WO2009105500A1 - Compounds that are erk inhibitors - Google Patents

Compounds that are erk inhibitors Download PDF

Info

Publication number
WO2009105500A1
WO2009105500A1 PCT/US2009/034447 US2009034447W WO2009105500A1 WO 2009105500 A1 WO2009105500 A1 WO 2009105500A1 US 2009034447 W US2009034447 W US 2009034447W WO 2009105500 A1 WO2009105500 A1 WO 2009105500A1
Authority
WO
WIPO (PCT)
Prior art keywords
substituted
group
triazolyl
nitrogen
compound
Prior art date
Application number
PCT/US2009/034447
Other languages
French (fr)
Inventor
Alan B. Cooper
Yang Nan
Yongqi Deng
Gerald W. Shipps, Jr.
Neng-Yang Shih
Hugh Y. Zhu
Joseph M. Kelly
Subrahmanyam Gudipati
Ronald J. Doll
Mehul F. Patel
Jagdish A. Desai
James J-S Wang
Sunil Paliwal
Hon-Chung Tsui
Sobhana Babu Boga
Abdul-Basit Alhassan
Xiaolei Gao
Liang Zhu
Xin Yao
Original Assignee
Schering Corporation
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
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=40601237&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=WO2009105500(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority to CN200980113870.7A priority Critical patent/CN102015693B/en
Priority to AU2009215534A priority patent/AU2009215534B8/en
Priority to BRPI0908120A priority patent/BRPI0908120A8/en
Priority to MX2010009268A priority patent/MX2010009268A/en
Priority to JP2010547732A priority patent/JP5276676B2/en
Priority to US12/918,099 priority patent/US8716483B2/en
Priority to ES09712601.5T priority patent/ES2556353T3/en
Application filed by Schering Corporation filed Critical Schering Corporation
Priority to EP09712601.5A priority patent/EP2260031B1/en
Priority to NZ587504A priority patent/NZ587504A/en
Priority to CA2714479A priority patent/CA2714479A1/en
Priority to RU2010138635/04A priority patent/RU2525389C2/en
Publication of WO2009105500A1 publication Critical patent/WO2009105500A1/en
Priority to IL207530A priority patent/IL207530A/en
Priority to ZA2010/05909A priority patent/ZA201005909B/en

