WO2009032703A1 - 2- (het) arylamino-6-aminopyridine derivatives and fused forms thereof as anaplastic lymphoma kinase inhibitors - Google Patents

2- (het) arylamino-6-aminopyridine derivatives and fused forms thereof as anaplastic lymphoma kinase inhibitors Download PDF

Info

Publication number
WO2009032703A1
WO2009032703A1 PCT/US2008/074490 US2008074490W WO2009032703A1 WO 2009032703 A1 WO2009032703 A1 WO 2009032703A1 US 2008074490 W US2008074490 W US 2008074490W WO 2009032703 A1 WO2009032703 A1 WO 2009032703A1
Authority
WO
WIPO (PCT)
Prior art keywords
phenyl
chloro
ylamino
phenylamino
isopropylsulfonyl
Prior art date
Application number
PCT/US2008/074490
Other languages
French (fr)
Inventor
Nathanael S. Gray
Pierre-Yves Michellys
Wei Pei
Thomas H. Marsilje
Wenshuo Lu
Bei Chen
Tetsuo Uno
Yunho Jin
Tao Jiang
Original Assignee
Irm Llc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Irm Llc filed Critical Irm Llc
Publication of WO2009032703A1 publication Critical patent/WO2009032703A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/46Two or more oxygen, sulphur or nitrogen atoms
    • C07D239/50Three nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Definitions

  • Cancer is a disease resulting from an abnormal growth of tissue. Certain cancers have the potential to invade into local tissues and also metastasize to distant organs. This disease can develop in a wide variety of different organs, tissues and cell types. Therefore, the term “cancer” refers to a collection of over a thousand different diseases.
  • Anaplastic lymphoma kinase (ALK), a member of the insulin receptor superfamily of receptor tyrosine kinases, has been implicated in oncogenesis in hematopoietic and non- hematopoietic tumors.
  • ALK insulin receptor superfamily of receptor tyrosine kinases
  • the aberrant expression of full-length ALK receptor proteins has been reported in neuroblastomas and glioblastomas; and ALK fusion proteins have occurred in anaplastic large cell lymphoma.
  • the study of ALK fusion proteins has also raised the possibility of new therapeutic treatments for patients with ALK-positive malignancies. (Pulford et al., Cell. MoI. Life Sci. 61:2939-2953 (2004)).
  • IGF-I Insulin- like growth factor
  • IGF-IR IGF-I receptor
  • ROSl c-ros oncogene 1
  • ROS a member of the tyrosine kinase insulin receptor gene family
  • mitotic assembly checkpoint kinase MPS 1 also known as TTK
  • TTK mitotic assembly checkpoint kinase
  • Normal cells have multiple redundant mechanisms for arresting in mitosis whereas cancer cells have a heightened dependence on TTK activity.
  • the invention relates to novel pyrimidine and pyridine derivatives and pharmaceutical compositions thereof, and their use as pharmaceuticals.
  • the invention provides compounds having the Formula (I): pa
  • a and B are each independently aryl or heteroaryl, each of which may be optionally substituted with one or more R4 groups;
  • Y is -SO2-, -SO2NH-, -NH-SO2-, -NH-C(O)-, -C(O)-NH-, -O-, or -NR2-;
  • X is NH, O or S
  • Z is N or CH
  • Rl is H, C1-C6 alkyl, halo-(Cl-C6 alkyl), C1-C6 cycloalkyl, halo-(Cl-C6 cycloalkyl), heterocyclyl, heterocyclylCl- ⁇ alkyl, aryl, arylCl- ⁇ alkyl, heteroaryl or heteroarylCl- ⁇ alkyl;
  • R2 is H, C1-C6 alkyl, halo-(Cl-C6 alkyl), C1-C6 cycloalkyl, halo-(Cl-C6 cycloalkyl), heterocyclyl, heterocyclylCl- ⁇ alkyl, aryl, arylCl- ⁇ alkyl, heteroaryl or heteroarylCl- ⁇ alkyl; R3
  • Ra is hydrogen and Rb is halogen, or Ra and Rb, taken together with the atoms to which they are bound form a pyrazolo or a pyrrolo ring fused to the pyrimidine ring, said pyrazolo or pyrrolo ring optionally bearing one or two R4 groups;
  • R4 is independently halogen, C1-C6 alkyl, halo-(Cl-C6 alkyl), Cl- C6 cycloalkyl, halo-(Cl-C6 cycloalkyl), heterocyclyl, heterocyclylCl- ⁇ alkyl, aryl, arylCl- ⁇ alkyl, heteroaryl or heteroarylCl- ⁇ alkyl, Cl-6alkoxy, Cl-6alkylthio, hydroxyl, nitro, azido, cyano, acyloxy, carboxy, ester, carbamoyl, carboxamide, ureido, amidino, guanidine, sulfonyl, sulphonylamino, aminosulphonyl;
  • R5 is independently is C1-C6 alkoxy, ⁇ — / , hydroxy, ⁇ — N N-(C 1 -C 6 alkyl) , dialkylamino, or -N-R7;
  • R6 is independently is hydroxy, ⁇ — ' , Nv — ⁇ , or -
  • R7 is independently is hydrogen, C1-C6 alkyl, C1-C6 alkoxy, hydroxyl, or C1-C6 hydroxyalkyl;
  • R 3 or R 4 if present in B are not nitro, azido, ureido, guanidine or sulphonylamino;
  • R 3 or R 4 if present in B are independently halogen, Ci-C 6 alkyl, halo-(d- Ce alkyl), C 3 -C 7 cycloalkyl, halo-(C 3 -C 7 cycloalkyl), heterocyclyl, heterocyclylCi_ 6 alkyl, aryl, arylCi_ 6 alkyl, heteroaryl or heteroarylCi_ 6 alkyl.
  • the invention provides compounds having Formula IA or III:
  • R 3 is -CO-R 5 , * alternatively, R 3 or R 4 if present in B are independently halogen, Ci-C 6 alkyl, halo-(d-
  • Y, X, R 1 , R 2 , R 5 , R 7 , R a and R b are as defined in Formula I.
  • the invention provides compounds having Formula III:
  • R 3 or R 4 if present in B are independently halogen, Ci-C 6 alkyl, 1IaIo-(C 1 - C 6 alkyl), C 3 -C 7 cycloalkyl, halo-(C 3 -C 7 cycloalkyl), heterocyclyl, heterocyclylCi_ 6 alkyl, aryl, arylCi_ 6 alkyl, heteroaryl or heteroarylCi_ 6 alkyl; n is an integer from 0-3; m is an integer from 0-4; p is an integer from 0-2; and
  • Y, X, R 1 , R 2 , R 5 , R 7 are as defined in Formula I.
  • the invention encompasses a compound of Formula I, IA, II or III, wherein X is O. In still other embodiments, the invention encompasses a compound of Formula I, IA, II or III, wherein R 2 is methyl, ethyl, propyl, isopropyl, cyclopropyl or cyclobutyl. In yet other embodiments, the invention encompasses a compound of Formula I, IA, II or III, wherein R is methyl. In another embodiment , the invention encompasses a compound of Formula I, IA, II or III, wherein Y is SO 2 .
  • the invention encompasses a compound of Formula I, IA, II or III, wherein R 1 is methyl, ethyl, propyl, isopropyl, cyclopropyl or cyclobutyl.
  • the invention encompasses a compound of Formula I, IA, II or III, wherein R 1 is isopropyl.
  • the invention encompasses a compound of Formula I, IA, II or III, wherein X is O, R 2 is methyl, Y is SO 2 and R 1 is isopropyl.
  • the invention provides compounds having Formula I wherein Z is N, and said compound is selected from the group consisting of: methyl 3-(4-amino-5-chloro-6-(2-(methylsulfonyl)phenylamino)pyrimidin-2-ylamino)-4- methoxybenzoate ;
  • the invention provides compounds having Formula I wherein Z is CH and said compound is selected from the group consisting of: l-(4-(4-(2-(isopropylsulfonyl)phenylamino)-lH-pyrrolo[2,3-b]pyridin-6-ylamino)-3- methoxyphenyl)piperidin-4-ol;
  • the invention provides compounds having Formula (III) and selected from the group consisting of:
  • Another aspect of the invention encompasses a pharmaceutical composition
  • a pharmaceutical composition comprising a compound according to Formula I, IA, II or III or a physiologically acceptable salt, solvate, hydrate or stereoisomer thereof and a pharmaceutically acceptable diluent or carrier.
  • the pharmaceutical composition the invention encompasses a composition wherein the compound is present in a therapeutically effective amount.
  • the pharmaceutical composition the invention further encompasses at least one further active compound, such as for example, an anti-hyperproliferative agent.
  • Another aspect of the invention encompasses a packaged pharmaceutical composition comprising a container, the pharmaceutical composition of the invention and instructions for using the pharmaceutical composition to treat a disease or condition in a mammal.
  • Yet another aspect of the invention encompasses a method of inhibiting kinase activity in a cell comprising contacting a cell with one or more compounds of the invention.
  • the kinase whose activity is inhibited is anaplastic lymphoma kinase, (ALK) hepatocyte growth factor receptor tyrosine kinase (c-Met), monopolar spindle (Mpsl) kinase, Ros, IGF-IR or InsR kinase.
  • ALK activity is inhibited.
  • c-Met or Mpsl kinase activity is inhibited.
  • the invention also provides methods for treating a condition mediated by ALK, c- Met, Mpsl, Ros, IGFlR or InsR, comprising administering to a cell or tissue system or to a mammalian subject, a therapeutically effective amount of a compound of Formula I, IA, II or III, or pharmaceutically acceptable salts or tautomers thereof, and optionally in combination with a second therapeutic agent; thereby treating said condition mediated by ALK, c-Met, Mpsl, Ros, IGFlR or InsR.
  • the compounds of the invention may be used for treating a condition mediated by ALK.
  • the present invention provides the use of a compound having Formula I, IA, II or III, in the manufacture of a medicament for treating a condition mediated by ALK, c- Met, Mpsl, Ros, IGFlR or InsR.
  • the compounds of the invention may be used alone or in combination with a second therapeutic agent to treat a condition mediated by ALK, c-Met, Mpsl, Ros, IGFlR or InsR, wherein said condition is an autoimmune disease, a transplantation disease, an infectious disease or a cell proliferative disorder.
  • the invention provides methods for treating a cell proliferative or angiogenesis disorder, comprising administering to a system or subject in need of such treatment an effective amount of a compound having Formula I, IA, II or III, or pharmaceutically acceptable salts or pharmaceutical compositions thereof, and optionally in combination with a second therapeutic agent, thereby treating said disorder.
  • the present invention provides the use of a compound having Formula I, IA, II or III, in the manufacture of a medicament for treating a cell-proliferative or angiogenesis disorder.
  • the compounds of the invention may be used alone or in combination with a chemotherapeutic agent to treat a cell proliferative disorder, including but not limited to, multiple myeloma, neuroblastoma, lymphoma, leukemia, melanoma, sarcoma, osteosarcoma, synovial sarcoma, Ewing's sarcoma, hepatoma, gastrointestinal stromal tumor or a solid tumor or cancer of breast, renal, prostate, colorectal, thyroid, ovarian, pancreas, lung, uterus, respiratory tract, brain, digestive tract, urinary tract, eye, liver, skin, head and neck, thyroid or parathyroid.
  • a chemotherapeutic agent to treat a cell proliferative disorder, including but not limited to, multiple myeloma, neuroblastoma, lymphoma, leukemia, melanoma, sarcoma, osteosarcoma, synovial sarcoma, Ewing'
  • a compound having Formula I, IA, II or III may be administered to a system comprising cells or tissues, or to a mammalian subject such as a human or animal subject.
  • halogen refers to radicals of fluorine, chlorine, bromine and iodine.
  • alkyl refers to a straight or branched hydrocarbon chain radical, containing solely carbon and hydrogen atoms, having in the range from one up to eight carbon atoms, and which is attached to the rest of the molecule by a single bond, such as illustratively, methyl, ethyl, n-propyl 1-methylethyl (iso-propyl), n-butyl, n-pentyl, and 1,1-dimethylethyl (tert-butyl).
  • cycloalkyl denotes a non-aromatic mono or multicyclic ring system having in the range of 3 up to 14 carbon atoms such as cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl.
  • multicyclic cycloalkyl groups include decahydronapththyl.
  • bridged cycloalkyl groups or sprirobicycloalkyl groups include adamantyl norbornyl, and spiro[4.4]nonyl groups.
  • Examples of carbocyclic rings include but are not limited to cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclopropylene, cyclohexanone, etc.
  • alkoxy denotes an alkyl group as defined herein attached via an oxygen linkage to the rest of the molecule. Representative examples of those groups are methoxy, ethoxy, iso-propoxy, n-butoxy, and tert-butoxy.
  • cycloalkoxy denotes a cycloalkyl group as defined herein attached via an oxygen linkage to the rest of the molecule. Representative examples of those groups are cyclopropoxy, cyclobutoxy, cyclopentoxy, cyclohexoxy, and cycloheptoxy.
  • aryl refers to aromatic radicals having in the range of 6 up to 14 carbon atoms such as phenyl, naphthyl, indanyl, and biphenyl.
  • heteroaryl refers to a stable 5- to 13-membered aromatic heterocycle having in the range of from 1 up to 4 heteroatoms from the group consisting of nitrogen, phosphorus, oxygen and sulfur, which ring or ring system can be linked via a carbon atom or a nitrogen atom, if such an atom is present.
  • the heteroaryl ring radical may be a monocyclic, bicyclic or tricyclic ring system.
  • heteroaryl radicals examples include: pyridyl, pyridyl N-oxide, pyrimidyl, pyridazinyl, pyrazinyl, thienyl, furyl, pyrrolyl, pyrazolyl, imidazolyl, thiazolyl, oxazolyl or isoxazolyl, indolicenyl, indolyl, benzo[b] thienyl, benzo[b]furyl, benzothiazolyl, benzothiadiazolyl, indazolyl, quinolyl, isoquinolyl, isoquinolyl, naphthyridinyl, quinazolinyl, oxadiazolyl, benzoxazolyl, tetrazoyl, triazolyl, thiadiazolyl, and benzimidazolyl.
  • heterocycloalkyl refers to a stable 3 to 13 membered saturated or partially unsaturated heterocycle having in the range from 1 up to 4 heteroatoms from the group consisting of nitrogen, phosphorus, oxygen and sulfur, which ring or ring system can be linked via a carbon atom or a nitrogen atom, if such an atom is present.
  • the heterocyclic ring radical may be a monocyclic, bicyclic or tricyclic ring system, which may include fused, bridged or spiro ring systems.
  • heterocyclyl radicals examples include: tetrahydropyranyl, aziridyl, azepanyl, tetrahydrofuryl, pyrrolidinyl, pyrrolinyl, piperidinyl, 1,2 dihydropyridinyl, 1,4 dihydropyridinyl, piperazinyl, morpholinyl, thiomorpholinyl, azepinyl, oxazolinyl, thiazolinyl and 1,4 diazepinyl.
  • nitrogen non-aromatic heterocyclyl radicals include but are not limited to piperidinyl, piperazinyl, pyrrolidinyl, azetidinyl, morpholinyl, etc.
  • alkylamino refers to an alkyl group as defined herein attached via amino linkage to the rest of the molecule.
  • alkylamino further includes dialkyl amino moieties in which two alkyl groups as define herein are attached via amino linkage to the rest of the molecule. Representative examples of those groups are methylamino and dimethylamino.
  • an H atom in any substituent groups encompasses all suitable isotopic variations, e.g., H, 2 H and 3 H.
  • a solvate for the purpose of this invention is a complex of a solvent and a compound of the invention in the solid state.
