US20140286902A1 - Combination therapy of hsp90 inhibitors with platinum-containing agents - Google Patents

Combination therapy of hsp90 inhibitors with platinum-containing agents Download PDF

Info

Publication number
US20140286902A1
US20140286902A1 US14/355,689 US201214355689A US2014286902A1 US 20140286902 A1 US20140286902 A1 US 20140286902A1 US 201214355689 A US201214355689 A US 201214355689A US 2014286902 A1 US2014286902 A1 US 2014286902A1
Authority
US
United States
Prior art keywords
cancer
optionally substituted
cisplatin
indol
triazole
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US14/355,689
Other languages
English (en)
Inventor
David Proia
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Synta Phamaceuticals Corp
Original Assignee
Synta Phamaceuticals Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Synta Phamaceuticals Corp filed Critical Synta Phamaceuticals Corp
Priority to US14/355,689 priority Critical patent/US20140286902A1/en
Publication of US20140286902A1 publication Critical patent/US20140286902A1/en
Assigned to SYNTA PHARMACEUTICALS CORP. reassignment SYNTA PHARMACEUTICALS CORP. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: PROIA, DAVID
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41961,2,4-Triazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/555Heterocyclic compounds containing heavy metals, e.g. hemin, hematin, melarsoprol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/661Phosphorus acids or esters thereof not having P—C bonds, e.g. fosfosal, dichlorvos, malathion or mevinphos
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/675Phosphorus compounds having nitrogen as a ring hetero atom, e.g. pyridoxal phosphate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • A61K33/243Platinum; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/28Compounds containing heavy metals
    • A61K31/282Platinum compounds
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41841,3-Diazoles condensed with carbocyclic rings, e.g. benzimidazoles