Links

Classifications

    • 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
    • C07D401/14Heterocyclic 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 three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the processes involved in tumor growth, progression, and metastasis are mediated by signaling pathways that are activated in cancer cells.
  • the ERK pathway plays a central role in regulating mammalian cell growth by relaying extracellular signals from ligand-bound cell surface tyrosine kinase receptors such as erbB family, PDGF, FGF, and VEGF receptor tyrosine kinase.
  • ligand-bound cell surface tyrosine kinase receptors such as erbB family, PDGF, FGF, and VEGF receptor tyrosine kinase.
  • Activation of the ERK pathway is via a cascade of phosphorylation events that begins with activation of Ras.
  • Activation of Ras leads to the recruitment and activation of Raf, a serine-threonine kinase.
  • Raf Activated Raf then phosphorylates and activates MEK1/2, which then phosphorylates and activates ERK1/2.
  • ERK1/2 phosphorylates several downstream targets involved in a multitude of cellular events including cytoskeletal changes and transcriptional activation.
  • the ERK/MAPK pathway is one of the most important for cell proliferation, and it is believed that the ERK/MAPK pathway is frequently activated in many tumors.
  • Ras genes, which are upstream of ERK1/2, are mutated in several cancers including colorectal, melanoma, breast and pancreatic tumors. The high Ras activity is accompanied by elevated ERK activity in many human tumors.
  • BRAF a serine-threonine kinase of the Raf family
  • ERK1/2 signalling pathway is an attractive pathway for anticancer therapies in a broad spectrum of human tumours.
  • small-molecules i.e., compounds that inhibit ERK activity (i.e., ERK1 and ERK2 activity), which small- molecules would be useful for treating a broad spectrum of cancers, such as, for example, melanoma, pancreatic cancer, thryroid cancer, colorectal cancer, lung cancer, breast cancer, and ovarian cancer.
  • ERK activity i.e., ERK1 and ERK2 activity
  • cancers such as, for example, melanoma, pancreatic cancer, thryroid cancer, colorectal cancer, lung cancer, breast cancer, and ovarian cancer.
  • This invention provides compounds that inhibit the activity of ERK1 and/or the activity of ERK2.
  • the compounds of this invention also inhibit the phosphorylation of ERK1 and ERK2.
  • this invention provides compounds that are ERK inhibitors (i.e., ERK1 inhibitors and/or ERK2 inhibitors), said compounds being of the formula 1.0:
  • Q is a tetrahydropyridinyl (e.g., 1 ,2,3,6-tetrahydopyridinyl), or a substituted tetrahydropyridinyl (e.g., a substituted 1 ,2,3,6-tetrahydo-pyridinyl); and R 1 and R 2 are as defined below.
  • This invention provides compounds of formula 1.0.
  • This invention provides compounds of formula 1.0 in pure or isolated form. This invention provides pharmaceutically acceptable salts of the compounds of formula 1.0.
  • This invention provides solvates of the compounds of formula 1.0.
  • This invention provides compounds of formula 1.0 wherein from one up to the total number of hydrogens are deuterium.
  • This invention provides compounds of formula 1.0 wherein at least one H is deuterium.
  • This invention provides compounds of formula 1.0 wherein 1 to 5 H are deuterium. This invention provides compounds of formula 1.0 wherein one H is deuterium.
  • This invention provides compounds A1 to A16 and A18 to A48.
  • This invention provides compounds A1 to A16 and A18 to A30.
  • This invention provides compounds A1 to A16 and A18 to A26. This invention provides compounds A31 to A48.
  • This invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount of at least one compound of formula 1.0 and a pharmaceutically acceptable carrier.
  • This invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount of at least one compound of formula 1.0 and an effective amount of at least one other pharmaceutically active ingredient (such as, for example, a chemotherapeutic agent), and a pharmaceutically acceptable carrier.
  • This invention also provides a method of inhibiting ERK (i.e., inhibiting the activity of ERK) in a patient in need of such treatment comprising administering to said patient an effective amount of at least one compound of formula 1.0.
  • This invention also provides a method of inhibiting ERK1 (i.e., inhibiting the activity of ERK1 ) in a patient in need of such treatment comprising administering to said patient an effective amount of at least one compound of formula 1.0.
  • This invention also provides a method of inhibiting ERK2 (i.e., inhibiting the activity of ERK2) in a patient in need of such treatment comprising administering to said patient an effective amount of at least one compound of formula 1.0.
  • This invention also provides a method of inhibiting ERK1 and ERK2 (i.e., inhibiting the activity of ERK1 and ERK2) in a patient in need of such treatment comprising administering to said patient an effective amount of at least one compound of formula 1.0.
  • This invention also provides a method for treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one compound of formula 1.0.
  • This invention also provides a method for treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one compound of formula 1.0.
  • This invention also provides a method for treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one compound of formula 1.0, in combination with an effective amount of at least one chemotherapeutic agent.
  • This invention also provides a method for treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one compound of formula 1.0, in combination with an effective amount of at least one chemotherapeutic agent.
  • This invention also provides a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one compound of formula 1.0 in combination with at least one signal transduction inhibitor.
  • This invention also provides a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least compound of formula 1.0 in combination with at least one signal transduction inhibitor.
  • the compounds of this invention can be administered concurrently or sequentially (i.e., consecutively) with the chemotherapeutic agents or the signal transduction inhibitor.
  • the methods of treating cancers described herein can optionally include the administration of an effective amount of radiation (i.e., the methods of treating cancers described herein optionally include the administration of radiation therapy).
  • a drug or compound in a specified period is per treatment cycle. For example, once a day means once per day of each day of the treatment cycle. Twice a day means twice per day each day of the treatment cycle. Once a week means one time per week during the treatment cycle. Once every three weeks means once per three weeks during the treatment cycle.
  • the following abbreviations have the following meanings unless defined otherwise:
  • anti-cancer agent means a drug (medicament or pharmaceutically active ingredient) for treating cancer
  • anti-plastic agent means a drug (medicament or pharmaceutically active ingredient) for treating cancer (i.e., a chemotherapeutic agent);
  • at least one as used in reference to the number of compounds of this invention means for example 1 -6, generally 1 -4, more generally 1 , 2 or 3, and usually one or two, and more usually one; thus, in one example "at least one” means one, in another example “at least one” means two, and in another example “at least one” means three;
  • "at least one”, as used in reference to the number of chemotherapeutic agents used, means for example 1 -6, generally 1-4, more generally 1 , 2 or 3, and usually one or two, or one; thus, in one example "at least one” means one, in another example “at least one” means two, and in another example “at least one” means three;
  • chemotherapeutic agent means a drug (medicament or pharmaceutically active ingredient) for treating cancer (i.e., and antineoplastic agent);
  • “compound” with reference to the antineoplastic agents includes the agents that are antibodies;
  • “concurrently” means (1) simultaneously in time (e.g., at the same time); or (2) at different times during the course of a common treatment schedule; "consecutively” means one following the other; "different” as used in the phrase “different antineoplastic agents” means that the agents are not the same compound or structure; preferably, “different” as used in the phrase “different antineoplastic agents” means not from the same class of antineoplastic agents; for example, one antineoplastic agent is a taxane, and another antineoplastic agent is a platinum coordinator compound;
  • an effective amount or a therapeutically effective amount is meant to describe an amount of compound or a composition of the present invention, or an amount of radiation, effective in treating or inhibiting the diseases or conditions described herein, and thus producing the desired therapeutic, ameliorative, inhibitory or preventative effect; thus, for example, in the methods of treating cancer described herein "effective amount” (or “therapeutically effective amount” ) means, for example, the amount of the compound (or drug), or radiation, that results in: (a) the reduction, alleviation or disappearance of one or more symptoms caused by the cancer, (b) the reduction of tumor size, (c) the elimination of the tumor, and/or (d) long-term disease stabilization (growth arrest) of the tumor; for example, in the treatment of lung cancer (e.g., non small cell lung cancer) a therapeutically effective amount is that amount that alleviates or eliminates cough, shortness of breath and/or pain; also, for example, an effective amount, or a therapeutically effective amount of the ERK inhibitor (i.e., a compound of this
  • patient means an animal, such as a mammal (e.g., a human being, and preferably a human being);
  • prodrug means compounds that are rapidly transformed, for example, by hydrolysis in blood, in vivo to the parent compound, i.e., to the compounds of formula 1.0 or to a salt and/or to a solvate thereof; a thorough discussion is provided in T. Higuchi and V. Stella, Pro-drugs as Novel Delivery Systems, Vol. 14 of the A.C.S. Symposium Series, and in Edward B.
  • the scope of this invention includes Prodrugs of the novel compounds of this invention; sequentially-represents (1) administration of one component of the method ((a) compound of the invention, or (b) chemotherapeutic agent, signal transduction inhibitor and/or radiation therapy) followed by administration of the other component or components; after adminsitration of one component, the next component can be administered substantially immediately after the first component, or the next component can be administered after an effective time period after the first component; the effective time period is the amount of time given for realization of maximum benefit from the administration of the first component; and
  • solvate means a physical association of a compound of this invention with one or more solvent molecules; this physical association involves varying degrees of ionic and covalent bonding, including hydrogen bonding; in certain instances the solvate will be capable of isolation, for example when one or more solvent molecules are incorporated in the crystal lattice of the crystalline solid; “solvate” encompasses both solution-phase and isolatable solvates; non-limiting examples of suitable solvates include ethanolates, methanolates, and the like; "hydrate” is a solvate wherein the solvent molecule is H 2 O.
  • acyl means an H-C(O)-, alkyl-C(O)-, alkenyl-C(O)-, Alkynyl-C(O)-, cycloalkyl-C(O)-, cycloalkenyl-C(O)-, or cycloalkynyl-C(O)- group in which the various groups are as defined below (and as defined below, the alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl and cycloalkynyl moieties can be substituted); the bond to the parent moiety is through the carbonyl; preferred acyls contain a lower alkyl; Non- limiting examples of suitable acyl groups include formyl, acetyl, propanoyl, 2- methylpropanoyl, butanoyl and cyclohexanoyl;
  • alkenyl means an aliphatic hydrocarbon group (chain) comprising at least one carbon to carbon double bond, wherein the chain can be straight or branched, and wherein said group comprises about 2 to about 15 carbon atoms; Preferred alkenyl groups comprise about 2 to about 12 carbon atoms in the chain; and more preferably about 2 to about 6 carbon atoms in the chain; branched means that one or more lower alkyl groups, such as methyl, ethyl or propyl, or alkenyl_groups are attached to a linear alkenyl chain; "lower alkenyl” means an alkenyl group comprising about 2 to about 6 carbon atoms in the chain, and the chain can be straight or branched; the term "substituted alkenyl” means that the alkenyl group is substituted by one or more independently selected substituents, and each substituent is independently selected from the group consisting of: halo, alkyl, aryl, cycloalkyl, cyano
  • alkoxycarbonyl means an alkyl-O-CO- group (i.e., the bond to the parent moiety is through the carbonyl) wherein the alkyl group is unsubstituted or substituted as previously defined; non-limiting examples of suitable alkoxycarbonyl groups include methoxycarbonyl and ethoxycarbonyl;
  • alkyl (including the alkyl portions of other moieties, such as trifluoroalkyl and alkyloxy) means an aliphatic hydrocarbon group (chain) that can be straight or branched wherein said group comprises about 1 to about 20 carbon atoms in the chain; preferred alkyl groups comprise about 1 to about 12 carbon atoms in the chain; more preferred alkyl groups comprise about 1 to about 6 carbon atoms in the chain; branched means that one or more lower alkyl groups, such as methyl, ethyl or propyl, are attached to a linear alkyl chain; "lower alkyl” means a group comprising about 1 to about 6 carbon atoms in the chain, and said chain can be straight or branched; the term "substituted alkyl” means that the alkyl group is substituted by one or more independently selected substituents, and wherein each substituent is independently selected from the group consisting of: halo, aryl, cycloalkyl, cyano,
  • alkylaryl (or alkaryl) means an alkyl-aryl- group (i.e., the bond to the parent moiety is through the aryl group) wherein the alkyl group is unsubstituted or substituted as defined above, and the aryl group is unsubstituted or substituted as defined below; preferred alkylaryls comprise a lower alkyl group; non-limiting examples of suitable alkylaryl groups include o-tolyl, p-tolyl and xylyl;
  • alkylheteroaryl means an alkyl-heteroaryl- group (i.e., the bond to the parent moiety is through the heteroaryl group) wherein the alkyl is unsubstituted or substituted as defined above and the heteroaryl group is unsubstituted or substituted as defined below;
  • alkylsulfinyl means an alkyl-S(O)- group (i.e., the bond to the parent moiety is through the sulfinyl) wherein the alkyl group is unsubstituted or substituted as previously defined; preferred groups are those in which the alkyl group is lower alkyl;
  • alkylsulfonyl means an alkyl-S(O 2 )- group (i.e., the bond to the parent moiety is through the sulfonyl) wherein the alkyl group is unsubstituted or substituted as previously defined; preferred groups are those in which the alkyl group is lower alkyl;
  • alkylthio means an alkyl-S- group (i.e., the bond to the parent moiety is through the sulfur) wherein the alkyl group is unsubstituted or substituted as previously described; non-limiting examples of suitable alkylthio groups include methylthio, ethylthio, i-propylthio and heptylthio; "alkynyl” means an aliphatic hydrocarbon group (chain) comprising at least one carbon to carbon triple bond, wherein the chain can be straight or branched, and wherein the group comprises about 2 to about 15 carbon atoms in the; preferred alkynyl groups comprise about 2 to about 12 carbon atoms in the chain; and more preferably about 2 to about 4 carbon atoms in the chain; Branched means that one or more lower alkyl groups, such as methyl, ethyl or propyl, are attached to a linear alkynyl chain; "lower alkynyl” means an alkynyl group
  • amino means a -NH 2 group
  • aralkenyl (or arylalkenyl) means an aryl-alkenyl- group (i.e., the bond to the parent moiety is through the alkenyl group) wherein the aryl group is unsubstituted or substituted as defined below, and the alkenyl group is unsubstituted or substituted as defined above; preferred aralkenyls contain a lower alkenyl group; non-limiting examples of suitable aralkenyl groups include 2-phenethenyl and 2-naphthylethenyl; "aralkyl” (or arylalkyl) means an aryl-alkyl- group (i.e., the bond to the parent moiety is through the alkyl group) wherein the aryl is unsubstituted or substituted as defined below and the alkyl is unsubstituted or substituted as defined above; preferred aralkyls comprise a lower alkyl group; non-limiting examples of suitable aral
  • aralkyloxy (or arylalkyloxy) means an aralkyl-O- group (i.e., the bond to the parent moiety is through the ether oxygen) wherein the aralkyl group is unsubstituted or substituted as previously described; non-limiting examples of suitable aralkyloxy groups include benzyloxy and 1- or 2-naphthalenemethoxy; "aralkoxycarbonyl” means an aralkyl-O-C(O)- group (i.e., the bond to the parent moiety is through the carbonyl) wherein the aralkyl group is unsubstituted or substituted as previously defined; a non-limiting example of a suitable aralkoxycarbonyl group is benzyloxycarbonyl;
  • aralkylthio means an aralkyl-S- group (i.e., the bond to the parent moiety is through the sulfur) wherein the aralkyl group is unsubstituted or substituted as previously described; a non-limiting example of a suitable aralkylthio group is benzylthio;
  • aroyl means an aryl-C(O)- group (i.e., the bond to the parent moiety is through the carbonyl) wherein the aryl group is unsubstituted or substituted as defined below; non-limiting examples of suitable groups include benzoyl and 1- and 2-naphthoyl;
  • aryl (sometimes abbreviated “ar”) means an aromatic monocyclic or multicyclic ring system comprising about 6 to about 14 carbon atoms, preferably about 6 to about 10 carbon atoms; the aryl group can be optionally substituted with one or more independently selected “ring system substituents” (defined below).
  • suitable aryl groups include phenyl and naphthyl;
  • arylalkynyl means an aryl-alkynyl- group (i.e., the bond to the parent moiety is through the alkynyl group) wherein the aryl group is unsubstituted or substituted as defined above, and the alkynyl group is unsubstituted or substitutedas defined above;
  • arylaminoheteroaryl means an aryl-amino-heteroaryl group (i.e., the bond to the parent moiety is through the heteroaryl group) wherein the aryl group is unsubstituted or substituted as defined above, the amino group is as defined above (i.e., a -NH- here), and the heteroaryl group is unsubstituted or substituted as defined below;
  • arylheteroaryl means an aryl-heteroarylgroup-(i.e., the bond to the parent moiety is through the heteroaryl group) wherein the aryl group is unsubstituted or substituted as defined above, and the heteroaryl group is unsubstituted or substituted as defined below;
  • aryloxy means an aryl-O- group (i.e., the bond to the parent moiety is through the ether oxygen) wherein the aryl group is unsubstituted or substituted as defined above; non-limiting examples of suitable aryloxy groups include phenoxy and naphthoxy;
  • aryloxycarbonyl means an aryl-O-C(O)- group (i.e., the bond to the parent moiety is through the carbonyl) wherein the aryl group is unsubstituted or substituted as previously defined; non-limiting examples of suitable aryloxycarbonyl groups include phenoxycarbonyl and naphthoxycarbonyl; "arylsulfinyl” means an aryl-S(O)- group (i.e., the bond to the parent moiety is through the sulfinyl) wherein aryl is unsubstituted or substituted as previously defined;
  • arylsulfonyl means an aryl-S(C> 2 )- group (i.e., the bond to the parent moiety is through the sulfonyl) wherein aryl is unsubstituted or substituted as previously defined;
  • arylthio means an aryl-S- group (i.e., the bond to the parent moiety is through the sulfur) wherein the aryl group is unsubstituted or substituted as previously described; non-limiting examples of suitable arylthio groups include phenylthio and naphthylthio; "cycloalkenyl” means a non-aromatic mono or multicyclic ring system comprising about 3 to about 10 carbon atoms, preferably about 5 to about 10 carbon atoms that contains at least one carbon-carbon double bond; preferred cycloalkenyl rings contain about 5 to about 7 ring atoms; the cycloalkenyl can be optionally substituted with one or more independently selected "ring system substituents" (defined below); Non-limiting examples of suitable monocyclic cycloalkenyls include cyclopentenyl, cyclohexenyl, cycloheptenyl, and the like; a non-limiting example of a suitable multicyclic cyclo
  • cycloalkyl means a non-aromatic mono- or multicyclic ring system comprising about 3 to about 7 carbon atoms, preferably about 3 to about 6 carbon atoms; the cycloalkyl can be optionally substituted with one or more independently selected "ring system substituents" (defined below); non-limiting examples of suitable monocyclic cycloalkyls include cyclopropyl, cyclopentyl, cyclohexyl, cycloheptyl and the like; non-limiting examples of suitable multicyclic cycloalkyls include 1 -decalin, norbornyl, adamantyl and the like;
  • cycloalkylalkyl means a cycloalkyl-alkyl-group (i.e., the bond to the parent moiety is through the alkyl group) wherein the cycloalkyl moiety is unsubstituted or substituted as defined above, and the alkyl moiety is unsubstituted or substituted as defined above;
  • halo means fluoro, chloro, bromo, or iodo groups; preferred halos are fluoro, chloro or bromo, and more preferred are fluoro and chloro;
  • halogen means fluorine, chlorine, bromine, or iodine; preferred halogens are fluorine, chlorine and bromine;
  • haloalkyl means an alkyl, as defined above, wherein one or more hydrogen atoms on the alkyl is replaced by a halo group, as defined above;
  • heteroarylkenyl means a heteroaryl-alkenyl- group (i.e., the bond to the parent moiety is through the alkenyl group) wherein the heteroaryl group is unsubstituted or substituted as defined below, and the alkenyl group is unsubstituted or substituted as defined above;
  • heteroarylkyl or heteroarylalkyl means a heteroaryl-alkyl- group (i.e., the bond to the parent moiety is through the alkyl group) in which the heteroaryl is unsubstituted or substituted as defined below, and the alkyl group is unsubstituted or substituted as defined above;
  • preferred heteroaralkyls comprise an alkyl group that is a lower alkyl group; non-limiting examples of suitable aralkyl groups include pyridylmethyl, 2-(furan-3-yl)ethyl and quinolin-3-ylmer.hyl;
  • heteroaralkylthio means a heteroaralkyl-S- group wherein the heteroaralkyl group is unsubstituted or substituted as defined above;
  • heteroaryl means an aromatic monocyclic or multicyclic ring system comprising about 5 to about 14 ring atoms, preferably about 5 to about 10 ring atoms, in which one or more of the ring atoms is an element other than carbon, for example nitrogen, oxygen or sulfur, alone or in combination; preferred heteroaryls comprise about 5 to about 6 ring atoms; the “heteroaryl” can be optionally substituted by one or more independently selected “ring system substituents” (defined below); the prefix aza, oxa or thia before the heteroaryl root name means that at least a nitrogen, oxygen or sulfur atom, respectively, is present as a ring atom; a nitrogen atom of a heteroaryl can be optionally oxidized to the corresponding N-oxide; non-limiting examples of suitable heteroaryls include pyridyl, pyrazinyl, furanyl, thienyl, pyrimidinyl, isoxazolyl, isothiazolyl
  • heteroarylalkynyl (or heteroaralkynyl) means a heteroaryl-alkynyl- group (i.e., the bond to the parent moiety is through the alkynyl group) wherein the heteroaryl group is unsubstituted or substituted as defined above, and the alkynyl group is unsubstituted or substituted as defined above;
  • heteroarylaryl (or heteroararyl) means a heteroaryl-aryl- group (i.e., the bond to the parent moiety is through the aryl group) wherein the heteroaryl group is unsubstituted or substituted as defined above, and the aryl group is unsubstituted or substituted as defined above;
  • heteroarylheteroarylaryl means a heteroaryl-heteroaryl- group (i.e., the bond to the parent moiety is through the last heteroaryl group) wherein each heteroaryl group is independently unsubstituted or substituted as defined above;
  • heteroarylsulfinyl means a heteroaryl-SO- group wherein the heteroaryl group is unsubstituted or substituted as defined above
  • heteroarylsulfonyl means a heteroaryl-SO 2 - group wherein the heteroaryl group is unsubstituted or substituted as defined above;
  • heteroarylthio means a heteroaryl-S- group wherein the heteroaryl group is unsubstituted or substituted as defined above;
  • heterocyclenyl (or heterocycloalkenyl) means a non-aromatic monocyclic or multicyclic ring system comprising about 3 to about 10 ring atoms, preferably about 5 to about 10 ring atoms, in which one or more of the atoms in the ring system is an element other than carbon (for example one or more heteroatoms independently selected from the group consisting of nitrogen, oxygen and sulfur atom), and which contains at least one carbon-carbon double bond or carbon-nitrogen double bond; there are no adjacent oxygen and/or sulfur atoms present in the ring system;
  • Preferred heterocyclenyl rings contain about 5 to about 6 ring atoms; the prefix aza, oxa or thia before the heterocyclenyl root name means that at least a nitrogen, oxygen or sulfur atom, respectively, is present as a ring atom; the heterocyclenyl can be optionally substituted by one or more independently selected "Ring system substituents" (defined below); the nitrogen or sulfur atom of the heterocyclenyl can be optionally oxidized to the corresponding N-oxide, S-oxide or S,S-dioxide; non-limiting examples of suitable monocyclic azaheterocyclenyl groups include 1 ,2,3,4- tetrahydropyridine, 1 ,2-dihydropyridyl, 1 ,4-dihydropyridyl, 1 ,2,3,6-tetrahydropyridine, 1 ,4,5,6-tetrahydropyrimidine, 2-pyrrolinyl, 3-pyrrolinyl, 2-imi
  • hydroxyalkyl means a HO-alkyl- group wherein the alkyl group is substituted or unsubstituted as defined above; preferred hydroxyalkyls comprise a lower alkyl; Non-limiting examples of suitable hydroxyalkyl groups include hydroxymethyl and 2-hydroxyethyl; and
  • ring system substituent means a substituent attached to an aromatic or non-aromatic ring system that, for example, replaces an available hydrogen on the ring system; ring system substituents are each independently selected from the group consisting of: alkyl, aryl, heteroaryl, aralkyl, alkylaryl, aralkenyl, heteroaralkyl, alkylheteroaryl, heteroaralkenyl, hydroxy, hydroxyalkyl, alkoxy, aryloxy, aralkoxy, acyl, aroyl, halo, nitro, cyano, carboxy, alkoxycarbonyl, aryloxycarbonyl, aralkoxycarbonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, alkylsulfinyl, arylsulfinyl, heteroarylsulfinyl, alkylthio, arylthio, heteroaryl
  • Lines drawn into a ring mean that the indicated bond may be attached to any of the substitutable ring carbon atoms.
  • Any carbon or heteroatom with unsatisfied valences in the text, schemes, examples, structural formulae, and any Tables herein is assumed to have the hydrogen atom or atoms to satisfy the valences. And any one or more of these hydrogen atoms can be deuterium.
  • One or more compounds of the invention may also exist as, or optionally converted to, a solvate. Preparation of solvates is generally known. Thus, for example, M. Caira et al, J. Pharmaceutical Sci., 93(3), 601-611 (2004) describe the preparation of the solvates of the antifungal fluconazole in ethyl acetate as well as from water. Similar preparations of solvates, hemisolvate, hydrates and the like are described by E.
  • a typical, non-limiting, process involves dissolving the inventive compound in desired amounts of the desired solvent (organic or water or mixtures thereof) at a higher than ambient temperature, and cooling the solution at a rate sufficient to form crystals which are then isolated by standard methods.
  • Analytical techniques such as, for example I. R. spectroscopy, show the presence of the solvent (or water) in the crystals as a solvate (or hydrate).
  • pharmaceutical composition is also intended to encompass both the bulk composition and individual dosage units comprised of more than one (e.g., two) pharmaceutically active agents such as, for example, a compound of the present invention and an additional agent selected from the lists of the additional agents described herein, along with any pharmaceutically inactive excipients.
  • the bulk composition and each individual dosage unit can contain fixed amounts of the aforesaid "more than one pharmaceutically active agents".
  • the bulk composition is material that has not yet been formed into individual dosage units.
  • An illustrative dosage unit is an oral dosage unit such as tablets, capsules, pills and the like.
  • the herein-described methods of treating a patient by administering a pharmaceutical composition of the present invention is also intended to encompass the administration of the afore-said bulk composition and individual dosage units.
  • Prodrugs of the compounds of the invention are also contemplated herein.
  • prodrug denotes a compound that is a drug precursor which, upon administration to a subject, undergoes chemical conversion by metabolic or chemical processes to yield a compound of formula 1.0 or a salt and/or solvate thereof.
  • a discussion of prodrugs is provided in T. Higuchi and V. Stella, Pro- drugs as Novel Delivery Systems (1987) 1_4 of the A.C.S. Symposium Series, and in Bioreversible Carriers in Drug Design, (1987) Edward B. Roche, ed., American Pharmaceutical Association and Pergamon Press, both of which are incorporated herein by reference thereto.
  • a prodrug can comprise an ester formed by the replacement of the hydrogen atom of the acid group with a group such as, for example, (Ci-C 8 )alkyl, (C 2 -Ci 2 )alkanoyloxy- methyl, 1 -(alkanoyloxy)ethyl having from 4 to 9 carbon atoms, 1 -methyl-1 -
  • alkanoyloxy)-ethyl having from 5 to 10 carbon atoms, alkoxycarbonyloxymethyl having from 3 to 6 carbon atoms, 1 -(alkoxycarbonyloxy)ethyl having from 4 to 7 carbon atoms, 1 -methyl-1 -(alkoxycarbonyloxy)ethyl having from 5 to 8 carbon atoms, N-(alkoxycarbonyl)aminomethyl having from 3 to 9 carbon atoms, 1-(N-(alkoxy- carbonyl)amino)ethyl having from 4 to 10 carbon atoms, 3-phthalidyl, 4- crotonolactonyl, gamma-butyrolacton-4-yl, di-N,N-(Ci-C 2 )alkylamino(C 2 -C 3 )alkyl (such as ⁇ -dimethylaminoethyl), carbamoyl-(Ci-C 2 )alkyl, N,N
  • a prodrug can be formed by the replacement of the hydrogen atom of the alcohol group with a group such as, for example, (Ci-C 6 )alkanoyloxymethyl, 1-((CrC 6 )alkanoyl- oxy)ethyl, 1 -methyl-1 -((Ci-C 6 )alkanoyloxy)ethyl, (Ci-C 6 )alkoxycarbonyloxymethyl, N- (Ci-C 6 )alkoxycarbonylaminomethyl, succinoyl, (CrC 6 )alkanoyl, ⁇ -amino(C r C 4 )alkanyl, arylacyl and ⁇ -aminoacyl, or ⁇ -aminoacyl- ⁇ -aminoacyl, where each ⁇ - aminoacyl group is independently selected from the naturally occurring L-amino acids, P(O)(OH) 2 ,
  • a prodrug can be formed by the replacement of a hydrogen atom in the amine group with a group such as, for example, R 70 -carbonyl, R 70 O-carbonyl, NR 70 R 75 -carbonyl where R 70 and R 75 are each independently (Ci-Ci O )alkyl, (C 3 -C 7 ) cycloalkyl, benzyl, or R 70 -carbonyl is a natural ⁇ -aminoacyl or natural ⁇ -aminoacyl, — C(OH)C(O)OY 80 wherein Y 80 is H, (CrC 6 )alkyl or benzyl, — C(OY 82 )Y 84 wherein Y 82 is (C r C 4 ) alkyl and Y 84 is (CrC 6 )alkyl, carboxy (d-C 6 )alkyl, amino(d-C 4 )al
  • This invention also includes the compounds of this invention in isolated and purified form.
  • Polymorphic forms of the compounds of formula 1.0, and of the salts, solvates and prodrugs of the compounds of formula 1.0, are intended to be included in the present invention.
  • Certain compounds of the invention may exist in different isomeric (e.g., enantiomers, diastereoisomers, atropisomers) forms.
  • the invention contemplates all such isomers both in pure form and in admixture, including racemic mixtures. Enol forms are also included.
  • All stereoisomers (for example, geometric isomers, optical isomers and the like) of the present compounds including those of the salts, solvates and prodrugs of the compounds as well as the salts and solvates of the prodrugs), such as those which may exist due to asymmetric carbons on various substituents, including enantiomeric forms (which may exist even in the absence of asymmetric carbons), rotameric forms, atropisomers, and diastereomeric forms, are contemplated within the scope of this invention.
  • Individual stereoisomers of the compounds of the invention may, for example, be substantially free of other isomers, or may be admixed, for example, as racemates or with all other, or other selected, stereoisomers.
  • the chiral centers of the present invention can have the S or R configuration as defined by the IUPAC 1974 Recommendations.
  • the use of the terms “salt”, “solvate” “prodrug” and the like, is intended to equally apply to the salt, solvate and prodrug of enantiomers, stereoisomers, rotamers, tautomers, racemates or prodrugs of the inventive compounds.
  • Diasteromeric mixtures can be separated into their individual diastereomers on the basis of their physical chemical differences by methods well known to those skilled in the art, such as, for example, by chromatography and/or fractional crystallization.
  • Enantiomers can be separated by converting the enantiomeric mixture into a diasteromeric mixture by reaction with an appropriate optically active compound (e.g., chiral auxiliary such as a chiral alcohol or Mosher's acid chloride), separating the diastereomers and converting (e.g., hydrolyzing) the individual diastereomers to the corresponding pure enantiomers.
  • an appropriate optically active compound e.g., chiral auxiliary such as a chiral alcohol or Mosher's acid chloride
  • some of the compounds of Formula (I) may be atropisomers (e.g., substituted biaryls) and are considered as part of this invention.
  • Enantiomers can also be separated by use of chiral HPLC column.
  • the compounds of formula 1.0 form salts that are also within the scope of this invention.
  • Reference to a compound of formula 1.0 herein is understood to include reference to salts thereof, unless otherwise indicated.
  • zwitterions inner salts may be formed and are included within the term
  • Salt(s) as used herein.
  • Pharmaceutically acceptable i.e., non-toxic, physiologically acceptable salts
  • Salts of the compounds of the formula 1.0 may be formed, for example, by reacting a compound of formula 1.0 with an amount of acid or base, such as an equivalent amount, in a medium such as one in which the salt precipitates or in an aqueous medium followed by lyophilization.
  • Acids (and bases) which are generally considered suitable for the formation of pharmaceutically useful salts from basic (or acidic) pharmaceutical compounds are discussed, for example, by S. Berge et al, Journal of Pharmaceutical Sciences (1977) 66(1 ) 1 -19; P. Gould, International J.
  • Exemplary acid addition salts include acetates, adipates, alginates, ascorbates, aspartates, benzoates, benzenesulfonates, bisulfates, borates, butyrates, citrates, camphorates, camphorsulfonates, cyclopentanepropionates, digluconates, dodecylsulfates, ethanesulfonates, fumarates, glucoheptanoates, glycerophosphates, hemisulfates, heptanoates, hexanoates, hydrochlorides, hydrobromides, hydroiodides, 2-hydroxyethanesulfonates, lactates, maleates, methanesulfonates, methyl sulfates, 2-naphthalenesulfonates, nicotinates, nitrates, oxalates, pamoates, pectinates, persulfates, 3-
  • Exemplary basic salts include ammonium salts, alkali metal salts such as sodium, lithium, and potassium salts, alkaline earth metal salts such as calcium and magnesium salts, aluminum salts, zinc salts, salts with organic bases (for example, organic amines) such as benzathines, diethylamine, dicyclohexylamines, hydrabamines (formed with N,N-bis(dehydroabietyl)ethylenediamine), N-methyl-D- glucamines, N-methyl-D-glucamides, t-butyl amines, piperazine, phenylcyclohexyl- amine, choline, tromethamine, and salts with amino acids such as arginine, lysine and the like.
  • organic bases for example, organic amines
  • organic bases for example, organic amines
  • benzathines diethylamine, dicyclohexylamines, hydrabamines (formed with N,
  • Basic nitrogen-containing groups may be quarternized with agents such as lower alkyl halides (e.g. methyl, ethyl, propyl, and butyl chlorides, bromides and iodides), dialkyl sulfates (e.g. dimethyl, diethyl, dibutyl, and diamyl sulfates), long chain halides (e.g. decyl, lauryl, myristyl and stearyl chlorides, bromides and iodides), aralkyl halides (e.g. benzyl and phenethyl bromides), and others.
  • lower alkyl halides e.g. methyl, ethyl, propyl, and butyl chlorides, bromides and iodides
  • dialkyl sulfates e.g. dimethyl, diethyl, dibutyl, and diamyl sulfates
  • long chain halides
  • hetero-atom containing ring systems of this invention there are no hydroxyl groups on carbon atoms adjacent to a N, O or S, and there are no N or S groups on carbon adjacent to another heteroatom.
  • the compounds of formula 1.0 may exist in different tautomeric forms, and all such forms are embraced within the scope of the invention. Also, for example, all keto-enol and imine-enamine forms of the compounds are included in the invention.
  • substituted means that one or more hydrogens on the designated atom is replaced with a selection from the indicated group, provided that the designated atom's normal valency under the existing circumstances is not exceeded, and that the substitution results in a stable compound. Combinations of substituents and/or variables are permissible only if such combinations result in stable compounds.
  • stable compound or “stable structure” is meant a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent.
  • purified refers to the physical state of said compound after being isolated from a synthetic process or natural source or combination thereof.
  • purified refers to the physical state of said compound after being obtained from a purification process or processes described herein or well known to the skilled artisan, in sufficient purity to be characterizable by standard analytical techniques described herein or well known to the skilled artisan.
  • protecting groups When a functional group in a compound is termed "protected", this means that the group is in modified form to preclude undesired side reactions at the protected site when the compound is subjected to a reaction. Suitable protecting groups will be recognized by those with ordinary skill in the art as well as by reference to standard textbooks such as, for example, T. W. Greene etal, Protective Groups in organic Synthesis (1991), Wiley, New York.
  • composition is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts.
  • the present invention also embraces isotopically-labelled compounds of the present invention which are identical to those recited herein, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature.
  • isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, fluorine, chlorine, and iodine, such as 2 H, 3 H, 11 C, 13 C, 14 C, 15 N, 18 0, 17 0, 31 P, 32 P, 35 S, 18 F, 36 CI, and 123 I, respectively.
  • Certain isotopically-labelled compounds of formula 1.0 are useful in compound and/or substrate tissue distribution assays. Tritiated (i.e., 3 H) and carbon-14 (i.e., 14 C) isotopes are particularly preferred for their ease of preparation and detectability.
  • Certain isotopically-labelled compounds of Formula (I) can be useful for medical imaging purposes. E.g., those labeled with positron-emitting isotopes like 11 C or 18 F can be useful for application in Positron Emission Tomography (PET) and those labeled with gamma ray emitting isotopes like 123 I can be useful for application in Single photon emission computed tomography
  • lsotopically labeled compounds of Formula (I) in particular those containing isotopes with longer half lives (T1/2 >1 day), can generally be prepared by following procedures analogous to those disclosed in the Schemes and/or in the Examples herein below, by substituting an appropriate isotopically labeled reagent for a non-isotopically labeled reagent.
  • This invention provides compounds of formula 1.0:
  • R 1 , R 2 and Q are independently selected, and wherein:
  • R 1 is selected from the group consisting of: heteroaryl and substituted heteroaryl, wherein said substituted heteroaryl is substituted with 1 to 3 (preferably 1 ) substituents independently selected from the group consisting of: -OH, alkoxy, and -O-alkylene-O-alkyl;
  • R 2 is selected from the group consisting of: -O-alkyl and -S-alkyl; and
  • R 5 is selected from the group consisting of:
  • substituted triazolyl-pyridyl- wherein: (1) said pyridyl is optionally substituted with 1 to 3 substituents independently selected from the group consisting of: halo (e.g., Br, Cl, F, and in one example F), alkyl, and alkoxy (e.g., CrC ⁇ alkoxy, and in one example, CrC 2 alkoxy, and in another example -OCH 3 ), provided that the carbon atoms adjacent to the nitrogen atom in said pyridyl are not substituted with halo, and (2) said triazolyl group is substituted with one or two substitutents independently selected from the group consisting of: alkyl, hydroxy substituted alkyl, -alkylene-O-alkyl, and amino (i.e., -NH 2 ), (j) triazolyl-thiazolyl-,
  • substituents independently selected from the group consisting of: halo (e.g., Br, Cl, F, and in one example F) and alkoxy (e.g., d-C ⁇ alkoxy, and in one example, Cr C 2 alkoxy, and in another example -OCH 3 ), and
  • R 1 is selected from the group consisting of: heteroaryl and substituted heteroaryl, wherein said substituted heteroaryl is substituted with 1 to 3 (preferably 1) substituents independently selected from the group consisting of: -OH, alkoxy, and -O-alkylene-O-alkyl;
  • R 2 is selected from the group consisting of: -O-alkyl and -S-alkyl; and R 5 is selected from the group consisting of: (a) triazolyl-phenyl-,
  • alkylene as used in the substituents -O-alkylene-O-alkyl and -alkylene-O-alkyl, means a divalent saturated hydrocarbon group.
  • an example of an alkylene moiety is -CH 2 -CH 2 -
  • an example of an -O-alkylene-O-alkyl moiety is -O-(CH 2 ) 2 -O-CH 3
  • an example of an -alkylene-O-alkyl moiety is -(CH 2 ) 2 -O-CH 3 .
  • alkylene also includes the moiety -CH 2 -.
  • R 1 heteroaryl group examples include, but are not limited to, pyridyl, pyrrolyl, pyrazolyl, imidazolyl, furanyl, thienyl, thiazolyl, pyridyl N-O, and pyrimidinyl.
  • R 1 substitued heteroaryl group examples include, but are not limited to, substituted pyridyl, substituted pyrrolyl, substituted pyrazolyl, substituted imidazolyl, substituted furanyl, substituted thienyl, substituted thiazolyl, substituted pyridyl N-O, and substituted pyrimidinyl.
  • R 1 is pyridyl. In another embodiment of this invention R 1 is substituted pyridyl. In another embodiment of this invention R 1 is pyridyl substituted with one substitutent.
  • the substitutents on the substituted R 1 groups are independently selected from the group consisting of: -OH, alkoxy, and -O-alkylene-O-alkyl.
  • alkoxy group include, for example, Ci to C 6 alkoxy (such as, for example, -0-CH 3 , -0-C 2 H 5 , and -O-CH(CH 3 ) 2 ).
  • Examples of the -O-alkylene-O-alkyl group include, for example, -O-(Ci-C 4 )alkylene-O-(C r C 6 )alkyl, -O-(CrC2)alkylene-O-(CrC 3 alkyl), and -0-(CH 2 )Z-O-CH 3 ).
  • R 1 examples include, for example,
  • R 1 is pyridyl substituted with alkoxy. In another embodiement of this invention R 1 is substituted with -OCH(CH 3 ) 2 . In another embodiement of this invention R 1 is substituted with -OC 2 H 5 . In another embodiment of this invention R 1 is:
  • R 1 is:
  • R 1 is substituted with -O-alkylene-O-alkyl.
  • R 1 is substituted with -OCH 2 CH 2 OCH 3 .
  • R 1 is:
  • R 2 -O-alkyl group examples include, for example, -O-(CrC 6 )alkyl,
  • R 2 -S-alkyl group examples include, for example, -S-(CrC 6 )alkyl, -S-(C 1 -C 2 )alkyl. and -SCH 3 .
  • R 2 is a -O-(C-i-C 2 )alkyl group. In another embodiment of this invention R 2 is -OCH 3 .
  • R 2 is a -S-(Ci -C 2 )alkyl group.
  • R 2 is -SCH 3 .
  • R 5 is a triazolyl-phenyl moiety wherein the triazolyl moiety is bonded to the phenyl moiety by a ring carbon of the triazolyl moiety
  • R 5 is a triazolyl-phenyl- moiety, such as, for example,
  • R 5 is a triazolyl-thienyl- moiety, such as, for example,
  • R 5 is a triazolyl-thienyl- moiety, such as, for example,
  • substituted triazolyl moiety of said R 5 group is substituted on the ring nitrogen.
  • examples of the alkyl groups include, for example, -CrC 6 alkyl, -CrC 4 alkyl, -CrC 2 alkyl, and -CH 3 . And in one embodiment, there is alkyl substitution on the triazolyl moiety of R 5 and said alkyl is -CH 3 .
  • examples of the -alkylene-O-alkyl groups include, for example, -Ci-C 4 alkylene-O-Cr C 6 alkyl, -Ci-C 2 alkylene-O-Ci-C 2 alkyl, -d-C-jalkylene-O-CHs, and -CH 2 CH 2 OCH 3 .
  • examples of the -alkylene-O- alkyl group includes, for example, -C 2 -C 4 alkylene-O-C r C 6 alkyl, -C 2 alkylene-O-C r C 2 alkyl, -C 2 -C4alkylene-O-CH 3 , and -CH 2 CH 2 OCH 3 .
  • hydroxy substituted alkyl groups include, for example, hydroxy substituted -CrC 4 alkyl, hydroxy substituted -Ci-C 2 alkyl, and hydroxy substituted -CH 3 .
  • examples also include, for example, -CH 2 COH(CH 3 ) 2 , and -CH 2 CH 2 OH.
  • halo atoms include, for example, Cl, F and Br.
  • the halo on the phenyl is F.
  • the phenyl is substituted with one F.
  • R 5 is a substituted triazolyl-phenyl- wherein said triazolyl is substituted and said phenyl is unsubstituted.
  • R 5 is a substituted triazolyl-phenyl- wherein said triazolyl is substituted on the nitrogen and said phenyl is unsubstituted.
  • R 5 is a substituted triazolyl-phenyl- wherein said triazolyl is substituted on the carbon and said phenyl is unsubstituted.
  • R 5 is a substituted triazolyl-phenyl- wherein said triazolyl is substituted on the nitrogen and on the carbon, and said phenyl is unsubstituted.
  • R 5 is a substituted triazolyl-phenyl- wherein said triazolyl is substituted and said phenyl is substituted.
  • R 5 is a substituted triazolyl-phenyl- wherein said triazolyl is substituted on the nitrogen and said phenyl is substituted. In another embodiment of this invention R 5 is a substituted triazolyl-phenyl- wherein said triazolyl is substituted on the carbon and said phenyl is substituted. In another embodiment of this invention R 5 is a substituted triazolyl-phenyl- wherein said triazolyl is substituted on the nitrogen and on the carbon, and said phenyl is substituted.
  • R 5 is a substituted triazolyl-phenyl- wherein said triazolyl is unsubstituted and said phenyl is substituted.
  • R 5 is an unsubstituted triazolyl-phenyk
  • R 5 is a substituted triazolyl-phenyl- wherein the triazolyl is substituted on the nitrogen with -CH 2 COH(CH 3 ) 2 .
  • R 5 is a substituted triazolyl-phenyl- wherein the triazolyl is substituted on the nitrogen with -CH 2 CH 2 OH.
  • R 5 is a substituted triazolyl-phenyl- wherein the triazolyl is substituted on the nitrogen with an alkyl group.
  • R 5 is a substituted triazolyl-phenyl- wherein the triazolyl is substituted on the nitrogen with an alkyl group and substituted on the carbon with an alkyl group, wherein each alkyl group is independently selected.
  • R 5 is a substituted triazolyl-phenyl- wherein the triazolyl is substituted on the nitrogen with a -CH 3 group.
  • R 5 is a substituted triazolyl-phenyl- wherein the triazolyl is substituted on the nitrogen with a -CH 3 group and on the carbon with a -CH 3 group.
  • R 5 moiety is a substituted triazolyl- phenyl- wherein the triazolyl is substituted on the carbon with a -NH 2 group.
  • R 5 is a substituted triazolyl-phenyl- wherein the triazolyl is substituted on the nitrogen with an -alkylene-O-alkyl group.
  • R 5 is a substituted triazolyl-phenyl- wherein the triazolyl is substituted on the nitrogen with a -CH 2 CH 2 OCH 3 group.
  • R 5 moiety is a substituted triazolyl- phenyl- wherein said phenyl moiety is substituted with halo, and said triazolyl moiety is substituted as described in any of the above embodiments.
  • R 5 is a substituted triazolyl-phenyl- wherein said phenyl moiety is substituted with one halo, and said triazolyl moiety is substituted as described in any one of the above embodiments.
  • R 5 is a substituted triazolyl-phenyl- wherein said phenyl moiety is substituted with F, and said triazolyl moiety is substituted as described in any one of the above embodiments.
  • R 5 is a substituted triazolyl-phenyl- wherein said phenyl moiety is substituted with one F, and said triazolyl moiety is substituted as described in any one of the above embodiments.
  • R 5 is a substituted triazolyl-phenyl- group wherein said phenyl moiety is substituted with one F, and said triazolyl moiety is substituted on the nitrogen with a hydroxyl substituted alkyl group.
  • R 5 is a substituted triazolyl-phenyl- wherein said phenyl moiety is substituted with one F, and said triazolyl moiety is substituted on the nitrogen with a -CH 2 CH 2 OH group.
  • R 5 is a substituted triazolyl-phenyl- wherein said phenyl moiety is substituted with one F, and said triazolyl moiety is substituted on the nitrogen with a -alkylene-O-alkyl group.
  • R 5 is a substituted triazolyl-phenyl- wherein said phenyl moiety is substituted with one F, and said triazolyl moiety is substituted on the nitrogen with a -CH 2 CH 2 OCH 3 group.
  • R 5 is a substituted triazolyl-phenyl- wherein said phenyl moiety is substituted with halo, and said triazolyl moiety is unsubstituted.
  • R 5 is a substituted triazolyl-phenyl- wherein said phenyl moiety is substituted with one halo, and said triazolyl moiety is unsubstituted.
  • R 5 is a substituted triazolyl-phenyl- wherein said phenyl moiety is substituted with F, and said triazolyl moiety is unsubstituted.
  • R 5 is a substituted triazolyl-phenyl- wherein said phenyl moiety is substituted with one F, and said triazolyl moiety is unsubstituted.
  • R 5 moiety is a substituted triazolyl- phenyl- wherein said phenyl moiety is substituted with alkoxy, and said triazolyl moiety is substituted as described in any of the above embodiments.
  • R 5 is a substituted triazolyl-phenyl- wherein said phenyl moiety is substituted with one alkoxy, and said triazolyl moiety is substituted as described in any one of the above embodiments.
  • R 5 is a substituted triazolyl-phenyl- wherein said phenyl moiety is substituted with -OCH 3 , and said triazolyl moiety is substituted as described in any one of the above embodiments.
  • R 5 is a substituted triazolyl-phenyl- wherein said phenyl moiety is substituted with one -OCH 3 , and said triazolyl moiety is substituted as described in any one of the above embodiments.
  • R 5 is a substituted triazolyl-phenyl- group wherein said phenyl moiety is substituted with one -OCH 3 , and said triazolyl moiety is substituted on the nitrogen with a hydroxyl substituted alkyl group.
  • R 5 is a substituted triazolyl-phenyl- wherein said phenyl moiety is substituted with one -OCH 3 , and said triazolyl moiety is substituted on the nitrogen with a -CH 2 CH 2 OH group.
  • R 5 is a substituted triazolyl-phenyl- wherein said phenyl moiety is substituted with one -OCH 3 , and said triazolyl moiety is substituted on the nitrogen with a -alkylene-O-alkyl group.
  • R 5 is a substituted triazolyl-phenyl- wherein said phenyl moiety is substituted with one -OCH 3 , and said triazolyl moiety is substituted on the nitrogen with a -CH 2 CH 2 OCH 3 group.
  • R 5 is a substituted triazolyl-phenyl- wherein said phenyl moiety is substituted with alkoxy, and said triazolyl moiety is unsubstituted.
  • R 5 is a substituted triazolyl-phenyl- wherein said phenyl moiety is substituted with one alkoxy, and said triazolyl moiety is unsubstituted.
  • R 5 is a substituted triazolyl-phenyl- wherein said phenyl moiety is substituted with with -OCH 3 , and said triazolyl moiety is unsubstituted.
  • R 5 is a substituted triazolyl-phenyl- wherein said phenyl moiety is substituted with one -OCH 3 , and said triazolyl moiety is unsubstituted.
  • R 5 is:
  • R ,5 is, In another embodiment of this invention R 5 is:
  • R 5 is,
  • R 5 is
  • R .5 is,
  • R 5 is,
  • R 5 is,
  • R 5 is:
  • R 5 is a substituted triazolyl-thienyl- wherein said triazolyl is substituted and said thienyl is unsubstituted.
  • R 5 is a substituted triazolyl-thienyl- wherein said triazolyl is substituted on the nitrogen and said thienyl is unsubstituted.
  • R 5 is a substituted triazolyl-thienyl- wherein said triazolyl is substituted on the carbon and said thienyl is unsubstituted.
  • R 5 is a substituted triazolyl-thienyl- wherein said triazolyl is substituted on the nitrogen and on the carbon, and said thienyl is unsubstituted.
  • R 5 is a substituted triazolyl-thienyl- wherein said triazolyl is substituted and said thienyl is substituted.
  • R 5 is a substituted triazolyl-thienyl- wherein said triazolyl is substituted on the nitrogen and said thienyl is substituted.
  • R 5 is a substituted triazolyl-thienyl- wherein said triazolyl is substituted on the carbon and said thienyl is substituted.
  • R 5 is a substituted triazolyl-thienyl- wherein said triazolyl is substituted on the nitrogen and on the carbon, and said thienyl is substituted.
  • R 5 is a substituted triazolyl-thienyl- wherein said triazolyl is unsubstituted and said thienyl is substituted.
  • R 5 is an unsubstituted ti ⁇ azolyl-thienyk
  • R 5 is a substituted triazolyl-thienyl- wherein the triazolyl is substituted on the nitrogen with -CH 2 COH(CH 3 )2.
  • R 5 is a substituted triazolyl-thienyl- wherein the triazolyl is substituted on the nitrogen with -CH 2 CH 2 OH.
  • R 5 is a substituted triazolyl-thienyl- wherein the triazolyl is substituted on the nitrogen with an alkyl group.
  • R 5 is a substituted triazolyl-thienyl- wherein the triazolyl is substituted on the nitrogen with an alkyl group and substituted on the carbon with an alkyl group, wherein each alkyl group is independently selected.
  • R 5 is a substituted triazolyl-thienyl- wherein the triazolyl is substituted on the nitrogen with a -CH 3 group.
  • R 5 is a substituted triazolyl-thienyl- wherein the triazolyl is substituted on the nitrogen with a -CH 3 group and on the carbon with a -CH 3 group.
  • R 5 moiety is a substituted triazolyl- thienyl- wherein the triazolyl is substituted on the carbon with a -NH 2 group.
  • R 5 is a substituted triazolyl-thienyl- wherein the triazolyl is substituted on the nitrogen with an -alkylene-O-alkyl group.
  • R 5 is a substituted triazolyl-thienyl- wherein the triazolyl is substituted on the nitrogen with a -CH 2 CH 2 OCH 3 group.
  • R 5 moiety is a substituted triazolyl- thienyl- wherein said thienyl moiety is substituted with halo, and said triazolyl moiety is substituted as described in any of the above embodiments.
  • R 5 is a substituted triazolyl-thienyl- wherein said thienyl moiety is substituted with one halo, and said triazolyl moiety is substituted as described in any one of the above embodiments.
  • R 5 is a substituted triazolyl-thienyl- wherein said thienyl moiety is substituted with F, and said triazolyl moiety is substituted as described in any one of the above embodiments.
  • R 5 is a substituted triazolyl-thienyl- wherein said thienyl moiety is substituted with one F, and said triazolyl moiety is substituted as described in any one of the above embodiments.
  • R 5 is a substituted triazolyl-thienyl- group wherein said thienyl moiety is substituted with one F, and said triazolyl moiety is substituted on the nitrogen with a hydroxyl substituted alkyl group.
  • R 5 is a substituted triazolyl-thienyl- wherein said thienyl moiety is substituted with one F, and said triazolyl moiety is substituted on the nitrogen with a -CH 2 CH 2 OH group.
  • R 5 is a substituted triazolyl-thienyl- wherein said thienyl moiety is substituted with one F, and said triazolyl moiety is substituted on the nitrogen with a -alkylene-O-alkyl group.
  • R 5 is a substituted triazolyl-thienyl- wherein said thienyl moiety is substituted with one F, and said triazolyl moiety is substituted on the nitrogen with a -CH 2 CH 2 OCH 3 group.
  • R 5 is a substituted triazolyl-thienyl- wherein said thienyl moiety is substituted with halo, and said triazolyl moiety is unsubstituted.
  • R 5 is a substituted triazolyl-thienyl- wherein said thienyl moiety is substituted with one halo, and said triazolyl moiety is unsubstituted.
  • R 5 is a substituted triazolyl-thienyl- wherein said thienyl moiety is substituted with F, and said triazolyl moiety is unsubstituted.
  • R 5 is a substituted triazolyl-thienyl- wherein said thienyl moiety is substituted with one F, and said triazolyl moiety is unsubstituted.
  • R 5 moiety is a substituted triazolyl- thienyl- wherein said thienyl moiety is substituted with alkoxy, and said triazolyl moiety is substituted as described in any of the above embodiments.
  • R 5 is a substituted triazolyl-thienyl- wherein said thienyl moiety is substituted with one alkoxy, and said triazolyl moiety is substituted as described in any one of the above embodiments.
  • R 5 is a substituted triazolyl-thienyl- wherein said thienyl moiety is substituted with -OCH 3 , and said triazolyl moiety is substituted as described in any one of the above embodiments.
  • R 5 is a substituted triazolyl-thienyl- wherein said thienyl moiety is substituted with one -OCH 3 , and said triazolyl moiety is substituted as described in any one of the above embodiments.
  • R 5 is a substituted triazolyl-thienyl- group wherein said thienyl moiety is substituted with one -OCH 3 , and said triazolyl moiety is substituted on the nitrogen with a hydroxyl substituted alkyl group.
  • R 5 is a substituted triazolyl-thienyl- wherein said thienyl moiety is substituted with one -OCH 3 , and said triazolyl moiety is substituted on the nitrogen with a -CH 2 CH 2 OH group.
  • R 5 is a substituted triazolyl-thienyl- wherein said thienyl moiety is substituted with one -OCH 3 , and said triazolyl moiety is substituted on the nitrogen with a -alkylene-O-alkyl group.
  • R 5 is a substituted triazolyl-thienyl- wherein said thienyl moiety is substituted with one -OCH 3 , and said triazolyl moiety is substituted on the nitrogen with a -CH 2 CH 2 OCH 3 group.
  • R 5 is a substituted triazolyl-thienyl- wherein said thienyl moiety is substituted with alkoxy, and said triazolyl moiety is unsubstituted.
  • R 5 is a substituted triazolyl-thienyl- wherein said thienyl moiety is substituted with one alkoxy, and said triazolyl moiety is unsubstituted.
  • R 5 is a substituted triazolyl-thienyl- wherein said thienyl moiety is substituted with -OCH 3 , and said triazolyl moiety is unsubstituted.
  • R 5 is a substituted triazolyl-thienyl- wherein said thienyl moiety is substituted with one -OCH 3 , and said triazolyl moiety is unsubstituted.
  • R 5 is triazolyl-pyridyl-.
  • R 5 is
  • R is a substituted triazolyl-pyridyl- wherein said triazolyl is substituted and said pyridyl is unsubstituted.
  • R 5 is a substituted triazolyl-pyridyl- wherein said triazolyl is substituted on the nitrogen and said pyridyl is unsubstituted.
  • R 5 is a substituted triazolyl-pyridyl- wherein said triazolyl is substituted on the carbon and said pyridyl is unsubstituted.
  • R 5 is a substituted triazolyl-pyridyl- wherein said triazolyl is substituted on the nitrogen and on the carbon, and said pyridyl is unsubstituted.
  • R 5 is a substituted triazolyl-pyridyl- wherein said triazolyl is substituted and said pyridyl is substituted.
  • R 5 is a substituted triazolyl-pyridyl- wherein said triazolyl is substituted on the nitrogen and said pyridyl is substituted.
  • R 5 is a substituted triazolyl-pyridyl- wherein said triazolyl is substituted on the carbon and said pyridyl is substituted.
  • R 5 is a substituted triazolyl-pyridyl- wherein said triazolyl is substituted on the nitrogen and on the carbon, and said pyridyl is substituted.
  • R 5 is a substituted triazolyl-pyridyl- wherein said triazolyl is unsubstituted and said pyridyl is substituted.
  • R 5 is a substituted triazolyl-pyridyl- wherein the triazolyl is substituted on the nitrogen with alkyl.
  • alkyl groups include, for example, -CrC 6 alkyl, -CrC 4 alkyl, -CrC 2 alkyl, and -CH 3 .
  • halo atoms include, for example, Cl, F and Br, provided that the carbon atoms adjacent to the nitrogen atom in said pyridyl are not substituted with halo.
  • the halo on the pyridyl is F.
  • the pyridyl is substituted with one F.
  • R 5 is a substituted triazolyl-pyridyl- wherein the triazolyl is substituted on the nitrogen with -CH 2 COH (CH 3 ) 2 , and the pyridyl is unsubstituted.
  • R 5 is a substituted triazolyl-pyridyl- wherein the triazolyl is substituted on the nitrogen with -CH 2 CH 2 OH, and the pyridyl is unsubstituted.
  • R 5 is a substituted triazolyl-pyridyl- wherein the triazolyl is substituted on the nitrogen with an alkyl group, and the pyridyl is unsubstituted.
  • R 5 is a substituted triazolyl-pyridyl- wherein the triazolyl is substituted on the nitrogen with an alkyl group and substituted on the carbon with an alkyl group, wherein each alkyl group is independently selected, and the pyridyl is unsubstituted.
  • R 5 is a substituted triazolyl-pyridyl- wherein the triazolyl is substituted on the nitrogen with a -CH 3 group, and the pyridyl is unsubstituted.
  • R 5 is a substituted triazolyl-pyridyl- wherein the triazolyl is substituted on the nitrogen with a -CH 3 group and on the carbon with a -CH 3 group, and the pyridyl is unsubstituted.
  • R 5 moiety is a substituted triazolyl- pyridyl- wherein the triazolyl is substituted on the carbon with a -NH 2 group, and the pyridyl is unsubstituted.
  • R 5 is a substituted triazolyl-pyridyl- wherein the triazolyl is substituted on the nitrogen with an -alkylene-O-alkyl group, and the pyridyl is unsubstituted, and provided that the alkylene moiety of said -alkylene-O-alkyl group is not -CH 2 - (i.e., the alkylene moiety is 2 or more carbons in length).
  • R 5 is a substituted triazolyl-pyridyl- wherein the triazolyl is substituted on the nitrogen with a -CH 2 CH 2 OCH 3 group, and the pyridyl is unsubstituted.
  • R 5 moiety is a substituted triazolyl- pyridyl- wherein said pyridyl moiety is substituted with halo, provided that the carbon atoms adjacent to the nitrogen atom in said pyridyl are not substituted with halo, and said triazolyl moiety is substituted as described in any of the above embodiments describing substituted triazolyl groups.
  • R 5 is a substituted triazolyl-pyridyl- wherein said pyridyl moiety is substituted with one halo, provided that a carbon atom adjacent to the nitrogen atom in said pyridyl is not substituted with said halo, and said triazolyl moiety is substituted as described in any one of the above embodiments, describing triazolyl groups.
  • R 5 is a substituted triazolyl-pyridyl- wherein said pyridyl moiety is substituted with F, provided that a carbon atom adjacent to the nitrogen atom in said pyridyl is not substituted with said F, and said triazolyl moiety is substituted as described in any one of the above embodiments, describing triazolyl groups.
  • R 5 is a substituted triazolyl-pyridyl- wherein said pyridyl moiety is substituted with one F, provided that a carbon atom adjacent to the nitrogen atom in said pyridyl is not substituted with said F, and said triazolyl moiety is substituted as described in any one of the above embodiments describing triazolyl groups.
  • R 5 is a substituted triazolyl-pyridyl- group wherein said pyridyl moiety is substituted with one F, provided that a carbon atom adjacent to the nitrogen atom in said pyridyl is not substituted with said F, and said triazolyl moiety is substituted on the nitrogen with a hydroxyl substituted alkyl group.
  • R 5 is a substituted triazolyl-pyridyl- wherein said pyridyl moiety is substituted with one F, provided that a carbon atom adjacent to the nitrogen atom in said pyridyl is not substituted with said F, and said triazolyl moiety is substituted on the nitrogen with a -CH 2 CH 2 OH group.
  • R 5 is a substituted triazolyl-pyridyl- wherein said pyridyl moiety is substituted with one F, provided that a carbon atom adjacent to the nitrogen atom in said pyridyl is not substituted with said F, and said triazolyl moiety is substituted on the nitrogen with an -alkylene-O-alkyl group, and the alkylene moiety of said -alkylene-O-alkyl group is not -CH 2 - (i.e., the alkylene moiety is 2 or more carbons in length).
  • R 5 is a substituted triazolyl-pyridyl- wherein said pyridyl moiety is substituted with one F, provided that a carbon atom adjacent to the nitrogen atom in said pyridyl is not substituted with said F, and said triazolyl moiety is substituted on the nitrogen with a -CH 2 CH 2 OCH 3 group.
  • R 5 is a substituted triazolyl-pyridyl- wherein said pyridyl moiety is substituted with halo, provided that the carbon atoms adjacent to the nitrogen atom in said pyridyl are not substituted with halo, and said triazolyl moiety is unsubstituted.
  • R 5 is a substituted triazolyl-pyridyl- wherein said pyridyl moiety is substituted with one halo, provided that a carbon atom adjacent to the nitrogen atom in said pyridyl is not substituted with said halo, and said triazolyl moiety is unsubstituted.
  • R 5 is a substituted triazolyl-pyridyl- wherein said pyridyl moiety is substituted with F, provided that a carbon atom adjacent to the nitrogen atom in said pyridyl is not substituted with said F, and said triazolyl moiety is unsubstituted.
  • R 5 is a substituted triazolyl-pyridyl- wherein said pyridyl moiety is substituted with one F, provided that a carbon atom adjacent to the nitrogen atom in said pyridyl is not substituted with said F, and said triazolyl moiety is unsubstituted.
  • R 5 moiety is a substituted triazolyl- pyridyl- wherein said pyridyl moiety is substituted with alkoxy, and said triazolyl moiety is substituted as described in any of the above embodiments describing substituted triazolyl groups.
  • R 5 is a substituted triazolyl-pyridyl- wherein said pyridyl moiety is substituted with one alkoxy, and said triazolyl moiety is substituted as described in any one of the above embodiments describing substituted triazolyl groups.
  • R 5 is a substituted triazolyl-pyridyl- wherein said pyridyl moiety is substituted with -OCH 3 , and said triazolyl moiety is substituted as described in any one of the above embodiments describing substituted triazolyl groups.
  • R 5 is a substituted triazolyl-pyridyl- wherein said pyridyl moiety is substituted with one -OCH 3 , and said triazolyl moiety is substituted as described in any one of the above embodiments describing substituted triazoly groups.
  • R 5 is a substituted triazolyl-pyridyl- group wherein said pyridyl moiety is substituted with one -OCH 3 , and said triazolyl moiety is substituted on the nitrogen with a hydroxyl substituted alkyl group.
  • R 5 is a substituted triazolyl-pyridyl- wherein said pyridyl moiety is substituted with one -OCH 3 , and said triazolyl moiety is substituted on the nitrogen with a -CH 2 CH 2 OH group.
  • R 5 is a substituted triazolyl-pyridyl- wherein said pyridyl moiety is substituted with one -OCH 3 , and said triazolyl moiety is substituted on the nitrogen with a -alkylene-O-alkyl group, and the alkylene moiety of said -alkylene-O-alkyl group is not -CH 2 - (i.e., the alkylene moiety is 2 or more carbons in length).
  • R 5 is a substituted triazolyl-pyridyl- wherein said pyridyl moiety is substituted with one -OCH 3 , and said triazolyl moiety is substituted on the nitrogen with a -CH 2 CH 2 OCH 3 group.
  • R 5 is a substituted triazolyl-pyridyl- wherein said pyridyl moiety is substituted with alkoxy, and said triazolyl moiety is unsubstituted.
  • R 5 is a substituted triazolyl-pyridyl- wherein said pyridyl moiety is substituted with one alkoxy, and said triazolyl moiety is unsubstituted.
  • R 5 is a substituted triazolyl-pyridyl- wherein said pyridyl moiety is substituted with -OCH 3 , and said triazolyl moiety is unsubstituted.
  • R 5 is a substituted triazolyl-pyridyl- wherein said pyridyl moiety is substituted with one -OCH 3 , and said triazolyl moiety is unsubstituted.
  • R 5 is a substituted triazolyl-pyridyl- wherein said triazolyl is substituted with 1 or 2 groups indepenendently selected from the group consisting of: (a) hydroxyl substituted alkyl group (e.g., -CH 2 COH(CH 3 ) 2) and -CH 2 CH 2 OH), (b) alkyl (e.g., -C r C 6 alkyl, -C r C 4 alkyl, -CrC 2 alkyl, and -CH 3 ), (c) -NH 2 , and (d) -alkylene-O-alkyl (e.g., -CH 2 CH 2 OCH 3 ), provided that the alkylene moiety of said -alkylene-O-alkyl group is not -CH 2 - (i.e., the alkylene moiety is 2 or more carbons in length) when said -alkylene-O-alkyl group is bound to the nitrogen
  • R 5 is
  • R is triazolyl-thiazolyl-.
  • R 5 is
  • R 5 is a substituted triazolyl-thiazolyl- wherein said triazolyl is substituted and said thiazolyl is unsubstituted.
  • R 5 is a substituted triazolyl-thiazolyl- wherein said triazolyl is substituted on the nitrogen and said thiazolyl is unsubstituted.
  • R 5 is a substituted triazolyl-thiazolyl- wherein said triazolyl is substituted on the carbon and said thiazolyl is unsubstituted.
  • R 5 is a substituted triazolyl-thiazolyl- wherein said triazolyl is substituted on the nitrogen and on the carbon, and said thiazolyl is unsubstituted.
  • alkyl groups include, for example, -Ci-C 6 alkyl, -Ci-C 4 alkyl, -CrC 2 alkyl, and -CH 3 .
  • halo atoms include, for example, Cl, F and Br.
  • the halo on the thiazolyl is F.
  • the thiazolyl is substituted with one F.
  • alkylamino group examples include, for example, Ci-C 6 alkyl-NH-, CrC 2 alkyl-NH-, CH 3 -NH-, and CH 3 CH 2 -NH-.
  • dialkylamino group examples include, for example, (CrC 6 alkyl) 2 -N- wherein each alkyl is independently selected, (CrC 2 alkyl) 2 -N- wherein each alkyl is independently selected, (CHg) 2 N-, (CH 3 CH 2 ) 2 -N-, and (CH 3 )(CH 3 CH 2 )N-.
  • R 5 is a substituted triazolyl-thiazolyl- wherein said triazolyl is substituted and said thiazolyl is substituted.
  • R 5 is a substituted triazolyl-thiazolyl- wherein said triazolyl is substituted on the nitrogen and said thiazolyl is substituted.
  • R 5 is a substituted triazolyl-thiazolyl- wherein said triazolyl is substituted on the carbon and said thiazolyl is substituted.
  • R 5 is a substituted triazolyl-thiazolyl- wherein said triazolyl is substituted on the nitrogen and on the carbon, and said thiazolyl is substituted.
  • R 5 is a substituted triazolyl-thiazolyl- wherein said triazolyl is unsubstituted and said thiazolyl is substituted.
  • R 5 is a substituted triazolyl-thiazolyl- wherein the triazolyl is substituted on the nitrogen with alkyl, and said thiazolyl is unsubstituted.
  • R 5 is a substituted triazolyl-thiazoly- wherein the triazolyl is substituted on the nitrogen with -CH 2 COH(CH 3 )2, and said thiazolyl is unsubstituted.
  • R 5 is a substituted triazolyl-thiazolyl- wherein the triazolyl is substituted on the nitrogen with -CH 2 CH 2 OH, and said thiazolyl is unsubstituted.
  • R 5 is a substituted triazolyl-thiazolyl- wherein the triazolyl is substituted on the nitrogen with an alkyl group and substituted on the carbon with an alkyl group, wherein each alkyl group is independently selected, and said thiazolyl is unsubstituted.
  • R 5 is a substituted triazolyl-thiazolyl- wherein the triazolyl is substituted on the nitrogen with a -CH 3 group, and said thiazolyl is unsubstituted.
  • R 5 is a substituted triazolyl-thiazolyl- wherein the triazolyl is substituted on the nitrogen with a -CH 3 group and on the carbon with a -CH 3 group, and said thiazolyl is unsubstituted.
  • R 5 moiety is a substituted triazolyl- thiazolyl- wherein the triazolyl is substituted on the carbon with a -MH 2 group, and said thiazolyl is unsubstituted.
  • R 5 is a substituted triazolyl-thiazolyl- wherein the triazolyl is substituted on the nitrogen with an -alkylene-O-alkyl group, provided that the alkylene moiety of said -alkylene-O-alkyl group is not -CH 2 - (i.e., the alkylene moiety is 2 or more carbons in length), and said thiazolyl is unsubstituted.
  • R 5 is a substituted triazolyl-thiazolyl- wherein the triazolyl is substituted on the nitrogen with a -CH 2 CH 2 OCH 3 group, and said thiazolyl is unsubstituted.
  • R 5 moiety is a substituted triazolyl- thiazolyl- wherein said thiazolyl moiety is substituted with halo, and said triazolyl moiety is substituted as described in any of the above embodiments describing substituted triazolyl groups.
  • R 5 is a substituted triazolyl-thiazolyl- wherein said thiazolyl moiety is substituted with F, and said triazolyl moiety is substituted as described in any one of the above embodiments describing substituted triazolyl groups.
  • R 5 is a substituted triazolyl-thiazolyl- group wherein said thiazolyl moiety is substituted with one F, and said triazolyl moiety is substituted on the nitrogen with a hydroxyl substituted alkyl group.
  • R 5 is a substituted triazolyl-thiazolyl- wherein said thiazolyl moiety is substituted with one F, and said triazolyl moiety is substituted on the nitrogen with a -CH 2 CH 2 OH group.
  • R 5 is a substituted triazolyl-thiazolyl- wherein said thiazolyl moiety is substituted with one F, and said triazolyl moiety is substituted on the nitrogen with an -alkylene-O-alkyl group.
  • R 5 is a substituted triazolyl-thiazolyl- wherein said thiazolyl moiety is substituted with one F, and said triazolyl moiety is substituted on the nitrogen with a -CH 2 CH 2 OCH 3 group.
  • R 5 is a substituted triazolyl-thiazolyl- wherein said thiazolyl moiety is substituted with halo, and said triazolyl moiety is unsubstituted.
  • R 5 is a substituted triazolyl-thiazolyl- wherein said thiazolyl moiety is substituted with F, and said triazolyl moiety is unsubstituted.
  • R 5 moiety is a substituted triazolyl- thiazolyl- wherein said thiazolyl moiety is substituted with alkoxy, and said triazolyl moiety is substituted as described in any of the above embodiments describing substituted triazolyl groups.
  • R 5 is a substituted triazolyl-thiazolyl- wherein said thiazolyl moiety is substituted with -OCH 3 , and said triazolyl moiety is substituted as described in any one of the above embodiments describing substituted triazolyl groups.
  • R 5 is a substituted triazolyl-thiazolyl- group wherein said thiazolyl moiety is substituted with one -OCH 3 , and said triazolyl moiety is substituted on the nitrogen with a hydroxyl substituted alkyl group.
  • R 5 is a substituted triazolyl-thiazolyl- wherein said thiazolyl moiety is substituted with one -OCH 3 , and said triazolyl moiety is substituted on the nitrogen with a -CH 2 CH 2 OH group.
  • R 5 is a substituted triazolyl-thiazolyl- wherein said thiazolyl moiety is substituted with one -OCH 3 , and said triazolyl moiety is substituted on the nitrogen with an -alkylene-O-alkyl group, and the alkylene moiety of said -alkylene-O-alkyl group is not -CH 2 - (i.e., the alkylene moiety is 2 or more carbons in length).
  • R 5 is a substituted triazolyl-thiazolyl- wherein said thiazolyl moiety is substituted with one -OCH 3 , and said triazolyl moiety is substituted on the nitrogen with a -CH 2 CH 2 OCH 3 group.
  • R 5 is a substituted triazolyl-thiazolyl- wherein said thiazolyl moiety is substituted with alkoxy, and said triazolyl moiety is unsubstituted.
  • R 5 is a substituted triazolyl-thiazolyl- wherein said thiazolyl moiety is substituted with -OCH 3 , and said triazolyl moiety is unsubstituted.
  • R 5 is a substituted triazolyl-thiazolyl- wherein said thiazolyl moiety is substituted with an alkylamino, and said triazolyl moiety is substituted as described in any of the above embodiments describing triazolyl groups.
  • R 5 is a substituted triazolyl-thiazolyl- wherein said thiazolyl moiety is substituted with a dialkylamino, and said triazolyl moiety is substituted as described in any of the above embodiments describing substituted triazolyl groups.
  • R 5 is a substituted triazolyl-thiazolyl- wherein said thiazolyl moiety is substituted with an alkylamino, and said triazolyl moiety is unsubstituted.
  • R 5 is a substituted triazolyl-thiazolyl- wherein said thiazolyl moiety is substituted with a dialkylamino, and said triazolyl moiety is unsubstituted.
  • R 5 is a substituted triazolyl-thiazolyl- wherein said triazolyl is substituted with 1 or 2 groups indepenendently selected from the group consisting of: (a) hydroxyl substituted alkyl group (e.g., -CH2COH(CH 3 ) 2 , and -CH 2 CH 2 OH) 1 (b) alkyl (e.g., -Ci-C 6 alkyl, -Ci-C 4 alkyl, -C r C 2 alkyl, and -CH 3 ), -NH 2 , and (c) -alkylene-O-alkyl (e.g., -CH 2 CH 2 OCH 3 ), provided that the alkylene moiety of said -alkylene-O-alkyl group is not -CH 2 - (i.e., the alkylene moiety is 2 or more carbons in length) when said -alkylene-O-alkyl group is bound to the nitrogen
  • R 5 is pyridazinyl-thienyl-. In another embodiment of this invention R 5 is
  • alkyl groups include, for example, -C r C 6 alkyl, -CrC 4 alkyl, -Ci-C 2 alkyl, and -CH 3 .
  • halo atoms include, for example, Cl, F and Br.
  • the halo on the pyridazinyl is F.
  • the pyridazinyl is substituted with one F.
  • alkylamino group examples include, for example, CrC 6 alkyl-NH-, CrC 2 alkyl-NH-, CH 3 -MH-, and CH 3 CH 2 -NH-.
  • dialkylamino group examples include, for example, (C r C 6 alkyl) 2 -N- wherein each alkyl is independently selected, (Ci-C 2 alkyl) 2 -N- wherein each alkyl is independently selected, (CHg) 2 N-, (CH 3 CH 2 ) 2 -N-, and (CH 3 )(CH 3 CH 2 )N-.
  • R 5 is a substituted pyridazinyl-thienyl- wherein said pyridazinyl is substituted and said thienyl is unsubstituted.
  • R 5 is a substituted pyridazinyl-thienyl- wherein said pyridazinyl is substituted with an alkyl group, and said thienyl is unsubstituted.
  • R 5 is a substituted pyridazinyl-thienyl- wherein said pyridazinyl is substituted with a methyl group, and said thienyl is unsubstituted.
  • R 5 is a substituted pyridazinyl-thienyl- wherein said pyridazinyl is substituted with an amino group, and said thienyl is unsubstituted.
  • R 5 is a substituted pyridazinyl-thienyl- wherein said pyridazinyl is substituted with an alkylamino group, and said thienyl is unsubstituted.
  • R 5 is a substituted pyridazinyl-thienyl- wherein said pyridazinyl is substituted with a dialkylamino group, and said thienyl is unsubstituted.
  • R 5 is a substituted pyridazinyl-thienyl- wherein said pyridazinyl is substituted and said thienyl is substituted.
  • R 5 is a substituted pyridazinyl-thienyl- wherein said pyridazinyl is unsubstituted and said thienyl is substituted.
  • R 5 is a substituted pyridazinyl-thienyl- wherein said pyridazinyl is unsubstituted and said thienyl is substituted with 1 to 2 independently selected halos.
  • R 5 is a substituted pyridazinyl-thienyl- wherein said pyridazinyl is unsubstituted and said thienyl is substituted with 1 to 2 halos independently selected from the group consisting of: Br, Cl and F.
  • R 5 is a substituted pyridazinyl-thienyl- wherein said pyridazinyl is unsubstituted and said thienyl is substituted with 1 to 2 independently selected alkoxy groups.
  • R 5 is a substituted pyridazinyl-thienyl- wherein said pyridazinyl is unsubstituted and said thienyl is substituted with 1 to 2 -OCH 3 groups.
  • R 5 is a substituted pyridazinyl-thienyl- wherein said pyridazinyl is substituted as described in any one of the embodiments above describing substituted pyridazinyl groups, and said thienyl is substituted.
  • R 5 is a substituted pyridazinyl-thienyl- wherein said pyridazinyl is substituted as described in any one of the embodiments above describing substituted pyridazinyl groups, and said thienyl is substituted with 1 to 2 independently selected halos.
  • R 5 is a substituted pyridazinyl-thienyl- wherein said pyridazinyl is substituted as described in any one of the embodiments above describing substituted pyridazinyl groups, and said thienyl is substituted with 1 to 2 halos independently selected from the group consisting of: Br, Cl and F.
  • R 5 is a substituted pyridazinyl-thienyl- wherein said pyridazinyl is substituted as described in any one of the embodiments above describing substituted pyridazinyl groups, and said thienyl is substituted with 1 to 2 independently selected alkoxy groups.
  • R 5 is a substituted pyridazinyl-thienyl- wherein said pyridazinyl is substituted as described in any one of the embodiments above describing substituted pyridazinyl groups, and said thienyl is substituted with 1 to 2 -OCH 3 groups.
  • alkoxy e.g., -OCH 3
  • halo e.g., Br, Cl and F
  • R 5 is
  • R 5 is
  • R 5 is a substituted triazolyl-phenyl- group wherein (a) said triazolyl moiety is optionally substituted on the nitrogen with a substituent selected from the group consisting of: -CH 2 COH(CH 3 ) 2 and -CH 2 CH 2 OH, (b) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is optionally substituted on the nitrogen with a -CH 3 group, (e) said triazolyl moiety is optionally substituted on the nitrogen with one -CH 3 group, and on the carbon with one -CH 3 group, (f) said triazolyl moiety is optionally substituted on the carbon with a -NH 2 group, or (g) said triazolyl moiety is optionally substituted on the nitrogen with a -NH 2 group, or
  • R 5 is a substituted triazolyl-phenyl- group wherein (a) said triazolyl moiety is optionally substituted on the nitrogen with a substituent selected from the group consisting of: -CH 2 COH(CH 3 ) 2 and -CH 2 CH 2 OH, (b) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is optionally substituted on the nitrogen with a -CH 3 group, (e) said triazolyl moiety is optionally substituted on the nitrogen with one -CH 3 group, and on the carbon with one -CH 3 group, (f) said triazolyl moiety is optionally substituted on the carbon with a -NH 2 group, or (g) said triazolyl moiety is optionally substituted on the nitrogen with a -NH 2 group, or
  • R 5 is a substituted triazolyl-phenyl- group wherein (a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH 2 COH(CH 3 ) 2 and -CH 2 CH 2 OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH 3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH 3 group, and on the carbon with one -CH 3 group, (f) said triazolyl moiety is substituted on the carbon with a -NH 2 group, or (g) said triazolyl moiety is substituted on the nitrogen with a
  • R 5 is a substituted triazolyl-phenyl- group wherein (a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH 2 COH(CH 3 ) 2 and -CH 2 CH 2 OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH 3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH 3 group, and on the carbon with one -CH 3 group, (f) said triazolyl moiety is substituted on the carbon with a -MH 2 group, or (g) said triazolyl moiety is substituted on the nitrogen with a -CH 2 CH 2 OCH 3 group; and wherein said triazolyl moiety
  • R 5 is a substituted triazolyl-phenyl- group wherein (a) said triazolyl moiety is substituted on the nitrogen with a substituent S ⁇ lect ⁇ d from the group consisting of: -CH 2 COH(CH 3 ⁇ and -CH 2 CH 2 OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH 3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH 3 group, and on the carbon with one -CH 3 group, (f) said triazolyl moiety is substituted on the carbon with a -NH 2 group, or (g) said triazolyl moiety is substituted on the nitrogen with a -CH 2 CH 2 OCH 3 group; and wherein (b) said
  • R 5 is a substituted triazolyl-phenyl- group wherein (a) said triazolyl moiety is optionally substituted on the nitrogen with a substituent selected from the group consisting of: -CH 2 COH(CH 3 ) 2 and -CH 2 CH 2 OH, (b) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is optionally substituted on the nitrogen with a -CH 3 group, (e) said triazolyl moiety is optionally substituted on the nitrogen with one -CH 3 group, and on the carbon with one -CH 3 group, (f) said triazolyl moiety is optionally substituted on the carbon with a -NH 2 group, or (g) said triazolyl moiety is optionally substituted on the nitrogen with a -NH 2 group, or
  • R 5 is a substituted triazolyl-phenyl- group wherein (a) said triazolyl moiety is optionally substituted on the nitrogen with a substituent selected from the group consisting of: -CH 2 COH(CH 3 ) 2 and -CH 2 CH 2 OH, (b) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is optionally substituted on the nitrogen with a -CH 3 group, (e) said triazolyl moiety is optionally substituted on the nitrogen with one -CH 3 group, and on the carbon with one -CH 3 group, (f) said triazolyl moiety is optionally substituted on the carbon with a -NH 2 group, or (g) said triazolyl moiety is optionally substituted on the nitrogen with a -NH 2 group, or
  • R 5 is a substituted triazolyl-phenyl- group wherein (a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH 2 COH(CH 3 ) 2 and -CH 2 CH 2 OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH 3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH 3 group, and on the carbon with one -CH 3 group, (f) said triazolyl moiety is substituted on the carbon with a -NH 2 group, or (g) said triazolyl moiety is substituted on the nitrogen with a
  • R 5 is a substituted triazolyl-phenyl- group wherein (a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH 2 COH(CH 3 ) 2 and -CH 2 CH 2 OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH 3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH 3 group, and on the carbon with one -CH 3 group, (f) said triazolyl moiety is substituted on the carbon with a -NH 2 group, or (g) said triazolyl moiety is substituted on the nitrogen with a -CH 2 CH 2 OCH 3 group; and wherein said triazolyl moiety
  • R 5 is a substituted triazolyl-thienyl- group wherein (a) said triazolyl moiety is optionally substituted on the nitrogen with a substituent selected from the group consisting of: -CH 2 COH(CH 3 ) 2 and -CH 2 CH 2 OH, (b) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is optionally substituted on the nitrogen with a -CH 3 group, (e) said triazolyl moiety is optionally substituted on the nitrogen with one -CH 3 group, and on the carbon with one -CH 3 group, (f) said triazolyl moiety is optionally substituted on the carbon with a -MH 2 group, or (g) said triazolyl moiety is optionally substituted on the nitrogen with a -MH 2 group,
  • R 5 is a substituted triazolyl-thienyl- group wherein (a) said triazolyl moiety is optionally substituted on the nitrogen with a substituent selected from the group consisting of: -CH 2 COH(CH 3 ) 2 and -CH 2 CH 2 OH, (b) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is optionally substituted on the nitrogen with a -CH 3 group, (e) said triazolyl moiety is optionally substituted on the nitrogen with one -CH 3 group, and on the carbon with one -CH 3 group, (f) said triazolyl moiety is optionally substituted on the carbon with a -NH 2 group, or (g) said triazolyl moiety is optionally substituted on the nitrogen with a -NH 2 group,
  • R 5 is a substituted triazolyl-thienyl- group wherein (a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH 2 COH(CH 3 ) 2 and -CH 2 CH 2 OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH 3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH 3 group, and on the carbon with one -CH 3 group, (f) said triazolyl moiety is substituted on the carbon with a -NH 2 group, or (g) said triazolyl moiety is substituted on the nitrogen with a -CH 2 CH 2 OCH 3 group; and wherein said triazolyl mo
  • R 5 is a substituted triazolyl-thienyl- group wherein (a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH 2 COH(CH 3 ) 2 and -CH 2 CH 2 OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH 3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH 3 group, and on the carbon with one -CH 3 group, (f) said triazolyl moiety is substituted on the carbon with a -NH 2 group, or (g) said triazolyl moiety is substituted on the nitrogen with a -CH 2 CH 2 OCH 3 group; and wherein said triazolyl mo
  • R 5 is a substituted triazolyl-thienyl- group wherein (a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH 2 COH(CH 3 ) 2 and -CH 2 CH 2 OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH 3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH 3 group, and on the carbon with one -CH 3 group, (f) said triazolyl moiety is substituted on the carbon with a -NH 2 group, or (g) said triazolyl moiety is substituted on the nitrogen with a -CH 2 CH 2 OCH 3 group; and wherein said triazolyl mo
  • R 5 is a substituted triazolyl-thienyl- group wherein (a) said triazolyl moiety is optionally substituted on the nitrogen with a substituent selected from the group consisting of: -CH 2 COH(CH 3 ) 2 , and -CH 2 CH 2 OH, (b) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is optionally substituted on the nitrogen with a -CH 3 group, (e) said triazolyl moiety is optionally substituted on the nitrogen with one -CH 3 group, and on the carbon with one -CH 3 group, (f) said triazolyl moiety is optionally substituted on the carbon with a -NH 2 group, or (g) said triazolyl moiety is optionally substituted on the nitrogen with a -NH 2
  • R 5 is a substituted triazolyl-thienyl- group wherein (a) said triazolyl moiety is optionally substituted on the nitrogen with a substituent selected from the group consisting of: -CH 2 COH(CH 3 ) 2 and -CH 2 CH 2 OH, (b) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is optionally substituted on the nitrogen with a -CH 3 group, (e) said triazolyl moiety is optionally substituted on the nitrogen with one -CH 3 group, and on the carbon with one -CH 3 group, (f) said triazolyl moiety is optionally substituted on the carbon with a -MH 2 group, or (g) said triazolyl moiety is optionally substituted on the nitrogen with a -MH 2 group,
  • R 5 is a substituted triazolyl-thienyl- group wherein (a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH 2 COH(CH 3 ) 2 and -CH 2 CH 2 OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH 3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH 3 group, and on the carbon with one -CH 3 group, (f) said triazolyl moiety is substituted on the carbon with a -NH 2 group, or (g) said triazolyl moiety is substituted on the nitrogen with a -CH 2 CH 2 OCH 3 group; and wherein said triazolyl mo
  • R 5 is a substituted triazolyl-thienyl- group wherein (a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH 2 COH(CH 3 ) 2 , and -CH 2 CH 2 OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH 3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH 3 group, and on the carbon with one -CH 3 group, (f) said triazolyl moiety is substituted on the carbon with a -NH 2 group, or (g) said triazolyl moiety is substituted on the nitrogen with a -CH 2 CH 2 OCH 3 group; and wherein (b) said
  • Embodiment No. 1 is directed to compounds of formula 1.0 wherein R 2 is a
  • R 1 is substituted pyridyl.
  • Embodiment No. 2 is directed to compounds of formula 1.0 wherein R 2 is a -O-(CrC2)alkyl group, and R 1 is pyridyl substituted with one substituent.
  • Embodiment No. 3 is directed to compounds of formula 1.0 wherein R 2 is a -O-(Ci-C 2 )alkyl group, and R 1 is selected from the group consisting of:
  • Embodiment No. 4 is directed to compounds of formula 1.0 wherein R 2 is a -O-(Ci-C 2 )alkyl group, and R 1 is:
  • Embodiment No. 5 is directed to compounds of formula 1.0 wherein R 2 is a
  • R J1 is, :
  • Embodiment No. 6 is directed to compounds of formula 1.0 wherein R 2 is a -O-(Ci-C 2 )alkyl group, and R 1 is: wherein said alkoxy group is -OCH(CH 3 ) 2 .
  • Embodiment No. 7 is directed to compounds of formula 1.0 wherein, R 2 is a -O-(Ci-C 2 )alkyl group, and R 1 is: wherein said alkoxy group is -OC 2 H 5 .
  • Embodiment No. 8 is directed to compounds of formula 1.0 wherein R 2 is a
  • R 1 is:
  • Embodiment No. 9 is directed to compounds of formula 1.0 wherein R 2 is a
  • R 1 is:
  • Embodiment No. 10 is directed to compounds of formula 1.0 wherein z is 1 , R 2 is a -O-(Ci-C 2 )alkyl group, and R 1 is: wherein said -O-alkylene-O-alkyl group is -OCH 2 CH 2 OCH 3 .
  • Embodiment No. 11 is directed to compounds of formula 1.0 wherein R 2 is a -O-(CrC 2 )alkyl group, and R 1 is:
  • Embodiment No. 12 is directed to compounds of formula 1.0 wherein R 2 is
  • Embodiment No. 13 is directed to compounds of formula 1.0 wherein R 2 . is, -OCH 3 , and R 1 is pyridyl substituted with one substituent.
  • Embodiment No. 14 is directed to compounds of formula 1.0 wherein R 2 is -OCH 3 , and R 1 is selected from the group consisting of:
  • Embodiment No. 15 is directed to compounds of formula 1.0 wherein R 2 is -OCH 3 , and R 1 is:
  • Embodiment No. 16 is directed to compounds of formula 1.0 wherein R 2 is -OCH 3 , and R 1 is:
  • Embodiment No. 17 is directed to compounds of formula 1.0 wherein R 2 is
  • R 1 is: wherein said alkoxy group is -OCH(CH 3 ) 2 .
  • Embodiment No. 18 is directed to compounds of formula 1.0 wherein R 2 is -OCH 3 , and R 1 is: wherein said alkoxy group is -OC 2 H 5 .
  • Embodiment No. 19 is directed to compounds of formula 1.0 wherein R 2 is -OCH 3 , and R 1 is: wherein said alkoxy group is -OC 2 H 5 .
  • Embodiment No. 19 is directed to compounds of formula 1.0 wherein R 2 is
  • R 1 is:
  • Embodiment No. 20 is directed to compounds of formula 1.0 wherein R 2 is
  • R 1 is:
  • Embodiment No. 21 is directed to compounds of formula 1.0 wherein R 2 is -OCH 3 , and R 1 is: wherein said -O-alkylene-O-alkyl group is -OCH 2 CH 2 OCH 3 .
  • EEmmbbooddiimmeernt No. 22 is directed to compounds of formula 1.0 wherein R 2 is -OCH 3 , and R 1 is:
  • Embodiment No. 23 is directed to compounds of formula 1.0 wherein R 2 is a -S-(Ci -C 2 )alkyl group, and R 1 is substituted pyridyl.
  • Embodiment No. 24 is directed to compounds of formula 1.0 wherein R 2 is a -S-(C 1 -C 2 )alkyl group, and R 1 is pyridyl substituted with one substituent.
  • Embodiment No. 25 is directed to compounds of formula 1.0 wherein R 2 is a
  • R 1 is selected from the group consisting of:
  • Embodiment No. 26 is directed to compounds of formula 1.0 wherein R 2 is a -S-(Ci -C 2 )alkyl group, and R 1 is:
  • Embodiment No. 27 is directed to compounds of formula 1.0 wherein R 2 is a -S-(Ci -C 2 )alkyl group, and R 1 is:
  • Embodiment No. 28 is directed to compounds of formula 1.0 wherein R 2 is a
  • R 1 is: wherein said alkoxy group is -OCH(CH 3 ) 2 .
  • alkoxy group is -OC 2 H 5 .
  • Embodiment No. 30 is directed to compounds of formula 1.0 wherein R 2 is a -S-(Ci -C 2 )alkyl group, and R 1 is:
  • Embodiment No. 31 is directed to compounds of formula 1.0 wherein R 2 is a -S-(Ci -C 2 )alkyl group, and R 1 is:
  • Embodiment No. 32 is directed to compounds of formula 1.0 wherein R 2 is a
  • R 1 is: wherein said -O-alkylene-O-alkyl group is -OCH 2 CH 2 OCH 3 .
  • Embodiment No. 33 is directed to compounds of formula 1.0 wherein R 2 is a S-(Ci -C 2 )alkyl group, and R 1 is:
  • Embodiment No. 34 is directed to compounds of formula 1.0 wherein R 2 is -SCH 3 , and R 1 is substituted pyridyl.
  • Embodiment No. 35 is directed to compounds of formula 1.0 wherein R 2 is -SCH 3 , and R 1 is pyridyl substituted with one substituent.
  • Embodiment No. 36 is directed to compounds of formula 1.0 wherein R 2 is -SCH 3 , and R 1 is selected from the group consisting of:
  • Embodiment No. 37 is directed to compounds of formula 1.0 wherein R 2 is
  • Embodiment No. 38 is directed to compounds of formula 1.0 wherein R 2 is
  • R 1 is:
  • Embodiment No. 39 is directed to compounds of formula 1.0 wherein R 2 is -SCH 3 , and R 1 is: wherein said alkoxy group is -OCH(CH 3 ) 2 .
  • Embodiment No. 40 is directed to compounds of formula 1.0 wherein R 2 is -SCH 3 , and R 1 is: wherein said alkoxy group is -OC 2 H 5 .
  • Embodiment No. 41 is directed to compounds of formula 1.0 wherein R 2 is -SCH 3 , and R 1 is:
  • Embodiment No. 42 is directed to compounds of formula 1.0 wherein R 2 is
  • R 1 is:
  • Embodimen itt NNoo.. 4433 iiss ddiirreecctteedd ttoo ccoompounds of formula 1.0 wherein R 2 is -SCH 3 , and R 1 is: wherein said -O-alkylene-0-alkyl group is -OCH 2 CH 2 OCH 3 .
  • Embodiment No. 44 is directed to compounds of formula 1.0 wherein R 2 is -SCH 3 , and R 1 is:
  • Embodiment No. 45 is directed to compounds of formula 1.0 having the formula 1.1 :
  • Embodiment No. 46 is directed to any one of Embodiment Numbers 1 to 44 wherein the compound of formula 1.0 is a compound of formula 1.1.
  • embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R 5 is a substituted triazolyl-phenyl- wherein the triazolyl moiety is substituted with one or two alkyl groups selected from the group consisting of: -CrC 6 alkyl, -CrC 4 alkyl, -CrC 2 alkyl, and -CH 3 . .
  • R 5 is a substituted triazolyl-phenyl- wherein the triazolyl moiety is substituted with one or two alkyl groups selected from the group consisting of: -Ci-C 4 alkyl, -CrC 2 alkyl, and -CH 3 .
  • R 5 is a substituted triazolyl- phenyl- wherein the triazolyl moiety is substituted with one or two -CH 3 groups.
  • R 5 is a substituted triazolyl-phenyl- wherein the triazolyl moiety is substituted on the nitrogen with an alkyl group selected from the group consisting of: -Ci-C 4 alkyl, -CrC 2 alkyl, and -CH 3 .
  • R 5 is a substituted triazolyl-phenyl- wherein the triazolyl moiety is substituted on the nitrogen with -CH 3 group.
  • R 5 is a substituted triazolyl-phenyl- wherein the triazolyl moiety is substituted on the nitrogen with an -alkylene-O-alkyl group selected from the group consisting of: -C 2 -C 4 alkylene-O-d- C 6 alkyl, -C 2 alkylene-O-C r C 2 alkyl, -C 2 -C 4 alkylene-O-CH 3 , and -CH 2 CH 2 OCH 3 .
  • R 5 is a substituted triazolyl-phenyl- wherein the triazolyl moiety is substituted on the nitrogen with an -alkylene-O-alkyl group selected from the group consisting of: -C 2 alkylene-O-C r C 2 alkyl, and -CH 2 CH 2 OCH 3 .
  • R 5 is a substituted triazolyl-phenyl- wherein the triazolyl moiety is substituted on the nitrogen with -CH 2 CH 2 OCH 3 .
  • R 5 is a substituted triazolyl-phenyl- wherein the triazolyl moiety is substituted on the nitrogen with a hydroxy substituted alkyl group selected from the group consisting of: hydroxy substituted -CrC 4 alkyl, hydroxy substituted -Ci-C 2 alkyl, and hydroxy substituted -CH 3 .
  • R 5 is a substituted triazolyl-phenyl- wherein the triazolyl moiety is substituted on the nitrogen with a hydroxy substituted alkyl group selected from the group consisting of: CH 2 COH(CH 3 ) 2 , and -CH 2 CH 2 OH.
  • R 5 is a substituted triazolyl-phenyl- wherein the triazolyl moiety is substituted on the nitrogen with a hydroxy substituted alkyl group selected from the group consisting of: CH 2 COH(CH 3 ) 2 , and -CH 2 CH 2 OH.
  • R 5 is a substituted triazolyl-phenyl- wherein the triazolyl moiety is substituted on the nitrogen with a hydroxy substituted alkyl group selected from the group consisting of: CH 2 COH(CH 3 ) 2 , and -CH 2 CH 2 OH.
  • R 5 is a substituted triazolyl-phenyl- wherein the triazolyl moiety is substituted on the nitrogen with
  • R 5 is a substituted triazolyl-phenyl- group wherein (a) said triazolyl moiety is optionally substituted on the nitrogen with a substituent selected from the group consisting of: -CH 2 COH(CH 3 ) 2 and -CH 2 CH 2 OH, (b) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is optionally substituted on the nitrogen with a -CH 3 group, (e) said triazolyl moiety is optionally substituted on the nitrogen with one -CH 3 group, and on the carbon with one -CH 3 group, (f) said triazolyl moiety is optionally substituted on the carbon with a -NH 2 group
  • R 5 is a substituted triazolyl-phenyl- group wherein (a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH 2 COH(CH 3 ) 2 and -CH 2 CH 2 OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH 3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH 3 group, and on the carbon with one -CH 3 group, (f) said triazolyl moiety is substituted on the carbon with a -NH 2 group, or (g) said triazolyl moiety
  • R 5 is a substituted triazolyl-phenyl- group wherein (a) said triazolyl moiety is optionally substituted on the nitrogen with a substituent selected from the group consisting of: -CH 2 COH(CH 3 ) 2 , and -CH 2 CH 2 OH, (b) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is optionally substituted on the nitrogen with a -CH 3 group, (e) said triazolyl moiety is optionally substituted on the nitrogen with one -CH 3 group, and on the carbon with one -CH 3 group, (f) said triazolyl moiety is optionally substituted on the carbon with a -NH
  • R 5 is a substituted triazolyl-phenyl- group wherein (a) said triazolyl moiety is optionally substituted on the nitrogen with a substituent selected from the group consisting of: -CH 2 COH(CH 3 ) 2 , and -CH 2 CH 2 OH, (b) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is optionally substituted on the nitrogen with a -CH 3 group, (e) said triazolyl moiety is optionally substituted on the nitrogen with one -CH 3 group, and on the carbon with one -CH 3 group, (f) said triazolyl moiety is optionally substituted on the carbon with a -NH
  • R 5 is a substituted triazolyl-phenyl- group wherein (a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH 2 COH(CH 3 ) 2> and -CH 2 CH 2 OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH 3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH 3 group, and on the carbon with one -CH 3 group, (f) said triazolyl moiety is substituted on the carbon with a -NH 2 group, or (g) said triazolyl mo
  • R 5 is a substituted triazolyl-phenyl- group wherein (a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH 2 COH(CH 3 ) 2 , and -CH 2 CH 2 OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH 3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH 3 group, and on the carbon with one -CH 3 group, (f) said triazolyl moiety is substituted on the carbon with a -NH 2 group, or (g) said triazolyl
  • R 5 is a substituted triazolyl-phenyl- group wherein (a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH 2 COH(CH 3 ) 2 , and -CH 2 CH 2 OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH 3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH 3 group, and on the carbon with one -CH 3 group, (f) said triazolyl moiety is substituted on the carbon with a -NH 2 group, or (g) said triazolyl
  • R 5 moiety is a substituted triazolyl-phenyl group wherein said triazolyl moiety is substituted with: (a) one substituent selected from the group consisting of: -CH 2 COH(CH 3 ) 2 , and -CH 2 CH 2 OH, (b) one alkyl group, (c) two alkyl groups, (d) one -CH 3 group, (e) two -CH 3 groups, (f) one -NH 2 group, or (g) one -CH 2 CH 2 OCH 3 group.
  • R 5 is a substituted triazolyl-thienyl- wherein the triazolyl moiety is substituted with one or two alkyl groups selected from the group consisting of: -CrC 6 alkyl, -Ci-C 4 alkyl, -CrC 2 alkyl, and -CH 3 .
  • R 5 is a substituted triazolyl-thienyl- wherein the triazolyl moiety is substituted with one or two alkyl groups selected from the group consisting of: -C- ⁇ -C 4 alkyl, -CrC 2 alkyl, and -CH 3 .
  • R 5 is a substituted triazolyl- thienyl- wherein the triazolyl moiety is substituted with one or two -CH 3 groups.
  • R 5 is a substituted triazolyl-thienyl- wherein the triazolyl moiety is substituted on the nitrogen with an alkyl group selected from the group consisting of: -Ci-C 4 alkyl, -C r C 2 alkyl, and -CH 3 .
  • R 5 is a substituted triazolyl-thienyl- wherein the triazolyl moiety is substituted on the nitrogen with -CH 3 .
  • R 5 is a substituted triazolyl-thienyl- wherein the triazolyl moiety is substituted on the nitrogen with an -alkylene-O-alkyl group selected from the group consisting of: -C 2 -C 4 alkylene-O-C r C 6 alkyl, -Cj.alkylene-O-CrCj.alkyl, -Cj.-C. t alkylene-0-CHs, and -CH 2 CH 2 OCH 3 .
  • R 5 is a substituted triazolyl-thienyl- wherein the triazolyl moiety is substituted on the nitrogen with an -alkylene-O-alkyl group selected from the group consisting of: -C 2 alkylene-O-d- C 2 alkyl, and-CH 2 CH 2 OCH 3 .
  • R 5 is a substituted triazolyl-thienyl- wherein the triazolyl moiety is substituted on the nitrogen with -CH 2 CH 2 OCH 3 .
  • R 5 is a substituted triazolyl-thienyl- wherein the triazolyl moiety is substituted on the nitrogen with a hydroxy substituted alkyl group selected from the group consisting of: hydroxy substituted -CrC 4 alkyl, hydroxy substituted -C r C 2 alkyl, and hydroxy substituted -CH 3 .
  • R 5 is a substituted triazolyl-thienyl- wherein the triazolyl moiety is substituted on the nitrogen with a hydroxy substituted alkyl group selected from the group consisting of: CH 2 COH(CH 3 ) 2 , and -CH 2 CH 2 OH.
  • inventions of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein the optional halo substituents for the thienyl moiety of R 5 are independently selected from the group consisting of: Cl, F and Br.
  • Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein the optional halo substituents for the thienyl moiety of R 5 are F.
  • Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein the optional halo substituent for the thienyl moiety of R 5 is one F.
  • R 5 is a substituted triazolyl-thienyl- group wherein (a) said triazolyl moiety is optionally substituted on the nitrogen with a substituent selected from the group consisting of: -CH 2 COH(CH 3 )2, and -CH 2 CH 2 OH, (b) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is optionally substituted on the nitrogen with a -CH 3 group, (e) said triazolyl moiety is optionally substituted on the nitrogen with one -CH 3 group, and on the carbon with one -CH 3 group, (f) said triazolyl moiety is optionally substituted on the carbon with a -NH 2
  • R 5 is a substituted triazolyl-thienyl- group wherein (a) said triazolyl moiety is optionally substituted on the nitrogen with a substituent selected from the group consisting of:-CH 2 COH(CH 3 ) 2 , -CH 2 CH 2 OH, (b) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is optionally substituted on the nitrogen with a -CH 3 group, (e) said triazolyl moiety is optionally substituted on the nitrogen with one -CH 3 group, and on the carbon with one -CH 3 group, (f) said triazolyl moiety is optionally substituted on the carbon with a -NH 2
  • R 5 is a substituted triazolyl-thienyl- group wherein (a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH 2 COH(CH 3 ) 2 , and -CH 2 CH 2 OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH 3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH 3 group, and on the carbon with one -CH 3 group, (f) said triazolyl moiety is substituted on the carbon with a -NH 2 group, or (g) said triazoly
  • R 5 is a substituted triazolyl-thienyl- group wherein (a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH 2 COH(CH 3 ) 2 , and -CH 2 CH 2 OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH 3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH 3 group, and on the carbon with one -CH 3 group, (f) said triazolyl moiety is substituted on the carbon with a -NH 2 group, or (g) said triazoly
  • R 5 is a substituted triazolyl-thienyl- group wherein (a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH 2 COH(CH 3 ) 2 , and -CH 2 CH 2 OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH 3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH 3 group, and on the carbon with one -CH 3 group, (f) said triazolyl moiety is substituted on the carbon with a -NH 2 group, or (g) said triazoly
  • R 5 moiety is a substituted triazolyl-thienyl group wherein said triazolyl moiety is substituted with: (a) one substituent selected from the group consisting of: -CH 2 COH(CH 3 )2, and -CH 2 CH 2 OH, (b) one alkyl group, (c) two alkyl groups, (d) one -CH 3 group, (e) two -CH 3 groups, (f) one -NH 2 group, or (g) one -CH 2 CH 2 OCH 3 group.
  • R 5 is a substituted triazolyl-thienyl- group wherein (a) said triazolyl moiety is optionally substituted on the nitrogen with a substituent selected from the group consisting of: -CH 2 COH(CH 3 ) 2 , and -CH 2 CH 2 OH 1 (b) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is optionally substituted on the nitrogen with a -CH 3 group, (e) said triazolyl moiety is optionally substituted on the nitrogen with one -CH 3 group, and on the carbon with one -CH 3 group, (f) said triazolyl moiety is optionally substituted on the carbon with a -
  • R 5 is a substituted triazolyl-thienyl- group wherein (a) said triazolyl moiety is optionally substituted on the nitrogen with a substituent selected from the group consisting of: -CH 2 COH(CH 3 ) 2 , and -CH 2 CH 2 OH, (b) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is optionally substituted on the nitrogen with a -CH 3 group, (e) said triazolyl moiety is optionally substituted on the nitrogen with one -CH 3 group, and on the carbon with one -CH 3 group, (f) said triazolyl moiety is optionally substituted on the carbon with a -
  • R 5 is a substituted triazolyl-thienyl- group wherein (a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH 2 COH(CH 3 ) 2 , and -CH 2 CH 2 OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH 3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH 3 group, and on the carbon with one -CH 3 group, (f) said triazolyl moiety is substituted on the carbon with a -MH 2 group, or (g) said triazoly
  • R 5 is a substituted triazolyl-thienyl- group wherein (a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH 2 COH(CH 3 ) 2 , and -CH 2 CH 2 OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH 3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH 3 group, and on the carbon with one -CH 3 group, (f) said triazolyl moiety is substituted on the carbon with a -NH 2 group, or (g) said triazoly
  • R 5 is a substituted triazolyl-pyridyl- wherein the triazolyl moiety is substituted with one or two alkyl groups selected from the group consisting of: -CrC 6 alkyl, -CrC 4 alkyl, -C r C 2 alkyl, and -CH 3 .
  • R 5 is a substituted triazolyl-pyridyl- wherein the triazolyl moiety is substituted with one or two alkyl groups selected from the group consisting of: -CrC 4 alkyl, -CrC 2 alkyl, and -CH 3 .
  • R 5 is a substituted triazolyl- pyridyl- wherein the triazolyl moiety is substituted with one or two -CH 3 groups.
  • R 5 is a substituted triazolyl-pyridyl- wherein the triazolyl moiety is substituted on the nitrogen with an alkyl group selected from the group consisting of: -CrC 4 alkyl, -Ci-C 2 alkyl, and -CH 3 .
  • R 5 is a substituted triazolyl-pyridyl- wherein the triazolyl moiety is substituted on the nitrogen with -CH 3 .
  • R 5 is a substituted triazolyl-pyridyl- wherein the triazolyl moiety is substituted on the nitrogen with an -alkylene-O-alkyl group selected from the group consisting of: -C 2 -C 4 alkylene-O-d- C 6 alkyl, -C 2 alkylene-O-C r C 2 alkyl, -CrC ⁇ lkylene-O-CHa, and -CH 2 CH 2 OCH 3 .
  • R 5 is a substituted triazolyl-pyridyl- wherein the triazolyl moiety is substituted on the nitrogen with an -alkylene-O-alkyl group selected from the group consisting of: -C 2 alkylene-O-Ci- C 2 alkyl, and -CH 2 CH 2 OCH 3 .
  • R 5 is a substituted triazolyl-pyridyl- wherein the triazolyl moiety is substituted on the nitrogen with a hydroxy substituted alkyl group selected from the group consisting of: hydroxy substituted -CrC 4 alkyl, hydroxy substituted -CrC 2 alkyl, and hydroxy substituted -CH 3 .
  • R 5 is a substituted triazolyl-pyridyl- wherein the triazolyl moiety is substituted on the nitrogen with a hydroxy substituted alkyl group selected from the group consisting of: CH 2 COH(CH 3 ) 2 , and -CH 2 CH 2 OH.
  • R 5 is a substituted triazolyl-pyridyl- group wherein (a) said triazolyl moiety is optionally substituted on the nitrogen with a substituent selected from the group consisting of: -CH 2 COH(CH 3 ) 2 , and -CH 2 CH 2 OH, (b) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is optionally substituted on the nitrogen with a -CH 3 group, (e) said triazolyl moiety is optionally substituted on the nitrogen with one -CH 3 group, and on the carbon with one -CH 3 group, (f) said triazolyl moiety is optionally substituted on the carbon with a
  • R 5 is a substituted triazolyl-pyridyl- group wherein (a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH 2 COH(CH 3 ) 2 , and -CH 2 CH 2 OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH 3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH 3 group, and on the carbon with one -CH 3 group, (f) said triazolyl moiety is substituted on the carbon with a -MH 2 group, or (g) said triazolyl moiety is substituted on the carbon with a -MH 2 group, or (g) said triazo
  • R 5 is a substituted triazolyl-thiazolyl- wherein the triazolyl moiety is substituted with one or two alkyl groups selected from the group consisting of: -CrC 6 alkyl, -Ci-C 4 alkyl, -Ci-C 2 alkyl, and -CH 3 .
  • R 5 is a substituted triazolyl-thiazolyl- wherein the triazolyl moiety is substituted with one or two alkyl groups selected from the group consisting of: -CrC 4 alkyl, -C r C 2 alkyl, and -CH 3 .
  • R 5 is a substituted triazolyl-thiazolyl- wherein the triazolyl moiety is substituted with one or two -CH 3 groups.
  • R 5 is a substituted triazolyl-thiazolyl- wherein the triazolyl moiety is substituted on the nitrogen with an alkyl group selected from the group consisting of: -CrC ⁇ alkyl, -d-C 2 alkyl, and -CH 3 .
  • R 5 is a substituted triazolyl-thiazolyl- wherein the triazolyl moiety is substituted on the nitrogen with -CH 3 .
  • R 5 is a substituted triazolyl-thiazolyl- wherein the triazolyl moiety is substituted on the nitrogen with an -alkylene-O-alkyl group selected from the group consisting of: -C 2 -C 4 alkylene-O-d- C 6 alkyl, -dalkylene-O-Crdalkyl, -C 2 -C 4 alkylene-O-CH 3 , and -CH 2 CH 2 OCH 3 .
  • R 5 is a substituted triazolyl-thiazolyl- wherein the triazolyl moiety is substituted on the nitrogen with an -alkylene-O-alkyl group selected from the group consisting of: -C 2 alkylene-O-d- C 2 alkyl, and -CH 2 CH 2 OCH 3 .
  • R 5 is a substituted triazolyl-thiazolyl- wherein the triazolyl moiety is substituted on the nitrogen with -CH 2 CH 2 OCH 3 .
  • R 5 is a substituted triazolyl-thiazolyl- wherein the triazolyl moiety is substituted on the nitrogen with a hydroxy substituted alkyl group selected from the group consisting of: hydroxy substituted -Ci-C 4 alkyl, hydroxy substituted -CrC 2 alkyl, and hydroxy substituted -CH 3 .
  • R 5 is a substituted triazolyl-thiazolyl- wherein the triazolyl moiety is substituted on the nitrogen with a hydroxy substituted alkyl group selected from the group consisting of: CH 2 COH(CH 3 ) 2 , and -CH 2 CH 2 OH.
  • R 5 is a substituted triazolyl-thiazolyl- group wherein (a) said triazolyl moiety is optionally substituted on the nitrogen with a substituent selected from the group consisting of: -CH 2 COH(CH 3 ) 2 , and -CH 2 CH 2 OH, (b) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is optionally substituted on the nitrogen with a -CH 3 group, (e) said triazolyl moiety is optionally substituted on the nitrogen with one -CH 3 group, and on the carbon with one -CH 3 group, (f) said triazolyl moiety is optionally substituted on the carbon with a -
  • R 5 is a substituted triazolyl-thiazolyl- group wherein (a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH 2 COH(CH 3 ) 2 , and -CH 2 CH 2 OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH 3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH 3 group, and on the carbon with one -CH 3 group, (f) said triazolyl moiety is substituted on the carbon with a -NH 2 group, or (g) said triazoly
  • R 1 and R 2 are as defined in any one of embodiment numbers 1 to 44, Q is:
  • R is selected from the group consisting of:
  • R 2 is a -O-(CrC 2 )alkyl group, and R' is:
  • R 5 is selected from the group consisting of:
  • R 2 is a -S-(Ci -C 2 )alkyl group, and R > ⁇ 1 i :s. :
  • R 5 is selected from the group consisting of:
  • inventions of this invention are directed to any one of the embodiments above (for example, any one of embodiment numbers 1 to 46, or any one of the embodiments following embodiment number 46) wherein one or more hydrogen atoms are deuterium.
  • Representative compounds of this invention include, but are not limited to:
  • Representative compounds of this invention include, but are not limited to:
  • Representative compounds of this invention include, but are not limited to:
  • Representative compounds of this invention wherein hydrogen has been replaced by deuterium include, but are not limited to:
  • Another embodiment of this invention is directed to compound A1. Another embodiment of this invention is directed to compound A2. Another embodiment of this invention is directed to compound A3. Another embodiment of this invention is directed to compound A4. Another embodiment of this invention is directed to compound A5. Another embodiment of this invention is directed to compound A6. Another embodiment of this invention is directed to compound A7. Another embodiment of this invention is directed to compound A8. Another embodiment of this invention is directed to compound A9. Another embodiment of this invention is directed to compound A10. Another embodiment of this invention is directed to compound A11. Another embodiment of this invention is directed to compound A12. Another embodiment of this invention is directed to compound A13. Another embodiment of this invention is directed to compound A14. Another embodiment of this invention is directed to compound A15. Another embodiment of this invention is directed to compound A16. Another embodiment of this invention is directed to compound A18. Another embodiment of this invention is directed to compound A19.
  • Another embodiment of this invention is directed to compound A20.
  • Another embodiment of this invention is directed to compound A21.
  • Another embodiment of this invention is directed to compound A22. Another embodiment of this invention is directed to compound A23.
  • Another embodiment of this invention is directed to compound A24.
  • Another embodiment of this invention is directed to compound A25.
  • Another embodiment of this invention is directed to compound A26.
  • Another embodiment of this invention is directed to compound A27. Another embodiment of this invention is directed to compound A28.
  • Another embodiment of this invention is directed to compound A29.
  • Another embodiment of this invention is directed to compound A30.
  • Another embodiment of this invention is directed to compound A31.
  • Another embodiment of this invention is directed to compound A32. Another embodiment of this invention is directed to compound A33.
  • Another embodiment of this invention is directed to compound A34.
  • Another embodiment of this invention is directed to compound A35.
  • Another embodiment of this invention is directed to compound A36.
  • Another embodiment of this invention is directed to compound A37.
  • Another embodiment of this invention is directed to compound A38.
  • Another embodiment of this invention is directed to compound A39.
  • Another embodiment of this invention is directed to compound A40.
  • Another embodiment of this invention is directed to compound A41.
  • Another embodiment of this invention is directed to compound A42. Another embodiment of this invention is directed to compound A43.
  • Another embodiment of this invention is directed to compound A44.
  • Another embodiment of this invention is directed to compound A45.
  • Another embodiment of this invention is directed to compound A46.
  • Another embodiment of this invention is directed to compound A47. Another embodiment of this invention is directed to compound A48.
  • Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A1.
  • Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A2.
  • Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A3.
  • Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A4. Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A5.
  • Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A6.
  • Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A7.
  • Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A8.
  • Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A9. Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A10.
  • Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A11.
  • Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A12.
  • Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A13.
  • Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A14.
  • Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A15.
  • Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A16.
  • Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A18.
  • Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A19.
  • Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A20. Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A21.
  • Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A22.
  • Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A23.
  • Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A24.
  • Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A25.
  • Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A26.
  • Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A27.
  • Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A28.
  • Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A29.
  • Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A30.
  • Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A31.
  • Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A32.
  • Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A33.
  • Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A34.
  • Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A35.
  • Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A36.
  • Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A37.
  • Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A38.
  • Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A39. Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A40.
  • Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A41.
  • Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A42.
  • Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A43.
  • Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A44. Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A45.
  • Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A46.
  • Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A47.
  • Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A48.
  • Another embodiment of this invention is directed to a solvate of compound A1. Another embodiment of this invention is directed to a solvate of compound A2. Another embodiment of this invention is directed to solvate of compound A3.
  • Another embodiment of this invention is directed to solvate of compound A4. Another embodiment of this invention is directed to a solvate of compound A5. Another embodiment of this invention is directed to a solvate of compound A6. Another embodiment of this invention is directed to a solvate of compound A7. Another embodiment of this invention is directed to a solvate of compound A8.
  • Another embodiment of this invention is directed to a solvate of compound A9. Another embodiment of this invention is directed to a solvate of compound A10.
  • Another embodiment of this invention is directed to solvate of compound A11.
  • Another embodiment of this invention is directed to a solvate of compound A12.
  • Another embodiment of this invention is directed to a solvate of compound A13.
  • Another embodiment of this invention is directed to a solvate of compound
  • Another embodiment of this invention is directed to a solvate of compound A15.
  • Another embodiment of this invention is directed to a solvate of compound A16.
  • Another embodiment of this invention is directed to a solvate of compound A18.
  • Another embodiment of this invention is directed to a solvate of compound A19.
  • Another embodiment of this invention is directed to a solvate of compound
  • Another embodiment of this invention is directed to a solvate of compound A21.
  • Another embodiment of this invention is directed to a solvate of compound A22.
  • Another embodiment of this invention is directed to a solvate of compound A23.
  • Another embodiment of this invention is directed to a solvate of compound A24.
  • Another embodiment of this invention is directed to a solvate of compound
  • Another embodiment of this invention is directed to a solvate of compound A26.
  • Another embodiment of this invention is directed to a solvate of compound A27.
  • Another embodiment of this invention is directed to a solvate of compound A28.
  • Another embodiment of this invention is directed to a solvate of compound A29.
  • Another embodiment of this invention is directed to a solvate of compound A30.
  • Another embodiment of this invention is directed to a solvate of compound A31.
  • Another embodiment of this invention is directed to a solvate of compound
  • Another embodiment of this invention is directed to a solvate of compound A33.
  • Another embodiment of this invention is directed to a solvate of compound A34.
  • Another embodiment of this invention is directed to a solvate of compound A35.
  • Another embodiment of this invention is directed to a solvate of compound A36. Another embodiment of this invention is directed to a solvate of compound
  • Another embodiment of this invention is directed to a solvate of compound A38.
  • Another embodiment of this invention is directed to a solvate of compound A39.
  • Another embodiment of this invention is directed to a solvate of compound A40.
  • Another embodiment of this invention is directed to a solvate of compound A41.
  • Another embodiment of this invention is directed to a solvate of compound
  • Another embodiment of this invention is directed to a solvate of compound A43.
  • Another embodiment of this invention is directed to a solvate of compound A44.
  • Another embodiment of this invention is directed to a solvate of compound A45.
  • Another embodiment of this invention is directed to a solvate of compound A46.
  • Another embodiment of this invention is directed to a solvate of compound A47.
  • Another embodiment of this invention is directed to a solvate of compound A48.
  • Other embodiments of this invention are directed to any one of the embodiments of formula 1.0 wherein the compound is in pure and isolated form.
  • inventions of this invention are directed to any one of the compounds of A31 to A48 in isolated form.
  • Another embodiment of this invention is directed to a pharmaceutical composition comprising an effective amount of at least one compound (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) of formula 1.0 (preferably of formula 1.1) and a pharmaceutically acceptable carrier.
  • Another embodiment of this invention is directed to a pharmaceutical composition comprising an effective amount of a compound of formula 1.0 (preferably of formula 1.1 ) and a pharmaceutically acceptable carrier.
  • Another embodiment of this invention is directed to a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount of at least one compound (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) selected from the group consisting of: A1 to A16 and A18 to A48, and a pharmaceutically acceptable carrier.
  • at least one compound e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 selected from the group consisting of: A1 to A16 and A18 to A48, and a pharmaceutically acceptable carrier.
  • Another embodiment of this invention is directed to a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount of at least one compound (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) selected from the group consisting of: A1 to A16 and A18 to A30, and a pharmaceutically acceptable carrier.
  • at least one compound e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 selected from the group consisting of: A1 to A16 and A18 to A30, and a pharmaceutically acceptable carrier.
  • Another embodiment of this invention is directed to a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount of at least one compound (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) selected from the group consisting of: A1 to A16 and A18 to A26, and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition comprising an effective amount of at least one compound (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) selected from the group consisting of: A31 to A48, and a pharmaceutically acceptable carrier.
  • Another embodiment of this invention is directed to a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount of a compound selected from the group consisting of: A1 to A16 and A18 to A48, and a pharmaceutically acceptable carrier.
  • Another embodiment of this invention is directed to a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount of at least one compound (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) of formula 1.0 (preferably of formula 1.1), at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) other active pharmaceutically active ingredient, and a pharmaceutically acceptable carrier.
  • Another embodiment of this invention is directed to a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount of a compound of formula 1.0 (preferably of formula 1.1), another active pharmaceutically active ingredient, and a pharmaceutically acceptable carrier.
  • Another embodiment of this invention is directed to a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount of at least one compound (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) selected from the group consisting of: A1 to A16 and A18 to A48, at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) other active pharmaceutically active ingredient, and a pharmaceutically acceptable carrier.
  • at least one compound e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 selected from the group consisting of: A1 to A16 and A18 to A48, at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) other active pharmaceutically active ingredient, and a pharmaceutically acceptable carrier.
  • Another embodiment of this invention is directed to a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount of at least one compound (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) selected from the group consisting of: A1 to A16 and A18 to A30, at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) other active pharmaceutically active ingredient, and a pharmaceutically acceptable carrier.
  • at least one compound e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 selected from the group consisting of: A1 to A16 and A18 to A30, at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) other active pharmaceutically active ingredient, and a pharmaceutically acceptable carrier.
  • Another embodiment of this invention is directed to a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount of at least one compound (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) selected from the group consisting of: A1 to A16 and A18 to A26, at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) other active pharmaceutically active ingredient, and a pharmaceutically acceptable carrier.
  • Another embodiment of this invention is directed to a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount of at least one compound (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) selected from the group consisting of: A31 to A48, at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) other active pharmaceutically active ingredient, and a pharmaceutically acceptable carrier.
  • at least one compound e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 selected from the group consisting of: A31 to A48, at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) other active pharmaceutically active ingredient, and a pharmaceutically acceptable carrier.
  • Another embodiment of this invention is directed to a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount of a compound selected from the group consisting of: A1 to A16 and A18 to A48, another active pharmaceutically active ingredient, and a pharmaceutically acceptable carrier.
  • Another embodiment of this invention is directed to a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount of a compound selected from the group consisting of: A1 to A16 and A18 to A30, another active pharmaceutically active ingredient, and a pharmaceutically acceptable carrier.
  • Another embodiment of this invention is directed to a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount of a compound selected from the group consisting of: A1 to A16 and A18 to A26, another active pharmaceutically active ingredient, and a pharmaceutically acceptable carrier.
  • Another embodiment of this invention is directed to a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount of a compound selected from the group consisting of: A31 to A48, another active pharmaceutically active ingredient, and a pharmaceutically acceptable carrier.
  • Another embodiment of this invention is directed to a pharmaceutical composition comprising an effective amount of at least one compound (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) of formula 1.0 (preferably of formula 1.1 ), at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) chemotherapeutic agent, and a pharmaceutically acceptable carrier.
  • Another embodiment of this invention is directed to a pharmaceutical composition comprising an effective amount of a compound of formula 1.0 (preferably of formula 1.1), a chemotherapeutic agent, and a pharmaceutically acceptable carrier.
  • Another embodiment of this invention is directed to a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount of at least one compound (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) selected from the group consisting of: A1 to A16 and A18 to A48, at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) chemotherapeutic agent, and a pharmaceutically acceptable carrier.
  • Another embodiment of this invention is directed to a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount of at least one compound (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) selected from the group consisting of: A1 to A16 and A18 to A30, at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) chemotherapeutic agent, and a pharmaceutically acceptable carrier.
  • Another embodiment of this invention is directed to a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount of at least one compound (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) selected from the group consisting of: A1 to A16 and A18 to A26, at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) chemotherapeutic agent, and a pharmaceutically acceptable carrier.
  • Another embodiment of this invention is directed to a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount of at least one compound (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) selected from the group consisting of: A31 to A48, at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) chemotherapeutic agent, and a pharmaceutically acceptable carrier.
  • Another embodiment of this invention is directed to a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount of a compound selected from the group consisting of: A1 to A16 and A18 to A48, a chemotherapeutic agent, and a pharmaceutically acceptable carrier.
  • Another embodiment of this invention is directed to a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount of a compound selected from the group consisting of: A1 to A16 and A18 to A30, a chemotherapeutic agent, and a pharmaceutically acceptable carrier.
  • Another embodiment of this invention is directed to a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount of a compound selected from the group consisting of: A1 to A16 and A18 to A26, a chemotherapeutic agent, and a pharmaceutically acceptable carrier.
  • Another embodiment of this invention is directed to a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount of a compound selected from the group consisting of: A31 to A48, a chemotherapeutic agent, and a pharmaceutically acceptable carrier.
  • Another embodiment of this invention is directed to a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) compound of formula 1.0 (preferably of formula 1.6).
  • Another embodiment of this invention is directed to a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of one compound of formula 1.0 (preferably of formula 1.6).
  • Another embodiment of this invention is directed to a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (1 , 2 or 3, or 1 or 2, or 1 , and usually 1) compound of A1 to A16 and A18 to A48.
  • Another embodiment of this invention is directed to a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (1 , 2 or 3, or 1 or 2, or 1 , and usually 1) compound of A1 to A16 and A18 to A30.
  • Another embodiment of this invention is directed to a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (1 , 2 or 3, or 1 or 2, or 1 , and usually 1) compound of A1 to A16 and A18 to A26.
  • Another embodiment of this invention is directed to a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) compound of A31 to A48.
  • Another embodiment of this invention is directed to a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of one compound of A1 to A16 and A18 to A48.
  • Another embodiment of this invention is directed to a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of one compound of A1 to A16 and A18 to A30.
  • Another embodiment of this invention is directed to a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of one compound of A1 to A16 and A18 to A26.
  • Another embodiment of this invention is directed to a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of one compound of A31 to A48.
  • Another embodiment of this invention is directed to a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) compound of formula 1.0, and an effective amount of at least one (1 , 2 or 3, or 1 or 2, or 1 , and usually 1) chemotherapeutic agent.
  • Another embodiment of this invention is directed to a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of one compound of formula 1.0, and an effective amount of at least one (1 , 2 or 3, or 1 or 2, or 1 , and usually 1) chemotherapeutic agent.
  • Another embodiment of this invention is directed to a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (1 , 2 or 3, or 1 or 2, or 1 , and usually 1) compound of A1 to A16 and A18 to A48, and an effective amount of at least one (1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) chemotherapeutic agent.
  • Another embodiment of this invention is directed to a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) compound of A1 to A16 and A18 to A30, and an effective amount of at least one (1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) chemotherapeutic agent.
  • Another embodiment of this invention is directed to a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) compound of A1 to A16 and A18 to A26, and an effective amount of at least one (1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) chemotherapeutic agent.
  • Another embodiment of this invention is directed to a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) compound of A31 to A48, and an effective amount of at least one (1 , 2 or 3, or 1 or 2, or 1 , and usually 1) chemotherapeutic agent.
  • Another embodiment of this invention is directed to a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of one compound of A1 to A16 and A18 to A48, and an effective amount of at least one (1 , 2 or 3, or 1 or 2, or 1 , and usually 1) chemotherapeutic agent.
  • Another embodiment of this invention is directed to a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of one compound of A1 to A16 and A18 to A30, and an effective amount of at least one (1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) chemotherapeutic agent.
  • Another embodiment of this invention is directed to a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of one compound of A1 to A16 and A18 to A26, and an effective amount of at least one (1 , 2 or 3, or 1 or 2, or 1 , and usually 1) chemotherapeutic agent.
  • Another embodiment of this invention is directed to a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of one compound of A31 to A48, and an effective amount of at least one (1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) chemotherapeutic agent.
  • this invention further provides a method of inhibiting ERK in mammals, especially humans, by the administration of an effective amount (e.g., a therapeutically effective amount) of one or more (e.g., one) compounds of this invention.
  • an effective amount e.g., a therapeutically effective amount
  • the methods can optionally include the administration of an effective amount of one or more (e.g., 1 , 2 or 3, or 1 or 2, or 1) chemotherapeutic agents.
  • the chemotherapeutic agents can be administered currently or sequentially with the compounds of this invention.
  • the methods of treating cancer described herein include methods wherein a combination of drugs (i.e., compounds, or pharmaceutically active ingredients, or pharmaceutical compositions) are used (i.e., the methods of treating cancer of this invention include combination therapies).
  • drugs i.e., compounds, or pharmaceutically active ingredients, or pharmaceutical compositions
  • the methods of treating cancer of this invention include combination therapies.
  • the drugs are generally administered individually as a pharmaceutical composition.
  • the use of a pharmaceutical composition comprising more than one drug is within the scope of this invention.
  • the methods can optionally include the administration of an effective amount of radiation therapy.
  • an effective amount of radiation therapy for radiation therapy, ⁇ -radiation is preferred.
  • cancers which may be treated by the methods of this invention include, but are not limited to: (A) lung cancer (e.g., lung adenocarcinoma and non small cell lung cancer), (B) pancreatic cancers (e.g., pancreatic carcinoma such as, for example, exocrine pancreatic carcinoma), (C) colon cancers (e.g., colorectal carcinomas, such as, for example, colon adenocarcinoma and colon adenoma), (D) myeloid leukemias (for example, acute myelogenous leukemia (AML), CML, and CMML), (E) thyroid cancer, (F) myelodysplastic syndrome (MDS), (G) bladder carcinoma, (H) epidermal carcinoma, (I) melanoma, (J) breast cancer, (K) prostate cancer, (L) head and neck cancers (e.g., squamous cell cancer of the head and neck), (M) ovarian cancer, (N) brain cancers
  • another embodiment of this invention is directed to a method for treating lung cancer, pancreatic cancer, colon cancer (e.g., colorectal cancer), myeloid leukemias (e.g., AML, CML, and CMML), thyroid cancer, myelodysplastic syndrome (MDS), bladder carcinoma, epidermal carcinoma, melanoma, breast cancer, prostate cancer, head and neck cancers (e.g., squamous cell cancer of the head and neck), ovarian cancer, brain cancers (e.g., gliomas, such as glioma blastoma multiforme), cancers of mesenchymal origin (e.g., fibrosarcomas and rhabdomyosarcomas), sarcomas, tetracarcinomas, nuroblastomas, kidney carcinomas, hepatomas, non- Hodgkin's lymphoma, multiple myeloma, or anaplastic thyroid carcinoma, in a patient in need of
  • Another embodiment of this invention is directed to a method for treating lung cancer, pancreatic cancer, colon cancer (e.g., colorectal cancer), myeloid leukemias (e.g., AML, CML, and CMML), thyroid cancer, myelodysplastic syndrome (MDS), bladder carcinoma, epidermal carcinoma, melanoma, breast cancer, prostate cancer, head and neck cancers (e.g., squamous cell cancer of the head and neck), ovarian cancer, brain cancers (e.g., gliomas, such as glioma blastoma multiforme), cancers of mesenchymal origin (e.g., fibrosarcomas and rhabdomyosarcomas), sarcomas, tetracarcinomas, nuroblastomas, kidney carcinomas, hepatomas, non-Hodgkin's lymphoma, multiple myeloma, or anaplastic thyroid carcinoma in a patient in need of such treatment
  • Another embodiment of this invention is directed to a method for treating lung cancer, pancreatic cancer, colon cancer (e.g., colorectal cancer), myeloid leukemias (e.g., AML, CML, and CMML), thyroid cancer, myelodysplastic syndrome (MDS), bladder carcinoma, epidermal carcinoma, melanoma, breast cancer, prostate cancer, head and neck cancers (e.g., squamous cell cancer of the head and neck), ovarian cancer, brain cancers (e.g., gliomas, such as glioma blastoma multiforme), cancers of mesenchymal origin (e.g., fibrosarcomas and rhabdomyosarcomas), sarcomas, tetracarcinomas, nuroblastomas, kidney carcinomas, hepatomas, non-Hodgkin's lymphoma, multiple myeloma, or anaplastic thyroid carcinoma in a patient in need of such treatment
  • Another embodiment of this invention is directed to a method for treating lung cancer, pancreatic cancer, colon cancer (e.g., colorectal cancer), myeloid leukemias (e.g., AML, CML, and CMML), thyroid cancer, myelodysplastic syndrome (MDS), bladder carcinoma, epidermal carcinoma, melanoma, breast cancer, prostate cancer, head and neck cancers (e.g., squamous cell cancer of the head and neck), ovarian cancer, brain cancers (e.g., gliomas, such as glioma blastoma multiforme), cancers of mesenchymal origin (e.g., fibrosarcomas and rhabdomyosarcomas), sarcomas, tetracarcinomas, nuroblastomas, kidney carcinomas, hepatomas, non-Hodgkin's lymphoma, multiple myeloma, or anaplastic thyroid carcinoma in a patient in need of such treatment
  • Another embodiment of this invention is directed to a method for treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, wherein said cancer is selected from the group consisting of: melanoma, pancreatic cancer, thryroid cancer, colorectal cancer, lung cancer, breast cancer, and ovarian cancer.
  • at least one e.g., 1 , 2 or 3, 1 or 2, and usually 1
  • said cancer is selected from the group consisting of: melanoma, pancreatic cancer, thryroid cancer, colorectal cancer, lung cancer, breast cancer, and ovarian cancer.
  • Another embodiment of this invention is directed to a method for treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1) chemotherapeutic agent wherein said cancer is selected from the group consisting of: melanoma, pancreatic cancer, thryroid cancer, colorectal cancer, lung cancer, breast cancer, and ovarian cancer.
  • Another embodiment of this invention is directed to a method for treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0, wherein said cancer is selected from the group consisting of: melanoma, pancreatic cancer, thryroid cancer, colorectal cancer, lung cancer, breast cancer, and ovarian cancer.
  • a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0, wherein said cancer is selected from the group consisting of: melanoma, pancreatic cancer, thryroid cancer, colorectal cancer, lung cancer, breast cancer, and ovarian cancer.
  • Another embodiment of this invention is directed to a method for treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1) chemotherapeutic agent wherein said cancer is selected from the group consisting of: melanoma, pancreatic cancer, thryroid cancer, colorectal cancer, lung cancer, breast cancer, and ovarian cancer.
  • a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1) chemotherapeutic agent wherein said cancer is selected from the group consisting of: melanoma, pancreatic cancer
  • Another embodiment of this invention is directed to a method for treating melanoma in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0.
  • Another embodiment of this invention is directed to a method for treating melanoma, in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
  • Another embodiment of this invention is directed to a method for treating melanoma in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
  • a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
  • Another embodiment of this invention is directed to a method for treating melanoma in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
  • a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
  • Another embodiment of this invention is directed to a method for treating pancreatic cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
  • at least one e.g., 1 , 2 or 3, 1 or 2, and usually 1
  • Another embodiment of this invention is directed to a method for treating pancreatic cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1) chemotherapeutic agent.
  • an effective amount of at least one e.g., 1 , 2 or 3, 1 or 2, and usually 1
  • chemotherapeutic agent e.g., 1 , 2 or 3, 1 or 2, or 1
  • Another embodiment of this invention is directed to a method for treating pancreatic cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0.
  • a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0.
  • Another embodiment of this invention is directed to a method for treating pancreatic cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
  • a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
  • Another embodiment of this invention is directed to a method for treating thyroid cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
  • at least one e.g., 1 , 2 or 3, 1 or 2, and usually 1
  • Another embodiment of this invention is directed to a method for treating thyroid cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1) chemotherapeutic agent.
  • an effective amount of at least one e.g., 1 , 2 or 3, 1 or 2, and usually 1 compound of formula 1.0
  • Another embodiment of this invention is directed to a method for treating thyroid cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
  • a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
  • Another embodiment of this invention is directed to a method for treating thyroid cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1) chemotherapeutic agent.
  • a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1) chemotherapeutic agent.
  • Another embodiment of this invention is directed to a method for treating colorectal cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0.
  • at least one e.g., 1 , 2 or 3, 1 or 2, and usually 1
  • Another embodiment of this invention is directed to a method for treating colorectal cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1) chemotherapeutic agent.
  • an effective amount of at least one e.g., 1 , 2 or 3, 1 or 2, and usually 1
  • chemotherapeutic agent e.g., 1 , 2 or 3, 1 or 2, or 1
  • Another embodiment of this invention is directed to a method for treating colorectal cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0.
  • a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0.
  • Another embodiment of this invention is directed to a method for treating colorectal cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
  • Another embodiment of this invention is directed to a method for treating lung cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
  • Another embodiment of this invention is directed to a method for treating lung cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
  • Another embodiment of this invention is directed to a method for treating lung cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
  • Another embodiment of this invention is directed to a method for treating lung cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
  • a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
  • Another embodiment of this invention is directed to a method for treating breast cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
  • Another embodiment of this invention is directed to a method for treating breast cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
  • Another embodiment of this invention is directed to a method for treating breast cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
  • Another embodiment of this invention is directed to a method for treating breast cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
  • Another embodiment of this invention is directed to a method for treating ovarian cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
  • at least one e.g., 1 , 2 or 3, 1 or 2, and usually 1
  • This invention also provides a method for treating ovarian cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
  • an effective amount of at least one e.g., 1 , 2 or 3, 1 or 2, and usually
  • Another embodiment of this invention is directed to a method for treating ovarian cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0.
  • a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0.
  • Another embodiment of this invention is directed to a method for treating ovarian cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
  • a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
  • inventions of this invention are directed to methods of treating breast cancer (i.e., post-menopausal and premenopausal breast cancer, e.g., hormone- dependent breast cancer) in a patient in need of such treatment, said treatment comprising the administration of an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0 in combination with hormonal therapies (i.e., antihormonal agents).
  • breast cancer i.e., post-menopausal and premenopausal breast cancer, e.g., hormone- dependent breast cancer
  • said treatment comprising the administration of an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0 in combination with hormonal therapies (i.e., antihormonal agents).
  • inventions of this invention are directed to methods of treating breast cancer (i.e., post-menopausal and premenopausal breast cancer, e.g., hormone- dependent breast cancer) in a patient in need of such treatment, said treatment comprising the administration of an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0 in combination with hormonal therapies (i.e., antihormonal agents).
  • breast cancer i.e., post-menopausal and premenopausal breast cancer, e.g., hormone- dependent breast cancer
  • a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0 in combination with hormonal therapies (i.e., antihormonal agents).
  • inventions of this invention are directed to methods of treating breast cancer (i.e., post-menopausal and premenopausal breast cancer, e.g., hormone- dependent breast cancer) in a patient in need of such treatment, said treatment comprising the administration of an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0 in combination with hormonal therapies (i.e., antihormonal agents), and in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1) chemotherapeutic agent.
  • breast cancer i.e., post-menopausal and premenopausal breast cancer, e.g., hormone- dependent breast cancer
  • said treatment comprising the administration of an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0 in combination with hormonal therapies (i.e., antihormonal agents), and in combination with an effective amount of at least one (e.g
  • inventions of this invention are directed to methods of treating breast cancer (i.e., post-menopausal and premenopausal breast cancer, e.g., hormone- dependent breast cancer) in a patient in need of such treatment, said treatment comprising the administration of an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0 in combination with hormonal therapies (i.e., antihormonal agents), and in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
  • a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0 in combination with hormonal therapies (i.e., antihormonal agents), and in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
  • the methods of treating breast cancer described herein include the treatment of hormone-dependent metastatic and advanced breast cancer, adjuvant therapy for hormone-dependent primary and early breast cancer, the treatment of ductal carcinoma in situ, and the treatment of inflammatory breast cancer in situ.
  • the methods of treating hormone-dependent breast cancer can also be used to prevent breast cancer in patients having a high risk of developing breast cancer.
  • breast cancer i.e., post-menopausal and premenopausal breast cancer, e.g., hormone-dependent breast cancer
  • said treatment comprising the administration of an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0 in combination with hormonal therapies (i.e., antihormonal agents).
  • inventions of this invention are directed to methods of preventing breast cancer (i.e., post-menopausal and premenopausal breast cancer, e.g., hormone-dependent breast cancer) in a patient in need of such treatment, said treatment comprising the administration of an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0 in combination with hormonal therapies (i.e., antihormonal agents).
  • breast cancer i.e., post-menopausal and premenopausal breast cancer, e.g., hormone-dependent breast cancer
  • a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0 in combination with hormonal therapies (i.e., antihormonal agents).
  • breast cancer i.e., post-menopausal and premenopausal breast cancer, e.g., hormone-dependent breast cancer
  • said treatment comprising the administration of an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0 in combination with hormonal therapies (i.e., antihormonal agents), and in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1) chemotherapeutic agent.
  • a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0 in combination with hormonal therapies (i.e., antihormonal agents), and in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1) chemotherapeutic agent.
  • Another embodiment of this invention is directed to a method for treating brain cancer (e.g., glioma, such as glioma blastoma multiforme) in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0.
  • brain cancer e.g., glioma, such as glioma blastoma multiforme
  • Another embodiment of this invention is directed to a method for treating brain cancer (e.g., glioma, such as glioma blastoma multiforme) in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1) chemotherapeutic agent.
  • brain cancer e.g., glioma, such as glioma blastoma multiforme
  • Another embodiment of this invention is directed to a method for treating brain cancer (e.g., glioma, such as glioma blastoma multiforme) a in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
  • brain cancer e.g., glioma, such as glioma blastoma multiforme
  • a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
  • Another embodiment of this invention is directed to a method for treating brain cancer (e.g., glioma, such as glioma blastoma multiforme) in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1) chemotherapeutic agent.
  • a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1) chemotherapeutic agent.
  • Another embodiment of this invention is directed to a method for treating brain cancer (e.g., glioma, such as glioma blastoma multiforme) in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of a chemotherapeutic agent wherein said chemotherapeutic agent is temozolomide.
  • brain cancer e.g., glioma, such as glioma blastoma multiforme
  • Another embodiment of this invention is directed to a method for treating brain cancer (e.g., glioma, such as glioma blastoma multiforme) in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of a chemotherapeutic agent, wherein said chemotherapeutic agent is temozolomide.
  • a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of a chemotherapeutic agent, wherein said chemotherapeutic agent is temozolomide.
  • Another embodiment of this invention is directed to a method for treating prostate cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
  • at least one e.g., 1 , 2 or 3, 1 or 2, and usually 1
  • Another embodiment of this invention is directed to a method for treating prostate cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
  • an effective amount of at least one e.g., 1 , 2 or 3, 1 or 2, and usually 1
  • compound of formula 1.0 in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
  • Another embodiment of this invention is directed to a method for treating prostate cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
  • a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
  • Another embodiment of this invention is directed to a method for treating prostate cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1) chemotherapeutic agent.
  • a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1) chemotherapeutic agent.
  • Another embodiment of this invention is directed to a method for treating myelodysplastic syndrome in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
  • at least one e.g., 1 , 2 or 3, 1 or 2, and usually 1
  • Another embodiment of this invention is directed to a method for treating myelodysplastic syndrome in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
  • an effective amount of at least one e.g., 1 , 2 or 3, 1 or 2, and usually 1
  • chemotherapeutic agent e.g., 1 , 2 or 3, 1 or 2, or 1
  • Another embodiment of this invention is directed to a method for treating myelodysplastic syndrome in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
  • a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
  • Another embodiment of this invention is directed to a method for treating myelodysplastic syndrome in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1 , in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
  • a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1 , in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
  • Another embodiment of this invention is directed to a method for treating myeloid leukemias in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
  • at least one e.g., 1 , 2 or 3, 1 or 2, and usually 1
  • Another embodiment of this invention is directed to a method for treating myeloid leukemias in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
  • Another embodiment of this invention is directed to a method for treating myeloid leukemias in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0.
  • a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0.
  • Another embodiment of this invention is directed to a method for treating myeloid leukemias in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
  • a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
  • Another embodiment of this invention is directed to a method for treating acute myelogenous leukemia (AML) in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
  • AML acute myelogenous leukemia
  • Another embodiment of this invention is directed to a method for treating acute myelogenous leukemia (AML) in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
  • AML acute myelogenous leukemia
  • Another embodiment of this invention is directed to a method for treating acute myelogenous leukemia (AML)in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
  • AML acute myelogenous leukemia
  • Another embodiment of this invention is directed to a method for treating acute myelogenous leukemia (AML)in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1) chemotherapeutic agent.
  • AML acute myelogenous leukemia
  • Another embodiment of this invention is directed to a method for treating chronic myelomonocytic leukemia (CMML) in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0.
  • CMML chronic myelomonocytic leukemia
  • Another embodiment of this invention is directed to a method for treating chronic myelomonocytic leukemia (CMML) in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1) chemotherapeutic agent.
  • CMML chronic myelomonocytic leukemia
  • Another embodiment of this invention is directed to a method for treating chronic myelomonocytic leukemia (CMML) in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0.
  • CMML chronic myelomonocytic leukemia
  • Another embodiment of this invention is directed to a method for treating chronic myelomonocytic leukemia (CMML) in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1) chemotherapeutic agent.
  • CMML chronic myelomonocytic leukemia
  • Another embodiment of this invention is directed to a method for treating chronic myelogenous leukemia (chronic myeloid leukemia, CML) in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
  • CML chronic myelogenous leukemia
  • Another embodiment of this invention is directed to a method for treating chronic myelogenous leukemia (chronic myeloid leukemia, CML) in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1) chemotherapeutic agent.
  • CML chronic myelogenous leukemia
  • Another embodiment of this invention is directed to a method for treating chronic myelogenous leukemia (chronic myeloid leukemia, CML) in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0.
  • CML chronic myelogenous leukemia
  • Another embodiment of this invention is directed to a method for treating chronic myelogenous leukemia (chronic myeloid leukemia, CML) in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1) chemotherapeutic agent.
  • CML chronic myelogenous leukemia
  • Another embodiment of this invention is directed to a method for treating myeloid leukemias in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
  • at least one e.g., 1 , 2 or 3, 1 or 2, and usually 1
  • Another embodiment of this invention is directed to a method for treating myeloid leukemias in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
  • an effective amount of at least one e.g., 1 , 2 or 3, 1 or 2, and usually 1
  • chemotherapeutic agent e.g., chemotherapeutic agent
  • Another embodiment of this invention is directed to a method for treating myeloid leukemias in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
  • a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
  • Another embodiment of this invention is directed to a method for treating myeloid leukemias in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
  • Another embodiment of this invention is directed to a method for treating bladder cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0.
  • Another embodiment of this invention is directed to a method for treating bladder cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1) chemotherapeutic agent.
  • an effective amount of at least one e.g., 1 , 2 or 3, 1 or 2, and usually 1
  • chemotherapeutic agent e.g., 1 , 2 or 3, 1 or 2, or 1
  • Another embodiment of this invention is directed to a method for treating bladder cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
  • a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
  • Another embodiment of this invention is directed to a method for treating bladder cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
  • a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
  • Another embodiment of this invention is directed to a method for treating non- Hodgkin's lymphoma in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
  • at least one e.g., 1 , 2 or 3, 1 or 2, and usually 1
  • Another embodiment of this invention is directed to a method for treating non- Hodgkin's lymphoma in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
  • Another embodiment of this invention is directed to a method for treating non- Hodgkin's lymphoma in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
  • a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
  • Another embodiment of this invention is directed to a method for treating non- Hodgkin's lymphoma in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
  • a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
  • Another embodiment of this invention is directed to a method for treating multiple myeloma in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
  • at least one e.g., 1 , 2 or 3, 1 or 2, and usually 1
  • Another embodiment of this invention is directed to a method for treating multiple myeloma in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
  • an effective amount of at least one e.g., 1 , 2 or 3, 1 or 2, and usually 1
  • chemotherapeutic agent e.g., 1 , 2 or 3, 1 or 2, or 1
  • Another embodiment of this invention is directed to a method for treating multiple myeloma in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0.
  • a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0.
  • Another embodiment of this invention is directed to a method for treating multiple myeloma in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
  • a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
  • Chemotherapeutic agents include but are not limited to: microtubule affecting agents, alkylating agents, antimetabolites, natural products and their derivatives, hormones and steroids (including synthetic analogs), and synthetics.
  • alkylating agents include nitrogen mustards, ethylenimine derivatives, alkyl sulfonates, nitrosoureas and triazenes
  • alkylating agents include: Uracil mustard, Chlormethine, Cyclophosphamide (Cytoxan ® ), Ifosfamide, Melphalan, Chlorambucil, Pipobroman, Triethylene-melamine, Triethylenethiophosphoramine, Busulfan, Carmustine, Lomustine, Streptozocin, dacarbazine, and Temozolomide.
  • antimetabolites include folic acid antagonists, pyrimidine analogs, purine analogs and adenosine deaminase inhibitors
  • examples of antimetabolites include: Methotrexate, 5-Fluorouracil, Floxuridine, Cytarabine, 6-Mercaptopurine, 6-Thioguanine, Fludarabine phosphate, Pentostatine, and Gemcitabine.
  • Examples of natural products and their derivatives include: Vinblastine, Vincristine, Vindesine, Bleomycin, Dactinomycin, Daunorubicin, Doxorubicin, Epirubicin, Idarubicin, Paclitaxel (paclitaxel is a microtubule affecting agent and is commercially available as Taxol ® ), Paclitaxel derivatives (e.g. taxotere), Mithramycin, Deoxyco-formycin, Mitomycin-C, L-Asparaginase, Interferons (especially IFN-a), Etoposide, and Teniposide.
  • Vinblastine Vincristine
  • Vindesine Bleomycin
  • Dactinomycin Daunorubicin
  • Doxorubicin Doxorubicin
  • Epirubicin Idarubicin
  • Paclitaxel Paclitaxel is a microtubule affecting agent and is commercially available as Taxol ®
  • Paclitaxel derivatives e.g
  • hormones and steroids include: 17 ⁇ - Ethinylestradiol, Diethylstilbestrol, Testosterone, Prednisone, Fluoxymesterone, Dromostanolone propionate, Testolactone, Megestrolacetate, Tamoxifen, Methylprednisolone, Methyl-testosterone, Prednisolone, Triamcinolone, Chlorotrianisene, Hydroxyprogesterone, Aminoglutethimide, Estramustine, Medroxyprogesteroneacetate, Leuprolide, Flutamide, Toremifene, and Zoladex.
  • synthetics including inorganic complexes such as platinum coordination complexes: Cisplatin, Carboplatin, Hydroxyurea, Amsacrine,
  • chemotherapeutics examples include: Navelbene, CPT-11 , Anastrazole, Letrazole, Capecitabinbe, Reloxafine, and Droloxafine.
  • a microtubule affecting agent e.g., paclitaxel, a paclitaxel derivative or a paclitaxel-like compound
  • paclitaxel e.g., paclitaxel, a paclitaxel derivative or a paclitaxel-like compound
  • agents can be, for instance, microtubule stabilizing agents or agents which disrupt microtubule formation.
  • Microtubule affecting agents useful in the methods of this invention, are well known to those skilled in the art and include, but are not limited to: Allocolchicine (NSC 406042), Halichondrin B (NSC 609395), Colchicine (NSC 757), Colchicine derivatives (e.g., NSC 33410), Dolastatin 10 (NSC 376128), Maytansine (NSC 153858), Rhizoxin (NSC 332598), Paclitaxel (Taxol ® , NSC 125973), Paclitaxel derivatives (e.g., Taxotere, NSC 608832), Thiocolchicine (NSC 361792), Trityl Cysteine (NSC 83265), Vinblastine Sulfate (NSC 49842), Vincristine Sulfate (NSC 67574), Epothilone A, Epothilone, Discodermolide (see Service, (1996) Science, 274:2009), Estramustine, Nocodazole
  • Chemotherapeutic agents with paclitaxel-like activity include, but are not limited to, paclitaxel and paclitaxel derivatives (paclitaxel-like compounds) and analogues.
  • Paclitaxel and its derivatives e.g. Taxol and Taxotere
  • methods of making paclitaxel and paclitaxel derivatives and analogues are well known to those of skill in the art (see, e.g., U.S.
  • paclitaxel refers to the drug commercially available as Taxol ® (NSC number: 125973). Taxol ® inhibits eukaryotic cell replication by enhancing polymerization of tubulin moieties into stabilized microtubule bundles that are unable to reorganize into the proper structures for mitosis.
  • Taxol ® inhibits eukaryotic cell replication by enhancing polymerization of tubulin moieties into stabilized microtubule bundles that are unable to reorganize into the proper structures for mitosis.
  • chemotherapeutic drugs paclitaxel has generated interest because of its efficacy in clinical trials against drug-refractory tumors, including ovarian and mammary gland tumors (Hawkins (1992) Oncology, 6: 17-23, Horwitz (1992) Trends Pharmacol. Sci. 13: 134-146, Rowinsky (1990) J. Natl. Cane.
  • microtubule affecting agents can be assessed using one of many such assays known in the art, e.g., a semiautomated assay which measures the tubulin-polymerizing activity of paclitaxel analogs in combination with a cellular assay to measure the potential of these compounds to block cells in mitosis (see Lopes (1997) Cancer Chemother. Pharmacol. 41 :37-47).
  • activity of a test compound is determined by contacting a cell with that compound and determining whether or not the cell cycle is disrupted, in particular, through the inhibition of a mitotic event. Such inhibition may be mediated by disruption of the mitotic apparatus, e.g., disruption of normal spindle formation.
  • Cells in which mitosis is interrupted may be characterized by altered morphology (e.g., microtubule compaction, increased chromosome number, etc.).
  • Compounds with possible tubulin polymerization activity can be screened in vitro.
  • the compounds are screened against cultured WR21 cells (derived from line 69-2 wap-ras mice) for inhibition of proliferation and/or for altered cellular morphology, in particular for microtubule compaction.
  • In vivo screening of positive-testing compounds can then be performed using nude mice bearing the WR21 tumor cells. Detailed protocols for this screening method are described by Porter (1995) Lab. Anim. ScL, 45(2): 145-150.
  • chemotherapeutic agents include those selected from the group consisting of: microtubule affecting agents, alkylating agents, antimetabolites, natural products and their derivatives, hormones and steroids (including synthetic analogs), and synthetics.
  • examples of said chemotherapeutic agents also include: (1) taxanes, (2) platinum coordinator compounds, (3) epidermal growth factor (EGF) inhibitors that are antibodies, (4) EGF inhibitors that are small molecules, (5) vascular endolithial growth factor (VEGF) inhibitors that are antibodies, (6) VEGF kinase inhibitors that are small molecules, (7) estrogen receptor antagonists or selective estrogen receptor modulators (SERMs), (8) anti-tumor nucleoside derivatives, (9) epothilones, (10) topoisomerase inhibitors, (11 ) vinca alkaloids, (12) antibodies that are inhibitors of ⁇ V ⁇ 3 integrins, (13) folate antagonists, (14) ribonucleotide reductase inhibitors, (15) anthracyclines, (16) biologies; (17) inhibitors of angiogenesis and/or suppressors of tumor necrosis factor alpha (TNF-alpha)
  • chemotherapeutic agents include:
  • taxanes such as paclitaxel (TAXOL ® ) and/or docetaxel (Taxotere ® );
  • platinum coordinator compounds such as, for example, carboplatin, cisplatin and oxaliplatin (e.g. Eloxatin);
  • EGF inhibitors that are antibodies, such as: HER2 antibodies (such as, for example trastuzumab (Herceptin ® ), Genentech, Inc.), Cetuximab (Erbitux, IMC-C225, ImClone Systems), EMD 72000 (Merck KGaA), anti-EFGR monoclonal antibody ABX (Abgenix), TheraCIM-h-R3 (Center of Molecular Immunology), monoclonal antibody 425 (Merck KGaA), monoclonal antibody ICR-62 (ICR, Sutton, England); Herzyme (Elan Pharmaceutical Technologies and Ribozyme Pharmaceuticals), PKI 166 (Novartis), EKB 569 (Wyeth-Ayerst), GW 572016 (GlaxoSmithKline), Cl 1033 (Pfizer Global Research and Development), trastuzmab-maytansinoid conjugate (Genentech, Inc.), mitumomab (Imclone Systems and Merck KG
  • EGF inhibitors that are small molecules, such as, Tarceva (TM) (OSI-774, OSI Pharmaceuticals, Inc.), and lressa (ZD 1839, Astra Zeneca); (5) VEGF inhibitors that are antibodies such as: bevacizumab (Genentech,
  • IMC-1C11 ImClone Systems
  • DC 101 a KDR VEGF Receptor 2 from ImClone Systems
  • VEGF kinase inhibitors that are small molecules such as SU 5416 (from Sugen, Inc), SU 6688 (from Sugen, Inc.), Bay 43-9006 (a dual VEGF and bRAF inhibitor from Bayer Pharmaceuticals and Onyx Pharmaceuticals);
  • estrogen receptor antagonists or selective estrogen receptor modulators such as tamoxifen, idoxifene, raloxifene, trans-2,3-dihydroraloxifene, levormeloxifene, droloxifene, MDL 103,323, and acolbifene (Schering Corp.);
  • anti-tumor nucleoside derivatives such as 5-fluorouracil, gemcitabine, capecitabine, cytarabine (Ara-C), fludarabine (F-Ara-A), decitabine, and chlorodeoxyadenosine (Cda, 2-Cda);
  • epothilones such as BMS-247550 (Bristol-Myers Squibb), and EPO906 (Novartis Pharmaceuticals);
  • topoisomerase inhibitors such as topotecan (Glaxo SmithKline), and Camptosar (Pharmacia);
  • vinca alkaloids such as, navelbine (Anvar and Fabre, France), vincristine and vinblastine
  • antibodies that are inhibitors of ⁇ V ⁇ 3 integrins such as, LM-609 (see, Clinical Cancer Research, Vol. 6, page 3056-3061 , August 2000, the disclosure of which is incorporated herein by reference thereto);
  • folate antagonists such as Methotrexate (MTX), and Premetrexed (Alimta);
  • ribonucleotide reductase inhibitors such as Hydroxyurea (HU);
  • anthracyclines such as Daunorubicin, Doxorubicin (Adriamycin), and Idarubicin;
  • biologies such as interferon (e.g., Intron-A and Roferon), pegylated interferon (e.g., Peg-lntron and Pegasys), and Rituximab (Rituxan, antibody used for the treatment of non-Hodgkin's lymphoma);
  • interferon e.g., Intron-A and Roferon
  • pegylated interferon e.g., Peg-lntron and Pegasys
  • Rituximab Rituxan, antibody used for the treatment of non-Hodgkin's lymphoma
  • Bcr/abl kinase inhibitors such as, for example Gleevec (STI-571 ), AMN- 17, ONO12380, SU11248 (Sunitinib) and BMS-354825 (19) MEK1 and/or MEK2 inhibitors, such as PD0325901 and Arry-142886
  • IGF-1 and IGF-2 inhibitors that are small molecules, such as, for example, NVP-AEW541 ;
  • (22) small molecule inhibitors of cell cycle dependent kinases such as CDK1 , CDK2, CDK4 and CDK6, such as, for example, CYC202, BMS387032, and Flavopiridol;
  • (23) alkylating agents such as, for example, Temodar® brand of temozolomide
  • chemotherapeutic agents include signal transduction inhibitors.
  • Typical signal transduction inhibitors that are chemotherapeutic agents, include but are not limited to: (i) Bcr/abl kinase inhibitors such as, for example, STI 571 (Gleevec), (ii) Epidermal growth factor (EGF) receptor inhibitor such as, for example, Kinase inhibitors (Iressa, OSI-774) and antibodies (Imclone: C225
  • HER-2/neu receptor inhibitors such as, for example, Herceptin® (trastuzumab).
  • the compound of formula 1.0 e.g., a pharmaceutical composition comprising the compound of formula 1.0
  • a pharmaceutical composition comprising more than one drug is within the scope of this invention.
  • the compound of formula 1.0 and the chemotherapeutic agents are administered in therapeutically effective dosages to obtain clinically acceptable results, e.g., reduction or elimination of symptoms or of the tumor.
  • the compound of formula 1.0 and chemotherapeutic agents can be administered concurrently or consecutively in a treatment protocol.
  • the administration of the chemotherapeutic agents can be made according to treatment protocols already known in the art. In general when more than one chemotherapeutic agent is used in the methods of this invention, the chemotherapeutic agents are administered on the same day either concurrently or consecutively in their standard dosage form.
  • the chemotherapeutic agents are usually administered intravenously, preferably by an IV drip using IV solutions well known in the art (e.g., isotonic saline (0.9% NaCI) or dextrose solution (e.g., 5% dextrose)).
  • IV solutions well known in the art (e.g., isotonic saline (0.9% NaCI) or dextrose solution (e.g., 5% dextrose)).
  • the chemotherapeutic agents are generally administered on the same day; however, those skilled in the art will appreciate that the chemotherapeutic agents can be administered on different days and in different weeks.
  • the skilled clinician can administer the chemotherapeutic agents according to their recommended dosage schedule from the manufacturer of the agent and can adjust the schedule according to the needs of the patient, e.g., based on the patient's response to the treatment.
  • gemcitabine is used in combination with a platinum coordinator compound, such as, for example, cisplatin, to treat lung cancer
  • a platinum coordinator compound such as, for example, cisplatin
  • the compounds of this invention and chemotherapeutic agents can be administered in a treatment protocol that usually lasts one to seven weeks, and is repeated typically from 6 to 12 times. Generally the treatment protocol can last one to four weeks. Treatment protocols of one to three weeks can also be used. A treatment protocol of one to two weeks can also be used. During this treatment protocol or cycle the compounds of this invention can be administered daily while the chemotherapeutic agents can be administered one or more times a week. Generally, a compound of this invention can be administered daily (i.e., once per day), and in one embodiment twice per day, and the chemotherapeutic agent is administered once a week or once every three weeks.
  • the taxanes e.g., Paclitaxel (e.g., Taxol ® ) or Docetaxel (e.g., Taxotere ® )
  • Paclitaxel e.g., Taxol ®
  • Docetaxel e.g., Taxotere ®
  • treatment protocols can be varied according to the needs of the patient.
  • the combination of compounds (drugs) used in the methods of this invention can be administered in variations of the protocols described above.
  • the compounds of this invention can be administered discontinuously rather than continuously during the treatment cycle.
  • the compounds of this invention can be administered daily for a week and then discontinued for a week, with this administration repeating during the treatment cycle.
  • the compounds of this invention can be administered daily for two weeks and discontinued for a week, with this administration repeating during the treatment cycle.
  • the compounds of this invention can be administered daily for one or more weeks during the cycle and discontinued for one or more weeks during the cycle, with this pattern of administration repeating during the treatment cycle.
  • This discontinuous treatment can also be based upon numbers of days rather than a full week. For example, daily dosing for 1 to 6 days, no dosing for 1 to 6 days, with this pattern repeating during the treatment protocol.
  • the number of days (or weeks) wherein the compounds of this invention are not dosed do not have to equal the number of days (or weeks) wherein the compounds of this invention are dosed.
  • the number of days or weeks that the compounds of this invention are dosed is at least equal or greater than the number of days or weeks that the compounds of this invention are not dosed.
  • the chemotherapeutic agent could be given by bolus or continuous infusion.
  • the chemotherapeutic agent could be given daily to once every week, or once every two weeks, or once every three weeks, or once every four weeks during the treatment cycle. If administered daily during a treatment cycle, this daily dosing can be discontinuous over the number of weeks of the treatment cycle. For example, dosed for a week (or a number of days), no dosing for a week (or a number of days, with the pattern repeating during the treatment cycle.
  • the compounds of this invention can be administered orally, preferably as a solid dosage form, and in one embodiment as a capsule, and while the total therapeutically effective daily dose can be administered in one to four, or one to two divided doses per day, generally, the therapeutically effective dose is given once or twice a day, and in one embodiment twice a day.
  • the compounds of this invention can be administered in an amount of about 50 to about 400 mg once per day, and can be administered in an amount of about 50 to about 300 mg once per day.
  • the compounds of this invention are generally administered in an amount of about 50 to about 350 mg twice a day, usually 50 mg to about 200 mg twice a day, and in one embodiment about 75 mg to about 125 mg administered twice a day, and in another embodiment about 100 mg administered twice a day.
  • the therapy cycle can be repeated according to the judgment of the skilled clinician.
  • the patient can be continued on the compounds of this invention at the same dose that was administered in the treatment protocol, or, if the dose was less than 200mg twice a day, the dose can be raised to 200 mg twice a day.
  • This maintenance dose can be continued until the patient progresses or can no longer tolerate the dose (in which case the dose can be reduced and the patient can be continued on the reduced dose).
  • chemotherapeutic agents used with the compounds of this invention, are administered in their normally prescribed dosages during the treatment cycle (i.e., the chemotherapeutic agents are administered according to the standard of practice for the administration of these drugs).
  • the chemotherapeutic agents are administered according to the standard of practice for the administration of these drugs.
  • the chemotherapeutic agents are administered according to the standard of practice for the administration of these drugs.
  • the chemotherapeutic agents are administered according to the standard of practice for the administration of these drugs.
  • the chemotherapeutic agents are administered according to the standard of practice for the administration of these drugs.
  • the chemotherapeutic agents are administered according to the standard of practice for the administration of these drugs.
  • the chemotherapeutic agents are administered according to the standard of practice for the administration of these drugs.
  • the chemotherapeutic agents are administered according to the standard of practice for the administration of these drugs.
  • the chemotherapeutic agents are administered according to the standard of practice for the administration of these drugs.
  • Thalidomide and related imids can be used orally in amounts of about 200 to about 800 mg/day, and can be contiuously dosed or used until releapse or toxicity. See for example Mitsiades et al., "Apoptotic signaling induced by immunomodulatory thalidomide analoqs in human multiple myeloma cells; therapeutic implications", Blood, 99(12):4525-30, June 15, 2002, the disclosure of which is incorporated herein by reference thereto.
  • the FPT inhibitor Sarasar® (brand of Ionifarnib) can be administered orally (e.g., capsule) in amounts of about 50 to about 200 mg given twice a day, or in amounts of about 75 to about 125 mg given twice a day, or in amounts of about 100 to about 200 mg given twice a day, or in an amount of about 100 mg given twice a day.
  • Paclitaxel e.g., Taxol ®
  • Paclitaxel can be administered once per week in an amount of about 50 to about 100 mg/m 2 and in another example about 60 to about 80 mg/m 2 .
  • Paclitaxel e.g., Taxol ®
  • Paclitaxel can be administered once every three weeks in an amount of about 150 to about 250 mg/m 2 and in another example about 175 to about 225 mg/m 2 .
  • Docetaxel e.g., Taxotere ®
  • Docetaxel can be administered once per week in an amount of about 10 to about 45 mg/m 2 .
  • Docetaxel e.g., Taxotere ®
  • Docetaxel can be administered once every three weeks in an amount of about 50 to about 100 mg/m 2 .
  • Cisplatin can be administered once per week in an amount of about 20 to about 40 mg/m 2 . In another example Cisplatin can be administered once every three weeks in an amount of about 60 to about 100 mg/m 2 . In another example Carboplatin can be administered once per week in an amount to provide an AUC of about 2 to about 3. In another example Carboplatin can be administered once every three weeks in an amount to provide an AUC of about 5 to about 8.
  • inventions of this invention are directed to any one of the method of treating cancer embodiments wherein the compounds of formula 1.0 and the chemotherapeutic agents are administered as a pharmaceutical composition comprising an effective amount of the compounds of formula 1.0, an effective amount of the chemotherapeutic agents, and a pharmaceutically acceptable carrier.
  • chemotherapeutic agent selected from the group consisting of: paclitaxel, docetaxel, carboplatin, cisplatin, gemcitabine, tamoxifen, Herceptin, Cetuximab, Tarceva, Iressa, bevacizumab, navelbine, IMC-1 C11 , SU5416 and SU6688.
  • chemotherapeutic agent is used wherein the the chemotherapeutic agent is selected from the group consisting of: paclitaxel, docetaxel, carboplatin, cisplatin, navelbine, gemcitabine, and Herceptin.
  • chemotherapeutic agent is selected from the group consisting of: Cyclophasphamide, 5-Fluorouracil, Temozolomide, Vincristine, Cisplatin, Carboplatin, and Gemcitabine.
  • chemotherapeutic agent selected from the group consisting of: Gemcitabine, Cisplatin and Carboplatin.
  • This invention also provides a method of treating cancer in a patient in need of such treatment, said treatment comprising administering to said patient a therapeutically effective amount at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) compound of formula 1.0, and therapeutically effective amounts of at least one (e.g., 1 , 2 or 3, or 1 or 2, or 2, or 1 ) chemotherapeutic agent selected from the group consisting of: (1) taxanes, (2) platinum coordinator compounds, (3) epidermal growth factor (EGF) inhibitors that are antibodies, (4) EGF inhibitors that are small molecules, (5) vascular endolithial growth factor (VEGF) inhibitors that are antibodies, (6) VEGF kinase inhibitors that are small molecules, (7) estrogen receptor antagonists or selective estrogen receptor modulators (SERMs), (8) anti-tumor nucleoside derivatives, (9) epothilones, (10) topoisomerase inhibitors, (11 ) vinca alkaloids, (12) antibodies that are
  • This invention also provides a method of treating cancer in a patient in need of such treatment, said treatment comprising administering to said patient a therapeutically effective amount at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) compound of formula 1.0, and therapeutically effective amounts of at least two (e.g., 2 or 3, or 2, and usually 2) different antineoplastic agents selected from the group consisting of: (1) taxanes, (2) platinum coordinator compounds, (3) epidermal growth factor (EGF) inhibitors that are antibodies, (4) EGF inhibitors that are small molecules, (5) vascular endolithial growth factor (VEGF) inhibitors that are antibodies, (6) VEGF kinase inhibitors that are small molecules, (7) estrogen receptor antagonists or selective estrogen receptor modulators (SERMs), (8) anti-tumor nucleoside derivatives, (9) epothilones, (10) topoisomerase inhibitors, (11) vinca alkaloids, (12) antibodies that are inhibitors of ⁇ V ⁇ 3 integrins
  • This invention also provides a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient therapeutically effective amounts at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) compound of formula 1.0, and an antineoplastic agent selected from the group consisting of: (1) EGF inhibitors that are antibodies, (2) EGF inhibitors that are small molecules, (3) VEGF inhibitors that are antibodies, and (4) VEGF inhibitors that are small molecules.
  • Radiation therapy can also be used in conjunction with this above combination therapy, i.e., the above method using a combination of compounds of the invention and antineoplastic agent can also comprise the administration of a therapeutically effect amount of radiation.
  • This invention also provides a method of treating leukemias (e.g., acute myeloid leukemia (AML), and chronic myeloid leukemia (CML)) in a patient in need of such treatment, said method comprising administering to said patient therapeutically effective amounts at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) compound of formula 1.0, and: (1 ) Gleevec and interferon to treat CML; (2) Gleevec and pegylated interferon to treat CML; (3) Gleevec to treat CML; (4) an anti-tumor nucleoside derivative (e.g., Ara-C) to treat AML; or (5) an anti-tumor nucleoside derivative (e.g., Ara-C) in combination with an anthracycline to treat AML.
  • leukemias e.g., acute myeloid leukemia (AML), and chronic myeloid leukemia (CML)
  • This invention also provides a method of treating non-Hodgkin's lymphoma in a patient in need of such treatment, said method comprising administering therapeutically effective amounts at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) compound of formula 1.0 and: (1 ) a biologic (e.g., Rituxan); (2) a biologic (e.g., Rituxan) and an anti-tumor nucleoside derivative (e.g., Fludarabine); or (3) Genasense (antisense to BCL-2).
  • a biologic e.g., Rituxan
  • an anti-tumor nucleoside derivative e.g., Fludarabine
  • Genasense antisense to BCL-2
  • This invention also provides a method of treating multiple myeloma in a patient in need of such treatment, said method comprising administering to said patient therapeutically effective amounts of at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) compound of formula 1.0 and: (1 ) a proteosome inhibitor (e.g., PS-341 from Millenium); or (2) Thalidomide (or related imid).
  • at least one e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually
  • a proteosome inhibitor e.g., PS-341 from Millenium
  • Thalidomide or related imid
  • This invention also provides a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient therapeutically effective amounts of: (a) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) compound of formula 1.0, and (b) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 2, or 1) antineoplastic agent selected from the group consisting of: (1 ) taxanes, (2) platinum coordinator compounds, (3) EGF inhibitors that are antibodies, (4) EGF inhibitors that are small molecules, (5) VEGF inhibitors that are antibodies, (6) VEGF kinase inhibitors that are small molecules, (7) estrogen receptor antagonists or selective estrogen receptor modulators, (8) anti-tumor nucleoside derivatives, (9) epothilones, (10) topoisomerase inhibitors, (11) vinca alkaloids, and (12) antibodies that are inhibitors of ⁇ V ⁇ 3 integrins.
  • antineoplastic agent selected
  • This invention also provides a method of treating non small cell lung cancer in a patient in need of such treatment, said method comprising administering to said patient therapeutically effective amounts of: (a) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) compound of formula 1.0, and (b) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 2, or 1) antineoplastic agent selected from the group consisting of: (1) taxanes, (2) platinum coordinator compounds, (3) EGF inhibitors that are antibodies, (4) EGF inhibitors that are small molecules, (5) VEGF inhibitors that are antibodies, (6) VEGF kinase inhibitors that are small molecules, (7) estrogen receptor antagonists or selective estrogen receptor modulators, (8) anti-tumor nucleoside derivatives, (9) epothilones, (10) topoisomerase inhibitors, (11) vinca alkaloids, and (12) antibodies that are inhibitors of ⁇ V ⁇ 3 integrins.
  • This invention also provides a method of treating non small cell lung cancer in a patient in need of such treatment, said method comprising administering to said patient therapeutically effective amounts of: (a) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) compound of formula 1.0, and (b) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 2, or 1 ) antineoplastic agent selected from the group consisting of: (1) taxanes, (2) platinum coordinator compounds, (3) anti-tumor nucleoside derivatives, (4) topoisomerase inhibitors, and (5) vinca alkaloids.
  • antineoplastic agent selected from the group consisting of: (1) taxanes, (2) platinum coordinator compounds, (3) anti-tumor nucleoside derivatives, (4) topoisomerase inhibitors, and (5) vinca alkaloids.
  • This invention also provides a method of treating non small cell lung cancer in a patient in need of such treatment, said method comprising administering therapeutically effective amounts of: (a) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) compound of formula 1.0, (b) carboplatin, and (c) paclitaxel.
  • This invention also provides a method of treating non small cell lung cancer in a patient in need of such treatment, said method comprising administering to said patient therapeutically effective amounts of: (a) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) compound of formula 1.0, (b) cisplatin, and (c) gemcitabine.
  • This invention also provides a method of treating non small cell lung cancer in a patient in need of such treatment, said method comprising administering therapeutically effective amounts of: (a) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) compound of formula 1.0, (b) carboplatin, and (c) gemcitabine.
  • This invention also provides a method of treating non small cell lung cancer in a patient in need of such treatment, said method comprising administering therapeutically effective amounts of: (a) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) compound of formula 1.0, (b) Carboplatin, and (c) Docetaxel.
  • This invention also provides a method of treating cancer in a patient in need of such treatment, said method comprising administering therapeutically effective amounts of: (a) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) compound of formula 1.0, and (b) an antineoplastic agent selected from the group consisting of: (1 ) EGF inhibitors that are antibodies, (2) EGF inhibitors that are small molecules, (3) VEGF inhibitors that are antibodies, (4) VEGF kinase inhibitors that are small molecules.
  • an antineoplastic agent selected from the group consisting of: (1 ) EGF inhibitors that are antibodies, (2) EGF inhibitors that are small molecules, (3) VEGF inhibitors that are antibodies, (4) VEGF kinase inhibitors that are small molecules.
  • This invention also provides a method of treating squamous cell cancer of the head and neck, in a patient in need of such treatment, said method comprising administering to said patient therapeutically effective amounts of: (a) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) compound of formula 1.0, and (b) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 2, or 1) antineoplastic agent selected from the group consisting of: (1 ) taxanes, and (2) platinum coordinator compounds.
  • at least one e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1
  • antineoplastic agent selected from the group consisting of: (1 ) taxanes, and (2) platinum coordinator compounds.
  • This invention also provides a method of treating squamous cell cancer of the head and neck, in a patient in need of such treatment, said method comprising administering to said patient therapeutically effective amounts of: (a) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) compound of formula 1.0, and (b) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 2, or 1 ) antineoplastic agent selected from the group consisting of: (1) taxanes, (2) platinum coordinator compounds, and (3) antitumor nucleoside derivatives (e.g., 5-Fluorouracil).
  • antineoplastic agent selected from the group consisting of: (1) taxanes, (2) platinum coordinator compounds, and (3) antitumor nucleoside derivatives (e.g., 5-Fluorouracil).
  • This invention also provides a method of treating CML in a patient in need of such treatment, said method comprising administering therapeutically effective amounts of: (a) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) compound of formula 1.0, (b) Gleevec, and (c) interferon (e.g., Intron-A).
  • This invention also provides a method of treating CML in a patient in need of such treatment comprising administering therapeutically effective amounts of: (a) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) compound of formula 1.0, (b) Gleevec; and (c) pegylated interferon (e.g., Peg-lntron, and Pegasys).
  • at least one e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1
  • compound of formula 1.0 e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1
  • pegylated interferon e.g., Peg-lntron, and Pegasys
  • This invention also provides a method of treating CML in a patient in need of such treatment comprising administering therapeutically effective amounts of: (a) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) compound of formula 1.0 and (b) Gleevec.
  • This invention also provides a method of treating CMML in a patient in need of such treatment, said method comprising administering to said patient therapeutically effective amounts of at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) compound of formula 1.0.
  • This invention also provides a method of treating AML in a patient in need of such treatment, said method comprising administering to said patient therapeutically effective amounts of: (a) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) compound of formula 1.0, and (b) an anti-tumor nucleoside derivative (e.g., Cytarabine (i.e., Ara-C)).
  • an anti-tumor nucleoside derivative e.g., Cytarabine (i.e., Ara-C)
  • This invention also provides a method of treating AML in a patient in need of such treatment, said method comprising administering to said patient therapeutically effective amounts of: (a) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) compound of formula 1.0, (b) an anti-tumor nucleoside derivative (e.g., Cytarabine (i.e., Ara-C)), and (c) an anthracycline.
  • a at least one
  • an anti-tumor nucleoside derivative e.g., Cytarabine (i.e., Ara-C)
  • an anthracycline e.g., Cytarabine (i.e., Ara-C)
  • This invention also provides a method of treating non-Hodgkin's lymphoma in a patient in need of such treatment, said method comprising administering to said patient therapeutically effective amounts of: (a) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) compound of formula 1.0, and (b) Rituximab (Rituxan).
  • This invention also provides a method of treating non-Hodgkin's lymphoma in a patient in need of such treatment, said method comprising administering to said patient therapeutically effective amounts of: (a) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) compound of formula 1.0, (b) Rituximab (Rituxan), and (c) an antitumor nucleoside derivative (e.g., Fludarabine (i.e., F-ara-A).
  • a) at least one e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 compound of formula 1.0
  • b Rituximab
  • an antitumor nucleoside derivative e.g., Fludarabine (i.e., F-ara-A).
  • This invention also provides a method of treating non-Hodgkin's lymphoma in a patient in need of such treatment, said method comprising administering to said patient therapeutically effective amounts of: (a) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) compound of formula 1.0, and (b) Genasense (antisense to BCL-
  • This invention also provides a method of treating multiple myeloma in a patient in need of such treatment, said method comprising administering therapeutically effective amounts of: (a) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) compound of formula 1.0, and (b) a proteosome inhibitor (e.g., PS-341 (Millenium)).
  • This invention also provides a method of treating multiple myeloma in a patient in need of such treatment, said method comprising administering to said patient therapeutically effective amounts of: (a) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) compound of formula 1.0, and (b) Thalidomide or related imid.
  • This invention also provides a method of treating multiple myeloma in a patient in need of such treatment, said method comprising administering therapeutically effective amounts of: (a) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) compound of formula 1.0, and (b) Thalidomide.
  • This invention is also directed to the methods of treating cancer described herein, particularly those described above, wherein in addition to the administration of the compound of formula 1.0 and antineoplastic agents, radiation therapy is also administered prior to, during, or after the treatment cycle.
  • This invention also provides a method for treating cancer (e.g., lung cancer, prostate cancer and myeloid leukemias) in a patient in need of such treatment, said method comprising administering to said patient (1 ) an effective amount of at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) compound of formula 1.0, in combination with (2) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 2, or 1) antineoplastic agent, microtubule affecting agent and/or radiation therapy.
  • cancer e.g., lung cancer, prostate cancer and myeloid leukemias
  • This invention also provides a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) compound of formula 1.0 in combination with an effective amount of at least one (e.g., 1 , 2 or 3, or
  • the compound of formula 1.0 is administered in an amount of about 50 mg to about 200 mg twice a day, and in another example about 75 mg to about 125 mg administered twice a day, and in yet another example about 100 mg administered twice a day, (2) Paclitaxel (e.g., Taxol ® is administered once per week in an amount of about 50 to about 100 mg/m 2 , and in another example about 60 to about 80 mg/m 2 , and (3) Carboplatin is administered once per week in an amount to provide an AUC of about
  • Paclitaxel e.g., Taxol ® is administered once per week in an amount of about 50 to about 100 mg/m 2 , and in another example about 60 to about 80 mg/m 2
  • Carboplatin is administered once per week in an amount to provide an AUC of about
  • the compound of formula 1.0 is administered in an amount of about 50 mg to about 200 mg twice a day, and in another example about 75 mg to about 125 mg administered twice a day, and yet in another example about 100 mg administered twice a day,
  • Paclitaxel e.g., Taxol ® is administered once per week in an amount of about 50 to about 100 mg/m 2 , and in another example about 60 to about 80 mg/m 2
  • Cisplatin is administered once per week in an amount of about 20 to about 40 mg/m 2 .
  • the compound of formula 1.0 is administered in an amount of about 50 mg to about 200 mg twice a day, and in another example about 75 mg to about 125 mg administered twice a day, and in yet another example about 100 mg administered twice a day, (2) Docetaxel (e.g., Taxotere ® ) is administered once per week in an amount of about 10 to about 45 mg/m 2 , and (3) Carboplatin is administered once per week in an amount to provide an AUC of about 2 to about 3.
  • Docetaxel e.g., Taxotere ®
  • Carboplatin is administered once per week in an amount to provide an AUC of about 2 to about 3.
  • the compound of formula 1.0 is administered in an amount of about 50 mg to about 200 mg twice a day, and in another example about 75 mg to about 125 mg administered twice a day, and in yet another example about 100 mg administered twice a day, (2) Docetaxel (e.g., Taxotere ® ) is administered once per week in an amount of about 10 to about 45 mg/m 2 , and (3) Cisplatin is administered once per week in an amount of about 20 to about 40 mg/m 2 .
  • Docetaxel e.g., Taxotere ®
  • Cisplatin is administered once per week in an amount of about 20 to about 40 mg/m 2 .
  • the compound of formula 1.0 is administered in an amount of about 50 mg to about 200 mg twice a day, and in another example about 75 mg to about 125 mg administered twice a day, and in yet another example about 100 mg administered twice a day,
  • Paclitaxel e.g., Taxol ® is administered once every three weeks in an amount of about 150 to about 250 mg/m 2 , and in another example about 175 to about 225 mg/m 2 , and in yet another example 175 mg/m 2
  • Carboplatin is administered once every three weeks in an amount to provide an AUC of about 5 to about 8, and in another example 6.
  • the compound of formula 1.0 is administered in an amount of 100 mg administered twice a day, (2) Paclitaxel (e.g., Taxol ® is administered once every three weeks in an amount of 175 mg/m 2 , and (3) Carboplatin is administered once every three weeks in an amount to provide an AUC of 6.
  • Paclitaxel e.g., Taxol ® is administered once every three weeks in an amount of 175 mg/m 2
  • Carboplatin is administered once every three weeks in an amount to provide an AUC of 6.
  • the compound of formula 1.0 is administered in an amount of about 50 mg to about 200 mg twice a day, and in another example about 75 mg to about 125 mg administered twice a day, and in yet another example about 100 mg administered twice a day, (2) Paclitaxel (e.g., Taxol ® is administered once every three weeks in an amount of about 150 to about 250 mg/m 2 , and in another example about 175 to about 225 mg/m 2 , and (3) Cisplatin is administered once every three weeks in an amount of about 60 to about 100 mg/m 2 .
  • Paclitaxel e.g., Taxol ® is administered once every three weeks in an amount of about 150 to about 250 mg/m 2 , and in another example about 175 to about 225 mg/m 2
  • Cisplatin is administered once every three weeks in an amount of about 60 to about 100 mg/m 2 .
  • the compound of formula 1.0 is administered in an amount of about 50 mg to about 200 mg twice a day, and in another example about 75 mg to about 125 mg administered twice a day, and in yet another example about 100 mg administered twice a day, (2) Docetaxel (e.g., Taxotere ® is administered once every three weeks in an amount of about 50 to about 100 mg/m 2 , and (3) Carboplatin is administered once every three weeks in an amount to provide an AUC of about 5 to about 8.
  • Docetaxel e.g., Taxotere ® is administered once every three weeks in an amount of about 50 to about 100 mg/m 2
  • Carboplatin is administered once every three weeks in an amount to provide an AUC of about 5 to about 8.
  • the compound of formula 1.0 is administered in an amount of about 50 mg to about 200 mg twice a day, in another example about 75 mg to about 125 mg administered twice a day, and in yet another example about 100 mg administered twice a day, (2) Docetaxel (e.g., Taxotere ® is administered once every three weeks in an amount of about 50 to about 100 mg/m 2 , and (3) Cisplatin is administered once every three weeks in an amount of about 60 to about 100 mg/m 2 .
  • Docetaxel e.g., Taxotere ® is administered once every three weeks in an amount of about 50 to about 100 mg/m 2
  • Cisplatin is administered once every three weeks in an amount of about 60 to about 100 mg/m 2 .
  • the compound of formula 1.0 is administered in an amount of about 50 mg to about 200 mg twice a day, and in another example about 75 mg to about 125 mg administered twice a day, and in yet another example about 100 mg administered twice a day, (2) Docetaxel (e.g., Taxotere ® is administered once every three weeks in an amount of about 75 mg/m 2 , and (3) Carboplatin is administered once every three weeks in an amount to provide an AUC of about 6.
  • Docetaxel e.g., Taxotere ® is administered once every three weeks in an amount of about 75 mg/m 2
  • Carboplatin is administered once every three weeks in an amount to provide an AUC of about 6.
  • the Docetaxel e.g., Taxotere ®
  • Cisplatin the Docetaxel and Cisplatin
  • the Docetaxel e.g., Taxotere ®
  • Carboplatin the Paclitaxel (e.g., Taxol ® ) and Carboplatin
  • the Paclitaxel e.g., Taxol ®
  • Cisplatin the Paclitaxel and Cisplatin
  • the compound of formula 1.0 is administered in an amount of about 100 mg to about 200 mg administered twice a day, (2) Gleevec is administered in an amount of about 400 to about 800 mg/day orally, and (3) interferon (Intron-A) is administered in an amount of about 5 to about 20 million IU three times per week.
  • the compound of formula 1.0 is administered in an amount of about 100 mg to about 200 mg administered twice a day, (2) Gleevec is administered in an amount of about 400 to about 800 mg/day orally, and (3) pegylated interferon (Peg-lntron or Pegasys) is administered in an amount of about 3 to about 6 micrograms/kg/day.
  • pegylated interferon Peg-lntron or Pegasys
  • non-Hodgkin's lymphoma (1 ) the compound of formula 1.0 is administered in an amount of about 50 mg to about 200 mg twice a day, and in another example about 75 mg to about 125 mg administered twice a day, and in yet another example about 100 mg administered twice a day, and (2) Genasense (antisense to BCL-2) is administered as a continuous IV infusion at a dose of about 2 to about 5 r ⁇ g/kg/day (e.g., 3 mg/kg/day) for 5 to 7 days every 3 to 4 weeks.
  • Genasense antisense to BCL-2
  • the compound of formula 1.0 is administered in an amount of about 50 mg to about 200 mg twice a day, and in another example about 75 mg to about 125 mg administered twice a day, and in yet another example about 100 mg administered twice a day, and (2) the proteosome inhibitor (e.g., PS-341 - Millenium) is administered in an amount of about 1.5mg/m 2 twice weekly for two consecutive weeks with a one week rest period.
  • the proteosome inhibitor e.g., PS-341 - Millenium
  • the compound of formula 1.0 is administered in an amount of about 50 mg to about 200 mg twice a day, and in another example about 75 mg to about 125 mg administered twice a day, and in yet another example about 100 mg administered twice a day, and (2) the Thalidomide (or related imid) is administered orally in an amount of about 200 to about 800 mg/day, with dosing being continuous until relapse or toxicity.
  • the chemotherapeutic agents are selected from the group consisting of: paclitaxel, docetaxel, carboplatin, cisplatin, gemcitabine, tamoxifen, Herceptin, Cetuximab, Tarceva, Iressa, bevacizumab, navelbine, IMC-1 C11 , SU5416 and SU6688.
  • the chemotherapeutic agents are selected from the group consisting of: paclitaxel, docetaxel, carboplatin, cisplatin, navelbine, gemcitabine, and Herceptin.
  • one embodiment of this invention is directed to a method of treating cancer comprising administering to a patient in need of such treatment therapeutically effective amounts of the compound of formula 1.0, a taxane, and a platinum coordination compound.
  • Another embodiment of this invention is directed to a method of treating cancer comprising administering to a patient in need of such treatment therapeutically effective amounts of the compound of formula 1.0, a taxane, and a platinum coordination compound, wherein said compound of formula 1.0 is administered every day, said taxane is administered once per week per cycle, and said platinum coordinator compound is administered once per week per cycle.
  • the treatment is for one to four weeks per cycle.
  • Another embodiment of this invention is directed to a method of treating cancer comprising administering to a patient in need of such treatment therapeutically effective amounts of the compound of formula 1.0, a taxane, and a platinum coordination compound, wherein said compound of formula 1.0 is administered every day, said taxane is administered once every three weeks per cycle, and said platinum coordinator compound is administered once every three weeks per cycle.
  • the treatment is for one to three weeks per cycle.
  • Another embodiment of this invention is directed to a method of treating cancer comprising administering to a patient in need of such treatment therapeutically effective amounts of the compound of formula 1.0, paclitaxel, and carboplatin.
  • said compound of formula 1.0 is administered every day, said paclitaxel is administered once per week per cycle, and said carboplatin is administered once per week per cycle.
  • the treatment is for one to four weeks per cycle.
  • Another embodiment of this invention is directed to a method of treating cancer comprising administering to a patient in need of such treatment therapeutically effective amounts of the compound of formula 1.0, paclitaxel, and carboplatin.
  • said compound of formula 1.0 is administered every day, said paclitaxel is administered once every three weeks per cycle, and said carboplatin is administered once every three weeks per cycle.
  • the treatment is for one to three weeks per cycle.
  • Another embodiment of this invention is directed to a method for treating non small cell lung cancer in a patient in need of such treatment comprising administering daily a therapeutically effective amount of the compound of formula 1.0, administering a therapeutically effective amount of carboplatin once a week per cycle, and administering a therapeutically effective amount of paclitaxel once a week per cycle, wherein the treatment is given for one to four weeks per cycle.
  • said compound of formula 1.0 is administered twice per day.
  • said carboplatin and said paclitaxel are administered on the same day, and in another embodiment said carboplatin and said paclitaxel are administered consecutively, and in another embodiment said carboplatin is administered after said paclitaxel.
  • Another embodiment of this invention is directed to a method for treating non small cell lung cancer in a patient in need of such treatment comprising administering daily a therapeutically effective amount of a compound of formula 1.0, administering a therapeutically effective amount of carboplatin once every three weeks per cycle, and administering a therapeutically effective amount of paclitaxel once every three weeks per cycle, wherein the treatment is given for one to three weeks.
  • compound of formula 1.0 is administered twice per day.
  • said carboplatin and said paclitaxel are administered on the same day, and in another embodiment said carboplatin and said paclitaxel are administered consecutively, and in another embodiment said carboplatin is administered after said paclitaxel.
  • Another embodiment of this invention is directed to a method for treating non small cell lung cancer in a patient in need of such treatment comprising administering about 50 to about 200 mg of a compound of formula 1.0 twice a day, administering carboplatin once per week per cycle in an amount to provide an AUC of about 2 to about 8 (and in another embodiment about 2 to about 3), and administering once per week per cycle about 60 to about 300 mg/m 2 (and in another embodiment about 50 to 100mg/m 2 , and in yet another embodiment about 60 to about 80 mg/m 2 ) of paclitaxel, wherein the treatment is given for one to four weeks per cycle.
  • said compound of formula 1.0 is administered in amount of about 75 to about 125 mg twice a day, and in another embodiment about 100 mg twice a day.
  • said carboplatin and said paclitaxel are administered on the same day, and in another embodiment said carboplatin and said paclitaxel are administered consecutively, and in another embodiment said carboplatin is administered after said paclitaxel.
  • this invention is directed to a method for treating non small cell lung cancer in a patient in need of such treatment comprising administering about 50 to about 200 mg of a compound of formula 1.0 twice a day, administering carboplatin once every three weeks per cycle in an amount to provide an AUC of about 2 to about 8 (in another embodiment about 5 to about 8, and in another embodiment 6), and administering once every three weeks per cycle about 150 to about 250 mg/m 2 (and in another embodiment about 175 to about 225 mg/m 2 , and in another embodiment 175 mg/m 2 ) of paclitaxel, wherein the treatment is given for one to three weeks.
  • said compound of formula 1.0 is administered in an amount of about 75 to about 125 mg twice a day, and in another embodiment about 100 mg twice a day.
  • said carboplatin and said paclitaxel are administered on the same day, and in another embodiment said carboplatin and said paclitaxel are administered consecutively, and in another embodiment said carboplatin is administered after said paclitaxel.
  • embodiments of this invention are directed to methods of treating cancer as described in the above embodiments (i.e., the embodiments directed to treating cancer and to treating non small cell lung cancer with a taxane and platinum coordinator compound) except that in place of paclitaxel and carboplatin the taxanes and platinum coordinator compounds used together in the methods are: (1 ) docetaxel (Taxotere®) and cisplatin; (2) paclitaxel and cisplatin; and (3) docetaxel and carboplatin.
  • cisplatin is used in amounts of about 30 to about 100 mg/m 2 .
  • docetaxel is used in amounts of about 30 to about 100 mg/m 2 .
  • this invention is directed to a method of treating cancer comprising administering to a patient in need of such treatment therapeutically effective amounts of a compound of formula 1.0, a taxane, and an EGF inhibitor that is an antibody.
  • the taxane used is paclitaxel
  • the EGF inhibitor is a HER2 antibody (in one embodiment Herceptin) or Cetuximab, and in another embodiment Herceptin is used.
  • the length of treatment, and the amounts and administration of said compound of formula 1.0 and the taxane are as described in the embodiments above.
  • the EGF inhibitor that is an antibody is administered once a week per cycle, and in another embodiment is administered on the same day as the taxane, and in another embodiment is administered consecutively with the taxane.
  • Herceptin is administered in a loading dose of about 3 to about 5 mg/m 2 (in another embodiment about 4 mg/m 2 ), and then is administered in a maintenance dose of about 2 mg/m 2 once per week per cycle for the remainder of the treatment cycle (usually the cycle is 1 to 4 weeks).
  • the cancer treated is breast cancer.
  • this invention is directed to a method of treating cancer comprising administering to a patient in need of such treatment therapeutically effective amounts of: (1) a compound of formula 1.0, (2) a taxane, and (3) an antineoplastic agent selected from the group consisting of: (a) an EGF inhibitor that is a small molecule, (b) a VEGF inhibitor that is an antibody, and (c) a VEGF kinase inhibitor that is a small molecule.
  • the taxane paclitaxel or docetaxel is used.
  • the antineoplastic agent is selected from the group consisting of: tarceva, Iressa, bevacizumab, SU5416, SU6688 and BAY 43- 9006.
  • the length of treatment, and the amounts and administration of said compound of formula 1.0 and the taxane are as described in the embodiments above.
  • the VEGF kinase inhibitor that is an antibody is usually given once per week per cycle.
  • the EGF and VEGF inhibitors that are small molecules are usually given daily per cycle.
  • the VEGF inhibitor that is an antibody is given on the same day as the taxane, and in another embodiment is administered concurrently with the taxane.
  • the EGF inhibitor that is a small molecule or the VEGF inhibitor that is a small molecule is administered on the same day as the taxane, the administration is concurrently with the taxane.
  • the EGF or VEGF kinase inhibitor is generally administered in an amount of about 10 to about 500 mg/m 2 .
  • this invention is directed to a method of treating cancer comprising administering to a patient in need of such treatment therapeutically effective amounts of a compound of formula 1.0, an anti-tumor nucleoside derivative, and a platinum coordination compound.
  • Another embodiment of this invention is directed to a method of treating cancer comprising administering to a patient in need of such treatment therapeutically effective amounts of a compound of formula 1.0, an anti-tumor nucleoside derivative, and a platinum coordination compound, wherein said compound of formula 1.0 is administered every day, said anti-tumor nucleoside derivative is administered once per week per cycle, and said platinum coordinator compound is administered once per week per cycle.
  • the treatment can be for one to four weeks per cycle, in one embodiment the treatment is for one to seven weeks per cycle.
  • Another embodiment of this invention is directed to a method of treating cancer comprising administering to a patient in need of such treatment therapeutically effective amounts of a compound of formula 1.0, an anti-tumor nucleoside derivative, and a platinum coordination compound, wherein said compound of formula 1.0 is administered every day, said an anti-tumor nucleoside derivative is administered once per week per cycle, and said platinum coordinator compound is administered once every three weeks per cycle.
  • the treatment can be for one to four weeks per cycle, in one embodiment the treatment is for one to seven weeks per cycle.
  • Another embodiment of this invention is directed to a method of treating cancer comprising administering to a patient in need of such treatment therapeutically effective amounts of a compound of formula 1.0, gemcitabine, and cisplatin.
  • said compound of formula 1.0 is administered every day, said gemcitabine is administered once per week per cycle, and said cisplatin is administered once per week per cycle.
  • the treatment is for one to seven weeks per cycle.
  • Another embodiment of this invention is directed to a method of treating cancer comprising administering to a patient in need of such treatment therapeutically effective amounts of a compound of formula 1.0, gemcitabine, and cisplatin.
  • said compound of formula 1.0 is administered every day, said gemcitabine is administered once per week per cycle, and said cisplatin is administered once every three weeks per cycle.
  • the treatment is for one to seven weeks.
  • Another embodiment of this invention is directed to a method of treating cancer comprising administering to a patient in need of such treatment therapeutically effective amounts of a compound of formula 1.0, gemcitabine, and carboplatin.
  • said compound of formula 1.0 is administered every day, said gemcitabine is administered once per week per cycle, and said carboplatin is administered once per week per cycle.
  • the treatment is for one to seven weeks per cycle.
  • Another embodiment of this invention is directed to a method of treating cancer comprising administering to a patient in need of such treatment therapeutically effective amounts of a compound of formula 1.0, gemcitabine, and carboplatin.
  • said compound of formula 1.0 is administered every day, said gemcitabine is administered once per week per cycle, and said carboplatin is administered once every three weeks per cycle.
  • the treatment is for one to seven weeks per cycle.
  • the compound of formula 1.0 and the platinum coordinator compound are administered as described above for the embodiments using taxanes.
  • Gemcitabine is administered in an amount of about 500 to about 1250 mg/m 2 .
  • the gemcitabine is administered on the same day as the platinum coordinator compound, and in another embodiment consecutively with the platinum coordinator compound, and in another embodiment the gemcitabine is administered after the platinum coordinator compound.
  • Another embodiment of this invention is directed to a method of treating cancer in a patient in need of such treatment comprising administering to said patient a compound of formula 1.0 and an antineoplastic agent selected from: (1 ) EGF inhibitors that are antibodies, (2) EGF inhibitors that are small molecules, (3) VEGF inhibitors that are antibodies, and (4) VEGF kinase inhibitors that are small molecules all as described above.
  • the treatment is for one to seven weeks per cycle, and generally for one to four weeks per cycle.
  • the compound of formula 1.0 is administered in the same mariner as described above for the other embodiments of this invention.
  • the small molecule antineoplastic agents are usually administered daily, and the antibody antineoplastic agents are usually administered once per week per cycle.
  • antineoplastic agents are selected from the group consisting of: Herceptin, Cetuximab, Tarceva, Iressa, bevacizumab, IMC-1C11 , SU5416, SU6688 and BAY 43-9006.
  • the platinum coordinator compound is generally administered after the other antineoplastic agents have been administered.
  • Other embodiments of this invention include the administration of a therapeutically effective amount of radiation to the patient in addition to the administration of a compound of formula 1.0 and antineoplastic agents in the embodiments described above. Radiation is administered according to techniques and protocols well know to those skilled in the art.
  • Another embodiment of this invention is directed to a pharmaceutical composition
  • a pharmaceutical composition comprising at least two different chemotherapeutic agents and a pharmaceutically acceptable carrier for intravenous administration.
  • the pharmaceutically acceptable carrier is an isotonic saline solution (0.9% NaCI) or a dextrose solution (e.g., 5% dextrose).
  • Another embodiment of this invention is directed to a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula 1.0 and at least two different antineoplastic agents and a pharmaceutically acceptable carrier for intravenous administration.
  • the pharmaceutically acceptable carrier is an isotonic saline solution (0.9% NaCI) or a dextrose solution (e.g., 5% dextrose).
  • Another embodiment of this invention is directed to a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula 1.0 and at least one antineoplastic agent and a pharmaceutically acceptable carrier for intravenous administration.
  • the pharmaceutically acceptable carrier is an isotonic saline solution (0.9%
  • dextrose solution e.g., 5% dextrose
  • compositions comprising more than one drug are within the scope of this invention.
  • another embodiment of this invention is directed to a method of treating
  • breast cancer i.e., postmenopausal and premenopausal breast cancer, e.g., hormone-dependent breast cancer
  • administering comprising administering to said patient a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0 and a therapeutically effective amount of at least one antihormonal agent selected from the group consisting of: (a) aromatase inhibitors, (b) antiestrogens, and (c) LHRH analogues; and said treatment optionally including the administration of at least one chemotherapeutic agent.
  • the compound of formula 1.0 is preferably administered orally, and in one embodiment is administered in capsule form.
  • aromatase inhibitors include but are not limited to: Anastrozole
  • Formestane e.g., Lentaron
  • antiestrogens examples include but are not limited to: Tamoxifen (e.g., Tamoxifen (e.g., Tamoxifen), e.g., Tamoxifen (e.g., Tamoxifen), e.g., Tamoxifen (e.g., Tamoxifen), e.g., Tamoxifen (e.g., Tamoxifen), e.g., Tamoxifen (e.g., Tamoxifen (e.g., Tamoxifen (e.g., Tamoxifen (e.g., Tamoxifen (e.g., Tamoxifen (e.g., Tamoxifen (e.g., Tamoxifen (e.g., Tamoxifen (e.g., Tamoxifen (e.g., Tamoxifen (e.g., Tamoxifen (e.g., Tamoxifen (e.g., Tamoxifen (e.g., Tamoxifen (e.
  • LHRH analogues include but are not limited to: Goserelin (e.g., Goserelin (e.g., Goserelin), FRserelin (FRserelin), FRserelin (FRserelin), FRserelin (FRserelin), FRserelin (FRserelin), FRserelin (FRserelin), FRserelin (e.g., Faslodex), Faslodex, Raloxifene (e.g., Evista), and Acolbifene.
  • LHRH analogues include but are not limited to: Goserelin (e.g., Goserelin (e.g., Goserelin (FRse., Faslodex), Faslodex), Raloxifene (e.g., Evista), and Acolbifene.
  • LHRH analogues include but are not limited
  • chemotherapeutic agents include but are not limited to:
  • Trastuzumab e.g., Herceptin
  • Gefitinib e.g., Iressa
  • Erlotinib e.g., Erlotinib HCI, such as Tarceva
  • Bevacizumab e.g., Avastin
  • Cetuximab e.g., Erbitux
  • Bortezomib e.g., Velcade
  • each agent is selected from a different category of agent.
  • one agent is an aromatase inhibitor (e.g., Anastrozole, Letrozole, or Exemestane) and one agent is an antiestrogen (e.g., Tamoxifen or Fulvestrant).
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0 and at least one antihormonal agent selected from the group consisting of: (a) aromatase inhibitors, (b) antiestrogens, and (c) LHRH analogues; and administering an effective amount of at least one chemotherapeutic agent.
  • at least one e.g., one
  • at least one antihormonal agent selected from the group consisting of: (a) aromatase inhibitors, (b) antiestrogens, and (c) LHRH analogues
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0 and at least one antihormonal agent selected from the group consisting of: (a) aromatase inhibitors, (b) antiestrogens, and (c) LHRH analogues.
  • at least one e.g., one
  • at least one antihormonal agent selected from the group consisting of: (a) aromatase inhibitors, (b) antiestrogens, and (c) LHRH analogues.
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0 and at least one antihormonal agent selected from the group consisting of: (a) aromatase inhibitors, and (b) antiestrogens.
  • at least one e.g., one
  • at least one antihormonal agent selected from the group consisting of: (a) aromatase inhibitors, and (b) antiestrogens.
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, at least one antihormonal agent selected from the group consisting of: (a) aromatase inhibitors and (b) antiestrogens; and at least one chemotherapeutic agent.
  • at least one e.g., one
  • at least one antihormonal agent selected from the group consisting of: (a) aromatase inhibitors and (b) antiestrogens
  • at least one chemotherapeutic agent selected from the group consisting of: (a) aromatase inhibitors and (b) antiestrogens.
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0 and at least one aromatase inhibitor.
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, at least one aromatase inhibitor, and at least one chemotherapeutic agent.
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of: (1) at least one (e.g., one) compound of formula 1.0; and (2) at least one antihormonal agent selected from the group consisting of: (a) aromatase inhibitors that are selected from the group consisting of Anastrozole, Letrozole, Exemestane, Fadrozole and Formestane, (b) antiestrogens that are selected from the group consisting of: Tamoxifen, Fulvestrant, Raloxifene, and Acolbifene, and (c) LHRH analogues that are selected from the group consisting of: Goserelin and Leuprolide; and administering an effective amount of at least one chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of: (1 ) at least one (e.g., one) compound of formula 1.0; and (2) at least one antihormonal agent selected from the group consisting of: (a) aromatase inhibitors that are selected from the group consisting of Anastrozole, Letrozole, Exemestane, Fadrozole and Formestane, (b) antiestrogens that are selected from the group consisting of: Tamoxifen, Fulvestrant, Raloxifene, and Acolbifene, and (c) LHRH analogues that are selected from the group consisting of: Goserelin and Leuprolide.
  • at least one antihormonal agent selected from the group consisting of: (a) aromatase inhibitors that are selected from the group consisting of Anastrozole, Letrozole, Exemestane, Fadrozo
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of: (1) at least one (e.g., one) compound of formula 1.0; and (2) at least one antihormonal agent selected from the group consisting of: (a) aromatase inhibitors that are selected from the group consisting of Anastrozole, Letrozole, Exemestane, Fadrozole and Formestane, and (b) antiestrogens that are selected from the group consisting of: Tamoxifen, Fulvestrant, Raloxifene, and Acolbifene.
  • at least one e.g., one
  • antihormonal agent selected from the group consisting of: (a) aromatase inhibitors that are selected from the group consisting of Anastrozole, Letrozole, Exemestane, Fadrozole and Formestane
  • antiestrogens that are selected from the group consisting of: Tam
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of: (1) at least one (e.g., one) compound of formula 1.0; and (2) at least one antihormonal agent selected from the group consisting of: (a) aromatase inhibitors that are selected from the group consisting of Anastrozole, Letrozole, Exemestane, Fadrozole and Formestane, (b) antiestrogens that are selected from the group consisting of: Tamoxifen, Fulvestrant, Raloxifene, and Acolbifene; and administering an effective amount of at least one chemotherapeutic agents are selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib.
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of: (1) at least one (e.g., one) compound of formula 1.0; and (2) at least one aromatase inhibitor selected from the group consisting of Anastrozole, Letrozole, Exemestane, Fadrozole and Formestane.
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of: (1) at least one (e.g., one) compound of formula 1.0; (2) at least one aromatase inhibitor that is selected from the group consisting of Anastrozole, Letrozole, Exemestane, Fadrozole and Formestane; and (3) administering an effective amount of at least one chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib.
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of: (1 ) at least one (e.g., one) compound of formula 1.0; (2) at least one aromatase inhibitor; and (3) at least one LHRH analogue.
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of:(1) at least one (e.g., one) compound of formula 1.0; (2) at least one antiestrogen ; and (3) at least one LHRH analogue.
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of: (1 ) at least one (e.g., one) compound of formula 1.0; (2) at least one aromatase inhibitor that is selected from the group consisting of Anastrozole, Letrozole, Exemestane, Fadrozole and Formestane; and (3) at least one LHRH analogue that is selected from the group consisting of: Goserelin and Leuprolide.
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of: (1 ) at least one (e.g., one) compound of formula 1.0; (2) at least one antiestrogen that is selected from the group consisting of: Tamoxifen, Fulvestrant, Raloxifene, and Acolbifene; and (3) at least one LHRH analogue that is selected from the group consisting of: Goserelin and Leuprolide.
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0 and Anastrozole.
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0 and Letrazole.
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0 and Exemestane.
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0 and and Fadrozole.
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0 and Formestane.
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0 and Tamoxifen.
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0 Fulvestrant.
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0 and Raloxifene.
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0 and Acolbifene.
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0 and Goserelin.
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0 and and Leuprolide.
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Anastrozole, and an antiestrogen selected from the group consisting of: Tamoxifen, Fulvestrant, Raloxifene, and Acolbifene.
  • at least one e.g., one
  • Anastrozole e.g., one
  • an antiestrogen selected from the group consisting of: Tamoxifen, Fulvestrant, Raloxifene, and Acolbifene.
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Letrozole, and an antiestrogen selected from the group consisting of: Tamoxifen, Fulvestrant, Raloxifene, and Acolbifene.
  • at least one e.g., one
  • an antiestrogen selected from the group consisting of: Tamoxifen, Fulvestrant, Raloxifene, and Acolbifene.
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Exemestane, and an antiestrogen selected from the group consisting of: Tamoxifen, Fulvestrant, Raloxifene, and Acolbifene.
  • at least one e.g., one
  • Exemestane Exemestane
  • an antiestrogen selected from the group consisting of: Tamoxifen, Fulvestrant, Raloxifene, and Acolbifene.
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Fadrozole, and an antiestrogen selected from the group consisting of: Tamoxifen, Fulvestrant, Raloxifene, and Acolbifene.
  • at least one e.g., one
  • Fadrozole e.g., Fadrozole
  • an antiestrogen selected from the group consisting of: Tamoxifen, Fulvestrant, Raloxifene, and Acolbifene.
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Formestane, and an antiestrogen selected from the group consisting of: Tamoxifen, Fulvestrant, Raloxifene, and Acolbifene.
  • at least one e.g., one
  • Formestane e.g., one
  • an antiestrogen selected from the group consisting of: Tamoxifen, Fulvestrant, Raloxifene, and Acolbifene.
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Anastrozole, and Tamoxifen.
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Letrozole, and Tamoxifen.
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Exemestane, and Tamoxifen.
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Fadrozole, and Tamoxifen.
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Formestane, and Tamoxifen.
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Anastrozole, and Fulvestrant.
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Letrozole, and Fulvestrant.
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Exemestane, and Fulvestrant.
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Fadrozole, and Fulvestrant.
  • at least one e.g., one
  • compound of formula 1.0 Fadrozole, and Fulvestrant.
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Formestane, and Fulvestrant.
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Anastrozole, and a chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib.
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Letrozole, and a chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib.
  • a chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib.
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Exemestane, and a chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib.
  • a chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib.
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Fadrozole, and a chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib.
  • a chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib.
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Formestane, and a chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib.
  • a chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib.
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Tamoxifen, and a chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib.
  • a chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib.
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Fulvestrant, and a chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib.
  • a chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib.
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Raloxifene, and a chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib.
  • a chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib.
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Acolbifene, and a chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib.
  • a chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib.
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Goserelin, and a chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib.
  • a chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib.
  • Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Leuprolein, and a chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib.
  • a chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib.

