 1991-05-10 1998-02-24 Rhone-Poulenc Rorer Pharmaceuticals Inc. Protein tyrosine kinase aryl and heteroaryl quinazoline compounds having selective inhibition of HER-2 autophosphorylation properties
US5739127A (en) * 1996-11-08 1998-04-14 Bayer Aktiengesellschaft 2,4'-bridged bis-2,4-diaminoquinazolines
US5747498A (en) * 1996-05-28 1998-05-05 Pfizer Inc. Alkynyl and azido-substituted 4-anilinoquinazolines
US5747486A (en) * 1994-11-08 1998-05-05 Takeda Chemical Industries Thienopyridine or thienopyrimidine derivatives and their use
US5760230A (en) * 1996-10-11 1998-06-02 Bayer Aktiengesellschaft 4, 4'-bridged bis-2, 4-diaminoquinazolines
US5874438A (en) * 1996-10-11 1999-02-23 Bayer Aktiengesellschaft 2,2'-bridged bis-2,4-diaminoquinazolines
US5948819A (en) * 1994-04-06 1999-09-07 Shionogi & Co., Ltd α-substituted phenylacetic acid derivative, its production and agricultural fungicide containing it
US5952346A (en) * 1996-10-14 1999-09-14 Hoechst Marion Roussel Deutschland Gmbh Use of non-peptide bradykinin antagonists for the treatment or prevention of Alzheimer's disease
US6080747A (en) * 1999-03-05 2000-06-27 Hughes Institute JAK-3 inhibitors for treating allergic disorders
US6084095A (en) * 1994-01-25 2000-07-04 Warner-Lambert Company Substituted pyrido[3,2-d]pyrimidines capable of inhibiting tyrosine kinases of the epidermal growth factor receptor family
US6124330A (en) * 1996-06-27 2000-09-26 Janssen-Cilag S.A. N-[4-(Heteroarylmethyl)phenyl]-heteroarylamines
US6184226B1 (en) * 1998-08-28 2001-02-06 Scios Inc. Quinazoline derivatives as inhibitors of P-38 α
US6204267B1 (en) * 1997-05-02 2001-03-20 Sugen, Inc. Methods of modulating serine/thereonine protein kinase function with quinazoline-based compounds
US6232312B1 (en) * 1995-06-07 2001-05-15 Cell Pathways, Inc. Method for treating patient having precancerous lesions with a combination of pyrimidopyrimidine derivatives and esters and amides of substituted indenyl acetic acides
US6242196B1 (en) * 1997-12-11 2001-06-05 Dana-Farber Cancer Institute Methods and pharmaceutical compositions for inhibiting tumor cell growth
US6251912B1 (en) * 1997-08-01 2001-06-26 American Cyanamid Company Substituted quinazoline derivatives
US6265425B1 (en) * 1997-12-19 2001-07-24 Janssen Pharmaceutica, N.V. Combination of a RAMBA and a tocopherol
US6284764B1 (en) * 1999-01-27 2001-09-04 Pfizer Inc. Substituted bicyclic derivatives useful as anticancer agents
US6344459B1 (en) * 1996-04-12 2002-02-05 Warner-Lambert Company Irreversible inhibitors of tyrosine kinases
USRE37650E1 (en) * 1991-05-10 2002-04-09 Aventis Pharmacetical Products, Inc. Aryl and heteroaryl quinazoline compounds which inhibit CSF-1R receptor tyrosine kinase
US20020048271A1 (en) * 1998-12-02 2002-04-25 Farzan Rastinejad Methods and composition for restoring conformational stability of a protein of the p53 family
US6384051B1 (en) * 2000-03-13 2002-05-07 American Cyanamid Company Method of treating or inhibiting colonic polyps
US6391874B1 (en) * 1996-07-13 2002-05-21 Smithkline Beecham Corporation Fused heterocyclic compounds as protein tyrosine kinase inhibitors
US20020082270A1 (en) * 2000-08-26 2002-06-27 Frank Himmelsbach Aminoquinazolines which inhibit signal transduction mediated by tyrosine kinases
US6432979B1 (en) * 1999-08-12 2002-08-13 American Cyanamid Company Method of treating or inhibiting colonic polyps and colorectal cancer
US6518283B1 (en) * 1999-05-28 2003-02-11 Celltech R&D Limited Squaric acid derivatives