Definitions

  • chemotherapeutic agents act on a specific molecular target thought to be involved in the development of the malignant phenotype.
  • a complex network of signaling pathways regulate cell proliferation and the majority of malignant cancers are facilitated by multiple genetic abnormalities in these pathways. Therefore, it is less likely that a therapeutic agent that acts on one molecular target will be fully effective in curing a patient who has cancer.
  • HSPs Heat shock proteins
  • HSPs are a class of chaperone proteins that are up-regulated in response to elevated temperature and other environmental stresses, such as ultraviolet light, nutrient deprivation and oxygen deprivation. HSPs act as chaperones to other cellular proteins (called client proteins), facilitate their proper folding and repair and aid in the refolding of mis-folded client proteins.
  • client proteins cellular proteins
  • the Hsp90 family is one of the most abundant HSP families, accounting for about 1-2% of proteins in a cell that is not under stress and increasing to about 4-6% in a cell under stress. Inhibition of Hsp90 results in the degradation of its client proteins via the ubiquitin proteasome pathway.
  • the client proteins of Hsp90 are mostly protein kinases or transcription factors involved in signal transduction, and a number of its client proteins have been shown to be involved in the progression of cancer.
  • the invention provides a method of utilizing Hsp90 inhibitors according to formulae (I) or (Ia), or a compound in Tables 1 or 2 for the treatment of proliferative disorders such as cancer, in combination with a platinum-containing anti-cancer agent.
  • a method of treating a subject with cancer includes administering to the subject an Hsp90 inhibitor according to formulae (I) or (Ia), or a compound in Tables 1 or 2, and a platinum-containing anti-cancer agent.
  • the administration of the Hsp90 inhibitor and the platinum-containing agent are done concurrently.
  • the administration of the Hsp90 inhibitor and the platinum-containing agent are done sequentially.
  • the administration of the Hsp90 inhibitor and the platinum-containing agent are dosed independently.
  • the platinum-containing agent may be cisplatin, carboplatin, oxaliplatin, or their analogues.
  • the Hsp90 inhibitor may be a compound represented in Tables 1 or 2.
  • the platinum-containing agent may be cisplatin.
  • the cancer may have mutations or translocations in the EGFR, K-Ras, c-Metc-Met, c-Kit, HER2, B-Raf, PI3K and/or ALK proteins.
  • the cancer may express wild-type EGFR and K-Ras.
  • the cancer may express mutated EGFR and wild-type K-Ras.
  • the cancer may express wild-type EGFR and mutated K-Ras protein.
  • the cancer may be ALK positive (“ALK+”.)
  • the cancer may have an EML4-ALK translocation.
  • the cancer may have a HER2 mutation.
  • the cancer may have a PI#K mutation.
  • the cancer may have a B-Raf protein mutation.
  • kits for administration of the combination therapy include separate pharmaceutical compositions containing the Hsp90 inhibitor according to formulae (I) or (Ia) or a compound in Tables 1 or 2, and a platinum-containing anti-cancer agent.
  • the kit includes one pharmaceutical composition containing both the Hsp90 inhibitor and the platinum-containing anti-cancer agent.
  • each pharmaceutical composition includes one or more pharmaceutically acceptable carrier or diluent.
  • the platinum-containing agent may be cisplatin, carboplatin, oxaliplatin, or their analogues.
  • the Hsp90 inhibitor may be a compound represented in Tables 1 or 2.
  • the platinum-containing agent may be cisplatin.
  • the invention provides the use of an Hsp90 inhibitor according to formulae (I) or (Ia) or a compound in Tables 1 or 2 for the manufacture of a medicament for treating cancer in combination with a platinum-containing anti-cancer agent.
  • a method of treating drug-resistant cancer in a subject includes administering to the subject an effective amount of the pharmaceutical combination including an Hsp90 compound according to formulae (I) or (Ia) or a compound in Tables 1 or 2 and a platinum-containing anti-cancer agent.
  • the method further includes the administration of one or more therapeutic agents.
  • the combination treatment utilizes an Hsp90 compound according to formulae (I) or (Ia) or a compound in Tables 1 or 2 with a platinum-containing anti-cancer agent to help to arrest, partially or fully, or reduce the development of drug resistant cancer in a subject.
  • the combinations described herein may allow a reduced dose of the platinum-containing agent given to a subject, because the Hsp90 inhibitor should inhibit the development of multidrug-resistant cancer cells.
  • the platinum-containing anti-cancer agent may be cisplatin, carboplatin, oxaliplatin, or their derivatives.
  • the platinum-containing agent may be cisplatin.
  • FIG. 1 shows a dose-dependent curve with the IC 50 of ganetespib, at about 32 nM.
  • FIG. 2 shows a dose-dependent curve with the IC 50 of cisplatin at about 5.7 ⁇ M.
  • FIG. 3 shows significant killing of HCT-116 cells by ganetespib in combination with cisplatin. Cells were exposed to the indicated single agent or combination, concurrently, for 3 days.
  • FIG. 4 shows significant killing of HCT-116 cells by the sequential combination of ganetespib with cisplatin.
  • Cells were exposed to ganetespib for 1 hour, washed and then treated with vehicle (DMSO) or indicated chemotherapeutic for 3 days. Single agent chemotherapeutic was dosed for 3 days.
  • vehicle DMSO
  • FIG. 5 shows substantial treatment improvement by a combination of ganetespib with cisplatin in an animal model of HCT-116 colon cancer xenograft.
  • Ganetespib 150 mg/kg was given as a single bolus injection 6 hours prior to cisplatin (2 mg/kg) administration.
  • FIG. 6 shows substantial treatment improvement by a combination of ganetespib with cisplatin in an animal model of MDA-MB-231 breast cancer tumor xenograft.
  • Ganetespib 150 mg/kg was given as a single bolus injection 6 hours prior to cisplatin (2 mg/kg) administration.
  • alkyl means a saturated or unsaturated, straight chain or branched, non-cyclic hydrocarbon having from 1 to 10 carbon atoms.
  • Representative straight chain alkyls include methyl, ethyl, n-propyl, n-butyl, n-pentyl, n-hexyl, n-heptyl, n-octyl, n-nonyl and n-decyl; while representative branched alkyls include isopropyl, sec-butyl, isobutyl, tert-butyl, isopentyl, 2-methylbutyl, 3-methylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2-methylhexyl, 3-methylhexyl, 4-methylhexyl, 5-methylhexyl, 2,3-dimethylbutyl, 2,3-dimethylpentyl, 2,4-dimethylp
  • (C 1 -C 6 )alkyl means a saturated, straight chain or branched, non-cyclic hydrocarbon having from 1 to 6 carbon atoms.
  • Alkyl groups included in compounds described herein may be optionally substituted with one or more substituents.
  • unsaturated alkyls include vinyl, allyl, 1-butenyl, 2-butenyl, isobutylenyl, 1-pentenyl, 2-pentenyl, 3-methyl-1-butenyl, 2-methyl-2-butenyl, 2,3-dimethyl-2-butenyl, 1-hexenyl, 2-hexenyl, 3-hexenyl, 1-heptenyl, 2-heptenyl, 3-heptenyl, 1-octenyl, 2-octenyl, 3-octenyl, 1-nonenyl, 2-nonenyl, 3-nonenyl, 1-decenyl, 2-decenyl, 3-decenyl, acetylenyl, propynyl, 1-butynyl, 2-butynyl, 1-pentynyl, 2-pentynyl, 3-methyl-1-butynyl, 4-pentynyl, 1-hexynyl,
  • cycloalkyl means a saturated or unsaturated, mono- or polycyclic, non-aromatic hydrocarbon having from 3 to 20 carbon atoms.
  • Representative cycloalkyls include cyclopropyl, 1-methylcyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl, octahydropentalenyl, cyclohexenyl, cyclooctenyl, cyclohexynyl, and the like. Cycloalkyl groups included in compounds described herein may be optionally substituted with one or more substituents.
  • alkylene refers to an alkyl group that has two points of attachment.
  • (C 1 -C 6 )alkylene refers to an alkylene group that has from one to six carbon atoms.
  • Straight chain (C 1 -C 6 )alkylene groups are preferred.
  • Non-limiting examples of alkylene groups include methylene (—CH 2 —), ethylene (—CH 2 CH 2 —), n-propylene (—CH 2 CH 2 CH 2 —), isopropylene (—CH 2 CH(CH 3 )—), and the like.
  • Alkylene groups may be saturated or unsaturated, and may be optionally substituted with one or more substituents.
  • lower refers to a group having up to four atoms.
  • a “lower alkyl” refers to an alkyl radical having from 1 to 4 carbon atoms
  • “lower alkoxy” refers to “—O—(C 1 -C 4 )alkyl.
  • haloalkyl means an alkyl group, in which one or more, including all, the hydrogen radicals are replaced by a halo group(s), wherein each halo group is independently selected from —F, —Cl, —Br, and —I.
  • halomethyl means a methyl in which one to three hydrogen radical(s) have been replaced by a halo group.
  • Representative haloalkyl groups include trifluoromethyl, bromomethyl, 1,2-dichloroethyl, 4-iodobutyl, 2-fluoropentyl, and the like.
  • alkoxy is an alkyl group which is attached to another moiety via an oxygen linker. Alkoxy groups included in compounds described herein may be optionally substituted with one or more substituents.
  • haloalkoxy is a haloalkyl group which is attached to another moiety via an oxygen linker.
  • an “aromatic ring” or “aryl” means a mono- or polycyclic hydrocarbon, containing from 6 to 15 carbon atoms, in which at least one ring is aromatic.
  • suitable aryl groups include phenyl, tolyl, anthracenyl, fluorenyl, indenyl, azulenyl, and naphthyl, as well as benzo-fused carbocyclic moieties such as 5,6,7,8-tetrahydronaphthyl.
  • Aryl groups included in compounds described herein may be optionally substituted with one or more substituents.
  • the aryl group is a monocyclic ring, wherein the ring comprises 6 carbon atoms, referred to herein as “(C 6 )aryl.”
  • aralkyl means an aryl group that is attached to another group by a (C 1 -C 6 )alkylene group.
  • Representative aralkyl groups include benzyl, 2-phenyl-ethyl, naphth-3-yl-methyl and the like.
  • Aralkyl groups included in compounds described herein may be optionally substituted with one or more substituents.
  • heterocyclyl means a monocyclic or a polycyclic, saturated or unsaturated, non-aromatic ring or ring system which typically contains 5- to 20-members and at least one heteroatom.
  • a heterocyclic ring system can contain saturated ring(s) or unsaturated non-aromatic ring(s), or a mixture thereof.
  • a 3- to 10-membered heterocycle can contain up to 5 heteroatoms, and a 7- to 20-membered heterocycle can contain up to 7 heteroatoms.
  • a heterocycle has at least one carbon atom ring member.
  • Each heteroatom is independently selected from nitrogen, which can be oxidized (e.g., N(O)) or quaternized, oxygen and sulfur, including sulfoxide and sulfone.
  • the heterocycle may be attached via any heteroatom or carbon atom.
  • heterocycles include morpholinyl, thiomorpholinyl, pyrrolidinonyl, pyrrolidinyl, piperidinyl, piperazinyl, hydantoinyl, valerolactamyl, oxiranyl, oxetanyl, tetrahydrofuranyl, tetrahydropyranyl, tetrahydropyrindinyl, tetrahydropyrimidinyl, tetrahydrothiophenyl, tetrahydrothiopyranyl, and the like.
  • a heteroatom may be substituted with a protecting group known to those of ordinary skill in the art, for example, a nitrogen atom may be substituted with a tert-butoxycarbonyl group.
  • the heterocycle included in compounds described herein may be optionally substituted with one or more substituents. Only stable isomers of such substituted heterocyclic groups are contemplated in this definition.
  • heteroaryl means a monocyclic or a polycyclic, unsaturated radical containing at least one heteroatom, in which at least one ring is aromatic.
  • Polycyclic heteroaryl rings must contain at least one heteroatom, but not all rings of a polycyclic heteroaryl moiety must contain heteroatoms.
  • Each heteroatom is independently selected from nitrogen, which can be oxidized (e.g., N(O)) or quaternized, oxygen and sulfur, including sulfoxide and sulfone.
  • heteroaryl groups include pyridyl, 1-oxo-pyridyl, furanyl, benzo[1,3]dioxolyl, benzo[1,4]dioxinyl, thienyl, pyrrolyl, oxazolyl, imidazolyl, thiazolyl, an isoxazolyl, quinolinyl, pyrazolyl, isothiazolyl, pyridazinyl, pyrimidinyl, pyrazinyl, a triazinyl, triazolyl, thiadiazolyl, isoquinolinyl, indazolyl, benzoxazolyl, benzofuryl, indolizinyl, imidazopyridyl, tetrazolyl, benzimidazolyl, benzothiazolyl, benzothiadiazolyl, benzoxadiazolyl, indolyl, tetrahydroindolyl
  • the heteroaromatic ring may be a 5-8 membered monocyclic heteroaryl ring.
  • the point of attachment of a heteroaromatic or heteroaryl ring may be at either a carbon atom or a heteroatom.
  • Heteroaryl groups included in compounds described herein may be optionally substituted with one or more substituents.
  • (C 5 )heteroaryl means an heteroaromatic ring of 5 members, wherein at least one carbon atom of the ring is replaced with a heteroatom, such as, for example, oxygen, sulfur or nitrogen.
  • Representative (C 5 )heteroaryls include furanyl, thienyl, pyrrolyl, oxazolyl, imidazolyl, thiazolyl, isoxazolyl, pyrazolyl, isothiazolyl, pyrazinyl, triazolyl, thiadiazolyl, and the like.
  • the term “(C 6 )heteroaryl” means an aromatic heterocyclic ring of 6 members, wherein at least one carbon atom of the ring is replaced with a heteroatom such as, for example, oxygen, nitrogen or sulfur.
  • Representative (C 6 )heteroaryls include pyridyl, pyridazinyl, pyrazinyl, triazinyl, tetrazinyl, and the like.
  • heteroarylkyl means a heteroaryl group that is attached to another group by a (C 1 -C 6 )alkylene.
  • Representative heteroaralkyls include 2-(pyridin-4-yl)-propyl, 2-(thien-3-yl)-ethyl, imidazol-4-yl-methyl, and the like.
  • Heteroaralkyl groups included in compounds described herein may be optionally substituted with one or more substituents.
  • halogen or “halo” means —F, —Cl, —Br or —I.
  • Suitable substituents for an alkyl, alkylene, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, heterocyclyl, aryl, aralkyl, heteroaryl, and heteroaralkyl groups include are those substituents which form a stable compound described herein without significantly adversely affecting the reactivity or biological activity of the compound described herein.
  • substituents for an alkyl, alkylene, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, heterocyclyl, aryl, aralkyl, heteroaryl, and heteroaralkyl include an alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, heterocyclyl, aryl, heteroaryl, aralkyl, heteraralkyl, heteroalkyl, alkoxy, (each of which can be optionally and independently substituted), —C(O)NR 28 R 29 , —C(S)NR 28 R 29 , —C(NR 32 )NR 28 R 29 , —NR 33 C(O)R 31 , —NR 33 C(S)R 31 , —NR 33 C(NR 32 )R 31 , halo, —OR 33 , cyano, nitro, —C(O)R 33 , —C(O
  • any saturated portion of an alkyl, cycloalkyl, alkylene, heterocyclyl, alkenyl, cycloalkenyl, alkynyl, aralkyl and heteroaralkyl groups may also be substituted with ⁇ O, ⁇ S, or ⁇ N—R 32 .
  • Each R 28 and R 29 is independently H, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, heterocyclyl, aryl, heteroaryl, aralkyl, or heteraralkyl, wherein each alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, heterocyclyl, aryl, heteroaryl, aralkyl, or heteroalkyl represented by R 28 or R 29 is optionally and independently substituted.
  • Each R 30 , R 31 and R 33 is independently H, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, heterocyclyl, aryl, heteroaryl, aralkyl, or heteraralkyl, wherein each alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, heterocyclyl, aryl, heteroaryl, aralkyl, and heteraralkyl represented by R 30 or R 31 or R 33 is optionally and independently unsubstituted.
  • Each R 32 is independently H, alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, heterocyclyl, aryl, heteroaryl, aralkyl, heteraralkyl, —C(O)R 33 , —C(O)NR 28 R 29 , —S(O) k R 33 , or —S(O) k NR 28 R 29 , wherein each alkyl, alkenyl, alkynyl, cycloalkyl, cycloalkenyl, heterocyclyl, aryl, heteroaryl, aralkyl and heteraralkyl represented by R 32 is optionally and independently substituted.
  • variable k is 0, 1 or 2.
  • suitable substituents include C1-C4 alkyl, C1-C4 haloalkyl, C1-C4 alkoxy, C1-C4 haloalkoxy, C1-C4 hydroxyalkyl, halo, or hydroxyl.
  • heterocyclyl, heteroaryl or heteroaralkyl group When a heterocyclyl, heteroaryl or heteroaralkyl group contains a nitrogen atom, it may be substituted or unsubstituted. When a nitrogen atom in the aromatic ring of a heteroaryl group has a substituent, the nitrogen may be oxidized or a quaternary nitrogen.
  • the compounds described herein containing reactive functional groups also include corresponding protected derivatives thereof.
  • “Protected derivatives” are those compounds in which a reactive site or sites are blocked with one or more protecting groups.
  • suitable protecting groups for hydroxyl groups include benzyl, methoxymethyl, allyl, trimethylsilyl, tert-butyldimethylsilyl, acetate, and the like.
  • suitable amine protecting groups include benzyloxycarbonyl, tert-butoxycarbonyl, tert-butyl, benzyl and fluorenylmethyloxy-carbonyl (Fmoc).
  • thiol protecting groups examples include benzyl, tert-butyl, acetyl, methoxymethyl and the like.
  • Other suitable protecting groups are well known to those of ordinary skill in the art and include those found in T. W. G REENE , P ROTECTING G ROUPS IN O RGANIC S YNTHESIS , (John Wiley & Sons, Inc., 1981).
  • the term “compound(s) described herein” or similar terms refers to a compound of formulae (I), or (Ia) or a compound in Tables 1 or 2 or a tautomer or pharmaceutically acceptable salt thereof. Also included in the scope of the embodiments are a solvate, clathrate, hydrate, polymorph, prodrug, or protected derivative of a compound of formulae (I), or (Ia), or a compound in Tables 1 or 2.
  • the compounds described herein may contain one or more chiral centers and/or double bonds and, therefore, exist as stereoisomers, such as double-bond isomers (i.e., geometric isomers), enantiomers or diastereomers.
  • stereoisomers such as double-bond isomers (i.e., geometric isomers), enantiomers or diastereomers.
  • Each chemical structure shown herein, including the compounds described herein encompass all of the corresponding compound's enantiomers, diastereomers and geometric isomers, that is, both the stereochemically pure form (e.g., geometrically pure, enantiomerically pure, or diastereomerically pure) and isomeric mixtures (e.g., enantiomeric, diastereomeric and geometric isomeric mixtures).
  • one enantiomer, diastereomer or geometric isomer will possess superior activity or an improved toxicity or kinetic profile compared to other isomers. In those cases, such enantiomers, diastereomers and geometric isomers of compounds described herein are preferred.
  • solvates e.g., hydrates
  • Solvates refer to crystalline forms wherein solvent molecules are incorporated into the crystal lattice during crystallization.
  • Solvates may include water or nonaqueous solvents such as ethanol, isopropanol, DMSO, acetic acid, ethanolamine and ethyl acetate.
  • water is the solvent molecule incorporated into the crystal lattice of a solvate, it is typically referred to as a “hydrate”. Hydrates include stoichiometric hydrates as well as compositions containing variable amounts of water.
  • the compound including solvates thereof, may exist in crystalline forms, non-crystalline forms or a mixture thereof.
  • the compounds or solvates may also exhibit polymorphism (i.e., the capacity to occur in different crystalline forms). These different crystalline forms are typically known as “polymorphs.”
  • polymorphs typically known as “polymorphs.”
  • the disclosed compounds and solvates e.g., hydrates
  • Polymorphs have the same chemical composition but differ in packing, geometrical arrangement and other descriptive properties of the crystalline solid state. Polymorphs, therefore, may have different physical properties such as shape, density, hardness, deformability, stability and dissolution properties.
  • Polymorphs typically exhibit different melting points, IR spectra and X-ray powder diffraction patterns, which may be used for identification.
  • different polymorphs may be produced, for example, by changing or adjusting the conditions used in crystallizing the compound. For example, changes in temperature, pressure or solvent may result in different polymorphs.
  • one polymorph may spontaneously convert to another polymorph under certain conditions.
  • clathrates include inclusion compounds, of the compound or its pharmaceutically acceptable salt, solvate or polymorph, are also included.
  • “Clathrate” means a compound described herein, or a salt thereof, in the form of a crystal lattice that contains spaces (e.g., channels) that have a guest molecule trapped within (e.g., a solvent or water).
  • prodrug means a derivative of a compound that can hydrolyze, oxidize, or otherwise react under biological conditions (in vitro or in vivo) to provide a compound described herein. Prodrugs may become active upon such reaction under biological conditions, or they may have activity in their unreacted forms.
  • prodrugs contemplated herein include analogs or derivatives of compounds of formulae (I) or (Ia) or a compound in Tables 1 or 2 that comprise biohydrolyzable moieties such as biohydrolyzable amides, biohydrolyzable esters, biohydrolyzable carbamates, biohydrolyzable carbonates, biohydrolyzable ureides and phosphate analogues.
  • Prodrugs can typically be prepared using well-known methods, such as those described by B URGER'S M EDICINAL C HEMISTRY AND D RUG D ISCOVERY , (Manfred E. Wolff Ed., 5th ed. (1995)) 172-178, 949-982.
  • an element means one element or more than one element.
  • the term “about” is understood as within a range of normal tolerance in the art, for example within 2 standard deviations of the mean. About can be understood as within 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, 0.1%, 0.05%, or 0.01% of the stated value. Unless otherwise clear from context, all numerical values provided herein can be modified by the term about.
  • the terms “subject”, “patient” and “mammal” are used interchangeably.
  • the terms “subject” and “patient” refer to an animal (e.g., a bird such as a chicken, quail or turkey, or a mammal), preferably a mammal including a non-primate (e.g., a cow, pig, horse, sheep, rabbit, guinea pig, rat, cat, dog, and mouse) and a primate (e.g., a monkey, chimpanzee and a human), and more preferably a human.
  • a non-primate e.g., a cow, pig, horse, sheep, rabbit, guinea pig, rat, cat, dog, and mouse
  • a primate e.g., a monkey, chimpanzee and a human
  • the subject is a non-human animal such as a farm animal (e.g., a horse, cow, pig or sheep), or a pet (e.g., a dog, cat, guinea pig or rabbit). In another embodiment, the subject is a human.
  • a farm animal e.g., a horse, cow, pig or sheep
  • a pet e.g., a dog, cat, guinea pig or rabbit.
  • the subject is a human.
  • Hsp90 includes each member of the family of heat shock proteins having a mass of about 90-kiloDaltons.
  • the highly conserved Hsp90 family includes the cytosolic Hsp90 ⁇ and Hsp90 ⁇ isoforms, as well as GRP94, which is found in the endoplasmic reticulum, and HSP75/TRAP1, which is found in the mitochondrial matrix.
  • c-Kit or “c-Kit kinase” refers to a membrane receptor protein tyrosine kinase which is preferably activated upon binding Stem Cell Factor (SCF) to its extracellular domain.
  • SCF Stem Cell Factor
  • c-Kit or “c-Kit kinase” and include those that fall into two classes: (1) having a single amino acid substitution at codon 816 of the human c-Kit kinase, or its equivalent position in other species (Ma, et al., J. Invest Dermatol ., (1999) 112:165-170), and (2) those which have mutations involving the putative juxtamembrane z-helix of the protein (Ma, et al., J. Biol. Chem ., (1999) 274:13399-13402). Both of these publications are incorporated by reference herein in their entirety, including any drawings.
  • BCR-ABL is a fusion protein that results from the translocation of gene sequences from c-ABL protein tyrosine kinase on chromosome 9 into BCR sequences on chromosome 22 producing the Philadelphia chromosome.
  • BCR-ABL fusion proteins can vary in size from 185-230 kD but they must contain at least the OLI domain from BCR and the TK domain from ABL for transforming activity.
  • the most common BCR-ABL gene products found in humans are P230 BCR-ABL, P210 BCR-ABL and P190 BCR-ABL.
  • P210 BCR-ABL is characteristic of CML and P190 BCR-ABL is characteristic of ALL.
  • FLT3 kinase is a tyrosine kinase receptor involved in the regulation and stimulation of cellular proliferation. Gilliland, et al., Blood (2002), 100:1532-42.
  • the FLT3 kinase has five immunoglobulin-like domains in its extracellular region, as well as an insert region of 75-100 amino acids in the middle of its cytoplasmic domain. FLT3 kinase is activated upon the binding of the FLT3 ligand which causes receptor dimerization.
  • Dimerization of the FLT3 kinase by FLT3 ligand activates the intracellular kinase activity as well as a cascade of downstream substrates including Stat5, Ras, phosphatidylinositol-3-kinase (PI3K), Erk2, Akt, MAPK, SHC, SHP2 and SHIP. Rosnet, et al., Acta Haematol . (1996), 95:218; Hayakawa, et al., Oncogene (2000), 19:624; Mizuki, et al., Blood (2000), 96:3907; Gilliand, et al., Curr. Opin. Hematol . (2002), 9: 274-81. Both membrane-bound and soluble FLT3 ligand bind, dimerize, and subsequently activate the FLT3 kinase.