Landscapes

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

Abstract

Disclosed are the ERK inhibitors of formula 1.0: and the pharmaceutically acceptable salts, and solvates thereof. Q is a tetrahydopyridinyl ring. All other substitutents are as defined herein. Also disclosed are methods of treating cancer using the compounds of formula 1.0.

Description

COMPOUNDS THAT ARE ERK INHIBITORS
REFERENCE TO RELATED APPLICATION
This Application claims the benefit of U.S. Provisional Application Serial No. 61/030407 filed February 21 , 2008.
BACKGROUND The processes involved in tumor growth, progression, and metastasis are mediated by signaling pathways that are activated in cancer cells. The ERK pathway plays a central role in regulating mammalian cell growth by relaying extracellular signals from ligand-bound cell surface tyrosine kinase receptors such as erbB family, PDGF, FGF, and VEGF receptor tyrosine kinase. Activation of the ERK pathway is via a cascade of phosphorylation events that begins with activation of Ras. Activation of Ras leads to the recruitment and activation of Raf, a serine-threonine kinase. Activated Raf then phosphorylates and activates MEK1/2, which then phosphorylates and activates ERK1/2. When activated, ERK1/2 phosphorylates several downstream targets involved in a multitude of cellular events including cytoskeletal changes and transcriptional activation. The ERK/MAPK pathway is one of the most important for cell proliferation, and it is believed that the ERK/MAPK pathway is frequently activated in many tumors. Ras genes, which are upstream of ERK1/2, are mutated in several cancers including colorectal, melanoma, breast and pancreatic tumors. The high Ras activity is accompanied by elevated ERK activity in many human tumors. In addition, mutations of BRAF, a serine-threonine kinase of the Raf family, are associated with increased kinase activity. Mutations in BRAF have been identified in melanomas (60%), thyroid cancers (greater than 40%) and colorectal cancers. These observations indicate that the ERK1/2 signalling pathway is an attractive pathway for anticancer therapies in a broad spectrum of human tumours. Therefore, a welcome contribution to the art would be small-molecules (i.e., compounds) that inhibit ERK activity (i.e., ERK1 and ERK2 activity), which small- molecules would be useful for treating a broad spectrum of cancers, such as, for example, melanoma, pancreatic cancer, thryroid cancer, colorectal cancer, lung cancer, breast cancer, and ovarian cancer. Such a contribution is provided by this invention.
SUMMARY OF THE INVENTION
This invention provides compounds that inhibit the activity of ERK1 and/or the activity of ERK2.
The compounds of this invention also inhibit the phosphorylation of ERK1 and ERK2.
Thus, this invention provides compounds that are ERK inhibitors (i.e., ERK1 inhibitors and/or ERK2 inhibitors), said compounds being of the formula 1.0:
Figure imgf000003_0001
or the pharmaceutically acceptable salts, and solvates thereof, wherein: Q is a tetrahydropyridinyl (e.g., 1 ,2,3,6-tetrahydopyridinyl), or a substituted tetrahydropyridinyl (e.g., a substituted 1 ,2,3,6-tetrahydo-pyridinyl); and R1 and R2 are as defined below. This invention provides compounds of formula 1.0.
This invention provides compounds of formula 1.0 in pure or isolated form. This invention provides pharmaceutically acceptable salts of the compounds of formula 1.0.
This invention provides solvates of the compounds of formula 1.0. This invention provides compounds of formula 1.0 wherein from one up to the total number of hydrogens are deuterium.
This invention provides compounds of formula 1.0 wherein at least one H is deuterium.
This invention provides compounds of formula 1.0 wherein 1 to 5 H are deuterium. This invention provides compounds of formula 1.0 wherein one H is deuterium.
This invention provides compounds A1 to A16 and A18 to A48.
This invention provides compounds A1 to A16 and A18 to A30.
This invention provides compounds A1 to A16 and A18 to A26. This invention provides compounds A31 to A48.
This invention also provides a pharmaceutical composition comprising an effective amount of at least one compound of formula 1.0 and a pharmaceutically acceptable carrier.
This invention also provides a pharmaceutical composition comprising an effective amount of at least one compound of formula 1.0 and an effective amount of at least one other pharmaceutically active ingredient (such as, for example, a chemotherapeutic agent), and a pharmaceutically acceptable carrier.
This invention also provides a method of inhibiting ERK (i.e., inhibiting the activity of ERK) in a patient in need of such treatment comprising administering to said patient an effective amount of at least one compound of formula 1.0.
This invention also provides a method of inhibiting ERK1 (i.e., inhibiting the activity of ERK1 ) in a patient in need of such treatment comprising administering to said patient an effective amount of at least one compound of formula 1.0.
This invention also provides a method of inhibiting ERK2 (i.e., inhibiting the activity of ERK2) in a patient in need of such treatment comprising administering to said patient an effective amount of at least one compound of formula 1.0.
This invention also provides a method of inhibiting ERK1 and ERK2 (i.e., inhibiting the activity of ERK1 and ERK2) in a patient in need of such treatment comprising administering to said patient an effective amount of at least one compound of formula 1.0.
This invention also provides a method for treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one compound of formula 1.0.
This invention also provides a method for treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one compound of formula 1.0.
This invention also provides a method for treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one compound of formula 1.0, in combination with an effective amount of at least one chemotherapeutic agent.
This invention also provides a method for treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one compound of formula 1.0, in combination with an effective amount of at least one chemotherapeutic agent.
This invention also provides a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one compound of formula 1.0 in combination with at least one signal transduction inhibitor.
This invention also provides a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least compound of formula 1.0 in combination with at least one signal transduction inhibitor.
In the methods of this invention the compounds of this invention can be administered concurrently or sequentially (i.e., consecutively) with the chemotherapeutic agents or the signal transduction inhibitor.
The methods of treating cancers described herein can optionally include the administration of an effective amount of radiation (i.e., the methods of treating cancers described herein optionally include the administration of radiation therapy).
DETAILED DESCRIPTION OF THE INVENTION
As described herein, unless otherwise indicated, the use of a drug or compound in a specified period is per treatment cycle. For example, once a day means once per day of each day of the treatment cycle. Twice a day means twice per day each day of the treatment cycle. Once a week means one time per week during the treatment cycle. Once every three weeks means once per three weeks during the treatment cycle. The following abbreviations have the following meanings unless defined otherwise:
ACN Acetonitrile
AcOH Acetic acid
Arihy Anhydrous DAST (diethylamino)sulfur trifluoride
DCC Dicyclohexylcarbodiimide
DCU Dicyclohexylurea
DCM Dichloromethane
Dl Deionized water
DIAD Diisopropylazodicarboxylate
DIEA Diisopropylethylamine
DMAP 4-Dimethylaminopyridine
DME Dimethoxyethane
DMF Dimethylformamide
DMFDMA N,N-Dimethylformamide dimethylacetal
DMSO Dimethyl sulfoxide
DTT Dithiothreitol
EDCI 1 -(3-dimethylamino-propyl)-3-ethylcarbodiimide hydrochloride
Et Ethyl
EtOAc Ethyl acetate
EtOH Ethanol
HATU N,N,Nl,N'-Tetramethyl-O-(7-Azabenzotriazol-1-yl)Uronium hexafluorophosphate
Hex hexanes
HOBt 1 -Hydroxylbenzotriazole
HPLC High pressure liquid chromatography
LCMS Liquid chromatography mass spectrometry
LDA Lithium diisopropylamide mCPBA me/a-Chloroperoxybenzoic acid
MeOH Methanol
MTT (3-[4,5-dimethyl-thiazol-2-yl]-2,5-diphenyltetrazolium bromide,
Thiazolyl blue)
NMR Nuclear magnetic resonance
PFP Pentafluorophenol
PMB p-methoxybenzyl
Py Pyridine
Pyr Pyridine
Rb Round bottom flask
Rbt Round bottom flask
RT (r.t.) Room temperature
SEMCI 2-(Trimethylsily)ethoxy methyl chloride
TEA Triethylamine Tr Triphenyl methane
Trt Triphenyl methane
TrCI Triphenyl methane chloride
TFA Trifluoroacetic acid THF Tetrahydrofuran
TLC Thin layer chromatography
TMS Trimethylsilyl
As used herein, unless otherwise specified, the following terms have the following meanings:
"anti-cancer agent" means a drug (medicament or pharmaceutically active ingredient) for treating cancer;
"antineoplastic agent" means a drug (medicament or pharmaceutically active ingredient) for treating cancer (i.e., a chemotherapeutic agent); "at least one", as used in reference to the number of compounds of this invention means for example 1 -6, generally 1 -4, more generally 1 , 2 or 3, and usually one or two, and more usually one; thus, in one example "at least one" means one, in another example "at least one" means two, and in another example "at least one" means three; "at least one", as used in reference to the number of chemotherapeutic agents used, means for example 1 -6, generally 1-4, more generally 1 , 2 or 3, and usually one or two, or one; thus, in one example "at least one" means one, in another example "at least one" means two, and in another example "at least one" means three; "chemotherapeutic agent" means a drug (medicament or pharmaceutically active ingredient) for treating cancer (i.e., and antineoplastic agent);
"compound" with reference to the antineoplastic agents, includes the agents that are antibodies;
"concurrently" means (1) simultaneously in time (e.g., at the same time); or (2) at different times during the course of a common treatment schedule; "consecutively" means one following the other; "different" as used in the phrase "different antineoplastic agents" means that the agents are not the same compound or structure; preferably, "different" as used in the phrase "different antineoplastic agents" means not from the same class of antineoplastic agents; for example, one antineoplastic agent is a taxane, and another antineoplastic agent is a platinum coordinator compound;
"effective amount" or "therapeutically effective amount" is meant to describe an amount of compound or a composition of the present invention, or an amount of radiation, effective in treating or inhibiting the diseases or conditions described herein, and thus producing the desired therapeutic, ameliorative, inhibitory or preventative effect; thus, for example, in the methods of treating cancer described herein "effective amount" (or "therapeutically effective amount" ) means, for example, the amount of the compound (or drug), or radiation, that results in: (a) the reduction, alleviation or disappearance of one or more symptoms caused by the cancer, (b) the reduction of tumor size, (c) the elimination of the tumor, and/or (d) long-term disease stabilization (growth arrest) of the tumor; for example, in the treatment of lung cancer (e.g., non small cell lung cancer) a therapeutically effective amount is that amount that alleviates or eliminates cough, shortness of breath and/or pain; also, for example, an effective amount, or a therapeutically effective amount of the ERK inhibitor (i.e., a compound of this invention) is that amount which results in the reduction in ERK (ERK1 and/or ERK2) activity and phosphorylation; the reduction in ERK activity may be determined by the analysis of pharmacodynamic markers such as phosphorylated RSK1 ,2 and phosphorylated ERK1 ,2, using techniques well known in the art; "Ex" in the tables represents "Example";
"one or more" has the same meaning as "at least one"; "patient" means an animal, such as a mammal (e.g., a human being, and preferably a human being);
"prodrug" means compounds that are rapidly transformed, for example, by hydrolysis in blood, in vivo to the parent compound, i.e., to the compounds of formula 1.0 or to a salt and/or to a solvate thereof; a thorough discussion is provided in T. Higuchi and V. Stella, Pro-drugs as Novel Delivery Systems, Vol. 14 of the A.C.S. Symposium Series, and in Edward B. Roche, ed., Bioreversible Carriers in Drug Design, American Pharmaceutical Association and Pergamon Press, 1987, both of which are incorporated herein by reference; the scope of this invention includes Prodrugs of the novel compounds of this invention; sequentially-represents (1) administration of one component of the method ((a) compound of the invention, or (b) chemotherapeutic agent, signal transduction inhibitor and/or radiation therapy) followed by administration of the other component or components; after adminsitration of one component, the next component can be administered substantially immediately after the first component, or the next component can be administered after an effective time period after the first component; the effective time period is the amount of time given for realization of maximum benefit from the administration of the first component; and
"solvate" means a physical association of a compound of this invention with one or more solvent molecules; this physical association involves varying degrees of ionic and covalent bonding, including hydrogen bonding; in certain instances the solvate will be capable of isolation, for example when one or more solvent molecules are incorporated in the crystal lattice of the crystalline solid; "solvate" encompasses both solution-phase and isolatable solvates; non-limiting examples of suitable solvates include ethanolates, methanolates, and the like; "hydrate" is a solvate wherein the solvent molecule is H2O.
As used herein, unless otherwise specified, the following terms have the following meanings, and unless otherwise specified, the definitions of each term (i.e., moiety or substituent) apply when that term is used individually or as a component of another term (e.g., the definition of aryl is the same for aryl and for the aryl portion of arylalkyl, alkylaryl, arylalkynyl, and the like):
"acyl" means an H-C(O)-, alkyl-C(O)-, alkenyl-C(O)-, Alkynyl-C(O)-, cycloalkyl-C(O)-, cycloalkenyl-C(O)-, or cycloalkynyl-C(O)- group in which the various groups are as defined below (and as defined below, the alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl and cycloalkynyl moieties can be substituted); the bond to the parent moiety is through the carbonyl; preferred acyls contain a lower alkyl; Non- limiting examples of suitable acyl groups include formyl, acetyl, propanoyl, 2- methylpropanoyl, butanoyl and cyclohexanoyl;
"alkenyl" means an aliphatic hydrocarbon group (chain) comprising at least one carbon to carbon double bond, wherein the chain can be straight or branched, and wherein said group comprises about 2 to about 15 carbon atoms; Preferred alkenyl groups comprise about 2 to about 12 carbon atoms in the chain; and more preferably about 2 to about 6 carbon atoms in the chain; branched means that one or more lower alkyl groups, such as methyl, ethyl or propyl, or alkenyl_groups are attached to a linear alkenyl chain; "lower alkenyl" means an alkenyl group comprising about 2 to about 6 carbon atoms in the chain, and the chain can be straight or branched; the term "substituted alkenyl" means that the alkenyl group is substituted by one or more independently selected substituents, and each substituent is independently selected from the group consisting of: halo, alkyl, aryl, cycloalkyl, cyano, alkoxy and -S(alkyl); non-limiting examples of suitable alkenyl groups include ethenyl, propenyl, n-butenyl, 3-methylbut-2-enyl, n-pentenyl, octenyl and decenyl; "alkoxy" means an alkyl-O- group (i.e., the bond to the parent moiety is through the ether oxygen) in which the alkyl group is unsubstituted or substituted as described below; non-limiting examples of suitable alkoxy groups include methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy and heptoxy;
"alkoxycarbonyl" means an alkyl-O-CO- group (i.e., the bond to the parent moiety is through the carbonyl) wherein the alkyl group is unsubstituted or substituted as previously defined; non-limiting examples of suitable alkoxycarbonyl groups include methoxycarbonyl and ethoxycarbonyl;
"alkyl" (including the alkyl portions of other moieties, such as trifluoroalkyl and alkyloxy) means an aliphatic hydrocarbon group (chain) that can be straight or branched wherein said group comprises about 1 to about 20 carbon atoms in the chain; preferred alkyl groups comprise about 1 to about 12 carbon atoms in the chain; more preferred alkyl groups comprise about 1 to about 6 carbon atoms in the chain; branched means that one or more lower alkyl groups, such as methyl, ethyl or propyl, are attached to a linear alkyl chain; "lower alkyl" means a group comprising about 1 to about 6 carbon atoms in the chain, and said chain can be straight or branched; the term "substituted alkyl" means that the alkyl group is substituted by one or more independently selected substituents, and wherein each substituent is independently selected from the group consisting of: halo, aryl, cycloalkyl, cyano, hydroxy, alkoxy, alkylthio, amino, -NH(alkyl), -NH(cycloalkyl), -N(alkyl)2, carboxy, -C(O)O-alkyl and -S(alkyl); non-limiting examples of suitable alkyl groups include methyl, ethyl, n- propyl, isopropyl, n-butyl, t-butyl, n-pentyl, heptyl, nonyl, decyl, fluoromethyl, trifluoromethyl and cyclopropylmethyl;
"alkylaryl" (or alkaryl) means an alkyl-aryl- group (i.e., the bond to the parent moiety is through the aryl group) wherein the alkyl group is unsubstituted or substituted as defined above, and the aryl group is unsubstituted or substituted as defined below; preferred alkylaryls comprise a lower alkyl group; non-limiting examples of suitable alkylaryl groups include o-tolyl, p-tolyl and xylyl;
"alkylheteroaryl" means an alkyl-heteroaryl- group (i.e., the bond to the parent moiety is through the heteroaryl group) wherein the alkyl is unsubstituted or substituted as defined above and the heteroaryl group is unsubstituted or substituted as defined below;
"alkylsulfinyl" means an alkyl-S(O)- group (i.e., the bond to the parent moiety is through the sulfinyl) wherein the alkyl group is unsubstituted or substituted as previously defined; preferred groups are those in which the alkyl group is lower alkyl;
"alkylsulfonyl" means an alkyl-S(O2)- group (i.e., the bond to the parent moiety is through the sulfonyl) wherein the alkyl group is unsubstituted or substituted as previously defined; preferred groups are those in which the alkyl group is lower alkyl;
"alkylthio" means an alkyl-S- group (i.e., the bond to the parent moiety is through the sulfur) wherein the alkyl group is unsubstituted or substituted as previously described; non-limiting examples of suitable alkylthio groups include methylthio, ethylthio, i-propylthio and heptylthio; "alkynyl" means an aliphatic hydrocarbon group (chain) comprising at least one carbon to carbon triple bond, wherein the chain can be straight or branched, and wherein the group comprises about 2 to about 15 carbon atoms in the; preferred alkynyl groups comprise about 2 to about 12 carbon atoms in the chain; and more preferably about 2 to about 4 carbon atoms in the chain; Branched means that one or more lower alkyl groups, such as methyl, ethyl or propyl, are attached to a linear alkynyl chain; "lower alkynyl" means an alkynyl group comprising about 2 to about 6 carbon atoms in the chain, and the chain can be straight or branched; non-limiting examples of suitable alkynyl groups include ethynyl, propynyl, 2-butynyl, 3- methylbutynyl, n-pentynyl, and decynyl; the term "substituted alkynyl" means that the alkynyl group is substituted by one or more independently selected, and each substituent is independently selected from the group consisting of alkyl; aryl and cycloalkyl;
"amino means a -NH2 group;
"aralkenyl" (or arylalkenyl) means an aryl-alkenyl- group (i.e., the bond to the parent moiety is through the alkenyl group) wherein the aryl group is unsubstituted or substituted as defined below, and the alkenyl group is unsubstituted or substituted as defined above; preferred aralkenyls contain a lower alkenyl group; non-limiting examples of suitable aralkenyl groups include 2-phenethenyl and 2-naphthylethenyl; "aralkyl" (or arylalkyl) means an aryl-alkyl- group (i.e., the bond to the parent moiety is through the alkyl group) wherein the aryl is unsubstituted or substituted as defined below and the alkyl is unsubstituted or substituted as defined above; preferred aralkyls comprise a lower alkyl group; non-limiting examples of suitable aralkyl groups include benzyl, 2-phenethyl and naphthalenylmethyl;
"aralkyloxy" (or arylalkyloxy) means an aralkyl-O- group (i.e., the bond to the parent moiety is through the ether oxygen) wherein the aralkyl group is unsubstituted or substituted as previously described; non-limiting examples of suitable aralkyloxy groups include benzyloxy and 1- or 2-naphthalenemethoxy; "aralkoxycarbonyl" means an aralkyl-O-C(O)- group (i.e., the bond to the parent moiety is through the carbonyl) wherein the aralkyl group is unsubstituted or substituted as previously defined; a non-limiting example of a suitable aralkoxycarbonyl group is benzyloxycarbonyl;
"aralkylthio" means an aralkyl-S- group (i.e., the bond to the parent moiety is through the sulfur) wherein the aralkyl group is unsubstituted or substituted as previously described; a non-limiting example of a suitable aralkylthio group is benzylthio;
"aroyl" means an aryl-C(O)- group (i.e., the bond to the parent moiety is through the carbonyl) wherein the aryl group is unsubstituted or substituted as defined below; non-limiting examples of suitable groups include benzoyl and 1- and 2-naphthoyl;
"aryl" (sometimes abbreviated "ar") means an aromatic monocyclic or multicyclic ring system comprising about 6 to about 14 carbon atoms, preferably about 6 to about 10 carbon atoms; the aryl group can be optionally substituted with one or more independently selected "ring system substituents" (defined below). Non-limiting examples of suitable aryl groups include phenyl and naphthyl;
"arylalkynyl" means an aryl-alkynyl- group (i.e., the bond to the parent moiety is through the alkynyl group) wherein the aryl group is unsubstituted or substituted as defined above, and the alkynyl group is unsubstituted or substitutedas defined above;
"arylaminoheteroaryl" means an aryl-amino-heteroaryl group (i.e., the bond to the parent moiety is through the heteroaryl group) wherein the aryl group is unsubstituted or substituted as defined above, the amino group is as defined above (i.e., a -NH- here), and the heteroaryl group is unsubstituted or substituted as defined below;
"arylheteroaryl" means an aryl-heteroarylgroup-(i.e., the bond to the parent moiety is through the heteroaryl group) wherein the aryl group is unsubstituted or substituted as defined above, and the heteroaryl group is unsubstituted or substituted as defined below;
"aryloxy" means an aryl-O- group (i.e., the bond to the parent moiety is through the ether oxygen) wherein the aryl group is unsubstituted or substituted as defined above; non-limiting examples of suitable aryloxy groups include phenoxy and naphthoxy;
"aryloxycarbonyl" means an aryl-O-C(O)- group (i.e., the bond to the parent moiety is through the carbonyl) wherein the aryl group is unsubstituted or substituted as previously defined; non-limiting examples of suitable aryloxycarbonyl groups include phenoxycarbonyl and naphthoxycarbonyl; "arylsulfinyl" means an aryl-S(O)- group (i.e., the bond to the parent moiety is through the sulfinyl) wherein aryl is unsubstituted or substituted as previously defined;
"arylsulfonyl" means an aryl-S(C>2)- group (i.e., the bond to the parent moiety is through the sulfonyl) wherein aryl is unsubstituted or substituted as previously defined;
"arylthio" means an aryl-S- group (i.e., the bond to the parent moiety is through the sulfur) wherein the aryl group is unsubstituted or substituted as previously described; non-limiting examples of suitable arylthio groups include phenylthio and naphthylthio; "cycloalkenyl" means a non-aromatic mono or multicyclic ring system comprising about 3 to about 10 carbon atoms, preferably about 5 to about 10 carbon atoms that contains at least one carbon-carbon double bond; preferred cycloalkenyl rings contain about 5 to about 7 ring atoms; the cycloalkenyl can be optionally substituted with one or more independently selected "ring system substituents" (defined below); Non-limiting examples of suitable monocyclic cycloalkenyls include cyclopentenyl, cyclohexenyl, cycloheptenyl, and the like; a non-limiting example of a suitable multicyclic cycloalkenyl is norbornylenyl;
"cycloalkyl" means a non-aromatic mono- or multicyclic ring system comprising about 3 to about 7 carbon atoms, preferably about 3 to about 6 carbon atoms; the cycloalkyl can be optionally substituted with one or more independently selected "ring system substituents" (defined below); non-limiting examples of suitable monocyclic cycloalkyls include cyclopropyl, cyclopentyl, cyclohexyl, cycloheptyl and the like; non-limiting examples of suitable multicyclic cycloalkyls include 1 -decalin, norbornyl, adamantyl and the like;
"cycloalkylalkyl" means a cycloalkyl-alkyl-group (i.e., the bond to the parent moiety is through the alkyl group) wherein the cycloalkyl moiety is unsubstituted or substituted as defined above, and the alkyl moiety is unsubstituted or substituted as defined above; "halo" means fluoro, chloro, bromo, or iodo groups; preferred halos are fluoro, chloro or bromo, and more preferred are fluoro and chloro;
"halogen" means fluorine, chlorine, bromine, or iodine; preferred halogens are fluorine, chlorine and bromine;
"haloalkyl" means an alkyl, as defined above, wherein one or more hydrogen atoms on the alkyl is replaced by a halo group, as defined above;
"heteroaralkenyl" means a heteroaryl-alkenyl- group (i.e., the bond to the parent moiety is through the alkenyl group) wherein the heteroaryl group is unsubstituted or substituted as defined below, and the alkenyl group is unsubstituted or substituted as defined above; "heteroaralkyl" (or heteroarylalkyl) means a heteroaryl-alkyl- group (i.e., the bond to the parent moiety is through the alkyl group) in which the heteroaryl is unsubstituted or substituted as defined below, and the alkyl group is unsubstituted or substituted as defined above; preferred heteroaralkyls comprise an alkyl group that is a lower alkyl group; non-limiting examples of suitable aralkyl groups include pyridylmethyl, 2-(furan-3-yl)ethyl and quinolin-3-ylmer.hyl;
"heteroaralkylthio" means a heteroaralkyl-S- group wherein the heteroaralkyl group is unsubstituted or substituted as defined above;
"heteroaryl" means an aromatic monocyclic or multicyclic ring system comprising about 5 to about 14 ring atoms, preferably about 5 to about 10 ring atoms, in which one or more of the ring atoms is an element other than carbon, for example nitrogen, oxygen or sulfur, alone or in combination; preferred heteroaryls comprise about 5 to about 6 ring atoms; the "heteroaryl" can be optionally substituted by one or more independently selected "ring system substituents" (defined below); the prefix aza, oxa or thia before the heteroaryl root name means that at least a nitrogen, oxygen or sulfur atom, respectively, is present as a ring atom; a nitrogen atom of a heteroaryl can be optionally oxidized to the corresponding N-oxide; non-limiting examples of suitable heteroaryls include pyridyl, pyrazinyl, furanyl, thienyl, pyrimidinyl, isoxazolyl, isothiazolyl, oxazolyl, thiazolyl, pyrazolyl, furazanyl, pyrrolyl, pyrazolyl, triazolyl, 1 ,2,4-thiadiazolyl, pyrazinyl, pyridazinyl, quinoxalinyl, phthalazinyl, imidazo[1 ,2-a]pyridinyl, imidazo[2,1-b]thiazolyl, benzofurazanyl, indolyl, azaindolyl, benzimidazolyl, benzothienyl, quinolinyl, imidazolyl, thienopyridyl, quinazolinyl, thienopyrimidyl, pyrrolopyridyl, imidazopyridyl, isoquinolinyl, benzoazaindolyl, 1 ,2,4- triazinyl, benzothiazolyl, furopyridine
Figure imgf000015_0001
and the like;
"heteroarylalkynyl" (or heteroaralkynyl) means a heteroaryl-alkynyl- group (i.e., the bond to the parent moiety is through the alkynyl group) wherein the heteroaryl group is unsubstituted or substituted as defined above, and the alkynyl group is unsubstituted or substituted as defined above;
"heteroarylaryl" (or heteroararyl) means a heteroaryl-aryl- group (i.e., the bond to the parent moiety is through the aryl group) wherein the heteroaryl group is unsubstituted or substituted as defined above, and the aryl group is unsubstituted or substituted as defined above; "heteroarylheteroarylaryl" means a heteroaryl-heteroaryl- group (i.e., the bond to the parent moiety is through the last heteroaryl group) wherein each heteroaryl group is independently unsubstituted or substituted as defined above;
"heteroarylsulfinyl" means a heteroaryl-SO- group wherein the heteroaryl group is unsubstituted or substituted as defined above; "heteroarylsulfonyl" means a heteroaryl-SO2- group wherein the heteroaryl group is unsubstituted or substituted as defined above;
"heteroarylthio" means a heteroaryl-S- group wherein the heteroaryl group is unsubstituted or substituted as defined above;
"heterocyclenyl" (or heterocycloalkenyl) means a non-aromatic monocyclic or multicyclic ring system comprising about 3 to about 10 ring atoms, preferably about 5 to about 10 ring atoms, in which one or more of the atoms in the ring system is an element other than carbon (for example one or more heteroatoms independently selected from the group consisting of nitrogen, oxygen and sulfur atom), and which contains at least one carbon-carbon double bond or carbon-nitrogen double bond; there are no adjacent oxygen and/or sulfur atoms present in the ring system;
Preferred heterocyclenyl rings contain about 5 to about 6 ring atoms; the prefix aza, oxa or thia before the heterocyclenyl root name means that at least a nitrogen, oxygen or sulfur atom, respectively, is present as a ring atom; the heterocyclenyl can be optionally substituted by one or more independently selected "Ring system substituents" (defined below); the nitrogen or sulfur atom of the heterocyclenyl can be optionally oxidized to the corresponding N-oxide, S-oxide or S,S-dioxide; non-limiting examples of suitable monocyclic azaheterocyclenyl groups include 1 ,2,3,4- tetrahydropyridine, 1 ,2-dihydropyridyl, 1 ,4-dihydropyridyl, 1 ,2,3,6-tetrahydropyridine, 1 ,4,5,6-tetrahydropyrimidine, 2-pyrrolinyl, 3-pyrrolinyl, 2-imidazolinyl, 2-pyrazolinyl, and the like; Non-limiting examples of suitable oxaheterocyclenyl groups include 3,4- dihydro-2H-pyran, dihydrofuranyl, fluorodihydrofuranyl, and the like; A non-limiting example of a suitable multicyclic oxaheterocyclenyl group is 7- oxabicyclo[2.2.1]heptenyl; non-limiting examples of suitable monocyclic thiaheterocyclenyl rings include dihydrothiophenyl, dihydrothiopyranyl, and the like; "heterocycloalkylalkyl" (or heterocyclylalkyl) means a heterocycloalkyl- alkyl- group (i.e., the bond to the parent moiety is through the alkyl group) wherein the heterocycloalkyl group (i.e., the heterocyclyl group) is unsubstituted or substituted as defined below, and the alkyl group is unsubstituted or substituted as defined above; "heterocyclyl" (or heterocycloalkyl) means a non-aromatic saturated monocyclic or multicyclic ring system comprising about 3 to about 10 ring atoms, preferably about 5 to about 10 ring atoms, in which one or more of the atoms in the ring system is an element other than carbon, for example nitrogen, oxygen or sulfur, alone or in combination; there are no adjacent oxygen and/or sulfur atoms present in the ring system; preferred heterocyclyls contain about 5 to about 6 ring atoms; the prefix aza, oxa or thia before the heterocyclyl root name means that at least a nitrogen, oxygen or sulfur atom respectively is present as a ring atom; the heterocyclyl can be optionally substituted by one or more independently selected "ring system substituents" (defined below); the nitrogen or sulfur atom of the heterocyclyl can be optionally oxidized to the corresponding N-oxide, S-oxide or S,S-dioxide; non-limiting examples of suitable monocyclic heterocyclyl rings include piperidyl, pyrrolidinyl, piperazinyl, morpholinyl, thiomorpholinyl, thiazolidinyl, 1 ,3-dioxolanyl, 1 ,4-dioxanyl, tetrahydrofuranyl, tetrahydrothiophenyl, tetrahydrothiopyranyl, and the like;
"hydroxyalkyl" means a HO-alkyl- group wherein the alkyl group is substituted or unsubstituted as defined above; preferred hydroxyalkyls comprise a lower alkyl; Non-limiting examples of suitable hydroxyalkyl groups include hydroxymethyl and 2-hydroxyethyl; and
"ring system substituent" means a substituent attached to an aromatic or non-aromatic ring system that, for example, replaces an available hydrogen on the ring system; ring system substituents are each independently selected from the group consisting of: alkyl, aryl, heteroaryl, aralkyl, alkylaryl, aralkenyl, heteroaralkyl, alkylheteroaryl, heteroaralkenyl, hydroxy, hydroxyalkyl, alkoxy, aryloxy, aralkoxy, acyl, aroyl, halo, nitro, cyano, carboxy, alkoxycarbonyl, aryloxycarbonyl, aralkoxycarbonyl, alkylsulfonyl, arylsulfonyl, heteroarylsulfonyl, alkylsulfinyl, arylsulfinyl, heteroarylsulfinyl, alkylthio, arylthio, heteroarylthio, aralkylthio, heteroaralkylthio, cycloalkyl, cycloalkenyl, heterocyclyl, heterocyclenyl, R60R65N-, R60R65N-alkyl-, R60R65NC(O)- and R60R65NSO2-, wherein R60 and R65 are each independently selected from the group consisting of: hydrogen, alkyl, aryl, and aralkyl; "Ring system substituent" also means a cyclic ring of 3 to 7 ring atoms, wherein 1-2 ring atoms can be heteroatoms, attached to an aryl, heteroaryl, heterocyclyl or heterocyclenyl ring by simultaneously substituting two ring hydrogen atoms on said aryl, heteroaryl, heterocyclyl or heterocyclenyl ring; Non-limiting examples include:
Figure imgf000017_0001
Lines drawn into a ring mean that the indicated bond may be attached to any of the substitutable ring carbon atoms.
Any carbon or heteroatom with unsatisfied valences in the text, schemes, examples, structural formulae, and any Tables herein is assumed to have the hydrogen atom or atoms to satisfy the valences. And any one or more of these hydrogen atoms can be deuterium. One or more compounds of the invention may also exist as, or optionally converted to, a solvate. Preparation of solvates is generally known. Thus, for example, M. Caira et al, J. Pharmaceutical Sci., 93(3), 601-611 (2004) describe the preparation of the solvates of the antifungal fluconazole in ethyl acetate as well as from water. Similar preparations of solvates, hemisolvate, hydrates and the like are described by E. C. van Tonder et al, AAPS PharmSciTech., 5(1), article 12 (2004); and A. L. Bingham et al, Chem. Commun., 603-604 (2001). A typical, non-limiting, process involves dissolving the inventive compound in desired amounts of the desired solvent (organic or water or mixtures thereof) at a higher than ambient temperature, and cooling the solution at a rate sufficient to form crystals which are then isolated by standard methods. Analytical techniques such as, for example I. R. spectroscopy, show the presence of the solvent (or water) in the crystals as a solvate (or hydrate).
The term "pharmaceutical composition" is also intended to encompass both the bulk composition and individual dosage units comprised of more than one (e.g., two) pharmaceutically active agents such as, for example, a compound of the present invention and an additional agent selected from the lists of the additional agents described herein, along with any pharmaceutically inactive excipients. The bulk composition and each individual dosage unit can contain fixed amounts of the aforesaid "more than one pharmaceutically active agents". The bulk composition is material that has not yet been formed into individual dosage units. An illustrative dosage unit is an oral dosage unit such as tablets, capsules, pills and the like. Similarly, the herein-described methods of treating a patient by administering a pharmaceutical composition of the present invention is also intended to encompass the administration of the afore-said bulk composition and individual dosage units. Prodrugs of the compounds of the invention are also contemplated herein.
The term "prodrug", as employed herein, denotes a compound that is a drug precursor which, upon administration to a subject, undergoes chemical conversion by metabolic or chemical processes to yield a compound of formula 1.0 or a salt and/or solvate thereof. A discussion of prodrugs is provided in T. Higuchi and V. Stella, Pro- drugs as Novel Delivery Systems (1987) 1_4 of the A.C.S. Symposium Series, and in Bioreversible Carriers in Drug Design, (1987) Edward B. Roche, ed., American Pharmaceutical Association and Pergamon Press, both of which are incorporated herein by reference thereto. For example, if a compound of formula 1.0, or a pharmaceutically acceptable salt, hydrate or solvate of the compound, contains a carboxylic acid functional group, a prodrug can comprise an ester formed by the replacement of the hydrogen atom of the acid group with a group such as, for example, (Ci-C8)alkyl, (C2-Ci 2)alkanoyloxy- methyl, 1 -(alkanoyloxy)ethyl having from 4 to 9 carbon atoms, 1 -methyl-1 -
(alkanoyloxy)-ethyl having from 5 to 10 carbon atoms, alkoxycarbonyloxymethyl having from 3 to 6 carbon atoms, 1 -(alkoxycarbonyloxy)ethyl having from 4 to 7 carbon atoms, 1 -methyl-1 -(alkoxycarbonyloxy)ethyl having from 5 to 8 carbon atoms, N-(alkoxycarbonyl)aminomethyl having from 3 to 9 carbon atoms, 1-(N-(alkoxy- carbonyl)amino)ethyl having from 4 to 10 carbon atoms, 3-phthalidyl, 4- crotonolactonyl, gamma-butyrolacton-4-yl, di-N,N-(Ci-C2)alkylamino(C2-C3)alkyl (such as β-dimethylaminoethyl), carbamoyl-(Ci-C2)alkyl, N,N-di (Ci-C2)alkylcarbamoyl-(C1 - C2)alkyl and piperidino-, pyrrolidino- or morpholino(C2-C3)alkyl, and the like.
Similarly, if a compound of formula 1.0 contains an alcohol functional group, a prodrug can be formed by the replacement of the hydrogen atom of the alcohol group with a group such as, for example, (Ci-C6)alkanoyloxymethyl, 1-((CrC6)alkanoyl- oxy)ethyl, 1 -methyl-1 -((Ci-C6)alkanoyloxy)ethyl, (Ci-C6)alkoxycarbonyloxymethyl, N- (Ci-C6)alkoxycarbonylaminomethyl, succinoyl, (CrC6)alkanoyl, α-amino(Cr C4)alkanyl, arylacyl and α-aminoacyl, or α-aminoacyl-α-aminoacyl, where each α- aminoacyl group is independently selected from the naturally occurring L-amino acids, P(O)(OH)2, -P(O)(O(Ci -C6)alkyl)2 or glycosyl (the radical resulting from the removal of a hydroxyl group of the hemiacetal form of a carbohydrate), and the like.
If a compound of formula 1.0 incorporates an amine functional group, a prodrug can be formed by the replacement of a hydrogen atom in the amine group with a group such as, for example, R70-carbonyl, R70O-carbonyl, NR70R75-carbonyl where R70 and R75 are each independently (Ci-CiO)alkyl, (C3-C7) cycloalkyl, benzyl, or R70-carbonyl is a natural α-aminoacyl or natural α-aminoacyl, — C(OH)C(O)OY80 wherein Y80 is H, (CrC6)alkyl or benzyl, — C(OY82)Y84 wherein Y82 is (CrC4) alkyl and Y84 is (CrC6)alkyl, carboxy (d-C6)alkyl, amino(d-C4)alkyl or mono-N— or di-N,N-(Cr C6)alkylaminoalkyl, -C(Y86) Y88 wherein Y86 is H or methyl and Y88 is mono-N— or di- N,N-(Ci-C6)alkylamino morpholino, piperidin-1 -yl or pyrrolidin-1 -yl, and the like.
This invention also includes the compounds of this invention in isolated and purified form. Polymorphic forms of the compounds of formula 1.0, and of the salts, solvates and prodrugs of the compounds of formula 1.0, are intended to be included in the present invention.
Certain compounds of the invention may exist in different isomeric (e.g., enantiomers, diastereoisomers, atropisomers) forms. The invention contemplates all such isomers both in pure form and in admixture, including racemic mixtures. Enol forms are also included.
All stereoisomers (for example, geometric isomers, optical isomers and the like) of the present compounds (including those of the salts, solvates and prodrugs of the compounds as well as the salts and solvates of the prodrugs), such as those which may exist due to asymmetric carbons on various substituents, including enantiomeric forms (which may exist even in the absence of asymmetric carbons), rotameric forms, atropisomers, and diastereomeric forms, are contemplated within the scope of this invention. Individual stereoisomers of the compounds of the invention may, for example, be substantially free of other isomers, or may be admixed, for example, as racemates or with all other, or other selected, stereoisomers. The chiral centers of the present invention can have the S or R configuration as defined by the IUPAC 1974 Recommendations. The use of the terms "salt", "solvate" "prodrug" and the like, is intended to equally apply to the salt, solvate and prodrug of enantiomers, stereoisomers, rotamers, tautomers, racemates or prodrugs of the inventive compounds.
Diasteromeric mixtures can be separated into their individual diastereomers on the basis of their physical chemical differences by methods well known to those skilled in the art, such as, for example, by chromatography and/or fractional crystallization. Enantiomers can be separated by converting the enantiomeric mixture into a diasteromeric mixture by reaction with an appropriate optically active compound (e.g., chiral auxiliary such as a chiral alcohol or Mosher's acid chloride), separating the diastereomers and converting (e.g., hydrolyzing) the individual diastereomers to the corresponding pure enantiomers. Also, some of the compounds of Formula (I) may be atropisomers (e.g., substituted biaryls) and are considered as part of this invention. Enantiomers can also be separated by use of chiral HPLC column.
The compounds of formula 1.0 form salts that are also within the scope of this invention. Reference to a compound of formula 1.0 herein is understood to include reference to salts thereof, unless otherwise indicated. The term "salt(s)", as employed herein, denotes acidic salts formed with inorganic and/or organic acids, as well as basic salts formed with inorganic and/or organic bases. In addition, when a compound of formula 1.0 contains both a basic moiety, such as, but not limited to a pyridine or imidazole, and an acidic moiety, such as, but not limited to a carboxylic acid, zwitterions ("inner salts") may be formed and are included within the term
"salt(s)" as used herein. Pharmaceutically acceptable (i.e., non-toxic, physiologically acceptable salts) are preferred. Salts of the compounds of the formula 1.0 may be formed, for example, by reacting a compound of formula 1.0 with an amount of acid or base, such as an equivalent amount, in a medium such as one in which the salt precipitates or in an aqueous medium followed by lyophilization. Acids (and bases) which are generally considered suitable for the formation of pharmaceutically useful salts from basic (or acidic) pharmaceutical compounds are discussed, for example, by S. Berge et al, Journal of Pharmaceutical Sciences (1977) 66(1 ) 1 -19; P. Gould, International J. of Pharmaceutics (1986) 33 201 -217; Anderson et al, The Practice of Medicinal Chemistry (1996), Academic Press, New York; in The Orange Book (Food & Drug Administration, Washington, D. C. on their website); and P. Heinrich Stahl, Camille G. Wermuth (Eds.), Handbook of Pharmaceutical Salts: Properties, Selection, and Use, (2002) Int'l. Union of Pure and Applied Chemistry, pp. 330-331. These disclosures are incorporated herein by reference thereto. Exemplary acid addition salts include acetates, adipates, alginates, ascorbates, aspartates, benzoates, benzenesulfonates, bisulfates, borates, butyrates, citrates, camphorates, camphorsulfonates, cyclopentanepropionates, digluconates, dodecylsulfates, ethanesulfonates, fumarates, glucoheptanoates, glycerophosphates, hemisulfates, heptanoates, hexanoates, hydrochlorides, hydrobromides, hydroiodides, 2-hydroxyethanesulfonates, lactates, maleates, methanesulfonates, methyl sulfates, 2-naphthalenesulfonates, nicotinates, nitrates, oxalates, pamoates, pectinates, persulfates, 3-phenylpropionates, phosphates, picrates, pivalates, propionates, salicylates, succinates, sulfates, sulfonates (such as those mentioned herein), tartarates, thiocyanates, toluenesulfonates (also known as tosylates,) undecanoates, and the like.
Exemplary basic salts include ammonium salts, alkali metal salts such as sodium, lithium, and potassium salts, alkaline earth metal salts such as calcium and magnesium salts, aluminum salts, zinc salts, salts with organic bases (for example, organic amines) such as benzathines, diethylamine, dicyclohexylamines, hydrabamines (formed with N,N-bis(dehydroabietyl)ethylenediamine), N-methyl-D- glucamines, N-methyl-D-glucamides, t-butyl amines, piperazine, phenylcyclohexyl- amine, choline, tromethamine, and salts with amino acids such as arginine, lysine and the like. Basic nitrogen-containing groups may be quarternized with agents such as lower alkyl halides (e.g. methyl, ethyl, propyl, and butyl chlorides, bromides and iodides), dialkyl sulfates (e.g. dimethyl, diethyl, dibutyl, and diamyl sulfates), long chain halides (e.g. decyl, lauryl, myristyl and stearyl chlorides, bromides and iodides), aralkyl halides (e.g. benzyl and phenethyl bromides), and others.
All such acid and base salts are intended to be pharmaceutically acceptable salts within the scope of the invention and all acid and base salts are considered equivalent to the free forms of the corresponding compounds for purposes of the invention.
Compounds of formula 1.0, and salts, solvates and prodrugs thereof, may exist in their tautomeric form (for example, as an amide or imino ether). All such tautomeric forms are contemplated herein as part of the present invention.
In hetero-atom containing ring systems of this invention, there are no hydroxyl groups on carbon atoms adjacent to a N, O or S, and there are no N or S groups on carbon adjacent to another heteroatom. Thus, for example, in the ring:
Figure imgf000022_0001
there is no -OH attached directly to carbons marked 2 and 5.
The compounds of formula 1.0 may exist in different tautomeric forms, and all such forms are embraced within the scope of the invention. Also, for example, all keto-enol and imine-enamine forms of the compounds are included in the invention.
Tautomeric forms such as, for example, the moieties:
Figure imgf000022_0002
are considered equivalent in certain embodiments of this invention.
The term "substituted" means that one or more hydrogens on the designated atom is replaced with a selection from the indicated group, provided that the designated atom's normal valency under the existing circumstances is not exceeded, and that the substitution results in a stable compound. Combinations of substituents and/or variables are permissible only if such combinations result in stable compounds. By "stable compound" or "stable structure" is meant a compound that is sufficiently robust to survive isolation to a useful degree of purity from a reaction mixture, and formulation into an efficacious therapeutic agent.
The term "optionally substituted" means optional substitution with the specified groups, radicals or moieties.
The term "purified", "in purified form" or "in isolated and purified form" for a compound refers to the physical state of said compound after being isolated from a synthetic process or natural source or combination thereof. Thus, the term "purified", "in purified form" or "in isolated and purified form" for a compound refers to the physical state of said compound after being obtained from a purification process or processes described herein or well known to the skilled artisan, in sufficient purity to be characterizable by standard analytical techniques described herein or well known to the skilled artisan.
When a functional group in a compound is termed "protected", this means that the group is in modified form to preclude undesired side reactions at the protected site when the compound is subjected to a reaction. Suitable protecting groups will be recognized by those with ordinary skill in the art as well as by reference to standard textbooks such as, for example, T. W. Greene etal, Protective Groups in organic Synthesis (1991), Wiley, New York.
When any variable (e.g., aryl, heterocycle, R3, etc.) occurs more than one time in any moiety or in any compound of formula 1.0, its definition on each occurrence is independent of its definition at every other occurrence. As used herein, the term "composition" is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, directly or indirectly, from combination of the specified ingredients in the specified amounts.
The present invention also embraces isotopically-labelled compounds of the present invention which are identical to those recited herein, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, fluorine, chlorine, and iodine, such as 2H, 3H, 11C, 13C, 14C, 15N, 180, 170, 31P, 32P, 35S, 18F, 36CI, and 123I, respectively.
Certain isotopically-labelled compounds of formula 1.0 (e.g., those labeled with 3H and 14C) are useful in compound and/or substrate tissue distribution assays. Tritiated (i.e., 3H) and carbon-14 (i.e., 14C) isotopes are particularly preferred for their ease of preparation and detectability. Certain isotopically-labelled compounds of Formula (I) can be useful for medical imaging purposes. E.g., those labeled with positron-emitting isotopes like 11C or 18F can be useful for application in Positron Emission Tomography (PET) and those labeled with gamma ray emitting isotopes like 123I can be useful for application in Single photon emission computed tomography
(SPECT). Further, substitution with heavier isotopes such as deuterium (i.e., 2H) may afford certain therapeutic advantages resulting from greater metabolic stability (e.g., increased in vivo half-life or reduced dosage requirements) and hence may be preferred in some circumstances, lsotopically labeled compounds of Formula (I), in particular those containing isotopes with longer half lives (T1/2 >1 day), can generally be prepared by following procedures analogous to those disclosed in the Schemes and/or in the Examples herein below, by substituting an appropriate isotopically labeled reagent for a non-isotopically labeled reagent.
This invention provides compounds of formula 1.0:
Figure imgf000024_0001
or the pharmaceutically acceptable salts, or solvates thereof, wherein R1, R2 and Q are independently selected, and wherein:
Figure imgf000024_0002
R1 is selected from the group consisting of: heteroaryl and substituted heteroaryl, wherein said substituted heteroaryl is substituted with 1 to 3 (preferably 1 ) substituents independently selected from the group consisting of: -OH, alkoxy, and -O-alkylene-O-alkyl; R2 is selected from the group consisting of: -O-alkyl and -S-alkyl; and R5 is selected from the group consisting of:
(a) triazolyl-phenyl-,
(b) triazolyl-phenyl- wherein said phenyl is optionally substituted with 1 to 3 substituents independently selected from the group consisting of: halo (e.g., Br, Cl, F, and in one example F) and alkoxy (e.g., CrC6alkoxy, and in one example, Cr C2alkoxy, and in another example -OCH3),
(c) substituted triazolyl-phenyl- wherein said phenyl is optionally substituted with 1 to 3 substituents independently selected from the group consisting of: halo (e.g., Br, Cl, F, and in one example F) and alkoxy (e.g., CrC6alkoxy, and in one example, Ci-C2alkoxy, and in another example -OCH3), and said triazolyl group is substituted with one or two substitutents independently selected from the group consisting of: alkyl, hydroxy substituted alkyl, -alkylene-O-alkyl, and amino (i.e., -NH2),
(d) triazolyl-thienyl-, (e) triazolyl-thienyl- wherein said thienyl is optionally substituted with 1 to 2 substituents independently selected from the group consisting of: halo (e.g., Br, Cl, F, and in one example F) and alkoxy (e.g., CrCδalkoxy, and in one example, C1- C2alkoxy, and in another example -OCH3),
(f) substituted triazolyl-thienyl- wherein said thienyl is optionally substituted with 1 to 2 substituents independently selected from the group consisting of: halo (e.g., Br, Cl, F, and in one example F) and alkoxy (e.g., CrCealkoxy, and in one example, CrC2alkoxy, and in another example -OCH3), and said triazolyl group is substituted with one or two substitutents independently selected from the group consisting of: alkyl, hydroxy substituted alkyl, -alkylene-O-alkyl, and amino (i.e., -NH2), (g) triazolyl-pyridyl-,
(h) triazolyl-pyridyl- wherein said pyridyl is optionally substituted with 1 to 3 substituents independently selected from the group consisting of: halo (e.g., Br, Cl, F, and in one example F), alkyl, and alkoxy (e.g., CrC6alkoxy, and in one example, Cr C2alkoxy, and in another example -OCH3), provided that the carbon atoms adjacent to the nitrogen atom in said pyridyl are not substituted with halo, and
(i) substituted triazolyl-pyridyl- wherein: (1) said pyridyl is optionally substituted with 1 to 3 substituents independently selected from the group consisting of: halo (e.g., Br, Cl, F, and in one example F), alkyl, and alkoxy (e.g., CrCβalkoxy, and in one example, CrC2alkoxy, and in another example -OCH3), provided that the carbon atoms adjacent to the nitrogen atom in said pyridyl are not substituted with halo, and (2) said triazolyl group is substituted with one or two substitutents independently selected from the group consisting of: alkyl, hydroxy substituted alkyl, -alkylene-O-alkyl, and amino (i.e., -NH2), (j) triazolyl-thiazolyl-,
(k) triazolyl-thiazolyl- wherein said thiazolyl is optionally substituted with 1 substituent independently selected from the group consisting of: halo (e.g., Br, Cl, F, and in one example F)1 alkyl, and alkoxy (e.g., d-Cβalkoxy, and in one example, C1- C2alkoxy, and in another example -OCH3), amino (i.e., NH2), alkylamino, and dialkylamino wherein each alkyl is independently selected, and
(I) substituted triazolyl-thiazolyl- wherein (1) said thiazolyl is optionally substituted with 1 substituent independently selected from the group consisting of: halo (e.g., Br, Cl, F, and in one example F), alkyl, and alkoxy (e.g., CrC6alkoxy, and in one example, Ci-C2alkoxy, and in another example -OCH3), amino (i.e., NH2), alkylamino, and dialkylamino wherein each alkyl is independently selected, and (2) said triazolyl group is substituted with one or two substitutents independently selected from the group consisting of: alkyl, hydroxy substituted alkyl, -alkylene-O-alkyl, and amino (i.e., -NH2),
(m) pyridazinyl-thienyl-, (n) pyridazinyl-thienyl- wherein said thienyl is optionally substituted with 1 to
2 substituents independently selected from the group consisting of: halo (e.g., Br, Cl, F, and in one example F) and alkoxy (e.g., d-Cβalkoxy, and in one example, Cr C2alkoxy, and in another example -OCH3), and
(o) substituted pyridazinyl-thienyl- wherein (1) said thienyl is optionally substituted with 1 to 2 substituents independently selected from the group consisting of: halo (e.g., Br, Cl, F, and in one example F) and alkoxy (e.g., CrCβalkoxy, and in one example, CrC2alkoxy, and in another example -OCH3), and (2) said pyridazinyl group is substituted with 1 to 3 substitutents independently selected from the group consisting of: =O, alkyl, amino (i.e., -NH2), alkylamino, dialkylamino wherein each alkyl is independently selected, and halo (e.g., Br, Cl, F, and in one example F), provided that the carbon atoms adjacent to the nitrogen atoms in said pyridazinyl are not substituted with halo, and provided that when said -alkylene-O-alkyl group is bound to the nitrogen of said triazolyl in (c), (f), (i) and (I) of R5 the alkylene moiety of said -alkylene-O-alkyl group is not -CH2- (i.e., the alkylene moiety is 2 or more carbons in length). This invention provides compounds of formula 1.0:
Figure imgf000027_0001
or the pharmaceutically acceptable salts, or solvates thereof, wherein: Q is:
Figure imgf000027_0002
R1 is selected from the group consisting of: heteroaryl and substituted heteroaryl, wherein said substituted heteroaryl is substituted with 1 to 3 (preferably 1) substituents independently selected from the group consisting of: -OH, alkoxy, and -O-alkylene-O-alkyl;
R2 is selected from the group consisting of: -O-alkyl and -S-alkyl; and R5 is selected from the group consisting of: (a) triazolyl-phenyl-,
(b) triazolyl-phenyl- wherein said phenyl is optionally substituted with 1 to 3 substituents independently selected from the group consisting of: halo (e.g., Br, Cl, F, and in one example F) and alkoxy (e.g., CrC6alkoxy, and in one example, d- C2alkoxy, and in another example -OCH3), (c) substituted triazolyl-phenyl- wherein said phenyl is optionally substituted with 1 to 3 substituents independently selected from the group consisting of: halo (e.g., Br, Cl, F, and in one example F) and alkoxy (e.g., CrCβalkoxy, and in one example, Ci-C2alkoxy, and in another example -OCH3), and said triazolyl group is substituted with one or two substitutents independently selected from the group consisting of: alkyl, hydroxy substituted alkyl, -alkylene-O-alkyl, and amino (i.e., -NH2); (d) triazolyl-thienyl-, (Θ) triazolyl-thienyl- wherein said thienyl is optionally substituted with 1 to 2 substituents independently selected from the group consisting of: halo (e.g., Br, Cl, F, and in one example F) and alkoxy (e.g., CrC6alkoxy, and in one example, Cr C2alkoxy, and in another example -OCH3), and (f) substituted triazolyl-thienyl- wherein said thienyl is optionally substituted with 1 to 2 substituents independently selected from the group consisting of: halo (e.g., Br, Cl, F, and in one example F) and alkoxy (e.g., CrC6alkoxy, and in one example, Ci-C2alkoxy, and in another example -OCH3), and said triazolyl group is substituted with one or two substitutents independently selected from the group consisting of: alkyl, hydroxy substituted alkyl, -alkylene-O-alkyl, and amino (i.e., -NH2), and provided that when said -alkylene-O-alkyl group is bound to the nitrogen of said triazolyl in (c) and (f) of R5 the alkylene moiety of said -alkylene-O-alkyl group is not -CH2- (i.e., the alkylene moiety is 2 or more carbons in length). Those skilled in the art will appreciate that the term "alkylene", as used in the substituents -O-alkylene-O-alkyl and -alkylene-O-alkyl, means a divalent saturated hydrocarbon group. Thus, an example of an alkylene moiety is -CH2-CH2-, and an example of an -O-alkylene-O-alkyl moiety is -O-(CH2)2-O-CH3, and an example of an -alkylene-O-alkyl moiety is -(CH2)2-O-CH3. Those skilled in the art will also appreciate that the term alkylene also includes the moiety -CH2-.
Examples of the R1 heteroaryl group include, but are not limited to, pyridyl, pyrrolyl, pyrazolyl, imidazolyl, furanyl, thienyl, thiazolyl, pyridyl N-O, and pyrimidinyl. Examples of the R1 substitued heteroaryl group include, but are not limited to, substituted pyridyl, substituted pyrrolyl, substituted pyrazolyl, substituted imidazolyl, substituted furanyl, substituted thienyl, substituted thiazolyl, substituted pyridyl N-O, and substituted pyrimidinyl.
In one embodiment of this invention R1 is pyridyl. In another embodiment of this invention R1 is substituted pyridyl. In another embodiment of this invention R1 is pyridyl substituted with one substitutent.
The substitutents on the substituted R1 groups (e.g., the substituted pyridyl) are independently selected from the group consisting of: -OH, alkoxy, and -O-alkylene-O-alkyl. Examples of the alkoxy group include, for example, Ci to C6alkoxy (such as, for example, -0-CH3, -0-C2H5, and -O-CH(CH3)2). Examples of the -O-alkylene-O-alkyl group include, for example, -O-(Ci-C4)alkylene-O-(CrC6)alkyl, -O-(CrC2)alkylene-O-(CrC3alkyl), and -0-(CH2)Z-O-CH3).
Examples of R1 include, for example,
Figure imgf000029_0001
In one embodiment of this invention R1 is pyridyl substituted with alkoxy. In another embodiement of this invention R1 is substituted with -OCH(CH3)2. In another embodiement of this invention R1 is substituted with -OC2H5. In another embodiment of this invention R1 is:
Figure imgf000029_0002
In another embodiment of this invention R1 is:
Figure imgf000029_0003
In another embodiment of this invention R1 is substituted with -O-alkylene-O-alkyl.
In another embodiment of this invention R1 is substituted with -OCH2CH2OCH3.
In another embodiment of this invention R1 is:
.
Figure imgf000029_0004
Examples of the R2 -O-alkyl group include, for example, -O-(CrC6)alkyl,
-O-(CrC2)alkyl, and -OCH3.
Examples of the R2 -S-alkyl group include, for example, -S-(CrC6)alkyl, -S-(C1 -C2)alkyl. and -SCH3.
In one embodiment of this invention R2 is a -O-(C-i-C2)alkyl group. In another embodiment of this invention R2 is -OCH3.
In another embodiment of this invention R2 is a -S-(Ci -C2)alkyl group.
In another embodiment of this invention R2 is -SCH3.
In one embodiment of this invention R5 is a triazolyl-phenyl moiety wherein the triazolyl moiety is bonded to the phenyl moiety by a ring carbon of the triazolyl moiety
IInn oonnee embodiment of this invention R5 is a triazolyl-phenyl- moiety, such as, for example,
Figure imgf000030_0001
In another embodiment of this invention R5 is a triazolyl-thienyl- moiety, such as, for example,
Figure imgf000030_0002
In another embodiment of this invention R5 is a triazolyl-thienyl- moiety, such as, for example,
Figure imgf000030_0003
.
In another embodiment of this invention the substituted triazolyl moiety of said R5 group is substituted on the ring nitrogen.
When the triazolyl moiety of R5 is substituted with alkyl, examples of the alkyl groups include, for example, -CrC6alkyl, -CrC4alkyl, -CrC2alkyl, and -CH3. And in one embodiment, there is alkyl substitution on the triazolyl moiety of R5 and said alkyl is -CH3. When the triazolyl moiety of R5 is substituted with -alkylene-O-alkyl groups, examples of the -alkylene-O-alkyl groups include, for example, -Ci-C4alkylene-O-Cr C6alkyl, -Ci-C2alkylene-O-Ci-C2alkyl, -d-C-jalkylene-O-CHs, and -CH2CH2OCH3. And in one embodiment, there is -alkylene-O-alkyl substitution on the triazolyl moiety of R5 and said -alkylene-O-alkyl is -CH2CH2OCH3. When the nitrogen of the triazolyl moiety of R5 is substituted with -alkylene-O-alkyl group, examples of the -alkylene-O- alkyl group includes, for example, -C2-C4alkylene-O-CrC6alkyl, -C2alkylene-O-Cr C2alkyl, -C2-C4alkylene-O-CH3, and -CH2CH2OCH3. And in one embodiment, there is -alkylene-O-alkyl substitution on the nitrogen of the triazolyl moiety of R5 and said -alkylene-O-alkyl is -CH2CH2OCH3.
When the triazolyl moiety of R5 is substituted with hydroxy substituted alkyl groups, examples of the hydroxy substituted alkyl groups include, for example, hydroxy substituted -CrC4alkyl, hydroxy substituted -Ci-C2alkyl, and hydroxy substituted -CH3. Examples also include, for example, -CH2COH(CH3)2, and -CH2CH2OH.
When the phenyl moiety of R5 is substituted with halo atoms, examples of the halo atoms include, for example, Cl, F and Br. In one embodiment of this invention the halo on the phenyl is F. In another embodiment of this invention the phenyl is substituted with one F. In one embodiment of this invention R5 is a substituted triazolyl-phenyl- wherein said triazolyl is substituted and said phenyl is unsubstituted.
In another embodiment of this invention R5 is a substituted triazolyl-phenyl- wherein said triazolyl is substituted on the nitrogen and said phenyl is unsubstituted.
In another embodiment of this invention R5 is a substituted triazolyl-phenyl- wherein said triazolyl is substituted on the carbon and said phenyl is unsubstituted. In another embodiment of this invention R5 is a substituted triazolyl-phenyl- wherein said triazolyl is substituted on the nitrogen and on the carbon, and said phenyl is unsubstituted.
In another embodiment of this invention R5 is a substituted triazolyl-phenyl- wherein said triazolyl is substituted and said phenyl is substituted.
In another embodiment of this invention R5 is a substituted triazolyl-phenyl- wherein said triazolyl is substituted on the nitrogen and said phenyl is substituted. In another embodiment of this invention R5 is a substituted triazolyl-phenyl- wherein said triazolyl is substituted on the carbon and said phenyl is substituted. In another embodiment of this invention R5 is a substituted triazolyl-phenyl- wherein said triazolyl is substituted on the nitrogen and on the carbon, and said phenyl is substituted.
In another embodiment of this invention R5 is a substituted triazolyl-phenyl- wherein said triazolyl is unsubstituted and said phenyl is substituted.
In another embodiment of this invention R5 is an unsubstituted triazolyl-phenyk
In another embodiment of this invention R5 is a substituted triazolyl-phenyl- wherein the triazolyl is substituted on the nitrogen with -CH2COH(CH3)2.
In another embodiment of this invention R5 is a substituted triazolyl-phenyl- wherein the triazolyl is substituted on the nitrogen with -CH2CH2OH.
In another embodiment of this invention R5 is a substituted triazolyl-phenyl- wherein the triazolyl is substituted on the nitrogen with an alkyl group.
In another embodiment of this invention R5 is a substituted triazolyl-phenyl- wherein the triazolyl is substituted on the nitrogen with an alkyl group and substituted on the carbon with an alkyl group, wherein each alkyl group is independently selected.
In another embodiment of this invention R5 is a substituted triazolyl-phenyl- wherein the triazolyl is substituted on the nitrogen with a -CH3 group.
In another embodiment of this invention R5 is a substituted triazolyl-phenyl- wherein the triazolyl is substituted on the nitrogen with a -CH3 group and on the carbon with a -CH3 group.
In another embodiment of this invention R5 moiety is a substituted triazolyl- phenyl- wherein the triazolyl is substituted on the carbon with a -NH2 group.
In another embodiment of this invention R5 is a substituted triazolyl-phenyl- wherein the triazolyl is substituted on the nitrogen with an -alkylene-O-alkyl group. In another embodiment of this invention R5 is a substituted triazolyl-phenyl- wherein the triazolyl is substituted on the nitrogen with a -CH2CH2OCH3 group.
In another embodiment of this invention R5 moiety is a substituted triazolyl- phenyl- wherein said phenyl moiety is substituted with halo, and said triazolyl moiety is substituted as described in any of the above embodiments. In another embodiment of this invention R5 is a substituted triazolyl-phenyl- wherein said phenyl moiety is substituted with one halo, and said triazolyl moiety is substituted as described in any one of the above embodiments. In another embodiment of this invention R5 is a substituted triazolyl-phenyl- wherein said phenyl moiety is substituted with F, and said triazolyl moiety is substituted as described in any one of the above embodiments.
In another embodiment of this invention R5 is a substituted triazolyl-phenyl- wherein said phenyl moiety is substituted with one F, and said triazolyl moiety is substituted as described in any one of the above embodiments.
In another embodiment of this invention R5 is a substituted triazolyl-phenyl- group wherein said phenyl moiety is substituted with one F, and said triazolyl moiety is substituted on the nitrogen with a hydroxyl substituted alkyl group. In another embodiment of this invention R5 is a substituted triazolyl-phenyl- wherein said phenyl moiety is substituted with one F, and said triazolyl moiety is substituted on the nitrogen with a -CH2CH2OH group.
In another embodiment of this invention R5 is a substituted triazolyl-phenyl- wherein said phenyl moiety is substituted with one F, and said triazolyl moiety is substituted on the nitrogen with a -alkylene-O-alkyl group.
In another embodiment of this invention R5 is a substituted triazolyl-phenyl- wherein said phenyl moiety is substituted with one F, and said triazolyl moiety is substituted on the nitrogen with a -CH2CH2OCH3 group.
In another embodiment of this invention R5 is a substituted triazolyl-phenyl- wherein said phenyl moiety is substituted with halo, and said triazolyl moiety is unsubstituted.
In another embodiment of this invention R5 is a substituted triazolyl-phenyl- wherein said phenyl moiety is substituted with one halo, and said triazolyl moiety is unsubstituted. In another embodiment of this invention R5 is a substituted triazolyl-phenyl- wherein said phenyl moiety is substituted with F, and said triazolyl moiety is unsubstituted.
In another embodiment of this invention R5 is a substituted triazolyl-phenyl- wherein said phenyl moiety is substituted with one F, and said triazolyl moiety is unsubstituted.
In another embodiment of this invention R5 moiety is a substituted triazolyl- phenyl- wherein said phenyl moiety is substituted with alkoxy, and said triazolyl moiety is substituted as described in any of the above embodiments. In another embodiment of this invention R5 is a substituted triazolyl-phenyl- wherein said phenyl moiety is substituted with one alkoxy, and said triazolyl moiety is substituted as described in any one of the above embodiments.
In another embodiment of this invention R5 is a substituted triazolyl-phenyl- wherein said phenyl moiety is substituted with -OCH3, and said triazolyl moiety is substituted as described in any one of the above embodiments.
In another embodiment of this invention R5 is a substituted triazolyl-phenyl- wherein said phenyl moiety is substituted with one -OCH3, and said triazolyl moiety is substituted as described in any one of the above embodiments. In another embodiment of this invention R5 is a substituted triazolyl-phenyl- group wherein said phenyl moiety is substituted with one -OCH3, and said triazolyl moiety is substituted on the nitrogen with a hydroxyl substituted alkyl group.
In another embodiment of this invention R5 is a substituted triazolyl-phenyl- wherein said phenyl moiety is substituted with one -OCH3, and said triazolyl moiety is substituted on the nitrogen with a -CH2CH2OH group.
In another embodiment of this invention R5 is a substituted triazolyl-phenyl- wherein said phenyl moiety is substituted with one -OCH3, and said triazolyl moiety is substituted on the nitrogen with a -alkylene-O-alkyl group.
In another embodiment of this invention R5 is a substituted triazolyl-phenyl- wherein said phenyl moiety is substituted with one -OCH3, and said triazolyl moiety is substituted on the nitrogen with a -CH2CH2OCH3 group.
In another embodiment of this invention R5 is a substituted triazolyl-phenyl- wherein said phenyl moiety is substituted with alkoxy, and said triazolyl moiety is unsubstituted. In another embodiment of this invention R5 is a substituted triazolyl-phenyl- wherein said phenyl moiety is substituted with one alkoxy, and said triazolyl moiety is unsubstituted.
In another embodiment of this invention R5 is a substituted triazolyl-phenyl- wherein said phenyl moiety is substituted with with -OCH3, and said triazolyl moiety is unsubstituted.
In another embodiment of this invention R5 is a substituted triazolyl-phenyl- wherein said phenyl moiety is substituted with one -OCH3, and said triazolyl moiety is unsubstituted.
In another embodiment of this invention R5 is:
Figure imgf000035_0001
In another embodiment of this invention R ,5 : is,
Figure imgf000035_0002
In another embodiment of this invention R5 is:
Figure imgf000035_0003
In another embodiment of this invention R is:
Figure imgf000035_0004
In another embodiment of this invention R 5 : is,
Figure imgf000036_0001
In another embodiment of this invention R 5 : is„
Figure imgf000036_0002
In another embodiment of this invention R .5 : is,
Figure imgf000036_0003
In another embodiment of this invention R 5 : is,
Figure imgf000036_0004
In another embodiment of this invention R is:
Figure imgf000036_0005
In another embodiment of this invention R 5 : is,
Figure imgf000037_0001
In another embodiment of this invention R5 is:
Figure imgf000037_0002
In another embodiment of this invention R5 is a substituted triazolyl-thienyl- wherein said triazolyl is substituted and said thienyl is unsubstituted.
In another embodiment of this invention R5 is a substituted triazolyl-thienyl- wherein said triazolyl is substituted on the nitrogen and said thienyl is unsubstituted.
In another embodiment of this invention R5 is a substituted triazolyl-thienyl- wherein said triazolyl is substituted on the carbon and said thienyl is unsubstituted. In another embodiment of this invention R5 is a substituted triazolyl-thienyl- wherein said triazolyl is substituted on the nitrogen and on the carbon, and said thienyl is unsubstituted.
In another embodiment of this invention R5 is a substituted triazolyl-thienyl- wherein said triazolyl is substituted and said thienyl is substituted. In another embodiment of this invention R5 is a substituted triazolyl-thienyl- wherein said triazolyl is substituted on the nitrogen and said thienyl is substituted. In another embodiment of this invention R5 is a substituted triazolyl-thienyl- wherein said triazolyl is substituted on the carbon and said thienyl is substituted.
In another embodiment of this invention R5 is a substituted triazolyl-thienyl- wherein said triazolyl is substituted on the nitrogen and on the carbon, and said thienyl is substituted.
In another embodiment of this invention R5 is a substituted triazolyl-thienyl- wherein said triazolyl is unsubstituted and said thienyl is substituted. In another embodiment of this invention R5 is an unsubstituted tiϊazolyl-thienyk
In another embodiment of this invention R5 is a substituted triazolyl-thienyl- wherein the triazolyl is substituted on the nitrogen with -CH2COH(CH3)2.
In another embodiment of this invention R5 is a substituted triazolyl-thienyl- wherein the triazolyl is substituted on the nitrogen with -CH2CH2OH.
In another embodiment of this invention R5 is a substituted triazolyl-thienyl- wherein the triazolyl is substituted on the nitrogen with an alkyl group.
In another embodiment of this invention R5 is a substituted triazolyl-thienyl- wherein the triazolyl is substituted on the nitrogen with an alkyl group and substituted on the carbon with an alkyl group, wherein each alkyl group is independently selected.
In another embodiment of this invention R5 is a substituted triazolyl-thienyl- wherein the triazolyl is substituted on the nitrogen with a -CH3 group.
In another embodiment of this invention R5 is a substituted triazolyl-thienyl- wherein the triazolyl is substituted on the nitrogen with a -CH3 group and on the carbon with a -CH3 group.
In another embodiment of this invention R5 moiety is a substituted triazolyl- thienyl- wherein the triazolyl is substituted on the carbon with a -NH2 group.
In another embodiment of this invention R5 is a substituted triazolyl-thienyl- wherein the triazolyl is substituted on the nitrogen with an -alkylene-O-alkyl group. In another embodiment of this invention R5 is a substituted triazolyl-thienyl- wherein the triazolyl is substituted on the nitrogen with a -CH2CH2OCH3 group.
In another embodiment of this invention R5 moiety is a substituted triazolyl- thienyl- wherein said thienyl moiety is substituted with halo, and said triazolyl moiety is substituted as described in any of the above embodiments. In another embodiment of this invention R5 is a substituted triazolyl-thienyl- wherein said thienyl moiety is substituted with one halo, and said triazolyl moiety is substituted as described in any one of the above embodiments.
In another embodiment of this invention R5 is a substituted triazolyl-thienyl- wherein said thienyl moiety is substituted with F, and said triazolyl moiety is substituted as described in any one of the above embodiments.
In another embodiment of this invention R5 is a substituted triazolyl-thienyl- wherein said thienyl moiety is substituted with one F, and said triazolyl moiety is substituted as described in any one of the above embodiments. In another embodiment of this invention R5 is a substituted triazolyl-thienyl- group wherein said thienyl moiety is substituted with one F, and said triazolyl moiety is substituted on the nitrogen with a hydroxyl substituted alkyl group.
In another embodiment of this invention R5 is a substituted triazolyl-thienyl- wherein said thienyl moiety is substituted with one F, and said triazolyl moiety is substituted on the nitrogen with a -CH2CH2OH group.
In another embodiment of this invention R5 is a substituted triazolyl-thienyl- wherein said thienyl moiety is substituted with one F, and said triazolyl moiety is substituted on the nitrogen with a -alkylene-O-alkyl group. In another embodiment of this invention R5 is a substituted triazolyl-thienyl- wherein said thienyl moiety is substituted with one F, and said triazolyl moiety is substituted on the nitrogen with a -CH2CH2OCH3 group.
In another embodiment of this invention R5 is a substituted triazolyl-thienyl- wherein said thienyl moiety is substituted with halo, and said triazolyl moiety is unsubstituted.
In another embodiment of this invention R5 is a substituted triazolyl-thienyl- wherein said thienyl moiety is substituted with one halo, and said triazolyl moiety is unsubstituted.
In another embodiment of this invention R5 is a substituted triazolyl-thienyl- wherein said thienyl moiety is substituted with F, and said triazolyl moiety is unsubstituted.
In another embodiment of this invention R5 is a substituted triazolyl-thienyl- wherein said thienyl moiety is substituted with one F, and said triazolyl moiety is unsubstituted. In another embodiment of this invention R5 moiety is a substituted triazolyl- thienyl- wherein said thienyl moiety is substituted with alkoxy, and said triazolyl moiety is substituted as described in any of the above embodiments.
In another embodiment of this invention R5 is a substituted triazolyl-thienyl- wherein said thienyl moiety is substituted with one alkoxy, and said triazolyl moiety is substituted as described in any one of the above embodiments.
In another embodiment of this invention R5 is a substituted triazolyl-thienyl- wherein said thienyl moiety is substituted with -OCH3, and said triazolyl moiety is substituted as described in any one of the above embodiments. In another embodiment of this invention R5 is a substituted triazolyl-thienyl- wherein said thienyl moiety is substituted with one -OCH3, and said triazolyl moiety is substituted as described in any one of the above embodiments.
In another embodiment of this invention R5 is a substituted triazolyl-thienyl- group wherein said thienyl moiety is substituted with one -OCH3, and said triazolyl moiety is substituted on the nitrogen with a hydroxyl substituted alkyl group.
In another embodiment of this invention R5 is a substituted triazolyl-thienyl- wherein said thienyl moiety is substituted with one -OCH3, and said triazolyl moiety is substituted on the nitrogen with a -CH2CH2OH group. In another embodiment of this invention R5 is a substituted triazolyl-thienyl- wherein said thienyl moiety is substituted with one -OCH3, and said triazolyl moiety is substituted on the nitrogen with a -alkylene-O-alkyl group.
In another embodiment of this invention R5 is a substituted triazolyl-thienyl- wherein said thienyl moiety is substituted with one -OCH3, and said triazolyl moiety is substituted on the nitrogen with a -CH2CH2OCH3 group.
In another embodiment of this invention R5 is a substituted triazolyl-thienyl- wherein said thienyl moiety is substituted with alkoxy, and said triazolyl moiety is unsubstituted.
In another embodiment of this invention R5 is a substituted triazolyl-thienyl- wherein said thienyl moiety is substituted with one alkoxy, and said triazolyl moiety is unsubstituted.
In another embodiment of this invention R5 is a substituted triazolyl-thienyl- wherein said thienyl moiety is substituted with -OCH3, and said triazolyl moiety is unsubstituted. In another embodiment of this invention R5 is a substituted triazolyl-thienyl- wherein said thienyl moiety is substituted with one -OCH3, and said triazolyl moiety is unsubstituted.
In another embodiment of this invention R5 is triazolyl-pyridyl-.
In another embodiment of this invention R5 is
Figure imgf000040_0001
In another embodiment of this invention R is a substituted triazolyl-pyridyl- wherein said triazolyl is substituted and said pyridyl is unsubstituted. In another embodiment of this invention R5 is a substituted triazolyl-pyridyl- wherein said triazolyl is substituted on the nitrogen and said pyridyl is unsubstituted.
In another embodiment of this invention R5 is a substituted triazolyl-pyridyl- wherein said triazolyl is substituted on the carbon and said pyridyl is unsubstituted. In another embodiment of this invention R5 is a substituted triazolyl-pyridyl- wherein said triazolyl is substituted on the nitrogen and on the carbon, and said pyridyl is unsubstituted.
In another embodiment of this invention R5 is a substituted triazolyl-pyridyl- wherein said triazolyl is substituted and said pyridyl is substituted. In another embodiment of this invention R5 is a substituted triazolyl-pyridyl- wherein said triazolyl is substituted on the nitrogen and said pyridyl is substituted.
In another embodiment of this invention R5 is a substituted triazolyl-pyridyl- wherein said triazolyl is substituted on the carbon and said pyridyl is substituted.
In another embodiment of this invention R5 is a substituted triazolyl-pyridyl- wherein said triazolyl is substituted on the nitrogen and on the carbon, and said pyridyl is substituted.
In another embodiment of this invention R5 is a substituted triazolyl-pyridyl- wherein said triazolyl is unsubstituted and said pyridyl is substituted.
In another embodiment of this invention R5 is a substituted triazolyl-pyridyl- wherein the triazolyl is substituted on the nitrogen with alkyl.
When the pyridyl moiety of R5 is substituted with alkyl, examples of the alkyl groups include, for example, -CrC6alkyl, -CrC4alkyl, -CrC2alkyl, and -CH3.
When the pyridyl moiety of R5 is substituted with halo atoms, examples of the halo atoms include, for example, Cl, F and Br, provided that the carbon atoms adjacent to the nitrogen atom in said pyridyl are not substituted with halo. In one embodiment of this invention the halo on the pyridyl is F. In another embodiment of this invention the pyridyl is substituted with one F.
In another embodiment of this invention R5 is a substituted triazolyl-pyridyl- wherein the triazolyl is substituted on the nitrogen with -CH2COH (CH3)2, and the pyridyl is unsubstituted.
In another embodiment of this invention R5 is a substituted triazolyl-pyridyl- wherein the triazolyl is substituted on the nitrogen with -CH2CH2OH, and the pyridyl is unsubstituted. In another embodiment of this invention R5 is a substituted triazolyl-pyridyl- wherein the triazolyl is substituted on the nitrogen with an alkyl group, and the pyridyl is unsubstituted.
In another embodiment of this invention R5 is a substituted triazolyl-pyridyl- wherein the triazolyl is substituted on the nitrogen with an alkyl group and substituted on the carbon with an alkyl group, wherein each alkyl group is independently selected, and the pyridyl is unsubstituted.
In another embodiment of this invention R5 is a substituted triazolyl-pyridyl- wherein the triazolyl is substituted on the nitrogen with a -CH3 group, and the pyridyl is unsubstituted.
In another embodiment of this invention R5 is a substituted triazolyl-pyridyl- wherein the triazolyl is substituted on the nitrogen with a -CH3 group and on the carbon with a -CH3 group, and the pyridyl is unsubstituted.
In another embodiment of this invention R5 moiety is a substituted triazolyl- pyridyl- wherein the triazolyl is substituted on the carbon with a -NH2 group, and the pyridyl is unsubstituted.
In another embodiment of this invention R5 is a substituted triazolyl-pyridyl- wherein the triazolyl is substituted on the nitrogen with an -alkylene-O-alkyl group, and the pyridyl is unsubstituted, and provided that the alkylene moiety of said -alkylene-O-alkyl group is not -CH2- (i.e., the alkylene moiety is 2 or more carbons in length).
In another embodiment of this invention R5 is a substituted triazolyl-pyridyl- wherein the triazolyl is substituted on the nitrogen with a -CH2CH2OCH3 group, and the pyridyl is unsubstituted. In another embodiment of this invention R5 moiety is a substituted triazolyl- pyridyl- wherein said pyridyl moiety is substituted with halo, provided that the carbon atoms adjacent to the nitrogen atom in said pyridyl are not substituted with halo, and said triazolyl moiety is substituted as described in any of the above embodiments describing substituted triazolyl groups. In another embodiment of this invention R5 is a substituted triazolyl-pyridyl- wherein said pyridyl moiety is substituted with one halo, provided that a carbon atom adjacent to the nitrogen atom in said pyridyl is not substituted with said halo, and said triazolyl moiety is substituted as described in any one of the above embodiments, describing triazolyl groups. In another embodiment of this invention R5 is a substituted triazolyl-pyridyl- wherein said pyridyl moiety is substituted with F, provided that a carbon atom adjacent to the nitrogen atom in said pyridyl is not substituted with said F, and said triazolyl moiety is substituted as described in any one of the above embodiments, describing triazolyl groups.
In another embodiment of this invention R5 is a substituted triazolyl-pyridyl- wherein said pyridyl moiety is substituted with one F, provided that a carbon atom adjacent to the nitrogen atom in said pyridyl is not substituted with said F, and said triazolyl moiety is substituted as described in any one of the above embodiments describing triazolyl groups.
In another embodiment of this invention R5 is a substituted triazolyl-pyridyl- group wherein said pyridyl moiety is substituted with one F, provided that a carbon atom adjacent to the nitrogen atom in said pyridyl is not substituted with said F, and said triazolyl moiety is substituted on the nitrogen with a hydroxyl substituted alkyl group.
In another embodiment of this invention R5 is a substituted triazolyl-pyridyl- wherein said pyridyl moiety is substituted with one F, provided that a carbon atom adjacent to the nitrogen atom in said pyridyl is not substituted with said F, and said triazolyl moiety is substituted on the nitrogen with a -CH2CH2OH group. In another embodiment of this invention R5 is a substituted triazolyl-pyridyl- wherein said pyridyl moiety is substituted with one F, provided that a carbon atom adjacent to the nitrogen atom in said pyridyl is not substituted with said F, and said triazolyl moiety is substituted on the nitrogen with an -alkylene-O-alkyl group, and the alkylene moiety of said -alkylene-O-alkyl group is not -CH2- (i.e., the alkylene moiety is 2 or more carbons in length).
In another embodiment of this invention R5 is a substituted triazolyl-pyridyl- wherein said pyridyl moiety is substituted with one F, provided that a carbon atom adjacent to the nitrogen atom in said pyridyl is not substituted with said F, and said triazolyl moiety is substituted on the nitrogen with a -CH2CH2OCH3 group. In another embodiment of this invention R5 is a substituted triazolyl-pyridyl- wherein said pyridyl moiety is substituted with halo, provided that the carbon atoms adjacent to the nitrogen atom in said pyridyl are not substituted with halo, and said triazolyl moiety is unsubstituted. In another embodiment of this invention R5 is a substituted triazolyl-pyridyl- wherein said pyridyl moiety is substituted with one halo, provided that a carbon atom adjacent to the nitrogen atom in said pyridyl is not substituted with said halo, and said triazolyl moiety is unsubstituted. In another embodiment of this invention R5 is a substituted triazolyl-pyridyl- wherein said pyridyl moiety is substituted with F, provided that a carbon atom adjacent to the nitrogen atom in said pyridyl is not substituted with said F, and said triazolyl moiety is unsubstituted.
In another embodiment of this invention R5 is a substituted triazolyl-pyridyl- wherein said pyridyl moiety is substituted with one F, provided that a carbon atom adjacent to the nitrogen atom in said pyridyl is not substituted with said F, and said triazolyl moiety is unsubstituted.
In another embodiment of this invention R5 moiety is a substituted triazolyl- pyridyl- wherein said pyridyl moiety is substituted with alkoxy, and said triazolyl moiety is substituted as described in any of the above embodiments describing substituted triazolyl groups.
In another embodiment of this invention R5 is a substituted triazolyl-pyridyl- wherein said pyridyl moiety is substituted with one alkoxy, and said triazolyl moiety is substituted as described in any one of the above embodiments describing substituted triazolyl groups.
In another embodiment of this invention R5 is a substituted triazolyl-pyridyl- wherein said pyridyl moiety is substituted with -OCH3, and said triazolyl moiety is substituted as described in any one of the above embodiments describing substituted triazolyl groups. In another embodiment of this invention R5 is a substituted triazolyl-pyridyl- wherein said pyridyl moiety is substituted with one -OCH3, and said triazolyl moiety is substituted as described in any one of the above embodiments describing substituted triazoly groups.
In another embodiment of this invention R5 is a substituted triazolyl-pyridyl- group wherein said pyridyl moiety is substituted with one -OCH3, and said triazolyl moiety is substituted on the nitrogen with a hydroxyl substituted alkyl group.
In another embodiment of this invention R5 is a substituted triazolyl-pyridyl- wherein said pyridyl moiety is substituted with one -OCH3, and said triazolyl moiety is substituted on the nitrogen with a -CH2CH2OH group. In another embodiment of this invention R5 is a substituted triazolyl-pyridyl- wherein said pyridyl moiety is substituted with one -OCH3, and said triazolyl moiety is substituted on the nitrogen with a -alkylene-O-alkyl group, and the alkylene moiety of said -alkylene-O-alkyl group is not -CH2- (i.e., the alkylene moiety is 2 or more carbons in length).
In another embodiment of this invention R5 is a substituted triazolyl-pyridyl- wherein said pyridyl moiety is substituted with one -OCH3, and said triazolyl moiety is substituted on the nitrogen with a -CH2CH2OCH3 group.
In another embodiment of this invention R5 is a substituted triazolyl-pyridyl- wherein said pyridyl moiety is substituted with alkoxy, and said triazolyl moiety is unsubstituted.
In another embodiment of this invention R5 is a substituted triazolyl-pyridyl- wherein said pyridyl moiety is substituted with one alkoxy, and said triazolyl moiety is unsubstituted. In another embodiment of this invention R5 is a substituted triazolyl-pyridyl- wherein said pyridyl moiety is substituted with -OCH3, and said triazolyl moiety is unsubstituted.
In another embodiment of this invention R5 is a substituted triazolyl-pyridyl- wherein said pyridyl moiety is substituted with one -OCH3, and said triazolyl moiety is unsubstituted.
In another embodiment of this invention R5 is a substituted triazolyl-pyridyl- wherein said triazolyl is substituted with 1 or 2 groups indepenendently selected from the group consisting of: (a) hydroxyl substituted alkyl group (e.g., -CH2COH(CH3)2) and -CH2CH2OH), (b) alkyl (e.g., -CrC6alkyl, -CrC4alkyl, -CrC2alkyl, and -CH3), (c) -NH2, and (d) -alkylene-O-alkyl (e.g., -CH2CH2OCH3), provided that the alkylene moiety of said -alkylene-O-alkyl group is not -CH2- (i.e., the alkylene moiety is 2 or more carbons in length) when said -alkylene-O-alkyl group is bound to the nitrogen of said triazolyl; and said pyridyl is substituted with 1 to 3 groups independently selected from the group consisting of: (a) alkyl (e.g., -Ci-C6alkyl, -CrC4alkyl, -CrC2alkyl, and -CH3), (b) halo (e.g., Cl, F and Br) and provided that carbon atoms adjacent to the nitrogen atom in said pyridyl are not substituted with halo, and (c) alkoxy (e.g., -OCH3).
In another embodiment of this invention R5 is
Figure imgf000046_0001
In another embodiment of this invention R is triazolyl-thiazolyl-. In another embodiment of this invention R5 is
Figure imgf000046_0002
In another embodiment of this invention R5 is a substituted triazolyl-thiazolyl- wherein said triazolyl is substituted and said thiazolyl is unsubstituted.
In another embodiment of this invention R5 is a substituted triazolyl-thiazolyl- wherein said triazolyl is substituted on the nitrogen and said thiazolyl is unsubstituted.
In another embodiment of this invention R5 is a substituted triazolyl-thiazolyl- wherein said triazolyl is substituted on the carbon and said thiazolyl is unsubstituted. In another embodiment of this invention R5 is a substituted triazolyl-thiazolyl- wherein said triazolyl is substituted on the nitrogen and on the carbon, and said thiazolyl is unsubstituted.
When the thiazolyl moiety of R5 is substituted with alkyl, examples of the alkyl groups include, for example, -Ci-C6alkyl, -Ci-C4alkyl, -CrC2alkyl, and -CH3.
When the thiazolyl moiety of R5 is substituted with halo atoms, examples of the halo atoms include, for example, Cl, F and Br. In one embodiment of this invention the halo on the thiazolyl is F. In another embodiment of this invention the thiazolyl is substituted with one F. When the thiazolyl moiety of R5 is substituted with an alkylamino group, examples of the alkylamino group include, for example, Ci-C6alkyl-NH-, CrC2alkyl-NH-, CH3-NH-, and CH3CH2-NH-.
When the thiazolyl moiety of R5 is substituted with a dialkylamino group, examples of the dialkylamino group include, for example, (CrC6alkyl)2-N- wherein each alkyl is independently selected, (CrC2alkyl)2-N- wherein each alkyl is independently selected, (CHg)2N-, (CH3CH2)2-N-, and (CH3)(CH3CH2)N-.
In another embodiment of this invention R5 is a substituted triazolyl-thiazolyl- wherein said triazolyl is substituted and said thiazolyl is substituted.
In another embodiment of this invention R5 is a substituted triazolyl-thiazolyl- wherein said triazolyl is substituted on the nitrogen and said thiazolyl is substituted. In another embodiment of this invention R5 is a substituted triazolyl-thiazolyl- wherein said triazolyl is substituted on the carbon and said thiazolyl is substituted.
In another embodiment of this invention R5 is a substituted triazolyl-thiazolyl- wherein said triazolyl is substituted on the nitrogen and on the carbon, and said thiazolyl is substituted.
In another embodiment of this invention R5 is a substituted triazolyl-thiazolyl- wherein said triazolyl is unsubstituted and said thiazolyl is substituted.
In another embodiment of this invention R5 is a substituted triazolyl-thiazolyl- wherein the triazolyl is substituted on the nitrogen with alkyl, and said thiazolyl is unsubstituted.
In another embodiment of this invention R5 is a substituted triazolyl-thiazoly- wherein the triazolyl is substituted on the nitrogen with -CH2COH(CH3)2, and said thiazolyl is unsubstituted.
In another embodiment of this invention R5 is a substituted triazolyl-thiazolyl- wherein the triazolyl is substituted on the nitrogen with -CH2CH2OH, and said thiazolyl is unsubstituted.
In another embodiment of this invention R5 is a substituted triazolyl-thiazolyl- wherein the triazolyl is substituted on the nitrogen with an alkyl group and substituted on the carbon with an alkyl group, wherein each alkyl group is independently selected, and said thiazolyl is unsubstituted.
In another embodiment of this invention R5 is a substituted triazolyl-thiazolyl- wherein the triazolyl is substituted on the nitrogen with a -CH3 group, and said thiazolyl is unsubstituted.
In another embodiment of this invention R5 is a substituted triazolyl-thiazolyl- wherein the triazolyl is substituted on the nitrogen with a -CH3 group and on the carbon with a -CH3 group, and said thiazolyl is unsubstituted.
In another embodiment of this invention R5 moiety is a substituted triazolyl- thiazolyl- wherein the triazolyl is substituted on the carbon with a -MH2 group, and said thiazolyl is unsubstituted. In another embodiment of this invention R5 is a substituted triazolyl-thiazolyl- wherein the triazolyl is substituted on the nitrogen with an -alkylene-O-alkyl group, provided that the alkylene moiety of said -alkylene-O-alkyl group is not -CH2- (i.e., the alkylene moiety is 2 or more carbons in length), and said thiazolyl is unsubstituted. In another embodiment of this invention R5 is a substituted triazolyl-thiazolyl- wherein the triazolyl is substituted on the nitrogen with a -CH2CH2OCH3 group, and said thiazolyl is unsubstituted.
In another embodiment of this invention R5 moiety is a substituted triazolyl- thiazolyl- wherein said thiazolyl moiety is substituted with halo, and said triazolyl moiety is substituted as described in any of the above embodiments describing substituted triazolyl groups.
In another embodiment of this invention R5 is a substituted triazolyl-thiazolyl- wherein said thiazolyl moiety is substituted with F, and said triazolyl moiety is substituted as described in any one of the above embodiments describing substituted triazolyl groups.
In another embodiment of this invention R5 is a substituted triazolyl-thiazolyl- group wherein said thiazolyl moiety is substituted with one F, and said triazolyl moiety is substituted on the nitrogen with a hydroxyl substituted alkyl group. In another embodiment of this invention R5 is a substituted triazolyl-thiazolyl- wherein said thiazolyl moiety is substituted with one F, and said triazolyl moiety is substituted on the nitrogen with a -CH2CH2OH group.
In another embodiment of this invention R5 is a substituted triazolyl-thiazolyl- wherein said thiazolyl moiety is substituted with one F, and said triazolyl moiety is substituted on the nitrogen with an -alkylene-O-alkyl group.
In another embodiment of this invention R5 is a substituted triazolyl-thiazolyl- wherein said thiazolyl moiety is substituted with one F, and said triazolyl moiety is substituted on the nitrogen with a -CH2CH2OCH3 group.
In another embodiment of this invention R5 is a substituted triazolyl-thiazolyl- wherein said thiazolyl moiety is substituted with halo, and said triazolyl moiety is unsubstituted.
In another embodiment of this invention R5 is a substituted triazolyl-thiazolyl- wherein said thiazolyl moiety is substituted with F, and said triazolyl moiety is unsubstituted. In another embodiment of this invention R5 moiety is a substituted triazolyl- thiazolyl- wherein said thiazolyl moiety is substituted with alkoxy, and said triazolyl moiety is substituted as described in any of the above embodiments describing substituted triazolyl groups. In another embodiment of this invention R5 is a substituted triazolyl-thiazolyl- wherein said thiazolyl moiety is substituted with -OCH3, and said triazolyl moiety is substituted as described in any one of the above embodiments describing substituted triazolyl groups. In another embodiment of this invention R5 is a substituted triazolyl-thiazolyl- group wherein said thiazolyl moiety is substituted with one -OCH3, and said triazolyl moiety is substituted on the nitrogen with a hydroxyl substituted alkyl group.
In another embodiment of this invention R5 is a substituted triazolyl-thiazolyl- wherein said thiazolyl moiety is substituted with one -OCH3, and said triazolyl moiety is substituted on the nitrogen with a -CH2CH2OH group.
In another embodiment of this invention R5 is a substituted triazolyl-thiazolyl- wherein said thiazolyl moiety is substituted with one -OCH3, and said triazolyl moiety is substituted on the nitrogen with an -alkylene-O-alkyl group, and the alkylene moiety of said -alkylene-O-alkyl group is not -CH2- (i.e., the alkylene moiety is 2 or more carbons in length).
In another embodiment of this invention R5 is a substituted triazolyl-thiazolyl- wherein said thiazolyl moiety is substituted with one -OCH3, and said triazolyl moiety is substituted on the nitrogen with a -CH2CH2OCH3 group.
In another embodiment of this invention R5 is a substituted triazolyl-thiazolyl- wherein said thiazolyl moiety is substituted with alkoxy, and said triazolyl moiety is unsubstituted.
In another embodiment of this invention R5 is a substituted triazolyl-thiazolyl- wherein said thiazolyl moiety is substituted with -OCH3, and said triazolyl moiety is unsubstituted. In another embodiment of this invention R5 is a substituted triazolyl-thiazolyl- wherein said thiazolyl moiety is substituted with an alkylamino, and said triazolyl moiety is substituted as described in any of the above embodiments describing triazolyl groups.
In another embodiment of this invention R5 is a substituted triazolyl-thiazolyl- wherein said thiazolyl moiety is substituted with a dialkylamino, and said triazolyl moiety is substituted as described in any of the above embodiments describing substituted triazolyl groups. In another embodiment of this invention R5 is a substituted triazolyl-thiazolyl- wherein said thiazolyl moiety is substituted with an alkylamino, and said triazolyl moiety is unsubstituted.
In another embodiment of this invention R5 is a substituted triazolyl-thiazolyl- wherein said thiazolyl moiety is substituted with a dialkylamino, and said triazolyl moiety is unsubstituted.
In another embodiment of this invention R5 is a substituted triazolyl-thiazolyl- wherein said triazolyl is substituted with 1 or 2 groups indepenendently selected from the group consisting of: (a) hydroxyl substituted alkyl group (e.g., -CH2COH(CH3)2, and -CH2CH2OH)1 (b) alkyl (e.g., -Ci-C6alkyl, -Ci-C4alkyl, -CrC2alkyl, and -CH3), -NH2, and (c) -alkylene-O-alkyl (e.g., -CH2CH2OCH3), provided that the alkylene moiety of said -alkylene-O-alkyl group is not -CH2- (i.e., the alkylene moiety is 2 or more carbons in length) when said -alkylene-O-alkyl group is bound to the nitrogen of said triazolyl; and said thiazolyl is substituted with 1 group selected from the group consisting of: (a) alkyl (e.g., -CrC6alkyl, or -CrC4alkyl, or -CrC2alkyl, or -CH3), (b) halo (e.g., Cl, F, or Br), (c) alkylamino (e.g., CrC6alkyl-NH-, or CrC2alkyl-NH-, or CH3-MH-, or CH3CH2-NH-), and (d) dialkylamino (e.g., (CrC6alkyl)2-N- wherein each alkyl is independently selected, or (CrC2alkyl)2-N- wherein each alkyl is independently selected, or (CH3)2N-, or (CH3CH2)2-N-, or (CH3)(CH3CH2)N-). In another embodiment of this invention R5 is
Figure imgf000050_0001
In another embodiment of this invention R5 is pyridazinyl-thienyl-. In another embodiment of this invention R5 is
Figure imgf000050_0002
When the pyridazinyl moiety of R is substituted with alkyl, examples of the alkyl groups include, for example, -CrC6alkyl, -CrC4alkyl, -Ci-C2alkyl, and -CH3.
When the pyridazinyl moiety of R5 is substituted with halo atoms, examples of the halo atoms include, for example, Cl, F and Br. In one embodiment of this invention the halo on the pyridazinyl is F. In another embodiment of this invention the pyridazinyl is substituted with one F. When the pyridazinyl moiety is substituted with halos, the carbons adjacent to the nitrogens are not substituted with halos.
When the pyridazinyl moiety of R5 is substituted with an alkylamino group, examples of the alkylamino group include, for example, CrC6alkyl-NH-, CrC2alkyl-NH-, CH3-MH-, and CH3CH2-NH-.