  • Exemplary solvates would include, but are not limited to, complexes of a compound of the invention with ethanol or methanol. Hydrates are a specific form of solvate wherein the solvent is water.
  • treating or “treatment” as used herein is used conventionally, e.g., the management or care of a subject for the purpose of combating, alleviating, reducing, relieving, improving the condition of, etc., of a disease or disorder, such as a carcinoma or cancer.
  • the present invention is based on the discovery that certain pyrimidine and pyrrolopyridine derivatives possess valuable, pharmacologically useful properties.
  • compounds and compositions described herein including salts, metabolites, solvates, solvates of salts, hydrates, prodrugs such as esters, polymorphs, and stereoisomeric forms thereof, may exhibit anti-proliferative activity and are thus useful to prevent or treat the disorders associated with hyper-proliferation.
  • the compounds of the invention are effective as protein tyrosine kinase inhibitors.
  • the compounds of the invention may inhibit the tyrosine kinase activity of anaplastic lymphoma kinase (ALK) and/or the fusion protein of NPM-ALK .
  • ALK anaplastic lymphoma kinase
  • NPM-ALK fusion protein of NPM-ALK .
  • NPM-ALK plays a key role in signal transmission in a number of hematopoetic and other human cells leading to hematological and neoplastic diseases, for example in anaplastic large-cell lymphoma (ALCL) and non-Hodgkin's lymphomas (NHL), specifically in ALK+NHL or Alkomas, in inflammatory myofibroblastic tumors (IMT) and neuroblastomas.
  • ACL anaplastic large-cell lymphoma
  • NHL non-Hodgkin's lymphomas
  • IMT myofibroblastic tumors
  • other gene fusions have been identified in human hematological and neoplastic diseases; mainly TPM3-ALK (a fusion of nonmuscle tropomyosin with ALK).
  • EML4- ALK fusion tyrosine kinase generated transformed foci in culture and subcutaneous tumors in mice. Therefore EML4-ALK, which is inhibited by the compounds described in this application, maybe a relevant target for NSCLC patients or other patients with this or related ALK-containing fusion proteins (Nature. 2007 Aug 2;448(7153):561-6)
  • the pyrimidine derivatives are useful for the inhibition of all such ALK-containing gene fusions.
  • a and B are each independently aryl or heteroaryl, each of which may be optionally substituted with one or more R4 groups;
  • Y is -SO2-, -SO2NH-, -NH-SO2-, -NH-C(O)-, -C(O)-NH-, -O-, or -NR2-;
  • X is NH, O or S
  • Z is N or CH
  • Rl is H, C1-C6 alkyl, halo-(Cl-C6 alkyl), C1-C6 cycloalkyl, halo-(Cl-C6 cycloalkyl), heterocyclyl, heterocyclylCl- ⁇ alkyl, aryl, arylCl- ⁇ alkyl, heteroaryl or heteroarylCl- ⁇ alkyl;
  • R2 is H, C1-C6 alkyl, halo-(Cl-C6 alkyl), C1-C6 cycloalkyl, halo-(Cl-C6 cycloalkyl), heterocyclyl, heterocyclylCl- ⁇ alkyl, aryl, arylCl- ⁇ alkyl, heteroaryl or heteroarylCl- ⁇ alkyl;
  • R3 is R4, -CO-R5,
  • Ra is hydrogen and Rb is halogen, or Ra and Rb, taken together with the atoms to which they are bound form a pyrazolo or a pyrrolo ring fused to the pyrimidine ring, said pyrazolo or pyrrolo ring optionally bearing one or two R4 groups;
  • R4 is independently halogen, C1-C6 alkyl, halo-(Cl-C6 alkyl), Cl- C6 cycloalkyl, halo-(Cl-C6 cycloalkyl), heterocyclyl, heterocyclylCl- ⁇ alkyl, aryl, arylCl- ⁇ alkyl, heteroaryl or heteroarylCl- ⁇ alkyl, Cl-6alkoxy, Cl-6alkylthio, hydroxyl, nitro, azido, cyano, acyloxy, carboxy, ester, carbamoyl, carboxamide, ureido, amidino, guanidine, sulfonyl, sulphonylamino, aminosulphonyl;
  • R5 is independently is C1-C6 alkoxy, ⁇ — / , hydroxy, N N-(C 1 -C 6 alkyl) , dialkylamino, or -N-R7;
  • R6 is independently is hydroxy, " ⁇ — ' , ⁇ — ⁇ , or -
  • R7 is independently is hydrogen, C1-C6 alkyl, C1-C6 alkoxy, hydroxyl, or C1-C6 hydroxyalkyl;
  • -X-R2 and -R3 may each independently represent hydrogen.
  • the invention encompasses a compound having the formula IA: wherein: n is an integer from 0-3; m is an integer from 0-4; and Y, X, R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R a and R b are as defined in Formula I. [0043] In yet another embodiment, the invention encompasses a compound having the formula II:
  • the invention encompasses a compound having the formula III:
  • Another aspect of the invention encompasses a compound having the IUPAC name: methyl 3-(4-amino-5-chloro-6-(2-(methylsulfonyl)phenylamino)pyrimidin-2-ylamino)-4- methoxybenzoate ;
  • Still another aspect of the invention encompasses a compound having the IUPAC name: l-(4-(4-(2-(isopropylsulfonyl)phenylamino)-lH-pyrazolo[3,4-d]pyrimidin-6-ylamino)-3- methoxyphenyl)piperidin-4-ol; l-(3-ethoxy-4-(4-(2-(isopropylsulfonyl)phenylamino)-lH-pyrazolo[3,4-d]pyrimidin-6- ylamino)phenyl)piperidin-4-ol;
  • Yet another aspect of the invention encompasses a compound having the IUPAC name: methyl 6-(4-(2-(isopropylsulfonyl)phenylamino)-lH-pyrrolo[2,3-b]pyridin-6- ylamino)nicotinate;
  • the compounds of this invention may contain one or more asymmetric centers, depending upon the location and nature of the various substituents desired.
  • Asymmetric carbon atoms may be present in the (R) or (S) configuration, resulting in racemic mixtures in the case of a single asymmetric center, and diastereomeric mixtures in the case of multiple asymmetric centers.
  • asymmetry may also be present due to restricted rotation about a given bond, for example, the central bond adjoining two substituted aromatic rings of the specified compounds.
  • Substituents on a ring may also be present in either cis or trans form. It is intended that all such configurations (including enantiomers and diastereomers), are included within the scope of the present invention.
  • the optical isomers can be obtained by resolution of the racemic mixtures according to conventional processes, for example, by the formation of diastereoisomeric salts using an optically active acid or base or formation of covalent diastereomers.
  • appropriate acids are tartaric, diacetyltartaric, ditoluoyltartaric and camphorsulfonic acid.
  • Mixtures of diastereoisomers can be separated into their individual diastereomers on the basis of their physical and/or chemical differences by methods known in the art, for example, by chromatography or fractional crystallization.
  • the optically active bases or acids are then liberated from the separated diastereomeric salts.
  • a different process for separation of optical isomers involves the use of chiral chromatography (e.g., chiral HPLC columns), with or without conventional derivitization, optimally chosen to maximize the separation of the enantiomers.
  • Suitable chiral HPLC columns are manufactured by Diacel, e.g., Chiracel OD and Chiracel OJ among many others, all routinely selectable.
  • Enzymatic separations, with or without derivitization are also useful.
  • the optically active compounds of this invention can likewise be obtained by chiral syntheses utilizing optically active starting materials.
  • the present invention also relates to useful forms of the compounds as disclosed herein, such as pharmaceutically acceptable salts, co-precipitates, metabolites, hydrates, solvates and prodrugs of all the compounds of examples.
  • pharmaceutically acceptable salt refers to a relatively non-toxic, inorganic or organic acid addition salt of a compound of the present invention. For example, see S. M. Berge, et al. "Pharmaceutical Salts," J. Pharm. Sci. 1977, 66, 1-19.
  • Pharmaceutically acceptable salts include those obtained by reacting the main compound, functioning as a base, with an inorganic or organic acid to form a salt, for example, salts of hydrochloric acid, sulfuric acid, phosphoric acid, methane sulfonic acid, camphor sulfonic acid, oxalic acid, maleic acid, succinic acid and citric acid.
  • Pharmaceutically acceptable salts also include those in which the main compound functions as an acid and is reacted with an appropriate base to form, e.g., sodium, potassium, calcium, magnesium, ammonium, and chorine salts.
  • acid addition salts of the claimed compounds may be prepared by reaction of the compounds with the appropriate inorganic or organic acid via any of a number of known methods.
  • alkali and alkaline earth metal salts of acidic compounds of the invention are prepared by reacting the compounds of the invention with the appropriate base via a variety of known methods.
  • Representative salts of the compounds of this invention include the conventional nontoxic salts and the quaternary ammonium salts which are formed, for example, from inorganic or organic acids or bases by means well known in the art.
  • acid addition salts include acetate, adipate, alginate, ascorbate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate, cinnamate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptanoate, glycerophosphate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, 2- hydroxyethanesulfonate, itaconate, lactate, maleate, mandelate, methanesul
  • Base salts include alkali metal salts such as potassium and sodium salts, alkaline earth metal salts such as calcium and magnesium salts, and ammonium salts with organic bases such as dicyclohexylamine and N-methyl-D-glucamine.
  • basic nitrogen containing groups may be quaternized with such agents as lower alkyl halides such as methyl, ethyl, propyl, and butyl chlorides, bromides and iodides; dialkyl sulfates like dimethyl, diethyl, and dibutyl sulfate; and diamyl sulfates, long chain halides such as decyl, lauryl, myristyl and strearyl chlorides, bromides and iodides, aralkyl halides like benzyl and phenethyl bromides and others.
  • lower alkyl halides such as methyl, ethyl, propyl, and butyl chlorides, bromides and iodides
  • dialkyl sulfates like dimethyl, diethyl, and dibutyl sulfate
  • diamyl sulfates long chain halides such as decyl, lauryl
  • the present invention also includes all suitable isotopic variations of the compounds of the invention, or pharmaceutically acceptable salts thereof.
  • An isotopic variation of a compound of the invention or a pharmaceutically acceptable salt thereof is defined as one in which at least one atom is replaced by an atom having the same atomic number but an atomic mass different from the atomic mass usually found in nature.
  • isotopes that may be incorporated into the compounds of the invention and pharmaceutically acceptable salts thereof include but are not limited to isotopes of hydrogen, carbon, nitrogen and oxygen such as 2 H, 3 H, 11 C, 13 C, 14 C, 15 N, 17 0, 18 0, 35 S, 18 F, 36 Cl and 123 I.
  • Certain isotopic variations of the compounds of the invention and pharmaceutically acceptable salts thereof for example, those in which a radioactive isotope such as 3 H or 14 C is incorporated, are useful in drug and/or substrate tissue distribution studies.
  • 3 H and 14 C isotopes may be used for their ease of preparation and detectability.
  • substitution with isotopes such as H may afford certain therapeutic advantages resulting from greater metabolic stability, such as increased in vivo half- life or reduced dosage requirements.
  • Isotopic variations of the compounds of the invention or pharmaceutically acceptable salts thereof can generally be prepared by conventional procedures using appropriate isotopic variations of suitable reagents.
  • Isotopic variations of the compounds have the potential to change a compound' s metabolic fate and/or create small changes in physical properties such as hydrophobicity, and the like.
  • Isotopic variation have the potential to enhance efficacy and safety, enhance bioavailability and half-life, alter protein binding, change biodistribution, increase the proportion of active metabolites and/or decrease the formation of reactive or toxic metabolites.
  • each optionally substituted moiety may be substituted with Ci-6 alkyl, C 2 -6 alkenyl or C3-6 alkynyl, each of which may be optionally halogenated or optionally having a carbon that may be replaced or substituted with N, S, O, or a combination thereof (for example, hydroxylCi-Csalkyl, Ci-CsalkoxyCi-Csalkyl); halo, amino, amidino, Ci_ 6 alkoxy; hydroxyl, methylenedioxy, carboxy; C 1-8 alkylcarbonyl, C 1-8 alkoxycarbonyl, carbamoyl, Ci_ 8 alkylcarbamoyl, sulfamoyl, cyano, oxo, nitro, or an optionally substituted carbocyclic ring, heterocyclic ring, aryl or heteroaryl as previously described.
  • the compounds of the invention may be selectively toxic or more toxic to rapidly proliferating cells than to normal cells, and may be used to treat hyper-proliferative disorders.
  • the compounds of the invention may have significant antiproliferative effects in human cancer cells, e.g., cancerous tumors, and promote differentiation, e.g., cell cycle arrest and apoptosis.
  • the compounds of the invention may be utilized to inhibit, block, reduce or decrease cell proliferation and/or cell division, and/or produce apoptosis.
  • the method comprises administering to a mammal in need thereof, including a human, an amount of a compound of this invention, or a pharmaceutically acceptable salt, isomer, polymorph, metabolite, hydrate, solvate or ester thereof; etc. which is effective to treat the disorder.
  • Hyper-proliferative disorders include but are not limited, e.g., psoriasis, keloids, and other hyperplasias affecting the skin, benign prostate hyperplasia (BPH), solid tumors, such as cancers of the breast, respiratory tract, brain, reproductive organs, digestive tract, urinary tract, eye, liver, skin, head and neck, thyroid, parathyroid and their distant metastases.
  • BPH benign prostate hyperplasia
  • solid tumors such as cancers of the breast, respiratory tract, brain, reproductive organs, digestive tract, urinary tract, eye, liver, skin, head and neck, thyroid, parathyroid and their distant metastases.
  • Those disorders also include lymphomas, sarcomas, and leukemias. These disorders have been well characterized in humans, but also exist with a similar etiology in other mammals, and can be treated by administering pharmaceutical compositions of the present invention.
  • breast cancer examples include, but are not limited to invasive ductal carcinoma, invasive lobular carcinoma, ductal carcinoma in situ, and lobular carcinoma in situ.
  • cancers of the respiratory tract include, but are not limited to small-cell and non-small-cell lung carcinoma, as well as bronchial adenoma and pleuropulmonary blastoma.
  • brain cancers include, but are not limited to brain stem and hypophtalmic glioma, cerebellar and cerebral astrocytoma, medulloblastoma, ependymoma, as well as neuroectodermal and pineal tumor.
  • Tumors of the male reproductive organs include, but are not limited to prostate and testicular cancer.
  • Tumors of the female reproductive organs include, but are not limited to endometrial, cervical, ovarian, vaginal, and vulvar cancer, as well as sarcoma of the uterus.
  • Tumors of the digestive tract include, but are not limited to anal, colon, colorectal, esophageal, gallbladder, gastric, pancreatic, rectal, small-intestine, and salivary gland cancers.
  • Tumors of the urinary tract include, but are not limited to bladder, penile, kidney, renal pelvis, ureter, urethral and human papillary renal cancers.
  • Eye cancers include, but are not limited to intraocular melanoma and retinoblastoma.
  • liver cancers include, but are not limited to hepatocellular carcinoma (liver cell carcinomas with or without fibrolamellar variant), cholangiocarcinoma (intrahepatic bile duct carcinoma), and mixed hepatocellular cholangiocarcinoma.
  • Skin cancers include, but are not limited to squamous cell carcinoma, Kaposi's sarcoma, malignant melanoma, Merkel cell skin cancer, and non-melanoma skin cancer.
  • Head-and-neck cancers include, but are not limited to laryngeal, hypopharyngeal, nasopharyngeal, oropharyngeal cancer, lip and oral cavity cancer and squamous cell.
  • Lymphomas include, but are not limited to AIDS-related lymphoma, non-Hodgkin's lymphoma, cutaneous T-cell lymphoma, Burkitt lymphoma, Hodgkin' s disease, and lymphoma of the central nervous system.