  • Normal cells that express FLT3 kinase include immature hematopoietic cells, typically CD34+ cells, placenta, gonads and brain. Rosnet, et al., Blood (1993), 82:1110-19; Small, et al., Proc. Natl. Acad. Sci. U.S.A . (1994), 91:459-63; Rosnet, et al., Leukemia (1996), 10:238-48.
  • efficient stimulation of proliferation via FLT3 kinase typically requires other hematopoietic growth factors or interleukins.
  • FLT3 kinase also plays a critical role in immune function through its regulation of dendritic cell proliferation and differentiation.
  • FLT3 kinase Numerous hematologic malignancies express FLT3 kinase, the most prominent of which is AML. Yokota, et al., Leukemia (1997), 11:1605-09. Other FLT3 expressing malignancies include B-precursor cell acute lymphoblastic leukemias, myelodysplastic leukemias, T-cell acute lymphoblastic leukemias, and chronic myelogenous leukemias. Rasko, et al., Leukemia (1995), 9:2058-66.
  • FLT3 kinase mutations associated with hematologic malignancies are activating mutations.
  • the FLT3 kinase is constitutively activated without the need for binding and dimerization by FLT3 ligand, and therefore stimulates the cell to grow continuously.
  • Two types of activating mutations have been identified: internal tandem duplications (ITDs) and point mutation in the activating loop of the kinase domain.
  • ITDs internal tandem duplications
  • FLT3 kinase refers to both wild type FLT3 kinase and mutant FLT3 kinases, such as FLT3 kinases that have activating mutations.
  • Inappropriate FLT3 activity includes enhanced FLT3 activity resulting from increased or de novo expression of FLT3 in cells, increased FLT3 expression or activity, and FLT3 mutations resulting in constitutive activation.
  • the existence of inappropriate or abnormal FLT3 ligand and FLT3 levels or activity can be determined using well-known methods in the art. For example, abnormally high FLT3 levels can be determined using commercially available ELISA kits. FLT3 levels can also be determined using flow cytometric analysis, immunohistochemical analysis and in situ hybridization techniques.
  • Epidermal growth factor receptor or “EGFR”, as used herein, means any epidermal growth factor receptor (EGFR) protein, peptide, or polypeptide having EGFR or EGFR family activity (e.g., Her1, Her2, Her3 and/or Her4), such as encoded by EGFR Genbank Accession Nos. shown in Table I of U.S. patent application Ser. No. 10/923,354, filed on Aug. 20, 2004, or any other EGFR transcript derived from a EGFR gene and/or generated by EGFR translocation.
  • EGFR epidermal growth factor receptor
  • EGFR is also meant to include other EGFR proteins, peptides, or polypeptides derived from EGFR isoforms (e.g., Her1, Her2, Her3 and/or Her4), mutant EGFR genes, splice variants of EGFR genes, and EGFR gene polymorphisms.
  • EGFR is a member of the type 1 subgroup of receptor tyrosine kinase family of growth factor receptors which play critical roles in cellular growth, differentiation and survival. Activation of these receptors typically occurs via specific ligand binding which results in hetero- or homodimerization between receptor family members, with subsequent autophosphorylation of the tyrosine kinase domain.
  • Specific ligands which bind to EGFR include epidermal growth factor (EGF), transforming growth factor ⁇ (TGF ⁇ ), amphiregulin and some viral growth factors.
  • EGFR Activation of EGFR triggers a cascade of intracellular signaling pathways involved in both cellular proliferation (the ras/raf/MAP kinase pathway) and survival (the PI3 kinase/Akt pathway).
  • ras/raf/MAP kinase pathway the ras/raf/MAP kinase pathway
  • survival the PI3 kinase/Akt pathway
  • EGFR Aberrant or overexpression of EGFR has been associated with an adverse prognosis in a number of human cancers, including head and neck, breast, colon, prostate, lung (e.g., NSCLC, adenocarcinoma and squamous lung cancer), ovarian, gastrointestinal cancers (gastric, colon, pancreatic), renal cell cancer, bladder cancer, glioma, gynecological carcinomas and prostate cancer.
  • overexpression of tumor EGFR has been correlated with both chemoresistance and a poor prognosis.
  • Mutations in EGFR are associated with many types of cancer as well. For example, EGFR mutations are highly prevalent in non-mucinous BAC patients. Finberg, et al., J. Mol. Diagnostics (2007) 9(3):320
  • c-Met is a receptor tyrosine kinase that is encoded by the Met protooncogene and transduces the biological effects of hepatocyte growth factor (HGF), which is also referred to as scatter factor (SF).
  • HGF hepatocyte growth factor
  • SF scatter factor
  • c-Met and HGF are required for normal mammalian development and have been shown to be important in cell migration, cell proliferation and survival, morphogenic differentiation, and organization of 3-dimensional tubular structures (e.g., renal tubular cells, gland formation, etc.).
  • the c-Met receptor has been shown to be expressed in a number of human cancers.
  • c-Met and its ligand, HGF have also been shown to be co-expressed at elevated levels in a variety of human cancers (particularly sarcomas).
  • c-Met signaling is most commonly regulated by tumor-stroma (tumor-host) interactions.
  • c-Met gene amplification, mutation, and rearrangement have been observed in a subset of human cancers. Families with germine mutations that activate c-Met kinase are prone to multiple kidney tumors as well as tumors in other tissues. Numerous studies have correlated the expression of c-Met and/or HGF/SF with the state of disease progression of different types of cancer (including lung, colon, breast, prostate, liver, pancreas, brain, kidney, ovaries, stomach, skin, and bone cancers). Furthermore, the overexpression of c-Met or HGF have been shown to correlate with poor prognosis and disease outcome in a number of major human cancers including lung, liver, gastric, and breast.
  • the anaplastic lymphoma kinase (ALK) tyrosine kinase receptor is an enzyme that, in humans, is encoded by the ALK gene.
  • the 2;5 chromosomal translocation is frequently associated with anaplastic large cell lymphomas (ALCLs).
  • the translocation creates a fusion gene consisting of the ALK (anaplastic lymphoma kinase) gene and the nucleophosmin (NPM) gene: the 3′ half of ALK, derived from chromosome 2, is fused to the 5′ portion of NPM from chromosome 5.
  • the product of the NPM-ALK fusion gene is oncogenic.
  • Other possible translocations of the ALK gene such as the em14 translocation, are also implicated in cancer.
  • ALK anaplastic lymphoma kinase
  • EML4-ALK and KIF5B-ALK translocations have been found in non-small cell lung cancer. See. e.g. Mano H., Cancer Sci. 2008 December;99(12):2349-55; Takeuchi K et al., Clin Cancer Res. 2009 May 1; 15(9):3143-9.
  • CLTC-ALK mutation has been found in DLBCL. See e.g. Rudzki Z et al., Pol J Pathol. 2005; 56 (1):37-45.
  • NPM-ALK, MSN-ALK, and other mutations have been found in ALCL. See e.g. Lamant L et al., Genes Chromosomes Cancer.
  • TPM4-ALK mutation has been found in esophageal squamous cell carcinoma (ESCC). See e.g. Li R, Morris S W., Med Res Rev. 2008 May; 28 (3):372-412. F1174L, R1275Q, and other point mutations have been found in NBL. See e.g. van Roy N et al. Genome Med 2009 July 27; 1 (7):74.
  • TPM3-ALK, TPM4-ALK, CLTC-ALK, RanBP2-ALK, and TPM4-ALK mutations have been found in IMT. See e.g.
  • the KRAS oncogene (the cellular homolog of the Kirsten rat sarcoma virus gene) is a critical gene in the development of a variety of cancers, and the mutation status of this gene is an important characteristic of many cancers. Mutation status of the gene can provide diagnostic, prognostic and predictive information for several cancers.
  • the KRAS gene is a member of a family of genes (KRAS, NRAS and HRAS). KRAS is a member of the RAS family of oncogenes, a collection of small guanosine triphosphate (GTP)-binding proteins that integrate extracellular cues and activate intracellular signaling pathways to regulate cell proliferation, differentiation, and survival.
  • GTP small guanosine triphosphate
  • KRAS Gain-of-function mutations that confer transforming capacity are frequently observed in KRAS, predominantly arising as single amino acid substitutions at amino acid residues G12, G13 or Q61. Constitutive activation of KRAS leads to the persistent stimulation of downstream signaling pathways that promote tumorigenesis, including the RAF/MEK/ERK and PI3K/AKT/mTOR cascades.
  • KRAS mutations are highly prevalent (20-30%) and are associated with unfavorable clinical outcomes. Mutations in KRAS appear mutually exclusive with those in EGFR in NSCLC tumors; more importantly, they can account for primary resistance to targeted EGFR TKI therapies. Mutations in the KRAS gene are common in many types of cancer, including pancreatic cancer ( ⁇ 65%), colon cancer ( ⁇ 40%), lung cancer ( ⁇ 20%) and ovarian cancer ( ⁇ 15%).
  • Thresholds of increased expression that constitute an EGFR mutation or an ALK mutation are well known in the art. Moreover, it is generally recognized that once an EGFR mutation is detected in a cancer, the KRAS mutation will be eliminated in the same cancer. Put reversely, if a KRAS mutation is positively identified in a cancer from a subject, it is then not necessary to engage in any further EGFR-related identification. Similar principle can be applied to an ALK mutation in a cancer.
  • a “proliferative disorder” or a “hyperproliferative disorder,” and other equivalent terms, means a disease or medical condition involving pathological growth of cells.
  • Proliferative disorders include cancer, smooth muscle cell proliferation, systemic sclerosis, cirrhosis of the liver, adult respiratory distress syndrome, idiopathic cardiomyopathy, lupus erythematosus, retinopathy, (e.g., diabetic retinopathy or other retinopathies), cardiac hyperplasia, reproductive system associated disorders such as benign prostatic hyperplasia and ovarian cysts, pulmonary fibrosis, endometriosis, fibromatosis, harmatomas, lymphangiomatosis, sarcoidosis and desmoid tumors.
  • Non-cancerous proliferative disorders also include hyperproliferation of cells in the skin such as psoriasis and its varied clinical forms, Reiter's syndrome, pityriasis rubra pilaris, hyperproliferative variants of disorders of keratinization (e.g., actinic keratosis, senile keratosis), scleroderma, and the like.
  • the proliferative disorder is cancer.
  • the invention provides a method of treating a proliferative disorder in a subject, comprising administering to the subject an effective amount of the combination of Hsp90 inhibitor and platinum-containing anti-cancer agent as described herein.
  • the proliferative disorder is cancer.
  • the cancer is breast cancer, gastric cancer, colorectal cancer, pancreatic cancer, ocular melanoma, prostate cancer, gastrointestinal stromal tumors (GIST), advanced esophagogastric cancer, melanoma, hepatocellular cancer, solid tumor, liver cancer, head and neck cancer, small cell lung cancer, non-small cell lung cancer, bladder cancer, testicular tumor, ovarian cancer, lymphoma, leukemia, multiple myeloma, or colon cancer.
  • the cancer is solid cancer, gastric cancer, bladder cancer, ovarian cancer, or colorectal cancer.
  • the cancer is colon cancer.
  • the cancer is metastatic colorectal cancer.
  • the cancer is bladder cancer. In an embodiment, the cancer is solid cancer. In an embodiment, the cancer is gastric cancer. In an embodiment, the cancer may have a mutation or translocation in EGFR, K-Ras, PI3K, ALK, HER2 and/or B-Raf proteins.
  • Some of the disclosed methods can be particularly effective at treating subjects whose cancer has become “drug resistant” or “multi-drug resistant”.
  • a cancer which initially responded to an anti-cancer drug becomes resistant to the anti-cancer drug when the anti-cancer drug is no longer effective in treating the subject with the cancer.
  • many tumors will initially respond to treatment with an anti-cancer drug by decreasing in size or even going into remission, only to develop resistance to the drug.
  • “Drug resistant” tumors are characterized by a resumption of their growth and/or reappearance after having seemingly gone into remission, despite the administration of increased dosages of the anti-cancer drug.
  • Cancers that have developed resistance to two or more anti-cancer drugs are said to be “multi-drug resistant”. For example, it is common for cancers to become resistant to three or more anti-cancer agents, often five or more anti-cancer agents and at times ten or more anti-cancer agents.
  • anti-proliferative or anti-cancer therapies may be combined with the pharmaceutical combination of this invention to treat proliferative diseases and cancer.
  • Other therapies or anti-cancer agents that may be used in combination with the inventive anti-cancer agents of the present invention include surgery, radiotherapy (including, but not limited to, gamma-radiation, neutron beam radiotherapy, electron beam radiotherapy, proton therapy, brachytherapy, and systemic radioactive isotopes), endocrine therapy, biologic response modifiers (including, but not limited to, interferons, interleukins, and tumor necrosis factor (TNF)), hyperthermia and cryotherapy, agents to attenuate any adverse effects (e.g., antiemetics), and other approved chemotherapeutic drugs.
  • radiotherapy including, but not limited to, gamma-radiation, neutron beam radiotherapy, electron beam radiotherapy, proton therapy, brachytherapy, and systemic radioactive isotopes
  • endocrine therapy including, but not limited to, inter
  • the pharmaceutical combination of the invention is administered with one or more therapeutic agent selected from DFMO, vandetanib, trastuzumab, temodar, dexamethasone, epirubicin, ifosfamide, mitoxantrone, vorinostat, interferon alpha, rituximab, prednisone, cyclophosphamide, bendamustine, adriamycin, valproate, celecoxib, thalidomide, nelarabine, methotrexate, filgrastim, gemtuzumab ozogamicin, testosterone, clofarabine, cytarabine, everolimus, rituxumab, busulfan, capecitabine, pegfilgrastim, mesna, amrubicin, obatoclax, gefitinib, cyclosporine, dasatinib, temozolomide, thiotepa, pler
  • the one or more therapeutic agent is selected from erlotinib, bevacizumab, bortezomib, paclitaxel, doxorubicin, docetaxel, mitoxantrone, cytarabine, 5-fluorouracil, leucovorin, pemetrexed and vincristine.
  • the term “pharmaceutically acceptable salt” refers to a salt prepared from a compound of formulae (I) or (Ia) or a compound in Tables 1 or 2 having an acidic functional group, such as a carboxylic acid functional group, and a pharmaceutically acceptable inorganic or organic base.
  • Suitable bases include hydroxides of alkali metals such as sodium, potassium, and lithium; hydroxides of alkaline earth metal such as calcium and magnesium; hydroxides of other metals, such as aluminum and zinc; ammonia, and organic amines, such as unsubstituted or hydroxy-substituted mono-, di-, or trialkylamines; dicyclohexylamine; tributyl amine; pyridine; N-methyl,N-ethylamine; diethylamine; triethylamine; mono-, bis-, or tris-(2-hydroxy-lower alkyl amines), such as mono-, bis-, or tris-(2-hydroxyethyl)amine, 2-hydroxy-tert-butylamine, or tris-(hydroxymethyl)methylamine, N,N,-di-lower alkyl-N-(hydroxy lower alkyl)-amines, such as N,N-dimethyl-N-(2-hydroxyethyl)amine, or
  • pharmaceutically acceptable salt also refers to a salt prepared from a compound of formulae (I) or (Ia) or a compound in Tables 1 or 2 having a basic functional group, such as an amine functional group, and a pharmaceutically acceptable inorganic or organic acid.
  • Suitable acids include hydrogen sulfate, citric acid, acetic acid, oxalic acid, hydrochloric acid (HCl), hydrogen bromide (HBr), hydrogen iodide (HI), nitric acid, hydrogen bisulfide, phosphoric acid, isonicotinic acid, oleic acid, tannic acid, pantothenic acid, saccharic acid, lactic acid, salicylic acid, tartaric acid, bitartratic acid, ascorbic acid, succinic acid, maleic acid, besylic acid, fumaric acid, gluconic acid, glucaronic acid, formic acid, benzoic acid, glutamic acid, methanesulfonic acid, ethanesulfonic acid, benzenesulfonic acid, pamoic acid and p-toluenesulfonic acid.
  • solvate is a solvate formed from the association of one or more pharmaceutically acceptable solvent molecules to one of the compounds of formulae (I) or (Ia) or a compound in Tables 1 or 2.
  • solvate includes hydrates, e.g., hemihydrate, monohydrate, dihydrate, trihydrate, tetrahydrate, and the like.
  • a pharmaceutically acceptable carrier may contain inert ingredients which do not unduly inhibit the biological activity of the compound(s) described herein.
  • the pharmaceutically acceptable carriers should be biocompatible, i.e., non-toxic, non-inflammatory, non-immunogenic and devoid of other undesired reactions upon the administration to a subject. Standard pharmaceutical formulation techniques can be employed, such as those described in R EMINGTON , J. P., R EMINGTON'S P HARMACEUTICAL S CIENCES (Mack Pub. Co., 17th ed., 1985).
  • Suitable pharmaceutical carriers for parenteral administration include, for example, sterile water, physiological saline, bacteriostatic saline (saline containing about 0.9% mg/ml benzyl alcohol), phosphate-buffered saline, Hank's solution, Ringer's-lactate, and the like.
  • Methods for encapsulating compositions, such as in a coating of hard gelatin or cyclodextran, are known in the art. See B AKER, ET AL ., C ONTROLLED R ELEASE OF B IOLOGICAL A CTIVE A GENTS , (John Wiley and Sons, 1986).
  • the term “effective amount” refers to an amount of a compound described herein which is sufficient to reduce or ameliorate the severity, duration, progression, or onset of a disease or disorder, delay onset of a disease or disorder, retard or halt the advancement of a disease or disorder, cause the regression of a disease or disorder, prevent or delay the recurrence, development, onset or progression of a symptom associated with a disease or disorder, or enhance or improve the therapeutic effect(s) of another therapy.
  • the disease or disorder is a proliferative disorder.
  • the precise amount of compound administered to a subject will depend on the mode of administration, the type and severity of the disease or condition and on the characteristics of the subject, such as general health, age, sex, body weight and tolerance to drugs. For example, for a proliferative disease or disorder, determination of an effective amount will also depend on the degree, severity and type of cell proliferation. The skilled artisan will be able to determine appropriate dosages depending on these and other factors.
  • an “effective amount” of any additional therapeutic agent(s) will depend on the type of drug used.
  • Suitable dosages are known for approved therapeutic agents and can be adjusted by the skilled artisan according to the condition of the subject, the type of condition(s) being treated and the amount of a compound of the invention being used. In cases where no amount is expressly noted, an effective amount should be assumed. Non-limiting examples of an effective amount of a compound described herein are provided herein below.
  • the invention provides a method of treating, managing, or ameliorating a disease or disorder, e.g.
  • a proliferative disorder or one or more symptoms thereof, the method comprising administering to a subject in need thereof a dose of the Hsp90 inhibitor at least 150 ⁇ g/kg, at least 250 ⁇ g/kg, at least 500 ⁇ g/kg, at least 1 mg/kg, at least 5 mg/kg, at least 10 mg/kg, at least 25 mg/kg, at least 50 mg/kg, at least 75 mg/kg, at least 100 mg/kg, at least 125 mg/kg, at least 150 mg/kg, or at least 200 mg/kg or more of one or more compounds described herein once every day, once every 2 days, once every 3 days, once every 4 days, once every 5 days, once every 6 days, once every 7 days, once every 8 days, once every 10 days, once every two weeks, once every three weeks, or once a month.
  • the dosage of the individual platinum-containing anti-cancer agent used in the pharmaceutical combination may be equal to or lower than the dose of an individual therapeutic agent when given independently to treat, manage, or ameliorate a disease or disorder, or one or more symptoms thereof.
  • the disease or disorder being treated with a combination therapy is a proliferative disorder.
  • the proliferative disorder is cancer.
  • the platinum-containing anticancer agent cisplatin is administered IV at a dose of between about 20 mg/m 2 to about 150 mg/m 2 .
  • cisplatin is administered once daily.
  • cisplatin is administered IV at 20 mg/m 2 once daily.
  • cisplatin is administered IV at a dose of between about 75 mg/m 2 to about 100 mg/m 2 .
  • a treatment cycle can last between one and 6 weeks.
  • the recommended dosages of therapeutic agents currently used for the treatment, management, or amelioration of a disease or disorder, or one or more symptoms thereof, can obtained from any reference in the art.
  • the terms “treat”, “treatment”, “inhibiting”, and “treating” refer to the reduction or amelioration of the progression, severity and/or duration of a disease or disorder, delay of the onset of a disease or disorder, or the amelioration of one or more symptoms (preferably, one or more discernible symptoms) of a disease or disorder, resulting from the administration of one or more therapies (e.g., one or more therapeutic agents such as a compound of the invention).
  • the terms “treat”, “treatment” and “treating” also encompass the reduction of the risk of developing a disease or disorder, and the delay or inhibition of the recurrence of a disease or disorder.
  • the disease or disorder being treated is a proliferative disorder such as cancer.
  • the terms “treat”, “treatment” and “treating” refer to the amelioration of at least one measurable physical parameter of a disease or disorder, such as growth of a tumor, not necessarily discernible by the patient.
  • the terms “treat”, “treatment” and “treating” refer to the inhibition of the progression of a disease or disorder, e.g., a proliferative disorder, either physically by the stabilization of a discernible symptom, physiologically by the stabilization of a physical parameter, or both.
  • the terms “treat”, “treatment” and “treating” of a proliferative disease or disorder refers to the reduction or stabilization of tumor size or cancerous cell count, and/or delay of tumor formation.
  • the terms “treat”, “treating” and “treatment” also encompass the administration of a compound described herein as a prophylactic measure to patients with a predisposition (genetic or environmental) to any disease or disorder described herein.
  • a therapeutic agent refers to any agent(s) that can be used in the treatment of a disease or disorder, e.g. a proliferative disorder, or one or more symptoms thereof.
  • the term “therapeutic agent” refers to a compound described herein.
  • the term “therapeutic agent” does not refer to a compound described herein.
  • a therapeutic agent is an agent that is known to be useful for, or has been or is currently being used for the treatment of a disease or disorder, e.g., a proliferative disorder, or one or more symptoms thereof.
  • the term “synergistic” refers to a combination of a compound described herein and another therapeutic agent, which, when taken together, is more effective than the additive effects of the individual therapies.
  • a synergistic effect of a combination of therapies permits the use of lower dosages of one or more of the therapeutic agent(s) and/or less frequent administration of the agent(s) to a subject with a disease or disorder, e.g., a proliferative disorder.
  • the ability to utilize lower the dosage of one or more therapeutic agent and/or to administer the therapeutic agent less frequently reduces the toxicity associated with the administration of the agent to a subject without reducing the efficacy of the therapy in the treatment of a disease or disorder.
  • a synergistic effect can result in improved efficacy of agents in the prevention, management or treatment of a disease or disorder, e.g. a proliferative disorder.
  • a synergistic effect of a combination of therapies may avoid or reduce adverse or unwanted side effects associated with the use of either therapeutic agent alone.
  • side effects encompasses unwanted and adverse effects of a therapeutic agent. Side effects are always unwanted, but unwanted effects are not necessarily adverse. An adverse effect from a therapeutic agent might be harmful or uncomfortable or risky to a subject. Side effects include fever, chills, lethargy, gastrointestinal toxicities (including gastric and intestinal ulcerations and erosions), nausea, vomiting, neurotoxicities, nephrotoxicities, renal toxicities (including such conditions as papillary necrosis and chronic interstitial nephritis), hepatic toxicities (including elevated serum liver enzyme levels), myelotoxicities (including leukopenia, myelosuppression, thrombocytopenia and anemia), dry mouth, metallic taste, prolongation of gestation, weakness, somnolence, pain (including muscle pain, bone pain and headache), hair loss, asthenia, dizziness, extra-pyramidal symptoms, akathisia, cardiovascular disturbances and sexual dysfunction.