When the pyridazinyl moiety of R5 is substituted with a dialkylamino group, examples of the dialkylamino group include, for example, (CrC6alkyl)2-N- wherein each alkyl is independently selected, (Ci-C2alkyl)2-N- wherein each alkyl is independently selected, (CHg)2N-, (CH3CH2)2-N-, and (CH3)(CH3CH2)N-. In another embodiment of this invention R5 is a substituted pyridazinyl-thienyl- wherein said pyridazinyl is substituted and said thienyl is unsubstituted.
In another embodiment of this invention R5 is a substituted pyridazinyl-thienyl- wherein said pyridazinyl is substituted with a =0 group, and said thienyl is unsubstituted. In another embodiment of this invention R5 is a substituted pyridazinyl-thienyl- wherein said pyridazinyl is substituted with an alkyl group, and said thienyl is unsubstituted.
In another embodiment of this invention R5 is a substituted pyridazinyl-thienyl- wherein said pyridazinyl is substituted with a methyl group, and said thienyl is unsubstituted.
In another embodiment of this invention R5 is a substituted pyridazinyl-thienyl- wherein said pyridazinyl is substituted with a =0 group, and with an alkyl group, and said thienyl is unsubstituted.
In another embodiment of this invention R5 is a substituted pyridazinyl-thienyl- wherein said pyridazinyl is substituted with a =0 group, and with a methyl group, and said thienyl is unsubstituted.
In another embodiment of this invention R5 is a substituted pyridazinyl-thienyl- wherein said pyridazinyl is substituted with an amino group, and said thienyl is unsubstituted. In another embodiment of this invention R5 is a substituted pyridazinyl-thienyl- wherein said pyridazinyl is substituted with an alkylamino group, and said thienyl is unsubstituted. In another embodiment of this invention R5 is a substituted pyridazinyl-thienyl- wherein said pyridazinyl is substituted with a dialkylamino group, and said thienyl is unsubstituted.
In another embodiment of this invention R5 is a substituted pyridazinyl-thienyl- wherein said pyridazinyl is substituted and said thienyl is substituted.
In another embodiment of this invention R5 is a substituted pyridazinyl-thienyl- wherein said pyridazinyl is unsubstituted and said thienyl is substituted.
In another embodiment of this invention R5 is a substituted pyridazinyl-thienyl- wherein said pyridazinyl is unsubstituted and said thienyl is substituted with 1 to 2 independently selected halos.
In another embodiment of this invention R5 is a substituted pyridazinyl-thienyl- wherein said pyridazinyl is unsubstituted and said thienyl is substituted with 1 to 2 halos independently selected from the group consisting of: Br, Cl and F.
In another embodiment of this invention R5 is a substituted pyridazinyl-thienyl- wherein said pyridazinyl is unsubstituted and said thienyl is substituted with 1 to 2 independently selected alkoxy groups.
In another embodiment of this invention R5 is a substituted pyridazinyl-thienyl- wherein said pyridazinyl is unsubstituted and said thienyl is substituted with 1 to 2 -OCH3 groups. In another embodiment of this invention R5 is a substituted pyridazinyl-thienyl- wherein said pyridazinyl is substituted as described in any one of the embodiments above describing substituted pyridazinyl groups, and said thienyl is substituted.
In another embodiment of this invention R5 is a substituted pyridazinyl-thienyl- wherein said pyridazinyl is substituted as described in any one of the embodiments above describing substituted pyridazinyl groups, and said thienyl is substituted with 1 to 2 independently selected halos.
In another embodiment of this invention R5 is a substituted pyridazinyl-thienyl- wherein said pyridazinyl is substituted as described in any one of the embodiments above describing substituted pyridazinyl groups, and said thienyl is substituted with 1 to 2 halos independently selected from the group consisting of: Br, Cl and F.
In another embodiment of this invention R5 is a substituted pyridazinyl-thienyl- wherein said pyridazinyl is substituted as described in any one of the embodiments above describing substituted pyridazinyl groups, and said thienyl is substituted with 1 to 2 independently selected alkoxy groups. In another embodiment of this invention R5 is a substituted pyridazinyl-thienyl- wherein said pyridazinyl is substituted as described in any one of the embodiments above describing substituted pyridazinyl groups, and said thienyl is substituted with 1 to 2 -OCH3 groups. In another embodiment of this invention R5 is a substituted pyridazinyl-thienyl- wherein said pyridazinyl is substituted with 1 or 2 groups indepenendently selected from the group consisting of alkyl (e.g., methyl) and =0, and said thienyl is substituted with 1 to 2 groups independently selected from the group consisting of: alkoxy (e.g., -OCH3), halo (e.g., Br, Cl and F). In another embodiment of this invention R5 is
Figure imgf000053_0001
In another embodiment of this invention R5 is
Figure imgf000053_0002
In another embodiment of this invention R5 is
Figure imgf000053_0003
In another embodiment of this invention R5 is a substituted triazolyl-phenyl- group wherein (a) said triazolyl moiety is optionally substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2 and -CH2CH2OH, (b) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is optionally substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is optionally substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is optionally substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is optionally substituted on the nitrogen with a -CH2CH2OCH3 group; and wherein said phenyl moiety is optionally substituted with halo. In another embodiment of this invention R5 is a substituted triazolyl-phenyl- group wherein (a) said triazolyl moiety is optionally substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2 and -CH2CH2OH, (b) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is optionally substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is optionally substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is optionally substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is optionally substituted on the nitrogen with a -CH2CH2OCH3 group; and wherein said phenyl moiety is substituted with halo.
In another embodiment of this invention R5 is a substituted triazolyl-phenyl- group wherein (a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2 and -CH2CH2OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is substituted on the nitrogen with a
-CH2CH2OCH3 group; and wherein said phenyl moiety is optionally substituted with halo.
In another embodiment of this invention R5 is a substituted triazolyl-phenyl- group wherein (a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2 and -CH2CH2OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is substituted on the carbon with a -MH2 group, or (g) said triazolyl moiety is substituted on the nitrogen with a -CH2CH2OCH3 group; and wherein said phenyl moiety is substituted with halo.
In another embodiment of this invention R5 is a substituted triazolyl-phenyl- group wherein (a) said triazolyl moiety is substituted on the nitrogen with a substituent Sθlectθd from the group consisting of: -CH2COH(CH3^ and -CH2CH2OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is substituted on the nitrogen with a -CH2CH2OCH3 group; and wherein said phenyl moiety is unsubstituted.
In another embodiment of this invention R5 is a substituted triazolyl-phenyl- group wherein (a) said triazolyl moiety is optionally substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2 and -CH2CH2OH, (b) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is optionally substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is optionally substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is optionally substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is optionally substituted on the nitrogen with a -CH2CH2OCH3 group; and wherein said phenyl moiety is optionally substituted with alkoxy. In another embodiment of this invention R5 is a substituted triazolyl-phenyl- group wherein (a) said triazolyl moiety is optionally substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2 and -CH2CH2OH, (b) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is optionally substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is optionally substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is optionally substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is optionally substituted on the nitrogen with a -CH2CH2OCH3 group; and wherein said phenyl moiety is substituted with alkoxy.
In another embodiment of this invention R5 is a substituted triazolyl-phenyl- group wherein (a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2 and -CH2CH2OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is substituted on the nitrogen with a
-CH2CH2OCH3 group; and wherein said phenyl moiety is optionally substituted with alkoxy.
In another embodiment of this invention R5 is a substituted triazolyl-phenyl- group wherein (a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2 and -CH2CH2OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is substituted on the nitrogen with a -CH2CH2OCH3 group; and wherein said phenyl moiety is substituted with alkoxy. In another embodiment of this invention R5 is a substituted triazolyl-thienyl- group wherein (a) said triazolyl moiety is optionally substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2 and -CH2CH2OH, (b) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is optionally substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is optionally substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is optionally substituted on the carbon with a -MH2 group, or (g) said triazolyl moiety is optionally substituted on the nitrogen with a -CH2CH2OCH3 group; and wherein said thienyl moiety is optionally substituted with halo.
In another embodiment of this invention R5 is a substituted triazolyl-thienyl- group wherein (a) said triazolyl moiety is optionally substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2 and -CH2CH2OH, (b) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is optionally substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is optionally substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is optionally substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is optionally substituted on the nitrogen with a -CH2CH2OCH3 group; and wherein said thienyl moiety is substituted with halo.
In another embodiment of this invention R5 is a substituted triazolyl-thienyl- group wherein (a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2 and -CH2CH2OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is substituted on the nitrogen with a -CH2CH2OCH3 group; and wherein said thienyl moiety is optionally substituted with halo.
In another embodiment of this invention R5 is a substituted triazolyl-thienyl- group wherein (a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2 and -CH2CH2OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is substituted on the nitrogen with a -CH2CH2OCH3 group; and wherein said thienyl moiety is substituted with halo.
In another embodiment of this invention R5 is a substituted triazolyl-thienyl- group wherein (a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2 and -CH2CH2OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is substituted on the nitrogen with a -CH2CH2OCH3 group; and wherein said thienyl moiety is unsubstituted.
In another embodiment of this invention R5 is a substituted triazolyl-thienyl- group wherein (a) said triazolyl moiety is optionally substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2, and -CH2CH2OH, (b) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is optionally substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is optionally substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is optionally substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is optionally substituted on the nitrogen with a -CH2CH2OCH3 group; and wherein said thienyl moiety is optionally substituted with alkoxy.
In another embodiment of this invention R5 is a substituted triazolyl-thienyl- group wherein (a) said triazolyl moiety is optionally substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2 and -CH2CH2OH, (b) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is optionally substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is optionally substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is optionally substituted on the carbon with a -MH2 group, or (g) said triazolyl moiety is optionally substituted on the nitrogen with a -CH2CH2OCH3 group; and wherein said thienyl moiety is substituted with alkoxy. In another embodiment of this invention R5 is a substituted triazolyl-thienyl- group wherein (a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2 and -CH2CH2OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is substituted on the nitrogen with a -CH2CH2OCH3 group; and wherein said thienyl moiety is optionally substituted with alkoxy.
In another embodiment of this invention R5 is a substituted triazolyl-thienyl- group wherein (a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2, and -CH2CH2OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is substituted on the nitrogen with a -CH2CH2OCH3 group; and wherein said thienyl moiety is substituted with alkoxy.
Other embodiments of the invention are described below. The embodiments have been numbered for ease of reference. Embodiment No. 1 is directed to compounds of formula 1.0 wherein R2 is a
-O-(CrC2)alkyl group, and R1 is substituted pyridyl.
Embodiment No. 2 is directed to compounds of formula 1.0 wherein R2 is a -O-(CrC2)alkyl group, and R1 is pyridyl substituted with one substituent.
Embodiment No. 3 is directed to compounds of formula 1.0 wherein R2 is a -O-(Ci-C2)alkyl group, and R1 is selected from the group consisting of:
Figure imgf000059_0001
Embodiment No. 4 is directed to compounds of formula 1.0 wherein R2 is a -O-(Ci-C2)alkyl group, and R1 is:
Figure imgf000059_0002
Embodiment No. 5 is directed to compounds of formula 1.0 wherein R2 is a
-O-(C-,-C2)alkyl group, and R J1 : is, :
Figure imgf000059_0003
Embodiment No. 6 is directed to compounds of formula 1.0 wherein R2 is a -O-(Ci-C2)alkyl group, and R1 is:
Figure imgf000060_0001
wherein said alkoxy group is -OCH(CH3)2.
Embodiment No. 7 is directed to compounds of formula 1.0 wherein, R2 is a -O-(Ci-C2)alkyl group, and R1 is:
Figure imgf000060_0002
wherein said alkoxy group is -OC2H5.
Embodiment No. 8 is directed to compounds of formula 1.0 wherein R2 is a
-O-(CrC2)alkyl group, and R1 is:
Figure imgf000060_0003
Embodiment No. 9 is directed to compounds of formula 1.0 wherein R2 is a
-O-(CrC2)alkyl group, and R1 is:
Figure imgf000060_0004
Embodiment No. 10 is directed to compounds of formula 1.0 wherein z is 1 , R2 is a -O-(Ci-C2)alkyl group, and R1 is:
Figure imgf000060_0005
wherein said -O-alkylene-O-alkyl group is -OCH2CH2OCH3.
Embodiment No. 11 is directed to compounds of formula 1.0 wherein R2 is a -O-(CrC2)alkyl group, and R1 is:
Figure imgf000060_0006
Embodiment No. 12 is directed to compounds of formula 1.0 wherein R 2 is
-OCH3, and R J : is substituted pyridyl. Embodiment No. 13 is directed to compounds of formula 1.0 wherein R 2 . is, -OCH3, and R1 is pyridyl substituted with one substituent.
Embodiment No. 14 is directed to compounds of formula 1.0 wherein R2 is -OCH3, and R1 is selected from the group consisting of:
Figure imgf000061_0001
Embodiment No. 15 is directed to compounds of formula 1.0 wherein R2 is -OCH3, and R1 is:
Figure imgf000061_0002
Embodiment No. 16 is directed to compounds of formula 1.0 wherein R2 is -OCH3, and R1 is:
Figure imgf000061_0003
Embodiment No. 17 is directed to compounds of formula 1.0 wherein R2 is
-OCH3, and R1 is:
Figure imgf000061_0004
wherein said alkoxy group is -OCH(CH3)2.
Embodiment No. 18 is directed to compounds of formula 1.0 wherein R2 is -OCH3, and R1 is:
Figure imgf000061_0005
wherein said alkoxy group is -OC2H5. Embodiment No. 19 is directed to compounds of formula 1.0 wherein R2 is
-OCH3, and R1 is:
Figure imgf000061_0006
Embodiment No. 20 is directed to compounds of formula 1.0 wherein R2 is
-OCH3, and R1 is:
Figure imgf000062_0001
Embodiment No. 21 is directed to compounds of formula 1.0 wherein R2 is -OCH3, and R1 is:
Figure imgf000062_0002
wherein said -O-alkylene-O-alkyl group is -OCH2CH2OCH3.
EEmmbbooddiimmeernt No. 22 is directed to compounds of formula 1.0 wherein R2 is -OCH3, and R1 is:
Figure imgf000062_0003
Embodiment No. 23 is directed to compounds of formula 1.0 wherein R2 is a -S-(Ci -C2)alkyl group, and R1 is substituted pyridyl.
Embodiment No. 24 is directed to compounds of formula 1.0 wherein R2 is a -S-(C1 -C2)alkyl group, and R1 is pyridyl substituted with one substituent. Embodiment No. 25 is directed to compounds of formula 1.0 wherein R2 is a
-S-(Ci -C2)alkyl group, and R1 is selected from the group consisting of:
Figure imgf000062_0004
Embodiment No. 26 is directed to compounds of formula 1.0 wherein R2 is a -S-(Ci -C2)alkyl group, and R1 is:
Figure imgf000062_0005
Embodiment No. 27 is directed to compounds of formula 1.0 wherein R2 is a -S-(Ci -C2)alkyl group, and R1 is:
Figure imgf000062_0006
Embodiment No. 28 is directed to compounds of formula 1.0 wherein R2 is a
-S-(Ci -C2)alkyl group, and R1 is:
Figure imgf000063_0001
wherein said alkoxy group is -OCH(CH3)2.
EEmmbbooddiimmeenntt NNoo.. 2299 iiss ddiirreected to compounds of formula 1.0 wherein R2 is a -S-(Ci -C2)alkyl group, and R1 is:
Figure imgf000063_0002
wherein said alkoxy group is -OC2H5.
Embodiment No. 30 is directed to compounds of formula 1.0 wherein R2 is a -S-(Ci -C2)alkyl group, and R1 is:
Figure imgf000063_0003
Embodiment No. 31 is directed to compounds of formula 1.0 wherein R2 is a -S-(Ci -C2)alkyl group, and R1 is:
Figure imgf000063_0004
Embodiment No. 32 is directed to compounds of formula 1.0 wherein R2 is a
-S-(Ci-C2)alkyl group, and R1 is:
Figure imgf000063_0005
wherein said -O-alkylene-O-alkyl group is -OCH2CH2OCH3.
Embodiment No. 33 is directed to compounds of formula 1.0 wherein R2 is a S-(Ci -C2)alkyl group, and R1 is:
Figure imgf000063_0006
Embodiment No. 34 is directed to compounds of formula 1.0 wherein R2 is -SCH3, and R1 is substituted pyridyl.
Embodiment No. 35 is directed to compounds of formula 1.0 wherein R2 is -SCH3, and R1 is pyridyl substituted with one substituent.
Embodiment No. 36 is directed to compounds of formula 1.0 wherein R2 is -SCH3, and R1 is selected from the group consisting of:
Figure imgf000064_0001
Embodiment No. 37 is directed to compounds of formula 1.0 wherein R2 is
-SCH3, and R is:
Figure imgf000064_0002
Embodiment No. 38 is directed to compounds of formula 1.0 wherein R2 is
-SCH3, and R1 is:
Figure imgf000064_0003
Embodiment No. 39 is directed to compounds of formula 1.0 wherein R2 is -SCH3, and R1 is:
Figure imgf000064_0004
wherein said alkoxy group is -OCH(CH3)2.
Embodiment No. 40 is directed to compounds of formula 1.0 wherein R2 is -SCH3, and R1 is:
Figure imgf000064_0005
wherein said alkoxy group is -OC2H5.
Embodiment No. 41 is directed to compounds of formula 1.0 wherein R2 is -SCH3, and R1 is:
Figure imgf000065_0001
Embodiment No. 42 is directed to compounds of formula 1.0 wherein R2 is
-SCH3, and R1 is:
Figure imgf000065_0002
Embodimen itt NNoo.. 4433 iiss ddiirreecctteedd ttoo ccoompounds of formula 1.0 wherein R2 is -SCH3, and R1 is:
Figure imgf000065_0003
wherein said -O-alkylene-0-alkyl group is -OCH2CH2OCH3.
Embodiment No. 44 is directed to compounds of formula 1.0 wherein R2 is -SCH3, and R1 is:
Figure imgf000065_0004
Embodiment No. 45 is directed to compounds of formula 1.0 having the formula 1.1 :
Figure imgf000065_0005
Embodiment No. 46 is directed to any one of Embodiment Numbers 1 to 44 wherein the compound of formula 1.0 is a compound of formula 1.1.
Other embodiments of the invention directed to the R5substituent are described below. The language "as described in any one of Embodiment Numbers 1 to 46" means that the embodiment being described is applicable to each one of Embodiment Numbers 1 to 46. For example, another embodiment of this invention is directed to compounds described in Embodiment No. 1 wherein R5 is as described in any one of the paragraphs below. In another example, another embodiment of this invention is directed to the compounds described in Embodiment No.2 wherein R5 is as described in any one of the paragraphs below, etc.
Thus, other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-phenyl- wherein the triazolyl moiety is substituted with one or two alkyl groups selected from the group consisting of: -CrC6alkyl, -CrC4alkyl, -CrC2alkyl, and -CH3. .
Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-phenyl- wherein the triazolyl moiety is substituted with one or two alkyl groups selected from the group consisting of: -Ci-C4alkyl, -CrC2alkyl, and -CH3. Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl- phenyl- wherein the triazolyl moiety is substituted with one or two -CH3 groups.
Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-phenyl- wherein the triazolyl moiety is substituted on the nitrogen with an alkyl group selected from the group consisting of: -Ci-C4alkyl, -CrC2alkyl, and -CH3. Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-phenyl- wherein the triazolyl moiety is substituted on the nitrogen with -CH3 group.
Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-phenyl- wherein the triazolyl moiety is substituted on the nitrogen with an -alkylene-O-alkyl group selected from the group consisting of: -C2-C4alkylene-O-d- C6alkyl, -C2alkylene-O-CrC2alkyl, -C2-C4alkylene-O-CH3, and -CH2CH2OCH3.
Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-phenyl- wherein the triazolyl moiety is substituted on the nitrogen with an -alkylene-O-alkyl group selected from the group consisting of: -C2alkylene-O-Cr C2alkyl, and -CH2CH2OCH3. Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-phenyl- wherein the triazolyl moiety is substituted on the nitrogen with -CH2CH2OCH3. Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-phenyl- wherein the triazolyl moiety is substituted on the nitrogen with a hydroxy substituted alkyl group selected from the group consisting of: hydroxy substituted -CrC4alkyl, hydroxy substituted -Ci-C2alkyl, and hydroxy substituted -CH3. Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-phenyl- wherein the triazolyl moiety is substituted on the nitrogen with a hydroxy substituted alkyl group selected from the group consisting of: CH2COH(CH3)2, and -CH2CH2OH. Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one oF Embodiment Numbers 1 to 46 wherein the optional halo substituents for the phenyl moiety of R5 are independently selected from the group consisting of: Cl, F and Br.
Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein the optional halo substituents for the phenyl moiety of R5 are F.
Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein the optional halo substituent for the phenyl moiety of R5 is one F. Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-phenyl- group wherein (a) said triazolyl moiety is optionally substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2 and -CH2CH2OH, (b) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is optionally substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is optionally substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is optionally substituted on the carbon with a -MH2 group, or (g) said triazolyl moiety is optionally substituted on the nitrogen with a -CH2CH2OCH3 group; and wherein said phenyl moiety is optionally substituted with halo (e.g., one halo, such as for example, F).
Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-phenyl- group wherein (a) said triazolyl moiety is optionally substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2 and -CH2CH2OH, (b) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is optionally substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is optionally substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is optionally substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is optionally substituted on the nitrogen with a -CH2CH2OCH3 group; and wherein said phenyl moiety is substituted with halo (e.g., one halo, such as for example, F).
Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-phenyl- group wherein (a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2 and
-CH2CH2OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is substituted on the carbon with a -MH2 group, or (g) said triazolyl moiety is substituted on the nitrogen with a-CH2CH2OCH3 group; and wherein said phenyl moiety is optionally substituted with halo (e.g., one halo, such as for example, F).
Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-phenyl- group wherein (a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2 and -CH2CH2OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is substituted on the nitrogen with a-CH2CH2OCH3 group; and wherein said phenyl moiety is substituted with halo (e.g., one halo, such as for example, F).
Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein the optional substituents for the phenyl moiety of R5 are independently selected from the group consisting of: alkoxy.
Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein the optional substituents for the phenyl moiety of R5 are -OCH3.
Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein the optional substituent for the phenyl moiety of R5 is one -OCH3.
Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-phenyl- group wherein (a) said triazolyl moiety is optionally substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2, and -CH2CH2OH, (b) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is optionally substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is optionally substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is optionally substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is optionally substituted on the nitrogen with a -CH2CH2OCH3 group; and wherein said phenyl moiety is optionally substituted with alkoxy (e.g., one -OCH3). Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-phenyl- group wherein (a) said triazolyl moiety is optionally substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2, and -CH2CH2OH, (b) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is optionally substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is optionally substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is optionally substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is optionally substituted on the nitrogen with a -CH2CH2OCH3 group; and wherein said phenyl moiety is substituted with alkoxy (e.g., one -OCH3).
Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-phenyl- group wherein (a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2> and -CH2CH2OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is substituted on the nitrogen with a-CH2CH2OCH3 group; and wherein said phenyl moiety is optionally substituted with alkoxy (e.g., one -OCH3).
Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-phenyl- group wherein (a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2, and -CH2CH2OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is substituted on the nitrogen with a-CH2CH2OCH3 group; and wherein said phenyl moiety is substituted with alkoxy (e.g., one -OCH3).
Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-phenyl- group wherein (a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2, and -CH2CH2OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is substituted on the nitrogen with a-CH2CH2OCH3 group; and wherein said phenyl moiety is unsubstituted.
Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein said substituted R5 moiety is a substituted triazolyl-phenyl group wherein said triazolyl moiety is substituted with: (a) one substituent selected from the group consisting of: -CH2COH(CH3)2, and -CH2CH2OH, (b) one alkyl group, (c) two alkyl groups, (d) one -CH3 group, (e) two -CH3 groups, (f) one -NH2 group, or (g) one -CH2CH2OCH3 group.
Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-thienyl- wherein the triazolyl moiety is substituted with one or two alkyl groups selected from the group consisting of: -CrC6alkyl, -Ci-C4alkyl, -CrC2alkyl, and -CH3. Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-thienyl- wherein the triazolyl moiety is substituted with one or two alkyl groups selected from the group consisting of: -C-ι-C4alkyl, -CrC2alkyl, and -CH3. Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl- thienyl- wherein the triazolyl moiety is substituted with one or two -CH3 groups.
Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-thienyl- wherein the triazolyl moiety is substituted on the nitrogen with an alkyl group selected from the group consisting of: -Ci-C4alkyl, -CrC2alkyl, and -CH3. Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-thienyl- wherein the triazolyl moiety is substituted on the nitrogen with -CH3. Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-thienyl- wherein the triazolyl moiety is substituted on the nitrogen with an -alkylene-O-alkyl group selected from the group consisting of: -C2-C4alkylene-O-Cr C6alkyl, -Cj.alkylene-O-CrCj.alkyl, -Cj.-C.talkylene-0-CHs, and -CH2CH2OCH3.
Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-thienyl- wherein the triazolyl moiety is substituted on the nitrogen with an -alkylene-O-alkyl group selected from the group consisting of: -C2alkylene-O-d- C2alkyl, and-CH2CH2OCH3. Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-thienyl- wherein the triazolyl moiety is substituted on the nitrogen with -CH2CH2OCH3.
Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-thienyl- wherein the triazolyl moiety is substituted on the nitrogen with a hydroxy substituted alkyl group selected from the group consisting of: hydroxy substituted -CrC4alkyl, hydroxy substituted -CrC2alkyl, and hydroxy substituted -CH3. Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-thienyl- wherein the triazolyl moiety is substituted on the nitrogen with a hydroxy substituted alkyl group selected from the group consisting of: CH2COH(CH3)2, and -CH2CH2OH.
Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein the optional halo substituents for the thienyl moiety of R5 are independently selected from the group consisting of: Cl, F and Br. Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein the optional halo substituents for the thienyl moiety of R5 are F. Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein the optional halo substituent for the thienyl moiety of R5 is one F.
Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-thienyl- group wherein (a) said triazolyl moiety is optionally substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2, and -CH2CH2OH, (b) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is optionally substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is optionally substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is optionally substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is optionally substituted on the nitrogen with a -CH2CH2OCH3 group; and wherein said thienyl moiety is optionally substituted with halo (e.g., one halo, such as for example, F).
Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-thienyl- group wherein (a) said triazolyl moiety is optionally substituted on the nitrogen with a substituent selected from the group consisting of:-CH2COH(CH3)2, -CH2CH2OH, (b) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is optionally substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is optionally substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is optionally substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is optionally substituted on the nitrogen with a -CH2CH2OCH3 group; and wherein said thienyl moiety is substituted with halo (e.g., one halo, such as for example, F). Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-thienyl- group wherein (a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2, and -CH2CH2OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is substituted on the nitrogen with a-CH2CH2OCH3 group; and wherein said thienyl moiety is optionally substituted with halo (e.g., one halo, such as for example, F).
Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-thienyl- group wherein (a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2, and -CH2CH2OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is substituted on the nitrogen with a-CH2CH2OCH3 group; and wherein said thienyl moiety is substituted with halo (e.g., one halo, such as for example, F). Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-thienyl- group wherein (a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2, and -CH2CH2OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is substituted on the nitrogen with a-CH2CH2OCH3 group; and wherein said thienyl moiety is unsubstituted. Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein said substituted R5 moiety is a substituted triazolyl-thienyl group wherein said triazolyl moiety is substituted with: (a) one substituent selected from the group consisting of: -CH2COH(CH3)2, and -CH2CH2OH, (b) one alkyl group, (c) two alkyl groups, (d) one -CH3 group, (e) two -CH3 groups, (f) one -NH2 group, or (g) one -CH2CH2OCH3 group.
Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein the optional substituents for the thienyl moiety of R5 are independently selected from the group consisting of: alkoxy.
Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein the optional substituents for the thienyl moiety of R5 are -OCH3.
Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein the optional substituent for the thienyl moiety of R5 is one -OCH3.
Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-thienyl- group wherein (a) said triazolyl moiety is optionally substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2, and -CH2CH2OH1 (b) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is optionally substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is optionally substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is optionally substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is optionally substituted on the nitrogen with a -CH2CH2OCH3 group; and wherein said thienyl moiety is optionally substituted with alkoxy (e.g., one -OCH3).
Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-thienyl- group wherein (a) said triazolyl moiety is optionally substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2, and -CH2CH2OH, (b) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is optionally substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is optionally substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is optionally substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is optionally substituted on the nitrogen with a -CH2CH2OCH3 group; and wherein said thienyl moiety is substituted with alkoxy (e.g., one -OCH3).
Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-thienyl- group wherein (a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2, and -CH2CH2OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is substituted on the carbon with a -MH2 group, or (g) said triazolyl moiety is substituted on the nitrogen with a-CH2CH2OCH3 group; and wherein said thienyl moiety is optionally substituted with alkoxy (e.g., one -OCH3).
Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-thienyl- group wherein (a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2, and -CH2CH2OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is substituted on the nitrogen with a-CH2CH2OCH3 group; and wherein said thienyl moiety is substituted with alkoxy (e.g., one -OCH3). Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-pyridyl- wherein the triazolyl moiety is substituted with one or two alkyl groups selected from the group consisting of: -CrC6alkyl, -CrC4alkyl, -CrC2alkyl, and -CH3. Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-pyridyl- wherein the triazolyl moiety is substituted with one or two alkyl groups selected from the group consisting of: -CrC4alkyl, -CrC2alkyl, and -CH3. Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl- pyridyl- wherein the triazolyl moiety is substituted with one or two -CH3 groups.
Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-pyridyl- wherein the triazolyl moiety is substituted on the nitrogen with an alkyl group selected from the group consisting of: -CrC4alkyl, -Ci-C2alkyl, and -CH3. Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-pyridyl- wherein the triazolyl moiety is substituted on the nitrogen with -CH3. Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-pyridyl- wherein the triazolyl moiety is substituted on the nitrogen with an -alkylene-O-alkyl group selected from the group consisting of: -C2-C4alkylene-O-d- C6alkyl, -C2alkylene-O-CrC2alkyl, -CrC^lkylene-O-CHa, and -CH2CH2OCH3. Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-pyridyl- wherein the triazolyl moiety is substituted on the nitrogen with an -alkylene-O-alkyl group selected from the group consisting of: -C2alkylene-O-Ci- C2alkyl, and -CH2CH2OCH3. Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-pyridyl- wherein the triazolyl moiety is substituted on the nitrogen with -CH2CH2OCH3.
Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-pyridyl- wherein the triazolyl moiety is substituted on the nitrogen with a hydroxy substituted alkyl group selected from the group consisting of: hydroxy substituted -CrC4alkyl, hydroxy substituted -CrC2alkyl, and hydroxy substituted -CH3. Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-pyridyl- wherein the triazolyl moiety is substituted on the nitrogen with a hydroxy substituted alkyl group selected from the group consisting of: CH2COH(CH3)2, and -CH2CH2OH.
Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-pyridyl- group wherein (a) said triazolyl moiety is optionally substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2, and -CH2CH2OH, (b) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is optionally substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is optionally substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is optionally substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is optionally substituted on the nitrogen with a -CH2CH2OCH3 group; and wherein said pyridyl moiety is unsubstitued.
Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-pyridyl- group wherein (a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2, and -CH2CH2OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is substituted on the carbon with a -MH2 group, or (g) said triazolyl moiety is substituted on the nitrogen with a-CH2CH2OCH3 group; and wherein said pyridyl moiety is unsubstitued. Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-thiazolyl- wherein the triazolyl moiety is substituted with one or two alkyl groups selected from the group consisting of: -CrC6alkyl, -Ci-C4alkyl, -Ci-C2alkyl, and -CH3.
Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-thiazolyl- wherein the triazolyl moiety is substituted with one or two alkyl groups selected from the group consisting of: -CrC4alkyl, -CrC2alkyl, and -CH3. Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-thiazolyl- wherein the triazolyl moiety is substituted with one or two -CH3 groups.
Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-thiazolyl- wherein the triazolyl moiety is substituted on the nitrogen with an alkyl group selected from the group consisting of: -CrC^alkyl, -d-C2alkyl, and -CH3. Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-thiazolyl- wherein the triazolyl moiety is substituted on the nitrogen with -CH3. Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-thiazolyl- wherein the triazolyl moiety is substituted on the nitrogen with an -alkylene-O-alkyl group selected from the group consisting of: -C2-C4alkylene-O-d- C6alkyl, -dalkylene-O-Crdalkyl, -C2-C4alkylene-O-CH3, and -CH2CH2OCH3.
Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-thiazolyl- wherein the triazolyl moiety is substituted on the nitrogen with an -alkylene-O-alkyl group selected from the group consisting of: -C2alkylene-O-d- C2alkyl, and -CH2CH2OCH3. Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-thiazolyl- wherein the triazolyl moiety is substituted on the nitrogen with -CH2CH2OCH3.
Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-thiazolyl- wherein the triazolyl moiety is substituted on the nitrogen with a hydroxy substituted alkyl group selected from the group consisting of: hydroxy substituted -Ci-C4alkyl, hydroxy substituted -CrC2alkyl, and hydroxy substituted -CH3. Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-thiazolyl- wherein the triazolyl moiety is substituted on the nitrogen with a hydroxy substituted alkyl group selected from the group consisting of: CH2COH(CH3)2, and -CH2CH2OH.
Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-thiazolyl- group wherein (a) said triazolyl moiety is optionally substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2, and -CH2CH2OH, (b) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is optionally substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is optionally substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is optionally substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is optionally substituted on the nitrogen with a -CH2CH2OCH3 group; and wherein said thiazolyl moiety is unsubstitued.
Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted triazolyl-thiazolyl- group wherein (a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2, and -CH2CH2OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is substituted on the nitrogen with a-CH2CH2OCH3 group; and wherein said thiazolyl moiety is unsubstitued.
Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted pyradizinyl-thienyl- wherein the pyridazinyl moiety is substituted with 1 or 2 groups indepenendently selected from the group consisting of alkyl (e.g., methyl) and =O, and said thienyl moiety is unsubstituted.
Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is a substituted pyradizinyl-thienyl- wherein the pyridazinyl moiety is substituted with an =0 moiety, or said pyridazinyl group is substituted with an alkyl (e.g., methyl), or said pyridazinyl is substituted with an =0 moiety and an an alkyl (e.g., methyl), and said thienyl moiety is unsubstituted.
Other embodiments of this invention are directed to compounds of formula 1.0 as described in any one of Embodiment Numbers 1 to 46 wherein R5 is selected from the group consisting of:
Figure imgf000081_0001
Figure imgf000081_0002
Figure imgf000081_0003
Figure imgf000082_0001
Figure imgf000082_0002
Figure imgf000082_0003
Other embodiments of this invention are directed to compounds of formula 1.0 aass ddeessccrriibbeedd iinn aannyy oonnee of Embodiment Numbers 1 to 46 wherein R5 is selected from the group consisting of:
Figure imgf000082_0004
Figure imgf000083_0001
Figure imgf000083_0002
Figure imgf000083_0003
Figure imgf000083_0004
Figure imgf000083_0005
Other embodiments of this invention are directed to compounds of formula 1.0 as des sccrriibbeedd iinn aannyy oonnee of Embodiment Numbers 1 to 46 wherein R5 is selected from the group consisting of:
Figure imgf000084_0001
Figure imgf000084_0002
Figure imgf000084_0003
Figure imgf000084_0004
and
Figure imgf000084_0005
Other embodiments of this invention are directed to compounds of formula 1.0 as des sccrriibbeedd iinn aannyy oonnee of Embodiment Numbers 1 to 46 wherein R5 is selected from the group consisting of:
Figure imgf000085_0001
Figure imgf000085_0002
Figure imgf000085_0003
Figure imgf000085_0004
, and
Figure imgf000085_0005
Other embodiments of this invention are directed to compounds of formula 1.1 :
Figure imgf000086_0001
wherein:
R1 and R2 are as defined in any one of embodiment numbers 1 to 44, Q is:
Figure imgf000086_0002
R is selected from the group consisting of:
Figure imgf000086_0003
Figure imgf000086_0004
Figure imgf000087_0001
Figure imgf000087_0002
Figure imgf000087_0003
Figure imgf000087_0004
Figure imgf000087_0005
Other embodiments of this invention are directed to compounds of formula 1.1 :
Figure imgf000088_0001
wherein:
R2 is a -O-(CrC2)alkyl group, and R' is:
Figure imgf000088_0002
Q is:
Figure imgf000088_0003
R5 is selected from the group consisting of:
Figure imgf000088_0004
Figure imgf000088_0005
Figure imgf000089_0001
Figure imgf000089_0002
Figure imgf000089_0003
Figure imgf000089_0004
Figure imgf000089_0005
Other embodiments of this invention are directed to compounds of formula 1.1:
Figure imgf000090_0001
wherein:
R2 is a -S-(Ci -C2)alkyl group, and R >η1 i :s. :
alkoxy
Figure imgf000090_0002
Q is:
Figure imgf000090_0003
R5 is selected from the group consisting of:
Figure imgf000090_0004
Figure imgf000090_0005
Figure imgf000091_0001
Figure imgf000091_0002
Figure imgf000091_0003
Figure imgf000091_0004
Figure imgf000091_0005
, and
Other embodiments of this invention are directed to any one of the embodiments above (for example, any one of embodiment numbers 1 to 46, or any one of the embodiments following embodiment number 46) wherein one or more hydrogen atoms are deuterium. Representative compounds of this invention include, but are not limited to:
Figure imgf000092_0001
Figure imgf000092_0002
Figure imgf000092_0003
Figure imgf000093_0001
Figure imgf000094_0001
Figure imgf000095_0001
Figure imgf000095_0002
Figure imgf000095_0003
Figure imgf000096_0001
Figure imgf000097_0001
Figure imgf000098_0001
Figure imgf000098_0002
Figure imgf000098_0003
Figure imgf000098_0004
Figure imgf000099_0001
Figure imgf000099_0002
Figure imgf000099_0003
Figure imgf000100_0001
Figure imgf000100_0002
Figure imgf000100_0003
Figure imgf000101_0001
Figure imgf000101_0002
Figure imgf000101_0003
Figure imgf000101_0004
Figure imgf000102_0001
Figure imgf000102_0002
Figure imgf000102_0003
Figure imgf000102_0004
Figure imgf000103_0001
Figure imgf000103_0002
Figure imgf000103_0003
Figure imgf000103_0004
Figure imgf000104_0001
Figure imgf000104_0002
Figure imgf000104_0003
Figure imgf000104_0004
Figure imgf000105_0001
Figure imgf000105_0002
Representative compounds of this invention include, but are not limited to:
Figure imgf000105_0003
Figure imgf000106_0001
Figure imgf000107_0001
Figure imgf000108_0001
Figure imgf000108_0002
Figure imgf000108_0003
Figure imgf000109_0001
Figure imgf000109_0002
Figure imgf000109_0003
Figure imgf000110_0001
Figure imgf000110_0002
Figure imgf000110_0003
Figure imgf000111_0001
Figure imgf000112_0001
Figure imgf000112_0002
Figure imgf000112_0003
Figure imgf000112_0004
Figure imgf000113_0001
Figure imgf000113_0002
Figure imgf000113_0003
Figure imgf000114_0001
Figure imgf000114_0002
Representative compounds of this invention include, but are not limited to:
Figure imgf000115_0001
Figure imgf000116_0001
Figure imgf000117_0001
Figure imgf000118_0001
Figure imgf000119_0001
Figure imgf000120_0001
Figure imgf000121_0001
Figure imgf000121_0002
Figure imgf000121_0003
Figure imgf000121_0004
Figure imgf000122_0001
Figure imgf000122_0002
Figure imgf000122_0003
Representative compounds of this invention wherein hydrogen has been replaced by deuterium include, but are not limited to:
Figure imgf000123_0001
Figure imgf000123_0002
Figure imgf000123_0003
Figure imgf000123_0004
Figure imgf000124_0001
Figure imgf000124_0002
Figure imgf000124_0003
Figure imgf000124_0004
Figure imgf000125_0001
Figure imgf000125_0002
Figure imgf000125_0003
Figure imgf000125_0004
Figure imgf000126_0001
Figure imgf000126_0002
Figure imgf000126_0003
Figure imgf000126_0004
Figure imgf000127_0001
Figure imgf000127_0002
Another embodiment of this invention is directed to compound A1. Another embodiment of this invention is directed to compound A2. Another embodiment of this invention is directed to compound A3. Another embodiment of this invention is directed to compound A4. Another embodiment of this invention is directed to compound A5. Another embodiment of this invention is directed to compound A6. Another embodiment of this invention is directed to compound A7. Another embodiment of this invention is directed to compound A8. Another embodiment of this invention is directed to compound A9. Another embodiment of this invention is directed to compound A10. Another embodiment of this invention is directed to compound A11. Another embodiment of this invention is directed to compound A12. Another embodiment of this invention is directed to compound A13. Another embodiment of this invention is directed to compound A14. Another embodiment of this invention is directed to compound A15. Another embodiment of this invention is directed to compound A16. Another embodiment of this invention is directed to compound A18. Another embodiment of this invention is directed to compound A19.
Another embodiment of this invention is directed to compound A20.
Another embodiment of this invention is directed to compound A21.
Another embodiment of this invention is directed to compound A22. Another embodiment of this invention is directed to compound A23.
Another embodiment of this invention is directed to compound A24.
Another embodiment of this invention is directed to compound A25.
Another embodiment of this invention is directed to compound A26.
Another embodiment of this invention is directed to compound A27. Another embodiment of this invention is directed to compound A28.
Another embodiment of this invention is directed to compound A29.
Another embodiment of this invention is directed to compound A30.
Another embodiment of this invention is directed to compound A31.
Another embodiment of this invention is directed to compound A32. Another embodiment of this invention is directed to compound A33.
Another embodiment of this invention is directed to compound A34.
Another embodiment of this invention is directed to compound A35.
Another embodiment of this invention is directed to compound A36.
Another embodiment of this invention is directed to compound A37. Another embodiment of this invention is directed to compound A38.
Another embodiment of this invention is directed to compound A39.
Another embodiment of this invention is directed to compound A40.
Another embodiment of this invention is directed to compound A41.
Another embodiment of this invention is directed to compound A42. Another embodiment of this invention is directed to compound A43.
Another embodiment of this invention is directed to compound A44.
Another embodiment of this invention is directed to compound A45.
Another embodiment of this invention is directed to compound A46.
Another embodiment of this invention is directed to compound A47. Another embodiment of this invention is directed to compound A48.
Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A1.
Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A2. Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A3.
Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A4. Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A5.
Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A6.
Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A7.
Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A8.
Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A9. Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A10.
Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A11.
Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A12.
Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A13.
Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A14. Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A15.
Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A16.
Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A18.
Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A19.
Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A20. Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A21.
Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A22. Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A23.
Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A24.
Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A25.
Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A26.
Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A27. Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A28.
Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A29.
Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A30.
Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A31.
Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A32. Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A33.
Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A34.
Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A35.
Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A36.
Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A37. Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A38.
Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A39. Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A40.
Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A41.
Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A42.
Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A43.
Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A44. Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A45.
Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A46.
Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A47.
Another embodiment of this invention is directed to a pharmaceutically acceptable salt of compound A48.
Another embodiment of this invention is directed to a solvate of compound A1. Another embodiment of this invention is directed to a solvate of compound A2. Another embodiment of this invention is directed to solvate of compound A3.
Another embodiment of this invention is directed to solvate of compound A4. Another embodiment of this invention is directed to a solvate of compound A5. Another embodiment of this invention is directed to a solvate of compound A6. Another embodiment of this invention is directed to a solvate of compound A7. Another embodiment of this invention is directed to a solvate of compound A8.
Another embodiment of this invention is directed to a solvate of compound A9. Another embodiment of this invention is directed to a solvate of compound A10.
Another embodiment of this invention is directed to solvate of compound A11. Another embodiment of this invention is directed to a solvate of compound A12.
Another embodiment of this invention is directed to a solvate of compound A13. Another embodiment of this invention is directed to a solvate of compound
A14.
Another embodiment of this invention is directed to a solvate of compound A15.
Another embodiment of this invention is directed to a solvate of compound A16.
Another embodiment of this invention is directed to a solvate of compound A18.
Another embodiment of this invention is directed to a solvate of compound A19. Another embodiment of this invention is directed to a solvate of compound
A20.
Another embodiment of this invention is directed to a solvate of compound A21.
Another embodiment of this invention is directed to a solvate of compound A22.
Another embodiment of this invention is directed to a solvate of compound A23.
Another embodiment of this invention is directed to a solvate of compound A24. Another embodiment of this invention is directed to a solvate of compound
A25.
Another embodiment of this invention is directed to a solvate of compound A26.
Another embodiment of this invention is directed to a solvate of compound A27.
Another embodiment of this invention is directed to a solvate of compound A28.
Another embodiment of this invention is directed to a solvate of compound A29. Another embodiment of this invention is directed to a solvate of compound A30.
Another embodiment of this invention is directed to a solvate of compound A31. Another embodiment of this invention is directed to a solvate of compound
A32.
Another embodiment of this invention is directed to a solvate of compound A33.
Another embodiment of this invention is directed to a solvate of compound A34.
Another embodiment of this invention is directed to a solvate of compound A35.
Another embodiment of this invention is directed to a solvate of compound A36. Another embodiment of this invention is directed to a solvate of compound
A37.
Another embodiment of this invention is directed to a solvate of compound A38.
Another embodiment of this invention is directed to a solvate of compound A39.
Another embodiment of this invention is directed to a solvate of compound A40.
Another embodiment of this invention is directed to a solvate of compound A41. Another embodiment of this invention is directed to a solvate of compound
A42.
Another embodiment of this invention is directed to a solvate of compound A43.
Another embodiment of this invention is directed to a solvate of compound A44.
Another embodiment of this invention is directed to a solvate of compound A45.
Another embodiment of this invention is directed to a solvate of compound A46. Another embodiment of this invention is directed to a solvate of compound A47.
Another embodiment of this invention is directed to a solvate of compound A48. Other embodiments of this invention are directed to any one of the embodiments of formula 1.0 wherein the compound is in pure and isolated form.
Other embodiments of this invention are directed to any one of the embodiments of formula 1.0 wherein the compound is in pure form.
Other embodiments of this invention are directed to any one of the embodiments of formula 1.0 wherein the compound is in isolated form.
Other embodiments of this invention are directed to any one of the compounds of A1 to A16 and A18 to A48 in pure and isolated form.
Other embodiments of this invention are directed to any one of the compounds of A1 to A16 and A18 to A30 in pure and isolated form. Other embodiments of this invention are directed to any one of the compounds of A1 to A16 and A18 to A26 in pure and isolated form.
Other embodiments of this invention are directed to any one of the compounds of A31 to A48 in pure and isolated form.
Other embodiments of this invention are directed to any one of the compounds of A1 to A16 and A18 to A48 in pure form.
Other embodiments of this invention are directed to any one of the compounds of A1 to A16 and A18 to A30 in pure form.
Other embodiments of this invention are directed to any one of the compounds of A1 to A16 and A18 to A26 in pure form. Other embodiments of this invention are directed to any one of the compounds of A31 to A48 in pure.
Other embodiments of this invention are directed to any one of the compounds of A1 to A16 and A18 to A48 in isolated form.
Other embodiments of this invention are directed to any one of the compounds of A1 to A16 and A18 to A30 in isolated form.
Other embodiments of this invention are directed to any one of the compounds of A1 to A16 and A18 to A26 in isolated form.
Other embodiments of this invention are directed to any one of the compounds of A31 to A48 in isolated form. Another embodiment of this invention is directed to a pharmaceutical composition comprising an effective amount of at least one compound (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) of formula 1.0 (preferably of formula 1.1) and a pharmaceutically acceptable carrier. Another embodiment of this invention is directed to a pharmaceutical composition comprising an effective amount of a compound of formula 1.0 (preferably of formula 1.1 ) and a pharmaceutically acceptable carrier.
Another embodiment of this invention is directed to a pharmaceutical composition comprising an effective amount of at least one compound (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) selected from the group consisting of: A1 to A16 and A18 to A48, and a pharmaceutically acceptable carrier.
Another embodiment of this invention is directed to a pharmaceutical composition comprising an effective amount of at least one compound (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) selected from the group consisting of: A1 to A16 and A18 to A30, and a pharmaceutically acceptable carrier.
Another embodiment of this invention is directed to a pharmaceutical composition comprising an effective amount of at least one compound (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) selected from the group consisting of: A1 to A16 and A18 to A26, and a pharmaceutically acceptable carrier. Another embodiment of this invention is directed to a pharmaceutical composition comprising an effective amount of at least one compound (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) selected from the group consisting of: A31 to A48, and a pharmaceutically acceptable carrier.
Another embodiment of this invention is directed to a pharmaceutical composition comprising an effective amount of a compound selected from the group consisting of: A1 to A16 and A18 to A48, and a pharmaceutically acceptable carrier.
Another embodiment of this invention is directed to a pharmaceutical composition comprising an effective amount of a compound selected from the group consisting of: A1 to A16 and A18 to A30, and a pharmaceutically acceptable carrier. Another embodiment of this invention is directed to a pharmaceutical composition comprising an effective amount of a compound selected from the group consisting of: A1 to A16 and A18 to A26, and a pharmaceutically acceptable carrier. Another embodiment of this invention is directed to a pharmaceutical composition comprising an effective amount of a compound selected from the group consisting of: A31 to A48, and a pharmaceutically acceptable carrier.
Another embodiment of this invention is directed to a pharmaceutical composition comprising an effective amount of at least one compound (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) of formula 1.0 (preferably of formula 1.1), at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) other active pharmaceutically active ingredient, and a pharmaceutically acceptable carrier.
Another embodiment of this invention is directed to a pharmaceutical composition comprising an effective amount of a compound of formula 1.0 (preferably of formula 1.1), another active pharmaceutically active ingredient, and a pharmaceutically acceptable carrier.
Another embodiment of this invention is directed to a pharmaceutical composition comprising an effective amount of at least one compound (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) selected from the group consisting of: A1 to A16 and A18 to A48, at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) other active pharmaceutically active ingredient, and a pharmaceutically acceptable carrier.
Another embodiment of this invention is directed to a pharmaceutical composition comprising an effective amount of at least one compound (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) selected from the group consisting of: A1 to A16 and A18 to A30, at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) other active pharmaceutically active ingredient, and a pharmaceutically acceptable carrier.
Another embodiment of this invention is directed to a pharmaceutical composition comprising an effective amount of at least one compound (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) selected from the group consisting of: A1 to A16 and A18 to A26, at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) other active pharmaceutically active ingredient, and a pharmaceutically acceptable carrier.
Another embodiment of this invention is directed to a pharmaceutical composition comprising an effective amount of at least one compound (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) selected from the group consisting of: A31 to A48, at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) other active pharmaceutically active ingredient, and a pharmaceutically acceptable carrier.
Another embodiment of this invention is directed to a pharmaceutical composition comprising an effective amount of a compound selected from the group consisting of: A1 to A16 and A18 to A48, another active pharmaceutically active ingredient, and a pharmaceutically acceptable carrier.
Another embodiment of this invention is directed to a pharmaceutical composition comprising an effective amount of a compound selected from the group consisting of: A1 to A16 and A18 to A30, another active pharmaceutically active ingredient, and a pharmaceutically acceptable carrier.
Another embodiment of this invention is directed to a pharmaceutical composition comprising an effective amount of a compound selected from the group consisting of: A1 to A16 and A18 to A26, another active pharmaceutically active ingredient, and a pharmaceutically acceptable carrier.
Another embodiment of this invention is directed to a pharmaceutical composition comprising an effective amount of a compound selected from the group consisting of: A31 to A48, another active pharmaceutically active ingredient, and a pharmaceutically acceptable carrier. Another embodiment of this invention is directed to a pharmaceutical composition comprising an effective amount of at least one compound (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) of formula 1.0 (preferably of formula 1.1 ), at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) chemotherapeutic agent, and a pharmaceutically acceptable carrier. Another embodiment of this invention is directed to a pharmaceutical composition comprising an effective amount of a compound of formula 1.0 (preferably of formula 1.1), a chemotherapeutic agent, and a pharmaceutically acceptable carrier.
Another embodiment of this invention is directed to a pharmaceutical composition comprising an effective amount of at least one compound (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) selected from the group consisting of: A1 to A16 and A18 to A48, at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) chemotherapeutic agent, and a pharmaceutically acceptable carrier.
Another embodiment of this invention is directed to a pharmaceutical composition comprising an effective amount of at least one compound (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) selected from the group consisting of: A1 to A16 and A18 to A30, at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) chemotherapeutic agent, and a pharmaceutically acceptable carrier.
Another embodiment of this invention is directed to a pharmaceutical composition comprising an effective amount of at least one compound (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) selected from the group consisting of: A1 to A16 and A18 to A26, at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) chemotherapeutic agent, and a pharmaceutically acceptable carrier.
Another embodiment of this invention is directed to a pharmaceutical composition comprising an effective amount of at least one compound (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) selected from the group consisting of: A31 to A48, at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) chemotherapeutic agent, and a pharmaceutically acceptable carrier.
Another embodiment of this invention is directed to a pharmaceutical composition comprising an effective amount of a compound selected from the group consisting of: A1 to A16 and A18 to A48, a chemotherapeutic agent, and a pharmaceutically acceptable carrier.
Another embodiment of this invention is directed to a pharmaceutical composition comprising an effective amount of a compound selected from the group consisting of: A1 to A16 and A18 to A30, a chemotherapeutic agent, and a pharmaceutically acceptable carrier.
Another embodiment of this invention is directed to a pharmaceutical composition comprising an effective amount of a compound selected from the group consisting of: A1 to A16 and A18 to A26, a chemotherapeutic agent, and a pharmaceutically acceptable carrier.
Another embodiment of this invention is directed to a pharmaceutical composition comprising an effective amount of a compound selected from the group consisting of: A31 to A48, a chemotherapeutic agent, and a pharmaceutically acceptable carrier. Another embodiment of this invention is directed to a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) compound of formula 1.0 (preferably of formula 1.6).
Another embodiment of this invention is directed to a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of one compound of formula 1.0 (preferably of formula 1.6).
Another embodiment of this invention is directed to a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (1 , 2 or 3, or 1 or 2, or 1 , and usually 1) compound of A1 to A16 and A18 to A48.
Another embodiment of this invention is directed to a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (1 , 2 or 3, or 1 or 2, or 1 , and usually 1) compound of A1 to A16 and A18 to A30.
Another embodiment of this invention is directed to a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (1 , 2 or 3, or 1 or 2, or 1 , and usually 1) compound of A1 to A16 and A18 to A26.
Another embodiment of this invention is directed to a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) compound of A31 to A48. Another embodiment of this invention is directed to a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of one compound of A1 to A16 and A18 to A48.
Another embodiment of this invention is directed to a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of one compound of A1 to A16 and A18 to A30.
Another embodiment of this invention is directed to a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of one compound of A1 to A16 and A18 to A26.
Another embodiment of this invention is directed to a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of one compound of A31 to A48.
Another embodiment of this invention is directed to a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) compound of formula 1.0, and an effective amount of at least one (1 , 2 or 3, or 1 or 2, or 1 , and usually 1) chemotherapeutic agent.
Another embodiment of this invention is directed to a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of one compound of formula 1.0, and an effective amount of at least one (1 , 2 or 3, or 1 or 2, or 1 , and usually 1) chemotherapeutic agent.
Another embodiment of this invention is directed to a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (1 , 2 or 3, or 1 or 2, or 1 , and usually 1) compound of A1 to A16 and A18 to A48, and an effective amount of at least one (1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) chemotherapeutic agent.
Another embodiment of this invention is directed to a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) compound of A1 to A16 and A18 to A30, and an effective amount of at least one (1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) chemotherapeutic agent.
Another embodiment of this invention is directed to a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) compound of A1 to A16 and A18 to A26, and an effective amount of at least one (1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) chemotherapeutic agent.
Another embodiment of this invention is directed to a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) compound of A31 to A48, and an effective amount of at least one (1 , 2 or 3, or 1 or 2, or 1 , and usually 1) chemotherapeutic agent.
Another embodiment of this invention is directed to a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of one compound of A1 to A16 and A18 to A48, and an effective amount of at least one (1 , 2 or 3, or 1 or 2, or 1 , and usually 1) chemotherapeutic agent.
Another embodiment of this invention is directed to a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of one compound of A1 to A16 and A18 to A30, and an effective amount of at least one (1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) chemotherapeutic agent.
Another embodiment of this invention is directed to a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of one compound of A1 to A16 and A18 to A26, and an effective amount of at least one (1 , 2 or 3, or 1 or 2, or 1 , and usually 1) chemotherapeutic agent.
Another embodiment of this invention is directed to a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of one compound of A31 to A48, and an effective amount of at least one (1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) chemotherapeutic agent.
The compounds of this invention inhibit the activity of ERK1 and ERK2 Thus, this invention further provides a method of inhibiting ERK in mammals, especially humans, by the administration of an effective amount (e.g., a therapeutically effective amount) of one or more (e.g., one) compounds of this invention. The administration of the compounds of this invention to patients, to inhibit ERK1 and/or ERK2, is useful in the treatment of cancer.
In any of the methods of treating cancer described herein, unless stated otherwise, the methods can optionally include the administration of an effective amount of one or more (e.g., 1 , 2 or 3, or 1 or 2, or 1) chemotherapeutic agents. The chemotherapeutic agents can be administered currently or sequentially with the compounds of this invention.
The methods of treating cancer described herein include methods wherein a combination of drugs (i.e., compounds, or pharmaceutically active ingredients, or pharmaceutical compositions) are used (i.e., the methods of treating cancer of this invention include combination therapies). Those skilled in the art will appreciate that the drugs are generally administered individually as a pharmaceutical composition. The use of a pharmaceutical composition comprising more than one drug is within the scope of this invention.
In any of the methods of treating cancer described herein, unless stated otherwise, the methods can optionally include the administration of an effective amount of radiation therapy. For radiation therapy, γ-radiation is preferred.
Examples of cancers which may be treated by the methods of this invention include, but are not limited to: (A) lung cancer (e.g., lung adenocarcinoma and non small cell lung cancer), (B) pancreatic cancers (e.g., pancreatic carcinoma such as, for example, exocrine pancreatic carcinoma), (C) colon cancers (e.g., colorectal carcinomas, such as, for example, colon adenocarcinoma and colon adenoma), (D) myeloid leukemias (for example, acute myelogenous leukemia (AML), CML, and CMML), (E) thyroid cancer, (F) myelodysplastic syndrome (MDS), (G) bladder carcinoma, (H) epidermal carcinoma, (I) melanoma, (J) breast cancer, (K) prostate cancer, (L) head and neck cancers (e.g., squamous cell cancer of the head and neck), (M) ovarian cancer, (N) brain cancers (e.g., gliomas, such as glioma blastoma multiforme), (O) cancers of mesenchymal origin (e.g., fibrosarcomas and rhabdomyosarcomas), (P) sarcomas, (Q) tetracarci nomas, (R) nuroblastomas, (S) kidney carcinomas, (T) hepatomas, (U) non-Hodgkin's lymphoma, (V) multiple myeloma, and (W) anaplastic thyroid carcinoma. Thus, another embodiment of this invention is directed to a method for treating lung cancer, pancreatic cancer, colon cancer (e.g., colorectal cancer), myeloid leukemias (e.g., AML, CML, and CMML), thyroid cancer, myelodysplastic syndrome (MDS), bladder carcinoma, epidermal carcinoma, melanoma, breast cancer, prostate cancer, head and neck cancers (e.g., squamous cell cancer of the head and neck), ovarian cancer, brain cancers (e.g., gliomas, such as glioma blastoma multiforme), cancers of mesenchymal origin (e.g., fibrosarcomas and rhabdomyosarcomas), sarcomas, tetracarcinomas, nuroblastomas, kidney carcinomas, hepatomas, non- Hodgkin's lymphoma, multiple myeloma, or anaplastic thyroid carcinoma, in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0.
Another embodiment of this invention is directed to a method for treating lung cancer, pancreatic cancer, colon cancer (e.g., colorectal cancer), myeloid leukemias (e.g., AML, CML, and CMML), thyroid cancer, myelodysplastic syndrome (MDS), bladder carcinoma, epidermal carcinoma, melanoma, breast cancer, prostate cancer, head and neck cancers (e.g., squamous cell cancer of the head and neck), ovarian cancer, brain cancers (e.g., gliomas, such as glioma blastoma multiforme), cancers of mesenchymal origin (e.g., fibrosarcomas and rhabdomyosarcomas), sarcomas, tetracarcinomas, nuroblastomas, kidney carcinomas, hepatomas, non-Hodgkin's lymphoma, multiple myeloma, or anaplastic thyroid carcinoma in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1) chemotherapeutic agent. Another embodiment of this invention is directed to a method for treating lung cancer, pancreatic cancer, colon cancer (e.g., colorectal cancer), myeloid leukemias (e.g., AML, CML, and CMML), thyroid cancer, myelodysplastic syndrome (MDS), bladder carcinoma, epidermal carcinoma, melanoma, breast cancer, prostate cancer, head and neck cancers (e.g., squamous cell cancer of the head and neck), ovarian cancer, brain cancers (e.g., gliomas, such as glioma blastoma multiforme), cancers of mesenchymal origin (e.g., fibrosarcomas and rhabdomyosarcomas), sarcomas, tetracarcinomas, nuroblastomas, kidney carcinomas, hepatomas, non-Hodgkin's lymphoma, multiple myeloma, or anaplastic thyroid carcinoma in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
Another embodiment of this invention is directed to a method for treating lung cancer, pancreatic cancer, colon cancer (e.g., colorectal cancer), myeloid leukemias (e.g., AML, CML, and CMML), thyroid cancer, myelodysplastic syndrome (MDS), bladder carcinoma, epidermal carcinoma, melanoma, breast cancer, prostate cancer, head and neck cancers (e.g., squamous cell cancer of the head and neck), ovarian cancer, brain cancers (e.g., gliomas, such as glioma blastoma multiforme), cancers of mesenchymal origin (e.g., fibrosarcomas and rhabdomyosarcomas), sarcomas, tetracarcinomas, nuroblastomas, kidney carcinomas, hepatomas, non-Hodgkin's lymphoma, multiple myeloma, or anaplastic thyroid carcinoma in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
Another embodiment of this invention is directed to a method for treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, wherein said cancer is selected from the group consisting of: melanoma, pancreatic cancer, thryroid cancer, colorectal cancer, lung cancer, breast cancer, and ovarian cancer.
Another embodiment of this invention is directed to a method for treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1) chemotherapeutic agent wherein said cancer is selected from the group consisting of: melanoma, pancreatic cancer, thryroid cancer, colorectal cancer, lung cancer, breast cancer, and ovarian cancer.
Another embodiment of this invention is directed to a method for treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0, wherein said cancer is selected from the group consisting of: melanoma, pancreatic cancer, thryroid cancer, colorectal cancer, lung cancer, breast cancer, and ovarian cancer.
Another embodiment of this invention is directed to a method for treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1) chemotherapeutic agent wherein said cancer is selected from the group consisting of: melanoma, pancreatic cancer, thryroid cancer, colorectal cancer, lung cancer, breast cancer, and ovarian cancer.
Another embodiment of this invention is directed to a method for treating melanoma in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0. Another embodiment of this invention is directed to a method for treating melanoma, in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent. Another embodiment of this invention is directed to a method for treating melanoma in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0. Another embodiment of this invention is directed to a method for treating melanoma in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
Another embodiment of this invention is directed to a method for treating pancreatic cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
Another embodiment of this invention is directed to a method for treating pancreatic cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1) chemotherapeutic agent.
Another embodiment of this invention is directed to a method for treating pancreatic cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0.
Another embodiment of this invention is directed to a method for treating pancreatic cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
Another embodiment of this invention is directed to a method for treating thyroid cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
Another embodiment of this invention is directed to a method for treating thyroid cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1) chemotherapeutic agent.
Another embodiment of this invention is directed to a method for treating thyroid cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
Another embodiment of this invention is directed to a method for treating thyroid cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1) chemotherapeutic agent.
Another embodiment of this invention is directed to a method for treating colorectal cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0.
Another embodiment of this invention is directed to a method for treating colorectal cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1) chemotherapeutic agent.
Another embodiment of this invention is directed to a method for treating colorectal cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0.
Another embodiment of this invention is directed to a method for treating colorectal cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent. Another embodiment of this invention is directed to a method for treating lung cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0. Another embodiment of this invention is directed to a method for treating lung cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent. Another embodiment of this invention is directed to a method for treating lung cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0. Another embodiment of this invention is directed to a method for treating lung cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
Another embodiment of this invention is directed to a method for treating breast cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0. Another embodiment of this invention is directed to a method for treating breast cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent. Another embodiment of this invention is directed to a method for treating breast cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0. Another embodiment of this invention is directed to a method for treating breast cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
Another embodiment of this invention is directed to a method for treating ovarian cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
This invention also provides a method for treating ovarian cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
Another embodiment of this invention is directed to a method for treating ovarian cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0.
Another embodiment of this invention is directed to a method for treating ovarian cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
Other embodiments of this invention are directed to methods of treating breast cancer (i.e., post-menopausal and premenopausal breast cancer, e.g., hormone- dependent breast cancer) in a patient in need of such treatment, said treatment comprising the administration of an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0 in combination with hormonal therapies (i.e., antihormonal agents).
Other embodiments of this invention are directed to methods of treating breast cancer (i.e., post-menopausal and premenopausal breast cancer, e.g., hormone- dependent breast cancer) in a patient in need of such treatment, said treatment comprising the administration of an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0 in combination with hormonal therapies (i.e., antihormonal agents).
Other embodiments of this invention are directed to methods of treating breast cancer (i.e., post-menopausal and premenopausal breast cancer, e.g., hormone- dependent breast cancer) in a patient in need of such treatment, said treatment comprising the administration of an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0 in combination with hormonal therapies (i.e., antihormonal agents), and in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1) chemotherapeutic agent.
Other embodiments of this invention are directed to methods of treating breast cancer (i.e., post-menopausal and premenopausal breast cancer, e.g., hormone- dependent breast cancer) in a patient in need of such treatment, said treatment comprising the administration of an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0 in combination with hormonal therapies (i.e., antihormonal agents), and in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
The methods of treating breast cancer described herein include the treatment of hormone-dependent metastatic and advanced breast cancer, adjuvant therapy for hormone-dependent primary and early breast cancer, the treatment of ductal carcinoma in situ, and the treatment of inflammatory breast cancer in situ. The methods of treating hormone-dependent breast cancer can also be used to prevent breast cancer in patients having a high risk of developing breast cancer.
Thus, other embodiment of this invention are directed to methods of preventing breast cancer (i.e., post-menopausal and premenopausal breast cancer, e.g., hormone-dependent breast cancer) in a patient in need of such treatment, said treatment comprising the administration of an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0 in combination with hormonal therapies (i.e., antihormonal agents).
Other embodiments of this invention are directed to methods of preventing breast cancer (i.e., post-menopausal and premenopausal breast cancer, e.g., hormone-dependent breast cancer) in a patient in need of such treatment, said treatment comprising the administration of an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0 in combination with hormonal therapies (i.e., antihormonal agents).
Other embodimenst of this invention are directed to methods of preventing breast cancer (i.e., post-menopausal and premenopausal breast cancer, e.g., hormone-dependent breast cancer) in a patient in need of such treatment, said treatment comprising the administration of an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0 in combination with hormonal therapies (i.e., antihormonal agents), and in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1) chemotherapeutic agent.
Other embodiments of this invention are directed to methods of preventing breast cancer (i.e., post-menopausal and premenopausal breast cancer, e.g., hormone-dependent breast cancer) in a patient in need of such treatment, said treatment comprising the administration of an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0 in combination with hormonal therapies (i.e., antihormonal agents), and in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1) chemotherapeutic agent.
Another embodiment of this invention is directed to a method for treating brain cancer (e.g., glioma, such as glioma blastoma multiforme) in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0. Another embodiment of this invention is directed to a method for treating brain cancer (e.g., glioma, such as glioma blastoma multiforme) in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1) chemotherapeutic agent.
Another embodiment of this invention is directed to a method for treating brain cancer (e.g., glioma, such as glioma blastoma multiforme) a in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
Another embodiment of this invention is directed to a method for treating brain cancer (e.g., glioma, such as glioma blastoma multiforme) in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1) chemotherapeutic agent. Another embodiment of this invention is directed to a method for treating brain cancer (e.g., glioma, such as glioma blastoma multiforme) in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of a chemotherapeutic agent wherein said chemotherapeutic agent is temozolomide. Another embodiment of this invention is directed to a method for treating brain cancer (e.g., glioma, such as glioma blastoma multiforme) in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of a chemotherapeutic agent, wherein said chemotherapeutic agent is temozolomide.
Another embodiment of this invention is directed to a method for treating prostate cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
Another embodiment of this invention is directed to a method for treating prostate cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
Another embodiment of this invention is directed to a method for treating prostate cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
Another embodiment of this invention is directed to a method for treating prostate cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1) chemotherapeutic agent.
Another embodiment of this invention is directed to a method for treating myelodysplastic syndrome in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
Another embodiment of this invention is directed to a method for treating myelodysplastic syndrome in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
Another embodiment of this invention is directed to a method for treating myelodysplastic syndrome in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
Another embodiment of this invention is directed to a method for treating myelodysplastic syndrome in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1 , in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
Another embodiment of this invention is directed to a method for treating myeloid leukemias in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
Another embodiment of this invention is directed to a method for treating myeloid leukemias in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
Another embodiment of this invention is directed to a method for treating myeloid leukemias in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0.
Another embodiment of this invention is directed to a method for treating myeloid leukemias in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent. Another embodiment of this invention is directed to a method for treating acute myelogenous leukemia (AML) in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
Another embodiment of this invention is directed to a method for treating acute myelogenous leukemia (AML) in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
Another embodiment of this invention is directed to a method for treating acute myelogenous leukemia (AML)in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
Another embodiment of this invention is directed to a method for treating acute myelogenous leukemia (AML)in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1) chemotherapeutic agent. Another embodiment of this invention is directed to a method for treating chronic myelomonocytic leukemia (CMML) in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0. Another embodiment of this invention is directed to a method for treating chronic myelomonocytic leukemia (CMML) in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1) chemotherapeutic agent. Another embodiment of this invention is directed to a method for treating chronic myelomonocytic leukemia (CMML) in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0. Another embodiment of this invention is directed to a method for treating chronic myelomonocytic leukemia (CMML) in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1) chemotherapeutic agent.
Another embodiment of this invention is directed to a method for treating chronic myelogenous leukemia (chronic myeloid leukemia, CML) in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0. Another embodiment of this invention is directed to a method for treating chronic myelogenous leukemia (chronic myeloid leukemia, CML) in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1) chemotherapeutic agent.
Another embodiment of this invention is directed to a method for treating chronic myelogenous leukemia (chronic myeloid leukemia, CML) in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0.
Another embodiment of this invention is directed to a method for treating chronic myelogenous leukemia (chronic myeloid leukemia, CML) in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1) chemotherapeutic agent. Another embodiment of this invention is directed to a method for treating myeloid leukemias in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
Another embodiment of this invention is directed to a method for treating myeloid leukemias in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
Another embodiment of this invention is directed to a method for treating myeloid leukemias in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
Another embodiment of this invention is directed to a method for treating myeloid leukemias in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent. Another embodiment of this invention is directed to a method for treating bladder cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0. Another embodiment of this invention is directed to a method for treating bladder cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1) chemotherapeutic agent.
Another embodiment of this invention is directed to a method for treating bladder cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
Another embodiment of this invention is directed to a method for treating bladder cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
Another embodiment of this invention is directed to a method for treating non- Hodgkin's lymphoma in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
Another embodiment of this invention is directed to a method for treating non- Hodgkin's lymphoma in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
Another embodiment of this invention is directed to a method for treating non- Hodgkin's lymphoma in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
Another embodiment of this invention is directed to a method for treating non- Hodgkin's lymphoma in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
Another embodiment of this invention is directed to a method for treating multiple myeloma in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0.
Another embodiment of this invention is directed to a method for treating multiple myeloma in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
Another embodiment of this invention is directed to a method for treating multiple myeloma in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1) compound of formula 1.0.
Another embodiment of this invention is directed to a method for treating multiple myeloma in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a pharmaceutical composition comprising an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, and usually 1 ) compound of formula 1.0, in combination with an effective amount of at least one (e.g., 1 , 2 or 3, 1 or 2, or 1 ) chemotherapeutic agent.
Chemotherapeutic agents (antineoplastic agent) include but are not limited to: microtubule affecting agents, alkylating agents, antimetabolites, natural products and their derivatives, hormones and steroids (including synthetic analogs), and synthetics.
Examples of alkylating agents (including nitrogen mustards, ethylenimine derivatives, alkyl sulfonates, nitrosoureas and triazenes) include: Uracil mustard, Chlormethine, Cyclophosphamide (Cytoxan®), Ifosfamide, Melphalan, Chlorambucil, Pipobroman, Triethylene-melamine, Triethylenethiophosphoramine, Busulfan, Carmustine, Lomustine, Streptozocin, Dacarbazine, and Temozolomide.
Examples of antimetabolites (including folic acid antagonists, pyrimidine analogs, purine analogs and adenosine deaminase inhibitors) include: Methotrexate, 5-Fluorouracil, Floxuridine, Cytarabine, 6-Mercaptopurine, 6-Thioguanine, Fludarabine phosphate, Pentostatine, and Gemcitabine. Examples of natural products and their derivatives (including vinca alkaloids, antitumor antibiotics, enzymes, lymphokines and epipodophyllotoxins) include: Vinblastine, Vincristine, Vindesine, Bleomycin, Dactinomycin, Daunorubicin, Doxorubicin, Epirubicin, Idarubicin, Paclitaxel (paclitaxel is a microtubule affecting agent and is commercially available as Taxol®), Paclitaxel derivatives (e.g. taxotere), Mithramycin, Deoxyco-formycin, Mitomycin-C, L-Asparaginase, Interferons (especially IFN-a), Etoposide, and Teniposide.
Examples of hormones and steroids (including synthetic analogs) include: 17α- Ethinylestradiol, Diethylstilbestrol, Testosterone, Prednisone, Fluoxymesterone, Dromostanolone propionate, Testolactone, Megestrolacetate, Tamoxifen, Methylprednisolone, Methyl-testosterone, Prednisolone, Triamcinolone, Chlorotrianisene, Hydroxyprogesterone, Aminoglutethimide, Estramustine, Medroxyprogesteroneacetate, Leuprolide, Flutamide, Toremifene, and Zoladex. Examples of synthetics (including inorganic complexes such as platinum coordination complexes): Cisplatin, Carboplatin, Hydroxyurea, Amsacrine,
Procarbazine, Mitotane, Mitoxantrone, Levamisole, and Hexamethylmelarnine.
Examples of other chemotherapeutics include: Navelbene, CPT-11 , Anastrazole, Letrazole, Capecitabinbe, Reloxafine, and Droloxafine.
A microtubule affecting agent (e.g., paclitaxel, a paclitaxel derivative or a paclitaxel-like compound), as used herein, is a compound that interferes with cellular mitosis, i.e., having an anti-mitotic effect, by affecting microtubule formation and/or action. Such agents can be, for instance, microtubule stabilizing agents or agents which disrupt microtubule formation.
Microtubule affecting agents, useful in the methods of this invention, are well known to those skilled in the art and include, but are not limited to: Allocolchicine (NSC 406042), Halichondrin B (NSC 609395), Colchicine (NSC 757), Colchicine derivatives (e.g., NSC 33410), Dolastatin 10 (NSC 376128), Maytansine (NSC 153858), Rhizoxin (NSC 332598), Paclitaxel (Taxol®, NSC 125973), Paclitaxel derivatives (e.g., Taxotere, NSC 608832), Thiocolchicine (NSC 361792), Trityl Cysteine (NSC 83265), Vinblastine Sulfate (NSC 49842), Vincristine Sulfate (NSC 67574), Epothilone A, Epothilone, Discodermolide (see Service, (1996) Science, 274:2009), Estramustine, Nocodazole, MAP4, and the like. Examples of such agents are described in, for example, Bulinski (1997) J. Cell Sci. 110:3055-3064, Panda (1997) Proc. Natl. Acad. Sci. USA 94:10560-10564, Muhlradt (1997) Cancer Res. 57:3344-3346, Nicolaou (1997) Nature 387:268-272, Vasquez (1997) MoI. Biol. Cell. 8:973-985, and Panda (1996) J. Biol. Chem. 271 :29807-29812.
Chemotherapeutic agents with paclitaxel-like activity include, but are not limited to, paclitaxel and paclitaxel derivatives (paclitaxel-like compounds) and analogues. Paclitaxel and its derivatives (e.g. Taxol and Taxotere) are available commercially. In addition, methods of making paclitaxel and paclitaxel derivatives and analogues are well known to those of skill in the art (see, e.g., U.S. Patent Nos: 5,569,729; 5,565,478; 5,530,020; 5,527,924; 5,508,447; 5,489,589; 5,488,116; 5,484,809; 5,478,854; 5,478,736; 5,475,120; 5,468,769; 5,461 ,169; 5,440,057; 5,422,364; 5,411 ,984; 5,405,972; and 5,296,506).
More specifically, the term "paclitaxel" as used herein refers to the drug commercially available as Taxol® (NSC number: 125973). Taxol® inhibits eukaryotic cell replication by enhancing polymerization of tubulin moieties into stabilized microtubule bundles that are unable to reorganize into the proper structures for mitosis. Of the many available chemotherapeutic drugs, paclitaxel has generated interest because of its efficacy in clinical trials against drug-refractory tumors, including ovarian and mammary gland tumors (Hawkins (1992) Oncology, 6: 17-23, Horwitz (1992) Trends Pharmacol. Sci. 13: 134-146, Rowinsky (1990) J. Natl. Cane. Inst. 82: 1247-1259). Additional microtubule affecting agents can be assessed using one of many such assays known in the art, e.g., a semiautomated assay which measures the tubulin-polymerizing activity of paclitaxel analogs in combination with a cellular assay to measure the potential of these compounds to block cells in mitosis (see Lopes (1997) Cancer Chemother. Pharmacol. 41 :37-47). Generally, activity of a test compound is determined by contacting a cell with that compound and determining whether or not the cell cycle is disrupted, in particular, through the inhibition of a mitotic event. Such inhibition may be mediated by disruption of the mitotic apparatus, e.g., disruption of normal spindle formation. Cells in which mitosis is interrupted may be characterized by altered morphology (e.g., microtubule compaction, increased chromosome number, etc.).
Compounds with possible tubulin polymerization activity can be screened in vitro. For example, the compounds are screened against cultured WR21 cells (derived from line 69-2 wap-ras mice) for inhibition of proliferation and/or for altered cellular morphology, in particular for microtubule compaction. In vivo screening of positive-testing compounds can then be performed using nude mice bearing the WR21 tumor cells. Detailed protocols for this screening method are described by Porter (1995) Lab. Anim. ScL, 45(2): 145-150.
Other methods of screening compounds for desired activity are well known to those of skill in the art. Typically such assays involve assays for inhibition of microtubule assembly and/or disassembly. Assays for microtubule assembly are described, for example, by Gaskin et al. (1974) J. Molec. Biol., 89: 737-758. U.S. Patent No. 5,569,720 also provides in vitro and in vivo assays for compounds with paclitaxel-like activity. Thus, in the methods of this invention wherein at least one chemotherapeutic agent is used, examples of said chemotherapeutic agents include those selected from the group consisting of: microtubule affecting agents, alkylating agents, antimetabolites, natural products and their derivatives, hormones and steroids (including synthetic analogs), and synthetics. In the methods of this invention wherein at least one chemotherapeutic agent is used, examples of said chemotherapeutic agents also include: (1) taxanes, (2) platinum coordinator compounds, (3) epidermal growth factor (EGF) inhibitors that are antibodies, (4) EGF inhibitors that are small molecules, (5) vascular endolithial growth factor (VEGF) inhibitors that are antibodies, (6) VEGF kinase inhibitors that are small molecules, (7) estrogen receptor antagonists or selective estrogen receptor modulators (SERMs), (8) anti-tumor nucleoside derivatives, (9) epothilones, (10) topoisomerase inhibitors, (11 ) vinca alkaloids, (12) antibodies that are inhibitors of αVβ3 integrins, (13) folate antagonists, (14) ribonucleotide reductase inhibitors, (15) anthracyclines, (16) biologies; (17) inhibitors of angiogenesis and/or suppressors of tumor necrosis factor alpha (TNF-alpha) such as thalidomide (or related imid), (18) Bcr/abl kinase inhibitors, (19) MEK1 and/or MEK 2 inhibitors that are small molecules, (20) IGF-1 and IGF-2 inhibitors that are small molecules, (21 ) small molecule inhibitors of RAF and BRAF kinases, (22) small molecule inhibitors of cell cycle dependent kinases such as CDK1 , CDK2, CDK4 and CDK6, (23) alkylating agents, and (24) farnesyl protein transferase inhibitors (also know as FPT inhibitors or FTI (i.e., farnesyl transfer inhibitors)).
In the methods of this invention wherein at least one chemotherapeutic agent is used, examples of such chemotherapeutic agents include:
(1) taxanes such as paclitaxel (TAXOL®) and/or docetaxel (Taxotere®); (2) platinum coordinator compounds, such as, for example, carboplatin, cisplatin and oxaliplatin (e.g. Eloxatin);