US20030055068A1 (en) * 2000-12-21 2003-03-20 David Bebbington Pyrazole compounds useful as protein kinase inhibitors
US6552055B2 (en) * 1996-12-11 2003-04-22 Dana-Farber Cancer Institute Methods and pharmaceutical compositions for inhibiting tumor cell growth
US6552027B2 (en) * 1998-05-28 2003-04-22 Parker Hughes Institute Quinazolines for treating brain tumor
US20030087931A1 (en) * 2001-03-23 2003-05-08 Patrick Mailliet Chemical derivatives and their application as antitelomerease agent
US20030087908A1 (en) * 2001-04-11 2003-05-08 Geuns-Meyer Stephanie D. Substituted triazinyl amide derivatives and methods of use
US6562319B2 (en) * 2001-03-12 2003-05-13 Yissum Research Development Company Of The Hebrew University Of Jerusalem Radiolabeled irreversible inhibitors of epidermal growth factor receptor tyrosine kinase and their use in radioimaging and radiotherapy
US6562818B1 (en) * 1997-07-29 2003-05-13 Warner-Lambert Company Irreversible inhibitors of tyrosine kinases
US20030134836A1 (en) * 2001-01-12 2003-07-17 Amgen Inc. Substituted arylamine derivatives and methods of use
US20030144330A1 (en) * 1996-12-11 2003-07-31 Spiegelman Bruce M. Methods and pharmaceutical compositions for inhibiting tumor cell growth
US20030144506A1 (en) * 2001-11-30 2003-07-31 Pfizer Inc. Processes for the preparation of substituted bicyclic derivatives for the treatment of abnormal cell growth
US20030144178A1 (en) * 1998-06-30 2003-07-31 Parker Hughes Institute Method for inhibiting C-jun expression using JAK-3 inhibitors
US20030149062A1 (en) * 2002-02-05 2003-08-07 Boehringer Ingelheim Pharma Gmbh & Co. Kg Use of tyrosine kinase inhibitors for the treatment of inflammatory processes
US20030149045A1 (en) * 1999-08-20 2003-08-07 Fatih M Uckun Therapeutic compounds
US20040014774A1 (en) * 1991-05-10 2004-01-22 Myers Michael R. Aryl and heteroaryl quinazoline compounds which inhibit CSF-1R receptor tyrosine kinase
US20040034044A1 (en) * 2000-11-02 2004-02-19 Masahiko Okano Quinazoline derivatives and drugs
US20040034045A1 (en) * 2000-11-29 2004-02-19 Parker Hughes Institute Inhibitors of thrombin induced platelet aggregation
US20040038856A1 (en) * 2002-05-17 2004-02-26 Sarvajit Chakravarty Treatment of fibroproliferative disorders using TGF-beta inhibitors
US20040043388A1 (en) * 2001-03-02 2004-03-04 Come Jon H. Three hybrid assay system
US6740651B2 (en) * 2000-08-26 2004-05-25 Boehringer Ingelheim Pharma Kg Aminoquinazolines which inhibit signal transduction mediated by tyrosine kinases
US6883375B2 (en) * 2001-06-29 2005-04-26 Harold L. Dunegan Detection of movement of termites in wood by acoustic emission techniques
US6890924B2 (en) * 2000-06-22 2005-05-10 Pfizer Inc Substituted bicyclic derivatives for the treatment of abnormal cell growth
US20050137213A1 (en) * 2003-07-03 2005-06-23 Myriad Genetics, Incorporated Compounds and therapeutical use thereof
US7087613B2 (en) * 1999-11-11 2006-08-08 Osi Pharmaceuticals, Inc. Treating abnormal cell growth with a stable polymorph of N-(3-ethynylphenyl)-6,7-bis(2-methoxyethoxy)-4-quinazolinamine hydrochloride

Family Cites Families (32)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CH421972A (de) 1960-02-02 1966-10-15 Thomae Gmbh Dr K Verfahren zur Herstellung von basisch substituierten Pyrimidincarbonsäuren-(4) bzw. ihren Derivaten
FR8491M (US20070244113A1-20071018-C00053.png) 1968-12-31 1973-07-27
DE2918591A1 (de) * 1979-05-09 1980-11-20 Hoechst Ag Neue 4-substituierte 5,6,7,8-tetrahydrochinoline, ihre herstellung und verwendung