  • Sarcomas include, but are not limited to sarcoma of the soft tissue, osteosarcoma, malignant fibrous histiocytoma, lymphosarcoma, and rhabdomyosarcoma.
  • Leukemias include, but are not limited to acute myeloid leukemia, acute lymphoblastic leukemia, chronic lymphocytic leukemia, chronic myelogenous leukemia, and hairy cell leukemia.
  • the present invention also provides methods for the treatment of disorders associated with aberrant expression of Ros, IGFlR, InsR, ALK, Hepatocyte growth factor receptor tyrosine kinase (c-Met), and/or Nucleophosmin-ALK.
  • the compounds described in this application are ATP-competitive kinase inhibitors. As such they competitively block ATP from binding to the kinase active site and thereby prevent phosphorylation of downstream substrates. This effectively blocks signal transduction from the targeted kinases.
  • Compounds in this application have the potential to interact with any kinase in the human kinome and have been tested for their ability to bind to a panel of 320 distinct protein kinases.
  • This panel includes the following kinases: AAKl, ABLl, ABL1(E255K), ABL1(H396P), ABL1(M351T), ABL1(Q252H), ABL1(T315I), ABL1(Y253F), AB L2, ACVRl, ACVRlB, ACVR2A, ACVR2B, ACVRLl, ADCK3,ADCK4,AKT1,AKT2, AKT3,ALK,AMPK-alphal,AMPK-alpha2, ANKKl, ARK5, AURKA, AURKB, AURKC, AXL, BIKE, BLK, BMPRlA, BMPR2, BMX, BRAF, BRAF(V600E), BRSKl, BRS K2, BTK, CAMKl, CAMKlD, CAMKlG, CAMK2A, CAMK2B, CAMK2D, CAMK2G, CAMK4, CAMKKl, CAMKK2, CDC2L
  • the compounds of the invention demonstrate inhibitory activity against: ADCK3, ADCK4, ALK, CLKl, CLK4, EGFR, EGFR(E746-A750del), EGFR(L747-E749del, A750P), EGFR(L747-S752del, P753S), EGFR(L747-T751del,Sins), EGFR(L858R), EGFR(L861Q), EGFR(S752-I759del), ERBB4, FER, FES, GAK, IGFlR, INSR, INSRR, LTK, PTK2, PTK2B, ROSl, RPS6KAl(Kin.Dom.2), TNKl, TNK2, TTK.
  • the ALK inhibitory activity and inhibitory activity against ALK-containing gene fusions of the compounds described herein make them useful pharmaceutical agents for the treatment of proliferative diseases.
  • the inventive compounds are particularly useful for treating a tumor which is a breast cancer, genitourinary cancer, lung cancer, gastrointestinal cancer, epidermoid cancer, melanoma, ovarian cancer, pancreas cancer, neuroblastoma, head and/or neck cancer or bladder cancer, or in a broader sense renal, brain or gastric cancer; in particular (i) a breast tumor; an epidermoid tumor, such as an epidermoid head and/or neck tumor or a mouth tumor; a lung tumor, for example a small cell or non-small cell lung tumor; a gastrointestinal tumor, for example, a colorectal tumor; or a genitourinary tumor, for example, a prostate tumor (especially a hormone-refractory prostate tumor); or (ii) a proliferative disease that is refractory to the treatment with
  • a proliferative disease may furthermore be a hyperproliferative condition such as leukemias, hyperplasias, fibrosis (especially pulmonary, but also other types of fibrosis, such as renal fibrosis), angiogenesis, psoriasis, atherosclerosis and smooth muscle proliferation in the blood vessels, such as stenosis or restenosis following angioplasty.
  • a hyperproliferative condition such as leukemias, hyperplasias, fibrosis (especially pulmonary, but also other types of fibrosis, such as renal fibrosis), angiogenesis, psoriasis, atherosclerosis and smooth muscle proliferation in the blood vessels, such as stenosis or restenosis following angioplasty.
  • Proliferative diseases treated according to the present method include tumors of blood and lymphatic system (e.g.
  • lymphoid leukemia acute or chronic myeloid leukemia, acute or chronic lymphocytic leukemia, monocytic leukemia, other leukemias of specified cell type, leukemia of unspecified cell type, other and unspecified malignant neoplasms of lymphoid, haematopoietic and related tissues, for example diffuse large cell lymphoma, T-cell lymphoma or cutaneous T-cell lymphoma).
  • Myeloid cancer includes e.g. acute or chronic myeloid leukaemia.
  • the present invention also provides methods of treating disorders and diseases associated with aberrant expression of the neurotrophic growth factor receptor tyrosine kinase A, B and C (Trk A, B, C also known as NTRKl, 2, and 3).
  • the compounds in the present invention exhibit inhibition of the neurotrophic growth factor receptor tyrosine kinase A, B and C (Trk A, B, C also known as NTRKl, 2, and 3).
  • Nrk A, B, C also known as NTRKl, 2, and 3
  • Several lines of evidence have implicated NTRKs in the development and progression of cancer through deregulation of tyrosine kinase activity by mutations, chromosomal rearrangements, upregulation of either the receptor, their ligand (Nerve Growth Factor, Brain Derived Neurotropic Factor, Neurotrophins) or both. Chromosomal translocations involving both NTRKl & 3 have been found in several different types of tumors.
  • TrkB is of central importance in preventing anoikis (detachment-induced apoptosis) which is believed to an important requirement in the metastatic process.
  • the present invention also provides methods of treating disorders and diseases associated with excessive and/or abnormal angiogenesis.
  • Inappropriate and ectopic expression of angiogenesis can be deleterious to an organism.
  • a number of pathological conditions are associated with the growth of extraneous blood vessels. These include, e.g., diabetic retinopathy, ischemic retinal-vein occlusion, and retinopathy of prematurity (Aiello et al. New Engl. J. Med. 1994, 331, 1480; Peer et al. Lab. Invest. 1995, 72, 638), age-related macular degeneration (AMD; see, Lopez et al. Invest. Opththalmol. Vis. Sci.
  • neovascular glaucoma neovascular glaucoma, psoriasis, retrolental fibroplasias, angiofibroma, inflammation, rheumatoid arthritis (RA), restenosis, in-stent restenosis, vascular graft restenosis, etc.
  • RA rheumatoid arthritis
  • restenosis in-stent restenosis
  • vascular graft restenosis etc.
  • the increased blood supply associated with cancerous and neoplastic tissue encourages growth, leading to rapid tumor enlargement and metastasis.
  • the growth of new blood and lymph vessels in a tumor provides an escape route for renegade cells, encouraging metastasis and the consequence spread of the cancer.
  • compounds of the present invention can be utilized to treat and/or prevent any of the aforementioned angiogenesis disorders, e.g., by inhibiting and/or reducing blood vessel formation; by inhibiting, blocking, reducing, decreasing, etc. endothelial cell proliferation or other types involved in angiogenesis, as well as causing cell death or apoptosis of such cell types.
  • the effective dosage of the compounds of this invention can readily be determined for treatment of each desired indication.
  • the amount of the active ingredient to be administered in the treatment of one of these conditions can vary widely according to such considerations as the particular compound and dosage unit employed, the mode of administration, the period of treatment, the age and sex of the patient treated, and the nature and extent of the condition treated.
  • the total amount of the active ingredient to be administered will generally range from about 0.001 mg/kg to about 200 mg/kg body weight per day, and preferably from about 0.01 mg/kg to about 20 mg/kg body weight per day.
  • the average daily oral dosing will be from about 200 mg/day to about 600 mg/day which corresponds to 2.8-8.6 mg/kg for a person with an average weight of 70 kgs.
  • Clinically useful dosing schedules will range from one to three times a day dosing to once every four weeks dosing.
  • "drug holidays" in which a patient is not dosed with a drug for a certain period of time may be beneficial to the overall balance between pharmacological effect and tolerability.
  • a unit dosage may contain from about 0.5 mg to about 1500 mg of active ingredient, and can be administered one or more times per day or less than once a day.
  • the average daily dosage for administration by injection including intravenous, intramuscular, subcutaneous and parenteral injections, and use of infusion techniques will preferably be from 0.01 to 200 mg/kg of total body weight.
  • the average daily rectal dosage regimen will preferably be from 0.01 to 200 mg/kg of total body weight.
  • the average daily vaginal dosage regimen will preferably be from 0.01 to 200 mg/kg of total body weight.
  • the average daily topical dosage regimen will preferably be from 0.1 to 200 mg administered between one to four times daily.
  • the transdermal concentration will preferably be that required to maintain a daily dose of from 0.01 to 200 mg/kg.
  • the average daily inhalation dosage regimen will preferably be from 0.01 to 100 mg/kg of total body weight.
  • the specific initial and continuing dosage regimen for each patient will vary according to the nature and severity of the condition as determined by the attending diagnostician, the activity of the specific compound employed, the age and general condition of the patient, time of administration, route of administration, rate of excretion of the drug, drug combinations, and the like.
  • the desired mode of treatment and number of doses of a compound of the present invention or a pharmaceutically acceptable salt or ester or composition thereof can be ascertained by those skilled in the art using conventional treatment tests.
  • the compounds of this invention can be administered as the sole pharmaceutical agent or in combination with one or more other pharmaceutical agents where the combination causes no unacceptable adverse effects.
  • the compounds of this invention can be combined with known anti-hyper-proliferative or other indication agents, and the like, as well as with admixtures and combinations thereof.
  • the additional pharmaceutical agent can be aldesleukin, alendronic acid, alfaferone, alitretinoin, allopurinol, aloprim, aloxi, altretamine, aminoglutethimide, amifostine, amrubicin, amsacrine, anastrozole, anzmet, aranesp, arglabin, arsenic trioxide, aromasin, 5-azacytidine, azathioprine, BCG or tice BCG, bestatin, betamethasone acetate, betamethasone sodium phosphate, bexarotene, bleomycin sulfate, broxuridine , bortezomib, busulfan, calcitonin, campath, capecitabine, carboplatin, casodex, cefesone, celmoleukin, cerubidine, chlorambucil, cisplatin, cladribine, cladribine, clo
  • Optional anti-hyper-proliferative agents which can be added to the composition include but are not limited to compounds listed on the cancer chemotherapy drug regimens in the 11 th Edition of the Merck Index, (1996), which is hereby incorporated by reference, such as asparaginase, bleomycin, carboplatin, carmustine, chlorambucil, cisplatin, colaspase, cyclophosphamide, cytarabine, dacarbazine, dactinomycin, daunorubicin, doxorubicin (adriamycine), epirubicin, etoposide, 5-fluorouracil, hexamethylmelamine, hydroxyurea, ifosfamide, irinotecan, leucovorin, lomustine, mechlorethamine, 6-mercaptopurine, mesna, methotrexate, mitomycin C, mitoxantrone, prednisolone, pred
  • composition of the invention include but are not limited to those compounds acknowledged to be used in the treatment of neoplastic diseases in Goodman and Gilman's The Pharmacological Basis of Therapeutics (Ninth Edition), editor Molinoff et al., publ.
  • anti-hyper-proliferative agents suitable for use with the composition of the invention include but are not limited to other anti-cancer agents such as epothilone and its derivatives, irinotecan, raloxifen and topotecan. Dosage Forms and Modes of Administration
  • compounds of the invention will be administered in therapeutically effective amounts via any of the usual and acceptable modes known in the art, either singly or in combination with one or more therapeutic agents.
  • a therapeutically effective amount may vary widely depending on the severity of the disease, the age and relative health of the subject, the potency of the compound used and other factors known to those of ordinary skill in the art.
  • the required dosage will also vary depending on the mode of administration, the particular condition to be treated and the effect desired.
  • compositions comprising them that are suitable for oral administration can be presented as discrete dosage forms, such as, but are not limited to, tablets (e.g., chewable tablets), caplets, capsules, and liquids (e.g., flavored syrups).
  • dosage forms contain predetermined amounts of active ingredients, and may be prepared by methods of pharmacy well known to those skilled in the art. See generally, Remington's Pharmaceutical Sciences, 18th ed., Mack Publishing, Easton Pa. (1990).
  • Typical oral dosage forms of the invention are prepared by combining the active ingredient(s) in an intimate admixture with at least one excipient according to conventional pharmaceutical compounding techniques.
  • Excipients can take a wide variety of forms depending on the form of preparation desired for administration.
  • excipients suitable for use in oral liquid or aerosol dosage forms include, but are not limited to, water, glycols, oils, alcohols, flavoring agents, preservatives, and coloring agents.
  • excipients suitable for use in solid oral dosage forms include, but are not limited to, starches, sugars, micro-crystalline cellulose, diluents, granulating agents, lubricants, binders, and disintegrating agents.
  • tablets and capsules represent very advantageous oral dosage unit forms, in which case solid excipients are employed. If desired, tablets can be coated by standard aqueous or nonaqueous techniques. Such dosage forms can be prepared by any of the methods of pharmacy. In general, pharmaceutical compositions and dosage forms are prepared by uniformly and intimately admixing the active ingredients with liquid carriers, finely divided solid carriers, or both, and then shaping the product into the desired presentation if necessary.
  • a tablet can be prepared by compression or molding. Compressed tablets can be prepared by compressing in a suitable machine the active ingredients in a free- flowing form such as powder or granules, optionally mixed with an excipient. Molded tablets can be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
  • excipients that can be used in oral dosage forms of the invention include, but are not limited to, binders, fillers, disintegrants, and lubricants.
  • Binders suitable for use in pharmaceutical compositions and dosage forms include, but are not limited to, corn starch, potato starch, or other starches, gelatin, natural and synthetic gums such as acacia, sodium alginate, alginic acid, other alginates, powdered tragacanth, guar gum, cellulose and its derivatives (e.g., ethyl cellulose, cellulose acetate, carboxymethyl cellulose calcium, sodium carboxymethyl cellulose), polyvinyl pyrrolidone, methyl cellulose, pre-gelatinized starch, hydroxypropyl methyl cellulose, (e.g., nos. 2208, 2906, 2910), microcrystalline cellulose, and mixtures thereof.
  • fillers suitable for use in the pharmaceutical compositions and dosage forms disclosed herein include, but are not limited to, talc, calcium carbonate (e.g., granules or powder), microcrystalline cellulose, powdered cellulose, dextrates, kaolin, mannitol, silicic acid, sorbitol, starch, pre-gelatinized starch, and mixtures thereof.
  • the binder or filler in pharmaceutical compositions of the invention is typically present in from about 50 to about 99 weight percent of the pharmaceutical composition or dosage form.
  • Suitable forms of microcrystalline cellulose include, but are not limited to, the materials sold as AVICEL-PH- 101, AVICEL-PH- 103 AVICEL RC-581, AVICEL-PH- 105 (available from FMC Corporation, American Viscose Division, Avicel Sales, Marcus Hook, Pa.), and mixtures thereof.
  • An specific binder is a mixture of microcrystalline cellulose and sodium carboxymethyl cellulose sold as AVICEL RC-581.
  • Suitable anhydrous or low moisture excipients or additives include AVICEL-PH- 103.TM and Starch 1500 LM.
  • Disintegrants are used in the compositions of the invention to provide tablets that disintegrate when exposed to an aqueous environment. Tablets that contain too much disintegrant may disintegrate in storage, while those that contain too little may not disintegrate at a desired rate or under the desired conditions. Thus, a sufficient amount of disintegrant that is neither too much nor too little to detrimentally alter the release of the active ingredients should be used to form solid oral dosage forms of the invention.
  • the amount of disintegrant used varies based upon the type of formulation, and is readily discernible to those of ordinary skill in the art. Typical pharmaceutical compositions comprise from about 0.5 to about 15 weight percent of disintegrant, specifically from about 1 to about 5 weight percent of disintegrant.