(3) EGF inhibitors that are antibodies, such as: HER2 antibodies (such as, for example trastuzumab (Herceptin®), Genentech, Inc.), Cetuximab (Erbitux, IMC-C225, ImClone Systems), EMD 72000 (Merck KGaA), anti-EFGR monoclonal antibody ABX (Abgenix), TheraCIM-h-R3 (Center of Molecular Immunology), monoclonal antibody 425 (Merck KGaA), monoclonal antibody ICR-62 (ICR, Sutton, England); Herzyme (Elan Pharmaceutical Technologies and Ribozyme Pharmaceuticals), PKI 166 (Novartis), EKB 569 (Wyeth-Ayerst), GW 572016 (GlaxoSmithKline), Cl 1033 (Pfizer Global Research and Development), trastuzmab-maytansinoid conjugate (Genentech, Inc.), mitumomab (Imclone Systems and Merck KGaA) and Melvax Il (Imclone Systems and Merck KgaA);
(4) EGF inhibitors that are small molecules, such as, Tarceva (TM) (OSI-774, OSI Pharmaceuticals, Inc.), and lressa (ZD 1839, Astra Zeneca); (5) VEGF inhibitors that are antibodies such as: bevacizumab (Genentech,
Inc.), and IMC-1C11 (ImClone Systems), DC 101 (a KDR VEGF Receptor 2 from ImClone Systems);
(6) VEGF kinase inhibitors that are small molecules such as SU 5416 (from Sugen, Inc), SU 6688 (from Sugen, Inc.), Bay 43-9006 (a dual VEGF and bRAF inhibitor from Bayer Pharmaceuticals and Onyx Pharmaceuticals);
(7) estrogen receptor antagonists or selective estrogen receptor modulators (SERMs), such as tamoxifen, idoxifene, raloxifene, trans-2,3-dihydroraloxifene, levormeloxifene, droloxifene, MDL 103,323, and acolbifene (Schering Corp.);
(8) anti-tumor nucleoside derivatives such as 5-fluorouracil, gemcitabine, capecitabine, cytarabine (Ara-C), fludarabine (F-Ara-A), decitabine, and chlorodeoxyadenosine (Cda, 2-Cda);
(9) epothilones such as BMS-247550 (Bristol-Myers Squibb), and EPO906 (Novartis Pharmaceuticals);
(10) topoisomerase inhibitors such as topotecan (Glaxo SmithKline), and Camptosar (Pharmacia);
(11) vinca alkaloids, such as, navelbine (Anvar and Fabre, France), vincristine and vinblastine; (12) antibodies that are inhibitors of αVβ3 integrins, such as, LM-609 (see, Clinical Cancer Research, Vol. 6, page 3056-3061 , August 2000, the disclosure of which is incorporated herein by reference thereto);
(13) folate antagonists, such as Methotrexate (MTX), and Premetrexed (Alimta);
(14) ribonucleotide reductase inhibitors, such as Hydroxyurea (HU);
(15) anthracyclines, such as Daunorubicin, Doxorubicin (Adriamycin), and Idarubicin;
(16) biologies, such as interferon (e.g., Intron-A and Roferon), pegylated interferon (e.g., Peg-lntron and Pegasys), and Rituximab (Rituxan, antibody used for the treatment of non-Hodgkin's lymphoma);
(17) thalidomide (or related imid);
(18) Bcr/abl kinase inhibitors, such as, for example Gleevec (STI-571 ), AMN- 17, ONO12380, SU11248 (Sunitinib) and BMS-354825 (19) MEK1 and/or MEK2 inhibitors, such as PD0325901 and Arry-142886
(AZD6244);
(20) IGF-1 and IGF-2 inhibitors that are small molecules, such as, for example, NVP-AEW541 ;
(21 ) small molecule inhibitors of RAF and BRAF kinases, such as, for example, BAY 43-9006 (Sorafenib);
(22) small molecule inhibitors of cell cycle dependent kinases such as CDK1 , CDK2, CDK4 and CDK6, such as, for example, CYC202, BMS387032, and Flavopiridol;
(23) alkylating agents, such as, for example, Temodar® brand of temozolomide;
(24) famesyl protein transferase inhibitors, such as, for example:
(a) Sarasar® brand of lonifarnib (i.e., 4-[2-[4-(3,10-dibromo-8-chloro-
6,11 -dihydro-5H-benzo[5,6]cyclohepta[1 ,2-b]byridin-11 -yl)-1 -piperidinyl)-2-oxoethyl]-1- piperidinecarboxamide, see for example, U.S. 5,874,442 issued February 23, 1999, and U.S. 6,632,455 issued October 14, 2003 the disclosures of each being incorporated herein by reference thereto),
(b) Zarnestra® brand of tipifamib (i.e., (R)-6-arπino[(4-chlorophenyl)(1 - methyl-1 H-imidazol-5-yl)methyl]-4-(3-chlorophenyl )-1 - methyl-2(1 H)-quinolinone, see for example, WO 97/16443 published May 9, 1997 and U.S. 5,968,952 issued October 19, 1999, the disclosures of each being incorporated herein by reference thereto), and
(c) Bristol-Myers Squibb 214662:
Figure imgf000163_0001
(see WO97/30992 published August 28, 1997, U.S. 6,011 ,029 issued January 4, 2000, and U.S. 6,455,523, the disclosures of each being incorporated herein by reference thereto).
The Bcr/abl kinase inhibitors, EGF receptor inhibitors, and HER-2 antibodies (EGF receptor inhibitors that are antibodies) described above are also known as signal transduction inhibitors. Therefore, chemotherapeutic agents, as used herein, include signal transduction inhibitors.
Typical signal transduction inhibitors, that are chemotherapeutic agents, include but are not limited to: (i) Bcr/abl kinase inhibitors such as, for example, STI 571 (Gleevec), (ii) Epidermal growth factor (EGF) receptor inhibitor such as, for example, Kinase inhibitors (Iressa, OSI-774) and antibodies (Imclone: C225
[Goldstein et al. (1995), Clin Cancer Res. 1 :1311 -1318], and Abgenix: ABX-EGF) and (iii) HER-2/neu receptor inhibitors such as, for example, Herceptin® (trastuzumab).
Methods for the safe and effective administration of most of these chemotherapeutic agents are known to those skilled in the art. In addition, their administration is described in the standard literature. For example, the administration of many of the chemotherapeutic agents is described in the "Physicians' Desk Reference" (PDR), e.g., 1996 edition (Medical Economics Company, Montvale, NJ 07645-1742, USA), the Physician's Desk Reference, 56th Edition, 2002 (published by Medical Economics company, Inc. Montvale, NJ 07645-1742), the Physician's Desk Reference, 57th Edition, 2003 (published by Thompson PDR, Montvale, NJ 07645- 1742); and the Physician's Desk Reference, 60th Edition, 2006 (published by Thompson PDR, Montvale, NJ 07645-1742); the disclosures of which are incorporated herein by reference thereto.
For example, the compound of formula 1.0 (e.g., a pharmaceutical composition comprising the compound of formula 1.0); can be administered orally (e.g., as a capsule), and the chemotherapeutic agents can be administered intravenously, usually as an IV solution. The use of a pharmaceutical composition comprising more than one drug is within the scope of this invention.
The compound of formula 1.0 and the chemotherapeutic agents are administered in therapeutically effective dosages to obtain clinically acceptable results, e.g., reduction or elimination of symptoms or of the tumor. Thus, the compound of formula 1.0 and chemotherapeutic agents can be administered concurrently or consecutively in a treatment protocol. The administration of the chemotherapeutic agents can be made according to treatment protocols already known in the art. In general when more than one chemotherapeutic agent is used in the methods of this invention, the chemotherapeutic agents are administered on the same day either concurrently or consecutively in their standard dosage form. For example, the chemotherapeutic agents are usually administered intravenously, preferably by an IV drip using IV solutions well known in the art (e.g., isotonic saline (0.9% NaCI) or dextrose solution (e.g., 5% dextrose)).
When two or more chemotherapeutic agents are used, the chemotherapeutic agents are generally administered on the same day; however, those skilled in the art will appreciate that the chemotherapeutic agents can be administered on different days and in different weeks. The skilled clinician can administer the chemotherapeutic agents according to their recommended dosage schedule from the manufacturer of the agent and can adjust the schedule according to the needs of the patient, e.g., based on the patient's response to the treatment. For example, when gemcitabine is used in combination with a platinum coordinator compound, such as, for example, cisplatin, to treat lung cancer, both the gemcitabine and the cisplatin are given on the same day on day one of the treatment cycle, and then gemcitabine is given alone on day 8 and given alone again on day 15
The compounds of this invention and chemotherapeutic agents can be administered in a treatment protocol that usually lasts one to seven weeks, and is repeated typically from 6 to 12 times. Generally the treatment protocol can last one to four weeks. Treatment protocols of one to three weeks can also be used. A treatment protocol of one to two weeks can also be used. During this treatment protocol or cycle the compounds of this invention can be administered daily while the chemotherapeutic agents can be administered one or more times a week. Generally, a compound of this invention can be administered daily (i.e., once per day), and in one embodiment twice per day, and the chemotherapeutic agent is administered once a week or once every three weeks. For example, the taxanes (e.g., Paclitaxel (e.g., Taxol®) or Docetaxel (e.g., Taxotere®)) can be administered once a week or once every three weeks. However, those skilled in the art will appreciate that treatment protocols can be varied according to the needs of the patient. Thus, the combination of compounds (drugs) used in the methods of this invention can be administered in variations of the protocols described above. For example, the compounds of this invention can be administered discontinuously rather than continuously during the treatment cycle. Thus, for example, during the treatment cycle the compounds of this invention can be administered daily for a week and then discontinued for a week, with this administration repeating during the treatment cycle. Or the compounds of this invention can be administered daily for two weeks and discontinued for a week, with this administration repeating during the treatment cycle. Thus, the compounds of this invention can be administered daily for one or more weeks during the cycle and discontinued for one or more weeks during the cycle, with this pattern of administration repeating during the treatment cycle. This discontinuous treatment can also be based upon numbers of days rather than a full week. For example, daily dosing for 1 to 6 days, no dosing for 1 to 6 days, with this pattern repeating during the treatment protocol. The number of days (or weeks) wherein the compounds of this invention are not dosed do not have to equal the number of days (or weeks) wherein the compounds of this invention are dosed. Usually, if a discontinuous dosing protocol is used, the number of days or weeks that the compounds of this invention are dosed is at least equal or greater than the number of days or weeks that the compounds of this invention are not dosed.
The chemotherapeutic agent could be given by bolus or continuous infusion. The chemotherapeutic agent could be given daily to once every week, or once every two weeks, or once every three weeks, or once every four weeks during the treatment cycle. If administered daily during a treatment cycle, this daily dosing can be discontinuous over the number of weeks of the treatment cycle. For example, dosed for a week (or a number of days), no dosing for a week (or a number of days, with the pattern repeating during the treatment cycle.
The compounds of this invention can be administered orally, preferably as a solid dosage form, and in one embodiment as a capsule, and while the total therapeutically effective daily dose can be administered in one to four, or one to two divided doses per day, generally, the therapeutically effective dose is given once or twice a day, and in one embodiment twice a day. The compounds of this invention can be administered in an amount of about 50 to about 400 mg once per day, and can be administered in an amount of about 50 to about 300 mg once per day. The compounds of this invention are generally administered in an amount of about 50 to about 350 mg twice a day, usually 50 mg to about 200 mg twice a day, and in one embodiment about 75 mg to about 125 mg administered twice a day, and in another embodiment about 100 mg administered twice a day. If the patient is responding, or is stable, after completion of the therapy cycle, the therapy cycle can be repeated according to the judgment of the skilled clinician. Upon completion of the therapy cycles, the patient can be continued on the compounds of this invention at the same dose that was administered in the treatment protocol, or, if the dose was less than 200mg twice a day, the dose can be raised to 200 mg twice a day. This maintenance dose can be continued until the patient progresses or can no longer tolerate the dose (in which case the dose can be reduced and the patient can be continued on the reduced dose).
The chemotherapeutic agents, used with the compounds of this invention, are administered in their normally prescribed dosages during the treatment cycle (i.e., the chemotherapeutic agents are administered according to the standard of practice for the administration of these drugs). For example: (a) about 30 to about 300 mg/m2 for the taxanes; (b) about 30 to about 100 mg/m2 for Cisplatin; (c) AUC of about 2 to about 8 for Carboplatin; (d) about 2 to about 4 mg/m2 for EGF inhibitors that are antibodies; (e) about 50 to about 500 mg/m2 for EGF inhibitors that are small molecules; (f) about 1 to about 10 mg/m2 for VEGF kinase inhibitors that are antibodies; (g) about 50 to about 2400 mg/m2 for VEGF inhibitors that are small molecules; (h) about 1 to about 20 mg for SERMs; (i) about 500 to about 1250 mg/m2 for the anti-tumor nucleosides 5-Fluorouracil, Gemcitabine and Capecitabine; (j) for the anti-tumor nucleoside Cytarabine (Ara-C) 100-200mg/m2/day for 7 to 10 days every 3 to 4 weeks, and high doses for refractory leukemia and lymphoma, i.e., 1 to 3 gm/m2 for one hour every 12 hours for 4-8 doses every 3 to four weeks; (k) for the anti-tumor nucleoside Fludarabine (F-ara-A) 10-25mg/m2/day every 3 to 4 weeks; (I) for the anti-tumor nucleoside Decitabine 30 to 75 mg/m2 for three days every 6 weeks for a maximum of 8 cycles; (m) for the anti-tumor nucleoside Chlorodeoxyadenosine (CdA, 2-CdA) 0.05-0.1 mg/kg/day as continuous infusion for up to 7 days every 3 to 4 weeks; (n) about 1 to about 100 mg/m2 for epothilones; (o) about 1 to about 350 mg/m2 for topoisomerase inhibitors; (p) about 1 to about 50 mg/m2 for vinca alkaloids; (q) for the folate antagonist Methotrexate (MTX) 20-60 mg/m2 by oral, IV or IM every 3 to 4 weeks, the intermediate dose regimen is 80-250 mg/m2 IV over 60 minutes every 3 to 4 weeks, and the high dose regimen is 250-1000mg/m2 IV given with leucovorin every 3 to 4 weeks; (r) for the folate antagonist Premetrexed (Alimta) 300-600 mg/m2 (10 minutes IV infusion day 1) every 3 weeks; (s) for the ribonucleotide reductase inhibitor Hydroxyurea (HU) 20-50 mg/kg/day (as needed to bring blood cell counts down); (t) the platinum coordinator compound Oxaliplatin (Eloxatin) 50-100 mg/m2 every 3 to 4 weeks (preferably used for solid tumors such as non-small cell lung cancer, colorectal cancer and ovarian cancer); (u) for the anthracycline daunorubicin 10-50 mg/m2/day IV for 3-5 days every 3 to 4 weeks; (v) for the anthracycline Doxorubicin (Adriamycin) 50-100 mg/m2 IV continuous infusion over 1 -4 days every 3 to 4 weeks, or 10-40 mg/m2 IV weekly; (w) for the anthracycline ldarubicin 10-30 mg/m2 daily for 1 -3 days as a slow IV infusion over 10-20 minutes every 3 to 4 weeks; (x) for the biologic interferon (Intron-A, Roferon) 5 to 20 million IU three times per week; (y) for the biologic pegylated interferon (Peg-intron, Pegasys) 3 to 4 micrograms/kg/day chronic sub cutaneous (until relapse or loss of activity); (z) for the biologic Rituximab (Rituxan) (antibody used for non-Hodgkin's lymphoma) 200- 400mg/m2 IV weekly over 4-8 weeks for 6 months; (aa) for the alkylating agent temozolomide 75 mg/m2 to 250mg/m2, for example, 150 mg/m2, or for example, 200 mg/m2, such as 200mg/m2 for 5 days; and (bb) for the MEK1 and/or MEK2 inhibitor PD0325901 , 15 mg to 30 mg, for example, 15 mg daily for 21 days every 4 weeks. Gleevec can be used orally in an amount of about 200 to about 800 mg/day.
Thalidomide (and related imids) can be used orally in amounts of about 200 to about 800 mg/day, and can be contiuously dosed or used until releapse or toxicity. See for example Mitsiades et al., "Apoptotic signaling induced by immunomodulatory thalidomide analoqs in human multiple myeloma cells; therapeutic implications", Blood, 99(12):4525-30, June 15, 2002, the disclosure of which is incorporated herein by reference thereto.
The FPT inhibitor Sarasar® (brand of Ionifarnib) can be administered orally (e.g., capsule) in amounts of about 50 to about 200 mg given twice a day, or in amounts of about 75 to about 125 mg given twice a day, or in amounts of about 100 to about 200 mg given twice a day, or in an amount of about 100 mg given twice a day.
Paclitaxel (e.g., Taxol®), for example, can be administered once per week in an amount of about 50 to about 100 mg/m2 and in another example about 60 to about 80 mg/m2. In another example Paclitaxel (e.g., Taxol®) can be administered once every three weeks in an amount of about 150 to about 250 mg/m2 and in another example about 175 to about 225 mg/m2.
In another example, Docetaxel (e.g., Taxotere®) can be administered once per week in an amount of about 10 to about 45 mg/m2. In another example Docetaxel (e.g., Taxotere®) can be administered once every three weeks in an amount of about 50 to about 100 mg/m2.
In another example Cisplatin can be administered once per week in an amount of about 20 to about 40 mg/m2. In another example Cisplatin can be administered once every three weeks in an amount of about 60 to about 100 mg/m2. In another example Carboplatin can be administered once per week in an amount to provide an AUC of about 2 to about 3. In another example Carboplatin can be administered once every three weeks in an amount to provide an AUC of about 5 to about 8.
Other embodiments of this invention are directed to any one of the method of treating cancer embodiments wherein the compounds of formula 1.0 and the chemotherapeutic agents are administered as a pharmaceutical composition comprising an effective amount of the compounds of formula 1.0, an effective amount of the chemotherapeutic agents, and a pharmaceutically acceptable carrier.
Other embodiments of this invention are directed to any one of the method of treating cancer embodiments wherein a chemotherapeutic agent is used wherein the the chemotherapeutic agent is selected from the group consisting of: paclitaxel, docetaxel, carboplatin, cisplatin, gemcitabine, tamoxifen, Herceptin, Cetuximab, Tarceva, Iressa, bevacizumab, navelbine, IMC-1 C11 , SU5416 and SU6688. Other embodiments of this invention are directed to any one of the method of treating cancer embodiments wherein a chemotherapeutic agent is used wherein the the chemotherapeutic agent is selected from the group consisting of: paclitaxel, docetaxel, carboplatin, cisplatin, navelbine, gemcitabine, and Herceptin. Other embodiments of this invention are directed to any one of the method of treating cancer embodiments wherein a chemotherapeutic agent is used wherein the the chemotherapeutic agent is selected from the group consisting of: Cyclophasphamide, 5-Fluorouracil, Temozolomide, Vincristine, Cisplatin, Carboplatin, and Gemcitabine. Other embodiments of this invention are directed to any one of the method of treating cancer embodiments wherein a chemotherapeutic agent is used wherein the the chemotherapeutic agent is selected from the group consisting of: Gemcitabine, Cisplatin and Carboplatin.
This invention also provides a method of treating cancer in a patient in need of such treatment, said treatment comprising administering to said patient a therapeutically effective amount at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) compound of formula 1.0, and therapeutically effective amounts of at least one (e.g., 1 , 2 or 3, or 1 or 2, or 2, or 1 ) chemotherapeutic agent selected from the group consisting of: (1) taxanes, (2) platinum coordinator compounds, (3) epidermal growth factor (EGF) inhibitors that are antibodies, (4) EGF inhibitors that are small molecules, (5) vascular endolithial growth factor (VEGF) inhibitors that are antibodies, (6) VEGF kinase inhibitors that are small molecules, (7) estrogen receptor antagonists or selective estrogen receptor modulators (SERMs), (8) anti-tumor nucleoside derivatives, (9) epothilones, (10) topoisomerase inhibitors, (11 ) vinca alkaloids, (12) antibodies that are inhibitors of αVβ3 integrins, (13) folate antagonists, (14) ribonucleotide reductase inhibitors, (15) anthracyclines, (16) biologies; (17) inhibitors of angiogenesis and/or suppressors of tumor necrosis factor alpha (TNF-alpha) such as thalidomide (or related imid), (18) Bcr/abl kinase inhibitors, (19) MEK1 and/or MEK 2 inhibitors that are small molecules, (20) IGF-1 and IGF-2 inhibitors that are small molecules, (21) small molecule inhibitors of RAF and BRAF kinases, (22) small molecule inhibitors of cell cycle dependent kinases such as CDK1 , CDK2, CDK4 and CDK6, (23) alkylating agents, and (24) farnesyl protein transferase inhibitors (also know as FPT inhibitors or FTI (i.e., farnesyl transfer inhibitors)). This invention also provides a method of treating cancer in a patient in need of such treatment, said treatment comprising administering to said patient a therapeutically effective amount at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) compound of formula 1.0, and therapeutically effective amounts of at least two (e.g., 2 or 3, or 2, and usually 2) different antineoplastic agents selected from the group consisting of: (1) taxanes, (2) platinum coordinator compounds, (3) epidermal growth factor (EGF) inhibitors that are antibodies, (4) EGF inhibitors that are small molecules, (5) vascular endolithial growth factor (VEGF) inhibitors that are antibodies, (6) VEGF kinase inhibitors that are small molecules, (7) estrogen receptor antagonists or selective estrogen receptor modulators (SERMs), (8) anti-tumor nucleoside derivatives, (9) epothilones, (10) topoisomerase inhibitors, (11) vinca alkaloids, (12) antibodies that are inhibitors of αVβ3 integrins, (13) folate antagonists, (14) ribonucleotide reductase inhibitors, (15) anthracyclines, (16) biologies; (17) inhibitors of angiogenesis and/or suppressors of tumor necrosis factor alpha (TNF-alpha) such as thalidomide (or related imid), (18) Bcr/abl kinase inhibitors, (19) MEK1 and/or MEK 2 inhibitors that are small molecules, (20) IGF-1 and IGF-2 inhibitors that are small molecules, (21 ) small molecule inhibitors of RAF and BRAF kinases, (22) small molecule inhibitors of cell cycle dependent kinases such as CDK1 , CDK2, CDK4 and CDK6, (23) alkylating agents, and (24) farnesyl protein transferase inhibitors (also know as FPT inhibitors or FTI (i.e., farnesyl transfer inhibitors)).
This invention also provides a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient therapeutically effective amounts at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) compound of formula 1.0, and an antineoplastic agent selected from the group consisting of: (1) EGF inhibitors that are antibodies, (2) EGF inhibitors that are small molecules, (3) VEGF inhibitors that are antibodies, and (4) VEGF inhibitors that are small molecules. Radiation therapy can also be used in conjunction with this above combination therapy, i.e., the above method using a combination of compounds of the invention and antineoplastic agent can also comprise the administration of a therapeutically effect amount of radiation.
This invention also provides a method of treating leukemias (e.g., acute myeloid leukemia (AML), and chronic myeloid leukemia (CML)) in a patient in need of such treatment, said method comprising administering to said patient therapeutically effective amounts at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) compound of formula 1.0, and: (1 ) Gleevec and interferon to treat CML; (2) Gleevec and pegylated interferon to treat CML; (3) Gleevec to treat CML; (4) an anti-tumor nucleoside derivative (e.g., Ara-C) to treat AML; or (5) an anti-tumor nucleoside derivative (e.g., Ara-C) in combination with an anthracycline to treat AML. This invention also provides a method of treating non-Hodgkin's lymphoma in a patient in need of such treatment, said method comprising administering therapeutically effective amounts at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) compound of formula 1.0 and: (1 ) a biologic (e.g., Rituxan); (2) a biologic (e.g., Rituxan) and an anti-tumor nucleoside derivative (e.g., Fludarabine); or (3) Genasense (antisense to BCL-2).
This invention also provides a method of treating multiple myeloma in a patient in need of such treatment, said method comprising administering to said patient therapeutically effective amounts of at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) compound of formula 1.0 and: (1 ) a proteosome inhibitor (e.g., PS-341 from Millenium); or (2) Thalidomide (or related imid).
This invention also provides a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient therapeutically effective amounts of: (a) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) compound of formula 1.0, and (b) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 2, or 1) antineoplastic agent selected from the group consisting of: (1 ) taxanes, (2) platinum coordinator compounds, (3) EGF inhibitors that are antibodies, (4) EGF inhibitors that are small molecules, (5) VEGF inhibitors that are antibodies, (6) VEGF kinase inhibitors that are small molecules, (7) estrogen receptor antagonists or selective estrogen receptor modulators, (8) anti-tumor nucleoside derivatives, (9) epothilones, (10) topoisomerase inhibitors, (11) vinca alkaloids, and (12) antibodies that are inhibitors of αVβ3 integrins.
This invention also provides a method of treating non small cell lung cancer in a patient in need of such treatment, said method comprising administering to said patient therapeutically effective amounts of: (a) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) compound of formula 1.0, and (b) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 2, or 1) antineoplastic agent selected from the group consisting of: (1) taxanes, (2) platinum coordinator compounds, (3) EGF inhibitors that are antibodies, (4) EGF inhibitors that are small molecules, (5) VEGF inhibitors that are antibodies, (6) VEGF kinase inhibitors that are small molecules, (7) estrogen receptor antagonists or selective estrogen receptor modulators, (8) anti-tumor nucleoside derivatives, (9) epothilones, (10) topoisomerase inhibitors, (11) vinca alkaloids, and (12) antibodies that are inhibitors of αVβ3 integrins.
This invention also provides a method of treating non small cell lung cancer in a patient in need of such treatment, said method comprising administering to said patient therapeutically effective amounts of: (a) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) compound of formula 1.0, and (b) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 2, or 1 ) antineoplastic agent selected from the group consisting of: (1) taxanes, (2) platinum coordinator compounds, (3) anti-tumor nucleoside derivatives, (4) topoisomerase inhibitors, and (5) vinca alkaloids.
This invention also provides a method of treating non small cell lung cancer in a patient in need of such treatment, said method comprising administering therapeutically effective amounts of: (a) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) compound of formula 1.0, (b) carboplatin, and (c) paclitaxel. This invention also provides a method of treating non small cell lung cancer in a patient in need of such treatment, said method comprising administering to said patient therapeutically effective amounts of: (a) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) compound of formula 1.0, (b) cisplatin, and (c) gemcitabine.
This invention also provides a method of treating non small cell lung cancer in a patient in need of such treatment, said method comprising administering therapeutically effective amounts of: (a) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) compound of formula 1.0, (b) carboplatin, and (c) gemcitabine.
This invention also provides a method of treating non small cell lung cancer in a patient in need of such treatment, said method comprising administering therapeutically effective amounts of: (a) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) compound of formula 1.0, (b) Carboplatin, and (c) Docetaxel.
This invention also provides a method of treating cancer in a patient in need of such treatment, said method comprising administering therapeutically effective amounts of: (a) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) compound of formula 1.0, and (b) an antineoplastic agent selected from the group consisting of: (1 ) EGF inhibitors that are antibodies, (2) EGF inhibitors that are small molecules, (3) VEGF inhibitors that are antibodies, (4) VEGF kinase inhibitors that are small molecules. This invention also provides a method of treating squamous cell cancer of the head and neck, in a patient in need of such treatment, said method comprising administering to said patient therapeutically effective amounts of: (a) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) compound of formula 1.0, and (b) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 2, or 1) antineoplastic agent selected from the group consisting of: (1 ) taxanes, and (2) platinum coordinator compounds.
This invention also provides a method of treating squamous cell cancer of the head and neck, in a patient in need of such treatment, said method comprising administering to said patient therapeutically effective amounts of: (a) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) compound of formula 1.0, and (b) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 2, or 1 ) antineoplastic agent selected from the group consisting of: (1) taxanes, (2) platinum coordinator compounds, and (3) antitumor nucleoside derivatives (e.g., 5-Fluorouracil).
This invention also provides a method of treating CML in a patient in need of such treatment, said method comprising administering therapeutically effective amounts of: (a) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) compound of formula 1.0, (b) Gleevec, and (c) interferon (e.g., Intron-A).
This invention also provides a method of treating CML in a patient in need of such treatment comprising administering therapeutically effective amounts of: (a) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) compound of formula 1.0, (b) Gleevec; and (c) pegylated interferon (e.g., Peg-lntron, and Pegasys).
This invention also provides a method of treating CML in a patient in need of such treatment comprising administering therapeutically effective amounts of: (a) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) compound of formula 1.0 and (b) Gleevec.
This invention also provides a method of treating CMML in a patient in need of such treatment, said method comprising administering to said patient therapeutically effective amounts of at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) compound of formula 1.0. This invention also provides a method of treating AML in a patient in need of such treatment, said method comprising administering to said patient therapeutically effective amounts of: (a) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) compound of formula 1.0, and (b) an anti-tumor nucleoside derivative (e.g., Cytarabine (i.e., Ara-C)). This invention also provides a method of treating AML in a patient in need of such treatment, said method comprising administering to said patient therapeutically effective amounts of: (a) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) compound of formula 1.0, (b) an anti-tumor nucleoside derivative (e.g., Cytarabine (i.e., Ara-C)), and (c) an anthracycline.
This invention also provides a method of treating non-Hodgkin's lymphoma in a patient in need of such treatment, said method comprising administering to said patient therapeutically effective amounts of: (a) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) compound of formula 1.0, and (b) Rituximab (Rituxan). This invention also provides a method of treating non-Hodgkin's lymphoma in a patient in need of such treatment, said method comprising administering to said patient therapeutically effective amounts of: (a) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) compound of formula 1.0, (b) Rituximab (Rituxan), and (c) an antitumor nucleoside derivative (e.g., Fludarabine (i.e., F-ara-A). This invention also provides a method of treating non-Hodgkin's lymphoma in a patient in need of such treatment, said method comprising administering to said patient therapeutically effective amounts of: (a) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) compound of formula 1.0, and (b) Genasense (antisense to BCL-
2). This invention also provides a method of treating multiple myeloma in a patient in need of such treatment, said method comprising administering therapeutically effective amounts of: (a) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) compound of formula 1.0, and (b) a proteosome inhibitor (e.g., PS-341 (Millenium)). This invention also provides a method of treating multiple myeloma in a patient in need of such treatment, said method comprising administering to said patient therapeutically effective amounts of: (a) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) compound of formula 1.0, and (b) Thalidomide or related imid.
This invention also provides a method of treating multiple myeloma in a patient in need of such treatment, said method comprising administering therapeutically effective amounts of: (a) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1) compound of formula 1.0, and (b) Thalidomide.
This invention is also directed to the methods of treating cancer described herein, particularly those described above, wherein in addition to the administration of the compound of formula 1.0 and antineoplastic agents, radiation therapy is also administered prior to, during, or after the treatment cycle.
This invention also provides a method for treating cancer (e.g., lung cancer, prostate cancer and myeloid leukemias) in a patient in need of such treatment, said method comprising administering to said patient (1 ) an effective amount of at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) compound of formula 1.0, in combination with (2) at least one (e.g., 1 , 2 or 3, or 1 or 2, or 2, or 1) antineoplastic agent, microtubule affecting agent and/or radiation therapy.
This invention also provides a method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one (e.g., 1 , 2 or 3, or 1 or 2, or 1 , and usually 1 ) compound of formula 1.0 in combination with an effective amount of at least one (e.g., 1 , 2 or 3, or
1 or 2, or 1 , and usually 1 ) signal transduction inhibitor.
Thus, in one example (e.g., treating non small cell lung cancer): (1 ) the compound of formula 1.0 is administered in an amount of about 50 mg to about 200 mg twice a day, and in another example about 75 mg to about 125 mg administered twice a day, and in yet another example about 100 mg administered twice a day, (2) Paclitaxel (e.g., Taxol® is administered once per week in an amount of about 50 to about 100 mg/m2, and in another example about 60 to about 80 mg/m2, and (3) Carboplatin is administered once per week in an amount to provide an AUC of about
2 to about 3.
In another example (e.g., treating non small cell lung cancer): (1 ) the compound of formula 1.0 is administered in an amount of about 50 mg to about 200 mg twice a day, and in another example about 75 mg to about 125 mg administered twice a day, and yet in another example about 100 mg administered twice a day, (2) Paclitaxel (e.g., Taxol® is administered once per week in an amount of about 50 to about 100 mg/m2, and in another example about 60 to about 80 mg/m2, and (3) Cisplatin is administered once per week in an amount of about 20 to about 40 mg/m2. In another example (e.g., treating non small cell lung cancer): (1 ) the compound of formula 1.0 is administered in an amount of about 50 mg to about 200 mg twice a day, and in another example about 75 mg to about 125 mg administered twice a day, and in yet another example about 100 mg administered twice a day, (2) Docetaxel (e.g., Taxotere®) is administered once per week in an amount of about 10 to about 45 mg/m2, and (3) Carboplatin is administered once per week in an amount to provide an AUC of about 2 to about 3.
In another example (e.g., treating non small cell lung cancer): (1) the compound of formula 1.0 is administered in an amount of about 50 mg to about 200 mg twice a day, and in another example about 75 mg to about 125 mg administered twice a day, and in yet another example about 100 mg administered twice a day, (2) Docetaxel (e.g., Taxotere®) is administered once per week in an amount of about 10 to about 45 mg/m2, and (3) Cisplatin is administered once per week in an amount of about 20 to about 40 mg/m2. In another example (e.g., treating non small cell lung cancer): (1) the compound of formula 1.0 is administered in an amount of about 50 mg to about 200 mg twice a day, and in another example about 75 mg to about 125 mg administered twice a day, and in yet another example about 100 mg administered twice a day, (2) Paclitaxel (e.g., Taxol® is administered once every three weeks in an amount of about 150 to about 250 mg/m2, and in another example about 175 to about 225 mg/m2, and in yet another example 175 mg/m2, and (3) Carboplatin is administered once every three weeks in an amount to provide an AUC of about 5 to about 8, and in another example 6.
In another example of treating non small cell lung cancer: (1) the compound of formula 1.0 is administered in an amount of 100 mg administered twice a day, (2) Paclitaxel (e.g., Taxol® is administered once every three weeks in an amount of 175 mg/m2, and (3) Carboplatin is administered once every three weeks in an amount to provide an AUC of 6.
In another example (e.g., treating non small cell lung cancer): (1) the compound of formula 1.0 is administered in an amount of about 50 mg to about 200 mg twice a day, and in another example about 75 mg to about 125 mg administered twice a day, and in yet another example about 100 mg administered twice a day, (2) Paclitaxel (e.g., Taxol® is administered once every three weeks in an amount of about 150 to about 250 mg/m2, and in another example about 175 to about 225 mg/m2, and (3) Cisplatin is administered once every three weeks in an amount of about 60 to about 100 mg/m2.
In another example (e.g., treating non small cell lung cancer): (1) the compound of formula 1.0 is administered in an amount of about 50 mg to about 200 mg twice a day, and in another example about 75 mg to about 125 mg administered twice a day, and in yet another example about 100 mg administered twice a day, (2) Docetaxel (e.g., Taxotere® is administered once every three weeks in an amount of about 50 to about 100 mg/m2, and (3) Carboplatin is administered once every three weeks in an amount to provide an AUC of about 5 to about 8.
In another example (e.g., treating non small cell lung cancer): (1) the compound of formula 1.0 is administered in an amount of about 50 mg to about 200 mg twice a day, in another example about 75 mg to about 125 mg administered twice a day, and in yet another example about 100 mg administered twice a day, (2) Docetaxel (e.g., Taxotere® is administered once every three weeks in an amount of about 50 to about 100 mg/m2, and (3) Cisplatin is administered once every three weeks in an amount of about 60 to about 100 mg/m2.
In another example for treating non small cell lung cancer using the compounds of formula 1.0, Docetaxel and Carboplatin: (1 ) the compound of formula 1.0 is administered in an amount of about 50 mg to about 200 mg twice a day, and in another example about 75 mg to about 125 mg administered twice a day, and in yet another example about 100 mg administered twice a day, (2) Docetaxel (e.g., Taxotere® is administered once every three weeks in an amount of about 75 mg/m2, and (3) Carboplatin is administered once every three weeks in an amount to provide an AUC of about 6.
In another example of the treatments of non-small cell lung cancer described above the Docetaxel (e.g., Taxotere®) and Cisplatin, the Docetaxel (e.g., Taxotere®) and Carboplatin, the Paclitaxel (e.g., Taxol®) and Carboplatin, or the Paclitaxel (e.g., Taxol®) and Cisplatin are administered on the same day. In another example (e.g., CML): (1 ) the compound of formula 1.0 is administered in an amount of about 100 mg to about 200 mg administered twice a day, (2) Gleevec is administered in an amount of about 400 to about 800 mg/day orally, and (3) interferon (Intron-A) is administered in an amount of about 5 to about 20 million IU three times per week. In another example (e.g., CML): (1 ) the compound of formula 1.0 is administered in an amount of about 100 mg to about 200 mg administered twice a day, (2) Gleevec is administered in an amount of about 400 to about 800 mg/day orally, and (3) pegylated interferon (Peg-lntron or Pegasys) is administered in an amount of about 3 to about 6 micrograms/kg/day.
In another example (e.g., non-Hodgkin's lymphoma): (1 ) the compound of formula 1.0 is administered in an amount of about 50 mg to about 200 mg twice a day, and in another example about 75 mg to about 125 mg administered twice a day, and in yet another example about 100 mg administered twice a day, and (2) Genasense (antisense to BCL-2) is administered as a continuous IV infusion at a dose of about 2 to about 5 rηg/kg/day (e.g., 3 mg/kg/day) for 5 to 7 days every 3 to 4 weeks. In another example (e.g., multiple myeloma): (1) the compound of formula 1.0 is administered in an amount of about 50 mg to about 200 mg twice a day, and in another example about 75 mg to about 125 mg administered twice a day, and in yet another example about 100 mg administered twice a day, and (2) the proteosome inhibitor (e.g., PS-341 - Millenium) is administered in an amount of about 1.5mg/m2 twice weekly for two consecutive weeks with a one week rest period.
In another example (e.g., multiple myeloma): (1 ) the compound of formula 1.0 is administered in an amount of about 50 mg to about 200 mg twice a day, and in another example about 75 mg to about 125 mg administered twice a day, and in yet another example about 100 mg administered twice a day, and (2) the Thalidomide (or related imid) is administered orally in an amount of about 200 to about 800 mg/day, with dosing being continuous until relapse or toxicity.
In one embodiment of the methods of treating cancer of this invention, the chemotherapeutic agents are selected from the group consisting of: paclitaxel, docetaxel, carboplatin, cisplatin, gemcitabine, tamoxifen, Herceptin, Cetuximab, Tarceva, Iressa, bevacizumab, navelbine, IMC-1 C11 , SU5416 and SU6688.
In another embodiment of the methods of treating cancer of this invention, the chemotherapeutic agents are selected from the group consisting of: paclitaxel, docetaxel, carboplatin, cisplatin, navelbine, gemcitabine, and Herceptin.
Thus, one embodiment of this invention is directed to a method of treating cancer comprising administering to a patient in need of such treatment therapeutically effective amounts of the compound of formula 1.0, a taxane, and a platinum coordination compound.
Another embodiment of this invention is directed to a method of treating cancer comprising administering to a patient in need of such treatment therapeutically effective amounts of the compound of formula 1.0, a taxane, and a platinum coordination compound, wherein said compound of formula 1.0 is administered every day, said taxane is administered once per week per cycle, and said platinum coordinator compound is administered once per week per cycle. In another embodiment the treatment is for one to four weeks per cycle.
Another embodiment of this invention is directed to a method of treating cancer comprising administering to a patient in need of such treatment therapeutically effective amounts of the compound of formula 1.0, a taxane, and a platinum coordination compound, wherein said compound of formula 1.0 is administered every day, said taxane is administered once every three weeks per cycle, and said platinum coordinator compound is administered once every three weeks per cycle. In another embodiment the treatment is for one to three weeks per cycle.
Another embodiment of this invention is directed to a method of treating cancer comprising administering to a patient in need of such treatment therapeutically effective amounts of the compound of formula 1.0, paclitaxel, and carboplatin. In another embodiment, said compound of formula 1.0 is administered every day, said paclitaxel is administered once per week per cycle, and said carboplatin is administered once per week per cycle. In another embodiment the treatment is for one to four weeks per cycle. Another embodiment of this invention is directed to a method of treating cancer comprising administering to a patient in need of such treatment therapeutically effective amounts of the compound of formula 1.0, paclitaxel, and carboplatin. In another embodiment, said compound of formula 1.0 is administered every day, said paclitaxel is administered once every three weeks per cycle, and said carboplatin is administered once every three weeks per cycle. In another embodiment the treatment is for one to three weeks per cycle.
Another embodiment of this invention is directed to a method for treating non small cell lung cancer in a patient in need of such treatment comprising administering daily a therapeutically effective amount of the compound of formula 1.0, administering a therapeutically effective amount of carboplatin once a week per cycle, and administering a therapeutically effective amount of paclitaxel once a week per cycle, wherein the treatment is given for one to four weeks per cycle. In another embodiment said compound of formula 1.0 is administered twice per day. In another embodiment said carboplatin and said paclitaxel are administered on the same day, and in another embodiment said carboplatin and said paclitaxel are administered consecutively, and in another embodiment said carboplatin is administered after said paclitaxel.
Another embodiment of this invention is directed to a method for treating non small cell lung cancer in a patient in need of such treatment comprising administering daily a therapeutically effective amount of a compound of formula 1.0, administering a therapeutically effective amount of carboplatin once every three weeks per cycle, and administering a therapeutically effective amount of paclitaxel once every three weeks per cycle, wherein the treatment is given for one to three weeks. In another embodiment compound of formula 1.0 is administered twice per day. In another embodiment said carboplatin and said paclitaxel are administered on the same day, and in another embodiment said carboplatin and said paclitaxel are administered consecutively, and in another embodiment said carboplatin is administered after said paclitaxel. Another embodiment of this invention is directed to a method for treating non small cell lung cancer in a patient in need of such treatment comprising administering about 50 to about 200 mg of a compound of formula 1.0 twice a day, administering carboplatin once per week per cycle in an amount to provide an AUC of about 2 to about 8 (and in another embodiment about 2 to about 3), and administering once per week per cycle about 60 to about 300 mg/m2 (and in another embodiment about 50 to 100mg/m2, and in yet another embodiment about 60 to about 80 mg/m2) of paclitaxel, wherein the treatment is given for one to four weeks per cycle. In another embodiment said compound of formula 1.0 is administered in amount of about 75 to about 125 mg twice a day, and in another embodiment about 100 mg twice a day. In another embodiment said carboplatin and said paclitaxel are administered on the same day, and in another embodiment said carboplatin and said paclitaxel are administered consecutively, and in another embodiment said carboplatin is administered after said paclitaxel.
In another embodiment, this invention is directed to a method for treating non small cell lung cancer in a patient in need of such treatment comprising administering about 50 to about 200 mg of a compound of formula 1.0 twice a day, administering carboplatin once every three weeks per cycle in an amount to provide an AUC of about 2 to about 8 (in another embodiment about 5 to about 8, and in another embodiment 6), and administering once every three weeks per cycle about 150 to about 250 mg/m2 (and in another embodiment about 175 to about 225 mg/m2, and in another embodiment 175 mg/m2) of paclitaxel, wherein the treatment is given for one to three weeks. In another embodiment said compound of formula 1.0 is administered in an amount of about 75 to about 125 mg twice a day, and in another embodiment about 100 mg twice a day. In another embodiment said carboplatin and said paclitaxel are administered on the same day, and in another embodiment said carboplatin and said paclitaxel are administered consecutively, and in another embodiment said carboplatin is administered after said paclitaxel.
Other embodiments of this invention are directed to methods of treating cancer as described in the above embodiments (i.e., the embodiments directed to treating cancer and to treating non small cell lung cancer with a taxane and platinum coordinator compound) except that in place of paclitaxel and carboplatin the taxanes and platinum coordinator compounds used together in the methods are: (1 ) docetaxel (Taxotere®) and cisplatin; (2) paclitaxel and cisplatin; and (3) docetaxel and carboplatin. In another embodiment of the methods of this invention cisplatin is used in amounts of about 30 to about 100 mg/m2. In another embodiment of the methods of this invention docetaxel is used in amounts of about 30 to about 100 mg/m2.
In another embodiment this invention is directed to a method of treating cancer comprising administering to a patient in need of such treatment therapeutically effective amounts of a compound of formula 1.0, a taxane, and an EGF inhibitor that is an antibody. In another embodiment the taxane used is paclitaxel, and the EGF inhibitor is a HER2 antibody (in one embodiment Herceptin) or Cetuximab, and in another embodiment Herceptin is used. The length of treatment, and the amounts and administration of said compound of formula 1.0 and the taxane are as described in the embodiments above. The EGF inhibitor that is an antibody is administered once a week per cycle, and in another embodiment is administered on the same day as the taxane, and in another embodiment is administered consecutively with the taxane. For example, Herceptin is administered in a loading dose of about 3 to about 5 mg/m2 (in another embodiment about 4 mg/m2), and then is administered in a maintenance dose of about 2 mg/m2 once per week per cycle for the remainder of the treatment cycle (usually the cycle is 1 to 4 weeks). In one embodiment the cancer treated is breast cancer.
In another embodiment this invention is directed to a method of treating cancer comprising administering to a patient in need of such treatment therapeutically effective amounts of: (1) a compound of formula 1.0, (2) a taxane, and (3) an antineoplastic agent selected from the group consisting of: (a) an EGF inhibitor that is a small molecule, (b) a VEGF inhibitor that is an antibody, and (c) a VEGF kinase inhibitor that is a small molecule. In another embodiment, the taxane paclitaxel or docetaxel is used. In another embodiment the antineoplastic agent is selected from the group consisting of: tarceva, Iressa, bevacizumab, SU5416, SU6688 and BAY 43- 9006. The length of treatment, and the amounts and administration of said compound of formula 1.0 and the taxane are as described in the embodiments above. The VEGF kinase inhibitor that is an antibody is usually given once per week per cycle. The EGF and VEGF inhibitors that are small molecules are usually given daily per cycle. In another embodiment, the VEGF inhibitor that is an antibody is given on the same day as the taxane, and in another embodiment is administered concurrently with the taxane. In another embodiment, when the EGF inhibitor that is a small molecule or the VEGF inhibitor that is a small molecule is administered on the same day as the taxane, the administration is concurrently with the taxane. The EGF or VEGF kinase inhibitor is generally administered in an amount of about 10 to about 500 mg/m2.
In another embodiment this invention is directed to a method of treating cancer comprising administering to a patient in need of such treatment therapeutically effective amounts of a compound of formula 1.0, an anti-tumor nucleoside derivative, and a platinum coordination compound.
Another embodiment of this invention is directed to a method of treating cancer comprising administering to a patient in need of such treatment therapeutically effective amounts of a compound of formula 1.0, an anti-tumor nucleoside derivative, and a platinum coordination compound, wherein said compound of formula 1.0 is administered every day, said anti-tumor nucleoside derivative is administered once per week per cycle, and said platinum coordinator compound is administered once per week per cycle. Although the treatment can be for one to four weeks per cycle, in one embodiment the treatment is for one to seven weeks per cycle. Another embodiment of this invention is directed to a method of treating cancer comprising administering to a patient in need of such treatment therapeutically effective amounts of a compound of formula 1.0, an anti-tumor nucleoside derivative, and a platinum coordination compound, wherein said compound of formula 1.0 is administered every day, said an anti-tumor nucleoside derivative is administered once per week per cycle, and said platinum coordinator compound is administered once every three weeks per cycle. Although the treatment can be for one to four weeks per cycle, in one embodiment the treatment is for one to seven weeks per cycle.
Another embodiment of this invention is directed to a method of treating cancer comprising administering to a patient in need of such treatment therapeutically effective amounts of a compound of formula 1.0, gemcitabine, and cisplatin. In another embodiment, said compound of formula 1.0 is administered every day, said gemcitabine is administered once per week per cycle, and said cisplatin is administered once per week per cycle. In one embodiment the treatment is for one to seven weeks per cycle.
Another embodiment of this invention is directed to a method of treating cancer comprising administering to a patient in need of such treatment therapeutically effective amounts of a compound of formula 1.0, gemcitabine, and cisplatin. In another embodiment, said compound of formula 1.0 is administered every day, said gemcitabine is administered once per week per cycle, and said cisplatin is administered once every three weeks per cycle. In another embodiment the treatment is for one to seven weeks.
Another embodiment of this invention is directed to a method of treating cancer comprising administering to a patient in need of such treatment therapeutically effective amounts of a compound of formula 1.0, gemcitabine, and carboplatin. In another embodiment said compound of formula 1.0 is administered every day, said gemcitabine is administered once per week per cycle, and said carboplatin is administered once per week per cycle. In another embodiment the treatment is for one to seven weeks per cycle. Another embodiment of this invention is directed to a method of treating cancer comprising administering to a patient in need of such treatment therapeutically effective amounts of a compound of formula 1.0, gemcitabine, and carboplatin. In another embodiment said compound of formula 1.0 is administered every day, said gemcitabine is administered once per week per cycle, and said carboplatin is administered once every three weeks per cycle. In another embodiment the treatment is for one to seven weeks per cycle.
In the above embodiments using gemcitabine, the compound of formula 1.0 and the platinum coordinator compound are administered as described above for the embodiments using taxanes. Gemcitabine is administered in an amount of about 500 to about 1250 mg/m2. In one embodiment the gemcitabine is administered on the same day as the platinum coordinator compound, and in another embodiment consecutively with the platinum coordinator compound, and in another embodiment the gemcitabine is administered after the platinum coordinator compound. Another embodiment of this invention is directed to a method of treating cancer in a patient in need of such treatment comprising administering to said patient a compound of formula 1.0 and an antineoplastic agent selected from: (1 ) EGF inhibitors that are antibodies, (2) EGF inhibitors that are small molecules, (3) VEGF inhibitors that are antibodies, and (4) VEGF kinase inhibitors that are small molecules all as described above. The treatment is for one to seven weeks per cycle, and generally for one to four weeks per cycle. The compound of formula 1.0 is administered in the same mariner as described above for the other embodiments of this invention. The small molecule antineoplastic agents are usually administered daily, and the antibody antineoplastic agents are usually administered once per week per cycle. In one embodiment the antineoplastic agents are selected from the group consisting of: Herceptin, Cetuximab, Tarceva, Iressa, bevacizumab, IMC-1C11 , SU5416, SU6688 and BAY 43-9006.
In the embodiments of this invention wherein a platinum coordinator compound is used as well as at least one other antineoplastic agent, and these drugs are administered consecutively, the platinum coordinator compound is generally administered after the other antineoplastic agents have been administered. Other embodiments of this invention include the administration of a therapeutically effective amount of radiation to the patient in addition to the administration of a compound of formula 1.0 and antineoplastic agents in the embodiments described above. Radiation is administered according to techniques and protocols well know to those skilled in the art.
Another embodiment of this invention is directed to a pharmaceutical composition comprising at least two different chemotherapeutic agents and a pharmaceutically acceptable carrier for intravenous administration. Preferably the pharmaceutically acceptable carrier is an isotonic saline solution (0.9% NaCI) or a dextrose solution (e.g., 5% dextrose).
Another embodiment of this invention is directed to a pharmaceutical composition comprising a compound of formula 1.0 and at least two different antineoplastic agents and a pharmaceutically acceptable carrier for intravenous administration. Preferably the pharmaceutically acceptable carrier is an isotonic saline solution (0.9% NaCI) or a dextrose solution (e.g., 5% dextrose).
Another embodiment of this invention is directed to a pharmaceutical composition comprising a compound of formula 1.0 and at least one antineoplastic agent and a pharmaceutically acceptable carrier for intravenous administration.
Preferably the pharmaceutically acceptable carrier is an isotonic saline solution (0.9%
NaCI) or a dextrose solution (e.g., 5% dextrose).
Other embodiments of this invention are directed to the use of a combination of at least one (e.g., one) compound of formula 1.0 and drugs for the treatment of breast cancer, i.e., this invention is directed to a combination therapy for the treatment of breast cancer. Those skilled in the art will appreciate that the compounds of formula
1.0 and drugs are generally administered as individual pharmaceutical compositions.
The use of a pharmaceutical composition comprising more than one drug is within the scope of this invention. Thus, another embodiment of this invention is directed to a method of treating
(or preventing) breast cancer (i.e., postmenopausal and premenopausal breast cancer, e.g., hormone-dependent breast cancer) in a patient in need of such treatment comprising administering to said patient a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0 and a therapeutically effective amount of at least one antihormonal agent selected from the group consisting of: (a) aromatase inhibitors, (b) antiestrogens, and (c) LHRH analogues; and said treatment optionally including the administration of at least one chemotherapeutic agent. The compound of formula 1.0 is preferably administered orally, and in one embodiment is administered in capsule form. Examples of aromatase inhibitors include but are not limited to: Anastrozole
(e.g., Arimidex), Letrozole (e.g., Femara), Exemestane (Aromasin), Fadrozole and
Formestane (e.g., Lentaron).
Examples of antiestrogens include but are not limited to: Tamoxifen (e.g.,
Nolvadex), Fulvestrant (e.g., Faslodex), Raloxifene (e.g., Evista), and Acolbifene. Examples of LHRH analogues include but are not limited to: Goserelin (e.g.,
Zoladex) and Leuprolide (e.g., Leuprolide Acetate, such as Lupron or Lupron Depot). Examples of chemotherapeutic agents include but are not limited to:
Trastuzumab (e.g., Herceptin), Gefitinib (e.g., Iressa), Erlotinib (e.g., Erlotinib HCI, such as Tarceva), Bevacizumab (e.g., Avastin), Cetuximab (e.g., Erbitux), and Bortezomib (e.g., Velcade).
Preferably, when more than one antihormonal agent is used, each agent is selected from a different category of agent. For example, one agent is an aromatase inhibitor (e.g., Anastrozole, Letrozole, or Exemestane) and one agent is an antiestrogen (e.g., Tamoxifen or Fulvestrant).
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0 and at least one antihormonal agent selected from the group consisting of: (a) aromatase inhibitors, (b) antiestrogens, and (c) LHRH analogues; and administering an effective amount of at least one chemotherapeutic agent.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0 and at least one antihormonal agent selected from the group consisting of: (a) aromatase inhibitors, (b) antiestrogens, and (c) LHRH analogues. Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0 and at least one antihormonal agent selected from the group consisting of: (a) aromatase inhibitors, and (b) antiestrogens. Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, at least one antihormonal agent selected from the group consisting of: (a) aromatase inhibitors and (b) antiestrogens; and at least one chemotherapeutic agent.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0 and at least one aromatase inhibitor. Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, at least one aromatase inhibitor, and at least one chemotherapeutic agent.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of: (1) at least one (e.g., one) compound of formula 1.0; and (2) at least one antihormonal agent selected from the group consisting of: (a) aromatase inhibitors that are selected from the group consisting of Anastrozole, Letrozole, Exemestane, Fadrozole and Formestane, (b) antiestrogens that are selected from the group consisting of: Tamoxifen, Fulvestrant, Raloxifene, and Acolbifene, and (c) LHRH analogues that are selected from the group consisting of: Goserelin and Leuprolide; and administering an effective amount of at least one chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of: (1 ) at least one (e.g., one) compound of formula 1.0; and (2) at least one antihormonal agent selected from the group consisting of: (a) aromatase inhibitors that are selected from the group consisting of Anastrozole, Letrozole, Exemestane, Fadrozole and Formestane, (b) antiestrogens that are selected from the group consisting of: Tamoxifen, Fulvestrant, Raloxifene, and Acolbifene, and (c) LHRH analogues that are selected from the group consisting of: Goserelin and Leuprolide.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of: (1) at least one (e.g., one) compound of formula 1.0; and (2) at least one antihormonal agent selected from the group consisting of: (a) aromatase inhibitors that are selected from the group consisting of Anastrozole, Letrozole, Exemestane, Fadrozole and Formestane, and (b) antiestrogens that are selected from the group consisting of: Tamoxifen, Fulvestrant, Raloxifene, and Acolbifene. Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of: (1) at least one (e.g., one) compound of formula 1.0; and (2) at least one antihormonal agent selected from the group consisting of: (a) aromatase inhibitors that are selected from the group consisting of Anastrozole, Letrozole, Exemestane, Fadrozole and Formestane, (b) antiestrogens that are selected from the group consisting of: Tamoxifen, Fulvestrant, Raloxifene, and Acolbifene; and administering an effective amount of at least one chemotherapeutic agents are selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of: (1) at least one (e.g., one) compound of formula 1.0; and (2) at least one aromatase inhibitor selected from the group consisting of Anastrozole, Letrozole, Exemestane, Fadrozole and Formestane.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of: (1) at least one (e.g., one) compound of formula 1.0; (2) at least one aromatase inhibitor that is selected from the group consisting of Anastrozole, Letrozole, Exemestane, Fadrozole and Formestane; and (3) administering an effective amount of at least one chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of: (1 ) at least one (e.g., one) compound of formula 1.0; (2) at least one aromatase inhibitor; and (3) at least one LHRH analogue.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of:(1) at least one (e.g., one) compound of formula 1.0; (2) at least one antiestrogen ; and (3) at least one LHRH analogue.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of: (1 ) at least one (e.g., one) compound of formula 1.0; (2) at least one aromatase inhibitor that is selected from the group consisting of Anastrozole, Letrozole, Exemestane, Fadrozole and Formestane; and (3) at least one LHRH analogue that is selected from the group consisting of: Goserelin and Leuprolide. Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of: (1 ) at least one (e.g., one) compound of formula 1.0; (2) at least one antiestrogen that is selected from the group consisting of: Tamoxifen, Fulvestrant, Raloxifene, and Acolbifene; and (3) at least one LHRH analogue that is selected from the group consisting of: Goserelin and Leuprolide.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0 and Anastrozole.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0 and Letrazole. Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0 and Exemestane.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0 and and Fadrozole.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0 and Formestane.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0 and Tamoxifen.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0 Fulvestrant.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0 and Raloxifene. Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0 and Acolbifene.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0 and Goserelin.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0 and and Leuprolide.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Anastrozole, and an antiestrogen selected from the group consisting of: Tamoxifen, Fulvestrant, Raloxifene, and Acolbifene.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Letrozole, and an antiestrogen selected from the group consisting of: Tamoxifen, Fulvestrant, Raloxifene, and Acolbifene.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Exemestane, and an antiestrogen selected from the group consisting of: Tamoxifen, Fulvestrant, Raloxifene, and Acolbifene.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Fadrozole, and an antiestrogen selected from the group consisting of: Tamoxifen, Fulvestrant, Raloxifene, and Acolbifene.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Formestane, and an antiestrogen selected from the group consisting of: Tamoxifen, Fulvestrant, Raloxifene, and Acolbifene.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Anastrozole, and Tamoxifen.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Letrozole, and Tamoxifen.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Exemestane, and Tamoxifen. Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Fadrozole, and Tamoxifen. Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Formestane, and Tamoxifen. Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Anastrozole, and Fulvestrant.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Letrozole, and Fulvestrant.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Exemestane, and Fulvestrant.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Fadrozole, and Fulvestrant.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Formestane, and Fulvestrant. Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Anastrozole, and a chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Letrozole, and a chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Exemestane, and a chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Fadrozole, and a chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib. Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Formestane, and a chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Tamoxifen, and a chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Fulvestrant, and a chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Raloxifene, and a chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib. Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Acolbifene, and a chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Goserelin, and a chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Leuprolein, and a chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Anastrozole, an antiestrogen selected from the group consisting of: Tamoxifen, Fulvestrant, Raloxifene, and Acolbifene, and a chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib. Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Letrozole, an antiestrogen selected from the group consisting of: Tamoxifen, Fulvestrant, Raloxifene, and Acolbifene, and a chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Exemestane, an antiestrogen selected from the group consisting of: Tamoxifen, Fulvestrant, Raloxifene, and Acolbifene, and a chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib. Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Fadrozole, an antiestrogen selected from the group consisting of: Tamoxifen, Fulvestrant, Raloxifene, and Acolbifene, and a chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Formestane, an antiestrogen selected from the group consisting of: Tamoxifen, Fulvestrant, Raloxifene, and Acolbifene, and a chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Anastrozole, Tamoxifen, and a chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib. Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Letrozole, Tamoxifen, and a chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Exemestane, Tamoxifen, and a chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Fadrozole, Tamoxifen, and a chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib. Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Formestane, Tamoxifen, and a chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Anastrozole, Fulvestrant, and a chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Letrozole, Fulvestrant, and a chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Exemestane, Fulvestrant, and a chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib. Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Fadrozole, Fulvestrant, and a chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Formestane, Fulvestrant, and a chemotherapeutic agent selected from the group consisting of: Trastuzumab, Gefitinib, Erlotinib, Bevacizumab, Cetuximab, and Bortezomib.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Goserelin and Tamoxifen.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Goserelin, and Fulvestrant. Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Goserelin, and Raloxifene.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Goserelin and Acolbifene.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Leuprolide, and Tamoxifen.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Leuprolide, and Fulvestrant.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Leuprolide, and Raloxifene.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Leuprolide and Acolbifene. Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Goserelin and Anastrozole.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Goserelin and Letrozole.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Goserelin and Exemestane.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Goserelin and Fadrozole.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Goserelin and Formestane. Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Leuprolide and Anastrozole. Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Leuprolide and Letrozole.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Leuprolide and Exemestane.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Leuprolide and Fadrozole.
Another embodiment of this invention is directed to a method of treating or preventing breast cancer in a patient in need of such treatment wherein said treatment comprises administering a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Leuprolide and Formestane.
Another embodiment of this invention is directed to the treatment or prevention of breast cancer in a patient in need of such treatment, said treatment comprising the administration of a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0 and Anastrozole. Another embodiment of this invention is directed to the treatment or prevention of breast cancer in a patient in need of such treatment, said treatment comprising the administration of a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0 and Letrozole.
Another embodiment of this invention is directed to the treatment or prevention of breast cancer in a patient in need of such treatment, said treatment comprising the administration of a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0 and Exemestane.
Another embodiment of this invention is directed to the treatment or prevention of breast cancer in a patient in need of such treatment, said treatment comprising the administration of a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0 and Tamoxifen.
Another embodiment of this invention is directed to the treatment or prevention of breast cancer in a patient in need of such treatment, said treatment comprising the administration of a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0 and Fulvestrant.
Another embodiment of this invention is directed to the treatment or prevention of breast cancer in a patient in need of such treatment, said treatment comprising the administration of a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Anastrozole, and Fulvestrant.
Another embodiment of this invention is directed to the treatment or prevention of breast cancer in a patient in need of such treatment, said treatment comprising the administration of a therapeutically effective amount of at least one compound of formula 1.0 (e.g., one), Letrozole, and Fulvestrant. Another embodiment of this invention is directed to the treatment or prevention of breast cancer in a patient in need of such treatment, said treatment comprising the administration of a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Exemestane, and Fulvestrant.
Another embodiment of this invention is directed to the treatment or prevention of breast cancer in a patient in need of such treatment, said treatment comprising the administration of a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Anastrozole, and Tamoxifen.
Another embodiment of this invention is directed to the treatment or prevention of breast cancer in a patient in need of such treatment, said treatment comprising the administration of a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Letrozole, and Tamoxifen.
Another embodiment of this invention is directed to the treatment or prevention of breast cancer in a patient in need of such treatment, said treatment comprising the administration of a therapeutically effective amount of at least one (e.g., one) compound of formula 1.0, Exemestane, and Tamoxifen.
Other embodiments of this invention are directed to any of the above described embodiments for the treatment of Breast Cancer wherein the chemotherapeutic agent is Trastuzumab. Other embodiments of this invention are directed to any of the above described embodiments for the treatment or prevention of Breast Cancer wherein the method is directed to the treatment of breast cancer.
The compound of formula 1.0, antihormonal agents and chemotherapeutic agents can be administered concurrently or sequentially.
The antihormonal agents and optional chemotherapeutic agents are administered according to their protocols, dosage amounts, and dosage forms that are well know to those skilled in the art (e.g., the Physician's Desk Reference or published literature). For example, for Tamoxifen, Fulvestrant, Raloxifene, Anastrozole, Letrozole, Exemestane, Leuprolide and Goserelin, see the Physician's Desk Reference, 57th Edition, 2003, published by Thomas PDR at Montvale, N.J. 07645-1742, the disclosure of which is incorporated herein by reference thereto. In general, in the embodiments directed to the methods of treating Breast Cancer: (1) the compound of formula 1.0 can be administered daily (e.g., once per day, and in one embodiment twice a day), (2) the aromatase inhibitors can be administered in accordance with the known protocol for the aromatase inhibitor used (e.g., once per day), (3) the antiestrogens can be administered in accordance with the known protocol for the antiestrogen used (e.g., from once a day to once a month), (4) the LHRH analogue can be administered in accordance with the known protocol for the LHRH analogue used (e.g., once a month to once every three months), and (5) the chemotherapeutic agent can be administered in accordance with the known protocol for the chemotherapeutic agent used (e.g., from once a day to once a week). Radiation therapy, if administered in the above treatments for breast cancer, is generally administered according to known protocols before administration of the compound of formula 1.0, antihormonal agents and optional chemotherapeutic agents.
Treatment according to the methods of treating breast cancer is continuous (i.e., a continuous dosing schedule is followed). The treatment is continued until there is a complete response, or until the skilled clinician determines that the patient is not benefiting from the treatment (for example, when there is disease progression). The continuous treatment protocol for breast cancer can be changed to a discontinuous treatment schedule if, in the judgment of the skilled clinician, the patient would benefit from a discontinuous treatment schedule with one or more of the administered drugs. For example, the compound of formula 1.0 can be given using a discontinous treatment schedule while the remaining drugs used in the treatment are given as described herein. An example of a discontinuous treatment protocol for the compound of formula 1.0 is a repeating cycle of three weeks with the compound of formula 1.0 followed by one week without the compound of formula 1.0. After a complete response is achieved with the breast cancer treatment, maintenance therapy with the compound of formula 1.0 can be continued using the dosing described in the methods of this invention. Maintenance therapy can also include administration of the antihormonal agents using the dosing described in the methods of this invention. Maintenance therapy can just be with the antihormonal agents. For example, after a complete response is achieved, an aromatase inhibitor (e.g., Anastrozole, Letrozole or Exemestane) can be continued for up to five years. Or, for example, an antiestrogen, e.g., Tamoxifen, may be used for up to five years after a complete response is achieved. Or, for example, an antiestrogen (e.g., Tamoxifen) can be used for up to five years after a complete response is achieved followed by the use of an aromatase inhibitor (e.g., Anastrozole, Letrozole or Exemestane) for up to five years.
In the embodiments directed to the treatment of breast cancer described above, the compound of formula 1.0 is administered continuously in a total daily dose of about 100 mg to about 600 mg. Usually this amount is administered in divided doses, and in one embodiment this amount is administered twice a day. In one embodiment the compound of formula 1.0 is dosed twice a day in an amount of about 50 mg to about 300 mg per dose. In another embodiment the compound of formula 1.0 is dosed twice a day in an amount of about 100 mg to about 200 mg per dose. Examples include the compound of formula 1.0 being dosed twice a day at 100 mg per dose. Examples also include the compound of formula 1.0 being dosed twice a day at 200 mg per dose.
Anastrozole is administered p.o. and is dosed once a day in amounts of about 0.5 to about 10 mg per dose, and in one embodiment in an amount of about 1.0 mg per dose. Letrozole is administered p.o. and is dosed once a day in amounts of about 1.0 to about 10 mg per dose, and in one embodiment in an amount of about 2.5 mg per dose. Exemestane is administered p.o. and is dosed once a day in amounts of about 10 to about 50 mg per dose, and in one embodiment in an amount of about 25 mg per dose.
Fadrozole is administered p.o. and is dosed twice a day in amounts of about 0.5 to about 10 mg per dose, and in one embodiment in an amount of about 2.0 mg per dose.
Formestane is administered i.m. and is dosed once every two weeks in amounts of about 100 to about 500 mg per dose, and in one embodiment in an amount of about 250 mg per dose. Tamoxifen is administered p.o. and is dosed once a day in amounts of about
10 to about 100 mg per dose, and in one embodiment in an amount of about 20 mg per dose.
Fulvestrant is administered i.m. and is dosed once a month in amounts of about 100 to about 1000 mg per dose, and in one embodiment in an amount of about 250 mg per dose.
Raloxifene is administered p.o. and is dosed once a day in amounts of about 10 to about 120 mg per dose, and in one embodiment in an amount of about 60 mg per dose.
Acolbifene is administered p.o. and is dosed once a day in amounts of about 5 to about 20 mg per dose, and in one embodiment in an amount of about 20 mg per dose.
Goserelin is administered s.c. and is dosed once a month, or once every three months, in amounts of about 2 to about 20 mg per dose, and in one embodiment in an amount of about 3.6 mg per dose when administered once a month, and in another embodiment in an amount of about 10.8 mg per dose when administered once every three months.
Leuprolide is administered s.c. and is dosed once a month, or once every three months, in amounts of about 2 to about 20 mg per dose, and in one embodiment in an amount of about 3.75 mg per dose when administered once a month, and in another embodiment in an amount of about 11.25 mg per dose when administered once every three months.
Trastuzumab is administered by i.v. and is dosed once a week in amounts of about 2 to about 20 mpk per dose, and in one embodiment in an amount of about 2 mpk per dose. Trastuzumab is generally initially administered in a loading dose that is generally twice the dose of the weekly dose. Thus, for example, a 4 mpk loading dose is administered and then dosing is 2 mpk per dose per week.
Gefitinib is administered p.o. and is dosed once a day in amounts of about 100 to about 1000 mg per dose, and in one embodiment in an amount of about 250 mg per dose.
Erlotinib is administered p.o. and is dosed once a day in amounts of about 100 to about 500 mg per dose, and in one embodiment in an amount of about 150 mg per dose.
Bevacizumab is administered i.v. and is dosed once every two weeks in amounts of about 2.5 to about 15 mg per kilogram of body weight per dose, and in one embodiment in an amount of about 10 mg per kilogram per dose.
Cetuximab is administered i.v. and is dosed once a week in amounts of about 200 to about 500 mg per meter squared dose, and in one embodiment in an amount of about 250 mg per meter squared per dose. Bortezomib is administered i.v. and is dosed twice a week for 2 weeks followed by a 10 day rest period (21 day treatment cycle) for a maximum of 8 treatment cycles in amounts of about 1.0 to about 2.5 mg per meter squared per dose, and in one embodiment in an amount of about 1.3 mg per meter squared per dose.
Thus in one embodiment of this invention breast cancer is treated (or prevented) in a patient in need of such treatment wherein said treatment comprises administering to said patient: (1 ) the compound of formula 1.0 orally in an amount of about 50 mg to about 300 mg per dose wherein each dose is administered twice a day, and (2) Anastrozole p.o. in an amount of about 0.5 to about 10 mg per dose wherein each dose is given once a day. In another embodiment of this invention breast cancer is treated (or prevented) in a patient in need of such treatment wherein said treatment comprises administering to said patient: (1) the compound of formula 1.0 orally in an amount of about 100 to 200 mg per dose, wherein each dose is administered twice a day, and (2) Anastrozole in an amount of about 1.0 mg per dose wherein each dose is given once a day. In another embodiment of this invention breast cancer is treated (or prevented) in a patient in need of such treatment wherein said treatment comprises administering to said patient: (1) the compound of formula 1.0 orally in an amount of about 50 mg to about 300 mg per dose wherein each dose is administered twice a day, and (2) Letrozole p.o. in an amount of about 1.0 to about 10 mg per dose wherein each dose is given once a day.
In another embodiment of this invention breast cancer is treated (or prevented) in a patient in need of such treatment wherein said treatment comprises administering to said patient: (1 ) the compound of formula 1.0 orally in an amount of about 100 to 200 mg per dose, wherein each dose is administered twice a day, and (2) Letrozole p.o. in an amount of about 2.5 mg per dose wherein each dose is given once a day.
In another embodiment of this invention breast cancer is treated (or prevented) in a patient in need of such treatment wherein said treatment comprises administering to said patient: (1 ) the compound of formula 1.0 orally in an amount of about 50 mg to about 300 rηg per dose wherein each dose is administered twice a day, and (2) Exemestane p.o. in an amount of about 10 to about 50 mg per dose wherein each dose is given once a day.
In another embodiment of this invention breast cancer is treated (or prevented) in a patient in need of such treatment wherein said treatment comprises administering to said patient: (1 ) the compound of formula 1.0 orally in an amount of about 100 to 200 mg per dose, wherein each dose is administered twice a day, and (2) Exemestane in an amount of about 25 mg per dose wherein each dose is given once a day. In another embodiment of this invention breast cancer is treated (or prevented) in a patient in need of such treatment wherein said treatment comprises administering to said patient: (1 ) the compound of formula 1.0 orally in an amount of about 50 mg to about 300 mg per dose wherein each dose is administered twice a day, and (2) Fulvestrant i.m. in an amount of about 100 to about 1000 mg per dose wherein each dose is given once a month.
In another embodiment of this invention breast cancer is treated (or prevented) in a patient in need of such treatment wherein said treatment comprises administering to said patient: (1 ) the compound of formula 1.0 orally in an amount of about 100 to 200 mg per dose, wherein each dose is administered twice a day, and (2) Fulvestrant i.m. in an amount of about 250 mg per dose wherein each dose is given once a month.
In another embodiment of this invention breast cancer is treated (or prevented) in a patient in need of such treatment wherein said treatment comprises administering to said patient: (1 ) the compound of formula 1.0 p.o. in an amount of about 50 mg to about 300 mg per dose wherein each dose is administered twice a day, and (2) Tamoxifen p.o. in an amount of about 10 to about 100 mg per dose wherein each dose is given once a day.
In another embodiment of this invention breast cancer is treated (or prevented) in a patient in need of such treatment wherein said treatment comprises administering to said patient: (1) the compound of formula 1.0 p.o. in an amount of about 100 to 200 mg per dose, wherein each dose is administered twice a day, and (2) Tamoxifen p.o. in an amount of about 20 mg per dose wherein each dose is given once a day.
In other embodiments of the invention breast cancer is treated in a patient in need of such treatment wherein said treatment comprises the administration of the compound of formula 1.0, one of the aromatase inhibitors (e.g., Anastrozole, Letrozole, or Exemestane, and in one embodiment Anastrozole), and one of the antiestrogens (e.g., Fulvestrant or Tamoxifen), wherein the compound of formula 1.0, aromatase inhibitor and antiestrogen are administered in the dosages described above.
Thus, for example in another embodiment of this invention breast cancer is treated (or prevented) in a patient in need of such treatment wherein said treatment comprises administering to said patient : (1 ) the compound of formula 1.0 p.o. in an amount of about 50 mg to about 300 mg per dose wherein each dose is administered twice a day, (2) Anastrozole p.o. in an amount of about 0.5 to about 10 mg per dose wherein each dose is given once a day, and (3) Fulvestrant i.m. in an amount of about 100 to about 1000 mg per dose wherein each dose is given once a month.
In another embodiment of this invention breast cancer is treated (or prevented) in a patient in need of such treatment wherein said treatment comprises administering to said patient: (1 ) the compound of formula 1.0 p.o in an amount of about 100 to 200 mg per dose, wherein each dose is administered twice a day, (2) Anastrozole p.o. in an amount of about 1.0 mg per dose wherein each dose is given once a day, and (3) Fulvestrant i.m. in an amount of about 250 mg per dose wherein each dose is given once a month. In another embodiment of this invention breast cancer is treated (or prevented) in a patient in need of such treatment wherein said treatment comprises administering to said patient: (1 ) the compound of formula 1.0 p.o. in an amount of about 50 mg to about 300 mg per dose wherein each dose is administered twice a day, (2) Letrozole p.o in an amount of about 1.0 to about 10 mg per dose wherein each dose is given once a day, and (3) Fulvestrant in an amount of about 100 to about 1000 mg per dose wherein each dose is given once a month.
In another embodiment of this invention breast cancer is treated (or prevented) in a patient in need of such treatment wherein said treatment comprises administering to said patient: (1 ) the compound of formula 1.0 p.o. in an amount of about 100 to 200 mg per dose, wherein each dose is administered twice a day, (2) Letrozole p.o. in an amount of about 2.5 mg per dose wherein each dose is given once a day, and (3) Fulvestrant i.m. in an amount of about 250 mg per dose wherein each dose is given once a month. In another embodiment of this invention breast cancer is treated (or prevented) in a patient in need of such treatment wherein said treatment comprises administering to said patient: (1) the compound of formula 1.0 p.o. in an amount of about 50 mg to about 300 mg per dose wherein each dose is administered twice a day, (2) Exemestane p.o. in an amount of about 10 to about 50 mg per dose wherein each dose is given once a day, and (3) Fulvestrant i.m. in an amount of about 100 to about 1000 mg per dose wherein each dose is given once a month.
In another embodiment of this invention breast cancer is treated (or prevented) in a patient in need of such treatment wherein said treatment comprises administering to said patient: (1 ) the compound of formula 1.0 p.o. in an amount of about 100 to 200 mg per dose, wherein each dose is administered twice a day, (2) Exemestane p.o. in an amount of about 25 mg per dose wherein each dose is given once a day, and (3) Fulvestrant i.m. in an amount of about 250 mg per dose wherein each dose is given once a month.
In another embodiment of this invention breast cancer is treated (or prevented) in a patient in need of such treatment wherein said treatment comprises administering to said patient: (1) the compound of formula 1.0 p.o. in an amount of about 50 mg to about 300 mg per dose wherein each dose is administered twice a day, (2) Anastrozole p.o. in an amount of about 0.5 to about 10 mg per dose wherein each dose is given once a day, and (3) Tamoxifen p.o. in an amount of about 10 to about 100 mg per dose wherein each dose is given once a day.
In another embodiment of this invention breast cancer is treated (or prevented) in a patient in need of such treatment wherein said treatment comprises administering to said patient: (1 ) the compound of formula 1.0 p.o. in an amount of about 100 to 200 mg per dose, wherein each dose is administered twice a day, (2) Anastrozole p.o. in an amount of about 1.0 mg per dose wherein each dose is given once a day, and (3) Tamoxifen p.o. in an amount of about 20 mg per dose wherein each dose is given once a day.
In another embodiment of this invention breast cancer is treated (or prevented) in a patient in need of such treatment wherein said treatment comprises administering to said patient: (1) the compound of formula 1.0 p.o. in an amount of about 50 mg to about 300 mg per dose wherein each dose is administered twice a day, (2) Letrozole p.o. in an amount of about 1.0 to about 10 mg per dose wherein each dose is given once a day, and (3) Tamoxifen p.o. in an amount of about 10 to about 100 mg per dose wherein each dose is given once a day.
In another embodiment of this invention breast cancer is treated (or prevented) in a patient in need of such treatment wherein said treatment comprises administering to said patient: (1 ) the compound of formula 1.0 p.o. in an amount of about 100 to 200 mg per dose, wherein each dose is administered twice a day, (2) Letrozole p.o. in an amount of about 2.5 mg per dose wherein each dose is given once a day, and (3) Tamoxifen p.o. in an amount of about 20 mg per dose wherein each dose is given once a day.
In another embodiment of this invention breast cancer is treated (or prevented) in a patient in need of such treatment wherein said treatment comprises administering to said patient: (1) the compound of formula 1.0 p.o. in an amount of about 50 mg to about 300 mg per dose wherein each dose is administered twice a day, (2) Exemestane p.o. in an amount of about 10 to about 50 mg per dose wherein each dose is given once a day, and (3) Tamoxifen p.o. in an amount of about 10 to about 100 mg per dose wherein each dose is given once a day. In another embodiment of this invention breast cancer is treated (or prevented) in a patient in need of such treatment wherein said treatment comprises administering to said patient: (1) the compound of formula 1.0 p.o. in an amount of about 100 to 200 mg per dose, wherein each dose is administered twice a day, (2) Exemestane p.o. in an amount of about 25 mg per dose wherein each dose is given once a day, and (3) Tamoxifen p.o. in an amount of about 20 mg per dose wherein each dose is given once a day.
Those skilled in the art will appreciate that when other combinations of antihormonal agents are used, the individual antihormonal agent is used in the amounts specified above for that individual antihormonal agent. Other embodiments of the treatment of Breast Cancer are directed to the methods of treating Breast Cancer described above wherein the compound of formula 1.0 is dosed twice a day in an amount of about 100 mg per dose.
Other embodiments of the treatment of Breast Cancer are directed to the methods of treating Breast Cancer described above wherein the compound of formula 1.0 is dosed twice a day in an amount of about 200 mg per dose.
Other embodiments of the treatment of Breast Cancer are directed to the methods of treating Breast Cancer described above wherein a chemotherapeutic agent is administered in addition to the compound of formula 1.0 and antihormonal agent (or antihormonal agents). In these embodiments the dosage ranges of the compound of formula 1.0 and antihormonal agents are as those described above in the combination therapies, or those described above for the individual compound of formula I and antihormonal agents, and the dosages of the chemotherapeutic agents are those described above for the individual chemotherapeutic agent. The dosages for the chemotherapeutic agents are well known in the art.
Other embodiments of this invention are directed to pharmaceutical compositions comprising the compound of formula 1.0 and at least one antihormonal agent and a pharmaceutically acceptable carrier.
Other embodiments of this invention are directed to pharmaceutical compositions comprising the compound of formula 1.0, at least one antihormonal agent, at least one chemotherapeutic agent, and a pharmaceutically acceptable carrier.
Other embodiments of this invention are directed to pharmaceutical compositions comprising the compound of formula 1.0, at least one chemotherapeutic agent, and a pharmaceutically acceptable carrier.
Those skilled in the art will appreciate that the compounds (drugs) used in the methods of this invention are available to the skilled clinician in pharmaceutical compositions (dosage forms) from the manufacturer and are used in those compositions. So, the recitation of the compound or class of compounds in the above described methods can be replaced with a recitation of a pharmaceutical composition comprising the particular compound or class of compounds. For example, the embodiment directed to a method of treating cancer comprising administering to a patient in need of such treatment therapeutically effective amounts of the compound of formula 1.0, a taxane, and a platinum coordination compound, includes within its scope a method of treating cancer comprising administering to a patient in need of such treatment therapeutically effective amounts of a pharmaceutical composition comprising the compound of formula 1.0, a pharmaceutical composition comprising a taxane, and a pharmaceutical composition comprising a platinum coordination compound.
Those skilled in the art will recognize that the actual dosages and protocols for administration employed in the methods of this invention may be varied according to the judgment of the skilled clinician. The actual dosage employed may be varied depending upon the requirements of the patient and the severity of the condition being treated. Determination of the proper dosage for a particular situation is within the skill of the art. A determination to vary the dosages and protocols for administration may be made after the skilled clinician takes into account such factors as the patient's age, condition and size, as well as the severity of the cancer being treated and the response of the patient to the treatment. The amount and frequency of administration of the compound of formula 1.0 and the chemotherapeutic agents will be regulated according to the judgment of the attending clinician (physician) considering such factors as age, condition and size of the patient as well as severity of the cancer being treated.
The chemotherapeutic agent can be administered according to therapeutic protocols well known in the art. It will be apparent to those skilled in the art that the administration of the chemotherapeutic agent can be varied depending on the cancer being treated and the known effects of the chemotherapeutic agent on that disease. Also, in accordance with the knowledge of the skilled clinician, the therapeutic protocols (e.g., dosage amounts and times of administration) can be varied in view of the observed effects of the administered therapeutic agents on the patient, and in view of the observed responses of the cancer to the administered therapeutic agents.
The initial administration can be made according to established protocols known in the art, and then, based upon the observed effects, the dosage, modes of administration and times of administration can be modified by the skilled clinician. The particular choice of chemotherapeutic agent will depend upon the diagnosis of the attending physicians and their judgement of the condition of the patient and the appropriate treatment protocol.
The determination of the order of administration, and the number of repetitions of administration of the chemotherapeutic agent during a treatment protocol, is well within the knowledge of the skilled physician after evaluation of the cancer being treated and the condition of the patient.
Thus, in accordance with experience and knowledge, the practicing physician can modify each protocol for the administration of an chemotherapeutic agent according to the individual patient's needs, as the treatment proceeds. All such modifications are within the scope of the present invention.
The particular choice of antihormonal agents, optional chemotherapeutic agents and optional radiation will depend upon the diagnosis of the attending physicians and their judgment of the condition of the patient and the appropriate treatment protocol.
The determination of the order of administration, and the number of repetitions of administration of the antihormonal agents, optional chemotherapeutic agents and optional radiation during a treatment protocol, is well within the knowledge of the skilled physician after evaluation of the breast cancer being treated and the condition of the patient.
Thus, in accordance with experience and knowledge, the practicing physician can modify each protocol for the administration of antihormonal agents, optional chemotherapeutic agents and optional radiation according to the individual patient's needs, as the treatment proceeds. All such modifications are within the scope of the present invention.
The attending clinician, in judging whether treatment is effective at the dosage administered, will consider the general well-being of the patient as well as more definite signs such as relief of cancer-related symptoms (e.g., pain, cough (for lung cancer), and shortness of breath (for lung cancer)), inhibition of tumor growth, actual shrinkage of the tumor, or inhibition of metastasis. Size of the tumor can be measured by standard methods such as radiological studies, e.g., CAT or MRI scan, and successive measurements can be used to judge whether or not growth of the tumor has been retarded or even reversed. Relief of disease-related symptoms such as pain, and improvement in overall condition can also be used to help judge effectiveness of treatment.
The compounds of the invention can be made according to the processes described in US 2007/0191604 published August 16, 2007, U.S. Serial No. 11/810282 filed June 5, 2007, as well as the processes described below. The disclosures of US 2007/0191604 and U.S. Serial No. 11/810282 are incorporated herein by reference thereto.
The LCMS conditions are: (1 ) column: C-18 reverse phase, 5um, 4.6 x 50 mm, (2) MSPE Sciex API-150EX, and (3) HPLC: Shimadzu LC-10 ADvp, 1 ml/min, linerar gradient 10% acetonitirle in water to 95% acetonitrile in water, both contain 0.05% TFA