DE3049207A1 (de) 1980-12-27 1982-07-29 Dr. Karl Thomae Gmbh, 7950 Biberach Neue trisubstituierte pyrimido (5,4-d) pyrimidine, ihre herstellung und ihre verwendung als arzneimittel
FR2498187A1 (fr) 1981-01-16 1982-07-23 Rhone Poulenc Sante Procede de preparation d'amino-4 chloro-7 quinoleines
DE3423092A1 (de) 1984-06-22 1986-01-02 Dr. Karl Thomae Gmbh, 7950 Biberach Neue 8-alkylthio-2-piperazino-pyrimido(5,4-d) pyrimidine, ihre herstellung und diese verbindungen enthaltende arzneimittel
IL88507A (en) * 1987-12-03 1993-02-21 Smithkline Beckman Intercredit 2,4-diaminoquinazolines, process for their preparation and pharmaceutical compositions comprising them
CA2015981A1 (en) * 1989-05-10 1990-11-10 Thomas H. Brown Compounds
GB8910722D0 (en) 1989-05-10 1989-06-28 Smithkline Beckman Intercredit Compounds
DE4029648A1 (de) * 1990-09-19 1992-03-26 Hoechst Ag 4-anilino-pyrimidine, verfahren zu ihrer herstellung, sie enthaltende mittel und ihre verwendung als fungizide
BR9205645A (pt) * 1991-02-20 1994-06-07 Pfizer Derivados de 2,4-diaminoquinazolinas para aumentar a atividade antitumoral
US5256781A (en) 1991-10-24 1993-10-26 American Home Products Corporation Substituted quinazolines as angiotensin II antagonists
GB9127252D0 (en) 1991-12-23 1992-02-19 Boots Co Plc Therapeutic agents
US5270466A (en) 1992-06-11 1993-12-14 American Cyanamid Company Substituted quinazoline fungicidal agents
US5565472A (en) 1992-12-21 1996-10-15 Pfizer Inc. 4-aryl-3-(heteroarylureido)-1,2-dihydro-2-oxo-quinoline derivatives as antihypercholesterolemic and antiatherosclerotic agents
DK60593D0 (da) 1993-05-26 1993-05-26 Novo Nordisk As Kemiske forbindelser, deres fremstilling og anvendelse
US6087160A (en) 1993-06-24 2000-07-11 The General Hospital Corporation Programmed cell death genes and proteins
US6313150B1 (en) 1994-04-06 2001-11-06 Shionogi & Co., Ltd. α-substituted phenylacetic acid derivative, its production and agricultural fungicide containing it
EP0889033B1 (en) 1995-09-29 2003-01-08 Shionogi & Co., Ltd. Alpha-substituted benzyl heterocyclic derivatives, intermediates for producing the same, and pesticides containing the same as active ingredient
US5874738A (en) * 1996-07-16 1999-02-23 Saint-Gobain Industrial Ceramics, Inc. Scintillation crystal modules and methods of making the same
CN1093130C (zh) 1996-08-06 2002-10-23 辉瑞大药厂 被取代的吡啶并6,6-二环衍生物
US6002008A (en) 1997-04-03 1999-12-14 American Cyanamid Company Substituted 3-cyano quinolines
AU8748798A (en) * 1997-08-22 1999-03-16 Kyowa Hakko Kogyo Co. Ltd. 4-aminoquinazoline derivatives
KR20010031056A (ko) 1997-10-10 2001-04-16 시토비아 인크. 신규 형광 리포터 분자 및 카스파제에 대한 분석을 비롯한이의 용도
US20020025968A1 (en) * 1998-04-15 2002-02-28 Rifat Pamukcu Method for inhibiting neoplastic cells and related conditions by exposure to 4-aminoquinazoline derivatives
KR20010089171A (ko) 1998-08-21 2001-09-29 추후제출 퀴나졸린 유도체
US6297258B1 (en) 1998-09-29 2001-10-02 American Cyanamid Company Substituted 3-cyanoquinolines
RS49836B (sr) 1999-03-31 2008-08-07 Pfizer Products Inc., Postupci i intermedijeri za dobijanje anti-kancernih jedinjenja
WO2002047690A1 (en) * 2000-12-12 2002-06-20 Cytovia, Inc. Substituted 2-aryl-4-arylaminopyrimidines and analogs as activators of caspases and inducers of apoptosis and the use thereof
EP1603884A4 (en) * 2003-02-28 2008-05-28 Encysive Pharmaceuticals Inc PYRIDINE, PYRIMIDINE, QUINOLINE, QUINAZOLINE AND NAPHTHALENE UROTENSIN II RECEPTOR ANTAGONISTS
WO2006074147A2 (en) * 2005-01-03 2006-07-13 Myriad Genetics, Inc. Nitrogen containing bicyclic compounds and therapeutical use thereof