  • Disintegrants that can be used in pharmaceutical compositions and dosage forms of the invention include, but are not limited to, agar-agar, alginic acid, calcium carbonate, microcrystalline cellulose, croscarmellose sodium, crospovidone, polacrilin potassium, sodium starch glycolate, potato or tapioca starch, pre-gelatinized starch, other starches, clays, other algins, other celluloses, gums, and mixtures thereof.
  • Lubricants that can be used in pharmaceutical compositions and dosage forms of the invention include, but are not limited to, calcium stearate, magnesium stearate, mineral oil, light mineral oil, glycerin, sorbitol, mannitol, polyethylene glycol, other glycols, stearic acid, sodium lauryl sulfate, talc, hydrogenated vegetable oil (e.g., peanut oil, cottonseed oil, sunflower oil, sesame oil, olive oil, corn oil, and soybean oil), zinc stearate, ethyl oleate, ethyl laureate, agar, and mixtures thereof.
  • calcium stearate e.g., magnesium stearate, mineral oil, light mineral oil, glycerin, sorbitol, mannitol, polyethylene glycol, other glycols, stearic acid, sodium lauryl sulfate, talc
  • hydrogenated vegetable oil e.g., peanut oil, cottonseed oil
  • Additional lubricants include, for example, a syloid silica gel (AEROSIL 200, manufactured by W. R. Grace Co. of Baltimore, Md.), a coagulated aerosol of synthetic silica (marketed by Degussa Co. of Piano, Tex.), CAB-O-SIL (a pyrogenic silicon dioxide product sold by Cabot Co. of Boston, Mass.), and mixtures thereof. If used at all, lubricants are typically used in an amount of less than about 1 weight percent of the pharmaceutical compositions or dosage forms into which they are incorporated.
  • AEROSIL 200 a syloid silica gel
  • a coagulated aerosol of synthetic silica marketed by Degussa Co. of Piano, Tex.
  • CAB-O-SIL a pyrogenic silicon dioxide product sold by Cabot Co. of Boston, Mass.
  • Parenteral dosage forms can be administered to patients by various routes including, but not limited to, subcutaneous, intravenous (including bolus injection and constant infusion), intramuscular, and intraarterial. Because their administration typically bypasses patients' natural defenses against contaminants, parenteral dosage forms are preferably sterile or capable of being sterilized prior to administration to a patient. Examples of parenteral dosage forms include, but are not limited to, solutions ready for injection, dry products (including, but not limited to lyophilized powders, pellets, and tablets) ready to be dissolved or suspended in a pharmaceutically acceptable vehicle for injection, suspensions ready for injection, and emulsions.
  • Suitable vehicles that can be used to provide parenteral dosage forms of the invention are well known to those skilled in the art. Examples include, but are not limited to: Water for Injection USP; aqueous vehicles such as, but not limited to, Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, and Lactated Ringer's Injection; water-miscible vehicles such as, but not limited to, ethyl alcohol, polyethylene glycol, and polypropylene glycol; and non-aqueous vehicles such as, but not limited to, corn oil, cottonseed oil, peanut oil, sesame oil, ethyl oleate, isopropyl myristate, and benzyl benzoate. Compounds that increase the solubility of one or more of the active ingredients disclosed herein can also be incorporated into the parenteral dosage forms of the invention.
  • Transdermal, Topical, And Mucosal Dosage Forms include, but are not limited to, ophthalmic solutions, sprays, aerosols, creams, lotions, ointments, gels, solutions, emulsions, suspensions, or other forms known to one of skill in the art. See, e.g., Remington's Pharmaceutical Sciences, 16th and 18th eds., Mack Publishing, Easton Pa. (1980 & 1990); and Introduction to Pharmaceutical Dosage Forms, 4th ed., Lea & Febiger, Philadelphia (1985).
  • transdermal dosage forms suitable for treating mucosal tissues within the oral cavity can be formulated as mouthwashes or as oral gels.
  • transdermal dosage forms include "reservoir type” or “matrix type” patches, which can be applied to the skin and worn for a specific period of time to permit the penetration of a desired amount of active ingredients.
  • Suitable excipients e.g., carriers and diluents
  • other materials that can be used to provide transdermal, topical, and mucosal dosage forms encompassed by this invention are well known to those skilled in the pharmaceutical arts, and depend on the particular tissue to which a given pharmaceutical composition or dosage form will be applied.
  • excipients include, but are not limited to, water, acetone, ethanol, ethylene glycol, propylene glycol, butane-l,3-diol, isopropyl myristate, isopropyl palmitate, mineral oil, and mixtures thereof to form lotions, tinctures, creams, emulsions, gels or ointments, which are nontoxic and pharmaceutically acceptable.
  • Moisturizers or humectants can also be added to pharmaceutical compositions and dosage forms if desired. Examples of such additional ingredients are well known in the art. See, e.g., Remington's Pharmaceutical Sciences, 16th and 18th eds., Mack Publishing, Easton Pa. (1980 & 1990).
  • penetration enhancers can be used to assist in delivering the active ingredients to the tissue.
  • Suitable penetration enhancers include, but are not limited to: acetone; various alcohols such as ethanol, oleyl, and tetrahydrofuryl; alkyl sulfoxides such as dimethyl sulfoxide; dimethyl acetamide; dimethyl formamide; polyethylene glycol; pyrrolidones such as polyvinylpyrrolidone; Kollidon grades (Povidone, Polyvidone); urea; and various water-soluble or insoluble sugar esters such as Tween 80 (polysorbate 80) and Span 60 (sorbitan monostearate).
  • the pH of a pharmaceutical composition or dosage form, or of the tissue to which the pharmaceutical composition or dosage form is applied may also be adjusted to improve delivery of one or more active ingredients.
  • the polarity of a solvent carrier, its ionic strength, or tonicity can be adjusted to improve delivery.
  • Compounds such as stearates can also be added to pharmaceutical compositions or dosage forms to advantageously alter the hydrophilicity or lipophilicity of one or more active ingredients so as to improve delivery.
  • stearates can serve as a lipid vehicle for the formulation, as an emulsifying agent or surfactant, and as a delivery-enhancing or penetration-enhancing agent.
  • Different salts, hydrates or solvates of the active ingredients can be used to further adjust the properties of the resulting composition.
  • kits which, when used by the medical practitioner, can simplify the administration of appropriate amounts of active ingredients to a patient.
  • a typical kit of the invention comprises one or more unit dosage forms of a compound of the invention or one or more compositions comprising a compound of the invention, or physiologically acceptable salts thereof, and instructions for use.
  • Kits of the invention can further comprise devices that are used to administer a compound of the invention to a patient.
  • devices include, but are not limited to, intravenous cannulation devices, syringes, drip bags, patches, topical gels, pumps, tubing, containers that provide protection from photodegredation, and inhalers.
  • Kits of the invention can further comprise pharmaceutically acceptable vehicles that can be used to administer one or more active ingredients.
  • the kit can comprise a sealed container of a suitable vehicle in which the active ingredient can be dissolved to form a particulate-free sterile solution that is suitable for parenteral administration.
  • Examples of pharmaceutically acceptable vehicles include, but are not limited to: Water for Injection USP; aqueous vehicles such as, but not limited to, Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, and Lactated Ringer's Injection; water-miscible vehicles such as, but not limited to, ethyl alcohol, polyethylene glycol, and polypropylene glycol; and non-aqueous vehicles such as, but not limited to, corn oil, cottonseed oil, peanut oil, sesame oil, ethyl oleate, isopropyl myristate, and benzyl benzoate.
  • aqueous vehicles such as, but not limited to, Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, and Lactated Ringer's Injection
  • water-miscible vehicles such as, but not limited to, ethyl alcohol
  • the compounds of Formula I, particularly Formula IA can be prepared through the preparation shown in the Reaction Scheme 2 below.
  • a sealed tube was charged with 2,3,5,6-tetrachloropyrimidine (2.18 g, 10.03 mmol), 2-(isopropylsulfonyl)benzenamine (2.0 mg, 10.03 mmol), 4N HC1 dioxane (2.5 mL) and dioxane (50.0 mL).
  • the tube was sealed and the reaction mixture was stirred at 120 0 C for 24 hours.
  • the reaction mixture was then partitioned between ethyl acetate and water. The organic layer was separated and the aqueous layer was neutralized with satd. ⁇ aHC ⁇ 3 solution and extracted with ethyl acetate.
  • reaction mixture was poured into ice water and the resulting white solid was filtered and dried, yielding (560 mg) mixture of two isomers 2,5- dichloro-N 4 -(2-(isopropylsulfonyl)phenyl)pyrimidine-4,6-diamine and 5,6-dichloro-N 4 -(2- (isopropylsulfonyl)phenyl)pyrimidine-2,4-diamine.
  • the compounds of Formula I, particularly Formula IA can be prepared through the preparation shown in the Reaction Scheme 3 below.
  • the compounds of Formula I may be prepared through the preparation of intermediates shown in the Reaction Scheme 4 below aq.)
  • This white solid was suspended in water (35 mL) and treated with sat. K 2 CO 3 slowly until a substantial amount of white solid precipitated. This slurry mixture was slowly stirred overnight and then cooled down to 0 0 C. The resulting white solid was collected, washed with water and then dried to give 5.2 g (58% yield) of the title product.
  • the reaction mixture was degassed using Argon for 10 min and then Pd 2 (dba) 2 (196 mg, 0.214 mmol) and 2- dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (153 mg, 0.32 mmol) were added.
  • the reaction flask was placed into the preheated oil-bath at 100 0 C.
  • the reaction mixture was further stirred at 100 0 C for a period of 4 hours after which, it was filtered and partitioned between ethyl acetate and water. The organic layer was separated and the aqueous layer was extracted with ethyl acetate.
  • the reaction mixture was degassed using Argon for 10 min after which Pd 2 (dba) 2 (26 mg, 0.03 mmol) and 2-dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (22 mg, 0.05 mmol) were added.
  • the reaction flask was put into the preheated oil-bath at 100 0 C.
  • the reaction mixture was further stirred at 100 0 C for a period of 6 hours after which, it was filtered and partitioned between ethyl acetate and water. The organic layer was separated and the aqueous layer was extracted with ethyl acetate.
  • the reaction mixture was stirred for 6h after which, it was neutralized with IN HCl solution and partitioned between ethyl acetate and water. The organic layer was separated and the aqueous layer was extracted with ethyl acetate. The combined organic extracts were washed with brine, dried over MgSO 4 , filtered and concentrated. The crude product was used without further purification.
  • a microwave tube was charged with 6-chloro-N-(2-(isopropylsulfonyl)pheny I)-I- (tetrahydro-2H-pyran-2-yl)-lH-pyrazolo[3,4-d]pyrimidin-4-amine (40 mg, 0.0917 mmol), 2- isopropoxy-5-methyl-4-(l-methylpiperidin-4-yl)benzenamine (72.3 mg, 0.275 mmol) 4N HCl in Dioxane (0.069 mL, 0.276 mmol) and ethylene glycol (1.5 mL). The tube was sealed and the reaction mixture was stirred at 180 0 C for 30 min in a microwave reactor.
  • the reaction mixture was purified by reverse-phase preparative HPLC.
  • the crude TFA salt of product was neutralized with a satd. aqueous solution of NaHC ⁇ 3 and extracted with ethyl acetate. The organic layer was dried over sodium sulfate, filtered and concentrated. The residue was purified by flash chromatography using 30:1:0.3 methylene chloride-methanol-triethylamine as solvent to afford the title compound (20.0 mg, 0.037 mmol).
  • Murine pro-B cell line Ba/F3 the human t(2,5)-positive Karpas-299 and TEL-ALK transformed Ba/F3 are maintained in RPMI medium 1640 supplemented with 10% FBS (Sigma-Aldrich, St. Louis, MO). Ba/F3 cells are grown in the presence of 10% of WEHI media. Cell lines expressing luciferase alone or in combination with TEL-kinase fusion constructs are generated by retroviral transduction of cells with pMSCV-IRES puro/Luc vector.
  • Luciferase-expressing Ba/F3 cells, Karpas-299, Tel_ALK transformed Ba/F3 stably expressing NPM-ALK and TEL-ALK are plated in 384-well plates (5,000 cells per well) and incubated with serial dilutions of ALK inhibitors or DMSO for 48 hours. Luciferase expression is used as a measure of cell proliferation/ survival and was evaluated with the Bright-Glo Luciferase Assay System (Promega, Madison, WI). Fifty percent inhibition values (IC 50 ) are generated by using XLFit software.
  • mice with established lymphomas are administered vehicle solution or test compound (typically 10 mg/kg) for 3 days.
  • test compound typically 10 mg/kg
  • ALK tyrosine kinase activity may also be measured using known methods, for example using the recombinant kinase domain of the ALK in analogy to the VEGF-R kinase assay described in J. Wood et al. Cancer Res. 60, 2178-2189 (2000).
  • the antiproliferative action of the compounds of the invention can also be determined in the human KARPAS-299 lymphoma cell line (described in WG Dirks et al. Int. J. Cancer 100, 49-56 (2002) using the same methodology described above).
  • Compounds 1-3, 1-13, 1-14, 2-1, 2-4, 2-7, 2-10, 2-14, 2-15, 3-14, 5-1, 5-7, 5-8, 5-9, 5-10, 5-11, 5-14, 6-1, 6-2, 6-5, 6-6, 6-9, 6-16, 6-18 are representative examples of inhibitors of NPM-ALK dependent cellular proliferation EC50's of 1 uM or less.
  • a cell-based assay consists of using a ETV6-NTRK3 transformed Ba/F3 cell line. This cell line may be used to discover compounds that are differentially cytotoxic as compared to parental Ba/F3 cells grown in the presence of IL-3. Compounds that are selectively cytotoxic to Ba/F3 ETV6-NTRK3 are confirmed using a biochemical NTRK3 kinase assay. Cellular inhibition of NTRK3 is confirmed using phosphospecific antibodies.
  • WO 2005016894 - Garcia-echeverria, Carlos; Kanazawa, Takanori; Kawahara, Eiji; Masuya, Keiichi; Matsuura, Naoko; Miyake, Takahiro; Ohmori, Osamu; Umemura, Ichiro; Steensma, Ruo; Chopiuk, Greg; Jiang, Jiqing; Wan, Yongqin; Ding, Qiang; Zhang, Qiong; Gray, Nathanael Schiander; Karanewsky, Donald. Preparation of 2,4-pyrimidinediamines useful in the treatment of neoplastic diseases, inflammatory and immune system disorders. PCT Int. Appl. (2005), 285 pp.

Abstract

This invention relates to novel amino substituted pyrimidine compounds of formula (I), pharmaceutical compositions containing such compounds and the use of those compounds or compositions for treating hyper-proliferative and/or angiogenesis disorders, as a sole agent or in combination with other active ingredients.

Description

2- (HET) ARYLAMINO-6-AMINOPYRIDINE DERIVATIVES AND FUSED FORMS THEREOF AS ANAPLASTIC LYMPHOMA KINASE INHIBITORS
Cross-Reference to Related Applications
[0001] This application claims the benefit of U.S. provisional application serial number 60/966,449, filed August 28, 2007, which is hereby incorporated by reference in its entirety.
Background of the Invention
[0002] Cancer is a disease resulting from an abnormal growth of tissue. Certain cancers have the potential to invade into local tissues and also metastasize to distant organs. This disease can develop in a wide variety of different organs, tissues and cell types. Therefore, the term "cancer" refers to a collection of over a thousand different diseases.