Example 1
Synthesis of 3-Methoχy-1 -(2-f 4-F4-(1 -methyl-1 H-f1 ,2,41triazol-3-yl)-phenvn-3.6- dihvdro-2H-pyridin-1 -yl!-2-oxo-ethyl)-pyrrolidine-3-carboxylic acid F3-(6- isopropoxy-pyridin-3-yl)-1 H-indazol-5-vll-amide
Synthesis of 2-chloro-1-(4-r4-(1 -methyl-1 H-Pl .2.41triazol-3-yl)-phenyl1-3.6- dihvdro-2H-pyridin-1-yl)-ethanone
Figure imgf000213_0002
Step 1 : Preparation of 4-bromo-benzimidic acid ethyl ester
4-Bromo-benzonitrile (5g) was suspended in absolute EtOH (100 ml) and cooled to 0-5 ° C. HCI gas was bubbled through, initially vigorously for several minutes and later slowly for 5 hours. The resulting solution was allowed to stir overnight. Most of solvent was removed and the precipitate was filtered, washed with EtOH twice and dried to afford compound 2BH (4.1 g) as white solid. Step 2: Preparation of Compound 3BH
Figure imgf000214_0001
The 4-bromo-benzimidic acid ethyl ester (2.12g, 8 mmols) was dissolved in pyridine (20 ml). Methylhydrazine (640 μl, 12 mmols) was added with stirring and the resulting mixture was allowed to stir overnight. The reaction mixture was concentrated under reduced preessure, and added ether, filtered, washed with ether three times and dried to provide the compound 3BH (2.2 g).
Step 3: Preparation of 3-(4-bromo-phenyl)-1 -methyl- 1 W-[I, 2, 4]triazole
Figure imgf000214_0002
A mixture of compound 3BH (2.2 g) in formic acid (30 ml) was refluxed overnight and concentrated. The residue was treated with sat. NaHCO3, and extracted with EtOAc three times. The combined organics were dried over MgSO4. After concentration, compound 4BH was obtained as colorless crystals (1.39 g). (Note: it was found that the reaction can be done in just two hours. In large scale synthesis, use 10% NaOH to replace NaHCO3).
Step 4: Preparation of 4-[4-(1 -methyl- 1 W-[1 , 2,4]triazol-3-yl)-phenyl]-3,6-dihydro- 2W-pyridine-1-carboxylic acid te/f-butyl ester
Figure imgf000214_0003
To a large pressure flask were charged compound 4BH (13.3 g, 55.9 mmols), 4-(4,4,5,5-tetramethyl-[1 ,3,2]dioxaborolan-2-yl)-3,6-dihydro-2H-pyridine-1 -carboxylic acid te/t-butyl ester (19 g, 61.5 mmols), [1 ,1'-bis(diphenylphosphino)- ferrocene]dichloropalladium(ll), complex with dichloromethane(1 :1) (2.3 g, 2.8 mmols), K2CO3 (23.2 g, 168 mmols) and DME/water (5:1 , 120 ml). The mixture was briefly degassed with Ar for -0.5 minute, capped and stirred at 8OC overnight. After cooling, the reaction mixture was diluted with EtOAc and brine. Organic layer was isolated, and dried (MgSO4). After concentration, the residue was purified on silica gel. Elution with MeOH/EtOAc (0-10%) gave the desired product 5BH (13.9 g, 73%).
Step 5: Preparation of 4-[4-(1 -methyl-1 H-[1 , 2,4]triazol-3-yl)-pheny|]-1 , 2,3,6- tetrahydro-pyridine hydrochloride
Figure imgf000215_0001
The Boc group can be removed by treating compound 5BH with 4N HCI in dioxane at rt for two hours. Removal of solvent under vacuum gave compound 6BH.
Step 6: Preparation of 2-chloro-1-{4-[4-(1 -methyl-1 H-[1 ,2,4]triazol-3-yl)-phenyl]- 3,6-dihydro-2H-pyridin-1 -yl}-ethanone
Figure imgf000215_0002
To a cold (00C) solution of 4-[4-(1 -methyl-1 H-[1 ,2,4]triazol-3-yl)-phenyl]- 1 ,2,3,6-tetrahydro-pyridine 6BH (13.7g, 44mmol) in dichloromethane (450ml) was added TEA (37ml, 264mmol) dropwise. After stirred at 0 0C for 10min, chloroacetyl chloride (10.5 ml, 132 mmols) was added to the reaction mixture. The resulting mixture was stirred at 0 0C for 1 hr., and quenched with water (165ml). The reaction mixture was diluted with dichloromethane (600ml). The organic layer was separated and washed with brine, dried over MgSO4. Reaction mixture was concentrated to ~50ml, ether was added and the solid was filtered out to get the desired product 7BH (8.74g). Svnthesis of S-Methoxy-pyrrolidine-S-carboxylic acid methyl ester Step 1 : Preparation of methyl α, α-dimethoxypropionate
Figure imgf000216_0001
The procedure by Ernest Wenkert, et al. (JACS, 1983, 705, 2021 -2029) was followed. A solution of methyl pyruvate (44g), trimethyl orthoformate (62 ml), concentrated H2SO4 (0.2 ml) in MeOH (120 ml) was relϊuxed for 4 hours. In the next one hour period, solvent (about 80 ml) was distilled out. The reaction mixture was cooled to 10 0C, poured into a KOH solution (1 .2 g KOH in 600 ml water), and extracted with ether (3x). Combined ether extracts were washed with brine and dried (MgSO4). After concentration, the residue was distilled under vacuum to provide the acetal (8BH) (4Og, 62%, 40-43C/1 torr).
Step 2: Preparation of 2-methoxyacrylate
Figure imgf000216_0002
The procedure by Ernest Wenkert, et al. (JACS, 1983, 705, 2021 -2029) was followed. To a one neck flask was charged α, α-dimethoxypropionate (8BH) (150 g) and Tolunesulfonic acid monohydrate (3g) and a short path distillation head was attached. The mixture was heated at 140 0C (oil bath temperature) and methanol began to come out first. The product (76 g) of (9BH) was then distilled out later after oil bath temperature was raised over 190 0C.
Step 3: Preparation of 1-benzyl-3-methoxy-pyrrolidine-3-carboxylic acid methyl ester
Figure imgf000216_0003
To a stirred solution of methyl 2-methoxyacrylate (20.8 g, 179 mmols) and N- (methoxymethyl)-Λ/-(trimethylsilylmethyl)benzylamine (55 ml, 215 mmols) in dichloromethane (160 ml) was added at 0 0C a solution of trifluoroacetic acid (2ml) in dichloromethane (10 ml). The resulting solution was warmed to room temperature and stirred overnight. After concentration, the crude product was purified by column chromatography on silica gel eluting with a solution of ethyl acetate/hexanes/Et3N (1000:3000:4 to 1000:1000:3) to give the title compound (10BH) (17.7 mg, 40 %). (Note: adding Et3N is essential to ensure sharp separation.)
Step 4: Preparation of S-methoxy-pyrrolidine-S-carboxylic acid methyl ester tartaric acid salt
acid
Figure imgf000217_0001
2.49 gm of 1 -benzyl-S-methoxy-pyrrolidine-S-carboxylic acid methyl ester (10BH) was hydrogenated in ethanol using 10% Pd/C at 55 psi hydrogen for 24 hrs. Filtration of the Pd/C followed by evaporation of the ethanol 1.6 gm of crude de- benzylated product (11 BH). The crude product was dissolved in 95 ml of methanol and 1.35 gm of L-tartaric acid added. After 24 hrs, the crystals were filtered and re- crystallized from methanol to give 13.4 grams of title product (12BH).
Step 5: Preparation of 3-Methoxy-pyrrolidine-1,3-dicarboxylic acid 1-tert-butyl ester
Figure imgf000217_0002
To a cold (O0C) solution of 12BH (28 g, 90.52 mmol) in dry CH2CI2 (250 ml_) was added triethylamine (31.5 ml_, 226.32 mmol, 2.5 equiv) followed by (Boc)2O (25.7 g, 117.68 mmol, 1.3 equiv). The resulting mixture was stirred from O0C to rt for overnight then diluted with CH2CI2, which was washed with saturated aqueous NaHCO3 solution and brine, dried (MgSO4) and concentrated. Chromatograph on silica gel (hexanes/ethyl acetate, 4:1 ) gave 13BH (23.5 mg, 90.52 mmol, 100%) as a colorless oil.
To a stirred solution of 13BH (23.5 mg, 90.52 mmol) in THF/MeOH (175 mL/175 ml_) was added 135 ml_ of LiOH (1 M in H2O, 135 mmol, 1.5 equiv). The reaction mixture was stirred at rt for overnight, to which 135 ml_ of 1 N HCI was added. The resulting mixture was stirred for additional 15 min and concentrated, azeotroped with dioxane (150 ml_ x 3) to give 14BH (42.32 g) as a white solid, which can be used in the next step without further purification.
Alternatively compound 11 BH can be prepared as follows:
Figure imgf000218_0001
To an ice cold solution of 3-Oxo-pyrrolidine-1-carboxylic acid benzyl ester 1a (25Og, 1.14 mol) in 3.5L anhydrous dichloromethane was added KCN (7.5g, 0.12 mol), followed by 18-crown-6 (3Og, 0.11 mol), though not completely dissolved, was added TMSCN (183 ml_, 1.37 mol) slowly over a period of 20 min. The reaction was stirred at ambient temperature for 1 overnight. A semi-saturated NaHCO3 solution (2L) was added at 150C, stirred for 10 min and then the organic layer was separated, dried over magnesium sulfate, filtered and evaporated to give 3-Cyano-3- trimethylsilanyloxy-pyrrolidine-1 -carboxylic acid benzyl ester 2a, 422 g (>100%). S-Cyano-S-trimethylsilanyloxy-pyrrolidine-i -carboxylic acid benzyl ester 2a (422 g) in 4L of anhydrous MeOH, was added 2.2 L of 4N HCI in dioxane. The reaction was refluxed for 13 h and stirred at ambient temperature for 1 overnight. Solvents were removed, suspended in 5 L of CH2Ck, washed with 4+3L of water, adjusted the pH to 6-7 with aq. NaHCO3, dried over magnesium sulfate, filtered and evaporated to give 3-Hydroxy-pyrrolidine-1 ,3-dicarboxylic acid 1 -benzyl ester 3-methyl ester 3a, 278 g (2 steps, 87%).
To a suspension of NaH (52g, 1.3 mol) in 2.2 L of anhydrous DMF at 80C, was added a solution of 3-Hydroxy-pyrrolidine-1 ,3-dicarboxylic acid 1 -benzyl ester 3- methyl ester 3a (278 g, 1.0 mol) in 700 DMF, keeping the reaction temperature below 110C. After complete addition (-20 min), ice bath was removed, stirred at 16-180C for 1 h, and then at ambient temperature for 1 h. Cooled back to 150C, added MeI (81 ml_, 1.3 mol) slowly. Reaction mixture was stirred at ambient temperature for 1 overnight. The reaction mixture was then poured in a cold water (4L) and then extracted with Et2O(6L) and EtOAc(2L), washed the organic later with water (5L), brine(700 mL), dried over magnesium sulfate, filtered and evaporated to give crude 3-Methoxy- pyrrolidine-1 ,3-dicarboxylic acid 1 -benzyl ester 3-methyl ester 4a 289 g (99% crude yield, contains mineral oil) (stirring/separation with pentane wash gave 4a, 268.2 g (92%). 3-Methoxy-pyrrolidine-1 ,3-dicarboxylic acid 1 -benzyl ester 3-methyl ester 4a in
MeOH (2200 mL), was added 14 g of 10% Pd/C (-50% in water). The reaction mixture was hydrogenated using H2 at -55 psi pressure (epen valve) for 1 overnight. The reaction mixture was filtered, dried to give 3-Methoxy-pyrrolidine-3-carboxylic acid methyl ester 11 BH 126 g. (overall yield of 70% for 4 steps, no column purification).
Synthesis of S-Methoxy-pyrrolidine-S-carboxylic acid r3-(6-isopropoxy-pyridin-3- VlH H-indazol-5-yll-amide
Figure imgf000219_0001
Step 1 :
5-bromo-1 H-pyridone 1 Bl (10Og, 0.58 mol), potassium carbonate (238g, 1.73mol) and 2-iodopropane (86ml, 0.86mol) were stirred in DMF (1 L) at r.t. for 1 day. The mixture were diluted with ethyl acetate and water, layers were separated. The separated organic layer was washed with water (X2), dried (Mg SO4) and filtered. Solvents were removed in vacuum and column purification [5% ethyl acetate in hexanes] gave first the less polar 5-isopropoxypyridine 2BIa (73g, 59%) as colourless liquid. Continuous elution with [50% ethyl acetate in hexanes] gave the more polar 5- bromo-1 -isopropylpyridone 2BIb as white solid (22g, 18%).
Step 2:
Figure imgf000220_0001
5-isopropoxypyridine 2BIa (1Og1 0.046mol), bis(pinacolato)diboron (14.1g, 0.056mol), potassium acetate (13.6g, 0.14mol) and PdCI2(dppf)2.CH2CI2 (3.78g, 0.0046mol) were weight into a 2-necked 1 L flask equipped with a water condenser. DMSO (100 ml) was added and the mixture was purged with nitrogen for 15 min. The mixture was heated at 100 0C under nitrogen for 2 hr. After being cooled to r.t., water (100ml), toluene (100ml), ethanol (100ml), potassium carbonate (32g, 0.23mol) and bromoindazole 3Bl (22.4g, 0.046mol) were added. The mixture were purged with nitrogen for 10 min at r.t. and Pd(Ph3P)4 (5.35g, 0.0046mol) was added. The final mixture were heated at 100 0C for 2 hr and cooled to r.t. Water and ethyl acetate were added. Solids were filtered through Celite. Layers were separated and the separated organic layer was washed with water (X2). The combined aqueous layers were back extracted with ethyl acetate (X1). The combined organic layers were dried (MgSO4), filtered and solvents were removed in vacuum. Column purification [Hexanes-ethyl acetate = 9:1 (v/v)] gave isopropoxyindazole 4Bl (2Og1 80%) as yellow solid.
Step 3:
Figure imgf000221_0001
Isopropoxyindazole 4Bl (2Og, 0.037mol) and Pd/C (10%, 50% wet, 7.8g, 0.0037mol) were stirred in toluene (100ml) and 2-propanol (200ml) under H2 (balloon) at r.t. for 1 day. The solid catalyst was filtered through Celite and solvents were removed in vacuum to give aminoindazole 5Bl (quant.) as off-white solid.
Step 4:
Figure imgf000221_0002
Aminoindazole 5Bl (39g, 0.076mol) and pyrrolidinecarboxylic acid 14BH (32g, 0.069mol) were dissolved in DMF (300ml) at r.t. HATU (29g, 0.076mol) followed by 1Pr2NEt (14.5ml, 0.083mol) were added. The mixture was stirred at r.t. overnight and was diluted with ethyl acetate and water. Layers were separated. The separated organic layer was washed with water (X2), dried (MgSO4) and filtered. Concentration in vacuum followed by column purification [hexanes - ethyl acetate = 4:1 (v/v)] gave crude 7Bl as off-white foam. Step 5:
Figure imgf000222_0001
The crude 7Bl was stirred in a mixture of dichloromethane (300ml), trifluoroacetic acid (100ml) and water (50ml) at r.t. overnight. The mixture was cooled at 0 0C and quenched carefully with saturared aqueous sodium bicarbonate. Solvents were removed in vacuum. Dilute with water and ethyl acetate. Layers were separated and the separated aqueous layer was extracted with ethyl acetate (X3). The combined organic layers were dried (MgSO4), filtered and solvents were removed in vacuum. Column purification [5 to 10% MeOH (7N ammonia) in dichloromethane] gave pyrrolidine 8Bl (23g, 84%) as off-white solid.
Alternatively:
Figure imgf000222_0002
Boronate 1 BJ (1.Og, 3.80mmol), bromoindazole 2BJ (1.84g, 3.80mmol), sodium bicarbonate (1.21 g, 11.4mmol) were charged into a sealed-tube. Toluene (30 ml), ethanol (30 ml) and water (15 ml) were added. The slurry was purged with nitrogen for 15 min. and Pd(Ph3P)4 (439 mg, 0.38mmol) was added in one portion. The mixture was heated in the sealed-tube at 90 0C overnight. After being cooled to r.t., water and ethyl acetate were added and the layers were separated. The separated organic layer was washed with water, dried (MgSO4) and filtered. Solvents were removed in vacuum. The residue was dissolved in toluene (100ml). Methanol (100ml) followed with Pd/C (809mg, 0.38mmol, 50% wet) were added. The mixture was stirred under H2 (balloon) overnight. The catalyst was filterd and solvents were removed in vacuum. Column purification [hexanes - ethyl acetate = 2:1 (v/v)] gave aniline 5Bl (1.2g, 61%, 2 steps) as off-white solid.
Svnthesis of 3-Methoxy-1 -(2-f 4-F4-(1 -methyl-1 H-M ,2,41triazol-3-yl)-phenyll-3.6- dihvdro-2H-pyridin-1 -yl)-2-oxo-ethyl)-pyrrolidine-3-carboxylic acid T3-(6- isopropoxy-pyridin-3-yl)-1 H-indazol-5-vπ-amide
Figure imgf000224_0001
A mixture of compound 7BH (7.88gm, 24.93mmol), compound 8Bl (9.86gm,
24.93mmol), and DIEA (26.1 ml, 149.62mmol) in DMF (200ml) was stirred at room temperature for 5hr. Reaction was completed to about 89% based on the LCMS. It was then heated at 50 0C for overnight (16hr.) LCMS shows the reaction is complete. DMF was removed under reduced pressure. The crude was dissolved in 700ml of DCM and washed with 35ml of water once. Aqueous layer was extracted with
20%MeOH/DCM (2 x 120ml). The combined organic extracts were homogenized with MeOH and dried over MgSO4. The solvent was removed and the crude was purified by column chromatography using 20%MeOH/EtoAc to get the desired product A1 as a yellow solid (70%). (LCMS M+1= 674, ret. time= 2.91 min.) 1 H NMR (400 MHz, CD3OD):δ 8.67 (S, 1 H)1 8.42 (S, 1 H), 8.35 (S, 1 H), 8.15-8.19 (m, 1 H), 7.94(dd, 2H, J= 8.4 & 10 Hz), 7.64 (m, 1 H), 7.49 (m, 3H), 6.84 (m, 1 H), 6.20 (d, 1 H, J= 11.2 Hz), 5.3 (m, 1 H)1 4.28 (s, 1 H), 4.23 (m, 1 H), 3.97 (s, 3H), 3.8 (m, 2H), 3.56 (d, 1 H, 2.4 Hz), 3.52(d, 1 H, 6.4 Hz), 3.31 (s, 3H), 3.17 (t, 1 H, J= 10 Hz), 3.07 (t, 1 H, J= 12 Hz), 2.81 (m, 1 H), 2.81 (m, 1 H), 2.65 (s, 1 H), 2.58 (s, 1 H), 2.43 (m, 1 H), 2.17 (m, 1 H), 1.32 (d, 6H, J=6.4 Hz )
Example 2
Synthesis of 3-Methoxy-1-(2-(4-r4-(1-methyl-1 H-M ,2,41triazol-3-yl)-phenyll-3,6- dihydro-2H-pyridin-1 -yl}-2-oxo-ethyl)-pyrrolidine-3-carboxylic acid r3-(6-hydroxy- pyridin-3-vπ-1 H-indazol-5-vll-amide
Figure imgf000225_0001
The above compound (A20) was isolated from the decomposition of di-HCL salt of 3-Methoxy-1 -(2-{4-[4-(1 -methyl-1 H-[1 ,2,4]triazol-3-yl)-pheny|]-3,6-dihydro-2H- pyridin-1 -ylJ^-oxo-ethyO-pyrrolidine-S-carboxylic acid [3-(6-isopropoxy-pyridin-3-yl)- 1 H-indazol-5-yl]-amide.
LCMS M+1 634, ret. time = 2.28 min. 1H-NMR (400 MHz, DMSO-d6): δ 13.09 (br, 1 H), 11.84 (br, 1 H), 10.02 (s, 1 H), 8.51 (s, 1 H), 8.37 (s, 1 H), 8.02 (dd, 1 H1 J=9.5 Hz & 2.5 Hz), 7.95 (m, 2H), 7.82 (d, 1 H, J=1.9 Hz), 7.71 (m, 1 H), 7.51 (m, 3H), 6.52 (d, 1 H, J=9.5 Hz), 6.27 (m, 1 H), 4.08-4.34 (m, 2H), 3.92 (s, 3H), 3.70 (m, 2H), 3.49 (m, 2H), 3.24 (s, 3H), 3.16 (d, 1 H, J=5.2 Hz), 2.84-3.13 (m, 3H), 2.55-2.76 (m, 2H), 2.36 (m, 1 H), 2.05 (m, 1 H). Example 3
Synthesis of 1-(2-(4-[4-(1.5-Dimethyl-i H-M ,2,41triazol-3-yl)-phenyll- S^-dihvdro-ΣH-pyridin-i-ylVΣ-oxo-ethvπ-S-methoxy-pyrrolidine-S-carboxylic acid r3-(6-isopropoxy-pyridin-3-yl)-1 H-indazol-S-yli-amide
Figure imgf000226_0001
Synthesis of (S)-1-(2-(4-(4-(1.5-dimethyl-1 H- 1.2.4-triazol-3-yl)phenyl)-5.6- dihvdropyridin-1(2H)-yl)-2-oxoethyl)-/v-(3-(6-isopropoxypyridin-3-yl)-1 H-indazol-
5-yl)-3-methoxypyrrolidine-3-carboxamide
Step 1 : Preparation of 3-(4-bromo-phenyl)-1 , 5-dimethyl-1 H-[1 ,2,4]triazole
Figure imgf000226_0002
A mixture of 4-bromo-/V-methylbenzimidohydrazide hydrochloride 1 (1.7 g) (prepared according to a procedure in synthesis of Sch-1499895) in acetic anhydride (10 ml) was heated at 100 0C for 0.5 hour. After cooling and concentration under reduced pressure, the residue was dissolved in EtOAc, washed with saturated NaHCO3 twice, brine and dried (MgSO4). After evaporation of solvent, the residue was purified on silica gel. Elution with EtOAc gave 3-(4-bromophenyl)-1 , 5-dimethyl- 1 H-[1 ,2,4]-triazole 2BK (1.04 g).
Step 2: Preparation of ferf-butyl 4-(4-(1 , 5-dimethyl-1 H-Λ ,2,4-triazol-3-yl)phenyl)- 5,6-dihydropyridine-1(2H)-carboxylate
Figure imgf000226_0003
To a pressure tube were charged compound 2BK (252 mg, 1 mmol), 4- (4,4,5, 5-tetramethyl-[1 ,3,2]dioxaborolan-2-yl)-3,6-dihydro-2/-/-pyridine-1-carboxylic acid te/t-butyl ester (403 mg, 1.3 mmol), [1 ,1 '-bis(diphenylphosphino)- ferrocene]dichloropalladium(ll), complex with dichloromethane(1 :1) (41 mg, 0.05 mmol), K2CO3 (410 mg, 3 mmol) and DME/water (5:1 , 6 ml). The mixture was briefly degassed with Ar for -0.5 minute, capped and stirred at 100 0C overnight. After cooling, the reaction mixture was diluted with EtOAc and brine. Organic layer was isolated, and dried (MgSO4). After concentration, the residue was purified on silica gel. Elution with MeOH/EtOAc (0-10%) gave the desired product 3BK (332 mg).
Step 3: Preparation of 4-(4-(1 ,5-dimethyl-1 H-1,2,4-triazol-3-yl)phenyl)-1 ,2,3,6- tetrahydropyridine dihydrochloride
Figure imgf000227_0001
The Boc group can be removed by treating compound 3BK with 4N HCI in dioxane at rt for two hours. Removal of solvent under vacuum gave compound 4BK.
Step 4: Preparation of (S)-terf-butyl 2-(3-(3-(6-isopropoxypyridin-3-yl)-1 H- indazol-5-ylcarbamoyl)-3-methoxypyrrolidin-1-yl)acetate
Figure imgf000227_0002
In a solution of compound 5BK (3.5 g, 6.6 mmol) (prepared according to a procedure in the synthesis of Sch-1499895) in acetonitrile (26 ml) was added DIEA (5.7 ml, 32.9 mmol). It was cooled to 0 0C and 0.47 ml (3.29 mmol) of te/t-butyl chloroacetate was added dropwise. After stirring for 4hr at 0 0C, 0.47 ml (3.29 mmol) of tert-butyl chloroacetate was added again. It was stirred further for 1 hr at 0 0C and then warmed up to r.t. After stirring overnight at room temperature, it was dissolved in EtOAc (200 ml) and washed with NaHCO3 (1 x 50ml), water (1x 50ml) and brine (1 x 50ml). The organic extracts were dried over MgSO4 and the solvent was removed. The crude was purified by column chromatography using 20%MeOH/EtOAC to get the desired product 6BK (2.4 g).
Figure imgf000228_0001
Step 5: Preparation of (S)-2-(3-(3-(6-isopropoxypyridin-3-yl)-1 H-indazol-5- ylcarbamoyl)-3-methoxypyrrolidin-1-yl) acetic acid
Figure imgf000228_0002
Compound 6BK (2.4 g) was treated with 40ml of TFA for 45 minutes at room temperature. TFA was removed under reduced pressure and the solid was washed with ether to get the compound 7BK as a TFA salt (4.4 g, 95%). Step 6: Preparation of (S)-1 -(2-(4-(4-(1 , 5-dimethyl-1 H-1,2,4-triazol-3-yl)phenyl)-
5,6-dihydropyridin-1(2H)-yl)-2-oxoethyl)-/V-(3-(6-isopropoxypyridin-3-yl)-1W- indazol-δ-yO-S-methoxypyrrolidine-S-carboxamide
Figure imgf000229_0001
To a mixture of compound 7BK (0.12 mmol), HATU (46 mg, 0.12 mmol) in DMF (2 ml) was added compound 4BK (39 mg, 0.12 mmol) and DIEA (0.063 ml). The mixture was stirred for 20 minutes and directly purified by HPLC to give compound A14. Mass spectrum: LCMS M+1= 690, retention time= 3.25 minutes;
1H NMR of A14 HCI salt (400 MHz, DMSO-d6):.δ10.5 (br, 1 H), 10.23 (d, J = 17.6 Hz, 1 H), 8.7 (m, 1 H), 8.45 (d, J = 1.2 Hz, 1 H), 8.18 (m, 1 H)1 7.94 (m, 2H)1 7.75 (dd, J = 8.8, 2.0 Hz, 1 H)1 7.56 (m, 3H), 6.93 (m, 1 H)1 6.3 (m, 1 H), 5.3 (m, 1 H), 4.6-4.53 (m, 2H)1 4.2-4.0 (m, 3H)1 3.82 (s, 3H)1 3.7-3.5 (m, 5H), 3.3 (s, 3H), 2.67-2.54 (m, 3H), 2.44 (s, 3H), 2.4(m, 1 H)1 1.33 (d, J = 6.4 Hz, 6H).
Example 4
Synthesis of 1-r2-f4-(3-Fluoro-4-ri-(2-methoxy-ethvn-1 H-M .2.41triazol-3-vn- phenyl)-3,6-dihvdro-2H-pyridin-1-yl)-2-oxo-ethvπ-3-methoxy-pyrrolidine-3- carboxylic acid r3-(6-isopropoxy-pyridin-3-yl)-1 H-indazol-5-yll-amide
Figure imgf000229_0002
Svnthesis of (S)-1-r2-(4-(3-fluoro-4-ri-(2-methoxy-ethyl)-1 H-\Λ .2,41triazol-3-yll- phenyl}-3,6-dihvdro-2^-pyridin-1-yl)-2-oxo-ethvn-3-methoxy-pyrrolidine-3- carboxylic acid r3-(6-isopropoxy-pyridin-3-yl)-1 H-indazol-S-yli-amide
Step 1 : Preparation of 4-bromo-2-f luoro-benzamide
Figure imgf000230_0001
At 0 0C, 1 ,1'-carbonyldiimidazole (8.8 g, 54.3 mmol) was added in portions to a stirred mixture of 4-bromo-2-fluorobenzoic acid (6 g, 27.3 mmol) in dichloromethane (100 ml). After 20 minutes, a clear solution was obtained. Ammonium hydroxide (28%, 30 ml) was added and the mixture was stirred overnight. Aqueous layer was isolated, extracted with dichloromethane twice. Combined organic extracts were washed with water, 1 N HCI twice, water, brine and dried (MgSO4). Solvent was removed under vacuum, and the solid was washed with hexane to afford 4-bromo-2- fluoro-benzamide 2BL (5.56 g).
Step 2: Preparation of compound 3BL
Figure imgf000230_0002
A mixture of compound 2BL (4.66 g, 21.48 mmol), Et3OPF6 (6.4 g, 25.77 mmol) in dichloroethane (86 ml) was refluxed for 1 hr. Solvent was removed under reduced pressure. The crude was cooled to 0 0C, triturated with ether and filtered to give the desired product 3BL (7.5 g).
Step 3: Preparation of compound 4BL
Figure imgf000230_0003
The compound 3BL (1.96 g) was dissolved in pyridine (10 ml). Hydroxyethylhydrazine (0.51 ml) was added with stirring and the resulting mixture was allowed to stir overnight. The reaction mixture was concentrated under reduced pressure to provide a crude 4BL as yellow gum which was directly used in next step synthesis without further purification.
Step 4: Preparation of compound 5BL
Figure imgf000231_0001
A mixture of the crude 4BL from the previous step in formic acid (30 ml) was refluxed overnight and concentrated under reduced pressure. The residue was treated with saturated NaHCO3, and extracted with EtOAc three times. The combined organics were dried over MgSO4. After concentration, compound residue was purified on silica gel. Elution with EtOAc gave compound 6BL (1.1 g), then 5BL (183 mg).
Compound 6BL can be easily converted to 5BL by aqueous basic hydrolysis.
Step 5: Preparation of compound 7BL
Figure imgf000231_0002
A solution of 5BL (429 mg, 1.5 mmol) in DMF (3 ml) was added with stirring into a flask containing NaH (60%, 66 mg, 1.65 mmol). After stirring for 30 minutes, MeI (0.103 ml, 1.65 mmol) was added slowly. After 30 minutes, the reaction mixture was diluted with EtOAc, washed with water three times, brine and dried (MgSO4). After concentration, the residue was purified on silica gel. Elution with 10% MeOH/EtOAc gave compound 7BL (88 mg).
Step 6: Preparation of compound A16
Figure imgf000232_0001
Compound A16 was prepared from 7BL following procedures similar to those for the synthesis of (S)-1 -(2-(4-(4-(1 ,5-dimethyl-1 H- 1 ,2,4-triazol-3-yl)phenyl)-5,6- dihydropyridin-1 (2/-/)-yl)-2-oxoethyl)-Λ/-(3-(6-isopropoxypyridin-3-yl)-1 H-indazol-5-yl)-3- methoxypyrrolidine-3-carboxamide (A14, Example 3). Mass spectrum: LCMS M+1= 738, retention time= 4 minutes.
1H NMR of A16 HCI salt (400 MHz, DMSO-d6):.δ10.45 (br, 1 H), 10.23 (d, J = 16.8 Hz, 1 H), 8.7 (m, 1 H), 8.6 (m, 1 H), 8.46 (m, 1 H), 8.18 (m, 1 H), 8.0 (m, 1 H), 7.74 (d, J = 8 Hz, 1 H), 7.57 (d, J = 8 Hz, 1 H), 7.42 (m, 1 H), 6.93 (m, 1 H), 6.43 (m, 1 H), 5.3 (m, 1 H), 4.6-4.5 (m, 2H), 4.41 (m, 2H), 4.21 -4.01 (m, 4H), 3.8-3.6 (br, 7H), 3.33 (m, 3H), 3.25 (m, 2H), 2.67-2.54 (m, 3H), 2.4(m, 1 H), 1.33 (d, J = 6.4 Hz, 6H).
Example 5
Synthesis of 1 -(2-{4-r4-(1 -Ethyl-1 H-M ,2,41triazol-3-vn-phenyll-3.6- dihvdro-2H-pyridin-1 -vD-Σ-oxo-ethvD-S-methoxy-pyrrolidine-S-carboxylic acid F3- (6-isopropoxy-pyridin-3-vh-1 H-indazol-5-vll-amide
Figure imgf000233_0001
Synthesis of 4-F4-(1 -ethyl-1 /-H1.2.41triazol-3-yl)-phenvn-1 ,2,3,6-tetrahvdro- pyridine hydrochloride Step 1 : Preparation of 4-bromo-benzimidic acid ethyl ester
Figure imgf000233_0002
4-Bromo-benzonitrile (5g) was suspended in absolute EtOH (100 ml) and cooled to 0-5 ° C. HCI gas was bubbled through, initially vigorously for several minutes and later slowly for 5 hours. The resulting solution was allowed to stir overnight. Most of solvent was removed and the precipitate was filtered, washed with EtOH twice and dried to afford compound 2BM (4.1 g) as white solid.
Step 2: Preparation of Compound 3BM
Figure imgf000233_0003
The 4-bromo-benzimidic acid ethyl ester (1 g) was dissolved in pyridine (20 ml). Ethyl hydrazine (550 mg) was added with stirring and the resulting mixture was allowed to stir overnight. The reaction mixture was concentrated under reduced preessure, and added ether, filtered, washed with ether three times and dried to provide the compound 3BM (1 g).
Step 3: Preparation of 3-(4-bromo-phenyl)-1 -ethyl- 1H-[\, 2, 4]triazole
Figure imgf000234_0001
A mixture of compound 3BM (1 g) in formic acid (10 ml) was refluxed overnight and concentrated. The residue was treated with sat. NaHCO3, and extracted with EtOAc three times. The combined organics were dried over MgSO4. After concentration, compound 4BM was obtained as colorless crystals (0.9 g).
Figure imgf000234_0002
Step 4: Preparation of 4-[4-(1-ethyl-7W-[1,2,4]triazol-3-yl)-phenyl]-3,6-dihydro-2W- pyridine-1-carboxylic acid ferf-butyl ester
Figure imgf000234_0003
To a large pressure flask were charged compound 4BM (400 mg), 4-(4,4,5,5- tetramethy|-[1 ,3,2]dioxaborolan-2-yl)-3,6-dihydro-2H-pyridine-1-carboxylic acid tert- butyl ester (540 mg), Pd(PPh3)4 (180 mg), Na2CO3 2N (3 ml) and Dioxane/EtOH/water (7:3:2, 10 ml). The mixture was briefly degassed with Ar for -0.5 minute, capped and microwaved at 12O0C for 20 mins. After cooling, the reaction mixture was diluted with EtOAc and brine. Organic layer was isolated, and dried (MgSO4). After concentration, the residue was purified on silica gel. Elution with MeOH/EtOAc (0- 10%) gave the desired product 5BM (310 mg). Step 5: Preparation of 4-[4-(1 -ethyl-1 H-[1 ,2,4]triazol-3-yl)-phenyl]-1 ,2,3,6- tetrahydro-pyridine hydrochloride
Figure imgf000235_0001
The Boc group can be removed by treating compound 5BWI with 4N HCI in dioxane at rt for two hours. Removal of solvent under vacuum gave compound 6BWI.
Synthesis of 1-(2-l4-r4-(1-Ethyl-1 H-H .2,41triazol-3-yl)-phenyll-3,6-dihvdro-2H- Pyridin-1-yl)-2-oxo-ethyl)-3-methoxy-pyrrolidine-3-carboxylic acid F3-(6- isopropoxy-pyridin-3-yl)-1 H-indazol-5-yl1-amide
Figure imgf000235_0002
The crude 8BWI (5.9 mmol) was stirred in a mixture of dichloromethane/MeOH
(1 :1 , 20 ml), Oxo-acetic acid ethyl ester (10 ml, 50%) and NaBH(OAc)3 (10 ml) at r.t. overnight. The mixture was quenched with saturared aqueous sodium bicarbonate.
Solvents were removed in vacuum. Dilute with water and ethyl acetate. Layers were separated and the separated aqueous layer was extracted with ethyl acetate (X3).
The combined organic layers were dried (MgSO4), filtered and solvents were removed in vacuum. Column purification gave 8BM as yellow oil.
Figure imgf000235_0003
The crude 9BM (2.35 g) was stirred in a solution of LiOH (1 M, 10 ml) and THF
(10 ml) at r.t. overnight. The mixture was adjusted to pH 3. Solvents were removed in vacuum. The product was used for next step without purification.
Figure imgf000236_0001
The crude 10BM (49 mg), 4-[4-(1 -Ethyl-1 H-[1 ,2,4]triazol-3-yl)-phenyl]-1 , 2,3,6- tetrahydro-pyridine (25.4 mg), HATU (45 mg) ang triethyl amine (0.1 ml) was stirred in DMF (1 ml at r.t. overnight. The mixture was purified by HPLC to give A3 as yellow oil. Mass spectrum: LCMS M+1 = 690, retention time= 3.47 minutes
Example 6
Figure imgf000236_0002
Svnthesis of 4-F4-(1 -isopropyl-1 H-\Λ ,2,4ltriazol-3-vD-phenyll-1 ,2,3,6-tetrahydro- pyridine hydrochloride
Step 1 : Preparation of 4-bromo-benzimidic acid ethyl ester
Figure imgf000237_0001
4-Bromo-benzonitrile (5g) was suspended in absolute EtOH (100 ml) and cooled to 0-5 ° C. HCI gas was bubbled through, initially vigorously for several minutes and later slowly for 5 hours. The resulting solution was allowed to stir overnight. Most of solvent was removed and the precipitate was filtered, washed with EtOH twice and dried to afford compound 2BN (4.1 g) as white solid. (Note: large scale synthesis may take longer time to get reaction complete. It is better to monitor the disappearance of starting material to get reaction end point.)
Step 2: Preparation of Compound 3BN
Figure imgf000237_0002
The 4-bromo-benzimidic acid ethyl ester (1 g) was dissolved in pyridine (20 ml).
Isopropylhydrazine (550 mg) was added with stirring and the resulting mixture was allowed to stir overnight. The reaction mixture was concentrated under reduced preessure, and added ether, filtered, washed with ether three times and dried to provide the compound 3BN (0.9 g).
Step 3: Preparation of 3-(4-bromo-phenyl)-1-isopropyl-7H-[1, 2, 4]triazole
Figure imgf000237_0003
A mixture of compound 3BN (1 g) in formic acid (10 ml) was refluxed overnight and concentrated. The residue was treated with sat. NaHCO3, and extracted with EtOAc three times. The combined organics were dried over MgSO4. After concentration, compound 4BN was obtained as colorless crystals (0.9 g).
Step 4: Preparation of 4-[4-(1-isopropyl-7H-[1,2,4]triazol-3-yl)-pheny|]-3,6- dihydro-2H-pyridine-1-carboxylic acid terf-butyl ester
Figure imgf000238_0001
To a pressure flask were charged compound 4BN (500 mg), 4-(4,4,5,5- tetramethyl-[1 ,3,2]dioxaborolan-2-yl)-3,6-dihydro-2/-/-pyridine-1 -carboxylic acid tert- butyl ester (583 mg), Pd(PPh3)4 (112 mg), Na2CO32N (3 ml) and Dioxane (10 ml). The mixture was briefly degassed with Ar for -0.5 minute, capped and stirred at 100C overnight. After cooling, the reaction mixture was diluted with EtOAc and brine. Organic layer was isolated, and dried (MgSO4). After concentration, the residue was purified on silica gel. Elution with MeOH/EtOAc (0-10%) gave the desired product 5BN (410 mg).
Step 5: Preparation of 4-[4-(1 -isopropy 1-1 W-[1, 2,4]triazol-3-yl)-phenyl]-1, 2,3,6- tetrahydro-pyridine hydrochloride
Figure imgf000238_0002
The Boc group can be removed by treating compound 5BN with 4N HCI in dioxane at rt for two hours. Removal of solvent under vacuum gave compound 6BN. Svnthesis of 1-(2-f4-r4-(1-lsopropyl-1 H-M ,2,41triazol-3-yl)-phenvπ-3.6-dihvdro-2H- pyridin-1 -yl)-2-oxo-ethvπ-3-methoxy-pyrrolidine-3-carboxylic acid T3-(6- isopropoxy-pyr id i n-3-yl)-1 H-i ndazol-5-yll-am ide
Figure imgf000239_0001
The crude 8BN (5.9 mmol) was stirred in a mixture of dichloromethane/MeOH (1 :1 , 20 ml), Oxo-acetic acid ethyl ester (10 ml, 50%) and NaBH(OAc)3 (10 ml) at r.t. overnight. The mixture was quenched with saturared aqueous sodium bicarbonate. Solvents were removed in vacuum. Dilute with water and ethyl acetate. Layers were separated and the separated aqueous layer was extracted with ethyl acetate (X3). The combined organic layers were dried (MgSO4), filtered and solvents were removed in vacuum. Column purification gave 9BN as yellow oil in 65% yield.
Figure imgf000239_0002
The crude 9 BN (2.35 g) was stirred in a solution of LiOH (1 M, 10 ml) and THF (10 ml) at r.t. overnight. The mixture was adjusted to pH 3. Solvents were removed in vacuum. The product was used for next step without purification, yield was quantitative.
Figure imgf000240_0001
The crude 10BN (60 mg), 4-[4-(1 -lsopropyl-1 H-[1 ,2,4]triazol-3-yl)-phenyl]- 1 ,2,3,6-tetrahydro-pyridine (35 mg), HATU (52 mg) ang triethyl amine (0.1 ml) was stirred in DMF (1 ml at r.t. overnight. The mixture was purified by HPLC to gave A5 as yellow oil. Mass spectrum: LCMS M+1= 704, retention time= 3.56 minutes
Example 7
Synthesis of 1-(2-f4-r4-(1-lsopropyl-1 H-M .2.41triazol-3-yl)-phenyll-3.6-dihydro-2H- pyridin-1-yl}-2-oxo-ethyl)-3-methoxy-pyrrolidine-3-carboxylic acid T3-(6- isopropoxy-pyridin-3-yl)-1 H-indazol-5-yll-amide
Figure imgf000240_0002
Svnthesis of 4-rø-d -isopropyl-1 H-\Λ ,2,41triazol-3-yl)-phenyll-1.2,3,6-tetrahvdro- pyridine hydrochloride
Step 1 : Preparation of 4-bromo-benzimidic acid ethyl ester
Figure imgf000241_0001
4-Bromo-benzonitrile (5g) was suspended in absolute EtOH (100 ml) and cooled to 0-5 ° C. HCI gas was bubbled through, initially vigorously for several minutes and later slowly for 5 hours. The resulting solution was allowed to stir overnight. Most of solvent was removed and the precipitate was filtered, washed with EtOH twice and dried to afford compound 2BO (4.1 g) as white solid.
Step 2: Preparation of Compound 3BO
Figure imgf000241_0002
The 4-bromo-benzimidic acid ethyl ester (1 g) was dissolved in pyridine (20 ml). Isopropylhydrazine (550 mg) was added with stirring and the resulting mixture was allowed to stir overnight. The reaction mixture was concentrated under reduced preessure, and added ether, filtered, washed with ether three times and dried to provide the compound 3BO (0.9 g).
Step 3: Preparation of 3-(4-bromo-phenyl)-1 -isopropyl- 7 W-[1 , 2, 4]triazole:
Figure imgf000241_0003
A mixture of compound 3BO (1 g) in formic acid (10 ml) was refluxed overnight and concentrated. The residue was treated with sat. NaHCO3, and extracted with EtOAc three times. The combined organics were dried over MgSO4. After concentration, compound 4BO was obtained as colorless crystals (0.9 g). (Note: it was found that the reaction can be done in just two hours. In large scale synthesis, use 10% NaOH to replace NaHCO3).
Step 4: Preparation of 4-[4-(1-isopropyl-7W-[1 ,2,4]triazol-3-yl)-phenyl]-3,6- dihydro-2H-pyridine-1-carboxylic acid tert-butyl ester
Figure imgf000242_0001
To a large pressure flask were charged compound 4BO (500 mg), 4-(4,4,5,5- tetramethyl-[1 ,3,2]dioxaborolan-2-yl)-3,6-dihydro-2/-/-pyridine-1 -carboxylic acid tert- butyl ester (583 mg), Pd(PPh3)4 (112 mg), Na2CO3 2N (3 ml) and Dioxane (10 ml). The mixture was briefly degassed with Ar for -0.5 minute, capped and stirred at 100C overnight. After cooling, the reaction mixture was diluted with EtOAc and brine. Organic layer was isolated, and dried (MgSO4). After concentration, the residue was purified on silica gel. Elution with MeOH/EtOAc (0-10%) gave the desired product 5BO (410 mg).
Step 5: Preparation of 4-[4-(1 -isopropyl-1 H-[Λ ,2,4]triazol-3-yl)-phenyl]-1 ,2,3,6- tetrahydro-pyridine hydrochloride
Figure imgf000242_0002
The Boc group can be removed by treating compound 5BO with 4N HCI in dioxane at rt for two hours. Removal of solvent under vacuum gave compound 6BO. Preparation and Chiral resolution of of S-Methylsulfanyl-pyrrolidine-S-carboxylic acid methyl ester 4BP
Figure imgf000243_0001
Pyrrolidine-1 ,3-dicarboxylic acid 1 -tert-butyl ester 1 BP (4.3 gm, 20 mmol) was dissolved in 28 mL of toluene and 3.5 ml of methanol. Trimethylsilyldiazomethane 2N solution in hexanes (13 ml, 26 mmol) was added drop wise at 0 C and the reaction mixture stirred for 10 min at ambient temperature. The mixture was evaporated to obtain 4.3 gm of oil. To the oil 2BP (0.5 gm, 2.1 mmol) dissolved in tetrahydrofuran (15 ml) 1.2 ml of lithium diisopropylamide 2N solution in hexanes was added drop wise and the reaction mixture stirred for 1 hr at -78 C. Dimethyldisulfide (0.48 mL, 5.4 mmol) was added slowly and let warm to ambient temperature gradually. The reaction mixture was stirred for 18 hrs. A saturated solution of Ammonium chloride (25 ml) was added and the reaction mixture stirred for 5 min. The reaction mixture was extracted with ethyl acetate three times (3x25 ml), dried over magnesium sulfate, filtered and evaporated to give 0.386g of title product 3BP after column chromatography.
3-Methylsulfanyl-pyrrolidine-1 ,3-dicarboxylic acid 1 -tert-butyl ester 3-methyl ester (2.15 gm, 8.8 mmol) was dissolved in 20 ml of 50% trifluoroacetic acid/dichloromethane and stirred for 2 hrs. The reaction mixture was evaporated to give 3.35 g of 3-Methylsulfanyl-pyrrolidine-3-carboxylic acid methyl ester 4BP as a gummy solid. Alternativelv 4BP can be prepared as follows:
Figure imgf000244_0001
2-Methylsulfanyl-propiorιic acid methyl ester 5BP (25 g, 0.1 mol) dissolved in chloroform was added sulfuryl chloride (15.1 ml_, 0.1 mol) slowly at 0 0C. The reaction mixture was stirred at 00C for 30 min and then refluxed at 65 0C for 30 min. The reaction mixture then concentrated to dryness to give 23.75 g of 2-Methylsulfanyl- acrylic acid methyl ester 6BP as a liquid.
To a stirred solution of 2-ethoxyacrylate 2-Methylsulfanyl-acrylic acid methyl ester 6BP (136 g, 1.03 mol) and benzyl-methoxymethyl-trimethylsilanylmethyl-amine 7BP (290 g, 1.22 mol) in dichloromethane (2.7 L ml) was added at 0 0C a solution of trilϊuroacetic acid (26 ml_, 0.3 mol). The resulting solution was warmed to room temperature and stirred for one overnight. The crude product was purified by column chromatography on silica gel eluting with a solution of ethyl acetate in hexane (1 :4) to give i -benzyl-S-methylsulfanyl-pyrrolidine-S-carboxylic acid methyl ester 8BP (131 g, 47%).
To 1 -benzyl-S-methylsulfanyl-pyrrolidine-S-carboxylic acid methyl ester 8BP (131 g, 493.7 mmol) dissolved in dichloroethane (2.6 L) at 0 0C was added N, N, N1, N1- Tetramethyl-naphthalene-1 ,8-diamine 9BP (31.8 g, 0.144 mmol) and then 2-Chloro- propionyl chloride 10BP (64 ml_, 593.1 mmol) sequentially. The reaction mixture was stirred for one overnight at ambient temperature and then concentrated to dryness. The residue was dissolved in 2.8 L of methanol and refluxed at 65 0C for 3.5 h. The reaction mixture was then concentrated to dryness and the residue was purified by column chromatography on silica gel eluting with a solution of methanol in dichloromethane (1 :9) to give 3-Methylsulfanyl-pyrrolidine-3-carboxylic acid methyl ester 4BP (75 g, 86%).
Chiral resolution of S-Methylsulfanyl-pyrrolidine-S-carboxylic acid methyl ester 4BP
Figure imgf000245_0001
S-Methylsulfanyl-pyrrolidine-S-carboxylic acid methyl ester 4BP (42.9g, 244.8 mmol) and L-tartaric acid (36.7 g, 244.8 mmol) were placed in a 1 L round bottomed flask and dissolved with 250 ml_ of methanol. The flask was then attached to a rotavapor at 75 0C. The mixture was allowed to gently spin at this temperature for about 20 min. to ensure complete dissolution. After the formation of a clear solution, about 10mg of authentic crystals of 3-Methylsulfanyl-pyrrolidine-3-carboxylic acid methyl ester 11 BP were added (to seed and aid the crystal formation) was allowed to settle gently for crystal formation. After 3 days, 19.4g of crystals were filtered which was then washed with cold methanol (20 -30 ml_) to give 18.2g of crystalline 3- Methylsulfanyl-pyrrolidine-3-carboxylic acid methyl ester 11 BP.
The chiral purity of the crystals 3-Methylsulfanyl-pyrrolidine-3-carboxylic acid methyl ester 11 BP was determined by derivatizing with 4-nitrobenzyl chloroformate and subjecting it to analytical HPLC (Chiracel AD column) under the conditions of 20% isopropanol/hexane solvent system with a flow rate of 1 mL/min. The purity was found to be >99.9% with a retention time of 16.58 min.
Preparation of 3-Methvlsulfanvl-Dvrrolidine-1 ,3-dicarboxvlic acid 1-tert-butvl ester
Figure imgf000246_0001
To a cold (O0C) solution of 12BP (28 g, 90.52 mmol) in dry CH2CI2 (250 mL) was added triethylamine (31.5 mL, 226.32 mmol, 2.5 equiv) followed by (BoC)2O (25.7 g, 117.68 mmol, 1.3 equiv). The resulting mixture was stirred from O0C to rt for overnight then diluted with CH2CI2, which was washed with saturated aqueous
NaHCO3 solution and brine, dried (MgSO4) and concentrated. Chromatograph on silica gel (hexanes/ethyl acetate, 4:1 ) gave 13BP (23.5 mg, 90.52 mmol, 100%) as a colorless oil.
To a stirred solution of 13BP (23.5 mg, 90.52 mmol) in THF/MeOH (175 mL/175 mL) was added 135 mL of LiOH (1 M in H2O, 135 mmol, 1.5 equiv). The reaction mixture was stirred at rt for overnight, to which 135 mL of 1 N HCI was added. The resulting mixture was stirred for additional 15 min and concentrated to give 14BP.
Step 1 :
Figure imgf000246_0002
Aminoindazole 5Bl (39g, 0.076mol) and pyrrolidinecarboxylic acid 14BP (32g, 0.069mol) were dissolved in DMF (300ml) at r.t. HATU (29g, 0.076mol) followed by 1Pr2NEt (14.5ml, 0.083mol) were added. The mixture was stirred at r.t. overnight and was diluted with ethyl acetate and water. Layers were separated. The separated organic layer was washed with water (X2), dried (MgSO4) and filtered. Concentration in vacuum followed by column purification [hexanes - ethyl acetate = 4:1 (v/v)] gave crude 7BO as off-white foam.
Step 2:
Figure imgf000247_0001
The crude 7BO was stirred in a mixture of dichloromethane (300ml), trifluoroacetic acid (100ml) and water (50ml) at r.t. overnight. The mixture was cooled at 0 0C and quenched carefully with saturared aqueous sodium bicarbonate. Solvents were removed in vacuum. Dilute with water and ethyl acetate. Layers were separated and the separated aqueous layer was extracted with ethyl acetate (X3). The combined organic layers were dried (MgSO4), filtered and solvents were removed in vacuum. Column purification [5 to 10% MeOH (7N ammonia) in dichloromethane] gave pyrrolidine 8BO (23g, 84%) as off-white solid.
Synthesis of 1 -(2-(4-F4-(1 -lsopropyl-1 H-M .2.41triazol-3-yl)-phenvn-3.6-dihvdro-2H- Pyridin-i-vD-Σ-oxo-ethvD-S-methylsulfanyl-pyrrolidine-S-carboxylic acid T3-(6- isopropoxy-pyridin-3-vO-1H-indazol-5-vπ-amide
Figure imgf000247_0002
The crude 8BO (5.9 mmol) was stirred in a mixture of dichloromethane/MeOH (1 :1 , 20 ml), Oxo-acetic acid ethyl ester (10 ml, 50%) and NaBH(OAc)3 (10 ml) at r.t. overnight. The mixture was quenched with saturared aqueous sodium bicarbonate. Solvents were removed in vacuum. Dilute with water and ethyl acetate. Layers were separated and the separated aqueous layer was extracted with ethyl acetate (X3). The combined organic layers were dried (MgSO4), filtered and solvents were removed in vacuum. Column purification gave 9BO as yellow oil.
Figure imgf000248_0001
The crude 9BO (2.35 g) was stirred in a solution of LiOH (1 M, 10 ml) and THF (10 ml) at r.t. overnight. The mixture was adjusted to pH 3. Solvents were removed in vacuum. The product was used for next step without purification.
Figure imgf000248_0002
The crude 10BO (60 mg), 4-[4-(1 -lsopropyl-1 H-[1 ,2,4]triazol-3-yl)-phenyl]- 1 ,2,3,6-tetrahydro-pyridine (35 mg), HATU (52 mg) ang triethyl amine (0.1 ml) was stirred in DMF (1 ml at r.t. overnight. The mixture was purified by HPLC to gave 11 BO as yellow oil. Mass spectrum: LCMS M+1 = 720, retention time= 3.68 minutes.
Example 8
Preparation of 1-r2-(4-{4-ri-(2-Hvdroxy-2-methyl-propyl)-1 H-M ,2,41triazol-3-vπ- phenvU-3.6-dihvdro-2H-pyridin-1-yl)-2-oxo-ethvn-3-methoxy-pyrrolidine-3- carboxylic acid r3-(6-isopropoxy-pyridin-3-yl)-1 H-indazol-5-yll-amide
Figure imgf000249_0001
Synthesis of 2-chloro-1-f4-r4-(1-ethyl-1 H-\λ ,2,41triazol-3-yl)-phenvπ-3,6-dihvdro- 2Af-pyridin-1 -yll-ethanone
Step 1 : Preparation of 4-bromo-benzimidic acid ethyl ester
Figure imgf000249_0002
4-Bromo-benzonitrile (5g) was suspended in absolute EtOH (100 ml) and cooled to 0-5 ° C. HCI gas was bubbled through, initially vigorously for several minutes and later slowly for 5 hours. The resulting solution was allowed to stir overnight. Most of solvent was removed and the precipitate was filtered, washed with EtOH twice and dried to afford compound 2BO (4.1 g) as white solid. Step 2: Preparation of Compound 3BQ:
Figure imgf000250_0001
The 4-bromo-benzimidic acid ethyl ester (1.68 g) was dissolved in pyridine (10 ml). 1 -Hydrazino-2-methyl-propan-2-ol (1 g) was added with stirring and the resulting mixture was allowed to stir overnight. The reaction mixture was concentrated under reduced pressure, and added ether, filtered, washed with ether three times and dried to provide the compound 3BQ (1 g).
Step 3: Preparation of 1-[3-(4-Bromo-phenyl)-[1,2,4]triazol-1-yl]-2-methyl-propan- 2-ol:
Figure imgf000250_0002
A mixture of compound 3BQ (1 g) in formic acid (10 ml) was refluxed overnight and concentrated. The residue was treated with sat. NaHCθ3, and extracted with EtOAc three times. The combined organics were dried over MgSO4. After concentration, compound 4BQ was obtained as colorless crystals (0.9 g).
Step 4: Preparation of 4-{4-[1-(2-Hydroxy-2-methyl-propyl)-1 H-[1 ,2,4]triazol-3-yl]- phenyl}-3,6-dihydro-2H-pyridine-1-carboxylic acid tert-butyl ester
Figure imgf000250_0003
To a large pressure flask were charged compound 4BQ (400 mg), 4-(4,4,5,5- tetramethyl-[1 ,3,2]dioxaborolan-2-yl)-3,6-dihydro-2H-pyridine-1-carboxylic acid tert- butyl ester (540 mg), Pd(PPh3)4 (180 mg), Na2CO3 2N (3 ml) and Dioxane/EtOH/water (7:3:2, 10 ml). The mixture was briefly degassed with Ar for -0.5 minute, capped and microwaved at 120C for 20 mins. After cooling, the reaction mixture was diluted with EtOAc and brine. Organic layer was isolated, and dried (MgSO4). After concentration, the residue was purified on silica gel. Elution with MeOH/EtOAc (0- 10%) gave the desired product 5BQ (310 mg).
Step 5: Preparation of 2-Methyl-1-{3-[4-(1 ,2,3,6-tetrahydro-pyridin-4-yl)-phenyl]- [1 ,2,4]triazol-1 -yl}-propan-2-ol
Figure imgf000251_0001
The Boc group can be removed by treating compound 5BQ with 4N HCI in dioxane at rt for two hours. Removal of solvent under vacuum gave compound 6BQ.
Synthesis of 1-r2-(4-{4-π-(2-Hvdroxy-2-methyl-ρropylV1 H-M ,2.41triazol-3-vn- phenyl)-3,6-dihvdro-2H-pyridin-1-yl)-2-oxo-ethyll-3-methoxy-pyrrolidine-3- carboxylic acid r3-(6-isopropoxy-pyridin-3-yl)-1 H-indazol-5-vn-amide
Figure imgf000251_0002
The crude 8BN (5.9 mmol) was stirred in a mixture of dichloromethane/MeOH (1 :1 , 20 ml), Oxo-acetic acid ethyl ester (10 ml, 50%) and NaBH(OAc)3 (10 ml) at r.t. overnight. The mixture was quenched with saturared aqueous sodium bicarbonate. Solvents were removed in vacuum. Dilute with water and ethyl acetate. Layers were separated and the separated aqueous layer was extracted with ethyl acetate (X3). The combined organic layers were dried (MgSO4), filtered and solvents were removed in vacuum. Column purification gave 9BN as yellow oil.
Figure imgf000252_0001
The crude 9BN (2.35 g) was stirred in a solution of LiOH (1 M, 10 ml) and THF (10 ml) at r.t. overnight. The mixture was adjusted to pH 3. Solvents were removed in vacuum. The product was used for next step without purification.
Figure imgf000252_0002
The crude 10BN (60 mg), 2-Methyl-1 -{3-[4-(1 ,2,3,6-tetrahydro-pyridin-4-yl)- pheny|]-[1 ,2,4]triazol-1 -yl}-propan-2-ol (38 mg), HATU (50 mg) ang triethyl amine (0.1 ml) was stirred in DMF (1 ml at r.t. overnight. The mixture was purified by HPLC to gave A9 as yellow oil, 50%. Mass spectrum: LCMS M+1= 734, retention time= 3.32 minutes. Example 9
Preparation of 3-Methoxy-1-(2-f4-r4-(1-methyl-1 H-F1.2,41triazol-3-yl)-phenvn-3.6- dihvdro-2H-pyridin-1-yl>-2-oxo-ethvπ-pyrrolicline-3-carboxylic acid (3-r6-(2- methoxy-ethoxy)-pyridin-3-yl1-1 H-indazol-5-yl)-arnide
Figure imgf000253_0001
Synthesis of S-Methoxy-pyrrolidine-S-carboxylic acid f3-r6-(2-methoxy-ethoxy)- pyridin-3-yll-1 H-indazol-5-yl)-amide
Step 1 :
Figure imgf000253_0002
To a pressure flask were charged compound 1 BR (1.75 g), 2BR (0.5 g), Pd(PPh3)4 (210 mg), Na2CO3 2N (10 ml) and Dioxane (10 ml). The mixture was briefly degassed with Ar for -0.5 minute, capped and stirred at 1000C overnight. After cooling, the reaction mixture was diluted with EtOAc and brine. Organic layer was isolated, and dried (MgSO4). After concentration, the residue was purified on silica gel. Elution gave the desired product 3BR (0.8 g). Step 2:
Figure imgf000254_0001
A mixture of compound 3BR (1.8 g), 2-Methoxy-ethanol (1 g), DEAD (0.8 g) and PPh3 (1.2 g) in THF (10 ml) was stirred overnight at rt and concentrated. The residue purified by silica gel to gave the desired product 4BR (0.8 g).
Step 3:
Figure imgf000254_0002
4BR (0.5 g) in MeOH (20 ml) was reduced by H-cube with Pd/C (10%) column.
Step 4:
Figure imgf000254_0003
Aminoindazole 5BR (80 mg) and pyrrolidinecarboxylic acid 14BH (38 mg) were dissolved in DMF (1 ml) at r.t. HATU (69 mg) followed by 1Pr2NEt (0.1 ml) were added. The mixture was stirred at r.t. overnight and was diluted with ethyl acetate and water. Layers were separated. The separated organic layer was washed with water (X2), dried (MgSO4) and filtered. Concentration in vacuum gave crude 7BR as off-white foam.
Step 5:
Figure imgf000255_0001
The crude 7BR was stirred in a mixture of dichloromethane (1 ml), trifluoroacetic acid (1 ml) at r.t. for 1 h. The mixture was cooled at 0 0C and quenched carefully with saturared aqueous sodium bicarbonate. Solvents were removed in vacuum. Dilute with water and ethyl acetate. Layers were separated and the separated aqueous layer was extracted with ethyl acetate (X3). The combined organic layers were dried (MgSO4), filtered and solvents were removed in vacuum and gave pyrrolidine 8BR as off-white solid.
Svnthesis of 3-Methoxy-1-(2-(4-r4-(1-methyl-1 H-f1 ,2,41triazol-3-vn-phenyll-3.6- dihvdro-ΣH-pyridin-i-vD-Σ-oxo-ethvD-pyrrolidine-a-carboxylic acid f3-F6-(2- methoxy-ethoxy)-pyridin-3-yll-1H-indazol-5-yl>-amide
Figure imgf000256_0001
Figure imgf000256_0002
A mixture of compound 7BH (52 mg), compound 8BR (62 mg), and DIEA (0.3 ml) in DMF (1.5 ml) was stirred at room temperature overnight. LCMS shows the reaction is complete. DMF was removed under reduced pressure. The crude was purified by HPLC. (LCMS M+1= 692, ret. time= 3.00 min.) 1 H NMR ( 400 MHz, CDCI3):.δ11.92 (S, 1 H), 11.11 (S, 1 H), 9.14 (S, 1 H), 8.58 (S, 1 H), 8.03 (S, 1 H), 7.99 (d, 1 H, J= 8.4 Hz), 7.90 (t, 2H, J= 6.8), 7.39 (S, 2H), 7.20 (S, 3H), 6.76 (d, 1 H, J=8.0 Hz), 5.85 (d, 1 H, J= 20 Hz), 4.4 (S, 4H), 4.05 (q, 4H, J= 7.2 Hz), 3.8 (S, 3H), 3.50-3.56 (m, 6H), 2.34-2.62 (m, 5H), 2.0 (S, 2H), 1.64-1.49 (m, 3H). Example 10
Preparation of 1-r2-(4-(4-ri-(2-Hvdroxy-ethyl)-1 H-M ,2,41triazol-3-vn-phenyli-3.6- dihvdro-ΣH-pyridin-i-vD-Σ-oxo-ethvn-S-methoxy-pyrrolidine-S-carboxylic acid \3-
(6-isopropoxy-pyridin-3-yl)-1 H-indazol-5-vπ-amide
Figure imgf000257_0001
Preparation of f3-r3-(6-lsopropoxy-pyridin-3-vD-1 Wnxazol-5-ylcarbamoyll-3- methoxy-pyrrolidin-1 -yl)-acetic acid
Figure imgf000257_0002
In a solution of compound 8Bl (3.5g, 6.6mmol) in acetonitrile (26ml) was added DIEA (5.7ml, 32.9mmol). It was cooled to 0 0C and 0.47ml (3.29mmol) of compound 13BS was added dropwise. After stirring for 4hr at 0 0C, 0.47ml (3.29mmol) of compound 13BS was added again. It was stirred further for 1 hr at 0 0C and then warmed up to r.t. After stirring overnight at room temperature, it was dissolved in EtOAc (200ml) and washed with NaHCO3 (1 x 50ml), water (1x 50ml) and brine (1 x 50ml). The organic extracts were dried over MgSO4 and the solvent was removed. The crude was purified by column chromatography using 20%MeOH/EtOAC to get the desired product 14BS (2.4g). Compound 14BS (2.4g) was treated with 40ml of TFA for 45 mins at room temperature. TFA was removed under reduced pressure and the solid was washed with ether to get the desired compound 15BS as a TFA salt (4.4gm, 95%). Compound 15BS was converted into the HCI salt by adding 4N HCI in water.
Synthesis of Preparation of 2-f3-rr4r(1.2.3,6-Tetrahvdro-pyridin-4-yl)-phenyl1-
H ,2,41triazol-1-yl)-ethanol hydrochloride
Figure imgf000258_0001
Preparation of 4-bromo-Λf-(2-hvdroxyethyl)benzimidohvdrazide hydrochloride
The 4-bromo-benzimidic acid ethyl ester 1 BH (5g) was dissolved in pyridine (100ml). Hydroxyethylhydrazine (1.92ml) was added with stirring and the resulting mixture was allowed to stir overnight. The precipitate was collected by filtration and mother liquor was concentrated to almost dryness and ether was added. Solid was collected by filtration. Combined solid was washed with ether twice and dried to afford 18BS (5.1g).
Figure imgf000258_0002
Preparation of 2-(3-(4-bromophenvQ-1 H-Λ ,2,4-trlazol-1-yl)ethanol A mixture of compound 18BS (3g) in formic acid (50ml) was stirred at room temperature for 30 minutes, at 100 0C for 1.5 hours and concentrated. The residue was treated with saturated NaHCO3, and extracted with EtOAc three times. The combined organic extracts were dried over MgSO4. After concentration, the crude was purified on silica gel. Elution with EtOAc gave compound 20BS (2g), and compound 19BS (262mg).
Compound 20BS can be easily converted to 19BS by aqueous hydrolysis
Preparation of 4-r4-(1-(2-Hvdroxy-ethvn-7tf-π ,2,41triazol-3-vn-phenvπ-3.6- dihvdro-2H-pyridine-1-carboxylic acid ferf-butyl ester
Figure imgf000259_0001
To a large pressure flask were charged compound 19BS (1.5 g, 5.6 mmols), A- (4 ,4,5,5-tetramethyl-[1 ,3,2]dioxaborolan-2-yl)-3,6-dihydro-2H-pyridine-1 -carboxylic acid te/t-butyl ester (1.9g, 6.2mmols), [1 ,1 '-bis(diphenylphosphino)- ferrocene]dichloropalladium(ll), complex with dichloromethane(1 :1 ) (0.22g, 0.28 mmols), K2CO3 (2.32g, 16.8mmols) and DME/water (5:1 , 12ml). The mixture was briefly degassed with Ar for -0.5 minute, capped and stirred at 800C overnight. After cooling, the reaction mixture was diluted with EtOAc and washed with water (1X) and brine (1x). Organic layer was isolated, and dried over MgSO4. After concentration, the residue was purified on silica gel. Elution with MeOH/EtOAc (0-10%) gave the desired product 21 BS (1.4g, 73%).
Preparation of 2-{3-[[4[(1 ,2,3,6-Tetrahydro-pyridin-4-yl)-pheny|]-[1 ,2,4]triazol-1- yl}-ethanol hydrochloride
Figure imgf000259_0002
The Boc group can be removed by treating compound 21 BS with 4N HCI in dioxane at rt for two hours. Removal of solvent under vacuum gave compound 22BS. Preparation of 1-r2-(4-f4-π-(2-Hvdroxy-ethylM H-M ,2,41tria2θl-3-vn-phenylV3.β- dihvdro-ΣH-pyridin-i-vD-Σ-oxo-ethvn-S-methoxy-pyrrolidine-S-carboxylic acid T3- (6-isopropoxy-pyrididn-3-yl)-1 H-inxa2θl-5-yll-amide
Figure imgf000260_0001
A mixture of compound 10BN (0.49g, 1 mmol), 1 -(3-Dimethyl-aminopropyl)-3- ethylcarboxiimide hydrochloride (0.38g, 2mmol), and 1-hydroxybenzotriazole (0.14g, 1 mmol) was dissolved in DMF (3ml). After stirring for 20 min at room temperature, compound 16BS (0.34g, 1 mmol) and DIEA (0.7ml, 4mmol) were added. After stirring for overnight at room temperature, it was diluted with DCM (45ml) and washed with NaHCO3 (1 X 7rrιl), water (3 x 7ml) and brine (1X1 OmI). The organic layer was dried over MgSO4. After concentration, the residue was purified on silica gel. Elution with 2% NH3 in 20 %MeOH/EtOAc gave the desired product A2 (0.3g). This compound was converted to HCI salt by adding 4N HCI in 1 ,4-dixoane. (LCMS: M+1 = 706, ret. time = 3.13 min.), 1H NMR (400 MHz, DMSO-d6): δ = 10.5 - 10.8(m, 1 H), 10.25 (d, 1 H, J = 19.2Hz), 8.60 - 8.75 (m, 2H), 8.43 (m, 1 H), 8.18 (m, 1 H), 7.98 (m, 2H), 7.78 (d, 2H, J = 20Hz), 7.5 - 7.6 (m, 4H), 6.9 (m, 2H), 6.3 (m, 1 H), 5.3 (m, 1 H), 4.5 - 4.6 (m, 2H), 4.15 - 4.3 (m, 4H), 4.0 - 4.15 (m, 2H), 3.75 (t, 4H, 5.2 Hz), 3.5 - 3.6 (m, 2H), 3.4 - 3.3 (m, 1 H), 3.29 (s, 2H), 2.5 - 2.7 (m, 3H), 2.3 - 2.4 (m, 1 H), 1.3 (d, 6H, J = 6.4Hz). Example 11
Synthesis of 3-Methoxy-1 -(2-f 4-rø-d -methyl-1 H-F1 ,2,41triazol-3-yl)-phenvπ-3.6- dihvdro-2H-pyridin-1 -yl)-2-oxo-ethyl)-pyrrolidine-3-carboxylic acid r3-(6-ethoxy- pyridin-3-yl)-1 H-indazol-5-yll-amide (Example 1)
Figure imgf000261_0001
Step 1 :
Figure imgf000261_0002
A mixture of 6-ethoxypyridine-3-boronic acid (2.5 g, 14.97 mmol), bromoindazole 3Bl (7.25 g, 14.97 mmol), potassium carbonate (6.2 g, 44.91 mmol), PdCI2(dppf)2.CH2Cl2 (1.22 g, 1.497 mmol), 1 ,4-dioxane (40 ml_) and water (10 rriL), was purged with nitrogen for 15 min at r.t. and then heated at 90 0C for 18 hrs and cooled to r.t. Water (100 ml_) and ethyl acetate (300 ml_) were added. Solids were filtered through Celite. Layers were separated and the separated organic layer was washed with water (100 ml_). The combined organic layers were dried (Na2SO4), filtered and solvents were removed in vacuum. Column purification [Hexanes-ethyl acetate = 9:1 (Wv)] gave Compound 2BU (7 g, 89%). Step 2:
Figure imgf000262_0001
Compound 2BU (2 g, 3.8 mmol) and Pd/C (10%, 50% wet, 0.7 g) were stirred in toluene (30 ml_) and MeOH (15 ml_) under H2 (balloon) at r.t. for 18 hrs. The solid catalyst was filtered through Celite and solvents were removed in vacuum. Column purification using 4%MeOH/CH2CI2 gave Compound 3BU (1.6 g, 85%).
Step 3:
Figure imgf000262_0002
Compound 3BU (1.46 g, 3.22 mmol) and pyrrolidinecarboxylic acid 14BH (0.56 g, 3.22 mmol) were dissolved in DMF (300ml) at r.t. HATU (1.68 g, 4.83 mmol) followed by 'Pr2NEt (0.78 ml_, 4.83 mmol) were added. The mixture was stirred at r.t. overnight and was diluted with ethyl acetate (200 ml_) and water (100 ml_). Layers were separated. The separated organic layer was washed with water (100 ml_), dried (Na2SO4) and filtered. Concentration in vacuum followed by column purification [hexanes - ethyl acetate = 9:1 (v/v)] gave product 4BU. Step 4:
Figure imgf000263_0001
Compound 4BU (1.6 g) was stirred in a mixture of dichloromethane (30 rriL), triiiuoroacetic acid (4 ml_) and few drops of water at r.t. overnight. The mixture was cooled at 0 0C and quenched carefully with 7% MeOH (MH3)ZCH2CI2. Solvents were removed in vacuum. Dilute with water (100 ml_) and ethyl acetate (200 ml_). Layers were separated. The organic layer was dried (Na2SO4), filtered and solvents were removed in vacuum. Column purification [7% MeOH (7N ammonia) in dichloromethane] gave Compound 5BU (0.44 g, 49%) which was converted to mono HCI salt by treating with 4 N HCI/dioxane and evaporating the solution to dryness.
Step 5:
Figure imgf000263_0002
A mixture of Compound 7BH (46 mg, 0.144 mmol), Compound 5BU (60 mg,
0.144 mmol), DMF (2 ml_) and N,N-diisopropylethylamine (0.076 ml_, 0.432 mmol) was stirred at room temperature for 18 hours. Diluted with EtOAc (100 ml_) and washed with water (2 x 100 ml_). The organic layer was dried over Na2SO4, filtered and concentrated. The residue was purified on silica gel eluting with 4% MeOH (MH3)/ CH2CI2 to give the desired product 6BU (40 mg, 42%). LCMass Spec M+1 @ ret. time = 662 @ 2.57 min
Example 12
Synthesis of 3-Methylsulf anyl-1 -(2-(444-(I -methyl-1 H-H .2,41triazol-3-yl)-phenyll-
3.6-dihvdro-2H-pyridin-1 -yll-2-oxo-ethyl)-pyrrolidine-3-carboxylic acid T3-(6- methoxy-pyridin-3-yl)-1 H-indazol-5-vll-amide
Figure imgf000264_0001
Step 1 :
Figure imgf000264_0002
Compound 8BU was prepared from Compound 3Bl using essentially the same procedure as described for the preparation of Compound 2BU from Compound 3Bl (Example 11 , Step 1), using 6-methoxypyridine-3-boronic acid in place of 6- ethoxypyridine-3-boronic acid. Step 2:
Figure imgf000265_0001
Compound 8BU (3 g, 3.8 mmol) and Pd/C (10%, 50% wet, 1.2g) were stirred in toluene (30 ml_) and MeOH (15 ml_) under H2 (balloon) at r.t. for 18 hrs. The solid catalyst was filtered through Celite and solvents were removed in vacuum to give Compound 10BU (2.8 g, 100%).
Step 3:
Figure imgf000265_0002
Compound 10BU (0.6 g, 1.24 mmol) and pyrrolidinecarboxylic acid 14BP (0.33 g, 1.24 mmol) were dissolved in DMF (5 ml_) at r.t. HATU (0.71 g, 1.86 mmol) followed by 'Pr2NEt (00.66 ml_, 3.72 mmol) were added. The mixture was stirred at r.t. overnight and was diluted with ethyl acetate (200 ml_) and water (100 ml_). Layers were separated. The separated organic layer was washed with water (100 ml_), dried (Na2SO4) and filtered. Concentration in vacuum followed by column purification [hexanes - ethyl acetate = 9:1 (v/v)] gave product 12BU (0.65 g, 72%).
Figure imgf000266_0001
Compound 12BU (0.65 g) was stirred in a mixture of dichloromethane (30 ml_), trifluoroacetic acid (4 ml_) and few drops of water at r.t. overnight. The mixture was cooled at 0 0C and quenched carefully with 7% MeOH (NH3)/CH2CI2. Solvents were removed in vacuum. Dilute with water (100 ml_) and ethyl acetate (200 ml_). Layers were separated. The organic layer was dried (Na2SO4), filtered and solvents were removed in vacuum. Column purification [15% MeOH (7N ammonia) in dichloromethane] gave Compound 13BU (0.22 g, 34%).
Step 5:
Figure imgf000266_0002
A mixture of Compound 7BH (40 mg, 0.126 mmol), Compound 13BU (50 mg, 0.126 mmol), DMF (2 ml_) and N,N-diisopropylethylamine (0.045 ml_, 0.25 mmol) was stirred at room temperature for 18 hours. Diluted with EtOAc (100 ml_) and washed with water (2 x 100 ml_). The organic layer was dried over Na2SO4, filtered and concentrated. The residue was purified on silica gel eluting with 3% MeOH (NH3)/ CH2CI2 to give the desired product A19 (50 mg, 60%).
LCMass Spec M+1 @ ret. time = 664 @ 2.88 min Example 13
Synthesis of 3-Methoxy-1 -(2-(4-[4-(1 -methyl-1 H-M ,2.41triazol-3-vn-phenyl1-3.6- dihvdro-2H-pyridin-1-yl)-2-oxo-ethyl)-pyrrolidine-3-carboxylic acid T3-(2- methoxy-pyridin-4-yl)-1 H-indazol-5-vll-amide
Figure imgf000267_0001
Step 1 :
Synthesis of 3-(2-Fluoro-pyridin-4-yl)-5-nitro-1-trityl-1 H-indazole
Figure imgf000267_0002
3-Bromo-5-nitro-1 -trityl-1 H-indazole 3Bl (15.64g, 32.3 mmol), 2-fluoro-4- pyridine boronic acid 2BV (5.Og, 35.5 mmol), K3PO4 (IT1Ig, 80.7 mmol) and
Pd(dppf)CI2 (2.64g, 3.23 mmol) was mixed in dioxane/H2O (240 ml_760 ml_) at rt and heated at 80 0C overnight. The reaction mixture was cooled to rt and concentrated to a small volume. The residue was partitioned between ethyl acetate (200 ml_) and brine (150 ml_). The organic layer was washed with brine, dried (MgSO4) and filtered. The resulting filtrate was concentrated and the residue was purified on silica gel column eluting with hexanes, 5% ethyl acetate in hexanes, 10% ethyl acetate in hexanes sequentially to yield a yellow solid 3BV (3.33g, 64%). Step 2:
Preparation of 3-(2-Methoxy-pyridin-4-yl)-5-nitro-1-trityl-1 H-indazole
Figure imgf000268_0001
In a 250 ml pressure vessel, 3-(2-Fluoro-pyridin-4-yl)-5-nitro-1 -trityl-1 H-indazole 3BV (5.005 g, 10.0 mmole) was dissolved in 0.1 N KOCH3/MeOH/Toluene(Acros, 150 mL, 15.0 mmole) under dry N2 gas. The pressure vessel was tightly sealed and heated under stirring at 70 0C for 24 hours. The pressure vessel was cooled to 0 0C in ice-bath before opening. The contents of the pressure vessel were transferred to 500 ml RBF and evaporated to dryness. The resulting solid was dissolved in CH2CI2 and washed with saturated NaCI solution and dried over MgSO4. The solvent was evaporated to dryness and dried under high vacuum to yield 4BV as a brown solid (5.12 g, 100%).
Step 3:
Preparation of 3-(2-methoxy-pyridin-4-yl)-1-trityl-1 H-indazol-5-ylamine
Figure imgf000268_0002
To the stirred suspension of 3-(2-Methoxy-pyridin-4-yl)-5-nitro-1 -trityl-1 H- indazole 4BV (5.125 g ,10.0 mmole) in MeOH/toluene (50 ml_/50 mL) was added 10% Pd/C (Degussa type, 0.5g) at r.t. under dry N2 gas. The mixture was degassed and was stirred under a balloon inflated with H2 gas overnight. The catalyst was filtered through microfiber filter and was washed with MeOH and CH2CI2. The filtrate was evaporated to dryness and a brown solid 5BV was obtained (5.0 g). Step 4
Preparation 3-Methoxy-3-[3-(2-methoxy-pyridin-4-yl)-1 -trityl-1 H-indazol-5- ylcarbamoyl]-pyrrolidine-1-carboxylic acid tert-butyl ester
Figure imgf000269_0001
Figure imgf000269_0002
To the stirred suspension of 3-methoxy-pyrrolidine-1 ,3-dicarboxylic acid 1 -tert- butyl ester 6BV (4.16 mmole, crude) in DMF/DCM (25 ml_/25 ml_), 3-(2-methoxy- pyridin-4-yl)-1 -trityl-1 H-indazol-5-ylamine 5BV (2.008 g, 4.16 mmole) and Et3N (2.9 ml_,21 mmole) were added at r. t. under dry N2 gas followed by HATU (3.16 g, 8.32 mmole). The mixture was stirred at r.t. under dry N2 gas overnight. The mixture was partitioned between 1 :1 EtOAc and sat. NaHCO3 solution. The organic phase was separated, washed with brine, dried over MgSO4 and evaporated to dryness. The crude solid was purified on RediSep 12Og cartridge eluting with 10-25% EtOAc/Hexanes to yield a brown solid 7BV (2.95 g, 100%). Step δ
Preparation S-Methoxy-pyrrolidine-S-carboxylic acid [3-(2-methoxy-pyridin-4-yl)-
1 H-indazol-5-yl]-amide
Figure imgf000270_0001
To the stirred solution 3-Methoxy-3-[3-(2-methoxy-pyridin-4-yl)-1 -trityl-1 H- indazol-5-ylcarbamoyl]-pyrrolidine-1 -carboxylic acid tert-butyl ester 7BV (2.95 g, 4.16 mmole) in DCM (50 rriL) at r.t. was added triethylsilane (1.00 ml, 6.24 mmole ) followed by TFA (10 ml_). The mixture was stirred at r.t. under dry N2 gas for 4-5 hours. The mixture was evaporated to dryness and co-evaporated with dry toluene (2x75 ml_). The resulting crude solid was purified on RediSep 120 g cartridge eluting with 2.5%-6% 2M NH3-MeOHZCH2CI2 to yield an off-white solid 8BV (1.23 g, 80%).
Step 6 Synthesis of 3-Methoxy-1 -(2-{4-[4-(1 -methyl-1 H-[1 ,2,4]triazol-3-yl)-phenyl]-3,6- dihydro-2H-pyridin-1-yl}-2-oxo-ethyl)-pyrrolidine-3-carboxylic acid [3-(2- methoxy-pyridin-4-yl)-1 H-indazol-5-yl]-amide
Figure imgf000270_0002
Figure imgf000271_0001
A mixture of compound 8BV (40 mg, 0.11 mmol), compound 9BV (41.6 mg, 0.13 rrimol) and tiϊethylamine (0.1 ml_) in DMF (3 ml_) was heated at 45 0C with stirring overnight. The reaction mixture was then concentrated in vacuo and the resulting crude was purified on silica gel column eluting with 2% and 4% 2N NHs/MeOH in CH2CI2 to isolate a yellow solid A21 (39.6 mg, 55%). (LCMS M+1 = 648, ret. time = 2.34 min.) 1H-NMR (400 MHz, CDCI3): δ 10.54 (br, 1 H), 9.51 & 9.44 (s, s, 1 H), 8.46 (dd, 1 H, J=8.2 Hz & 1.4 Hz), 8.27 (d, 1 H, J=5.3 Hz), 8.07 (s, 1 H), 8.05 (m, 2H), 7.76 (m, 1 H), 7.54 (m, 1 H)1 7.44 (m, 3H), 7.37 (br, 1 H), 6.20 & 6.11 (t, t, 1 H, J=2.5 Hz), 4.27 (m, 2H), 4.00 & 3.98 (s, s, 6H), 3.96 & 3.86 (m, m, 1 H), 3.74 (m, 1 H), 3.47-3.60 (m, 2H), 3.453 & 3.446 (s, s, 3H), 3.32 (d, 1 H, 9.9 Hz), 3.02 (m, 1 H), 2.97 (dd, 1 H, J=10.1 Hz & 1.0 Hz), 2.91 (m, 1 H), 2.64 (m, 2H), 2.47 (m, 1 H), 2.19 (m, 1 H).
Example 14 Synthesis of 3-Methylsulfanyl-1-(2-f4-r4-(1-methyl-1 H-M ,2.41triazol-3-yl)-phenvn- S.S-dihvdro-ΣH-pyridin-i-vD-Σ-oxo-ethvD-pyrrolidine-S-carboxylic acid \3-l2- ethoxy-pyridin-4-vl)-1 H-indazol-5-vll-amide
Figure imgf000271_0002
Step i :
Preparation of 3-(2-Ethoxy-pyridin-4-yl)-5-nitro-1-trityl-1 H-indazole
Figure imgf000272_0001
In a 150 mL pressure vessel, to the stirred solution of 3-(2-Fluoro-pyridin-4-yl)- 5-nitro-1 -trityl-1 H-indazole 3BV (2.0 g, 4.0 mmole) in anhydrous EtOH (40 mL), solid potassium tert-butoxide (12 g, 10.0 mmole) was added under dry N2 gas. The pressure vessel was tightly sealed and heated at 80 0C for 24 hours. The pressure vessel was cooled to 0 0C in ice-bath before opening. The contents of the pressure vessel were transferred to 250 mL RBF and concentrated to a small volume. The resulting mixture was partitioned between EtOAc and H2O. The organic phase was separated, washed with saturated NaCI solution and dried over MgSO4. The solvent was evaporated to dryness to give a crude solid 11 BV (1.5 g, 71 %).
Step 2: Preparation of 3-(2-ethoxy-pyridin-4-yl)-1 -trityl-1 H-indazol-5-ylamine
Figure imgf000272_0002
12BV was prepared by using essentially the same procedure as in Example 13, Step 3, except using 11 BV instead of 4BV. Step 3:
Preparation of 3-[3-(2-Ethoxy-pyridin-4-yl)-1-trityl-1 H-indazol-5-ylcarbamoyl]-3- methylsulfanyl-pyrrolidine-1-carboxylic acid tert-butyl ester
Figure imgf000273_0001
Figure imgf000273_0002
14BV, with a yield of 88%, was prepared using essentially the same procedure as in Example 13, Step 4, except using 12BV and 14BP instead of 5BV and 6BV.
Step 4:
Preparation of 3-Methylsulfanyl-pyrrolidine-3-carboxylic acid [3-(2-ethoxy- pyridin-4-yl)-1 H-indazol-5-yl]-amide
Figure imgf000273_0003
15BV, with a yield of 80%, was prepared using essentially the same procedure as in Example 13, Step 5, except using 14BV instead of 7BV. Step 5:
Preparation of 3-Methylsulfanyl-1-(2-{4-[4-(1-methyl-1 H-[1 ,2,4]triazol-3-yl)- phenyll-SjG-dihydro^H-pyridin-i-yl^-oxo-ethyO-pyrrolidine-S-carboxylic acid
[3-(2-ethoxy-pyridin-4-yl)-1 H-indazol-5-yl]-amide
Figure imgf000274_0001
A mixture of compound 15BV (40 mg, 0.10 mmol), compound 7BH (41.6 mg, 0.13 mmol) and triethylamine (0.1 ml_) in DMF (3 ml_) was heated at 45 0C with stirring overnight. The reaction mixture was then concentrated in vacuo and the resulting crude was purified on silica gel column eluting with 2% and 4% 2N NrVMeOH in CH2CI2 to isolate a yellow solid A22 (36.0 mg, 53%). (LCMS M+1 =
678, ret. time = 2.41 rnin.) 1H-NMR (400 MHz), CDCI3): δ 10.55 (br, 1 H), 10.01 & 9.84 (s, s, 1 H), 8.50 & 8.44 (d, d, 1 H, J=1 Hz), 8.25 (d, 1 H, J=5.2 Hz), 8.08 (s, 1 H), 8.04 (d, 1 H, J=8.2 Hz), 8.00 (d, 1 H, J=8.2 Hz), 7.75 (m, 1 H), 7.53 (m, 1 H), 7.43 (d, 1 H, J=8.2 Hz), 7.39 (m, 2H), 7.34 (d, 1 H, J=5.2 Hz), 6.18 & 6.11 (t, t, 1 H, J=2.5 Hz), 4.42 (q, 2H, J=7.0 Hz), 4.35 & 4.30 (m, m, 1 H), 4.22 (m, 1 H), 3.99 (s, 3H), 3.92 (t, 1 H, J=5.6 Hz), 3.72 (m, 3H), 3.42 (m, 1 H), 3.12 (m, 1 H), 2.84 (q, 1 H, J=7.9 Hz)1 2.76 (d, 1 H, J=9.9 Hz), 2.70 (m, 1 H), 2.63 (m, 2H), 2.15 (s, 3H)1 2.11 (m, 1 H)1 1.44 (t, 3H, J=7.0 Hz). Example 15
Synthesis of 3-Methoxy-1-(2-(4-r4-(1-methyl-1 H-π .2.41triazol-3-yl)-phenvπ-3,6- dihvdro-ΣH-pyridin-i-ylVΣ-oxo-ethvO-pyrrolidine^-carboxylic acid T3-(2- isopropoxy-pyridin-4-yl)-1 H-indazol-5-yll-amide
Figure imgf000275_0001
Step 1 :
Preparation of 4-Bromo-2-lsopropoxy-pyridine
Figure imgf000275_0002
To the stirred solution of 4-bromo-2-fluoro-pyridine 17BV (4.12 g, 23.41 mmole) in 50 mL anhydrous IPA in a 150 mL pressure vessel was added 2.627 g
(23.41 mmole) of solid potassium tert-butoxide under dry N2 gas. The pressure vessel was tightly sealed and heated at 80 0C for 3 hours. The pressure vessel was cooled to 0 0C in ice-bath before opening. The contents of the pressure vessel were transferred to 250 mL RBF and concentrated to a small volume. The resulting mixture was partitioned between EtOAc and H2O. The organic phase was separated, washed with saturated NaCI solution and dried over MgSO4. The solvent was evaporated and resulting clear oil was purified on RediSep 80 g cartridge eluting with 20:1 Hexanes/EtOAc to give clear oil 18BV (4.2 g, 83%). Step 2
Preparation of 3-(2-lsopropoxy-pyridin-4-yl)-5-nitro-1-trityl-1 H-indazole
Figure imgf000276_0001
To the stirred solution of 4.20 g (19.4 mmole) 4-bromo-2-isopropoxy-pyridine 18BV in 150 ml_ anhydrous DMSO, 7.39 g (29.1 mmole) of bis(pinacolato)diboron, 5.704 g (58.2 mmole) of potassium acetate, and 1.584 g (1.94 mmole) of Pd(dppf)CI2 were added at r.t. under dry N2 gas. The mixture was degassed couple of times with dry N2 gas. The dark orange mixture was heated at 100 0C for 2 hours. The dark colored mixture was allowed to cool to r.t. 75 ml_ of H2O was added followed by 9.396 g (19.4 mmole) of i -Trityl-5-Nitroindazole, 13.401 g (96.96 mmole) of potassium carbonate, and 2.246 g (1.94 mmole) of PdTetrakis(Triphenylphosphine) were added at r.t. under dry N2 gas. The mixture was degassed couple of times with dry N2 gas. The dark colored mixture was heated at 100 0C overnight. The mixture was allowed to cool to r.t. and diluted with 1 :1 mixture of H2O/EtOAc.The diluted mixture was filtered through the pad of Celite and Celite pad was liberally washed with EtOAc. The contents were transferred to a separation funnel and shaken well. The organic phase was separated and washed couple of times with saturated NaCI solution, dried over MgSO4 and evaporated to dryness. The dark colored gum was purified on RediSep 330 g cartridge eluting with Hexanes, 5% EtOAc/Hexane and 10% EtOAc/Hexanes, gave 3.24 g (31 %) pale yellow solid 19BV. Step 3
Preparation of 3-(2-lsopropoxy-pyridin-4-yl)-1-trityl-1 H-indazol-5-ylamine
Figure imgf000277_0001
20BV was prepared, with a crude yield of 100%, using essentially the same procedure as in Example 13, Step 3, except using 19BV instead of 4BV.
Step 4:
Preparation of 3-[3-(2-lsopropoxy-pyridin-4-yl)-1-trityl-1 H-indazol-5-ylcarbamoyl]-
3-methoxy-pyrrolidine-1-carboxylic acid tert-butyl ester
Figure imgf000277_0002
Figure imgf000277_0003
21 BV was prepared using essentially the same procedure as in Example 13, Step 4, starting with 20BV instead of 5BV. Step 5:
Preparation of 3-Methoxy-pyrrolidine-3-carboxylic acid [3-(2-isopropoxy-pyridin- 4-yl)-1 H-indazol-5-yl]-amide
Figure imgf000278_0001
22BV was prepared using essentially the same procedure as in Example 13, Step 5, using 21 BV instead of 7BV.
Step 6: Preparation of 3-Methoxy-1 -(2-{4-[4-(1 -methyl-1 H-[1 ,2,4]triazol-3-yl)-phenyl]-3,6- dihydro-2H-pyridin-1 -yl}-2-oxo-ethyl)-pyrrolidine-3-carboxylic acid [3-(2- isopropoxy-pyridin-4-yl)-1H-indazol-5-yl]-amide
Figure imgf000279_0001
A mixture of compound 22BV (40 mg, 0.10 mmol), compound 7BH (41.6 mg, 0.13 mmol) and triethylamine (0.1 mL) in DMF (3 mL) was heated at 45 0C with stirring overnight. The reaction mixture was then concentrated in vacuo and the resulting crude was purified on silica gel column eluting with 2% and 4% 2N NH3MeOH in CH2CI2 to isolate a yellow solid 23BV (35.0 mg, 52%). LCMS M+1 = 676, ret. time = 2.45 min. 1H-NMR (400 MHz, CDCI3): δ 10.42 (br, 1 H), 9.41 & 9.34 (s, S1I H)1 8.43 (dd, 1 H, J=10.7 Hz & 1.2 Hz), 8.25 (d, 1 H, J=5.2 Hz), 8.07 (s, 1 H), 8.05 (m, 2H), 7.74 (m, 1 H), 7.49 (m,1 H), 7.44 ( m, 3H)1 7.29 (br, 1 H), 6.20 & 6.11 (t, t, 1 H, J=2.5 Hz)1 5.37 (m, 1 H)1 4.27 (m, 2H)1 3.98 (s, 3H)1 3.80-3.98 (m, 1 H), 3.74 (m, 1 H), 3.53 (d, 1 H, J=6.6 Hz), 3.49 (br, 1 H), 3.47 & 3.46 (s, s, 3H), 3.28 (d, 1 H, J=10 Hz), 3.02 (m, 2H), 2.88 (m, 1 H), 2.64 (m, 2H), 2.47 (m, 1 H), 2.19 (m, 1 H), 1.40 (d, 6H, J=6.1 Hz).
Example 16
Synthesis of 3-Methylsulfanyl-1-(2-{4-r5-(1-methyl-1 H-F1 ,2,41triazol-3-vD- thiophen-Σ-vn-S.S-dlhvdro-ΣH-pyridin-i-vD-Σ-oxo-ethvπ-pyrrolidine-S-carboxylic acid r3-(6-isopropoxy-pyridin-3-yl)-1 H-indazol-5-vll-amide
Figure imgf000280_0001
Synthesis of 2-Chloro-1-(4-r5-(1-methyl-1 H- M .2.41triazol-3-yl)-thiophen-2-vπ-3.6- dihvdro-2Afpyridin-1-yl)-ethanone
Step i :
Preparation of δ-bromo-thiophene^-carboxylic acid amide
Figure imgf000280_0002
5-Bromo-thiophene 2-carboxylic acid (1 BW) (6g) was suspended in dichloromethane (30 ml) and thionyl chloride (30ml). The resulting solution was allowed to reflux for overnight at 80 0C. Solvent was removed under reduced pressure and the precipitate was dissolved in dichloromethane which was added dropwise to the cooled solution of ammonia in methanol (100 mL) reaction was monitored by TLC and LC-MS. Solvent was removed under reduced pressure to yield 5-bromo-thiophene2-carboxylic acid amide (2BW).
Step 2: Preparation of 5-bromo-thiophene 2- carboximidic acid ethyl ester
Figure imgf000280_0003
The 5-bromo-thiophene2-carboxylic acid amide (2BW) (7g, 34.1 mmols) was dissolved in dichloroethane (170 ml). Triethyloxonium hexaflourophosphate (10.16g, 40.97 mmols) was added and the resulting mixture was refluxed for 1 hr at 90 0C. The reaction mixture was concentrated under reduced pressure, and was taken directly to next step.
Step 3:
Preparation of 5-bromo-thiophene 2-carboximidic-N'-methyl-hydrazide
Figure imgf000281_0001
The 5-bromo-thiophene 2- carboximidic acid ethyl ester (3BW) (13g, 34.3 mmols) was dissolved in pyridine (100 ml). Methylhydrazine (2.7, 51.45 mmols) was added with stirring and the resulted mixture was allowed to stir overnight. The reaction mixture was concentrated under reduced pressure, and added ether, filtered, washed with ether three times and dried to provide the titled compound (4BW) (13 g).
Step 4:
Preparation of 3-(5-bromo-thiophen-2-yl)-1 -methyl- 1H-[\ ,2, 4]triazole
Figure imgf000281_0002
A mixture of compound 5-bromo-thiophene 2-carboximidic-N'-methyl-hydrazide (4BW) (13 g) in formic acid (100 ml) was refluxed overnight and concentrated. The residue was treated with sat. NaHCO3, and extracted with EtOAc three times. The combined organics were dried over MgSO4. After concentration, compound 5BW was purified by column using 80% EtoAc/hexane to yield light yellow colored solid. Step 5:
Preparation of 4-[5-(1 -methyl- 7 H-[1 ,2,4]triazol-3-yl)-thiophen-2-yl]-3,6-dihydro-
2H- pyridine-1-carboxylic acid ferf-butyl ester
Figure imgf000282_0001
To a large pressure flask were charged compound 3-(5-bromo-thiophen-2-yl)-
1 -methyl-1 H-[1 ,2, 4]triazole (3.1 g, 12.8 mmols) (5BW), 4-(4,4,5,5-tetramethyl- [1 ,3,2]dioxaborolan-2-yl)-3,6-dihydro-2/-/-pyridine-1 -carboxylic acid tørt-butyl ester (3.6g, 11.65 mmols), [1 ,1 '-bis(diphenylphosphino)-ferrocene]dichloro-palladium (II), complex with dichloromethane(1 :1) (0.475 g, 0.89 mmols), Na2CO3 (8.5ml) and dioxane (40ml). The mixture was briefly degassed with Ar for -0.5 minute, capped and stirred at 800C overnight. After cooling, the reaction mixture was diluted with EtOAc and brine. Organic layer was isolated, and dried MgSO4. After concentration, the residue was purified on silica gel. Elution with EtOAc (100%) gave the desired product (6BW) (3.5 g).
Step 6:
Preparation of 4-[5-(1 -methyl-1 H- [1 ,2,4]triazol-3-yl)-thiophen-2-y|]-1 ,2,3,6- tetrahydro-pyridine hydrochloride
Figure imgf000282_0002
The Boc group on 6BW can be removed by treating compound 4-[5-(1 -methyl-
1 H-[1 ,2,4]triazol-3-yl)-thiophen-2-yl]-3,6-dihydro-2H-pyridine-1 -carboxylic acid tert- butyl ester with 4N HCI in dioxane at room temperature for two hours. Removal of solvent under vacuum followed by ether wash yielded the desired compound (7BW). Step 7:
Preparation of 2-chloro-1 -{4-[5-(1 -methyl-1 H-[\ ,2,4]triazol-3-yl)-thiophen-2-yl]-
3,6-dihydro-2H-pyridin-1-yl}-ethanone
Figure imgf000283_0001
To a cold (0 0C) solution of 4-[5-(1 -methyl-1 H-[1 ,2,4]triazol-3-yl)-thiophen-2-yl]-
1 ,2,3,6-tetrahydro-pyridine (7BW) (1.5g, 4.72mmol) in dichloromethane (50ml) was added TEA (4.5ml, 28.32mmol) dropwise. After stirred at 0 0C for 10min, chloroacetyl chloride (1.12 ml, 14.2 mmol) was added to the reaction mixture. The resulting mixture was stirred at 0 0C for 1 hr., and quenched with water (15.6ml). The reaction mixture was diluted with dichloromethane (200ml). The organic layer was separated and washed with brine, dried over MgSO4. Reaction mixture was concentrated to ~50ml, ether was added and the solid was filtered out to get the desired product (8BW).
Step 8:
Synthesis of 3-Methoxy-1 -(2-{4-[4-(1 -methyl-1 H-[1 ,2,4]triazol-3-yl)-phenyl]-3,6- dihydro-2H-pyridin-1 -yl}-2-oxo-ethyl)-pyrrolidine-3-carboxylic acid [3-(6- isopropoxy-pyridin-3-yl)-1 H-indazol-5-yl]-amide
Figure imgf000283_0002
Figure imgf000284_0001
A mixture of 2-chloro-1 -{4-[5-(1 -methyl-1 H-[1 ,2,4]triazol-3-yl)-thiophen-2-yl]-3,6- dihydro-2H-pyridin-1 -yl}-ethanone (8BW) (0.522gm, 0.863mmol), 3-Methylsulfanyl- pyrrolidine-3-carboxylic acid [3-(6-isopropoxy-pyridin-3-yl)-1 H-indazol-5-yl]-amide (8BO) (0.463gm, 0.95 mmol), and DIEA (0.9ml, 5.2mmol) in DMF (10ml) was stirred at room temperature for overnight. DMF was removed under reduced pressure. The crude reaction mixture was precipitated in 30ml ice cold water, filtered, dried, and purified by column chromatography using 10% MeOH/EtOAc to get desired product A24 as a yellow solid (70%). (LCMS M+1 = 698, ret. time= 3.6 min.) 1 H NMR ( 400 MHz, DMSO):D8.67 (S, 1 H), 8.64(S,1 H), 8.36 (S, 1 H), 8.15 (S, 1 H), 7.64(d, 1 H, J= 8.8Hz), 7.52 (S, 1 H), 7.49 (d, 1 H J= 5.2Hz), 7.40( d, 1 H J=8.8Hz), 7.02 (m, 1 H), 6.18 (d, 1 H, J= 18.8 Hz), 5.08 (m, 1 H), 4.41 (d, 2H, J=7.6Hz), 4.34 (d, 1 H, J=6.4Hz)), 4.12( S, 1 H), 3.96 (s, 2H), 3.84 (S, 3H), 3.76 (S, 2H), 3.52(S, 2H), 2.76 (S, 1 H), 2.58 (d, 2H J = 20.4Hz), 2.13 (S, 3H), 1.94 (t, 2H J= 2.8Hz), 1.39 (d, 6H, J=4.8 Hz ).
Examples 17 to 25
Compounds A4, A6, A8, A10-A12, A18, A25 and A26 were prepared following the procedures indicated in the Table 1 below. Table 1
Figure imgf000285_0001
The LCMS data for compounds in Table 1 are given in Table 2 below.
Table 2
Figure imgf000285_0002
Figure imgf000286_0003
EXAMPLE 26
Preparation of (S)-N-(3-(6-isopropoxypyridin-3-yl)-1 H-indazol-5-vD-3-methoxy-1- (2-(4-(5-(1 -methyl-1 H-1 ,2,4-triazol-3-yl)pyridin-2-yl)-5,6-dihvdropyridin-1 (2H)-yl)-2- oxoethyl)pyrrolidine-3-carboxamide:
Figure imgf000286_0001
Step 1 : Methyl 6-chloronicotinimidate (1 BX)
Figure imgf000286_0002
Sodium methoxide (725mg, 13.42mmol) was added to a solution of 2-Chloro pyridine-5-carbonitrile (1.8g,13.04mmol) in MeOH:dioxane ( 40ml,1 :1 ) at O0C, then stirred for 30 minutes at O0C, and 1 hour at room temperature.The reaction was diluted with EtOAc (200ml) and H2O (100ml), organic layer separated, dried over Na2SO4, filtered and solvent evaporated to yield title compound 1 BX as a white solid (2.6g,100%) MS (MH 171 ) Step 2: β-chloro-N'-methylnicotinimidohydrazide (2BX)
Figure imgf000287_0001
Methyl hydrazine (750mg, 16.30mmol) was added to a solution of Methyl 6- chloronicotinimidate (1 BX) (2.6g, 15.29mmol) in Pyridine (10ml) at room temperature, then stirred for 1 hour. The solvent was evaporated, and residual solid triturated with cold Ether (2x1 OmI) yielding title product 2BX as a yellow powder (2.4g, 85%) MS (MH, 185)
Step 3: 2-chloro-5-(1-methyl-1 H-1 ,2,4-triazol-3-yl)pyridine (3BX)
Figure imgf000287_0002
A solution of β-chloro-N'-methylnicotinimidohydrazide (2BX) (2.4g, 13mmol) in Formic Acid (99%, 10ml) was stirred at reflux temperature for 1 hour. Reaction was cooled and solvent evaporated. The residue was extracted with EtOAc ( 100ml) and Aqueous NaHCO3 (50ml), organic layer was separated.dried over Na2SO4, filtered and solvent evaporated.The residue chromatographed on silica gel eluting with 10% MeOHiCH2CI2 yielding title product as a solid (1.8g, 72%) MS (MH, 195)
Step 4: Tert-butyl 4-(5-(1 -methyl-1 H-1 ,2,4-triazol-3-yl)pyridin-2-yl)-5,6- dihydropyridine-1 (2H)-carboxylate (4BX)
Figure imgf000287_0003
A mixture of 2-chloro-5-(1 -methyl-1 H-1 ,2,4-triazol-3-yl)pyridine (3BX) ( 400mg, 2.06mmol); N-Tert-Butoxycarbonyl-1 ,2,3,6-tetrahydropyridine-4-boronic acid.pinacol ester (1.4g, 4.53mmol); Cesium carbonate ( 2.3g,7.07mmol) and PdCI2dppf (100mg) in dioxane/H2O ( v/v10:1 , 20ml) was stirred at 1000C for 2 hours.The reaction was cooled, diluted with CH2CI2 (300ml) and H2O (100ml), organic layer separated, dried over Na2SO4, filtered and solvent evaporated yielding a residue which chromatographed on silica gel eluting with EtOAc yielding title product 4BX as a solid (400mg,57%) MS (MH.342).
Step 5: 5-(1 -methyl-1 H-1 ,2,4-triazol-3-yl)-2-(1 ,2,3,6-tetrahydropyridin-4-yl)pyridine dihydrochloride (5BX)
Figure imgf000288_0001
4M HCI in dioxane ( 20ml) was added to a solution of Tert-butyl 4-(5-(1 -methyl- 1 H-1 ,2,4-triazol-3-yl)pyridin-2-yl)-5,6-dihydropyridine-1 (2H)-carboxylate (4BX) (4g, 11.67mmol) in CH2CI2 ( 50ml) at room temperature. The mixture was stirred for 3 hours then solvent was evaporated yielding title product as a white solid (3.8g).
Step 6: 2-chloro-1-(4-(5-(1 -methyl-1 H-1 ,2,4-triazol-3-yl)pyridin-2-yl)-5,6- dihydropyridin-1 (2H)-yl)ethanone (6BX)
Figure imgf000288_0002
Added 1 N NaOH (50ml,50mmol) and Chloroacetyl chloride (3ml, 37.7mmol) in CH2CI2 (50ml) dropwise to a solution of 5-(1 -methyl-1 H-1 ,2,4-triazol-3-yl)-2-(1 ,2,3,6- tetrahydropyridin-4-yl)pyridine dihydrochloride (5BX) ( 1g, 3.60mmol) in CH2CI2( 50ml at O0C, maintaining pH at >12. Mixture was stirred for 2 hours at O0C, then reaction was diluted with CH2CI2 (200ml) and H2O (100ml). The organic layer was separated, washed with H2O (50ml), dried (Na2SO4), filtered and solvent evaporated yielding title product as a white solid (1.1 g, 100%) MS (MH, 318) Step 7
Figure imgf000289_0001
Added a solution of 2-chloro-1-(4-(5-(1 -methyl- 1 H-1 ,2,4-triazol-3-yl)pyridin-2- yl)-5,6-dihydropyridin-1 (2H)-yl)ethanone (6BX) (0.85g,2.68mmol) in CH2CI2 (10ml) to a solution of the Indazole ( 1 g,2.53mmol) in DMF (10ml) at room temperature, then stirred at 500C for 3 hours. The reaction was diluted with EtOAc (300ml) and H2O (100ml), then the organic layer was separated, dried over Na2SO4, filtered and evaporated solvent. The residue chromatographed on silica gel eluting with 10% v/v MeOH/CH2CI2/NH4OH yielding product A27 as a white solid (1.3g,76%) MS (MH,677)
LCMS Elution time = 2.61 minutes
EXAMPLE 27 Preparation of (S)-N-(3-(6-isopropoxypyridin-3-yl)-1 H-indazol-5-yl)-3-methoxy-1- (2-(4-(5-(1 -methyl-1 H-1 ,2.4-triazol-3-yl)thiazol-2-yl)-5.6-dihvdropyridin-1 (2H)-yl)-2- oxoethvOpvrrol8dine-3-carboxamide:
Figure imgf000289_0002
Step i
Figure imgf000290_0001
To a stirred mixture of 2-chlorothiazole-carbonitrile (1 g, 6.92 mmol) in MeOH was added NaOMe (745 mg, 13.8 mmol, 2 equiv.) and the the reaction mixture was stirred at O -C for 15 min and warmed to rt for another 15 min, quenched with water, extracted with EtOAc. The combined organic layer was washed with brine, dried over Na2SO4 and concentrated to give crude 2BY (1.2 g) as a yellow oil. To the stirred solution of crude 2BY in pyridine (1 ml) was added methyl hydrazine (363 μl_, 6.9 mmol, 1 equiv.), the resulting mixture was stirred at 0 QC for 15 min and quenched with HCOOH (5 ml). The resulting mixture was then transferred to a sealed tube and stirred at 110 SC for overnight, cooled to rt, to which water was added. The resulting mixture was extracted with CH2CI2. The combined organic layer was washed with brine, dried and concentrated to give a yellow solid, which was filtered and washed with CH2CI2. The resulting yellow solid (375 mg) is desired product 4BY. The filtrate was concentrated and could be further purified on column to provide more product.
Step 2
Figure imgf000290_0002
A mixture of 4BY (130 mg) in POCI3 was stirred under N2 at 120 QC for 2 d. The initial heterogeneous mixture became a cleaer brown solution, which was concentrated and purified on silica gel (CH2CI2/MeOH, 50/1 ) to give 5BY (143 mg) as a yellow solid.
Step 3
Figure imgf000291_0001
A mixture of 5BY (1 18 mg, 0.59 mmol), 6BY (274 mg, 0.89 mmol, 1.5 equiv.), 2
M Na2CO3 (590 μl_, 1.18 mmol, 2 equiv.) and Pd(PPh3)4 (34 mg, 0.05 equiv.) in benzene/MeOH (5 ml_, 4/1 ) was degassed and stirred under N2 at 80 QC for overnight. The reaction mixture was then concentrated and purified on silica gel (CH2CI2/MeOH, 30/1) to give 7BY (150 mg) as a yellow solid.
Step 4
Figure imgf000291_0002
Figure imgf000291_0003
A mixture of 7BY (150 mg, 0.43 mmol) and TFA (1.5 ml_) was stirred at rt for 1 h and concentrated. Chromatograph on silica gel (CH2CI2/MeOH, 15/1 ) gave the 8BY (98 mg). To a stirred mixture of 8BY (98 mg, 0.4 mmol) and triethylamine (335 μl_, 2.4 mmol, 6 equiv.) in CH2CI2/MeOH (6 ml_, 2/1 ) at 0 5C was added chloroacetyl chloride (126 μl_, 1.6 mmol, 4 equiv.). The reaction mixture was stirred at 0 3C for 1 h and concentrated. Chromatograph on silica gel (CH2CI2/MeOH, 25/1 ) gave the 9BY (103 mg) as a white solid.
Step 5
Figure imgf000292_0001
Compound 9BY was substituted for compound 7BH in Example 15 step 6 to obtain A28. LCMS MH = 683.4, Retention time = 2.77 minutes
EXAMPLE 28 Preparation of (S)-N-(3-(6-isopropoxypyridin-3-yl)-1 H-indaol-5-vD-3-
(methylthio)-i -(2-oxo-2-(4-(5-(6-oxopyridazin-1 (6H)-yl)-5,6-dihvdropyridin-1 (2H)- yl)ethyl)pyrrolidine-3-carboxamide) (A29)
Figure imgf000292_0002
Step 1 : Preparation of 2-(5-bromothiophen-2-yl)pyridazin-3(2H)-one
Figure imgf000293_0001
A mixture of 2,5-dibromothiophene (1.5 g, 6.2 mmol), pyridazin-3(2H)-one (0.4 g, 4.1 mmol), copper(l) iodide (0.24 g, 1.2 mmol), potassium carbonate (1.7 g, 12.4 mmol), trans-N,N'-dimethylcyclohexane-1 ,2-diamine (0.2 ml_, 1.2 mmol) and toluene (15 ml_) was degassed for 15 minutes and then heated in a sealed tube at 110 0C for 18 hours. Cooled to room temperature, filtered through celite and washed with EtOAc. The filterate was washed with water (100 ml_ x 2). The organic layer was dried over Na2SO4, filtered and concentrated give the desired product 2BZ (0.9 g, 90%). The residue was purified on silica gel eluting with 80% EtOAc/ hexane to give the desired product 2BZ (0.7 g, 67%).
Step 2: Preparation of tert-butyl 4-(5-(6-oxopyridazin-1(6H)-yl)thiophen-2-yl)-5,6- dihydropyridine-1(2H)-carboxylate
Figure imgf000293_0002
A mixture of Compound 2BZ (0.7 g, 2.7 mmol), 4-(4,4,5,5-tetramethyl- [1 ,3,2]dioxaborolan-2-yl)-3,6-dihydro-2H-pyridine-1 -carboxylic acid te/t-butyl ester (0.84 g, 2.7 mmol), 2M aq. sodium carbonate solution (6.8 ml_, 13.6 mmol), Pd(PPh3)4 (0.31 g, 0.27 mmol) and 1/1 /toluene/ethanol (20 ml_) was degassed for 15 minutes. Then it was heated at 90 °C for overnight. Cooled to room temperature and diluted with EtOAc (200 ml). The organic layer was washed with water (100 ml), dried over Na2SO4, filtered and concentrated. The residue was purified on silica gel eluting with 60% EtOAc/hexane to give the desired product 3BZ (0.63 g, 65%). Step 4: Preparation of 2-(5-(1,2,3,6-tetrahydropyridin-4-yl)thiophen-2- yl)pyridazin-3(2W)-one
Figure imgf000294_0001
A mixture of Compound 3BZ (0.63 g, 1.75 mmol), CH2CI2 (20 ml_) and TFA (23ml_) was stirred at room temperature for 18 hours. Concentrated and purified on silica gel eluting with 5% MeOH (MH3)/ CH2CI2 to give the desired product 4BZ (0.4 g, 89%).
Step 5: Preparation of 2-(5-(1-(2-chloroacetyl)-1 ,2,3,6-tetrahydropyridin-4- yl)thiophen-2-yl)pyridazin-3(2H)-one
Figure imgf000294_0002
To a mixture of Compound 4BZ (0.62 g, 2.39 mmol), CH2CI2 (10 ml_), MeOH (3 ml_) and triethyl amine (0.67 ml_, 4.78 mmol) at -78 0C was added chloroacetyl chloride (0.19 ml_( 2.39 mmol). Reaction mixture was stirred at -78 °C for 10 minutes then warm to 0 0C and stirred for 1 hour. Diluted with CH2CI2 (100 ml_) and washed with saturated aq. NaHCO3 (100 ml_). The organic layer was dried over Na2SO4, filtered and concentrated. The residue was taken in CH2CI2 and added ether. Resulting solid was filtered and washed with ether and dried to give the desired product 5BZ (0.56 g, 70%). Step 6
Figure imgf000295_0001
A mixture of Compound 5BZ (0.04 g, 0.12 mmol), compound 6BZ (0.05 g, 0.12 mmol), DMF (2 ml_) and N,N-diisopropylethylamine (0.042 ml_, 0.24 mmol) was stirred at room temperature for 18 hours. Diluted with EtOAc (100 ml_) and washed with water (2 x 100 ml_). The organic layer was dried over Na2SO4, filtered and concentrated. The residue was purified on silica gel eluting with 3% MeOH (NH3)/ CH2CI2 to give the desired product A29 (0.083 g, 97%).
LCMS MH = 711 , Retention time = 3.26 minutes.
EXAMPLE 29
Preparation of (S)-N-(3-(6-isopropoxypyridin-3-yl)-1 H-indaol-5-vh-1-(2-(4-(5-(3- methyl-6-oxopyridazin-1(6H)-yl)thiophen-2-vn-5.6-dihvdropyridin-1(2A/)-yl)-2- oxoethyl)-3-(methylthio)pyrrolidine-3-carboxamide (A30)
Figure imgf000295_0002
Compound A30 was prepared using a procedure similar to that in Example 28 except that 6-methylpyridazin-3(2/-/)-one was used in place of pyridazin-3(2/-/)-one in Step 1.
LCMS MH = 725, Retention time = 3.21 minutes.
EXAMPLE 30
Figure imgf000296_0001
8BO (4.25g, 10.33 mmol) was dissolved in N,N-dimethylformamide (40 ml) at room temperature. Diisopropylethylamine (5.1 mL, 30.99 mmol) was added followed by 7BH (3.27g, 10.33 mmol). The reaction mixture was stirred for 1 hr. overnite at ambient temperature. Brine was added to the reaction mixture which was then extracted with ethylacetate (3X100 ml_). The ethylacetate extracts were dried over magnesium sulfate, filtered and evaporated to obtain crude title product. The crude product was chromatographed to obtain 4.95g (69%) of title product (A6)(10% 2Λ/ NH3 in MeOHiCH2CI2).
EXAMPLE 31
Figure imgf000297_0001
Figure imgf000297_0002
To CH3Ph (3 ml_) solution of 1CA (140 mg, 1.4 mmol) was added a drop of DMF followed by addition of oxalyl chloride (0.14 ml_, 1.6 mmol) at r.t. After 30 mins, the resulting clear solution was added to a CH2CI2 solution of 3CA (240 mg, 1 mmol) and triethyl amine (0.5 ml_). After stirring at r.t. for 30 mins, it was quenched with sat. NaHCO3, extracted with CH2CI2 and cone, to get an off white solid as a crude 4CA (312 mg) which was used directly in next reactions. In 1CA, 2CA, and 4CA "D" represents deuterium.
Compounds A31 and A32, were prepared by a method similar to the method for preparing Compound A1 and A6 substituting 4CA for 7BH. Compounds A45 and A48 were prepared by a method similar to the method for preparing A23 and A25 substituting 4CA for 7BH. In A31 , A32, A45 and A48 "D" represents deuterium.
Figure imgf000298_0001
Figure imgf000299_0001
EXAMPLE 32
If one were to follow a procedure similar to the procedure in Example 31 , Compounds A33 to A44, A46, and A47 below would be obtained. In A33 to A44, A46, and A47 "D" represents deuterium.
Figure imgf000299_0002
Figure imgf000299_0003
Figure imgf000300_0001
Figure imgf000300_0002
Figure imgf000300_0003
Figure imgf000300_0004
Figure imgf000301_0001
Figure imgf000301_0002
Figure imgf000301_0003
Figure imgf000301_0004
Figure imgf000302_0001
Figure imgf000302_0002
Figure imgf000302_0003
Figure imgf000302_0004
Figure imgf000303_0001
Figure imgf000303_0002
In A33 to A44, A46, and A47 "D" represents deuterium.
ASSAYS
Coupled ERK2 Assay:
Activity of compounds against inactive ERK2 can be tested in a coupled MEK1/ERK2 IMAP assay as follows: Compounds can be diluted to 25x final test concentration in 100% DMSO. 14μl of kinase buffer (1OmM Tris.HCI pH 7.2, 1OmM MgCI2, 0.01% Tween-20, 1 mM DTT) containing 0.4ng unphosphorylated Mouse ERK2 protein can be added to each well of a black 384-well assay plate. 1 μl of 25x compound can be added to each well and incubated at room temperature for 30 minutes to allow an opportunity for the compound to bind to the inactive enzyme. DMSO concentration during initial incubation is 6.7%. ERK2 activity can be determined to be insensitive to DMSO concentrations up to 20%. ERK2 can then be activated and it's kinase activity can be measured by the addition of 10μl kinase buffer with the following components (final concentration per reaction): 2ng active (phosphorylated) human MEK1 protein and 4μM (total) ERK2 IMAP substrate peptides (3.9μM unlabeled IPTTPITTTYFFFK-CONH2 and 10OnM
IPTTPITTTYFFFK(5-carboxylluorescein)-CONH2 ) and 30μM ATP. DMSO concentration during ERK activation can be 4%. After one hour, reactions can be terminated by addition of 60μl IMAP detections beads in binding buffer (Molecular Devices). Binding can be allowed to equilibrate for 30 minutes before reading the plate on an LJL Analyst Fluorescence Polarization plate reader. Compound inhibition can be calculated relative to DMSO and fully inhibited standards. Active compounds can be reconfirmed in an independent assay.
Active ERK2 Assay:
Activated ERK2 activity was also determined in the IMAP assay format using the procedure outlined above. 1 μl of 25x compound was added to 14μl of kinase buffer containing 0.25ng fully phosphorylated, active Mouse ERK2 protein. Following a 30 minute incubation, the reactions were initiated by addition of 10μl of kinase buffer containing 1 μM ERK2 IMAP substrate peptide (0.9μM unlabeled IPTTPITTTYFFFK-COMH2 and 10OnM IPTTPITTTYFFFK(5-carboxyfluorescein)- CONH2 ) and 30μM ATP. Reactions proceeded for 30 minutes before termination by addition of 60μl IMAP detection beads in binding buffer. Plates were read as above after 30 minute binding equilibration. Active compounds were reconfirmed in an independent assay.
Soft Agar Assay:
Anchorage-independent growth is a characteristic of tumorigenic cell lines. Human tumor cells can be suspended in growth medium containing 0.3% agarose and an indicated concentration of a farnesyl transferase inhibitor. The solution can be overlayed onto growth medium solidified with 0.6% agarose containing the same concentration of ERK1 and ERK2 inhibitor as the top layer. After the top layer is solidified, plates can be incubated for 10-16 days at 370C under 5% CO2 to allow colony outgrowth. After incubation, the colonies can be stained by overlaying the agar with a solution of MTT (3-[4,5-dimethyl-thiazol-2-yl]-2,5-diphenyltetrazolium bromide, Thiazolyl blue) (1 mg/mL in PBS). Colonies can be counted and the ICso's can be determined. The AUC (area under the concentration-time curve during the first 6 hours (AUCfihr) was determined using the Protocol of Cassette Accelerating Rapid Rat screen (CARRS) Animal dosing and sample collection Male Sprague-Dawley rats (Charles River, Co.) were pre-cannulated (femoral artery) in order to facilitate precise blood sampling times, and to reduce the stress on the animals caused by serial bleedings. Following an overnight fast, two rats were dosed orally with one compound at a dose of 10 mg/kg in a 5-mL/kg dose volume. Blood was collected into heparin-containing tubes serially from each animal at 0.5, 1 , 2, 3, 4 and 6 h post-dosing and centrifuged to generate plasma. Approximately 100 μl_ of plasma were collected at the individual time points. The plasma samples were stored at -20 0C until analysis.
Plasma sample and standard curve preparation A set of 12 rat plasma samples was generated for each NCE (i.e. 6 timepoints and n = 2 rats). These 12 samples were pooled across the two rats at each timepoint to provide 6 pooled samples (one sample per time point) for each NCE. The pooled samples were assayed as cassettes of six (36 samples total) to provide data on the six compounds. The50-μL aliquots of the 36 plasma samples were placed into individual wells of a 96-well plate. An additional compound (often a structural analog of the test compounds) was selected as the internal standard. A mini-calibration curve was prepared (three points plus a zero) for each compound assayed. Drug-free rat plasma was measured into 1-mL aliquots and each aliquot was spiked with known concentrations of the compounds to generate standards of the desired concentrations. The concentrations of the standards were chosen to bracket the expected concentration of the pooled samples based on historical data from previous studies on other compounds. For this work, the standards were set to contain concentrations of 25, 250 and 2500 ng NCE/mL plasma. The plasma standards were precipitated in duplicate along with the samples. Protein precipitation occurred after addition of 150 μl_ of acetonitrile containing the internal standard at a concentration of 1 ng/mL into each sample well using the Tomtec Quadra 96 system. The precipitated samples and standards were vortexed and centrifuged in the 96-well plate. Approximately 50-100 μl_ of the supernatant were removed and placed into a fresh 96-well plate using the Tomtec Quadra 96 system. A volume of 5-10 μl_ of the supernatant was used for analysis by HPLC-MS/MS. The mini-standard curve was run in duplicate, once before and once after the samples. Thus, a total of 14 study samples plus standards were analyzed per compound. In addition, solvent blanks were injected before and after each set of 14 and after the highest calibration standard for each compound; therefore, a total of 103 injections were made into each HPLC system for each set of six compounds. Multiple solvent blank injections could be made from a single well. Twelve solvent blank wells were designated in each 96- well plate. Thus, one batch (cassette) of six NCEs was prepared and assayed using one 96-well plate format.
HPLC-MS/MS analysis
All the compounds were analyzed using selected reaction monitoring (SRM) methods with LC/MS/MS instruments. Once the method development had been completed, the assay was quickly set up using a standard injection sequence template for the CARRS assay.
Compounds A1 to A16, A18, A20, A21 , A23, A25, A26, and A27 to A30 had an AERK2 IC50 in the range of 1.2 to 50 riM.
Compounds A1 -A3, A6, A8-A11 , A13-A16, A20-A24, A26 and A27 to A30 had an AUC in the range of 36 to 50,999nM.hr in the CARRS assay. For preparing pharmaceutical compositions from the compounds described by this invention, inert, pharmaceutically acceptable carriers can be either solid or liquid. Solid form preparations include powders, tablets, dispersible granules, capsules, cachets and suppositories. The powders and tablets may be comprised of from about 5 to about 95 percent active ingredient. Suitable solid carriers are known in the art, e.g. magnesium carbonate, magnesium stearate, talc, sugar or lactose. Tablets, powders, cachets and capsules can be used as solid dosage forms suitable for oral administration. Examples of pharmaceutically acceptable carriers and methods of manufacture for various compositions may be found in A. Gennaro (ed.), Remington: The Science and Practice of Pharmacy, 20th Edition, (2000), Lippincott Williams & Wilkins, Baltimore, MD.
Liquid form preparations include solutions, suspensions and emulsions. As an example may be mentioned water or water-propylene glycol solutions for parenteral injection or addition of sweeteners and opacifiers for oral solutions, suspensions and emulsions. Liquid form preparations may also include solutions for intranasal administration.
Aerosol preparations suitable for inhalation may include solutions and solids in powder form, which may be in combination with a pharmaceutically acceptable carrier, such as an inert compressed gas, e.g. nitrogen.
Also included are solid form preparations which are intended to be converted, shortly before use, to liquid form preparations for either oral or parenteral administration. Such liquid forms include solutions, suspensions and emulsions.
The compounds of the invention may also be deliverable transdermally. The transdermal compositions can take the form of creams, lotions, aerosols and/or emulsions and can be included in a transdermal patch of the matrix or reservoir type as are conventional in the art for this purpose.
Preferably the compound is administered orally.
Preferably, the pharmaceutical preparation is in a unit dosage form. In such form, the preparations subdivided into suitably sized unit doses containing appropriate quantities of the active component, e.g., an effective amount to achieve the desired purpose.
The quantity of active compound in a unit dose of preparation may be varied or adjusted from about 0.01 mg to about 1000 mg, preferably from about 0.01 mg to about 750 mg, more preferably from about 0.01 mg to about 500 mg, and most preferably from about 0.01 mg to about 250 mg according to the particular application.
The actual dosage employed may be varied depending upon the requirements of the patient and the severity of the condition being treated. Determination of the proper dosage regimen for a particular situation is within the skill in the art. For convenience, the total daily dosage may be divided and administered in portions during the day as required.
The amount and frequency of administration of the compounds of the invention and/or the pharmaceutically acceptable salts thereof will be regulated according to the judgment of the attending clinician considering such factors as age, condition and size of the patient as well as severity of the symptoms being treated. A typical recommended daily dosage regimen for oral administration can range from about 0.04 mg/day to about 4000 mg/day, in two to four divided doses.
While the present invention has been described in conjunction with the specific embodiments set forth above, many alternatives, modifications and variations thereof will be apparent to those of ordinary skill in the art. All such alternatives, modifications and variations are intended to fall within the spirit and scope of the present invention.