WO2005037825A2 (en) * 2003-10-14 2005-04-28 Arizona Board Of Regents On Behalf Of The University Of Arizona Protein kinase inhibitors

Patent Citations (99)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2883382A (en) * 1957-08-29 1959-04-21 Parke Davis & Co Benzo-quinolylamino-2-[di(beta-chloroethyl)aminomethyl]-phenols
US3031450A (en) * 1959-04-30 1962-04-24 Thomae Gmbh Dr K Substituted pyrimido-[5, 4-d]-pyrimidines
US3502681A (en) * 1968-04-10 1970-03-24 Roussel Uclaf 7- or 8-chloro-4-phenylamino-chloroquinolines
US3632761A (en) * 1969-08-08 1972-01-04 Upjohn Co Method of obtaining antihypertensive and antianxiety effects
US3971783A (en) * 1973-03-07 1976-07-27 Pfizer Inc. 4-Aminoquinazoline derivatives as cardiac stimulants
US4025629A (en) * 1974-01-07 1977-05-24 The Upjohn Company P-(trifluoromethylquinolylamino)benzamides, pharmaceutical dosage forms and method of treatment
US4322420A (en) * 1978-09-11 1982-03-30 Sankyo Company Limited Method of using 4-anilinoquinazoline derivatives as analgesic and anti-inflammatory agents
US4464375A (en) * 1978-09-11 1984-08-07 Sankyo Co., Ltd. 4-Anilinoquinazoline compounds and pharmaceutical compositions thereof
US4435003A (en) * 1980-01-31 1984-03-06 Ciba-Geigy Corporation Chromogenic quinazolines
US4675047A (en) * 1980-07-01 1987-06-23 Alexander Serban Substituted phenylamine- and phenyloxy-quinazolines as herbicides
US4510307A (en) * 1980-08-20 1985-04-09 Asahi Kasei Kogyo Kabushiki Kaisha 6-Quinazolinesulfonyl derivatives and process for preparation thereof
US5294622A (en) * 1988-01-29 1994-03-15 Dowelanco Substituted quinolines and cinnolines
US5145843A (en) * 1988-01-29 1992-09-08 Dowelanco Quinoline and cinnoline fungicides
US5114939A (en) * 1988-01-29 1992-05-19 Dowelanco Substituted quinolines and cinnolines as fungicides
US5240940A (en) * 1988-01-29 1993-08-31 Dowelanco Quinoline and cinnoline fungicide compositions
US5223505A (en) * 1989-04-21 1993-06-29 Imperial Chemical Industries Plc Pyrimidine derivatives
US5654298A (en) * 1990-04-19 1997-08-05 Imperial Chemical Industries Amine derivatives
US20040014774A1 (en) * 1991-05-10 2004-01-22 Myers Michael R. Aryl and heteroaryl quinazoline compounds which inhibit CSF-1R receptor tyrosine kinase
US5646153A (en) * 1991-05-10 1997-07-08 Rhone-Poulenc Rorer Pharmaceuticals Inc. Bis mono- and bicyclic aryl and heteroaryl compounds which inhibit EGF and/or PDGF receptor tyrosine kinase
USRE36256E (en) * 1991-05-10 1999-07-20 Rhone-Poulenc Rorer Pharmaceuticals, Inc. Bis mono- and bicyclic aryl and heteroaryl compounds which inhibit EGF and/or PDGF receptor tyrosine kinase
US5409930A (en) * 1991-05-10 1995-04-25 Rhone-Poulenc Rorer Pharmaceuticals Inc. Bis mono- and bicyclic aryl and heteroaryl compounds which inhibit EGF and/or PDGF receptor tyrosine kinase
US6057320A (en) * 1991-05-10 2000-05-02 Aventis Pharmaceuticals Products Inc. Bis mono-and bicyclic aryl and heteroaryl compounds which inhibit EGF and/or PDGF receptor tyrosine kinase
US5480883A (en) * 1991-05-10 1996-01-02 Rhone-Poulenc Rorer Pharmaceuticals Inc. Bis mono- and bicyclic aryl and heteroaryl compounds which inhibit EGF and/or PDGF receptor tyrosine kinase
US5721237A (en) * 1991-05-10 1998-02-24 Rhone-Poulenc Rorer Pharmaceuticals Inc. Protein tyrosine kinase aryl and heteroaryl quinazoline compounds having selective inhibition of HER-2 autophosphorylation properties
US5714493A (en) * 1991-05-10 1998-02-03 Rhone-Poulenc Rorer Pharmaceuticals, Inc. Aryl and heteroaryl quinazoline compounds which inhibit CSF-1R receptor tyrosine kinase