[0003] Anaplastic lymphoma kinase (ALK), a member of the insulin receptor superfamily of receptor tyrosine kinases, has been implicated in oncogenesis in hematopoietic and non- hematopoietic tumors. The aberrant expression of full-length ALK receptor proteins has been reported in neuroblastomas and glioblastomas; and ALK fusion proteins have occurred in anaplastic large cell lymphoma. The study of ALK fusion proteins has also raised the possibility of new therapeutic treatments for patients with ALK-positive malignancies. (Pulford et al., Cell. MoI. Life Sci. 61:2939-2953 (2004)).
[0004] Insulin- like growth factor (IGF-I) signaling is highly implicated in cancer, with the IGF-I receptor (IGF-IR) as the predominating factor. IGR-IR is important for tumor transformation and survival of malignant cells, but is only partially involved in normal cell growth. Targeting of IGF-IR has been suggested to be a promising option for cancer therapy. (Larsson et al., Br. J. Cancer 92:2097-2101 (2005)).
[0005] The c-ros oncogene 1 (ROSl, also known as ROS), a member of the tyrosine kinase insulin receptor gene family, is highly expressed in a variety of tumor cell lines.
[0006] The enzymatic activity of mitotic assembly checkpoint kinase MPS 1 (also known as TTK) is required for cells to undergo a mitotic checkpoint arrest in response to agents that disrupt mitosis such as compounds that interfere with tubulin polymerization/depolymerization. Normal cells have multiple redundant mechanisms for arresting in mitosis whereas cancer cells have a heightened dependence on TTK activity. [0007] Despite advancements in the art, there remains a need for cancer treatments and anticancer compounds.
Summary of the Invention
[0008] The invention relates to novel pyrimidine and pyridine derivatives and pharmaceutical compositions thereof, and their use as pharmaceuticals.
[0009] In one aspect, the invention provides compounds having the Formula (I): pa
HN-R
«\* R-X-(B)NΛZΛN®Y-B'
R3 H H ^
or a physiologically acceptable salt, solvate, hydrate or stereoisomer thereof, wherein:
A and B are each independently aryl or heteroaryl, each of which may be optionally substituted with one or more R4 groups;
Y is -SO2-, -SO2NH-, -NH-SO2-, -NH-C(O)-, -C(O)-NH-, -O-, or -NR2-;
X is NH, O or S;
Z is N or CH;
Rl is H, C1-C6 alkyl, halo-(Cl-C6 alkyl), C1-C6 cycloalkyl, halo-(Cl-C6 cycloalkyl), heterocyclyl, heterocyclylCl-όalkyl, aryl, arylCl-όalkyl, heteroaryl or heteroarylCl-όalkyl;
R2 is H, C1-C6 alkyl, halo-(Cl-C6 alkyl), C1-C6 cycloalkyl, halo-(Cl-C6 cycloalkyl), heterocyclyl, heterocyclylCl-όalkyl, aryl, arylCl-όalkyl, heteroaryl or heteroarylCl-όalkyl; R3
Figure imgf000003_0001
Figure imgf000004_0001
Ra is hydrogen and Rb is halogen, or Ra and Rb, taken together with the atoms to which they are bound form a pyrazolo or a pyrrolo ring fused to the pyrimidine ring, said pyrazolo or pyrrolo ring optionally bearing one or two R4 groups;
Each occurrence of R4 is independently halogen, C1-C6 alkyl, halo-(Cl-C6 alkyl), Cl- C6 cycloalkyl, halo-(Cl-C6 cycloalkyl), heterocyclyl, heterocyclylCl-όalkyl, aryl, arylCl- όalkyl, heteroaryl or heteroarylCl-όalkyl, Cl-6alkoxy, Cl-6alkylthio, hydroxyl, nitro, azido, cyano, acyloxy, carboxy, ester, carbamoyl, carboxamide, ureido, amidino, guanidine, sulfonyl, sulphonylamino, aminosulphonyl;
I— N NH Each occurrence of R5 is independently is C1-C6 alkoxy, ^ — / , hydroxy, \— N N-(C1-C6 alkyl)
Figure imgf000004_0002
, dialkylamino, or -N-R7;
\— N \ I— N N-R7 Each occurrence of R6 is independently is hydroxy, ^ — ' , Nv — ^ , or -
C0NH2; and
Each occurrence of R7 is independently is hydrogen, C1-C6 alkyl, C1-C6 alkoxy, hydroxyl, or C1-C6 hydroxyalkyl;
With the proviso that when B is heteroaryl, -X-R2 and -R3 may each independently represent hydrogen. [0010] In particular, the invention provides compounds having Formula I, further provided
<— N V- R6 \— N O \— N N-R' that i) if B is phenyl and Z is N, R is not
Figure imgf000005_0001
alternatively, R3 or R4 if present in B are not nitro, azido, ureido, guanidine or sulphonylamino; and
(ii) if B is phenyl, pyridyl or thiazolyl and Z is CH, R3 is
Figure imgf000005_0002
Figure imgf000005_0003
alternatively, R3 or R4 if present in B are independently halogen, Ci-C6 alkyl, halo-(d- Ce alkyl), C3-C7 cycloalkyl, halo-(C3-C7 cycloalkyl), heterocyclyl, heterocyclylCi_6alkyl, aryl, arylCi_6alkyl, heteroaryl or heteroarylCi_6alkyl.
[0011] In one embodiment, the invention provides compounds having Formula IA or III:
Figure imgf000005_0004
wherein R3 is -CO-R5, *
Figure imgf000006_0001
Figure imgf000006_0002
alternatively, R3 or R4 if present in B are independently halogen, Ci-C6 alkyl, halo-(d-
C6 alkyl), C3-C7 cycloalkyl, halo-(C3-C7 cycloalkyl), heterocyclyl, heterocyclylCi-όalkyl, aryl, arylCi_6alkyl, heteroaryl, heteroarylCi_6alkyl, Ci-6alkoxy, Ci_6alkylthio, hydroxyl, cyano, acyloxy, carboxy, ester, carbamoyl, carboxamide, amidino, sulfonyl, or aminosulphonyl; n is an integer from 0-3; m is an integer from 0-4; p is an integer from 0- 1 ; and
Y, X, R1, R2, R5, R7, Ra and Rb are as defined in Formula I. [0012] In yet another embodiment, the invention provides compounds having Formula III:
Figure imgf000006_0003
|— N Y- R6 I— N O I — ( I N-R' I— N N-R7 wherein R3 is
Figure imgf000006_0004
alternatively, R3 or R4 if present in B are independently halogen, Ci-C6 alkyl, 1IaIo-(C1- C6 alkyl), C3-C7 cycloalkyl, halo-(C3-C7 cycloalkyl), heterocyclyl, heterocyclylCi_6alkyl, aryl, arylCi_6alkyl, heteroaryl or heteroarylCi_6alkyl; n is an integer from 0-3; m is an integer from 0-4; p is an integer from 0-2; and
Y, X, R1, R2, R5, R7 are as defined in Formula I.
[0013] In some embodiments, the invention encompasses a compound of Formula I, IA, II or III, wherein X is O. In still other embodiments, the invention encompasses a compound of Formula I, IA, II or III, wherein R2 is methyl, ethyl, propyl, isopropyl, cyclopropyl or cyclobutyl. In yet other embodiments, the invention encompasses a compound of Formula I, IA, II or III, wherein R is methyl. In another embodiment , the invention encompasses a compound of Formula I, IA, II or III, wherein Y is SO2.
[0014] In still another embodiment, the invention encompasses a compound of Formula I, IA, II or III, wherein R1 is methyl, ethyl, propyl, isopropyl, cyclopropyl or cyclobutyl. In still yet other embodiments, the invention encompasses a compound of Formula I, IA, II or III, wherein R1 is isopropyl. In yet another embodiment, the invention encompasses a compound of Formula I, IA, II or III, wherein X is O, R2 is methyl, Y is SO2 and R1 is isopropyl.
[0015] In particular embodiments, the invention provides compounds having Formula I wherein Z is N, and said compound is selected from the group consisting of: methyl 3-(4-amino-5-chloro-6-(2-(methylsulfonyl)phenylamino)pyrimidin-2-ylamino)-4- methoxybenzoate ;
(3-(4-amino-5-chloro-6-(2-(methylsulfonyl)phenylamino)pyrimidin-2-ylamino)-4- methoxyphenyl)(piperazin-l-yl)methanone;
5-chloro-N2-(2-methoxy-5-(2-(pyrrolidin-l-yl)ethoxy)phenyl)-N4-(2- (methylsulfonyl)phenyl)pyrimidine-2,4,6-triamine; l,4'-bipiperidin-l'-yl(4-(4-amino-5-chloro-6-(2-(methylsulfonyl)phenylamino)pyrimidin- 2-ylamino)-3-isopropoxyphenyl)methanone;
(4-(4-amino-5-chloro-6-(2-(methylsulfonyl)phenylamino)pyrimidin-2-ylamino)-3- isopropoxyphenyl)(4-(4-methylpiperazin- 1 -yl)piperidin- 1 -yl)methanone; l,4'-bipiperidin-r-yl(4-(4-amino-5-chloro-6-(2-(methylsulfonyl)phenylamino)pyrimidin- 2-ylamino)-3-methoxyphenyl)methanone;
5-chloro-N4-(2-(isopropylsulfonyl)phenyl)-N2-(2-methoxy-5-(2-(pyrrolidin-l- yl)ethoxy)phenyl)pyrimidine-2,4,6-triamine; l,4'-bipiperidin-l'-yl(4-(4-amino-5-chloro-6-(2-(isopropylsulfonyl)phenylamino) pyrimidin-2-ylamino)-3-isopropoxyphenyl)methanone; 5-chloro-N2-(2-isopropoxy-5-methyl-4-(l-methylpiperidin-4-yl)phenyl)-N4-(2- (methylsulfonyl)phenyl)pyrimidine-2,4,6-triamine;
4'-(4-amino-5-chloro-6-(2-(methylsulfonyl)phenylamino)pyrimidin-2-ylamino)-5'- methoxy-n,2'-dimethylbiphenyl-4-carboxamide;
5-chloro-N2-(2-isopropoxy-5-methyl-4-(l-methylpiperidin-4-yl)phenyl)-N4-(2- (isopropylsulfonyl)phenyl)pyrimidine-2,4,6-triamine;
5-chloro-N4-(2-(isopropylsulfonyl)phenyl)-N2-(2-methoxy-4-(l-methylpiperidin-3- yl)phenyl)pyrimidine-2,4,6-triamine;
5-chloro-N4-(2-(isopropylsulfonyl)phenyl)-N2-(2-methoxy-4-(l-methylpiperidin-4- yloxy)phenyl)pyrimidine-2,4,6-triamine;
5-chloro-N2-(6-(2,6-dimethylmorpholino)pyridin-3-yl)-N4-(2- (isopropylsulfonyl)phenyl)pyrimidine-2,4,6-triamine; l,4'-bipiperidin-l'-yl(4-(4-amino-5-chloro-6-(2- (isopropylsulfonyl)phenylamino)pyrimidin-2-ylamino)-3-methoxyphenyl)methanone;
5-chloro-N2-(2-isopropoxy-5-methyl-4-(l-methylpiperidin-3-yl)phenyl)-N4-(2- (isopropylsulfonyl)phenyl)pyrimidine-2,4,6-triamine;
5-chloro-N2-(lH-indol-6-yl)-N4-(2-(methylsulfonyl)phenyl)pyrimidine-2,4,6-triamine;
5-chloro-N2-(lH-indazol-6-yl)-N4-(2-(methylsulfonyl)phenyl)pyrimidine-2,4,6- triamine;
4-(4-amino-5-chloro-6-(2-(isopropylsulfonyl)phenylamino)pyrimidin-2-ylamino)-3- methoxy-N-(4-(4-methylpiperazin-l-yl)phenyl)benzamide;
4-{4-Amino-5-chloro-6-[2-(propane-2-sulfonyl)-phenylamino]-pyrimidin-2-ylamino}-3- methoxy-N-(4-morpholin-4-yl-phenyl)-benzamide;
4-{4-Amino-5-chloro-6-[2-(propane-2-sulfonyl)-phenylamino]-pyrimidin-2-ylamino}-3- methoxy-N- [4-(4-methyl-piperazin- 1 -yl)-phenyl] -benzamide ;
(4- { 4- Amino-5-chloro-ό- [2-(propane-2-sulfonyl)-phenylamino]-pyrimidin-2-ylamino } - 3-methoxy-phenyl)-(4-pyrrolidin-l-yl-piperidin-l-yl)-methanone;
(4- { 4- Amino-5-chloro-ό- [2-(propane-2-sulfonyl)-phenylamino]-pyrimidin-2-ylamino } - 3 -methoxy-phenyl)- [4-(4-methyl-piperazin- 1 -yl)-piperidin- 1 -yl] -methanone ;
3-[4-(4-{4-Amino-5-chloro-6-[2-(propane-2-sulfonyl)-phenylamino]-pyrimidin-2- ylamino } -3-methoxy-benzoyl)-piperazin- 1 -yl]-propionitrile; (4-{4-Amino-5-chloro-6-[2-(propane-2-sulfonyl)-phenylamino]-pyrimidin-2-ylamino}- 3-methoxy-phenyl)-(2-methyl-morpholin-4-yl)-methanone;
(4- { 4-Amino-5-chloro-6- [2-(propane-2-sulfonyl)-phenylamino]-pyrimidin-2-ylamino } - 3-methoxy-phenyl)-(3-dimethylamino-pyrrolidin-l-yl)-methanone;
4-{4-Amino-5-chloro-6-[2-(propane-2-sulfonyl)-phenylamino]-pyrimidin-2-ylamino}-N- (2-dimethylamino-ethyl)-3-methoxy-benzamide;
4-{4-Amino-5-chloro-6-[2-(propane-2-sulfonyl)-phenylamino]-pyrimidin-2-ylamino}-N- (4-amino-cyclohexyl)-3-methoxy-benzamide;
N-(4-Acetylamino-cyclohexyl)-4-{4-amino-5-chloro-6-[2-(propane-2-sulfonyl)- phenylamino]-pyrimidin-2-ylamino}-3-methoxy-benzamide;
4-{4-Amino-5-chloro-6-[2-(propane-2-sulfonyl)-phenylamino]-pyrimidin-2-ylamino}-N- (4-dimethylamino-cyclohexyl)-3-methoxy-benzamide;
4-{4-Amino-5-chloro-6-[2-(propane-2-sulfonyl)-phenylamino]-pyrimidin-2-ylamino}-N- (3-amino-cyclohexyl)-3-methoxy-benzamide;
N-(3-Acetylamino-cyclohexyl)-4-{4-amino-5-chloro-6-[2-(propane-2-sulfonyl)- phenylamino]-pyrimidin-2-ylamino}-3-methoxy-benzamide; and
4-{4-Amino-5-chloro-6-[2-(propane-2-sulfonyl)-phenylamino]-pyrimidin-2-ylamino}-N- (3-dimethylamino-cyclohexyl)-3-methoxy-benzamide; or a physiologically acceptable salt, solvate, hydrate or stereoisomer thereof. [0016] In yet other particular embodiments, the invention provides compounds having Formula I wherein Z is CH and said compound is selected from the group consisting of: l-(4-(4-(2-(isopropylsulfonyl)phenylamino)-lH-pyrrolo[2,3-b]pyridin-6-ylamino)-3- methoxyphenyl)piperidin-4-ol;
N4-(2-(isopropylsulfonyl)phenyl)-N6-(2-methoxy-4-(4-(4-methylpiperazin-l- yl)piperidin-l-yl)phenyl)-lH-pyrrolo[2,3-b]pyridine-4,6-diamine;
N6-(2-isopropoxy-5-methyl-4-(piperidin-4-yl)phenyl)-N4-(2-(isopropylsulfonyl)phenyl)- lH-pyrrolo[2,3-b]pyridine-4,6-diamine;
N6-(2-isopropoxy-5-methyl-4-(l-methylpiperidin-4-yl)phenyl)-N4-(2- (isopropylsulfonyl)phenyl)-lH-pyrrolo[2,3-b]pyridine-4,6-diamine;
N4-(2-(isopropylsulfonyl)phenyl)-N6-(2-methoxy-4-(4-methylpiperazin-l-yl)phenyl)- lH-pyrrolo[2,3-b]pyridine-4,6-diamine; 2-(4-(4-(4-(2-(isopropylsulfonyl)phenylamino)-lH-pyrrolo[2,3-b]pyridin-6-ylamino)-3- methoxyphenyl)piperazin- 1 -yl)ethanol;
N6-(4-(4-aminopiperidin-l-yl)-2-methoxyphenyl)-N4-(2-(isopropylsulfonyl)phenyl)-lH- pyrrolo[2,3-b]pyridine-4,6-diamine; l-(3-isopropoxy-4-(4-(2-(isopropylsulfonyl)phenylamino)-lH-pyrrolo[2,3-b]pyridin-6- ylamino)phenyl)piperidin-4-ol; l-(3-ethoxy-4-(4-(2-(isopropylsulfonyl)phenylamino)-lH-pyrrolo[2,3-b]pyridin-6- ylamino)phenyl)piperidin-4-ol; l-(4-(4-(2-(isopropylsulfonyl)phenylamino)-lH-pyrrolo[2,3-b]pyridin-6-ylamino)-3- methoxyphenyl)piperidin-3-ol;
N4-(2-(isopropylsulfonyl)phenyl)-N6-(2-methoxy-4-morpholinophenyl)-lH-pyrrolo[2,3- b]pyridine-4,6-diamine; and
N6-(2-isopropoxy-5-methyl-4-(l-methylpiperidin-4-yl)phenyl)-N4-(2- (isopropylsulfonyl)phenyl)-lH-pyrrolo[2,3-b]pyridine-4,6-diamine; or a physiologically acceptable salt, solvate, hydrate or stereoisomer thereof. [0017] In other particular embodiments, the invention provides compounds having Formula (III) and selected from the group consisting of:
N4-(2-(isopropylsulfonyl)phenyl)-N6-(2-methoxy-4-(l-methylpiperidin-4- yloxy)phenyl)-lH-pyrazolo[3,4-d]pyrimidine-4,6-diamine;
N4-(2-(isopropylsulfonyl)phenyl)-N6-(2-methoxy-4-(l-methylpiperidin-3-yl)phenyl)- lH-pyrazolo[3,4-d]pyrimidine-4,6-diamine;
N6-(2-isopropoxy-5-methyl-4-(l-methylpiperidin-4-yl)phenyl)-N4-(2- (isopropylsulfonyl)phenyl)-lH-pyrazolo[3,4-d]pyrimidine-4,6-diamine;
N6-(2-isopropoxy-5-methyl-4-(piperidin-4-yl)phenyl)-N4-(2-(isopropylsulfonyl)phenyl)- lH-pyrazolo[3,4-d]pyrimidine-4,6-diamine;
N6-(2-isopropoxy-5-methyl-4-(l-methylpiperidin-3-yl)phenyl)-N4-(2- (isopropylsulfonyl)phenyl)-lH-pyrazolo[3,4-d]pyrimidine-4,6-diamine; l,4'-bipiperidin-l'-yl(4-(4-(2-(isopropylsulfonyl)phenylamino)-lH-pyrazolo[3,4- d]pyrimidin-6-ylamino)-3-methoxyphenyl)methanone;
N6-(2-isopropoxy-5-methyl-4-(piperidin-4-yl)phenyl)-N4-(2-(methylsulfonyl)phenyl)- lH-pyrazolo[3,4-d]pyrimidine-4,6-diamine; 5'-methoxy-N,2'-dimethyl-4'-(4-(2-(methylsulfonyl)phenylamino)-lH-pyrazolo[3,4- d]pyrimidin-6-ylamino)biphenyl-4-carboxamide; and
N6-(2-isopropoxy-5-methyl-4-(l-methylpiperidin-4-yl)phenyl)-N4-(2- (methylsulfonyl)phenyl)-lH-pyrazolo[3,4-d]pyrimidine-4,6-diamine; or a physiologically acceptable salt, solvate, hydrate or stereoisomer thereof.