Claims

WHAT IS CLAIMED IS:
1. A compound of formula 1.0:
Figure imgf000308_0001
or a pharmaceutically acceptable salt or solvate thereof, wherein R , R and Q are independently selected, and wherein: Q is:
Figure imgf000308_0002
R1 is s from the. group consisting of: heteroaryl and substituted heteroaryl, wherein said substituted heteroaryl is substituted with 1 to 3 substituents independently selected from the group consisting of: -OH, alkoxy, and -O-alkylene-O-alkyl;
R2 is selected from the group consisting of: -O-alkyl and -S-alkyl; and R5 is selected from the group consisting of: (a) triazolyl-phenyl-,
(b) triazolyl-phenyl- wherein said phenyl is optionally substituted with 1 to 3 substituents independently selected from the group consisting of: halo and alkoxy,
(c) substituted triazolyl-phenyl- wherein said phenyl is optionally substituted with 1 to 3 substituents independently selected from the group consisting of: halo and alkoxy, and said triazolyl group is substituted with one or two substitutents independently selected from the group consisting of: alkyl, hydroxy substituted alkyl, -alkylene-O-alkyl, and amino,
(d) triazolyl-thienyl-,
(e) triazolyl-thienyl- wherein said thienyl is optionally substituted with 1 to 2 substituents independently selected from the group consisting of: halo and alkoxy,
(f) substituted triazolyl-thienyl- wherein said thienyl is optionally substituted with 1 to 2 substituents independently selected from the group consisting of: halo and alkoxy, and said triazolyl group is substituted with one or two substitutents independently selected from the group consisting of: alkyl, hydroxy substituted alkyl, -alkylene-O-alkyl, and amino, (g) triazolyl-pyridyl-, (h) triazolyl-pyridyl- wherein said pyridyl is optionally substituted with 1 to 3 substituents independently selected from the group consisting of: halo, alkyl, and alkoxy, provided that the carbon atoms adjacent to the nitrogen atom in said pyridyl are not substituted with halo, and
(i) substituted triazolyl-pyridyl- wherein: (1) said pyridyl is optionally substituted with 1 to 3 substituents independently selected from the group consisting of: halo, alkyl, and alkoxy, provided that the carbon atoms adjacent to the nitrogen atom in said pyridyl are not substituted with halo, and (2) said triazolyl group is substituted with one or two substitutents independently selected from the group consisting of: alkyl, hydroxy substituted alkyl, -alkylene-O-alkyl, and amino, 0) triazolyl-thiazolyl-,
(k) triazolyl-thiazolyl- wherein said thiazolyl is optionally substituted with 1 substituent independently selected from the group consisting of: halo, alkyl, and alkoxy, amino, alkylamino, and dialkylamino wherein each alkyl is independently selected, and (I) substituted triazolyl-thiazolyl- wherein (1 ) said thiazolyl is optionally substituted with 1 substituent independently selected from the group consisting of: halo, alkyl, and alkoxy, amino, alkylamino, and dialkylamino wherein each alkyl is independently selected, and (2) said triazolyl group is substituted with one or two substitutents independently selected from the group consisting of: alkyl, hydroxy substituted alkyl, -alkylene-O-alkyl, and amino, (m) pyridazinyl-thienyl-,
(n) pyridazinyl-thienyl- wherein said thienyl is optionally substituted with 1 to 2 substituents independently selected from the group consisting of: halo and alkoxy, and (o) substituted pyridazinyl-thienyl- wherein (1 ) said thienyl is optionally substituted with 1 to 2 substituents independently selected from the group consisting of: halo and alkoxy, and (2) said pyridazinyl group is substituted with 1 to 3 substitutents independently selected from the group consisting of: =0, alkyl, amino, alkylamino, dialkylamino wherein each alkyl is independently selected, and halo, provided that the carbon atoms adjacent to the nitrogen atoms in said pyridazinyl are not substituted with halo, and provided that when said -alkylene-O-alkyl group is bound to the nitrogen of said triazolyl in (c), (f), (i) and (I) of R5 the alkylene moiety of said -alkylene-O-alkyl group is not -CH2-.
2. The compound of Claim 1 wherein R1 is a pyridyl substituted with a substituent selected from the group consisting of: -OH, alkoxy, and -O-alkylene-O-alkyl.
3. The compound of Claim 2 wherein said substituent is selected from the group consisting of: -0-CH3, -0-C2H5, -O-CH(CH3)2, and -0-(CH2J2-O-CH3.
4. The compound of Claim 1 wherein R1 is selected from the group consisting of:
Figure imgf000310_0001
5. The compound of Claim 1 wherein R2 is selected from the group consisting of: -OCH3 and -SCH3.
6. The compound of Claim 1 wherein Rs is selected from the group consisting of:
(a) a substituted triazolyl-phenyl- wherein the triazolyl moiety is substituted with one or two alkyl groups selected from the group consisting of: -Ci-C6alkyl, -C1-C4SlM. -Ci-C2alkyl, and -CH3;
(b) a substituted triazolyl-phenyl- wherein the triazolyl moiety is substituted with one or two alkyl groups selected from the group consisting of: -CrC4alkyl, -CrC2alkyl, and -CH3; (c) a substituted triazolyl-phenyl- wherein the triazolyl moiety is substituted on the nitrogen with an alkyl group selected from the group consisting of: -Ci-C4alkyl, -d-C2alkyl, and -CH3;
(d) a substituted triazolyl-phenyl- wherein the triazolyl moiety is substituted on the nitrogen with an -alkylene-O-alkyl group selected from the group consisting of: -C2-C4alkylene-O-Ci-C6alkyl, -C2alkylene-O-Ci-C2alkyl, -Cz^alkylene-O-CHa, and -CH2CH2OCH3;
(e) a substituted triazolyl-phenyl- wherein the triazolyl moiety is substituted on the nitrogen with an -alkylene-O-alkyl group selected from the group consisting of: -C2alkylene-O-Ci-C2alkyl, and -CH2CH2OCH3;
(f) a substituted triazolyl-phenyl- wherein the triazolyl moiety is substituted on the nitrogen with a hydroxy substituted alkyl group selected from the group consisting of: hydroxy substituted -C1-C4BlRyI, hydroxy substituted -d-C2alkyl, and hydroxy substituted -CH3; and (g) a substituted triazolyl-phenyl- wherein the triazolyl moiety is substituted on the nitrogen with a hydroxy substituted alkyl group selected from the group consisting of: CH2COH(CH3)2, and -CH2CH2OH.
7. The compound of Claim 1 wherein the optional halo substituents for the phenyl moiety of R5 are F.
8. The compound of Claim 1 wherein R5 is a substituted triazolyl-phenyl- group wherein:
(1 ) (a) said triazolyl moiety is optionally substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2, and -CH2CH2OH, (b) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is optionally substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is optionally substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is optionally substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is optionally substituted on the nitrogen with a -CH2CH2OCH3 group; and wherein said phenyl moiety is optionally substituted with halo; or
(2)
(a) said triazolyl moiety is optionally substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2, and -CH2CH2OH, (b) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is optionally substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is optionally substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is optionally substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is optionally substituted on the nitrogen with a -CH2CH2OCH3 group; and wherein said phenyl moiety is optionally substituted with alkoxy; or
(3) (a) said triazolyl moiety is optionally substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2, and -CH2CH2OH, (b) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is optionally substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is optionally substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is optionally substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is optionally substituted on the nitrogen with a -CH2CH2OCH3 group; and wherein said phenyl moiety is substituted with halo; or
(4)
(a) said triazolyl moiety is optionally substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2, and -CH2CH2OH, (b) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is optionally substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is optionally substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is optionally substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is optionally substituted on the nitrogen with a -CH2CH2OCH3 group; and wherein said phenyl moiety is substituted with alkoxy; or
(5) (a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2, and -CH2CH2OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is substituted on the nitrogen with a -CH2CH2OCH3 group; and wherein said phenyl moiety is optionally substituted with halo; or
(6)
(a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2, and -CH2CH2OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is substituted on the nitrogen with a -CH2CH2OCH3 group; and wherein said phenyl moiety is optionally substituted with alkoxy; or
(7)
(a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2, and -CH2CH2OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is substituted on the nitrogen with a -CH2CH2OCH3 group; and wherein said phenyl moiety is substituted with halo; or
(8)
(a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CHs)2, and -CH2CH2OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is substituted on the nitrogen with a -CH2CH2OCH3 group; and wherein said phenyl moiety is substituted with alkoxy; or
O)
(a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CHg)2, and -CH2CH2OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH3 group, (β) said triazolyl moiety is substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is substituted on the nitrogen with a -CH2CH2OCH3 group; and wherein said phenyl moiety is unsubstituted.
9. The compound of Claim 1 wherein R5 is a substituted triazolyl-phenyl group wherein said triazolyl moiety Is substituted with: (a) one substituent selected from the group consisting of: -CH2COH(CH3)2, and -CH2CH2OH, (b) one alkyl group, (c) two alkyl groups, (d) one -CH3 group, (e) two -CH3 groups, (f) one -NH2 group, or (g) one -CH2CH2OCH3 group.
10. The compound of Claim 1 wherein the optional alkoxy substituents for the phenyl moiety of R5 are -OCH3.
11. The compound of Claim 1 wherein R5 is selected from the group consisting of:
(a) a substituted triazolyl-thienyl- wherein the triazolyl moiety is substituted with one or two alkyl groups selected from the group consisting of:
-Ci-C6alkyl, -d-C4alkyl, -d-C2alkyl, and -CH3;
(b) a substituted triazolyl-thienyl- wherein the triazolyl moiety is substituted with one or two alkyl groups selected from the group consisting of: -CrC4alkyl, -Ci-C2alkyl, and -CH3; (c) a substituted triazolyl-thienyl- wherein the triazolyl moiety is substituted on the nitrogen with an alkyl group selected from the group consisting of: -Ci-C4alkyl, -Ci-C2alkyl, and -CH3;
(d) a substituted triazolyl-thienyl- wherein the triazolyl moiety is substituted on the nitrogen with an -alkylene-O-alkyl group selected from the group consisting of: -^^alkylene-O-CrCβalkyl, -C2alkylene-O-Ci-C2alkyl, -C2-C4alkylene-O-CH3, and -CH2CH2OCH3;
(e) a substituted triazolyl-thienyl- wherein the triazolyl moiety is substituted on the nitrogen with an -alkylene-O-alkyl group selected from the group consisting of: -C2alkylene-O-Ci-C2alkyl, and -CH2CH2OCH3; (f) a substituted triazolyl-thienyl- wherein the triazolyl moiety is substituted on the nitrogen with a hydroxy substituted alkyl group selected from the group consisting of: hydroxy substituted -CrC4alkyl, hydroxy substituted -CrC2alkyl, and hydroxy substituted -CH3; and
(g) a substituted triazolyl-thienyl- wherein the triazolyl moiety is substituted on the nitrogen with a hydroxy substituted alkyl group selected from the group consisting of: CH2COH(CH3)2, and -CH2CH2OH.
12. The compound of Claim 1 wherein R5 is a substituted triazolyl-thienyl- group wherein: (A)
(a) said triazolyl moiety is optionally substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2, and -CH2CH2OH, (b) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is optionally substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is optionally substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is optionally substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is optionally substituted on the nitrogen with a -CH2CH2OCH3 group; and wherein said thienyl moiety is optionally substituted with halo; or
(B)
(a) said triazolyl moiety is optionally substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2, and -CH2CH2OH, (b) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is optionally substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is optionally substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is optionally substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is optionally substituted on the nitrogen with a -CH2CH2OCH3 group; and wherein said thienyl moiety is optionally substituted with alkoxy; or
(C)
(a) said triazolyl moiety is optionally substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2, and -CH2CH2OH, (b) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is optionally substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is optionally substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is optionally substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is optionally substituted on the nitrogen with a -CH2CH2OCH3 group; and wherein said thienyl moiety is substituted with halo; or
(D)
(a) said triazolyl moiety is optionally substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2, and -CH2CH2OH, (b) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is optionally substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is optionally substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is optionally substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is optionally substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is optionally substituted on the nitrogen with a -CH2CH2OCH3 group; and wherein said thienyl moiety is substituted with alkoxy; or
(E)
(a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2, and -CH2CH2OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is substituted on the nitrogen with a -CH2CH2OCH3 group; and wherein said thienyl moiety is optionally substituted with halo; or
(F)
(a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2, and -CH2CH2OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is substituted on the nitrogen with a
-CH2CH2OCH3 group; and wherein said thienyl moiety is optionally substituted with alkoxy; or (G)
(a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2, and -CH2CH2OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is substituted on the nitrogen with a -CH2CH2OCH3 group; and wherein said thienyl moiety is substituted with halo; or
(H)
(a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3J2, and -CH2CH2OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is substituted on the nitrogen with a
-CH2CH2OCH3 group; and wherein said thienyl moiety is substituted with alkoxy; or
(I)
(a) said triazolyl moiety is substituted on the nitrogen with a substituent selected from the group consisting of: -CH2COH(CH3)2, and -CH2CH2OH, (b) said triazolyl moiety is substituted on the nitrogen with an alkyl group, (c) said triazolyl moiety is substituted on the nitrogen with an alkyl group, and on the carbon with an alkyl group, (d) said triazolyl moiety is substituted on the nitrogen with a -CH3 group, (e) said triazolyl moiety is substituted on the nitrogen with one -CH3 group, and on the carbon with one -CH3 group, (f) said triazolyl moiety is substituted on the carbon with a -NH2 group, or (g) said triazolyl moiety is substituted on the nitrogen with a -CH2CH2OCH3 group; and wherein said thienyl moiety is unsubstituted.
13. The compound of Claim 1 wherein R5 is a substituted triazolyl-thienyl group wherein said triazolyl moiety is substituted with: (a) one substituent selected from the group consisting of:-CH2COH(CH3)2 and -CH2CH2OH, (b) one alkyl group, (c) two alkyl groups, (d) one -CH3 group, (e) two -CH3 groups, (f) one -NH2 group, or (g) one -CH2CH2OCH3 group.
14. The compound of Claim 1 wherein the optional alkoxy substituents for the phenyl moiety of R5 are -OCH3.
15. The compound of Claim 8 wherein R1 is a pyridyl substituted with a substituent selected from the group consisting of: -0-CH3, -0-C2H5, -O-CH(CH3)2, and -O-(CH2)2-O-CH3, and R2 is selected From the group consisting of: -OCH3 and -SCH3.
16. The compound of Claim 8 wherein R1 is selected from the group consisting of:
Figure imgf000319_0001
and R2 is selected from the group consisting of: -OCH3 and -SCH3.
17. The compound of Claim 1 wherein: (a) R5 is a substituted triazolyl-pyridyl- wherein said triazolyl is substituted and said pyridyl is substituted; or
(b) R5 is a substituted triazolyl-pyridyl- wherein said triazolyl is substituted on the nitrogen and said pyridyl is substituted; or
(c) R5 is a substituted triazolyl-pyridyl- wherein said triazolyl is substituted on the carbon and said pyridyl is substituted; or
(d) R5 is a substituted triazolyl-pyridyl- wherein said triazolyl is substituted on the nitrogen and on the carbon, and said pyridyl is substituted; or
(e) R5 is a substituted triazolyl-pyridyl- wherein said triazolyl is unsubstituted and said pyridyl is substituted.
18. The compound of Claim 1 wherein:
(a) R5 is a substituted triazolyl-pyridyl- wherein the triazolyl is substituted on the nitrogen with an alkyl group, and the pyridyl is unsubstituted; or
(b) R5 is a substituted triazolyl-pyridyl- wherein the triazolyl is substituted on the nitrogen with an alkyl group and substituted on the carbon with an alkyl group, wherein each alkyl group is independently selected, and the pyridyl is unsubstituted; or
(c) R5 is a substituted triazolyl-pyridyl- wherein the triazolyl is substituted on the nitrogen with a -CH3 group, and the pyridyl is unsubstituted; or (d) R5 is a substituted triazolyl-pyridyl- wherein the triazolyl is substituted on the nitrogen with a -CH3 group and on the carbon with a -CH3 group, and the pyridyl is unsubstituted.
19. The compound of Claim 1 wherein: (a) R5 is a substituted triazolyl-thiazolyl- wherein said triazolyl is substituted and said thiazolyl is unsubstituted; or
(b) R5 is a substituted triazolyl-thiazolyl- wherein said triazolyl is substituted on the nitrogen and said thiazolyl is unsubstituted; or
(c) R5 is a substituted triazolyl-thiazolyl- wherein said triazolyl is substituted on the carbon and said thiazolyl is unsubstituted; or
(d) R5 is a substituted triazolyl-thiazolyl- wherein said triazolyl is substituted on the nitrogen and on the carbon, and said thiazolyl is unsubstituted; or
(e) R5 is a substituted triazolyl-thiazolyl- wherein said triazolyl is substituted and said thiazolyl is substituted; or (f) R5 is a substituted triazolyl-thiazolyl- wherein said triazolyl is substituted on the nitrogen and said thiazolyl is substituted; or
(g) R5 is a substituted triazolyl-thiazolyl- wherein said triazolyl is substituted on the carbon and said thiazolyl is substituted; or
(f) R5 is a substituted triazolyl-thiazolyl- wherein said triazolyl is substituted on the nitrogen and on the carbon, and said thiazolyl is substituted; or
(h) R5 is a substituted triazolyl-thiazolyl- wherein said triazolyl is unsubstituted and said thiazolyl is substituted.
20. The compound of Claim 1 wherein: (a) R5 is a substituted triazolyl-thiazolyl- wherein the triazolyl is substituted on the nitrogen with alkyl, and said thiazolyl is unsubstituted; or
(b) R5 is a substituted triazolyl-thiazolyl- wherein the triazolyl is substituted on the nitrogen with an alkyl group and substituted on the carbon with an alkyl group, wherein each alkyl group is independently selected, and said thiazolyl is unsubstituted; or
(c) R5 is a substituted triazolyl-thiazolyl- wherein the triazolyl is substituted on the nitrogen with a -CH3 group, and said thiazolyl is unsubstituted; or
(d) R5 is a substituted triazolyl-thiazolyl- wherein the triazolyl is substituted on the nitrogen with a -CH3 group and on the carbon with a -CH3 group, and said thiazolyl is unsubstituted.
21. The compound of Claim 1 wherein R5 is a substituted pyridazinyl-thienyl- wherein said pyridazinyl is substituted and said thienyl is unsubstituted; or R5 is a substituted pyridazinyl-thienyl- wherein said pyridazinyl is unsubstituted and said thienyl is substituted; or R5 is a substituted pyridazinyl-thienyl- wherein said pyridazinyl is substituted and said thienyl is substituted.
22. The compound of Claim 1 wherein: (a) R5 is a substituted pyridazinyl-thienyl-wherein said pyridazinyl is substituted with a =0 group, and said thienyl is unsubstituted; or
(b) R5 is a substituted pyridazinyl-thienyl-wherein said pyridazinyl is substituted with an alkyl group, and said thienyl is unsubstituted; or
(c) R5 is a substituted pyridazinyl-thienyl-wherein said pyridazinyl is substituted with a methyl group, and said thienyl is unsubstituted; or
In another embodiment of this invention R5 is a substituted pyridazinyl-thienyl- wherein said pyridazinyl is substituted with a =0 group, and with an alkyl group, and said thienyl is unsubstituted.
(d) R5 is a substituted pyridazinyl-thienyl-wherein said pyridazinyl is substituted with a =0 group, and with a methyl group, and said thienyl is unsubstituted.
23. The compound of Claim 1 wherein:
Figure imgf000322_0001
Figure imgf000323_0001
24. The compound of Claim 23 wherein R1 is a pyridyl substituted with a substituent selected from the group consisting of: -0-CH3, -0-C2H5, -0-CH(CH3J2, and -O-(CH2)2-O-CH3, and R2 is selected from the group consisting of: -OCH3 and -SCH3.
25. The compound of Claim 24 wherein R1 is selected from the group consisting of:
Figure imgf000323_0002
and R is selected from the group consisting of: -OCH3 and -SCH3.
26. The compound of Claim 11 wherein the compound of formula 1.0 is a compound of formula 1.1 :
Figure imgf000323_0003
27. The compound of Claim 26 wherein R5 is selected from the group consisting of:
Figure imgf000324_0001
Figure imgf000325_0001
28. The compound of Claim 27 wherein R1 is a pyridyl substituted with a substituent selected from the group consisting of: -0-CH3, -0-C2H5, -O-CH(CH3)2, and -O-(CH2)2-O-CH3, and R2 is selected from the group consisting of: -OCH3 and -SCH3.
29. The compound of Claim 28 wherein R1 is selected from the group consisting of:
Figure imgf000325_0002
30. The compound of Claim 1 wherein R5 is selected from the group consisting of:
Figure imgf000325_0003
,
Figure imgf000326_0001
31. The compound of Claim 29 wherein R5 is selected from the group consisting of:
Figure imgf000326_0002
Figure imgf000327_0001
32. The compound of Claim 1 wherein R5 is selected from the group consisting of:
Figure imgf000327_0002
Figure imgf000328_0001
Figure imgf000328_0002
33. The compound of Claim 29 wherein R5 is selected from the group consisting of:
Figure imgf000328_0003
Figure imgf000328_0004
Figure imgf000329_0001
34. The compound of Claim 1 wherein R5 is triazolyl-phenyk
35. The compound of Claim 1 wherein R5 is triazolyl-thienyl-.
36. The compound of Claim 1 wherein R5 is
Figure imgf000329_0002
37. The compound of Claim 1 wherein R5 is:
Figure imgf000329_0003
38. The compound of Claim 1 wherein R is
Figure imgf000329_0004
39. The compound of Claim 1 wherein R5 is
Figure imgf000329_0005
40. The compound of Claim 1 wherein R5 is
Figure imgf000329_0006
41. A compound selected from the group consisting of compounds A1 to A16 and A18 to A30.
42. A compound selected from the group consisting of compounds A31 to
A48.
43. The compound of Claim 41 wherein said compound is compound A1.
44. The compound of Claim 41 wherein said compound is compound A2.
45. The compound of Claim 41 wherein said compound is compound A3.
46. The compound of Claim 41 wherein said compound is compound A6.
47. The compound of Claim 41 wherein said compound is compound A7.
48. The compound of Claim 41 wherein said compound is compound A13.
49. The compound of Claim 41 wherein said compound is compound A19.
50. The compound of Claim 41 wherein said compound is compound A22.
51. The compound of Claim 41 wherein said compound is compound A24.
52. The compound of Claim 41 wherein said compound is compound A25.
53. The compound of Claim 41 wherein said compound is compound A27.
54. The compound of Claim 41 wherein said compound is compound A28.
55. The compound of Claim 41 wherein said compound is compound A29.
56. The compound of Claim 41 wherein said compound is compound A30.
57. A solvate, or a pharmaceutically acceptable salt, of a compound selected from the group consisting of compounds A1 to A16 and A18 to A48.
58. A pharmaceutically acceptable salt of a compound of Claim 41.
59. A solvate of a compound of Claim 41.
60. A solvate, or a pharmaceutically acceptable salt, of a compound of Claim 42.
61. The compound of Claim 1 in pure and isolated form.
62. The compound of Claim 41 in pure and isolated form.
63. The compound of Claim 42 in pure and isolated form.
64. A pharmaceutically acceptable salt of a compound of Claim 42.
65. A pharmaceutical composition comprising at least one compound of
Claim 1 and a pharmaceutically acceptable carrier.
66. A pharmaceutical composition comprising at least one compound of Claim 41 and a pharmaceutically acceptable carrier.
67. A pharmaceutical composition comprising at least one compound of Claim 42 and a pharmaceutically acceptable carrier.
68. A method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one compound of Claim 1.
69. A method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one compound of Claim 1 in combination with an effective amount of at least one chemotherapeutic agent.
70. A method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of a compound of Claim 1 in combination with an effective amount of at least one chemotherapeutic agent, and an effective amount of radiation therapy.
71. A method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of at least one compound of Claim 1 , and therapeutically effective amounts of at least one chemotherapeutic agent selected from the group consisting of: (1) taxanes, (2) platinum coordinator compounds, (3) epidermal growth factor (EGF) inhibitors that are antibodies, (4) EGF inhibitors that are small molecules, (5) vascular endolithial growth factor (VEGF) inhibitors that are antibodies, (6) VEGF kinase inhibitors that are small molecules, (7) estrogen receptor antagonists or selective estrogen receptor modulators (SERMs), (8) anti-tumor nucleoside derivatives, (9) epothilones, (10) topoisomerase inhibitors, (11) vinca alkaloids, (12) antibodies that are inhibitors of αVβ3 integrins, (13) folate antagonists, (14) ribonucleotide reductase inhibitors, (15) anthracyclines, (16) biologies; (17) inhibitors of angiogenesls and/or suppressors of tumor necrosis factor alpha (TNF-alpha) such as thalidomide (or related imid), (18) Bcr/abl kinase inhibitors, (19) MEK1 and/or MEK 2 inhibitors that are small molecules, (20) IGF-1 and IGF-2 inhibitors that are small molecules, (21) small molecule inhibitors of RAF and BRAF kinases, (22) small molecule inhibitors of cell cycle dependent kinases such as CDK1 , CDK2, CDK4 and CDK6, (23) alkylating agents, and (24) farnesyl protein transferase inhibitors.
72. A method of treating cancer in a patient in need of such treatment, said method comprising administering to said patient an effective amount of: (a) at least one compound of Claim 1 in combination with at least one signal transduction inhibitor;
(b) at least one compound of Claim 1 , said cancer being selected from the group consisting of: lung cancer, pancreatic cancer, colon cancer, myeloid leukemias, thyroid cancer, myelodysplasia syndrome, bladder carcinoma, epidermal carcinoma, mβlanoma, breast cancer, prostate cancer, head and neck cancers, ovarian cancer, brain cancers, cancers of mesenchymal origin, sarcomas, tetracarcinomas, myoblastomas, kidney carcinomas, hepatomas, non-Hodgkin's lymphoma, multiple myeloma, and anaplastic thyroid carcinoma; (c) at least one compound of Claim 1 , wherein said cancer is selected from the group consisting of: melanoma, pancreatic cancer, thryroid cancer, colorectal cancer, lung cancer, breast cancer, and ovarian cancer;
(d) at least one compound of Claim 1 , in combination with an effective amount of at least one chemotherapeutic agent, wherein said cancer is selected from the group consisting of: melanoma, pancreatic cancer, thryroid cancer, colorectal cancer, lung cancer, breast cancer, and ovarian cancer;
(e) at least one compound of Claim 1 , and said cancer is melanoma;
(f) at least one compound of Claim 1 , in combination with an effective amount of at least one chemotherapeutic agent, and said cancer is melanoma; (g) at least one compound of Claim 1 , and said cancer is pancreatic cancer;
(h) at least one compound of Claim 1 , in combination with an effective amount of at least one chemotherapeutic agent, and said cancer is pancreatic cancer;
(i) at least one compound of Claim 1 , and said cancer is thyroid cancer;
(j) at least one compound of Claim 1 , in combination with an effective amount of at least one chemotherapeutic agent, and said cancer is thyroid cancer;
(k) at least one compound of Claim 1 , and said cancer is colorectal cancer;
(I) at least one compound of Claim 1 , in combination with an effective amount of at least one chemotherapeutic agent, and said cancer is colorectal cancer;
(m) at least one compound of Claim 1 , and said cancer is lung cancer; (n) at least one compound of Claim 1 , in combination with an effective amount of at least one chemotherapeutic agent, wherein said cancer is lung cancer;
(o) at least one compound of Claim 1 , and said cancer is breast cancer;
(p) at least one compound of Claim 1 , in combination with an effective amount of at least one chemotherapeutic agent, and said cancer is breast cancer; (q) at least one compound of Claim 1 , and said cancer is ovarian cancer;
(r) at least one compound of Claim 1 , in combination with an effective amount of at least one chemotherapeutic agent, and said cancer is ovarian cancer;
(s) at least one compound of Claim 1 , in combination with antihormonal agents, wherein said cancer is hormone-dependent breast cancer; (t) at least one compound of Claim 1 , in combination with antihormonal agents, and in combination with an effective amount of at least one chemotherapeutic agent wherein said cancer is hormone-dependent breast cancer;
(u) at least one compound of Claim 1 , and said cancer is brain cancer; (v) at least one compound of Claim 1 , in combination with an effective amount of at least one chemotherapeutic agent, and said cancer is brain cancer;
(w) at least one compound of Claim 1 , in combination with an effective amount of at least one chemotherapeutic agent, and said chemotherapeutic agent is temozolomide, and wherein said cancer is brain cancer; (x) at least one compound of Claim 1 , and said cancer is prostate cancer;
(y) at least one compound of Claim 1 , in combination with an effective amount of at least one chemotherapeutic agent, and said cancer is prostate cancer;
(z) at least one compound of Claim 1 , and said cancer is myelodysplastic syndrome; (aa) at least one compound of Claim 1 , in combination with an effective amount of at least one chemotherapeutic agent, and said cancer is myelodysplastic syndrome cancer;
(ab) at least one compound of Claim 1 , and said cancer is a myeloid leukemia;
(ac) at least one compound of Claim 1 , in combination with an effective amount of at least one chemotherapeutic agent, and said cancer is a myeloid leukemia;
(ad) at least one compound of Claim 1 , and said cancer is acute myelogenous leukemia;
(ae) at least one compound of Claim 1 , in combination with an effective amount of at least one chemotherapeutic agent, and said cancer is acute myelogenous leukemia;
(af) at least one compound of Claim 1 , and said cancer is chronic myelomonocytic leukemia;
(ag) at least one compound of Claim 1 , in combination with an effective amount of at least one chemotherapeutic agent, and said cancer is chronic myelomonocytic leukemia;
(ah) at least one compound of Claim 1 , and said cancer is chronic myelogenous leukemia; (ai) at least one compound of Claim 1 , in combination with an effective amount of at least one chemotherapeutic agent, and said cancer is chronic myelogenous leukemia;
(aj) at least one compound of Claim 1 , and said cancer is bladder cancer; (ak) at least one compound of Claim 1 , in combination with an effective amount of at least one chemotherapeutic agent, and said cancer is bladder cancer;
(al) at least one compound of Claim 1 , and said cancer is non-Hodgkin's lymphoma;
(am) at least one compound of Claim 1 , in combination with an effective amount of at least one chemotherapeutic agent, and said cancer is non-Hodgkin's lymphoma;
(an) at least one compound of Claim 1 , and said cancer is multiple myeloma; or
(ao) at least one compound of Claim 1 , in combination with an effective amount of at least one chemotherapeutic agent, and said cancer is multiple myeloma;
73. A method of preventing hormone-dependent breast cancer in a patient in need of such treatment, said treatment comprising the administration of an effective amount of: (a) at least one compound of Claim 1 in combination with antihormonal agents; or
(b) at least one compound of Claim 1 in combination with antihormonal agents, and in combination with an effective amount of at least one chemotherapeutic agent.
PCT/US2009/034447 2008-02-21 2009-02-19 Compounds that are erk inhibitors WO2009105500A1 (en)

Priority Applications (13)

Application Number Priority Date Filing Date Title
RU2010138635/04A RU2525389C2 (en) 2008-02-21 2009-02-19 Compounds which are erk inhibitors
ES09712601.5T ES2556353T3 (en) 2008-02-21 2009-02-19 Compounds that are ERK inhibitors
BRPI0908120A BRPI0908120A8 (en) 2008-02-21 2009-02-19 COMPOUNDS WHICH ARE ERK INHIBITORS, PHARMACEUTICAL COMPOSITION AND USE
MX2010009268A MX2010009268A (en) 2008-02-21 2009-02-19 Compounds that are erk inhibitors.
JP2010547732A JP5276676B2 (en) 2008-02-21 2009-02-19 Compounds that are ERK inhibitors
US12/918,099 US8716483B2 (en) 2008-02-21 2009-02-19 Compounds that are ERK inhibitors
EP09712601.5A EP2260031B1 (en) 2008-02-21 2009-02-19 Compounds that are erk inhibitors
CN200980113870.7A CN102015693B (en) 2008-02-21 2009-02-19 Compounds that are ERK inhibitors
AU2009215534A AU2009215534B8 (en) 2008-02-21 2009-02-19 Compounds that are ERK inhibitors
NZ587504A NZ587504A (en) 2008-02-21 2009-02-19 Benzopyrazole derivatives as ERK inhibitors
CA2714479A CA2714479A1 (en) 2008-02-21 2009-02-19 Compounds that are erk inhibitors
IL207530A IL207530A (en) 2008-02-21 2010-08-10 Triazolyl-phenyl compounds and use thereof for the manufacture of medicaments for treating cancer
ZA2010/05909A ZA201005909B (en) 2008-02-21 2010-08-18 Compounds that are erk inhibitors

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US3040708P 2008-02-21 2008-02-21
US61/030,407 2008-02-21

Publications (1)

Publication Number Publication Date
WO2009105500A1 true WO2009105500A1 (en) 2009-08-27

Family

ID=40601237

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2009/034447 WO2009105500A1 (en) 2008-02-21 2009-02-19 Compounds that are erk inhibitors

Country Status (23)

Country Link
US (1) US8716483B2 (en)
EP (1) EP2260031B1 (en)
JP (1) JP5276676B2 (en)
KR (1) KR20100117123A (en)
CN (1) CN102015693B (en)
AR (1) AR070460A1 (en)
AU (1) AU2009215534B8 (en)
BR (1) BRPI0908120A8 (en)
CA (1) CA2714479A1 (en)
CL (1) CL2009000394A1 (en)
CO (1) CO6300939A2 (en)
EC (1) ECSP10010415A (en)
ES (1) ES2556353T3 (en)
IL (1) IL207530A (en)
MX (1) MX2010009268A (en)
MY (1) MY152271A (en)
NZ (1) NZ587504A (en)
PE (1) PE20091491A1 (en)
RU (1) RU2525389C2 (en)
SG (1) SG188179A1 (en)
TW (1) TWI398441B (en)
WO (1) WO2009105500A1 (en)
ZA (2) ZA201005909B (en)

Cited By (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010021978A2 (en) * 2008-08-19 2010-02-25 Schering Corporation Il-8 biomarker
US7807672B2 (en) 2006-02-16 2010-10-05 Schering Corporation Compounds that are ERK inhibitors
WO2011002887A1 (en) 2009-07-02 2011-01-06 Schering Corporation FUSED TRICYCLIC COMPOUNDS AS NOVEL mTOR INHIBITORS
WO2012036997A1 (en) * 2010-09-16 2012-03-22 Schering Corporation Fused pyrazole derivatives as novel erk inhibitors
WO2012052412A1 (en) 2010-10-22 2012-04-26 Bayer Cropscience Ag Novel heterocyclic compounds as pesticides
EP2584903A1 (en) * 2010-06-24 2013-05-01 Merck Sharp & Dohme Corp. Novel heterocyclic compounds as erk inhibitors
US8546404B2 (en) 2005-12-13 2013-10-01 Merck Sharp & Dohme Compounds that are ERK inhibitors
US8871966B2 (en) 2013-03-15 2014-10-28 Iowa State University Research Foundation, Inc. Regiospecific synthesis of terephthalates
US9000209B2 (en) 2011-07-22 2015-04-07 Iowa State University Research Foundation, Inc. Method of regioselective synthesis of substituted benzoates
CN105050598A (en) * 2012-09-28 2015-11-11 默沙东公司 Novel compounds that are erk inhibitors
US9229008B2 (en) 2008-08-19 2016-01-05 Merck Sharp & Dohme Corp. IL-8 level as a determinant of responsivity of a cancer to treatment
CN105377856A (en) * 2013-03-13 2016-03-02 豪夫迈·罗氏有限公司 Process for making benzoxazepin compounds
EP2875001A4 (en) * 2012-07-18 2016-06-01 Sunshine Lake Pharma Co Ltd Nitrogenous heterocyclic derivatives and their application in drugs
WO2016100147A1 (en) 2014-12-18 2016-06-23 Merck Sharp & Dohme Corp. (s)-n-(3-(6-isopropoxypyridin-3-yl)-1h-indazol-5-yl)-1-(2-(4-(4-(1-methyl-1h-1,2,4-triazol-3-yl)phenyl)-3,6-dihydropyridin-1(2h)-yl)-2-oxoethyl)-3- (methylthio)pyrrolidine-3-carboxamide compositions for pharmaceutical preparations
WO2016100152A1 (en) 2014-12-19 2016-06-23 Merck Sharp & Dohme Corp. (s)-n-(3-(6-isopropoxypyridin-3-yl)-1h-indazol-5-yl)-1-(2-(4-(4-(1-methyl-1h-1,2,4-triazol-3-yl)phenyl)-3,6-dihydropyridin-1(2h)-yl)-2-oxoethyl)-3-(methylthio)pyrrolidine-3-carboxamide compositions for pharmaceutical preparations
WO2016095089A1 (en) * 2014-12-15 2016-06-23 Merck Sharp & Dohme Corp. Erk inhibitors
WO2016095088A1 (en) * 2014-12-15 2016-06-23 Merck Sharp & Dohme Corp. Erk inhibitors
US9624228B2 (en) 2013-10-03 2017-04-18 Kura Oncology, Inc. Inhibitors of ERK and methods of use
US10519126B2 (en) 2012-03-01 2019-12-31 Array Biopharma Inc. Serine/threonine kinase inhibitors
US10525036B2 (en) 2015-04-03 2020-01-07 Recurium Ip Holdings, Llc Spirocyclic compounds
WO2021067266A1 (en) * 2019-10-01 2021-04-08 Recurium Ip Holdings, Llc Pyrrolidinyl-based compounds
WO2021069567A1 (en) 2019-10-09 2021-04-15 Bayer Aktiengesellschaft Novel heteroaryl-triazole compounds as pesticides

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8658651B2 (en) 2009-09-30 2014-02-25 Merck Sharp & Dohme Corp. Compounds that are ERK inhibitors
WO2012068148A1 (en) * 2010-11-16 2012-05-24 Medimmune, Llc Regimens for treatments using anti-igf antibodies
WO2013169858A1 (en) 2012-05-08 2013-11-14 The Broad Institute, Inc. Diagnostic and treatment methods in patients having or at risk of developing resistance to cancer therapy
US9226922B2 (en) 2012-09-28 2016-01-05 Merck Sharp & Dohme Corp. Compounds that are ERK inhibitors
US10428387B2 (en) 2014-05-16 2019-10-01 University Of Massachusetts Treating chronic myelogenous leukemia (CML)
WO2016025639A1 (en) * 2014-08-13 2016-02-18 Celgene Avilomics Research, Inc. Combinations of an erk inhibitor and a chemotherapeutic agent and related methods
US20180250232A1 (en) * 2015-09-03 2018-09-06 Merck Sharp & Dohme Corp. Process for preparing spray dried solid dispersions of (s)-n-(3-(6-isopropoxypyridin-3-yl)-1h-indazol-5-yl)-1-(2-(4-(4-(1-methyl-1h-1,2,4-triazol-3-yl)phenyl)-3,6-dihydropyridin-1(2h)-yl)-2-oxoethyl)-3-(methylthio)pyrrolidine-3-carboxamide for pharmaceutical preparations
CN106432182B (en) * 2016-09-06 2019-04-30 铜仁学院 Specially the synthetic method of azoles amine intermediate
CN109020789B (en) * 2017-06-12 2021-08-13 浙江医药股份有限公司新昌制药厂 Method for preparing 2-methoxypropene

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007070398A1 (en) * 2005-12-13 2007-06-21 Schering Corporation Polycyclic indazole derivatives that are erk inhibitors
WO2008153858A1 (en) * 2007-06-05 2008-12-18 Schering Corporation Polycyclic indazole derivatives and their use as erk inhibitors for the treatment of cancer

Family Cites Families (54)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB2306108A (en) 1995-10-13 1997-04-30 Merck & Co Inc Treatment of Raf-mediated cancers with imidazole derivatives
AU3215197A (en) 1996-05-30 1998-01-05 Merck & Co., Inc. A method of treating cancer
GB2323845A (en) 1997-03-31 1998-10-07 Merck & Co Inc MEK inhibiting lactones
JP2001510168A (en) 1997-07-18 2001-07-31 ノボ ノルディスク アクティーゼルスカブ Use of FVIIa or FVIIai for the treatment of adverse conditions associated with FVIIa-mediated intracellular signaling pathway
GB9716557D0 (en) 1997-08-06 1997-10-08 Glaxo Group Ltd Benzylidene-1,3-dihydro-indol-2-one derivatives having anti-cancer activity
CA2369502A1 (en) 2000-02-05 2001-08-09 Vertex Pharmaceuticals Incorporated Compositions useful as inhibitors of erk
EP1200422A2 (en) 2000-02-05 2002-05-02 Vertex Pharmaceuticals Incorporated Pyrazole compositions useful as inhibitors of erk
WO2001068619A1 (en) 2000-03-15 2001-09-20 Warner-Lambert Company 5-amide substituted diarylamines as mex inhibitors
TWI310684B (en) 2000-03-27 2009-06-11 Bristol Myers Squibb Co Synergistic pharmaceutical kits for treating cancer
US20050009876A1 (en) 2000-07-31 2005-01-13 Bhagwat Shripad S. Indazole compounds, compositions thereof and methods of treatment therewith
US7211594B2 (en) 2000-07-31 2007-05-01 Signal Pharmaceuticals, Llc Indazole compounds and compositions thereof as JNK inhibitors and for the treatment of diseases associated therewith
US6897231B2 (en) * 2000-07-31 2005-05-24 Signal Pharmaceuticals, Inc. Indazole derivatives as JNK inhibitors and compositions and methods related thereto
MXPA03002299A (en) 2000-09-15 2003-06-06 Vertex Pharma Pyrazole compounds useful as protein kinase inhibitors.
ATE335737T1 (en) 2000-09-15 2006-09-15 Vertex Pharma ISOXAZOLES AND THEIR USE AS ERK INHIBITORS
EP1353905B1 (en) 2000-11-20 2007-01-10 Scios Inc. Piperidine/piperazine-type inhibitors of p38 kinase
ATE528303T1 (en) 2000-12-21 2011-10-15 Vertex Pharma PYRAZOLE COMPOUNDS AS PROTEIN KINASE INHIBITORS
MY130778A (en) 2001-02-09 2007-07-31 Vertex Pharma Heterocyclic inhibitiors of erk2 and uses thereof
JP4535680B2 (en) 2001-04-16 2010-09-01 エーザイ・アール・アンド・ディー・マネジメント株式会社 New 1H-indazole compounds
JP2004538266A (en) 2001-04-27 2004-12-24 バーテックス ファーマシューティカルズ インコーポレイテッド Pyrazole-derived kinase inhibitors
US6962936B2 (en) 2001-04-27 2005-11-08 Vertex Pharmaceuticals Incorporated Triazole-derived kinase inhibitors and uses thereof
DE60238620D1 (en) 2001-08-03 2011-01-27 Vertex Pharma KINASEIN HIBITORS DERIVED FROM PYRAZOL AND THEIR USE
CA2456192A1 (en) 2001-08-03 2003-02-13 Qing Tang Pyrazole-derived kinase inhibitors and uses thereof
AU2002359291C1 (en) 2001-10-23 2008-11-20 Merck Serono Sa Azole derivatives and pharmaceutical compositions containing them
US20030187026A1 (en) 2001-12-13 2003-10-02 Qun Li Kinase inhibitors
EP1506189A1 (en) 2002-04-26 2005-02-16 Vertex Pharmaceuticals Incorporated Pyrrole derivatives as inhibitors of erk2 and uses thereof
WO2003099212A2 (en) 2002-05-24 2003-12-04 The University Of Utah Research Foundation Mitogen activated protein kinase inhibitor compositions for lymphoma therapy
WO2003101968A1 (en) 2002-05-31 2003-12-11 Eisai Co., Ltd. Pyrazole compound and medicinal composition containing the same
US7205308B2 (en) 2002-09-04 2007-04-17 Schering Corporation Trisubstituted 7-aminopyrazolopyrimidines as cyclin dependent kinase inhibitors
US7196092B2 (en) 2002-09-04 2007-03-27 Schering Corporation N-heteroaryl pyrazolopyrimidines as cyclin dependent kinase inhibitors
EP1539756B1 (en) 2002-09-19 2007-11-14 Schering Corporation Imidazopyridines as cyclin dependent kinase inhibitors
AU2004221881A1 (en) 2003-03-13 2004-09-30 Vertex Pharmaceuticals Incorporated Compositions useful as protein kinase inhibitors
GB2400101A (en) 2003-03-28 2004-10-06 Biofocus Discovery Ltd Compounds capable of binding to the active site of protein kinases
WO2005002673A1 (en) 2003-07-03 2005-01-13 Astex Therapeutics Limited Raf kinase inhibitors
WO2005051308A2 (en) 2003-11-19 2005-06-09 Signal Pharmaceuticals, Llc Methods of treating diseases and disorders by targeting multiple kinases
EP1715871A1 (en) 2003-12-22 2006-11-02 Gilead Sciences, Inc. Kinase inhibitor phosphonate conjugates
US7462612B2 (en) 2004-03-26 2008-12-09 Vertex Pharmaceuticals Incorporated Pyridine inhibitors of ERK2 and uses thereof
US20070208015A1 (en) 2004-04-13 2007-09-06 Astex Therapeutics, Ltd. 5-Morpholinylmethylthiophenyl Pharmaceutical Compounds As P38 MAP Kinase Modulators
PL3305776T3 (en) 2004-05-14 2020-03-31 Vertex Pharmaceuticals Incorporated Pyrrole compounds as inhibitors of erk protein kinases and pharmaceutical compositions containing these compounds
EP1753744B1 (en) 2004-05-14 2009-07-08 Vertex Pharmaceuticals Incorporated Prodrugs of pyrrolylpyrimidine erk protein kinase inhibitors
WO2006040569A1 (en) 2004-10-14 2006-04-20 Astex Therapeutics Limited Thiophene amide compounds for use in the treatment or prophylaxis of cancers
JP2008525461A (en) 2004-12-23 2008-07-17 バーテックス ファーマシューティカルズ インコーポレイテッド Selective inhibitor of ERK protein kinase and use thereof
CN100377868C (en) 2005-03-24 2008-04-02 中国科学院物理研究所 Nuclear composite film for magnetic, nonmagnetic and magnetic multilayer film and use thereof
WO2006136008A1 (en) 2005-05-24 2006-12-28 University Health Network Salicylic acid hydrazones as inhibitors of the erk mapkinase pathway and for the treatment of cancer
US8217042B2 (en) 2005-11-11 2012-07-10 Zentaris Gmbh Pyridopyrazines and their use as modulators of kinases
US8546404B2 (en) 2005-12-13 2013-10-01 Merck Sharp & Dohme Compounds that are ERK inhibitors
JP2009528280A (en) 2006-02-16 2009-08-06 シェーリング コーポレイション Pyrrolidine derivatives as ERK inhibitors
EP2260869A3 (en) 2006-04-20 2011-03-23 Takeda Pharmaceutical Company Limited Pharmaceutical product
US7601852B2 (en) 2006-05-11 2009-10-13 Kosan Biosciences Incorporated Macrocyclic kinase inhibitors
WO2008005268A1 (en) 2006-06-30 2008-01-10 Schering Corporation Substituted piperidines that increase p53 activity and the uses thereof
US7671832B2 (en) 2006-07-10 2010-03-02 Philips Lumileds Lighting Company, Llc Multi-colored LED backlight with color-compensated clusters near edge
JP5347202B2 (en) 2007-03-28 2013-11-20 ファーマサイクリックス,インク. Breton tyrosine kinase inhibitor
AU2008262038A1 (en) 2007-06-08 2008-12-18 AbbVie Deutschland GmbH & Co. KG 5-heteroaryl substituted indazoles as kinase inhibitors
JP2010530421A (en) 2007-06-18 2010-09-09 シェーリング コーポレイション Heterocyclic compounds and their use as ERK inhibitors
US8658651B2 (en) 2009-09-30 2014-02-25 Merck Sharp & Dohme Corp. Compounds that are ERK inhibitors

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007070398A1 (en) * 2005-12-13 2007-06-21 Schering Corporation Polycyclic indazole derivatives that are erk inhibitors
WO2008153858A1 (en) * 2007-06-05 2008-12-18 Schering Corporation Polycyclic indazole derivatives and their use as erk inhibitors for the treatment of cancer

Cited By (42)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8546404B2 (en) 2005-12-13 2013-10-01 Merck Sharp & Dohme Compounds that are ERK inhibitors
US7807672B2 (en) 2006-02-16 2010-10-05 Schering Corporation Compounds that are ERK inhibitors
WO2010021978A3 (en) * 2008-08-19 2010-05-06 Schering Corporation Il-8 biomarker for monitoring cancer treatment
WO2010021978A2 (en) * 2008-08-19 2010-02-25 Schering Corporation Il-8 biomarker
US9229008B2 (en) 2008-08-19 2016-01-05 Merck Sharp & Dohme Corp. IL-8 level as a determinant of responsivity of a cancer to treatment
US8586543B2 (en) 2008-08-19 2013-11-19 Merck Sharp & Dohme Corp. IL-8 biomarker for monitoring cancer treatment with certain ERK inhibitors
WO2011002887A1 (en) 2009-07-02 2011-01-06 Schering Corporation FUSED TRICYCLIC COMPOUNDS AS NOVEL mTOR INHIBITORS
EP2584903A4 (en) * 2010-06-24 2014-12-31 Merck Sharp & Dohme Novel heterocyclic compounds as erk inhibitors
EP2584903A1 (en) * 2010-06-24 2013-05-01 Merck Sharp & Dohme Corp. Novel heterocyclic compounds as erk inhibitors
US9242981B2 (en) 2010-09-16 2016-01-26 Merck Sharp & Dohme Corp. Fused pyrazole derivatives as novel ERK inhibitors
WO2012036997A1 (en) * 2010-09-16 2012-03-22 Schering Corporation Fused pyrazole derivatives as novel erk inhibitors
US9173396B2 (en) 2010-10-22 2015-11-03 Bayer Intellectual Property Gmbh Heterocyclic compounds as pesticides
WO2012052412A1 (en) 2010-10-22 2012-04-26 Bayer Cropscience Ag Novel heterocyclic compounds as pesticides
US9000209B2 (en) 2011-07-22 2015-04-07 Iowa State University Research Foundation, Inc. Method of regioselective synthesis of substituted benzoates
US10519126B2 (en) 2012-03-01 2019-12-31 Array Biopharma Inc. Serine/threonine kinase inhibitors
EP2875001A4 (en) * 2012-07-18 2016-06-01 Sunshine Lake Pharma Co Ltd Nitrogenous heterocyclic derivatives and their application in drugs
CN105050598A (en) * 2012-09-28 2015-11-11 默沙东公司 Novel compounds that are erk inhibitors
CN105050598B (en) * 2012-09-28 2018-04-27 默沙东公司 New compound as ERK inhibitor
CN105377856A (en) * 2013-03-13 2016-03-02 豪夫迈·罗氏有限公司 Process for making benzoxazepin compounds
CN105377856B (en) * 2013-03-13 2018-05-22 豪夫迈·罗氏有限公司 The method for preparing benzo oxygen azepine * compounds
US8871966B2 (en) 2013-03-15 2014-10-28 Iowa State University Research Foundation, Inc. Regiospecific synthesis of terephthalates
US10301317B2 (en) 2013-10-03 2019-05-28 Kura Oncology, Inc. Inhibitors of ERK and methods of use
US9624228B2 (en) 2013-10-03 2017-04-18 Kura Oncology, Inc. Inhibitors of ERK and methods of use
US9951078B2 (en) 2013-10-03 2018-04-24 Kura Oncology, Inc. Inhibitors of ERK and methods of use
US9988397B2 (en) 2014-12-15 2018-06-05 Merck Sharp & Dohme Corp. ERK inhibitors
US9884048B2 (en) 2014-12-15 2018-02-06 Merck Sharp & Dohme Corp. ERK inhibitors
WO2016095088A1 (en) * 2014-12-15 2016-06-23 Merck Sharp & Dohme Corp. Erk inhibitors
WO2016095089A1 (en) * 2014-12-15 2016-06-23 Merck Sharp & Dohme Corp. Erk inhibitors
CN111662272A (en) * 2014-12-18 2020-09-15 默沙东公司 Compounds for pharmaceutical formulations
US11034673B2 (en) 2014-12-18 2021-06-15 Merck Sharp & Dohme Corp. (S)-N-(3-(6-isopropoxypyridin-3-yl)-1H-indazol-5-yl)-1-(2-(4-(4-(1-methyl-1H-1,2,4-triazol-3-yl)phenyl)-3,6-dihydropyridin-1(2H)-yl)-2-oxoethyl)-3-(methylthio)pyrrolidine-3-carboxamide compositions for pharmaceutical preparations
WO2016100147A1 (en) 2014-12-18 2016-06-23 Merck Sharp & Dohme Corp. (s)-n-(3-(6-isopropoxypyridin-3-yl)-1h-indazol-5-yl)-1-(2-(4-(4-(1-methyl-1h-1,2,4-triazol-3-yl)phenyl)-3,6-dihydropyridin-1(2h)-yl)-2-oxoethyl)-3- (methylthio)pyrrolidine-3-carboxamide compositions for pharmaceutical preparations
US10577348B2 (en) 2014-12-18 2020-03-03 Merck Sharp & Dohme Corp. (S)-N-(3-(6-isopropoxypyridin-3-yl)-1H-indazol-5-yl)-1-(2-(4-(4-(1-methyl-1H-1,2,4-triazol-3-yl)phenyl)-3,6-dihydropyridin-1(2H)-yl)-2-oxoethyl)-3-(methylthio)pyrrolidine-3-carboxamide compositions for pharmaceutical preparations
US10710982B2 (en) 2014-12-18 2020-07-14 Merck Sharp & Dohme Corp. (S)-N-(3-6-isopropoxypyridin-3-3YL)-1H-indazol-5-yl)-1-(2-(4-4(1-methyl-1H-1,2,4-triazol-3-yl)phenyl)-3,6-dihydropyridin-1(2H-yl)-2-oxoethyl)-3(methylthio)pyrrolidine-3-carboxamide compositions for pharmaceutical preparations
EP3998258A1 (en) 2014-12-18 2022-05-18 Merck Sharp & Dohme Corp. (s)-n-(3-(6-isopropoxypyridin-3-yl)-1h-indazol-5-yl)-1-(2-(4-(4-(1-methyl- 1h-1,2,4-triazol-3-yl)phenyl)-3,6-dihydropyridin-1(2h)-yl)-2-oxoethyl)-3- (methylthio)pyrrolidine-3-carboxamide compositions for pharmaceutical preparations
WO2016100152A1 (en) 2014-12-19 2016-06-23 Merck Sharp & Dohme Corp. (s)-n-(3-(6-isopropoxypyridin-3-yl)-1h-indazol-5-yl)-1-(2-(4-(4-(1-methyl-1h-1,2,4-triazol-3-yl)phenyl)-3,6-dihydropyridin-1(2h)-yl)-2-oxoethyl)-3-(methylthio)pyrrolidine-3-carboxamide compositions for pharmaceutical preparations
US10058544B2 (en) 2014-12-19 2018-08-28 Merck Sharp & Dohme Corp. (S)-N-(3-(6-isopropoxypyridin-3-yl)-1H-indazol-5-yl)-1-(2-(4-(4-(1-methyl-1H-1,2,4-triazol-3-yl)phenyl)-3,6-dihydropyridin-1(2H)-yl)-2-oxoethyl)-3-(methylthio)pyrrolidine-3-carboxamide compositions for pharmaceutical preparations
RU2712148C2 (en) * 2014-12-19 2020-01-24 Мерк Шарп И Доум Корп. (s)-n-(3-(6-isopropoxypyridin-3-yl)-1h-indazol-5-yl)-1-(2-(4-(4-(1-methyl-1h-1,2,4-triazol-3-yl)phenyl)-3,6-dihydropyridine-1(2n)-yl)-2-oxoethyl)-3-(methylthio)pyrrolidine-3-carboxamide compositions for pharmaceutical preparations
AU2015362849B2 (en) * 2014-12-19 2020-05-07 Merck Sharp & Dohme Corp. (S)-N-(3-(6-isopropoxypyridin-3-yl)-1H-indazol-5-yl)-1-(2-(4-(4-(1-methyl-1H-1,2,4-triazol-3-yl)phenyl)-3,6-dihydropyridin-1(2H)-yl)-2-oxoethyl)-3-(methylthio)pyrrolidine-3-carboxamide compositions for pharmaceutical preparations
CN106999474A (en) * 2014-12-19 2017-08-01 默沙东公司 (2 (4 (4 (the 1 methyl 1H 1 of (S) N (base of 3 (base of 6 isopropoxypyrid 3) 1H indazoles 5) 1 for pharmaceutical preparation, 2, the base of 4 triazole 3) phenyl) 3,6 dihydropyridine 1 (2H) bases) 2 oxoethyls) 3 (methyl mercapto) pyrrolidines, 3 formyl amine composition
US10525036B2 (en) 2015-04-03 2020-01-07 Recurium Ip Holdings, Llc Spirocyclic compounds
WO2021067266A1 (en) * 2019-10-01 2021-04-08 Recurium Ip Holdings, Llc Pyrrolidinyl-based compounds
WO2021069567A1 (en) 2019-10-09 2021-04-15 Bayer Aktiengesellschaft Novel heteroaryl-triazole compounds as pesticides

Also Published As

Publication number Publication date
IL207530A0 (en) 2010-12-30
TW200948799A (en) 2009-12-01
MY152271A (en) 2014-09-15
AU2009215534A1 (en) 2009-08-27
CO6300939A2 (en) 2011-07-21
CN102015693B (en) 2014-10-29
JP5276676B2 (en) 2013-08-28
ECSP10010415A (en) 2010-09-30
BRPI0908120A8 (en) 2017-10-24
AU2009215534A8 (en) 2014-07-10
CL2009000394A1 (en) 2011-02-11
ES2556353T3 (en) 2016-01-15
CN102015693A (en) 2011-04-13
RU2525389C2 (en) 2014-08-10
BRPI0908120A2 (en) 2017-08-22
TWI398441B (en) 2013-06-11
AR070460A1 (en) 2010-04-07
ES2556353T8 (en) 2017-10-13
CA2714479A1 (en) 2009-08-27
PE20091491A1 (en) 2009-09-25
SG188179A1 (en) 2013-03-28
AU2009215534B8 (en) 2014-07-10
US20110189192A1 (en) 2011-08-04
EP2260031B1 (en) 2015-10-07
RU2010138635A (en) 2012-03-27
AU2009215534A2 (en) 2010-12-23
NZ587504A (en) 2012-09-28
ZA201005909B (en) 2014-03-26
ZA201400396B (en) 2014-12-23
US8716483B2 (en) 2014-05-06
IL207530A (en) 2014-08-31
EP2260031A1 (en) 2010-12-15
AU2009215534B2 (en) 2014-06-26
MX2010009268A (en) 2010-09-14
JP2011513225A (en) 2011-04-28
KR20100117123A (en) 2010-11-02

Similar Documents

Publication Publication Date Title
EP2260031B1 (en) Compounds that are erk inhibitors
EP2483263B1 (en) Heterocyclic compounds that are erk inhibitors
EP1984331B1 (en) Pyrrolidine derivatives as erk inhibitors
AU2006326616B2 (en) Polycyclic indazole derivatives that are ERK inhibitors
EP2155722B1 (en) Polycyclic indazole derivatives and their use as erk inhibitors for the treatment of cancer
US20110038876A1 (en) Heterocyclic compounds and use thereof as erk inhibitors
US8546404B2 (en) Compounds that are ERK inhibitors
CA2590979A1 (en) Novel farnesyl protein transferase inhibitors and their use to treat cancer

Legal Events

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

Ref document number: 200980113870.7

Country of ref document: CN

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

Ref document number: 09712601

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 2714479

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 207530

Country of ref document: IL

WWE Wipo information: entry into national phase

Ref document number: 12010501874

Country of ref document: PH

WWE Wipo information: entry into national phase

Ref document number: 2009215534

Country of ref document: AU

Ref document number: 5162/CHENP/2010

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 587504

Country of ref document: NZ

Ref document number: 2010547732

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: MX/A/2010/009268

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: PI 2010003904

Country of ref document: MY

WWE Wipo information: entry into national phase

Ref document number: 2009712601

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2009215534

Country of ref document: AU

Date of ref document: 20090219

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 20107020896

Country of ref document: KR

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2010138635

Country of ref document: RU

Ref document number: 10116702

Country of ref document: CO

WWE Wipo information: entry into national phase

Ref document number: 12918099

Country of ref document: US

ENP Entry into the national phase

Ref document number: PI0908120

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20100820