US5710158A (en) * 1991-05-10 1998-01-20 Rhone-Poulenc Rorer Pharmaceuticals Inc. Aryl and heteroaryl quinazoline compounds which inhibit EGF and/or PDGF receptor tyrosine kinase
US5795889A (en) * 1991-05-10 1998-08-18 Rhone-Poulenc Rorer Pharmaceuticals Inc. Bis mono- and bicyclic aryl and heteroaryl compounds which inhibit EGF and/or PDGF receptor tyrosine kinase
USRE37650E1 (en) * 1991-05-10 2002-04-09 Aventis Pharmacetical Products, Inc. Aryl and heteroaryl quinazoline compounds which inhibit CSF-1R receptor tyrosine kinase
US5276148A (en) * 1991-05-23 1994-01-04 Basf Aktiengesellschaft Phenylazobenzenes or naphthylazobenzenes having a plurality of fiber-reactive groups
US5236925A (en) * 1991-10-24 1993-08-17 American Home Products Corporation Fused pyrimidines as angiotensin II antagonists
US5187168A (en) * 1991-10-24 1993-02-16 American Home Products Corporation Substituted quinazolines as angiotensin II antagonists
US5330989A (en) * 1991-10-24 1994-07-19 American Home Products Corporation Heterocycles substituted with biphenyl-3-cyclobutene-1,2-dione derivatives
US5604251A (en) * 1992-03-07 1997-02-18 Hoechst Aktiengesellschaft Imidazole derivatives with a biphenylsulfonylurea or biphenylsulfonylurethane side chain and their use as angitensin II receptors
US5436233A (en) * 1992-07-15 1995-07-25 Ono Pharmaceutical Co., Ltd. 4-aminoquinazoline derivatives
US5618829A (en) * 1993-01-28 1997-04-08 Mitsubishi Chemical Corporation Tyrosine kinase inhibitors and benzoylacrylamide derivatives
US5618814A (en) * 1993-08-02 1997-04-08 Dr. Karl Thomae Gmbh Trisubstituted pyrimido [5,4-d] pyrimidines for modulating multi-drug resistance and pharmaceutical compositions containing these compounds
US5656643A (en) * 1993-11-08 1997-08-12 Rhone-Poulenc Rorer Pharmaceuticals Inc. Bis mono-and bicyclic aryl and heteroaryl compounds which inhibit EGF and/or PDGF receptor tyrosine kinase
US6713484B2 (en) * 1994-01-25 2004-03-30 Warner-Lambert Company Bicyclic compounds capable of inhibiting tyrosine kinases of the epidermal growth factor receptor family
US6265410B1 (en) * 1994-01-25 2001-07-24 Warner-Lambert Company Bicyclic compounds capable of inhibiting tyrosine kinases of the epidermal growth factor receptor family
US5654307A (en) * 1994-01-25 1997-08-05 Warner-Lambert Company Bicyclic compounds capable of inhibiting tyrosine kinases of the epidermal growth factor receptor family
US6521620B1 (en) * 1994-01-25 2003-02-18 Warner-Lambert Company Bicyclic compounds capable of inhibiting tyrosine kinases of the epidermal growth factor receptor family
US6084095A (en) * 1994-01-25 2000-07-04 Warner-Lambert Company Substituted pyrido[3,2-d]pyrimidines capable of inhibiting tyrosine kinases of the epidermal growth factor receptor family
US5948819A (en) * 1994-04-06 1999-09-07 Shionogi & Co., Ltd α-substituted phenylacetic acid derivative, its production and agricultural fungicide containing it
US5707989A (en) * 1994-09-07 1998-01-13 Dr. Karl Thomae Gmbh Pyrimido 5,4-D!pyrimidines, medicaments comprising these compounds, their use and processes for their preparation
US5747486A (en) * 1994-11-08 1998-05-05 Takeda Chemical Industries Thienopyridine or thienopyrimidine derivatives and their use
US6232312B1 (en) * 1995-06-07 2001-05-15 Cell Pathways, Inc. Method for treating patient having precancerous lesions with a combination of pyrimidopyrimidine derivatives and esters and amides of substituted indenyl acetic acides