  • the term “in combination” refers to the use of more than one therapeutic agent.
  • the use of the term “in combination” does not restrict the order in which the therapeutic agents are administered to a subject with a disease or disorder, e.g., a proliferative disorder.
  • a first therapeutic agent such as a compound described herein, can be administered prior to (e.g., 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks before), concomitantly with, or subsequent to (e.g., 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks after) the administration of a second therapeutic agent, such as an anti-cancer agent, to a subject with a disease or disorder, e.g.
  • a second therapeutic agent such as an anti-cancer agent
  • the Hsp90 inhibitor and the platinum-containing anti-cancer agent are dosed on independent schedules. In another embodiment, the Hsp90 inhibitor and the platinum-containing anti-cancer agent are dosed on approximately the same schedule. In another embodiment, the Hsp90 inhibitor and the platinum-containing anticancer agent are dosed concurrently or sequentially on the same day.
  • therapies can refer to any protocol(s), method(s), and/or agent(s) that can be used in the prevention, treatment, management, or amelioration of a disease or disorder, e.g., a proliferative disorder, or one or more symptoms thereof.
  • a disease or disorder e.g., a proliferative disorder, or one or more symptoms thereof.
  • a “protocol” includes dosing schedules and dosing regimens.
  • the protocols herein are methods of use and include therapeutic protocols.
  • composition that “substantially” comprises a compound means that the composition contains more than about 80% by weight, more preferably more than about 90% by weight, even more preferably more than about 95% by weight, and most preferably more than about 97% by weight of the compound.
  • a “racemic mixture” means about 50% of one enantiomer and about 50% of is corresponding enantiomer of the molecule.
  • the combination encompasses all enantiomerically-pure, enantiomerically-enriched, diastereomerically pure, diastereomerically enriched, and racemic mixtures of the compounds described herein.
  • Enantiomeric and diastereomeric mixtures can be resolved into their component enantiomers or diastereomers by well-known methods, such as chiral-phase gas chromatography, chiral-phase high performance liquid chromatography, crystallizing the compound as a chiral salt complex, or crystallizing the compound in a chiral solvent.
  • Enantiomers and diastereomers can also be obtained from diastereomerically- or enantiomerically-pure intermediates, reagents, and catalysts by well-known asymmetric synthetic methods.
  • the compounds described herein are defined by their chemical structures and/or chemical names. Where a compound is referred to by both a chemical structure and a chemical name, and the chemical structure and the chemical name conflict, the chemical structure is determinative of the compound's identity.
  • the compounds described herein When administered to a subject (e.g., a non-human animal for veterinary use or for improvement of livestock or to a human for clinical use), the compounds described herein are administered in an isolated form, or as the isolated form in a pharmaceutical composition.
  • isolated means that the compounds described herein are separated from other components of either: (a) a natural source, such as a plant or cell, preferably bacterial culture, or (b) a synthetic organic chemical reaction mixture.
  • the compounds described herein are purified via conventional techniques.
  • purified means that when isolated, the isolate contains at least 95%, preferably at least 98%, of a compound described herein by weight of the isolate either as a mixture of stereoisomers, or as a diastereomerically or enantiomerically pure isolate.
  • X is CR 4 . In another embodiment, in formula (I) or (Ia), X is N.
  • R 1 may be —H, lower alkyl, lower alkoxy, lower cycloalkyl, or lower cycloalkoxy.
  • R 1 may be —H, methyl, ethyl, propyl, isopropyl, cyclopropyl, methoxy, ethoxy, propoxy, or cyclopropoxy.
  • R 3 may be —H, a lower alkyl, a lower cycloalkyl, —C(O)N(R 27 ) 2 , or —C(O)OH, wherein R 27 is —H or a lower alkyl.
  • R 3 may be —H, methyl, ethyl, n-propyl, isopropyl, cyclopropyl, n-butyl, sec-butyl, tert-butyl, n-pentyl, n-hexyl, —C(O)OH, —(CH 2 ) m C(O)OH, —CH 2 OCH 3 , —CH 2 CH 2 OCH 3 , or —C(O)N(CH 3 ) 2 .
  • R 4 may be H or a lower alkyl.
  • R 4 in formula (I) or (Ia), R 4 may be —H, methyl, ethyl, propyl, isopropyl or cyclopropyl.
  • R 1 may be —H, —OH, —SH, —NH 2 , a lower alkoxy or a lower alkyl amino. In another embodiment, in formula (I) or (Ia), R 1 may be —H, —OH, methoxy or ethoxy.
  • Z is —OH
  • Z is —SH.
  • R 2 may be —H, —OH, —SH, —NH 2 , a lower alkoxy or a lower alkyl amino.
  • R 2 may be —H, —OH, methoxy, or ethoxy.
  • R 1 may be —H, methyl, ethyl, propyl, isopropyl, cyclopropyl, methoxy, ethoxy, propoxy, or cyclopropoxy
  • R 3 may be —H, methyl, ethyl, n-propyl, isopropyl, cyclopropyl, n-butyl, sec-butyl, tert-butyl, n-pentyl, n-hexyl, —C(O)OH, —(CH 2 ) m C(O)OH, —CH 2 OCH 3 , —CH 2 CH 2 OCH 3 , or —C(O)N(CH 3 ) 2
  • R 4 may be —H, methyl, ethyl, propyl, isopropyl or cyclopropyl
  • R 2 may be —H, —OH, —SH, —NH 2
  • R 1 may be —H, methyl, ethyl, propyl, isopropyl, cyclopropyl, methoxy, ethoxy, propoxy, or cyclopropoxy
  • R 3 may be of —H, methyl, ethyl, n-propyl, isopropyl, cyclopropyl, n-butyl, sec-butyl, tert-butyl, n-pentyl, n-hexyl, —C(O)OH, —(CH 2 ) m C(O)OH, —CH 2 OCH 3 , —CH 2 CH 2 OCH 3 , or —C(O)N(CH 3 ) 2
  • R 4 may be —H, methyl, ethyl, propyl, isopropyl or cyclopropyl
  • R 2 may be of —H, —OH, —SH, —NH
  • the compound may be:
  • the compound may be:
  • Hsp90 inhibitory compounds as well as tautomers or pharmaceutically acceptable salts thereof that may be used in the methods described herein are depicted in Tables 1 or 2.
  • Hsp90 inhibitory compounds used in the disclosed combination methods can be prepared according to the procedures disclosed in U.S. Patent Publication No. 2006/0,167,070, and WO2009/023,211.
  • triazolone compounds typically can form a tautomeric structure as shown below and as exemplified by the tautomeric structures shown in Tables 1 and 2:
  • the present invention provides a pharmaceutical combination for the treatment, prophylaxis, and amelioration of proliferative disorders, such as cancer.
  • the combination comprises one or more Hsp90 inhibitors according to formulae (I) or (Ia), or a compound in Tables 1 or 2, or a tautomer or a pharmaceutically acceptable salt thereof in addition to a platinum-containing anti-cancer agent.
  • the combination includes a pharmaceutical composition or a single unit dosage form containing both an Hsp90 inhibitor and a platinum-containing anti-cancer agent.
  • Pharmaceutical combinations and dosage forms described herein comprise the two active ingredients in relative amounts and formulated in such a way that a given pharmaceutical combination or dosage form can be used to treat proliferative disorders, such as cancer.
  • Preferred pharmaceutical combinations and dosage forms comprise a compound of formulae (I) or (Ia), or a compound in Tables 1 or 2, or a tautomer or pharmaceutically acceptable salt thereof, in combination with a platinum-containing anticancer agent.
  • the Hsp90 inhibitor and the platinum-containing anticancer agent may be in individual or separate pharmaceutical compositions, depending on the dosing schedules, preferred routes of administration, and available formulations of the two compounds.
  • these embodiments can also contain one or more additional therapeutic agents.
  • the pharmaceutical combinations described herein are formulated to be compatible with its intended route of administration.
  • routes of administration include parenteral, e.g., intravenous, intradermal, subcutaneous, oral, intranasal (e.g., inhalation), transdermal (topical), transmucosal, and rectal administration.
  • the combination is formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous, subcutaneous, intramuscular, oral, intranasal or topical administration to human beings.
  • the combination is formulated in accordance with routine procedures for subcutaneous administration to human beings.
  • the combination therapies described herein comprise one or more compounds and at least one other therapy which has the same mechanism of action as the compounds.
  • the combination therapies described herein comprise one or more compounds described herein and at least one other therapy which has a different mechanism of action than the compounds.
  • the combination therapies described herein improve the therapeutic effect of one or more triazolone compounds described herein by functioning together with the platinum-containing anticancer agent to have an additive or synergistic effect.
  • the combination therapies described herein reduce the side effects associated with the therapies.
  • the combination therapies described herein reduce the effective dosage of one or more of the therapies.
  • the combination comprising one or more triazolone compounds described herein is administered to a subject, preferably a human, to prevent, treat, manage, or ameliorate cancer, or one or more symptom thereof.
  • the pharmaceutical combinations described herein may also comprise one or more other agents being used, have been used, or are known to be useful in the treatment or amelioration of cancer, particularly breast cancer, gastric cancer, colorectal cancer, pancreatic cancer, ocular melanoma, prostate cancer, gastrointestinal stromal tumors (GIST), advanced esophagogastric cancer, melanoma, hepatocellular cancer, solid tumor, liver cancer, head and neck cancer, small cell lung cancer, non-small cell lung cancer, bladder cancer, or colon cancer.
  • the pharmaceutical combinations described herein utilize pharmaceutical compositions and dosage forms which comprise one or more excipients. Suitable excipients are well known to those skilled in the art of pharmacy.
  • triazolone compounds described herein can be also formulated into or administered by controlled release means or by delivery devices that are well known to those of ordinary skill in the art. Examples include those described in U.S. Pat. Nos. 3,845,770; 3,916,899; 3,536,809; 3,598,123; and 4,008,719, 5,674,533, 5,059,595, 5,591,767, 5,120,548, 5,073,543, 5,639,476, 5,354,556, and 5,733,566.
  • the present invention also provides a method of treating a proliferative disorder in a subject, comprising administering to the subject an effective amount of the combination of an Hsp90 inhibitor and a platinum-containing anticancer agent as described herein.
  • the proliferative disorder is cancer.
  • the cancer is breast cancer, gastric cancer, colorectal cancer, pancreatic cancer, ocular melanoma, prostate cancer, gastrointestinal stromal tumors (GIST), advanced esophagogastric cancer, melanoma, hepatocellular cancer, solid tumor, liver cancer, head and neck cancer, small cell lung cancer, non-small cell lung cancer, bladder cancer, testicular tumor, ovarian cancer, lymphoma, leukemia, multiple myeloma, or colon cancer.
  • GIST gastrointestinal stromal tumors
  • anti-proliferative or anti-cancer therapies may be combined with the compounds described herein to treat proliferative diseases and cancer.
  • Other therapies or anti-cancer agents that may be used in combination with the inventive anti-cancer agents described herein include surgery, radiotherapy (including gamma-radiation, neutron beam radiotherapy, electron beam radiotherapy, proton therapy, brachytherapy, and systemic radioactive isotopes), endocrine therapy, biologic response modifiers (including interferons, interleukins, and tumor necrosis factor (TNF)), hyperthermia and cryotherapy, agents to attenuate any adverse effects (e.g., antiemetics), and other approved chemotherapeutic drugs.
  • radiotherapy including gamma-radiation, neutron beam radiotherapy, electron beam radiotherapy, proton therapy, brachytherapy, and systemic radioactive isotopes
  • endocrine therapy including interferons, interleukins, and tumor necrosis factor (TNF)
  • TNF tumor necros
  • the therapeutic agents of the combination therapies described herein can be administered sequentially or concurrently.
  • the administration of the Hsp90 inhibitor and the platinum-containing anticancer agent are done concurrently.
  • the administration of the Hsp90 inhibitor and the platinum-containing anticancer agent are done separately.
  • the administration of the Hsp90 inhibitor and the platinum-containing anticancer agent are done sequentially.
  • the administration of the Hsp90 inhibitor and the platinum-containing anticancer agent are done until the cancer is cured or stabilized or improved.
  • the present method includes treating, managing, or ameliorating cancer, or one or more symptoms thereof, comprising administering to a subject in need thereof one or more compounds represented by the structural formulae (I) or (Ia) or a compound in Table 1 or Table 2, in combination with a platinum-containing anticancer agent such as cisplatin, carboplatin, or oxaliplatin, wherein the cancer is breast cancer, gastric cancer, colorectal cancer, pancreatic cancer, ocular melanoma, prostate cancer, gastrointestinal stromal tumors (GIST), advanced esophagogastric cancer, melanoma, hepatocellular cancer, solid tumor, liver cancer, head and neck cancer, small cell lung cancer, non-small cell lung cancer, bladder cancer, testicular tumor, ovarian cancer, lymphoma, leukemia, multiple myeloma, or colon cancer.
  • a platinum-containing anticancer agent such as cisplatin, carboplatin, or oxaliplatin
  • the method of treating a subject with cancer includes administering to the subject an effective amount of a triazolone compound of 3-(2,4-dihydroxy-5-isopropyl-phenyl)-4-(1-methyl-indol-5-yl)-5-hydroxy-[1,2,4]triazole, or a tautomer, or a pharmaceutically acceptable salt thereof, in combination with an effective amount of a platinum-containing anticancer agent such as cisplatin, carboplatin, or oxaliplatin.
  • the cancer has a KRAS mutation.
  • the cancer has an ALK mutation.
  • the cancer has a BRAF mutation.
  • the method of treating a subject with cancer includes administering to the subject an effective amount of a triazolone compound of 3-(2,4-dihydroxy-5-isopropyl-phenyl)-4-(1-methyl-indol-5-yl)-5-hydroxy-[1,2,4]triazole, or a tautomer, or a pharmaceutically acceptable salt thereof, in combination with an effective amount of cisplatin.
  • the method of treating a subject with cancer includes administering to the subject an effective amount of a triazolone compound of 3-(2,4-dihydroxy-5-isopropyl-phenyl)-4-(1-methyl-indol-5-yl)-5-hydroxy-[1,2,4]triazole, or a tautomer, or a pharmaceutically acceptable salt thereof, in combination with a synergistic amount of cisplatin.
  • cisplatin is administered IV at a dose of between about 20 mg/m 2 to about 150 mg/m 2 .
  • cisplatin is administered at a dose of about 20 mg/m 2 once daily.
  • cisplatin is administered IV at about 20 mg/m 2 once daily for 5 days per cycle. In an embodiment, cisplatin is administered IV at a dose of between about 75 mg/m 2 to about 100 mg/m 2 . In an embodiment, cisplatin is administered IV at a dose of between about 75 mg/m 2 to about 100 mg/m 2 once every 4 weeks per cycle. In an embodiment, cisplatin is administered IV at a dose of about 100 mg/m 2 once every 4 weeks per cycle. In an embodiment, cisplatin is administered IV at a dose of between about 50 mg/m 2 to about 70 mg/m 2 once every 3 or 4 weeks per cycle.
  • the amount of the Hsp90 inhibitor is from about 2 mg/m 2 to about 260 mg/m 2 . In an embodiment, the amount of the Hsp90 inhibitor is about 75 mg/m 2 , about 85 mg/m 2 , about 100 mg/m 2 , about 110 mg/m 2 , about 115 mg/m 2 , about 120 mg/m 2 , about 145 mg/m 2 , about 150 mg/m 2 , about 175 mg/m 2 , about 180 mg/m 2 , about 200 mg/m 2 , about 215 mg/m 2 or about 260 mg/m 2 . In an embodiment, the Hsp90 inhibitor is administered IV once weekly or twice weekly. In any one of the above embodiments, the cancer may have a KRAS mutation. In any one of the above embodiments, the cancer may have an ALK mutation. In any one of the above embodiments, the cancer may have a BRAF mutation.
  • the method of treating a subject with cancer includes administering to the subject an effective amount of a triazolone compound of 5-hydroxy-4-(5-hydroxy-4-(1-methyl-1H-indol-5-yl)-4H-1,2,4-triazol-3-yl)-2-isopropylphenyl dihydrogen phosphate, or a tautomer, or a pharmaceutically acceptable salt thereof, in combination with an effective amount of a platinum-containing anticancer agent such as cisplatin, carboplatin, or oxaliplatin.
  • the cancer has a KRAS mutation.
  • the cancer has an ALK mutation.
  • the cancer has a BRAF mutation.
  • the method of treating a subject with cancer includes administering to the subject an effective amount of a triazolone compound of 5-hydroxy-4-(5-hydroxy-4-(1-methyl-1H-indol-5-yl)-4H-1,2,4-triazol-3-yl)-2-isopropylphenyl dihydrogen phosphate, or a tautomer, or a pharmaceutically acceptable salt thereof, in combination with an effective amount of cisplatin.
  • the method of treating a subject with cancer includes administering to the subject an effective amount of a triazolone compound of 5-hydroxy-4-(5-hydroxy-4-(1-methyl-1H-indol-5-yl)-4H-1,2,4-triazol-3-yl)-2-isopropylphenyl dihydrogen phosphate, or a tautomer, or a pharmaceutically acceptable salt thereof, in combination with a synergistic amount of cisplatin.
  • cisplatin is administered IV at a dose of between about 20 mg/m 2 to about 150 mg/m 2 .
  • cisplatin is administered at a dose of about 20 mg/m 2 once daily.
  • cisplatin is administered IV at about 20 mg/m 2 once daily for 5 days per cycle. In an embodiment, cisplatin is administered IV at a dose of between about 75 mg/m 2 to about 100 mg/m 2 . In an embodiment, cisplatin is administered IV at a dose of between about 75 mg/m 2 to about 100 mg/m 2 once every 4 weeks per cycle. In an embodiment, cisplatin is administered IV at a dose of about 100 mg/m 2 once every 4 weeks per cycle. In an embodiment, cisplatin is administered IV at a dose of between about 50 mg/m 2 to about 70 mg/m 2 once every 3 or 4 weeks per cycle.
  • the amount of the Hsp90 inhibitor is from about 2 mg/m 2 to about 260 mg/m 2 . In an embodiment, the amount of the Hsp90 inhibitor is about 75 mg/m 2 , about 85 mg/m 2 , about 100 mg/m 2 , about 110 mg/m 2 , about 115 mg/m 2 , about 120 mg/m 2 , about 145 mg/m 2 , about 150 mg/m 2 , about 175 mg/m 2 , about 180 mg/m 2 , about 200 mg/m 2 , about 215 mg/m 2 or about 260 mg/m 2 . In an embodiment, the Hsp90 inhibitor is administered IV once weekly or twice weekly. In any one of the above embodiments, the cancer may have a KRAS mutation. In any one of the above embodiments, the cancer may have an ALK mutation. In any one of the above embodiments, the cancer may have a BRAF mutation.
  • the method of treating a subject with cancer includes administering to the subject an effective amount of a triazolone compound of 3-(2,4-dihydroxy-5-isopropyl-phenyl)-4-(1-methyl-indol-5-yl)-5-hydroxy-[1,2,4]triazole, or a tautomer, or a pharmaceutically acceptable salt thereof, in combination with a platinum-containing anticancer agent such as cisplatin, carboplatin, or oxaliplatin, wherein the cancer is breast cancer, gastric cancer, colorectal cancer, pancreatic cancer, ocular melanoma, prostate cancer, gastrointestinal stromal tumors (GIST), advanced esophagogastric cancer, melanoma, hepatocellular cancer, solid tumor, liver cancer, head and neck cancer, small cell lung cancer, non-small cell lung cancer, bladder cancer, testicular tumor, ovarian cancer, lymphoma, leukemia, multiple myeloma, or colon
  • the method of treating a subject with cancer includes administering to the subject an effective amount of a triazolone compound of 5-hydroxy-4-(5-hydroxy-4-(1-methyl-1H-indol-5-yl)-4H-1,2,4-triazol-3-yl)-2-isopropylphenyl dihydrogen phosphate, or a tautomer, or a pharmaceutically acceptable salt thereof, in combination with a platinum-containing anticancer agent such as cisplatin, carboplatin, or oxaliplatin, wherein the cancer is breast cancer, gastric cancer, colorectal cancer, pancreatic cancer, ocular melanoma, prostate cancer, gastrointestinal stromal tumors (GIST), advanced esophagogastric cancer, melanoma, hepatocellular cancer, solid tumor, liver cancer, head and neck cancer, small cell lung cancer, non-small cell lung cancer, bladder cancer, testicular tumor, ovarian cancer, lymphoma, leukemia, multiple
  • the method of treating a subject with cancer includes administering to the subject an effective amount of a triazolone compound represented by the structural formulae (I) or (Ia) or a compound in Table 1 or Table 2, in combination with a platinum-containing anticancer agent such as cisplatin, carboplatin, or oxaliplatin.
  • a platinum-containing anticancer agent such as cisplatin, carboplatin, or oxaliplatin.
  • the cancer has a KRAS mutation.
  • the cancer has an ALK mutation.
  • the cancer has a BRAF mutation.
  • the method of treating a subject with cancer includes administering to the subject an effective amount of a triazolone compound represented by the structural formulae (I) or (Ia) or a compound in Table 1 or Table 2, in combination with a platinum-containing anticancer agent such as cisplatin, carboplatin, or oxaliplatin, wherein the cancer is breast cancer, gastric cancer, colorectal cancer, pancreatic cancer, ocular melanoma, prostate cancer, gastrointestinal stromal tumors (GIST), advanced esophagogastric cancer, melanoma, hepatocellular cancer, solid tumor, liver cancer, head and neck cancer, small cell lung cancer, non-small cell lung cancer, bladder cancer, testicular tumor, ovarian cancer, lymphoma, leukemia, multiple myeloma, or colon cancer.
  • the cancer has a KRAS mutation.
  • the cancer has a KRAS mutation.
  • the cancer has a KRAS mutation.
  • the cancer has a K
  • the method of treating a subject with cancer includes administering to the subject an effective amount of 3-(2,4-dihydroxy-5-isopropyl-phenyl)-4-(1-methyl-indol-5-yl)-5-hydroxy-[1,2,4]triazole, or a tautomer, or a pharmaceutically acceptable salt thereof, in combination with a platinum-containing anticancer agent such as cisplatin, carboplatin, or oxaliplatin.
  • the cancer has a KRAS mutation.
  • the cancer has an ALK mutation.
  • the cancer has a BRAF mutation.
  • the method of treating a subject with cancer, wherein the subject is being or has been treated with a chemotherapeutic agent includes administering to the subject an effective amount of 3-(2,4-dihydroxy-5-isopropyl-phenyl)-4-(1-methyl-indol-5-yl)-5-hydroxy-[1,2,4]triazole, or a tautomer, or a pharmaceutically acceptable salt thereof, in combination with cisplatin.
  • cisplatin is administered IV at a dose of between about 20 mg/m 2 to about 150 m g/m 2 .
  • cisplatin is administered at a dose of about 20 mg/m 2 once daily.
  • cisplatin is administered IV at about 20 mg/m 2 once daily for 5 days per cycle. In an embodiment, cisplatin is administered IV at a dose of between about 75 mg/m 2 to about 100 mg/m 2 . In an embodiment, cisplatin is administered IV at a dose of between about 75 mg/m 2 to about 100 mg/m 2 once every 4 weeks per cycle. In an embodiment, cisplatin is administered IV at a dose of about 100 mg/m 2 once every 4 weeks per cycle. In an embodiment, cisplatin is administered IV at a dose of between about 50 m g/m 2 to about 70 mg/m 2 once every 3 or 4 weeks per cycle.
  • the amount of the Hsp90 inhibitor is from about 2 mg/m 2 to about 260 mg/m 2 . In an embodiment, the amount of the Hsp90 inhibitor is about 75 mg/m 2 , about 85 mg/m 2 , about 100 mg/m 2 , about 110 mg/m 2 , about 115 mg/m 2 , about 120 mg/m 2 , about 145 mg/m 2 , about 150 mg/m 2 , about 175 mg/m 2 , about 180 mg/m 2 , about 200 m g/m 2 , about 215 mg/m 2 or about 260 mg/m 2 . In an embodiment, the Hsp90 inhibitor is administered IV once weekly or twice weekly. In any one of the above embodiments, the cancer may have a KRAS mutation. In any one of the above embodiments, the cancer may have an ALK mutation. In any one of the above embodiments, the cancer may have a BRAF mutation.
  • the method of treating a subject with cancer includes administering to the subject an effective amount of 5-hydroxy-4-(5-hydroxy-4-(1-methyl-1H-indol-5-yl)-4H-1,2,4-triazol-3-yl)-2-isopropylphenyl dihydrogen phosphate, or a tautomer, or a pharmaceutically acceptable salt thereof, in combination with a platinum-containing anticancer agent such as cisplatin, carboplatin, or oxaliplatin.
  • the cancer has a KRAS mutation.
  • the cancer has an ALK mutation.
  • the cancer has a BRAF mutation.
  • the method of treating a subject with cancer, wherein the subject is being or has been treated with a chemotherapeutic agent includes administering to the subject an effective amount of 5-hydroxy-4-(5-hydroxy-4-(1-methyl-1H-indol-5-yl)-4H-1,2,4-triazol-3-yl)-2-isopropylphenyl dihydrogen phosphate, or a tautomer, or a pharmaceutically acceptable salt thereof, in combination with cisplatin.
  • cisplatin is administered IV at a dose of between about 20 mg/m 2 to about 150 mg/m 2 .
  • cisplatin is administered at a dose of about 20 mg/m 2 once daily.