[0018] Another aspect of the invention encompasses a pharmaceutical composition comprising a compound according to Formula I, IA, II or III or a physiologically acceptable salt, solvate, hydrate or stereoisomer thereof and a pharmaceutically acceptable diluent or carrier. In some embodiments, the pharmaceutical composition the invention encompasses a composition wherein the compound is present in a therapeutically effective amount. In still another embodiment, the pharmaceutical composition the invention further encompasses at least one further active compound, such as for example, an anti-hyperproliferative agent. Another aspect of the invention encompasses a packaged pharmaceutical composition comprising a container, the pharmaceutical composition of the invention and instructions for using the pharmaceutical composition to treat a disease or condition in a mammal.
[0019] Yet another aspect of the invention encompasses a method of inhibiting kinase activity in a cell comprising contacting a cell with one or more compounds of the invention. In one embodiment, the kinase whose activity is inhibited is anaplastic lymphoma kinase, (ALK) hepatocyte growth factor receptor tyrosine kinase (c-Met), monopolar spindle (Mpsl) kinase, Ros, IGF-IR or InsR kinase. In particular examples, ALK activity is inhibited. In other examples, c-Met or Mpsl kinase activity is inhibited.
[0020] The invention also provides methods for treating a condition mediated by ALK, c- Met, Mpsl, Ros, IGFlR or InsR, comprising administering to a cell or tissue system or to a mammalian subject, a therapeutically effective amount of a compound of Formula I, IA, II or III, or pharmaceutically acceptable salts or tautomers thereof, and optionally in combination with a second therapeutic agent; thereby treating said condition mediated by ALK, c-Met, Mpsl, Ros, IGFlR or InsR. In particular embodiments, the compounds of the invention may be used for treating a condition mediated by ALK.
[0021] Alternatively, the present invention provides the use of a compound having Formula I, IA, II or III, in the manufacture of a medicament for treating a condition mediated by ALK, c- Met, Mpsl, Ros, IGFlR or InsR. In particular embodiments, the compounds of the invention may be used alone or in combination with a second therapeutic agent to treat a condition mediated by ALK, c-Met, Mpsl, Ros, IGFlR or InsR, wherein said condition is an autoimmune disease, a transplantation disease, an infectious disease or a cell proliferative disorder.
[0022] Furthermore, the invention provides methods for treating a cell proliferative or angiogenesis disorder, comprising administering to a system or subject in need of such treatment an effective amount of a compound having Formula I, IA, II or III, or pharmaceutically acceptable salts or pharmaceutical compositions thereof, and optionally in combination with a second therapeutic agent, thereby treating said disorder. Alternatively, the present invention provides the use of a compound having Formula I, IA, II or III, in the manufacture of a medicament for treating a cell-proliferative or angiogenesis disorder. In particular examples, the compounds of the invention may be used alone or in combination with a chemotherapeutic agent to treat a cell proliferative disorder, including but not limited to, multiple myeloma, neuroblastoma, lymphoma, leukemia, melanoma, sarcoma, osteosarcoma, synovial sarcoma, Ewing's sarcoma, hepatoma, gastrointestinal stromal tumor or a solid tumor or cancer of breast, renal, prostate, colorectal, thyroid, ovarian, pancreas, lung, uterus, respiratory tract, brain, digestive tract, urinary tract, eye, liver, skin, head and neck, thyroid or parathyroid.
[0023] In the above methods for using the compounds of the invention, a compound having Formula I, IA, II or III, may be administered to a system comprising cells or tissues, or to a mammalian subject such as a human or animal subject.
Definitions
[0024] The term "halogen" refers to radicals of fluorine, chlorine, bromine and iodine.
[0025] The term "alkyl" refers to a straight or branched hydrocarbon chain radical, containing solely carbon and hydrogen atoms, having in the range from one up to eight carbon atoms, and which is attached to the rest of the molecule by a single bond, such as illustratively, methyl, ethyl, n-propyl 1-methylethyl (iso-propyl), n-butyl, n-pentyl, and 1,1-dimethylethyl (tert-butyl).
[0026] The term "cycloalkyl" denotes a non-aromatic mono or multicyclic ring system having in the range of 3 up to 14 carbon atoms such as cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl. Examples of multicyclic cycloalkyl groups include decahydronapththyl. Examples of bridged cycloalkyl groups or sprirobicycloalkyl groups include adamantyl norbornyl, and spiro[4.4]nonyl groups. [0027] A "carbocyclic ring" as used herein refers to a saturated or partially unsaturated, monocyclic, fused bicyclic or bridged polycyclic ring containing carbon atoms, which may optionally be substituted, for example, with =0. Examples of carbocyclic rings include but are not limited to cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclopropylene, cyclohexanone, etc.
[0028] The term "alkoxy" denotes an alkyl group as defined herein attached via an oxygen linkage to the rest of the molecule. Representative examples of those groups are methoxy, ethoxy, iso-propoxy, n-butoxy, and tert-butoxy.
[0029] The term "cycloalkoxy" denotes a cycloalkyl group as defined herein attached via an oxygen linkage to the rest of the molecule. Representative examples of those groups are cyclopropoxy, cyclobutoxy, cyclopentoxy, cyclohexoxy, and cycloheptoxy.
[0030] The term "aryl" refers to aromatic radicals having in the range of 6 up to 14 carbon atoms such as phenyl, naphthyl, indanyl, and biphenyl.
[0031] The term "heteroaryl" refers to a stable 5- to 13-membered aromatic heterocycle having in the range of from 1 up to 4 heteroatoms from the group consisting of nitrogen, phosphorus, oxygen and sulfur, which ring or ring system can be linked via a carbon atom or a nitrogen atom, if such an atom is present. For purposes of this invention, the heteroaryl ring radical may be a monocyclic, bicyclic or tricyclic ring system. Examples of such heteroaryl radicals are: pyridyl, pyridyl N-oxide, pyrimidyl, pyridazinyl, pyrazinyl, thienyl, furyl, pyrrolyl, pyrazolyl, imidazolyl, thiazolyl, oxazolyl or isoxazolyl, indolicenyl, indolyl, benzo[b] thienyl, benzo[b]furyl, benzothiazolyl, benzothiadiazolyl, indazolyl, quinolyl, isoquinolyl, isoquinolyl, naphthyridinyl, quinazolinyl, oxadiazolyl, benzoxazolyl, tetrazoyl, triazolyl, thiadiazolyl, and benzimidazolyl.
[0032] The term "heterocycloalkyl" refers to a stable 3 to 13 membered saturated or partially unsaturated heterocycle having in the range from 1 up to 4 heteroatoms from the group consisting of nitrogen, phosphorus, oxygen and sulfur, which ring or ring system can be linked via a carbon atom or a nitrogen atom, if such an atom is present. For purposes of this invention, the heterocyclic ring radical may be a monocyclic, bicyclic or tricyclic ring system, which may include fused, bridged or spiro ring systems. Examples of such heterocyclyl radicals are: tetrahydropyranyl, aziridyl, azepanyl, tetrahydrofuryl, pyrrolidinyl, pyrrolinyl, piperidinyl, 1,2 dihydropyridinyl, 1,4 dihydropyridinyl, piperazinyl, morpholinyl, thiomorpholinyl, azepinyl, oxazolinyl, thiazolinyl and 1,4 diazepinyl. Examples of nitrogen non-aromatic heterocyclyl radicals include but are not limited to piperidinyl, piperazinyl, pyrrolidinyl, azetidinyl, morpholinyl, etc.
[0033] The term "alkylamino" refers to an alkyl group as defined herein attached via amino linkage to the rest of the molecule. The term alkylamino further includes dialkyl amino moieties in which two alkyl groups as define herein are attached via amino linkage to the rest of the molecule. Representative examples of those groups are methylamino and dimethylamino.
[0034] As used herein, an H atom in any substituent groups (e.g., CH2) encompasses all suitable isotopic variations, e.g., H, 2H and 3H.
[0035] Where the plural form of the word compounds, salts, polymorphs, hydrates, solvates and the like, is used herein, this is taken to mean also a single compound, salt, polymorph, isomer, hydrate, solvate or the like.
[0036] A solvate for the purpose of this invention is a complex of a solvent and a compound of the invention in the solid state. Exemplary solvates would include, but are not limited to, complexes of a compound of the invention with ethanol or methanol. Hydrates are a specific form of solvate wherein the solvent is water.
[0037] The term "treating" or "treatment" as used herein is used conventionally, e.g., the management or care of a subject for the purpose of combating, alleviating, reducing, relieving, improving the condition of, etc., of a disease or disorder, such as a carcinoma or cancer.
[0038] Where a tumor, a tumor disease, a carcinoma or a cancer is mentioned, metastasis in the original organ or tissue and/or in any other location is encompassed.
Detailed Description of the Invention
[0039] The present invention is based on the discovery that certain pyrimidine and pyrrolopyridine derivatives possess valuable, pharmacologically useful properties. For example, compounds and compositions described herein, including salts, metabolites, solvates, solvates of salts, hydrates, prodrugs such as esters, polymorphs, and stereoisomeric forms thereof, may exhibit anti-proliferative activity and are thus useful to prevent or treat the disorders associated with hyper-proliferation.
[0040] In one embodiment, the compounds of the invention are effective as protein tyrosine kinase inhibitors. For example, the compounds of the invention may inhibit the tyrosine kinase activity of anaplastic lymphoma kinase (ALK) and/or the fusion protein of NPM-ALK .This protein tyrosine kinase results from a gene fusion of nucleophosmin (NPM) and the anaplastic lymphoma kinase (ALK), rendering the protein tyrosine kinase activity of ALK ligand- independent. NPM-ALK plays a key role in signal transmission in a number of hematopoetic and other human cells leading to hematological and neoplastic diseases, for example in anaplastic large-cell lymphoma (ALCL) and non-Hodgkin's lymphomas (NHL), specifically in ALK+NHL or Alkomas, in inflammatory myofibroblastic tumors (IMT) and neuroblastomas. In addition to NPM-ALK, other gene fusions have been identified in human hematological and neoplastic diseases; mainly TPM3-ALK (a fusion of nonmuscle tropomyosin with ALK). ALK has been shown to become translocated to the echinoderm microtubule-associate protein-like 4 (EML4) in a subset of patients afflicted with Non small cell lung cancer (NSCLC). The EML4- ALK fusion tyrosine kinase generated transformed foci in culture and subcutaneous tumors in mice. Therefore EML4-ALK, which is inhibited by the compounds described in this application, maybe a relevant target for NSCLC patients or other patients with this or related ALK-containing fusion proteins (Nature. 2007 Aug 2;448(7153):561-6) The pyrimidine derivatives are useful for the inhibition of all such ALK-containing gene fusions. [0041] In one aspect, the invention provides compounds having the Formula I: pa
HN-R
R-X-(B)NΛZΛN®Y-R'
R3 H H φ
or a physiologically acceptable salt, solvate, hydrate or stereoisomer thereof, wherein:
A and B are each independently aryl or heteroaryl, each of which may be optionally substituted with one or more R4 groups;
Y is -SO2-, -SO2NH-, -NH-SO2-, -NH-C(O)-, -C(O)-NH-, -O-, or -NR2-;
X is NH, O or S;
Z is N or CH;
Rl is H, C1-C6 alkyl, halo-(Cl-C6 alkyl), C1-C6 cycloalkyl, halo-(Cl-C6 cycloalkyl), heterocyclyl, heterocyclylCl-όalkyl, aryl, arylCl-όalkyl, heteroaryl or heteroarylCl-όalkyl;
R2 is H, C1-C6 alkyl, halo-(Cl-C6 alkyl), C1-C6 cycloalkyl, halo-(Cl-C6 cycloalkyl), heterocyclyl, heterocyclylCl-όalkyl, aryl, arylCl-όalkyl, heteroaryl or heteroarylCl-όalkyl; R3 is R4, -CO-R5,
Figure imgf000016_0001
Figure imgf000016_0002
Ra is hydrogen and Rb is halogen, or Ra and Rb, taken together with the atoms to which they are bound form a pyrazolo or a pyrrolo ring fused to the pyrimidine ring, said pyrazolo or pyrrolo ring optionally bearing one or two R4 groups;
Each occurrence of R4 is independently halogen, C1-C6 alkyl, halo-(Cl-C6 alkyl), Cl- C6 cycloalkyl, halo-(Cl-C6 cycloalkyl), heterocyclyl, heterocyclylCl-όalkyl, aryl, arylCl- όalkyl, heteroaryl or heteroarylCl-όalkyl, Cl-6alkoxy, Cl-6alkylthio, hydroxyl, nitro, azido, cyano, acyloxy, carboxy, ester, carbamoyl, carboxamide, ureido, amidino, guanidine, sulfonyl, sulphonylamino, aminosulphonyl;
\— N NH Each occurrence of R5 is independently is C1-C6 alkoxy, ^ — / , hydroxy, N N-(C1-C6 alkyl)
Figure imgf000016_0003
, dialkylamino, or -N-R7;
I— N \ t— N N-R7 Each occurrence of R6 is independently is hydroxy, "^ — ' , ^^ — ^ , or -
C0NH2; and
Each occurrence of R7 is independently is hydrogen, C1-C6 alkyl, C1-C6 alkoxy, hydroxyl, or C1-C6 hydroxyalkyl;
With the proviso that when B is heteroaryl, -X-R2 and -R3 may each independently represent hydrogen.