US6602863B1 (en) * 1996-04-12 2003-08-05 Warner-Lambert Company Irreversible inhibitors of tyrosine kinases
US6344459B1 (en) * 1996-04-12 2002-02-05 Warner-Lambert Company Irreversible inhibitors of tyrosine kinases
US5747498A (en) * 1996-05-28 1998-05-05 Pfizer Inc. Alkynyl and azido-substituted 4-anilinoquinazolines
US6124330A (en) * 1996-06-27 2000-09-26 Janssen-Cilag S.A. N-[4-(Heteroarylmethyl)phenyl]-heteroarylamines
US6391874B1 (en) * 1996-07-13 2002-05-21 Smithkline Beecham Corporation Fused heterocyclic compounds as protein tyrosine kinase inhibitors
US5760230A (en) * 1996-10-11 1998-06-02 Bayer Aktiengesellschaft 4, 4'-bridged bis-2, 4-diaminoquinazolines
US5874438A (en) * 1996-10-11 1999-02-23 Bayer Aktiengesellschaft 2,2'-bridged bis-2,4-diaminoquinazolines
US5952346A (en) * 1996-10-14 1999-09-14 Hoechst Marion Roussel Deutschland Gmbh Use of non-peptide bradykinin antagonists for the treatment or prevention of Alzheimer's disease
US5739127A (en) * 1996-11-08 1998-04-14 Bayer Aktiengesellschaft 2,4'-bridged bis-2,4-diaminoquinazolines
US20030144330A1 (en) * 1996-12-11 2003-07-31 Spiegelman Bruce M. Methods and pharmaceutical compositions for inhibiting tumor cell growth
US6552055B2 (en) * 1996-12-11 2003-04-22 Dana-Farber Cancer Institute Methods and pharmaceutical compositions for inhibiting tumor cell growth
US20010014679A1 (en) * 1997-05-02 2001-08-16 Tang Peng C. Methods of modulating serine/threonine protein kinase function with quinazoline-based compounds
US6204267B1 (en) * 1997-05-02 2001-03-20 Sugen, Inc. Methods of modulating serine/thereonine protein kinase function with quinazoline-based compounds
US6562818B1 (en) * 1997-07-29 2003-05-13 Warner-Lambert Company Irreversible inhibitors of tyrosine kinases
US6251912B1 (en) * 1997-08-01 2001-06-26 American Cyanamid Company Substituted quinazoline derivatives
US6242196B1 (en) * 1997-12-11 2001-06-05 Dana-Farber Cancer Institute Methods and pharmaceutical compositions for inhibiting tumor cell growth
US6265425B1 (en) * 1997-12-19 2001-07-24 Janssen Pharmaceutica, N.V. Combination of a RAMBA and a tocopherol
US6552027B2 (en) * 1998-05-28 2003-04-22 Parker Hughes Institute Quinazolines for treating brain tumor
US20030144178A1 (en) * 1998-06-30 2003-07-31 Parker Hughes Institute Method for inhibiting C-jun expression using JAK-3 inhibitors
US20030069248A1 (en) * 1998-08-28 2003-04-10 Sarvajit Chakravarty Quinazoline derivatives as medicaments
US6184226B1 (en) * 1998-08-28 2001-02-06 Scios Inc. Quinazoline derivatives as inhibitors of P-38 α
US6277989B1 (en) * 1998-08-28 2001-08-21 Scios, Inc. Quinazoline derivatives as medicaments
US20020048271A1 (en) * 1998-12-02 2002-04-25 Farzan Rastinejad Methods and composition for restoring conformational stability of a protein of the p53 family
US6541481B2 (en) * 1999-01-27 2003-04-01 Pfizer Inc Substituted bicyclic derivatives useful as anticancer agents
US6284764B1 (en) * 1999-01-27 2001-09-04 Pfizer Inc. Substituted bicyclic derivatives useful as anticancer agents
US6177433B1 (en) * 1999-03-05 2001-01-23 Parker Hughes Institute JAK-3 inhibitors for treating allergic disorders
US6080747A (en) * 1999-03-05 2000-06-27 Hughes Institute JAK-3 inhibitors for treating allergic disorders
US6080748A (en) * 1999-03-05 2000-06-27 Parker Hughes Institute Therapeutic use of JAK-3 inhibitors
US6518283B1 (en) * 1999-05-28 2003-02-11 Celltech R&D Limited Squaric acid derivatives
US20030162799A1 (en) * 1999-05-28 2003-08-28 Langham Barry John Squaric acid derivatives