  • cisplatin is administered IV at about 20 mg/m 2 once daily for 5 days per cycle. In an embodiment, cisplatin is administered IV at a dose of between about 75 mg/m 2 to about 100 mg/m 2 . In an embodiment, cisplatin is administered IV at a dose of between about 75 mg/m 2 to about 100 mg/m 2 once every 4 weeks per cycle. In an embodiment, cisplatin is administered IV at a dose of about 100 mg/m 2 once every 4 weeks per cycle. In an embodiment, cisplatin is administered IV at a dose of between about 50 mg/m 2 to about 70 mg/m 2 once every 3 or 4 weeks per cycle.
  • the amount of the Hsp90 inhibitor is from about 2 mg/m 2 to about 260 mg/m 2 . In an embodiment, the amount of the Hsp90 inhibitor is about 75 mg/m 2 , about 85 mg/m 2 , about 100 mg/m 2 , about 110 mg/m 2 , about 115 mg/m 2 , about 120 mg/m 2 , about 145 mg/m 2 , about 150 mg/m 2 , about 175 mg/m 2 , about 180 mg/m 2 , about 200 mg/m 2 , about 215 mg/m 2 or about 260 mg/m 2 . In an embodiment, the Hsp90 inhibitor is administered IV once weekly or twice weekly. In any one of the above embodiments, the cancer may have a KRAS mutation. In any one of the above embodiments, the cancer may have an ALK mutation. In any one of the above embodiments, the cancer may have a BRAF mutation.
  • the method of treating a subject with cancer includes administering to the subject an effective amount of a triazolone compound of 3-(2,4-dihydroxy-5-isopropyl-phenyl)-4-(1-methyl-indol-5-yl)-5-hydroxy-[1,2,4]triazole, or a tautomer, or a pharmaceutically acceptable salt thereof, in combination with a platinum-containing anticancer agent such as cisplatin, carboplatin, or oxaliplatin, wherein the cancer is breast cancer, gastric cancer, colorectal cancer, pancreatic cancer, ocular melanoma, prostate cancer, gastrointestinal stromal tumors (GIST), advanced esophagogastric cancer, melanoma, hepatocellular cancer, solid tumor, liver cancer, head and neck cancer, small cell lung cancer, non-small cell lung cancer, bladder cancer, testicular tumor, ovarian
  • a platinum-containing anticancer agent such as cisplatin, carboplatin,
  • the method of treating a subject with cancer includes administering to the subject an effective amount of a triazolone compound of 5-hydroxy-4-(5-hydroxy-4-(1-methyl-1H-indol-5-yl)-4H-1,2,4-triazol-3-yl)-2-isopropylphenyl dihydrogen phosphate, or a tautomer, or a pharmaceutically acceptable salt thereof, in combination with a platinum-containing anticancer agent such as cisplatin, carboplatin, or oxaliplatin, wherein the cancer is breast cancer, gastric cancer, colorectal cancer, pancreatic cancer, ocular melanoma, prostate cancer, gastrointestinal stromal tumors (GIST), advanced esophagogastric cancer, melanoma, hepatocellular cancer, solid tumor, liver cancer, head and neck cancer, small cell lung cancer, non-small cell lung cancer, bladder cancer
  • a platinum-containing anticancer agent such as cisplatin, carboplatin,
  • the method of treating a subject with cancer includes administering to the subject an effective amount of a triazolone compound represented by the structural formulae (I) or (Ia) or a compound in Table 1 or Table 2, in combination with a platinum-containing anticancer agent such as cisplatin, carboplatin, or oxaliplatin, wherein the cancer is breast cancer, gastric cancer, colorectal cancer, pancreatic cancer, ocular melanoma, prostate cancer, gastrointestinal stromal tumors (GIST), advanced esophagogastric cancer, melanoma, hepatocellular cancer, solid tumor, liver cancer, head and neck cancer, small cell lung cancer, non-small cell lung cancer, bladder cancer, testicular tumor, ovarian cancer, lymphoma, leukemia, multiple myeloma, or colon cancer.
  • the cancer has a KRAS mutation.
  • the cancer has a KRAS mutation.
  • the method of treating a subject with cancer includes administering to the subject an effective amount of 3-(2,4-dihydroxy-5-isopropyl-phenyl)-4-(1-methyl-indol-5-yl)-5-hydroxy-[1,2,4]triazole, or a tautomer, or a pharmaceutically acceptable salt thereof, in combination with a platinum-containing anticancer agent such as cisplatin, carboplatin, or oxaliplatin.
  • the cancer has a KRAS mutation.
  • the cancer has an ALK mutation.
  • the cancer has a BRAF mutation.
  • the method of treating a subject with cancer includes administering to the subject an effective amount of 3-(2,4-dihydroxy-5-isopropyl-phenyl)-4-(1-methyl-indol-5-yl)-5-hydroxy-[1,2,4]triazole, or a tautomer, or a pharmaceutically acceptable salt thereof, in combination with cisplatin.
  • cisplatin is administered IV at a dose of between about 20 mg/m 2 to about 150 mg/m 2 .
  • cisplatin is administered at a dose of about 20 mg/m 2 once daily.
  • cisplatin is administered IV at about 20 mg/m 2 once daily for 5 days per cycle. In an embodiment, cisplatin is administered IV at a dose from about 75 mg/m 2 to about 100 mg/m 2 . In an embodiment, cisplatin is administered IV at a dose from about 75 m g/m 2 to about 100 mg/m 2 once every 4 weeks per cycle. In an embodiment, cisplatin is administered IV at a dose of about 100 mg/m 2 once every 4 weeks per cycle. In an embodiment, cisplatin is administered IV at a dose of between about 50 mg/m 2 to about 70 mg/m 2 once every 3 or 4 weeks per cycle.
  • the amount of the Hsp90 inhibitor is from about 2 mg/m 2 to about 260 mg/m 2 . In an embodiment, the amount of the Hsp90 inhibitor is about 75 mg/m 2 , about 85 mg/m 2 , about 100 mg/m 2 , about 110 m g/m 2 , about 115 mg/m 2 , about 120 mg/m 2 , about 145 mg/m 2 , about 150 mg/m 2 , about 175 mg/m 2 , about 180 mg/m 2 , about 200 mg/m 2 , about 215 mg/m 2 or about 260 mg/m 2 . In an embodiment, the Hsp90 inhibitor is administered IV once weekly or twice weekly. In any one of the above embodiments, the cancer may have a KRAS mutation. In any one of the above embodiments, the cancer may have an ALK mutation. In any one of the above embodiments, the cancer may have a BRAF mutation.
  • the method of treating a subject with cancer includes administering to the subject an effective amount of 5-hydroxy-4-(5-hydroxy-4-(1-methyl-1H-indol-5-yl)-4H-1,2,4-triazol-3-yl)-2-isopropylphenyl dihydrogen phosphate, or a tautomer, or a pharmaceutically acceptable salt thereof, in combination with a platinum-containing anticancer agent such as cisplatin, carboplatin, or oxaliplatin.
  • the cancer has a KRAS mutation.
  • the cancer has an ALK mutation.
  • the cancer has a BRAF mutation.
  • the method of treating a subject with cancer includes administering to the subject an effective amount of 5-hydroxy-4-(5-hydroxy-4-(1-methyl-1H-indol-5-yl)-4H-1,2,4-triazol-3-yl)-2-isopropylphenyl dihydrogen phosphate, or a tautomer, or a pharmaceutically acceptable salt thereof, in combination with cisplatin.
  • cisplatin is administered IV at a dose of between about 20 mg/m 2 to about 150 mg/m 2 .
  • cisplatin is administered at a dose of about 20 mg/m 2 once daily.
  • cisplatin is administered IV at about 20 mg/m 2 once daily for 5 days per cycle. In an embodiment, cisplatin is administered IV at a dose from about 75 mg/m 2 to about 100 mg/m 2 . In an embodiment, cisplatin is administered IV at a dose from about 75 mg/m 2 to about 100 mg/m 2 once every 4 weeks per cycle. In an embodiment, cisplatin is administered IV at a dose of about 100 mg/m 2 once every 4 weeks per cycle. In an embodiment, cisplatin is administered IV at a dose from about 50 mg/m 2 to about 70 mg/m 2 once every 3 or 4 weeks per cycle.
  • the amount of the Hsp90 inhibitor is from about 2 mg/m 2 to about 260 mg/m 2 . In an embodiment, the amount of the Hsp90 inhibitor is about 75 mg/m 2 , about 85 mg/m 2 , about 100 mg/m 2 , about 110 mg/m 2 , about 115 mg/m 2 , about 120 mg/m 2 , about 145 mg/m 2 , about 150 mg/m 2 , about 175 mg/m 2 , about 180 mg/m 2 , about 200 m g/m 2 , about 215 mg/m 2 or about 260 mg/m 2 . In an embodiment, the Hsp90 inhibitor is administered IV once weekly or twice weekly. In any one of the above embodiments, the cancer may have a KRAS mutation. In any one of the above embodiments, the cancer may have an ALK mutation. In any one of the above embodiments, the cancer may have a BRAF mutation.
  • the method of treating a subject with cancer includes administering to the subject an effective amount of a triazolone compound of 3-(2,4-dihydroxy-5-isopropyl-phenyl)-4-(1-methyl-indol-5-yl)-5-hydroxy-[1,2,4]triazole, or a tautomer, or a pharmaceutically acceptable salt thereof, in combination with a platinum-containing anticancer agent such as cisplatin, carboplatin, or oxaliplatin, wherein the cancer is breast cancer, gastric cancer, colorectal cancer, pancreatic cancer, ocular melanoma, prostate cancer, gastrointestinal stromal tumors (GIST), advanced esophagogastric cancer, melanoma, hepatocellular cancer, solid tumor, liver cancer, head and neck cancer, small cell lung cancer, non-small cell lung cancer, bladder cancer, testicular tumor, ova
  • the method of treating a subject with cancer includes administering to the subject an effective amount of a triazolone compound of 5-hydroxy-4-(5-hydroxy-4-(1-methyl-1H-indol-5-yl)-4H-1,2,4-triazol-3-yl)-2-isopropylphenyl dihydrogen phosphate, or a tautomer, or a pharmaceutically acceptable salt thereof, in combination with a platinum-containing anticancer agent such as cisplatin, carboplatin, or oxaliplatin, wherein the cancer is breast cancer, gastric cancer, colorectal cancer, pancreatic cancer, ocular melanoma, prostate cancer, gastrointestinal stromal tumors (GIST), advanced esophagogastric cancer, melanoma, hepatocellular cancer, solid tumor, liver cancer, head and neck cancer, small cell lung cancer, non-small cell lung cancer, bladder
  • the method includes inhibiting the growth of a cancer or tumor cell comprising the steps of: (a) contacting the cell with an effective amount of a compound of formulae (I) or (Ia) or a compound in Table (1) or Table (2), or tautomer or a pharmaceutically acceptable salt thereof; and (b) exposing the cell to an effective amount of a platinum-containing anticancer agent such as cisplatin, carboplatin, or oxaliplatin.
  • a platinum-containing anticancer agent such as cisplatin, carboplatin, or oxaliplatin.
  • the method includes inhibiting the growth of a cancer or tumor cell comprising the steps of: (a) contacting the cell with an effective amount of a compound of -(2,4-dihydroxy-5-isopropyl-phenyl)-4-(1-methyl-indol-5-yl)-5-hydroxy-[1,2,4]triazole, or a tautomer, or a pharmaceutically acceptable salt thereof; and (b) exposing the cell to an effective amount of a platinum-containing anticancer agent such as cisplatin, carboplatin, or oxaliplatin.
  • the cancer has a KRAS mutation.
  • the cancer has an ALK mutation.
  • the cancer has a BRAF mutation.
  • the invention also provides a method of inhibiting the growth of a cancer or tumor cell, comprising the steps of: (a) contacting the cell with an effective amount of a compound of -(2,4-dihydroxy-5-isopropyl-phenyl)-4-(1-methyl-indol-5-yl)-5-hydroxy-[1,2,4]triazole, or a tautomer, or a pharmaceutically acceptable salt thereof; and (b) exposing the cell to an effective amount of cisplatin.
  • the cancer has a KRAS mutation.
  • the cancer has an ALK mutation.
  • the cancer has a BRAF mutation.
  • the invention also provides a method of inhibiting the growth of a cancer or tumor cell, comprising the steps of: (a) contacting the cell with an effective amount of a compound of 5-hydroxy-4-(5-hydroxy-4-(1-methyl-1H-indol-5-yl)-4H-1,2,4-triazol-3-yl)-2-isopropylphenyl dihydrogen phosphate, or tautomer or a pharmaceutically acceptable salt thereof; and (b) exposing the cell to an effective amount of a platinum-containing anticancer agent such as cisplatin, carboplatin, or oxaliplatin.
  • the cancer has a KRAS mutation.
  • the cancer has an ALK mutation.
  • the cancer has a BRAF mutation.
  • the invention also provides a method of inhibiting the growth of a cancer or tumor cell, comprising the steps of: (a) contacting the cell with an effective amount of a compound of 5-hydroxy-4-(5-hydroxy-4-(1-methyl-1H-indol-5-yl)-4H-1,2,4-triazol-3-yl)-2-isopropylphenyl dihydrogen phosphate, or tautomer or a pharmaceutically acceptable salt thereof; and (b) exposing the cell to an effective amount of cisplatin.
  • the cancer has a KRAS mutation.
  • the cancer has an ALK mutation.
  • the cancer has a BRAF mutation.
  • the recommended daily dose range of a triazolone compound for the conditions described herein lie within the range of from about 0.01 mg to about 1000 mg per day, given as a single once-a-day dose preferably as divided doses throughout a day.
  • the daily dose is administered twice daily in equally divided doses.
  • a daily dose range should be from about 5 mg to about 500 mg per day, more specifically, between about 10 mg and about 200 mg per day.
  • the therapy should be initiated at a lower dose, perhaps about 1 mg to about 25 mg, and increased if necessary up to about 200 mg to about 1000 mg per day as either a single dose or divided doses, depending on the patient's global response.
  • the dosage of the composition comprising a triazolone compound described herein administered to prevent, treat, manage, or ameliorate cancer, or one or more symptoms thereof in a patient is 150 ⁇ g/kg, preferably 250 ⁇ g/kg, 500 ⁇ g/kg, 1 mg/kg, 5 mg/kg, 10 mg/kg, 25 mg/kg, 50 mg/kg, 75 mg/kg, 100 mg/kg, 125 mg/kg, 150 mg/kg, or 200 mg/kg or more of a patient's body weight.
  • the dosage of the composition comprising a compound described herein administered to prevent, treat, manage, or ameliorate cancer, or one or more symptoms thereof in a patient is a unit dose of 0.1 mg to 20 mg, 0.1 mg to 15 mg, 0.1 mg to 12 mg, 0.1 mg to 10 mg, 0.1 mg to 8 mg, 0.1 mg to 7 mg, 0.1 mg to 5 mg, 0.1 to 2.5 mg, 0.25 mg to 20 mg, 0.25 to 15 mg, 0.25 to 12 mg, 0.25 to 10 mg, 0.25 to 8 mg, 0.25 mg to 7 mg, 0.25 mg to 5 mg, 0.5 mg to 2.5 mg, 1 mg to 20 mg, 1 mg to 15 mg, 1 mg to 12 mg, 1 mg to 10 mg, 1 mg to 8 mg, 1 mg to 7 mg, 1 mg to 5 mg, or 1 mg to 2.5 mg.
  • the unit dose can be administered 1, 2, 3, 4 or more times daily, or once every 2, 3, 4, 5, 6 or 7 days, or once weekly, once every two weeks, once every three weeks or once monthly.
  • the therapies are administered less than 5 minutes apart, less than 30 minutes apart, 1 hour apart, at about 1 hour apart, at about 1 to about 2 hours apart, at about 2 hours to about 3 hours apart, at about 3 hours to about 4 hours apart, at about 4 hours to about 5 hours apart, at about 5 hours to about 6 hours apart, at about 6 hours to about 7 hours apart, at about 7 hours to about 8 hours apart, at about 8 hours to about 9 hours apart, at about 9 hours to about 10 hours apart, at about 10 hours to about 11 hours apart, at about 11 hours to about 12 hours apart, at about 12 hours to 18 hours apart, 18 hours to 24 hours apart, 24 hours to 36 hours apart, 36 hours to 48 hours apart, 48 hours to 52 hours apart, 52 hours to 60 hours apart, 60 hours to 72 hours apart, 72 hours to 84 hours apart, 84 hours to 96 hours apart, or 96 hours to 120 hours part.
  • two or more therapies are administered within the same
  • one or more compounds described herein and one or more other the therapies are cyclically administered. Cycling therapy involves the administration of a first therapy (e.g., a first prophylactic or therapeutic agents) for a period of time, followed by the administration of a second therapy (e.g., a second prophylactic or therapeutic agents) for a period of time, followed by the administration of a third therapy (e.g., a third prophylactic or therapeutic agents) for a period of time and so forth, and repeating this sequential administration, i.e., the cycle in order to reduce the development of resistance to one of the agents, to avoid or reduce the side effects of one of the agents, and/or to improve the efficacy of the treatment.
  • a first therapy e.g., a first prophylactic or therapeutic agents
  • a second therapy e.g., a second prophylactic or therapeutic agents
  • a third therapy e.g., a third prophylactic or therapeutic agents
  • administration of the same compound described herein may be repeated and the administrations may be separated by at least 1 day, 2 days, 3 days, 5 days, 10 days, 15 days, 30 days, 45 days, 2 months, 75 days, 3 months, or 6 months.
  • administration of the same prophylactic or therapeutic agent may be repeated and the administration may be separated by at least at least 1 day, 2 days, 3 days, 5 days, 10 days, 15 days, 30 days, 45 days, 2 months, 75 days, 3 months, or 6 months.
  • a method of preventing, treating, managing, or ameliorating a proliferative disorders, such as cancer, or one or more symptoms thereof comprising administering to a subject in need thereof a dose of at least 150 ⁇ g/kg, preferably at least 250 ⁇ g/kg, at least 500 ⁇ g/kg, at least 1 mg/kg, at least 5 mg/kg, at least 10 mg/kg, at least 25 mg/kg, at least 50 mg/kg, at least 75 mg/kg, at least 100 mg/kg, at least 125 mg/kg, at least 150 mg/kg, or at least 200 mg/kg or more of one or more compounds described herein once every day, preferably, once every 2 days, once every 3 days, once every 4 days, once every 5 days, once every 6 days, once every 7 days, once every 8 days, once every 10 days, once every two weeks, once every three weeks, or once a month.
  • the dose can be divided into portions (typically equal portions) administered two, three, four or more times
  • the invention also provides the use of a compound of formulae (I) or (Ia), or a compound in Tables 1 or 2, or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for the treatment of a subject with cancer.
  • the invention further provides the use of a compound of formulae (I) or (Ia), or a compound in Tables 1 or 2, or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for the treatment of a subject with a cancer, in combination with a platinum-containing anticancer agent such as cisplatin, carboplatin, or oxaliplatin.
  • the invention further provides the use of a compound of 3-(2,4-dihydroxy-5-isopropyl-phenyl)-4-(1-methyl-indol-5-yl)-5-hydroxy-[1,2,4]triazole, or a tautomer, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment of a subject with a cancer, in combination with a platinum-containing anticancer agent such as cisplatin, carboplatin, or oxaliplatin.
  • a platinum-containing anticancer agent such as cisplatin, carboplatin, or oxaliplatin.
  • the invention further provides the use of a compound of 5-hydroxy-4-(5-hydroxy-4-(1-methyl-1H-indol-5-yl)-4H-1,2,4-triazol-3-yl)-2-isopropylphenyl dihydrogen phosphate, or a tautomer, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment of a subject with a cancer, in combination with a platinum-containing anticancer agent such as cisplatin, carboplatin, or oxaliplatin.
  • a platinum-containing anticancer agent such as cisplatin, carboplatin, or oxaliplatin.
  • the invention further provides the use of the compound of 3-(2,4-dihydroxy-5-isopropyl-phenyl)-4-(1-methyl-indol-5-yl)-5-hydroxy-[1,2,4]triazole, or a tautomer, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment of a subject with a cancer, in combination with cisplatin.
  • the invention further provides the synergistic use of the compound of 3-(2,4-dihydroxy-5-isopropyl-phenyl)-4-(1-methyl-indol-5-yl)-5-hydroxy-[1,2,4]triazole, or a tautomer, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment of a subject with a cancer, in combination with cisplatin.
  • the invention also provides a compound of formulae (I) or (Ia) or a pharmaceutically acceptable salt thereof for use in treating a subject with a cancer.
  • the invention also provides a compound of formulae (I) or (Ia) or a pharmaceutically acceptable salt thereof for use in treating a subject with cancer in combination with a platinum-containing anticancer agent such as cisplatin, carboplatin, or oxaliplatin.
  • the invention also provides a compound of 3-(2,4-dihydroxy-5-isopropyl-phenyl)-4-(1-methyl-indol-5-yl)-5-hydroxy-[1,2,4]triazole, or a tautomer, or a pharmaceutically acceptable salt thereof, for use in treating a subject with cancer in combination with a platinum-containing anticancer agent such as cisplatin, carboplatin, or oxaliplatin.
  • a platinum-containing anticancer agent such as cisplatin, carboplatin, or oxaliplatin.
  • the invention also provides a compound of 5-hydroxy-4-(5-hydroxy-4-(1-methyl-1H-indol-5-yl)-4H-1,2,4-triazol-3-yl)-2-isopropylphenyl dihydrogen phosphate, or a tautomer, or a pharmaceutically acceptable salt thereof, for use in treating a subject with cancer in combination with a platinum-containing anticancer agent such as cisplatin, carboplatin, or oxaliplatin.
  • a platinum-containing anticancer agent such as cisplatin, carboplatin, or oxaliplatin.
  • the invention also provides a compound of 3-(2,4-dihydroxy-5-isopropyl-phenyl)-4-(1-methyl-indol-5-yl)-5-hydroxy-[1,2,4]triazole, or a tautomer, or a pharmaceutically acceptable salt thereof, for use in treating a subject with cancer in combination with cisplatin.
  • the invention also provides a compound of 3-(2,4-dihydroxy-5-isopropyl-phenyl)-4-(1-methyl-indol-5-yl)-5-hydroxy-[1,2,4]triazole, or a tautomer, or a pharmaceutically acceptable salt thereof, for synergistic use in treating a subject with cancer in combination with cisplatin.
  • Human HCT-116 colorectal cancer cells (CRC) and MDA-MB-231 breast cancer cells were purchased from the American Type Culture Collection (Manassas, Va.) and grown following ATCC recommendations, in the presence of fetal bovine serum (10%), 2 mM L-glutamine and antibiotics (100 IU/ml penicillin and 100 ⁇ g/ml streptomycin, Sigma). Cells were maintained at 37° C., 5% CO 2 atmosphere.
  • Cell viability was measured using the alamarBlue assay (Invitrogen). In brief, cells were plated in 96-well plates in triplicate at 5K cells per well and incubated at 37° C., 5% CO 2 atmosphere for 24 hr prior to the addition of drug or vehicle (0.3% DMSO) to the culture medium. After 72 hr, 10 ⁇ l/well alamarBlue was added to the wells and incubated for an additional 3 hr at 37° C., 5% CO 2 atmosphere. Fluorescence (560 EX /590 EM nM) was measured with a SpectraMax microplate reader (Molecular Devices) and the resulting data were used to calculate cell viability, normalized to vehicle control.
  • SpectraMax microplate reader Molecular Devices
  • mice Six to seven week old, female CB17/Icr-Prkdc scid /Crl (SCID) mice were obtained from Charles River Laboratories (Wilmington, Mass., USA). Animals were housed 4-5/cage in micro-isolators, with a 12 hr/12 hr light/dark cycle, acclimated for at least 1 week prior to use and fed normal laboratory chow ad libitum. Animals were between seven to eight weeks of age at implantation.
  • HCT-116 or MDA-MB-231 tumor cells were harvested by trypsinization, washed in PBS and re-susupended at a concentration of 5 ⁇ 10(7) cells/mL in 50% non-supplemented medium and 50% Matrigel Basement Membrane Matrix (BD Biosciences; Bedford, Mass., USA).
  • 5 ⁇ 10(6) cells in 0.1 mL of a cell suspension were injected subcutaneously into the flanks of SCID mice.
  • Ganetespib was prepared by dissolving the appropriate amounts of the compound in dimethyl sulfoxide (DMSO) by sonication in an ultrasonic water bath. Stock solutions were prepared weekly, stored at ⁇ 20° C. and diluted fresh each day for dosing. A solution of 20% Cremophor RH40 (polyoxyl 40 hydrogenated castor oil; BASF Corp., Aktiengesellschaft, Ludwigshafen, Germany) in 5% dextrose in water (Abbott Laboratories, North Chicago, Ill., USA) was also prepared by first heating 100% Cremophor RH40 at 50-60° C. until liquefied and clear, diluting 1:5 with 100% D5W, reheating again until clear and then mixing well.
  • DMSO dimethyl sulfoxide
  • This solution can be stored at room temperature for up to 3 months prior to use.
  • DMSO stock solutions were diluted 1:10 with 20% Cremophor RH40.
  • the final DRD formulation for dosing contained 10% DMSO, 18% Cremophor RH40, 3.6% dextrose, 68.4% water and the appropriate amount of test article.
  • Animals were intravenously (i.v.) injected with this formulation at 10 mL per kg body weight 1 day each week.
  • Cisplatin was prepared fresh in saline and given intravenously 6 hours after ganetespib injection, once per week.
  • IC 50 half maximal inhibitory concentration
  • Combinations between ganetespib and cisplatin were then performed in HCT-116 cells concurrently based on the IC 50 for each agent in matrix format with 54 combination pairs for each drug.
  • the combined drugs, as well as each drug alone, were incubated with the cells for 3 days and the surviving fraction of cells relative to control was determined using the alamarBlue assay. Representative figures are shown in FIGS. 3 and 4 .
  • the combination of ganetespib with cisplatin displayed enhanced cytotoxicity relative to single agent drugs alone. Similar results were observed when cells were exposed to ganetespib for just one hour, washed and then treated with cisplatin for 3 days.
  • Ganetespib 150 mg/kg was given as a single bolus injection 6 hours prior to cisplatin (2 mg/kg) administration. Representative figures are shown in FIGS. 5 and 6 . In both cancer xenograft models, the combination of ganetespib with cisplatin displayed greater antitumor activity relative to monotherapy.
US14/355,689 2011-11-02 2012-11-01 Combination therapy of hsp90 inhibitors with platinum-containing agents Abandoned US20140286902A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US14/355,689 US20140286902A1 (en) 2011-11-02 2012-11-01 Combination therapy of hsp90 inhibitors with platinum-containing agents