[0042] In another embodiment, the invention encompasses a compound having the formula IA:
Figure imgf000017_0001
wherein: n is an integer from 0-3; m is an integer from 0-4; and Y, X, R1, R2, R3, R4, R5, R6, R7, Ra and Rb are as defined in Formula I. [0043] In yet another embodiment, the invention encompasses a compound having the formula II:
Figure imgf000017_0002
wherein:
N is an integer from 0-3 ; M is an integer from 0-4; P is an integer from 0-2; and Y, X, R1, R2, R3, R4, R5, R6, and R7 are as defined in Formula I. [0044] In still another embodiment, the invention encompasses a compound having the formula III:
Figure imgf000017_0003
wherein:
N is an integer from 0-3 ; M is an integer from 0-4; P is an integer from 0-1; and Y, X, R1, R2, R3, R4, R5, R6, and R7 are as defined in Formula I. [0045] Another aspect of the invention encompasses a compound having the IUPAC name: methyl 3-(4-amino-5-chloro-6-(2-(methylsulfonyl)phenylamino)pyrimidin-2-ylamino)-4- methoxybenzoate ;
(3-(4-amino-5-chloro-6-(2-(methylsulfonyl)phenylamino)pyrimidin-2-ylamino)-4- methoxyphenyl)(piperazin-l-yl)methanone; l-(4-(4-amino-5-chloro-6-(2-(methylsulfonyl)phenylamino)pyrimidin-2-ylamino)-3- methoxyphenyl)piperidin-4-ol; l-(4-(4-amino-5-chloro-6-(2-(methylsulfonyl)phenylamino)pyrimidin-2-ylamino)-3- methoxyphenyl)piperidin-4-ol;
5-chloro-N2-(2-methoxy-5-(2-(pyrrolidin-l-yl)ethoxy)phenyl)-N4-(2- (methylsulfonyl)phenyl)pyrimidine-2,4,6-triamine;
5-chloro-N2-(4-(4-(diethylamino)piperidin-l-yl)-2-isopropoxyphenyl)-N4-(2- (methylsulfonyl)phenyl)pyrimidine-2,4,6-triamine; l,4'-bipiperidin-l'-yl(4-(4-amino-5-chloro-6-(2-(methylsulfonyl)phenylamino)pyrimidin- 2-ylamino)-3-isopropoxyphenyl)methanone;
(4-(4-amino-5-chloro-6-(2-(methylsulfonyl)phenylamino)pyrimidin-2-ylamino)-3- isopropoxyphenyl)(4-(4-methylpiperazin- 1 -yl)piperidin- 1 -yl)methanone; l,4'-bipiperidin-l'-yl(4-(4-amino-5-chloro-6-(2-(methylsulfonyl)phenylamino)pyrimidin- 2-ylamino)-3-methoxyphenyl)methanone; l-(4-(4-amino-5-chloro-6-(2-(isopropylsulfonyl)phenylamino)pyrimidin-2-ylamino)-3- methoxyphenyl)piperidin-4-ol;
5-chloro-N4-(2-(isopropylsulfonyl)phenyl)-N2-(2-methoxy-5-(2-(pyrrolidin-l- yl)ethoxy)phenyl)pyrimidine-2,4,6-triamine;
5-chloro-N2-(4-(4-(diethylamino)piperidin-l-yl)-2-isopropoxyphenyl)-N4-(2- (isopropylsulfonyl)phenyl)pyrimidine-2,4,6-triamine; l,4'-bipiperidin-l'-yl(4-(4-amino-5-chloro-6-(2- (isopropylsulfonyl)phenylamino)pyrimidin-2-ylamino)-3-isopropoxyphenyl)methanone;
5-chloro-N4-(2-(isopropylsulfonyl)phenyl)-N2-(2-methoxy-4- morpholinophenyl)pyrimidine-2,4,6-triamine; l-(4-(4-amino-5-chloro-6-(2-(methylsulfonyl)phenylamino)pyrimidin-2-ylamino)-3- isopropoxyphenyl)piperidin-4-ol; l-(4-(4-amino-5-chloro-6-(2-(isopropylsulfonyl)phenylamino)pyrimidin-2-ylamino)-3- isopropoxyphenyl)piperidin-4-ol; l-(4-(6-amino-5-chloro-2-(2-(isopropylsulfonyl)phenylamino)pyrimidin-4-ylamino)-3- isopropoxyphenyl)piperidin-4-ol;
5-chloro-N2-(2-isopropoxy-5-methyl-4-(l-methylpiperidin-4-yl)phenyl)-N4-(2- (methylsulfonyl)phenyl)pyrimidine-2,4,6-triamine;
4'-(4-amino-5-chloro-6-(5-methyl-lH-pyrazol-3-ylamino)pyrimidin-2-ylamino)-5'- methoxy-N,2'-dimethylbiphenyl-4-carboxamide;
4'-(4-amino-5-chloro-6-(2-(methylsulfonyl)phenylamino)pyrimidin-2-ylamino)-5'- methoxy-N,2'-dimethylbiphenyl-4-carboxamide;
5-chloro-N2-(2-isopropoxy-5-methyl-4-(l-methylpiperidin-4-yl)phenyl)-N4-(2- (isopropylsulfonyl)phenyl)pyrimidine-2,4,6-triamine;
5-chloro-N2-(2-ethoxy-4-morpholinophenyl)-N4-(2-(methylsulfonyl)phenyl)pyrimidine- 2,4,6-triamine;
N2-(4-(l,4'-bipiperidin-l'-yl)-2-ethoxyphenyl)-5-chloro-N4-(2- (methylsulfonyl)phenyl)pyrimidine-2,4,6-triamine;
5-chloro-N4-(2-(isopropylsulfonyl)phenyl)-N2-(2-methoxy-4-(4-methylpiperazin-l- yl)phenyl)pyrimidine-2,4,6-triamine;
5-chloro-N2-(2-ethoxy-4-morpholinophenyl)-N4-(2- (isopropylsulfonyl)phenyl)pyrimidine-2,4,6-triamine; l-(4-(4-amino-5-chloro-6-(2-(isopropylsulfonyl)phenylamino)pyrimidin-2-ylamino)-3- ethoxyphenyl)piperidin-4-ol;
5-chloro-N4-(2-(isopropylsulfonyl)phenyl)-N2-(2-methoxy-4-(4-(4-methylpiperazin-l- yl)piperidin-l-yl)phenyl)pyrimidine-2,4,6-triamine;
2-(4-(4-(4-amino-5-chloro-6-(2-(isopropylsulfonyl)phenylamino)pyrimidin-2-ylamino)- 3-methoxyphenyl)piperazin-l-yl)ethanol;
(R)-l-(4-(4-amino-5-chloro-6-(2-(isopropylsulfonyl)phenylamino)pyrimidin-2-ylamino)- 3 -methoxypheny l)pyrrolidin- 3 -ol ; l-(4-(4-amino-5-chloro-6-(2-(isopropylsulfonyl)phenylamino)pyrimidin-2-ylamino)-3- methoxyphenyl)piperidin-3-ol;
5-chloro-N4-(2-(isopropylsulfonyl)phenyl)-N2-(2-methoxy-4-(l-methylpiperidin-3- yl)phenyl)pyrimidine-2,4,6-triamine; 5-chloro-N4-(2-(isopropylsulfonyl)phenyl)-N2-(2-methoxy-4-(l-methylpiperidin-4- yloxy)phenyl)pyrimidine-2,4,6-triamine;
N2-(4-(l,4'-bipiperidin-l'-yl)-2-ethoxyphenyl)-5-chloro-N4-(2- (isopropylsulfonyl)phenyl)pyrimidine-2,4,6-triamine;
5-chloro-N2-(6-(2,6-dimethylmorpholino)pyridin-3-yl)-N4-(2- (isopropylsulfonyl)phenyl)pyrimidine-2,4,6-triamine; l-(4-(4-amino-5-chloro-6-(2-(isopropylsulfonyl)phenylamino)pyrimidin-2-ylamino)-3- methoxyphenyl)piperidine-4-carboxamide; l,4'-bipiperidin-l'-yl(4-(4-amino-5-chloro-6-(2- (isopropylsulfonyl)phenylamino)pyrimidin-2-ylamino)-3-methoxyphenyl)methanone;
5-chloro-N2-(2-isopropoxy-5-methyl-4-(l-methylpiperidin-3-yl)phenyl)-N4-(2- (isopropylsulfonyl)phenyl)pyrimidine-2,4,6-triamine;
5-chloro-N2-(lH-indol-6-yl)-N4-(2-(methylsulfonyl)phenyl)pyrimidine-2,4,6-triamine;
4-(4-amino-5-chloro-6-(2-(isopropylsulfonyl)phenylamino)pyrimidin-2-ylamino)-3- methoxy-N-(4-(4-methylpiperazin-l-yl)phenyl)benzamide;
4-{4-Amino-5-chloro-6-[2-(propane-2-sulfonyl)-phenylamino]-pyrimidin-2-ylamino}-3- methoxy-N-(4-morpholin-4-yl-phenyl)-benzamide;
4-{4-Amino-5-chloro-6-[2-(propane-2-sulfonyl)-phenylamino]-pyrimidin-2-ylamino}-3- methoxy-N- [4-(4-methyl-piperazin- 1 -yl)-phenyl]-benzamide;
(4- { 4-Amino-5-chloro-6- [2-(propane-2-sulfonyl)-phenylamino]-pyrimidin-2-ylamino } - 3-methoxy-phenyl)-(4-pyrrolidin-l-yl-piperidin-l-yl)-methanone;
(4- { 4-Amino-5-chloro-6- [2-(propane-2-sulfonyl)-phenylamino]-pyrimidin-2-ylamino } - 3 -methoxy-phenyl)- [4-(4-methyl-piperazin- 1 -yl)-piperidin- 1 -yl] -methanone ;
3-[4-(4-{4-Amino-5-chloro-6-[2-(propane-2-sulfonyl)-phenylamino]-pyrimidin-2- ylamino } -3 -methoxy-benzoyl)-piperazin- 1 -yl] -propionitrile;
(4- { 4- Amino-5-chloro-ό- [2-(propane-2-sulfonyl)-phenylamino]-pyrimidin-2-ylamino } - 3-methoxy-phenyl)-(2-methyl-morpholin-4-yl)-methanone;
(4- { 4- Amino-5-chloro-ό- [2-(propane-2-sulfonyl)-phenylamino]-pyrimidin-2-ylamino } - 3-methoxy-phenyl)-(3-dimethylamino-pyrrolidin-l-yl)-methanone;
4-{4-Amino-5-chloro-6-[2-(propane-2-sulfonyl)-phenylamino]-pyrimidin-2-ylamino}-N- (2-dimethylamino-ethyl)-3-methoxy-benzamide; 4-{4-Amino-5-chloro-6-[2-(propane-2-sulfonyl)-phenylamino]-pyrimidin-2-ylamino}-N- (4-amino-cyclohexyl)-3-methoxy-benzamide;
N-(4-Acetylamino-cyclohexyl)-4-{4-amino-5-chloro-6-[2-(propane-2-sulfonyl)- phenylamino]-pyrimidin-2-ylamino}-3-methoxy-benzamide;
4-{4-Amino-5-chloro-6-[2-(propane-2-sulfonyl)-phenylamino]-pyrimidin-2-ylamino}-N- (4-dimethylamino-cyclohexyl)-3-methoxy-benzamide;
4-{4-Amino-5-chloro-6-[2-(propane-2-sulfonyl)-phenylamino]-pyrimidin-2-ylamino}-N- (3-amino-cyclohexyl)-3-methoxy-benzamide;
N-(3-Acetylamino-cyclohexyl)-4-{4-amino-5-chloro-6-[2-(propane-2-sulfonyl)- phenylamino]-pyrimidin-2-ylamino}-3-methoxy-benzamide;
4-{4-Amino-5-chloro-6-[2-(propane-2-sulfonyl)-phenylamino]-pyrimidin-2-ylamino}-N- (S-dimethylamino-cyclohexy^-S-methoxy-benzamide; or a physiologically acceptable salt, solvate, hydrate or stereoisomer thereof. [0046] Still another aspect of the invention encompasses a compound having the IUPAC name: l-(4-(4-(2-(isopropylsulfonyl)phenylamino)-lH-pyrazolo[3,4-d]pyrimidin-6-ylamino)-3- methoxyphenyl)piperidin-4-ol; l-(3-ethoxy-4-(4-(2-(isopropylsulfonyl)phenylamino)-lH-pyrazolo[3,4-d]pyrimidin-6- ylamino)phenyl)piperidin-4-ol;
N4-(2-(isopropylsulfonyl)phenyl)-N6-(2-methoxy-4-(l-methylpiperidin-4- yloxy)phenyl)-lH-pyrazolo[3,4-d]pyrimidine-4,6-diamine;
N4-(2-(isopropylsulfonyl)phenyl)-N6-(2-methoxy-4-(4-methylpiperazin-l-yl)phenyl)- lH-pyrazolo[3,4-d]pyrimidine-4,6-diamine;
N4-(2-(isopropylsulfonyl)phenyl)-N6-(2-methoxy-4-(4-(4-methylpiperazin-l- yl)piperidin-l-yl)phenyl)-lH-pyrazolo[3,4-d]pyrimidine-4,6-diamine;
N4-(2-(isopropylsulfonyl)phenyl)-N6-(2-methoxy-4-(l-methylpiperidin-3-yl)phenyl)- lH-pyrazolo[3,4-d]pyrimidine-4,6-diamine; l-(4-(4-(2-(isopropylsulfonyl)phenylamino)-lH-pyrazolo[3,4-d]pyrimidin-6-ylamino)-3- methoxyphenyl)piperidine-4-carboxamide;
N6-(4-(l,4'-bipiperidin-l'-yl)-2-ethoxyphenyl)-N4-(2-(isopropylsulfonyl)phenyl)-lH- pyrazolo[3,4-d]pyrimidine-4,6-diamine; N6-(2-isopropoxy-5-methyl-4-(l-methylpiperidin-4-yl)phenyl)-N4-(2- (isopropylsulfonyl)phenyl)-lH-pyrazolo[3,4-d]pyrimidine-4,6-diamine;
N6-(2-isopropoxy-5-methyl-4-(piperidin-4-yl)phenyl)-N4-(2-(isopropylsulfonyl)phenyl)- lH-pyrazolo[3,4-d]pyrimidine-4,6-diamine;
N6-(2-isopropoxy-5-methyl-4-(l-methylpiperidin-3-yl)phenyl)-N4-(2- (isopropylsulfonyl)phenyl)-lH-pyrazolo[3,4-d]pyrimidine-4,6-diamine; l,4'-bipiperidin-l'-yl(4-(4-(2-(isopropylsulfonyl)phenylamino)-lH-pyrazolo[3,4- d]pyrimidin-6-ylamino)-3-methoxyphenyl)methanone;
N6-(2-ethoxy-4-morpholinophenyl)-N4-(2-(methylsulfonyl)phenyl)-lH-pyrazolo[3,4- d]pyrimidine-4,6-diamine; l-(3-methoxy-4-(4-(2-(methylsulfonyl)phenylamino)-lH-pyrazolo[3,4-d]pyrimidin-6- ylamino)phenyl)piperidin-4-ol;
N6-(4-(4-(diethylamino)piperidin-l-yl)-2-isopropoxyphenyl)-N4-(2- (methylsulfonyl)phenyl)-lH-pyrazolo[3,4-d]pyrimidine-4,6-diamine; l-(3-isopropoxy-4-(4-(2-(methylsulfonyl)phenylamino)-lH-pyrazolo[3,4-d]pyrimidin-6- ylamino)phenyl)piperidin-4-ol;
N6-(2-methoxy-4-(4-(4-methylpiperazin-l-yl)piperidin-l-yl)phenyl)-N4-(2- (methylsulfonyl)phenyl)-lH-pyrazolo[3,4-d]pyrimidine-4,6-diamine;
N6-(2-isopropoxy-5-methyl-4-(piperidin-4-yl)phenyl)-N4-(2-(methylsulfonyl)phenyl)- lH-pyrazolo[3,4-d]pyrimidine-4,6-diamine;
5'-methoxy-N,2'-dimethyl-4'-(4-(2-(methylsulfonyl)phenylamino)-lH-pyrazolo[3,4- d]pyrimidin-6-ylamino)biphenyl-4-carboxamide;
N6-(2-isopropoxy-5-methyl-4-(l-methylpiperidin-4-yl)phenyl)-N4-(2- (methylsulfonyl)phenyl)-lH-pyrazolo[3,4-d]pyrimidine-4,6-diamine; or a physiologically acceptable salt, solvate, hydrate or stereoisomer thereof. [0047] Yet another aspect of the invention encompasses a compound having the IUPAC name: methyl 6-(4-(2-(isopropylsulfonyl)phenylamino)-lH-pyrrolo[2,3-b]pyridin-6- ylamino)nicotinate;
6-(4-(2-(isopropylsulfonyl)phenylamino)-lH-pyrrolo[2,3-b]pyridin-6-ylamino)-N- methylnicotinamide; (6-(4-(2-(isopropylsulfonyl)phenylamino)-lH-pyrrolo[2,3-b]pyridin-6-ylamino)pyridin- 3-yl)(4-(4-methylpiperazin- l-yl)piperidin- l-yl)methanone; l-(4-(4-(2-(isopropylsulfonyl)phenylamino)-lH-pyrrolo[2,3-b]pyridin-6-ylamino)-3- methoxyphenyl)piperidin-4-ol;
N4-(2-(isopropylsulfonyl)phenyl)-N6-(2-methoxy-4-(4-(4-methylpiperazin-l- yl)piperidin-l-yl)phenyl)-lH-pyrrolo[2,3-b]pyridine-4,6-diamine;
N6-(2-isopropoxy-5-methyl-4-(piperidin-4-yl)phenyl)-N4-(2-(isopropylsulfonyl)phenyl)- lH-pyrrolo[2,3-b]pyridine-4,6-diamine;
N6-(2-isopropoxy-5-methyl-4-(l-methylpiperidin-4-yl)phenyl)-N4-(2- (isopropylsulfonyl)phenyl)-lH-pyrrolo[2,3-b]pyridine-4,6-diamine;
N4-(2-(isopropylsulfonyl)phenyl)-N6-(2-methoxy-4-(4-methylpiperazin-l-yl)phenyl)- lH-pyrrolo[2,3-b]pyridine-4,6-diamine;
2-(4-(4-(4-(2-(isopropylsulfonyl)phenylamino)-lH-pyrrolo[2,3-b]pyridin-6-ylamino)-3- methoxyphenyl)piperazin- 1 -yl)ethanol;