US6432979B1 (en) * 1999-08-12 2002-08-13 American Cyanamid Company Method of treating or inhibiting colonic polyps and colorectal cancer
US20030149045A1 (en) * 1999-08-20 2003-08-07 Fatih M Uckun Therapeutic compounds
US7087613B2 (en) * 1999-11-11 2006-08-08 Osi Pharmaceuticals, Inc. Treating abnormal cell growth with a stable polymorph of N-(3-ethynylphenyl)-6,7-bis(2-methoxyethoxy)-4-quinazolinamine hydrochloride
US6384051B1 (en) * 2000-03-13 2002-05-07 American Cyanamid Company Method of treating or inhibiting colonic polyps
US6890924B2 (en) * 2000-06-22 2005-05-10 Pfizer Inc Substituted bicyclic derivatives for the treatment of abnormal cell growth
US20020082270A1 (en) * 2000-08-26 2002-06-27 Frank Himmelsbach Aminoquinazolines which inhibit signal transduction mediated by tyrosine kinases
US6740651B2 (en) * 2000-08-26 2004-05-25 Boehringer Ingelheim Pharma Kg Aminoquinazolines which inhibit signal transduction mediated by tyrosine kinases
US20040034044A1 (en) * 2000-11-02 2004-02-19 Masahiko Okano Quinazoline derivatives and drugs
US20040034045A1 (en) * 2000-11-29 2004-02-19 Parker Hughes Institute Inhibitors of thrombin induced platelet aggregation
US20030055068A1 (en) * 2000-12-21 2003-03-20 David Bebbington Pyrazole compounds useful as protein kinase inhibitors
US20030105090A1 (en) * 2000-12-21 2003-06-05 David Bebbington Pyrazole compounds useful as protein kinase inhibitors
US6727251B2 (en) * 2000-12-21 2004-04-27 Vertex Pharmaceuticals Incorporated Pyrazole compounds useful as protein kinase inhibitors
US20030134836A1 (en) * 2001-01-12 2003-07-17 Amgen Inc. Substituted arylamine derivatives and methods of use
US20040043388A1 (en) * 2001-03-02 2004-03-04 Come Jon H. Three hybrid assay system
US6562319B2 (en) * 2001-03-12 2003-05-13 Yissum Research Development Company Of The Hebrew University Of Jerusalem Radiolabeled irreversible inhibitors of epidermal growth factor receptor tyrosine kinase and their use in radioimaging and radiotherapy
US20030087931A1 (en) * 2001-03-23 2003-05-08 Patrick Mailliet Chemical derivatives and their application as antitelomerease agent
US20030087908A1 (en) * 2001-04-11 2003-05-08 Geuns-Meyer Stephanie D. Substituted triazinyl amide derivatives and methods of use
US6864255B2 (en) * 2001-04-11 2005-03-08 Amgen Inc. Substituted triazinyl amide derivatives and methods of use
US6883375B2 (en) * 2001-06-29 2005-04-26 Harold L. Dunegan Detection of movement of termites in wood by acoustic emission techniques
US20030144506A1 (en) * 2001-11-30 2003-07-31 Pfizer Inc. Processes for the preparation of substituted bicyclic derivatives for the treatment of abnormal cell growth
US20030149062A1 (en) * 2002-02-05 2003-08-07 Boehringer Ingelheim Pharma Gmbh & Co. Kg Use of tyrosine kinase inhibitors for the treatment of inflammatory processes
US20040038856A1 (en) * 2002-05-17 2004-02-26 Sarvajit Chakravarty Treatment of fibroproliferative disorders using TGF-beta inhibitors
US20050137213A1 (en) * 2003-07-03 2005-06-23 Myriad Genetics, Incorporated Compounds and therapeutical use thereof

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7989462B2 (en) 2003-07-03 2011-08-02 Myrexis, Inc. 4-arylamin-or-4-heteroarylamino-quinazolines and analogs as activators of caspases and inducers of apoptosis and the use thereof
US20080004297A1 (en) * 2003-07-03 2008-01-03 Myriad Genetics, Inc. Compounds and therapeutical use thereof
US20080032974A1 (en) * 2003-07-03 2008-02-07 Myriad Genetics, Incorporated Compounds and therapeutical use thereof