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201161554706P 2011-11-02 2011-11-02
US14/355,689 US20140286902A1 (en) 2011-11-02 2012-11-01 Combination therapy of hsp90 inhibitors with platinum-containing agents
PCT/US2012/063039 WO2013067165A1 (fr) 2011-11-02 2012-11-01 Polythérapie d'inhibiteurs de hsp 90 avec des agents contenant du platine

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2012/063039 A-371-Of-International WO2013067165A1 (fr) 2011-11-02 2012-11-01 Polythérapie d'inhibiteurs de hsp 90 avec des agents contenant du platine

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/940,055 Continuation US10500193B2 (en) 2011-11-02 2018-03-29 Combination therapy of HSP90 inhibitors with platinum-containing agents

Publications (1)

Publication Number Publication Date
US20140286902A1 true US20140286902A1 (en) 2014-09-25

Family

ID=47178975

Family Applications (2)

Application Number Title Priority Date Filing Date
US14/355,689 Abandoned US20140286902A1 (en) 2011-11-02 2012-11-01 Combination therapy of hsp90 inhibitors with platinum-containing agents
US15/940,055 Active US10500193B2 (en) 2011-11-02 2018-03-29 Combination therapy of HSP90 inhibitors with platinum-containing agents

Family Applications After (1)

Application Number Title Priority Date Filing Date
US15/940,055 Active US10500193B2 (en) 2011-11-02 2018-03-29 Combination therapy of HSP90 inhibitors with platinum-containing agents

Country Status (6)

Country Link
US (2) US20140286902A1 (fr)
EP (1) EP2776025A1 (fr)
JP (1) JP2014534228A (fr)
AU (1) AU2012332424A1 (fr)
CA (1) CA2853806C (fr)
WO (1) WO2013067165A1 (fr)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9006277B2 (en) 2006-05-25 2015-04-14 Synta Pharmaceuticals Corp. Triazole compounds that modulate HSP90 activity
US9205086B2 (en) 2010-04-19 2015-12-08 Synta Pharmaceuticals Corp. Cancer therapy using a combination of a Hsp90 inhibitory compounds and a EGFR inhibitor
US9402831B2 (en) 2011-11-14 2016-08-02 Synta Pharmaceutical Corp. Combination therapy of HSP90 inhibitors with BRAF inhibitors
US9439899B2 (en) 2011-11-02 2016-09-13 Synta Pharmaceuticals Corp. Cancer therapy using a combination of HSP90 inhibitors with topoisomerase I inhibitors
US9539243B2 (en) 2008-08-08 2017-01-10 Synta Pharmaceuticals Corp. Triazole compounds that modulate HSP90 activity

Families Citing this family (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SG182662A1 (en) 2010-01-28 2012-08-30 Harvard College Compositions and methods for enhancing proteasome activity
WO2012154967A1 (fr) 2011-05-12 2012-11-15 Proteostasis Therapeutics, Inc. Régulateurs de la protéostasie
WO2014116228A1 (fr) 2013-01-25 2014-07-31 President And Fellows Of Harvard College Inhibiteurs de l'usp14 utilisables en vue du traitement ou de la prévention d'infections virales
WO2015073528A1 (fr) 2013-11-12 2015-05-21 Proteostasis Therapeutics, Inc. Composés renforçant l'activité des protéasomes
EP3127544B1 (fr) * 2014-04-04 2021-09-15 Taiho Pharmaceutical Co., Ltd. Médicament antitumoral contenant un complexe de platine antitumoral et promoteur de l'effet antitumoral
PL3179991T3 (pl) 2014-08-11 2022-02-14 Acerta Pharma B.V. Kombinacje terapeutyczne inhibitora btk i inhibitora bcl-2
WO2016024231A1 (fr) 2014-08-11 2016-02-18 Acerta Pharma B.V. Combinaisons thérapeutiques d'un inhibiteur de btk, d'un inhibiteur de pi3k, d'un inhibiteur de jak-2, d'un inhibiteur de pd-1 et/ou d'un inhibiteur de pd-l1
WO2016024232A1 (fr) 2014-08-11 2016-02-18 Acerta Pharma B.V. Combinaisons thérapeutiques d'un inhibiteur de la btk, d'un inhibiteur de la pi3k, d'un inhibiteur de la jak-2 et/ou d'un inhibiteur de la cdk 4/6
PT3300500T (pt) 2015-05-20 2020-05-19 Amgen Inc Agonistas triazóis do receptor apj
US9988369B2 (en) 2016-05-03 2018-06-05 Amgen Inc. Heterocyclic triazole compounds as agonists of the APJ receptor
WO2018093580A1 (fr) 2016-11-16 2018-05-24 Amgen Inc. Composés de triazole pyridyle en tant qu'agonistes du récepteur apj
US11191762B2 (en) 2016-11-16 2021-12-07 Amgen Inc. Alkyl substituted triazole compounds as agonists of the APJ Receptor
US10736883B2 (en) 2016-11-16 2020-08-11 Amgen Inc. Triazole furan compounds as agonists of the APJ receptor
WO2018097945A1 (fr) 2016-11-16 2018-05-31 Amgen Inc. Triazoles substitués par hétéroaryle utilisés en tant qu'agonistes du récepteur apj
EP3541810B1 (fr) 2016-11-16 2020-12-23 Amgen Inc. Composés phényle triazole en tant qu'agonistes du récepteur apj
MA46827A (fr) 2016-11-16 2019-09-25 Amgen Inc Composés de triazole à substitution cycloalkyle en tant qu'agonistes du récepteur apj
MA50509A (fr) 2017-11-03 2021-06-02 Amgen Inc Agonistes de triazole fusionnés du récepteur apj
WO2019213006A1 (fr) 2018-05-01 2019-11-07 Amgen Inc. Pyrimidinones substituées en tant qu'agonistes du récepteur apj
WO2021007512A1 (fr) * 2019-07-11 2021-01-14 Emory University Chimiothérapie à base de platine, agents de liaison à la mast, agents de liaison au récepteur des glucocorticoïdes (gr) et/ou agents de liaison à hsp90 destinés à être utilisés dans le traitement du cancer

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7825148B2 (en) * 2004-11-18 2010-11-02 Synta Pharmaceuticals Corp. Triazole compounds that modulate Hsp90 activity
US8742133B2 (en) * 2007-08-13 2014-06-03 Synta Pharmaceuticals Corp. Triazole compounds that modulate HSP90 activity

Family Cites Families (216)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE1001992C2 (de) 1954-05-10 1957-07-11 Hoechst Ag Verfahren zur Herstellung von 3, 5-Di-(2'-anthrachinonyl)-4-phenyl-1, 2, 4-triazolen
US2898343A (en) 1957-11-18 1959-08-04 American Cyanamid Co Anthraquinone triazoles
GB928919A (en) 1960-08-16 1963-06-19 Bellon Labor Sa Roger Triazole derivatives and a process for their preparation
US3898272A (en) 1966-05-06 1975-08-05 Bayer Ag Thionosalicylic acid anilides
DE1805156B2 (de) 1968-10-25 1976-01-29 Bayer Ag, 5090 Leverkusen Verfahren zur herstellung von 2- acyloxythionobenzamiden
US3536809A (en) 1969-02-17 1970-10-27 Alza Corp Medication method
US3598123A (en) 1969-04-01 1971-08-10 Alza Corp Bandage for administering drugs
US3845770A (en) 1972-06-05 1974-11-05 Alza Corp Osmatic dispensing device for releasing beneficial agent
US3916899A (en) 1973-04-25 1975-11-04 Alza Corp Osmotic dispensing device with maximum and minimum sizes for the passageway
US4008719A (en) 1976-02-02 1977-02-22 Alza Corporation Osmotic system having laminar arrangement for programming delivery of active agent
US4178253A (en) 1977-04-05 1979-12-11 Ciba-Geigy Corporation Corrosion inhibited lubricant compositions
US4269846A (en) 1979-10-29 1981-05-26 Usv Pharmaceutical Corporation Heterocyclic compounds useful as anti-allergy agents
JPS5770820A (en) 1980-10-20 1982-05-01 Haruo Ogura Immunoregulating agent
JPS5910574A (ja) 1982-07-07 1984-01-20 Fujisawa Pharmaceut Co Ltd トリアゾ−ル誘導体およびその製造法
FR2546887B1 (fr) 1983-05-30 1985-08-30 Paris 7 Universite Procede de preparation de dihydro-2,4 triazol-1,2,4 thiones-3 disubstituees en positions 4 et 5 et nouveaux composes pouvant etre prepares par ce procede
IE58110B1 (en) 1984-10-30 1993-07-14 Elan Corp Plc Controlled release powder and process for its preparation
US4624995A (en) 1985-04-09 1986-11-25 Minnesota Mining And Manufacturing Company Triazolinethione-containing polymer
US4740568A (en) 1985-04-09 1988-04-26 Minnesota Mining And Manufacturing Company Triazolinethione-containing polymer
US5436252A (en) 1986-12-19 1995-07-25 Merrell Dow Pharmaceuticals Inc. 5-aryl-3H-1,2,4-triazol-3-ones and their use in the treatment of neurodegenerative disorders
US5006650A (en) 1987-02-11 1991-04-09 The Upjohn Company Novel N-1 substituted beta-lactams as antibiotics
DE3729070A1 (de) 1987-09-01 1989-03-09 Bayer Ag Substituierte triazolinone
US5241074A (en) 1988-05-09 1993-08-31 Bayer Aktiengesellschaft Sulphonylaminocarbonyltriazolinones
US5073543A (en) 1988-07-21 1991-12-17 G. D. Searle & Co. Controlled release formulations of trophic factors in ganglioside-lipsome vehicle
IT1229203B (it) 1989-03-22 1991-07-25 Bioresearch Spa Impiego di acido 5 metiltetraidrofolico, di acido 5 formiltetraidrofolico e dei loro sali farmaceuticamente accettabili per la preparazione di composizioni farmaceutiche in forma a rilascio controllato attive nella terapia dei disturbi mentali organici e composizioni farmaceutiche relative.
US5120548A (en) 1989-11-07 1992-06-09 Merck & Co., Inc. Swelling modulated polymeric drug delivery device
US5733566A (en) 1990-05-15 1998-03-31 Alkermes Controlled Therapeutics Inc. Ii Controlled release of antiparasitic agents in animals
US5219722A (en) 1990-09-21 1993-06-15 Konica Corporation Silver halide color photographic light-sensitive material
SK278998B6 (sk) 1991-02-01 1998-05-06 Merck Sharp & Dohme Limited Deriváty imidazolu, triazolu a tetrazolu, spôsob i
US5356897A (en) 1991-09-09 1994-10-18 Fujisawa Pharmaceutical Co., Ltd. 3-(heteroaryl)-pyrazololi[1,5-a]pyrimidines
US5580578A (en) 1992-01-27 1996-12-03 Euro-Celtique, S.A. Controlled release formulations coated with aqueous dispersions of acrylic polymers
WO1993018029A1 (fr) 1992-03-13 1993-09-16 Merck Sharp & Dohme Limited Derives d'imidazole, de triazole et de tetrazole
TW218017B (fr) 1992-04-28 1993-12-21 Takeda Pharm Industry Co Ltd
DE4222771A1 (de) 1992-07-10 1994-01-13 Bayer Ag Heterocyclyltriazolinone
DE4234801A1 (de) 1992-10-15 1994-04-21 Bayer Ag Sulfonylaminocarbonyltriazolinone
US5591767A (en) 1993-01-25 1997-01-07 Pharmetrix Corporation Liquid reservoir transdermal patch for the administration of ketorolac
DE4303376A1 (de) 1993-02-05 1994-08-11 Bayer Ag Substituierte Triazolinone
DE4303676A1 (de) 1993-02-09 1994-08-11 Bayer Ag 1-Aryltriazolin(thi)one
US5529925A (en) 1993-12-03 1996-06-25 St. Jude Children's Research Hospital Nucleic acid sequences and fusion proteins present in human t(2;5) lymphoma
DE4411913A1 (de) 1994-04-07 1995-10-12 Bayer Ag Substituierte Sulfonylaminocarbonyltriazolinone
US5538988A (en) 1994-04-26 1996-07-23 Martinez; Gregory R. Benzocycloalkylazolethione derivatives
US5489598A (en) 1994-06-08 1996-02-06 Warner-Lambert Company Cytoprotection utilizing aryltriazol-3-thiones
IT1270594B (it) 1994-07-07 1997-05-07 Recordati Chem Pharm Composizione farmaceutica a rilascio controllato di moguisteina in sospensione liquida
JP3372365B2 (ja) 1994-08-19 2003-02-04 富士写真フイルム株式会社 ハロゲン化銀写真感光材料およびそれを用いた画像形成方法
US5525625A (en) 1995-01-24 1996-06-11 Warner-Lambert Company 2-(2-Amino-3-methoxyphenyl)-4-oxo-4H-[1]benzopyran for treating proliferative disorders
DE19502579A1 (de) 1995-01-27 1996-08-01 Bayer Ag Sulfonylamino(thio)carbonyl-triazolin(thi)one
US6080772A (en) 1995-06-07 2000-06-27 Sugen, Inc. Thiazole compounds and methods of modulating signal transduction
DE19521162A1 (de) 1995-06-09 1996-12-12 Bayer Ag N-Aryl-1,2,4-triazolin-5-one
DE19525162A1 (de) 1995-07-11 1997-01-16 Bayer Ag Sulfonylamino(thio)carbonylverbindungen
DE69615376T2 (de) 1995-07-13 2002-09-05 Knoll Gmbh Piperazin-derivate als heilmittel
DE19540737A1 (de) 1995-11-02 1997-05-07 Bayer Ag Substituierte Sulfonylamino(thio)carbonylverbindungen
DE19609059A1 (de) 1996-03-08 1997-09-11 Bayer Ag Substituierte Arylsulfonylamino(thio)carbonyl-triazolin(thi)one
ZA974703B (en) 1996-05-30 1997-12-30 Bayer Ag Substituted sulfonylamino(thio)carbonyl compounds.
TW467902B (en) 1996-07-31 2001-12-11 Bristol Myers Squibb Co Diphenyl heterocycles as potassium channel modulators
DE19632945A1 (de) 1996-08-16 1998-02-19 Bayer Ag Substituierte Sulfonylamino(thio)carbonylverbindungen
CN1237172A (zh) 1996-11-12 1999-12-01 塞普拉科公司 2r,4s,r,s-和2s,4r,r,s-羟基伊曲康唑-和羟基沙波康唑衍生物
US6458373B1 (en) 1997-01-07 2002-10-01 Sonus Pharmaceuticals, Inc. Emulsion vehicle for poorly soluble drugs
US5968921A (en) 1997-10-24 1999-10-19 Orgegon Health Sciences University Compositions and methods for promoting nerve regeneration
JP3788676B2 (ja) 1997-11-11 2006-06-21 富士写真フイルム株式会社 有機エレクトロルミネツセンス素子材料およびそれを使用した有機エレクトロルミネツセンス素子
AR017200A1 (es) 1997-12-23 2001-08-22 Astrazeneca Ab Compuestos inhibidores de la proteina cinasa c, sales farmaceuticamente aceptables de los mismos, formulaciones farmaceuitcas que los comprenden, usode las mismas y proceso para la sintesis de dichos compuestos
EP1062208A1 (fr) 1998-03-09 2000-12-27 Basf Aktiengesellschaft Benzylphenylethers a substitution hetaryle, leur procede de preparation et leur utilisation pour lutter contre les champignons parasites et les parasites animaux
US6747055B1 (en) 1998-07-17 2004-06-08 The United States Of America As Represented By The Department Of Health And Human Services Water-soluble drugs and methods for their production
US6858598B1 (en) 1998-12-23 2005-02-22 G. D. Searle & Co. Method of using a matrix metalloproteinase inhibitor and one or more antineoplastic agents as a combination therapy in the treatment of neoplasia
JP2000284412A (ja) 1999-03-30 2000-10-13 Fuji Photo Film Co Ltd 熱現像写真材料
US6492406B1 (en) 1999-05-21 2002-12-10 Astrazeneca Ab Pharmaceutically active compounds
SE9902387D0 (sv) 1999-06-22 1999-06-22 Astra Ab New pharmaceutically active compounds
US20040110684A1 (en) 1999-08-02 2004-06-10 Universite Catholique De Louvain Novel pharmaceutical compositions for modulating angiogenesis
US6921763B2 (en) 1999-09-17 2005-07-26 Abbott Laboratories Pyrazolopyrimidines as therapeutic agents
AU7989400A (en) 1999-10-01 2001-05-10 Smithkline Beecham Corporation Compounds and methods
AU2001247759A1 (en) 2000-03-24 2001-10-08 Duke University Characterization of grp94-ligand interactions and purification, screening, and therapeutic methods relating thereto
US20030216385A1 (en) 2000-05-19 2003-11-20 Takahiko Tobe Triazole derivatives
US6887853B2 (en) 2000-06-29 2005-05-03 The Trustees Of Boston University Use of geldanamycin and related compounds for treatment of fibrogenic disorders
SI1301472T1 (sl) 2000-07-19 2014-05-30 Warner-Lambert Company Llc Oksigenirani estri 4-jodo fenilamino benzihidroksamskih kislin
US6946456B2 (en) 2000-07-28 2005-09-20 Sloan-Kettering Institute For Cancer Research Methods for treating cell proliferative disorders and viral infections
WO2002036171A1 (fr) 2000-11-02 2002-05-10 Sloan Kettering Institute For Cancer Research Methode permettant d'ameliorer l'efficacite d'agents cytotoxiques a l'aide d'inhibiteurs de la hsp 90
WO2002066447A1 (fr) 2001-02-21 2002-08-29 Ono Pharmaceutical Co., Ltd. Derives de 4h-1,2,4-triazole-3(2h)-thione tenant lieu d'inhibiteurs de sphingomyelinase
EP1423080A4 (fr) 2001-03-01 2009-06-03 Conforma Therapeutics Corp Procede de traitement de troubles proliferatifs d'origine genetique par des inhibiteurs de hsp90
MXPA03008142A (es) 2001-03-09 2003-12-12 Pfizer Prod Inc Nuevos compuestos antiinflamatorios de bencimidazol.
WO2002094259A1 (fr) 2001-05-03 2002-11-28 MAX-PLANCK-Gesellschaft zur Förderung der Wissenschaften e.V. Composes inhibant hsp90 et stimulant hsp70 et hsp40, utiles pour prevenir ou traiter des maladies associees a l'agregation de proteines et a la formation d'amyloides
WO2002094196A2 (fr) 2001-05-23 2002-11-28 Sloan Kettering Institute For Cancer Research Methode de traitement de cancers associes a des niveaux de her-2 eleves
CN1951939A (zh) 2001-05-24 2007-04-25 伊莱利利公司 作为药物的新的吡唑衍生物
JP4102124B2 (ja) 2001-08-01 2008-06-18 富士フイルム株式会社 ハロゲン化銀乳剤およびハロゲン化銀写真感光材料
AU2002330998A1 (en) 2001-08-06 2003-02-24 Kosan Biosciences, Inc. Benzoquinone ansamycins
AU2002323475B2 (en) 2001-09-13 2006-09-28 Synta Pharmaceuticals Corp 2-aroylimidazole compounds for treating cancer
US7473695B2 (en) 2001-10-22 2009-01-06 Mitsubishi Tanabe Pharma Corporation 4-imidazolin-2-one compounds
BR0214553A (pt) 2001-11-28 2004-10-13 Sod Conseils Rech Applic Compostos, processo de preparação, em fase lìquida, dos compostos, e, composições farmacêuticas
US7307071B2 (en) 2001-12-04 2007-12-11 Onyx Pharmaceuticals, Inc RAF-MEK-ERK pathway inhibitors to treat cancer
WO2003050295A2 (fr) 2001-12-12 2003-06-19 Conforma Therapeutics Corporation Essais et mises en oeuvre permettant de determiner et de moduler une activite de liaison hsp90
AU2002356301A1 (en) 2001-12-21 2003-07-15 Cancer Research Technology Ltd. 3,4-diarylpyrazoles and their use in the therapy of cancer
PT2275102E (pt) 2002-03-13 2015-10-27 Array Biopharma Inc Derivados de benzimidazole alquilado n3 como inibidores de mek
GB0207362D0 (en) 2002-03-28 2002-05-08 Univ Liverpool Chemotherapy
GB0208516D0 (en) 2002-04-15 2002-05-22 Univ Liverpool Chemotherapy
JP2006508974A (ja) 2002-11-15 2006-03-16 ワーナー−ランバート・カンパニー、リミテッド、ライアビリティ、カンパニー 癌を治療するためのmek阻害剤およびカペシタビン(capecitabine)を含む併用化学療法
GB0228417D0 (en) 2002-12-05 2003-01-08 Cancer Rec Tech Ltd Pyrazole compounds
GB0229618D0 (en) 2002-12-19 2003-01-22 Cancer Rec Tech Ltd Pyrazole compounds
CA2515726C (fr) 2003-02-11 2012-07-10 Vernalis (Cambridge) Limited Isoxazoles
EP1457499A1 (fr) 2003-03-12 2004-09-15 Tufts University School Of Medicine Inhibiteurs de protéine de choc thermique (HSP90) extracellulaire
EP1605931A4 (fr) 2003-03-13 2009-04-29 Conforma Therapeutics Corp Preparations medicamenteuses contenant des triglycerides a longue chaine et a chaine moyenne
WO2004089415A2 (fr) 2003-04-11 2004-10-21 Novo Nordisk A/S Therapie combinatoire utilisant un inhibiteur de 11$g(b)-hydroxysteroide deshydrogenase de type 1 et agoniste du recepteur de glucocorticoides pour minimiser les effets secondaires associes a la therapie a base d'agoniste du recepteur de glucocorticoides
WO2004089416A2 (fr) 2003-04-11 2004-10-21 Novo Nordisk A/S Polytherapie utilisant un inhibiteur de type 1 de la 11beta-hydroxysteroide deshydrogenase et un agent hypotenseur dans le traitement du syndrome metabolique et des troubles et maladies associes
EP1615637A1 (fr) 2003-04-11 2006-01-18 Novo Nordisk A/S Utilisation pharmaceutique de 1,2,4-triazoles substituees
US7378233B2 (en) 2003-04-12 2008-05-27 The Johns Hopkins University BRAF mutation T1796A in thyroid cancers
DE10318020A1 (de) 2003-04-19 2004-11-11 Repower Systems Ag Gitterturm für eine Windkraftanlage
SE0301232D0 (sv) 2003-04-25 2003-04-25 Astrazeneca Ab Novel use
GB0309637D0 (en) 2003-04-28 2003-06-04 Cancer Rec Tech Ltd Pyrazole compounds
US20050054625A1 (en) 2003-05-30 2005-03-10 Kosan Biosciences, Inc. Method for treating diseases using HSP90-inhibiting agents in combination with nuclear export inhibitors
US20050026893A1 (en) 2003-05-30 2005-02-03 Kosan Biosciences, Inc. Method for treating diseases using HSP90-inhibiting agents in combination with immunosuppressants
US20050020557A1 (en) 2003-05-30 2005-01-27 Kosan Biosciences, Inc. Method for treating diseases using HSP90-inhibiting agents in combination with enzyme inhibitors
US7691838B2 (en) 2003-05-30 2010-04-06 Kosan Biosciences Incorporated Method for treating diseases using HSP90-inhibiting agents in combination with antimitotics
US20050020556A1 (en) * 2003-05-30 2005-01-27 Kosan Biosciences, Inc. Method for treating diseases using HSP90-inhibiting agents in combination with platinum coordination complexes
US20050054589A1 (en) 2003-05-30 2005-03-10 Kosan Biosciences, Inc. Method for treating diseases using HSP90-inhibiting agents in combination with antibiotics
US20050020534A1 (en) 2003-05-30 2005-01-27 Kosan Biosciences, Inc. Method for treating diseases using HSP90-inhibiting agents in combination with antimetabolites
US20050019918A1 (en) 2003-06-03 2005-01-27 Hidetoshi Sumimoto Treatment of cancer by inhibiting BRAF expression
JP4580932B2 (ja) 2003-06-12 2010-11-17 メルク・シャープ・エンド・ドーム・コーポレイション 有糸***キネシン阻害剤のプロドラッグ
GB0315111D0 (en) 2003-06-27 2003-07-30 Cancer Rec Tech Ltd Substituted 5-membered ring compounds and their use
WO2005016920A1 (fr) 2003-08-15 2005-02-24 Vertex Pharmaceuticals Incorporated Compositions de pyrrole convenant comme inhibiteurs de c-met
JP2005084612A (ja) 2003-09-11 2005-03-31 Fuji Photo Film Co Ltd ハロゲン化銀乳剤、ハロゲン化銀感光材料、および熱現像感光材料
WO2005027972A2 (fr) 2003-09-23 2005-03-31 Novartis Ag Polytherapie combinant un inhibiteur des recepteurs du facteur vegf avec un agent chimiotherapeutique
JP2007506763A (ja) 2003-09-24 2007-03-22 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 プロテインキナーゼのインヒビターとして有用なイミダゾール組成物
US20050085531A1 (en) 2003-10-03 2005-04-21 Hodge Carl N. Thiophene-based compounds exhibiting ATP-utilizing enzyme inhibitory activity, and compositions, and uses thereof
CA2542682A1 (fr) 2003-10-18 2005-05-06 Bayer Healthcare Ag Derives de 2-(phenylmethyl)thio-4-phenyl-4h-1,2,4-triazole substitues en 5 et composes associes utilises en tant que gaba-agonistes dans le traitement de l'incontinence urinaire et des maladies associees
WO2005044194A2 (fr) 2003-10-28 2005-05-19 Pharmacia Corporation Traitement ou prevention de la neoplasie a l'aide d'un inhibiteur de la proteine hsp90
JP2007509968A (ja) 2003-10-28 2007-04-19 ファルマシア・コーポレーション 新形成を治療または予防するためのhsp90阻害剤およびホスホジエステラーゼ阻害剤を含む組み合わせ
US6855705B1 (en) 2003-11-12 2005-02-15 Kosan Biosciences, Inc. 11-O-methylgeldanamycin compounds
AU2005222402A1 (en) 2004-03-08 2005-09-22 Wyeth Ion channel modulators
EP1730124A4 (fr) 2004-03-26 2009-04-01 Amphora Discovery Corp Composes et compositions a base de triazoles et leurs applications
WO2005105077A1 (fr) 2004-04-28 2005-11-10 Massachusetts Eye & Ear Infirmary Maladie inflammatoire de l'oeil
EP1766068A4 (fr) 2004-06-04 2010-03-03 Genentech Inc Mutations de egfr
GB0416168D0 (en) 2004-07-20 2004-08-18 Vernalis Cambridge Ltd Pyrmidothiophene compounds
DE102004039280A1 (de) 2004-08-13 2006-02-23 Merck Patent Gmbh 1,5-Diphenyl-pyrazole
WO2006036892A2 (fr) 2004-09-24 2006-04-06 Sapphire Therapeutics, Inc. Utilisation d'inhibiteurs de 24-hydroxylase dans le traitement du cancer
DE102004049078A1 (de) 2004-10-08 2006-04-13 Merck Patent Gmbh Phenylpyrazole
WO2006047631A2 (fr) 2004-10-25 2006-05-04 University Of Medicine And Dentistry Of New Jersey Composes antimitotiques antiproliferatifs
US20060235034A1 (en) 2004-11-01 2006-10-19 Nouri Neamati Novel compounds for treatment of cancer and disorders associated with angiogenesis function
WO2006061712A2 (fr) 2004-12-10 2006-06-15 Pfizer Inc. Utilisation d'inhibiteurs de mek dans le traitement d'une croissance cellulaire anormale
DE102005007304A1 (de) 2005-02-17 2006-08-24 Merck Patent Gmbh Triazolderivate
PT1856057E (pt) 2005-02-25 2015-02-04 Serenex Inc Derivados de tetraidroindolona e tetraidroindazolona, sua composição e usos relacionados
CN101160291B (zh) 2005-03-09 2012-09-05 日本化药株式会社 作为hsp90抑制剂的***衍生物
US8399464B2 (en) 2005-03-09 2013-03-19 Nippon Kayaku Kabushiki Kaisha HSP90 inhibitor
JP2008137894A (ja) 2005-03-22 2008-06-19 Nippon Kayaku Co Ltd 新規なアセチレン誘導体
US7705159B2 (en) 2005-07-06 2010-04-27 Sicor, Inc. Process for the preparation of letrozole
CA2614919C (fr) 2005-07-25 2015-06-23 Zachary Demko Inhibiteurs de 1, 2, 3 triazoles de polymerisation de la tubuline pour le traitement des troubles proliferants
US7473784B2 (en) 2005-08-01 2009-01-06 Bristol-Myers Squibb Company Benzothiazole and azabenzothiazole compounds useful as kinase inhibitors
US7608635B2 (en) 2005-08-12 2009-10-27 Synta Pharmaceuticals Corp. Pyrazole compounds that modulate HSP90 activity
JP5683787B2 (ja) 2005-08-18 2015-03-11 シンタ ファーマシューティカルズ コーポレーション Hsp90活性を調節するイミダゾール化合物
EP1928846B1 (fr) 2005-08-18 2014-06-04 Synta Pharmaceuticals Corp. Dérivés de triazole modulant l'activité de hsp90
ATE453635T1 (de) 2006-03-22 2010-01-15 Vertex Pharma C-met-proteinkinasehemmer zur behandlung proliferativer erkrankungen
EP2447359B1 (fr) 2006-04-14 2015-11-04 Cell Signaling Technology, Inc. Défauts de gène et ALK kinase mutante dans des tumeurs solides humaines
JPWO2007132867A1 (ja) 2006-05-15 2009-09-24 杉本 芳一 癌の予防及び治療剤
DE102006023337A1 (de) 2006-05-18 2007-11-22 Merck Patent Gmbh Triazolderivate II
US20080125587A1 (en) 2006-05-25 2008-05-29 Chimmanamada Dinesh U Synthesis of triazole compounds that modulate HSP90 activity
CA2653217A1 (fr) * 2006-05-25 2007-12-06 Synta Pharmaceuticals Corp. Procede pour traiter des troubles proliferants associes a des produits protooncogenes
JP5441690B2 (ja) 2006-05-25 2014-03-12 シンタ ファーマシューティカルズ コーポレーション Hsp90活性を調節するトリアゾール化合物
AU2007267860B2 (en) 2006-05-25 2012-08-09 Synta Pharmaceuticals Corp. Triazole compounds that modulate Hsp90 activity
AU2012200157B2 (en) 2006-05-25 2014-08-21 Synta Pharmaceuticals Corp. Method for treating proliferative disorders associated with protooncogene products
US20080027047A1 (en) 2006-05-25 2008-01-31 Weiwen Ying Compounds that modulate HSP90 activity and methods for identifying same
TW200804314A (en) 2006-05-25 2008-01-16 Synta Pharmaceuticals Corp Triazole compounds that modulate Hsp90 activity
CA2653336C (fr) 2006-05-25 2014-10-28 Synta Pharmaceuticals Corp. Methode pour traiter un lymphome non hodgkinien
US8188075B2 (en) * 2006-08-17 2012-05-29 Synta Pharmaceuticals Corp. Triazole compounds that modulate HSP90 activity
WO2008051416A2 (fr) 2006-10-19 2008-05-02 Synta Pharmaceuticals Corp. Procede permettant de traiter des infections
WO2008057246A2 (fr) 2006-10-26 2008-05-15 Synta Pharmaceuticals Corp. Procédé de traitement de troubles inflammatoires
DE102007002715A1 (de) 2007-01-18 2008-07-24 Merck Patent Gmbh Triazolderivat
EP2118077B1 (fr) 2007-02-08 2014-12-24 Synta Pharmaceuticals Corp. Composés à base de triazole modulant l'activité de hsp90
JPWO2008108386A1 (ja) 2007-03-05 2010-06-17 協和発酵キリン株式会社 医薬組成物
WO2008112199A1 (fr) 2007-03-12 2008-09-18 Synta Pharmaceuticals Corp. Procédé d'inhibition de la topoisomérase ii
AU2008232354B9 (en) 2007-03-27 2012-07-26 Synta Pharmaceuticals Corp. Triazinone and diazinone derivatives useful as Hsp90 inhibitors
WO2008125633A2 (fr) 2007-04-13 2008-10-23 Rikshospitalet - Radiumhospitalet Hf Traitement et diagnostic du cancer métastatique de la prostate par des inhibiteurs de récepteur de facteur de croissance epidermique (egfr)
US8648104B2 (en) 2007-05-25 2014-02-11 Synta Pharmaceuticals Corp. Method for treating proliferative disorders associated with mutations in c-Met
AU2008267081B2 (en) 2007-06-12 2013-05-09 Provid Pharmaceuticals, Inc. Kinase inhibitors, compositions thereof, and methods of use therewith
US20080318241A1 (en) 2007-06-18 2008-12-25 The Regents Of The University Of Michigan Methods and Systems for Detecting Antiangiogenesis
US7732491B2 (en) 2007-11-12 2010-06-08 Bipar Sciences, Inc. Treatment of breast cancer with a PARP inhibitor alone or in combination with anti-tumor agents
CA2715353A1 (fr) 2008-02-08 2009-08-13 Poniard Pharmaceuticals, Inc. Utilisation de picoplatine et de cetuximab dans le traitement du cancer colorectal
US20110110923A1 (en) 2008-02-12 2011-05-12 The Brigham And Women's Hospital, Inc. Fish assay for eml4 and alk fusion in lung cancer
US20090232906A1 (en) 2008-03-14 2009-09-17 Bionumerik Pharmaceuticals, Inc. Treatment methods and compositions for lung cancer, adenocarcinoma, and other medical conditions
WO2009139916A1 (fr) 2008-05-16 2009-11-19 Synta Pharmaceuticals Corp. Composés triazoles tricycliques modulant l'activité hsp90
US8450500B2 (en) 2008-06-04 2013-05-28 Synta Pharmaceuticals Corp. Pyrrole compounds that modulate HSP90 activity
WO2009158026A1 (fr) 2008-06-27 2009-12-30 Synta Pharmaceuticals Corp. Composés d’hydrazonamide modulant l’activité des hsp90
DK2328893T3 (da) 2008-08-08 2013-06-17 Synta Pharmaceuticals Corp Triazolforbindelser som modulerer hsp90 aktivitet
EP2323737A2 (fr) 2008-08-08 2011-05-25 Synta Pharmaceuticals Corp. Composés de triazole qui modulent l'activité hsp90
CA2734428A1 (fr) 2008-08-18 2010-02-25 Max-Planck-Gesellschaft Zur Foerderung Der Wissenschaften E.V. Susceptibilite aux inhibiteurs hsp90
CN102227221A (zh) 2008-11-28 2011-10-26 诺瓦提斯公司 包含hsp90抑制剂和mtor抑制剂的药物组合
US20100209929A1 (en) 2009-01-14 2010-08-19 Nodality, Inc., A Delaware Corporation Multiple mechanisms for modulation of jak/stat activity
WO2010138377A1 (fr) 2009-05-28 2010-12-02 Merck Sharp & Dohme Corp. Compositions et procédés de traitement du cancer
RS53716B1 (en) 2009-10-19 2015-04-30 Synta Pharmaceuticals Corp. COMBINED CANCER THERAPY WITH HSP90 INHIBITORS
EP2499486A4 (fr) 2009-11-13 2013-11-27 Infinity Pharmaceuticals Inc Compositions, kits, et procédés pour l'identification, l'évaluation, la prévention, et la thérapie d'un cancer
EP2333103A1 (fr) 2009-12-11 2011-06-15 Max-Planck-Gesellschaft zur Förderung der Wissenschaften e.V. Diagnostic différentiel et thérapie pour les inhibiteurs de la kinase
EP2560640A1 (fr) 2010-04-19 2013-02-27 Synta Pharmaceuticals Corp. Thérapie anticancéreuse à l'aide d'une combinaison d'un composé inhibiteur de hsp90 et d'un inhibiteur d'egfr
WO2011133521A2 (fr) 2010-04-19 2011-10-27 Synta Pharmaceuticals Corp. Traitement anticancéreux utilisant une combinaison de composés inhibiteurs de hsp90 et un inhibiteur de vegf
WO2011146803A1 (fr) 2010-05-20 2011-11-24 Synta Pharmaceuticals Corp. Méthode de traitement d'adénocarcinomes pulmonaires par composés inhibiteurs de hsp90
WO2011146801A1 (fr) 2010-05-20 2011-11-24 Synta Pharmaceuticals Corp. Formulation et analyse de composés inhibiteurs de hsp90
EP2575810A1 (fr) 2010-05-24 2013-04-10 Synta Pharmaceuticals Corp. Traitement anticancéreux utilisant une combinaison d'un composé inhibiteur de hsp90 et d'un inhibiteur de la topoisomérase ii
WO2012026931A1 (fr) 2010-08-25 2012-03-01 Synta Pharmaceuticals Corp. Procédé de synthèse de 2-alkylphénols substitués
EP2616063A1 (fr) 2010-09-13 2013-07-24 Synta Pharmaceuticals Corp. Inhibiteurs de hsp90 pour le traitement de cancers du poumon non à petites cellules chez des patients présentant un egfr et/ou kras de type sauvage
WO2012068487A1 (fr) 2010-11-18 2012-05-24 Synta Pharmaceuticals Corp. Présélection de patients pour un traitement thérapeutique fondé sur un état hypoxique, avec des agents sensibles à l'oxygène
EP2640385A1 (fr) 2010-11-18 2013-09-25 Synta Pharmaceuticals Corp. Présélection de patients pour un traitement thérapeutique fondé sur un état hypoxique
WO2012078757A2 (fr) 2010-12-08 2012-06-14 Synta Pharmaceuticals Corp. Traitement combinatoire du cancer du sein avec des composés inhibiteurs d'hsp90
EP2663305A1 (fr) 2011-01-11 2013-11-20 Synta Pharmaceuticals Corp. Plurithérapie associant des composés inhibiteurs de hsp90 à des inhibiteurs du protéasome
US20140051664A1 (en) 2011-02-23 2014-02-20 Synta Pharmaceuticals Corp. Combination therapy of hsp90 inhibitory compounds with radiotherapy
US20140051665A1 (en) 2011-02-24 2014-02-20 Synta Pharmaceuticals Corp. Prostate cancer therapy with hsp90 inhibitory compounds
US20140045908A1 (en) 2011-02-25 2014-02-13 Synta Pharmaceuticals Corp. Hsp90 inhibitory compounds in treating jak/stat signaling-mediated cancers
CA2820709C (fr) 2011-02-28 2016-02-16 Sunshine Lake Pharma Co., Ltd. Quinoleines substituees et leurs methodes d'utilisation
WO2012155063A1 (fr) 2011-05-11 2012-11-15 Synta Pharmaceuticals Corp. Traitement du cancer avec un composé inhibiteur de hsp90
EP2714039A1 (fr) 2011-05-23 2014-04-09 Synta Pharmaceuticals Corp. Polythérapie de composés inhibiteurs de hsp90 avec inhibiteurs de mek
US20140315943A1 (en) 2011-05-24 2014-10-23 Synta Pharmaceuticals Corp. Combination therapy of hsp90 inhibitory compounds with mtor/p13k inhibitors
US20140194388A1 (en) 2011-05-26 2014-07-10 Synta Pharmaceuticals Corp. Combination therapy of hsp90 inhibitory compounds with chk inhibitors
EP2729144A2 (fr) 2011-07-07 2014-05-14 Synta Pharmaceuticals Corp. Traitement du cancer au moyen de composés inhibiteurs de hsp90
WO2013028505A1 (fr) 2011-08-19 2013-02-28 Synta Pharmaceuticals Corp. Polythérapie anticancéreuse d'inhibiteur de hsp90 comprenant un anti-métabolite
US8628752B2 (en) 2011-10-28 2014-01-14 Synta Pharmaceuticals Corp. Methods of identifying HSP90 inhibitors with less ocular toxicity
CA2853799A1 (fr) 2011-11-02 2013-05-10 Synta Pharmaceuticals Corp. Therapie anticancereuse utilisant une combinaison d'inhibiteurs de hsp 90 et d'inhibiteurs de topoisomerase i
US9402831B2 (en) 2011-11-14 2016-08-02 Synta Pharmaceutical Corp. Combination therapy of HSP90 inhibitors with BRAF inhibitors
CA2868258A1 (fr) 2012-03-28 2013-10-03 Synta Pharmaceuticals Corp. Derives de triazole comme inhibiteurs de hsp90
CA2868323A1 (fr) 2012-04-04 2013-10-10 Synta Pharmaceuticals Corp. Nouveaux composes triazoles qui modulent l'activite hsp90
CA2871540A1 (fr) 2012-05-10 2013-11-14 Synta Pharmaceuticals Corp. Traitement du cancer avec des composes inhibiteurs de hsp90
US10155987B2 (en) 2012-06-12 2018-12-18 Dana-Farber Cancer Institute, Inc. Methods of predicting resistance to JAK inhibitor therapy
US20150283147A1 (en) 2012-10-19 2015-10-08 Synta Pharmaceuticals Corp. Treating polycystic kidney disease with hsp90 inhibitory compounds
US20160120848A1 (en) 2013-05-21 2016-05-05 Synta Pharmaceuticals Corp. Specific cancer treatment regimes with ganetespib
WO2015109218A1 (fr) 2014-01-17 2015-07-23 Synta Pharmaceuticals Corp. Traitement ciblé du cancer par ganetespib et de nvp-auy922 inhibiteurs d'hsp90