US8309562B2 (en) 2003-07-03 2012-11-13 Myrexis, Inc. Compounds and therapeutical use thereof
US20070244114A1 (en) * 2004-07-06 2007-10-18 Myriad Genetics, Incorporated Compounds and therapeutical use thereof
US8258145B2 (en) 2005-01-03 2012-09-04 Myrexis, Inc. Method of treating brain cancer
US20080051398A1 (en) * 2005-01-03 2008-02-28 Myriad Genetics, Inc. Method of treating brain cancer
US20080319048A1 (en) * 2007-06-22 2008-12-25 Scidose Llc Solubilized formulation of docetaxel without tween 80
US20110092580A1 (en) * 2009-10-19 2011-04-21 Scidose Llc Docetaxel formulations with lipoic acid and/or dihydrolipoic acid
US20110092579A1 (en) * 2009-10-19 2011-04-21 Scidose Llc Solubilized formulation of docetaxel
US7772274B1 (en) 2009-10-19 2010-08-10 Scidose, Llc Docetaxel formulations with lipoic acid
US8541465B2 (en) 2009-10-19 2013-09-24 Scidose, Llc Docetaxel formulations with lipoic acid and/or dihydrolipoic acid
US8912228B2 (en) 2009-10-19 2014-12-16 Scidose Llc Docetaxel formulations with lipoic acid

Also Published As

Publication number Publication date
KR20060052775A (ko) 2006-05-19
WO2005003100A3 (en) 2005-05-12
US20050137213A1 (en) 2005-06-23
NZ544472A (en) 2009-04-30
CN1984660B (zh) 2010-12-15
US7618975B2 (en) 2009-11-17
JP5129957B2 (ja) 2013-01-30
US20080032974A1 (en) 2008-02-07
EP1660092A2 (en) 2006-05-31
CN1984660A (zh) 2007-06-20
US20070208044A1 (en) 2007-09-06
KR101218213B1 (ko) 2013-01-04
AU2004253967A1 (en) 2005-01-13
US20170050937A1 (en) 2017-02-23
US20070249601A1 (en) 2007-10-25
AU2004253967A8 (en) 2010-02-04
JP2012207044A (ja) 2012-10-25
AU2004253967B2 (en) 2010-02-18
WO2005003100A2 (en) 2005-01-13
CA2531327A1 (en) 2005-01-13
JP2007524637A (ja) 2007-08-30
US20080039479A1 (en) 2008-02-14

Similar Documents

Publication Publication Date Title
US7618975B2 (en) 4-arylamino-quinazolines and analogs as activators of caspases and inducers of apoptosis and the use thereof
US8309562B2 (en) Compounds and therapeutical use thereof
US7989462B2 (en) 4-arylamin-or-4-heteroarylamino-quinazolines and analogs as activators of caspases and inducers of apoptosis and the use thereof
US8258145B2 (en) Method of treating brain cancer
US20070099877A1 (en) N-aryl-thienopyrimidin-4-amines and analogs as activators of caspases and inducers of apoptosis and the use thereof
CA2592971A1 (en) Method of treating brain cancer
US20070213305A1 (en) N-alkyl-N-aryl-thienopyrimidin-4-amines and analogs as activators of caspases and inducers of apoptosis and the use thereof
WO2008057402A2 (en) N-aryl-isoxazolopyrimidin-4-amines and related compounds as activators of caspases and inducers of apoptosis and the use thereof
HU223313B1 (hu) Kinazolinszármazékok, ezeket tartalmazó gyógyszerkészítmények és eljárás előállításukra
US7842805B2 (en) Pharmaceutical compounds as activators of caspases and inducers of apoptosis and the use thereof
US20070244114A1 (en) Compounds and therapeutical use thereof
US20090280133A1 (en) Pharmaceutical compounds as inhibitors of cell proliferation and the use thereof
Yilmaz Synthesis and biological studies of some 3-substituted-2, 4 (1H, 3H)-quinazolinedione derivatives

Legal Events

Date Code Title Description
AS Assignment

Owner name: MYRIAD PHARMACEUTICALS, INC., UTAH

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:MYRIAD GENETICS, INC.;REEL/FRAME:023138/0115

Effective date: 20090630

STCB Information on status: application discontinuation

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