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7825148B2 (en) * 2004-11-18 2010-11-02 Synta Pharmaceuticals Corp. Triazole compounds that modulate Hsp90 activity
US8742133B2 (en) * 2007-08-13 2014-06-03 Synta Pharmaceuticals Corp. Triazole compounds that modulate HSP90 activity

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9006277B2 (en) 2006-05-25 2015-04-14 Synta Pharmaceuticals Corp. Triazole compounds that modulate HSP90 activity
US9206162B2 (en) 2006-05-25 2015-12-08 Synta Pharmaceuticals Corp. Triazole compounds that modulate Hsp90 activity
US9539243B2 (en) 2008-08-08 2017-01-10 Synta Pharmaceuticals Corp. Triazole compounds that modulate HSP90 activity
US9205086B2 (en) 2010-04-19 2015-12-08 Synta Pharmaceuticals Corp. Cancer therapy using a combination of a Hsp90 inhibitory compounds and a EGFR inhibitor
US9439899B2 (en) 2011-11-02 2016-09-13 Synta Pharmaceuticals Corp. Cancer therapy using a combination of HSP90 inhibitors with topoisomerase I inhibitors
US9402831B2 (en) 2011-11-14 2016-08-02 Synta Pharmaceutical Corp. Combination therapy of HSP90 inhibitors with BRAF inhibitors

Also Published As

Publication number Publication date
WO2013067165A1 (fr) 2013-05-10
US10500193B2 (en) 2019-12-10
AU2012332424A1 (en) 2014-06-05
US20180221345A1 (en) 2018-08-09
CA2853806A1 (fr) 2013-05-10
CA2853806C (fr) 2020-07-14
EP2776025A1 (fr) 2014-09-17
JP2014534228A (ja) 2014-12-18

Similar Documents

Publication Publication Date Title
US10500193B2 (en) Combination therapy of HSP90 inhibitors with platinum-containing agents
US9439899B2 (en) Cancer therapy using a combination of HSP90 inhibitors with topoisomerase I inhibitors
US9402831B2 (en) Combination therapy of HSP90 inhibitors with BRAF inhibitors
US20130171105A1 (en) Cancer therapy using a combination of a hsp90 inhibitory compound and a topoisomerase ii inhibitor
US9205086B2 (en) Cancer therapy using a combination of a Hsp90 inhibitory compounds and a EGFR inhibitor
AU2011302344B2 (en) HSP90 inhibitors for treating non-small cell lung cancers in wild-type EGFR and/or KRAS patients
US20150099721A1 (en) Treating cancer with hsp90 inhibitory compounds
US20140315943A1 (en) Combination therapy of hsp90 inhibitory compounds with mtor/p13k inhibitors
US20140228418A1 (en) Combination therapy of hsp90 inhibitory compounds with mek inhibitors
US20150150850A1 (en) Treating cancer with hsp90 inhibitory compounds
US20130331357A1 (en) Combination therapy of hsp90 inhibitory compounds with proteasome inhibitors
US20140194388A1 (en) Combination therapy of hsp90 inhibitory compounds with chk inhibitors

Legal Events

Date Code Title Description
AS Assignment

Owner name: SYNTA PHARMACEUTICALS CORP., MASSACHUSETTS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:PROIA, DAVID;REEL/FRAME:035163/0061

Effective date: 20150312

STCB Information on status: application discontinuation

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