N6-(4-(4-aminopiperidin-l-yl)-2-methoxyphenyl)-N4-(2-(isopropylsulfonyl)phenyl)-lH- pyrrolo[2,3-b]pyridine-4,6-diamine; l,4'-bipiperidin-l'-yl(4-(4-(2-(isopropylsulfonyl)phenylamino)-lH-pyrrolo[2,3- b]pyridin-6-ylamino)-3-methoxyphenyl)methanone;
(2-(4-(2-(isopropylsulfonyl)phenylamino)-lH-pyrrolo[2,3-b]pyridin-6-ylamino)pyridin- 4-yl)(4-(4-methylpiperazin- l-yl)piperidin- l-yl)methanone;
(2-(4-(2-(isopropylsulfonyl)phenylamino)-lH-pyrrolo[2,3-b]pyridin-6-ylamino)pyridin- 4-yl)(4-methylpiperazin-l-yl)methanone;
2-(4-(2-(isopropylsulfonyl)phenylamino)-lH-pyrrolo[2,3-b]pyridin-6-ylamino)-N-(3- morpholinopropyl)isonicotinamide;
(4-(4-methylpiperazin-l-yl)piperidin-l-yl)(6-(4-(2-(methylsulfonyl)phenylamino)-lH- pyrrolo[2,3-b]pyridin-6-ylamino)pyridin-3-yl)methanone; l-(3-isopropoxy-4-(4-(2-(isopropylsulfonyl)phenylamino)-lH-pyrrolo[2,3-b]pyridin-6- ylamino)phenyl)piperidin-4-ol; l-(3-ethoxy-4-(4-(2-(isopropylsulfonyl)phenylamino)-lH-pyrrolo[2,3-b]pyridin-6- ylamino)phenyl)piperidin-4-ol; l-(4-(4-(2-(isopropylsulfonyl)phenylamino)-lH-pyrrolo[2,3-b]pyridin-6-ylamino)-3- methoxyphenyl)piperidin-3-ol; or a physiologically acceptable salt, solvate, hydrate or stereoisomer thereof.
[0048] The compounds of this invention may contain one or more asymmetric centers, depending upon the location and nature of the various substituents desired. Asymmetric carbon atoms may be present in the (R) or (S) configuration, resulting in racemic mixtures in the case of a single asymmetric center, and diastereomeric mixtures in the case of multiple asymmetric centers. In certain instances, asymmetry may also be present due to restricted rotation about a given bond, for example, the central bond adjoining two substituted aromatic rings of the specified compounds. Substituents on a ring may also be present in either cis or trans form. It is intended that all such configurations (including enantiomers and diastereomers), are included within the scope of the present invention. Separated, pure or partially purified isomers and stereoisomers or racemic or diastereomeric mixtures of the compounds of this invention are also included within the scope of the present invention. The purification and the separation of such materials can be accomplished by standard techniques known in the art.
[0049] The optical isomers can be obtained by resolution of the racemic mixtures according to conventional processes, for example, by the formation of diastereoisomeric salts using an optically active acid or base or formation of covalent diastereomers. Examples of appropriate acids are tartaric, diacetyltartaric, ditoluoyltartaric and camphorsulfonic acid. Mixtures of diastereoisomers can be separated into their individual diastereomers on the basis of their physical and/or chemical differences by methods known in the art, for example, by chromatography or fractional crystallization. The optically active bases or acids are then liberated from the separated diastereomeric salts. A different process for separation of optical isomers involves the use of chiral chromatography (e.g., chiral HPLC columns), with or without conventional derivitization, optimally chosen to maximize the separation of the enantiomers. Suitable chiral HPLC columns are manufactured by Diacel, e.g., Chiracel OD and Chiracel OJ among many others, all routinely selectable. Enzymatic separations, with or without derivitization, are also useful. The optically active compounds of this invention can likewise be obtained by chiral syntheses utilizing optically active starting materials.
[0050] The present invention also relates to useful forms of the compounds as disclosed herein, such as pharmaceutically acceptable salts, co-precipitates, metabolites, hydrates, solvates and prodrugs of all the compounds of examples. The term "pharmaceutically acceptable salt" refers to a relatively non-toxic, inorganic or organic acid addition salt of a compound of the present invention. For example, see S. M. Berge, et al. "Pharmaceutical Salts," J. Pharm. Sci. 1977, 66, 1-19. Pharmaceutically acceptable salts include those obtained by reacting the main compound, functioning as a base, with an inorganic or organic acid to form a salt, for example, salts of hydrochloric acid, sulfuric acid, phosphoric acid, methane sulfonic acid, camphor sulfonic acid, oxalic acid, maleic acid, succinic acid and citric acid. Pharmaceutically acceptable salts also include those in which the main compound functions as an acid and is reacted with an appropriate base to form, e.g., sodium, potassium, calcium, magnesium, ammonium, and chorine salts. Those skilled in the art will further recognize that acid addition salts of the claimed compounds may be prepared by reaction of the compounds with the appropriate inorganic or organic acid via any of a number of known methods. Alternatively, alkali and alkaline earth metal salts of acidic compounds of the invention are prepared by reacting the compounds of the invention with the appropriate base via a variety of known methods.
[0051] Representative salts of the compounds of this invention include the conventional nontoxic salts and the quaternary ammonium salts which are formed, for example, from inorganic or organic acids or bases by means well known in the art. For example, such acid addition salts include acetate, adipate, alginate, ascorbate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate, cinnamate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate, glucoheptanoate, glycerophosphate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, 2- hydroxyethanesulfonate, itaconate, lactate, maleate, mandelate, methanesulfonate, 2- naphthalenesulfonate, nicotinate, nitrate, oxalate, pamoate, pectinate, persulfate, 3- phenylpropionate, picrate, pivalate, propionate, succinate, sulfonate, tartrate, thiocyanate, tosylate, and undecanoate.
[0052] Base salts include alkali metal salts such as potassium and sodium salts, alkaline earth metal salts such as calcium and magnesium salts, and ammonium salts with organic bases such as dicyclohexylamine and N-methyl-D-glucamine. Additionally, basic nitrogen containing groups may be quaternized with such agents as lower alkyl halides such as methyl, ethyl, propyl, and butyl chlorides, bromides and iodides; dialkyl sulfates like dimethyl, diethyl, and dibutyl sulfate; and diamyl sulfates, long chain halides such as decyl, lauryl, myristyl and strearyl chlorides, bromides and iodides, aralkyl halides like benzyl and phenethyl bromides and others.
[0053] The present invention also includes all suitable isotopic variations of the compounds of the invention, or pharmaceutically acceptable salts thereof. An isotopic variation of a compound of the invention or a pharmaceutically acceptable salt thereof is defined as one in which at least one atom is replaced by an atom having the same atomic number but an atomic mass different from the atomic mass usually found in nature. Examples of isotopes that may be incorporated into the compounds of the invention and pharmaceutically acceptable salts thereof include but are not limited to isotopes of hydrogen, carbon, nitrogen and oxygen such as 2H, 3H, 11C, 13C, 14C, 15N, 170, 180, 35S, 18F, 36Cl and 123I. Certain isotopic variations of the compounds of the invention and pharmaceutically acceptable salts thereof, for example, those in which a radioactive isotope such as 3H or 14C is incorporated, are useful in drug and/or substrate tissue distribution studies.
[0054] In particular examples, 3H and 14C isotopes may be used for their ease of preparation and detectability. In other examples, substitution with isotopes such as H may afford certain therapeutic advantages resulting from greater metabolic stability, such as increased in vivo half- life or reduced dosage requirements. Isotopic variations of the compounds of the invention or pharmaceutically acceptable salts thereof can generally be prepared by conventional procedures using appropriate isotopic variations of suitable reagents. Isotopic variations of the compounds have the potential to change a compound' s metabolic fate and/or create small changes in physical properties such as hydrophobicity, and the like. Isotopic variation have the potential to enhance efficacy and safety, enhance bioavailability and half-life, alter protein binding, change biodistribution, increase the proportion of active metabolites and/or decrease the formation of reactive or toxic metabolites.
[0055] In each of the above formula, each optionally substituted moiety may be substituted with Ci-6 alkyl, C2-6 alkenyl or C3-6 alkynyl, each of which may be optionally halogenated or optionally having a carbon that may be replaced or substituted with N, S, O, or a combination thereof (for example, hydroxylCi-Csalkyl, Ci-CsalkoxyCi-Csalkyl); halo, amino, amidino, Ci_6 alkoxy; hydroxyl, methylenedioxy, carboxy; C1-8 alkylcarbonyl, C1-8 alkoxycarbonyl, carbamoyl, Ci_8 alkylcarbamoyl, sulfamoyl, cyano, oxo, nitro, or an optionally substituted carbocyclic ring, heterocyclic ring, aryl or heteroaryl as previously described.
Method of treating hyper-proliferative disorders
[0056] The compounds of the invention may be selectively toxic or more toxic to rapidly proliferating cells than to normal cells, and may be used to treat hyper-proliferative disorders. For example, the compounds of the invention may have significant antiproliferative effects in human cancer cells, e.g., cancerous tumors, and promote differentiation, e.g., cell cycle arrest and apoptosis. The compounds of the invention may be utilized to inhibit, block, reduce or decrease cell proliferation and/or cell division, and/or produce apoptosis. In one embodiment, the method comprises administering to a mammal in need thereof, including a human, an amount of a compound of this invention, or a pharmaceutically acceptable salt, isomer, polymorph, metabolite, hydrate, solvate or ester thereof; etc. which is effective to treat the disorder.
[0057] Hyper-proliferative disorders include but are not limited, e.g., psoriasis, keloids, and other hyperplasias affecting the skin, benign prostate hyperplasia (BPH), solid tumors, such as cancers of the breast, respiratory tract, brain, reproductive organs, digestive tract, urinary tract, eye, liver, skin, head and neck, thyroid, parathyroid and their distant metastases. Those disorders also include lymphomas, sarcomas, and leukemias. These disorders have been well characterized in humans, but also exist with a similar etiology in other mammals, and can be treated by administering pharmaceutical compositions of the present invention.
[0058] Examples of breast cancer include, but are not limited to invasive ductal carcinoma, invasive lobular carcinoma, ductal carcinoma in situ, and lobular carcinoma in situ.
[0059] Examples of cancers of the respiratory tract include, but are not limited to small-cell and non-small-cell lung carcinoma, as well as bronchial adenoma and pleuropulmonary blastoma.
[0060] Examples of brain cancers include, but are not limited to brain stem and hypophtalmic glioma, cerebellar and cerebral astrocytoma, medulloblastoma, ependymoma, as well as neuroectodermal and pineal tumor.
[0061] Tumors of the male reproductive organs include, but are not limited to prostate and testicular cancer. Tumors of the female reproductive organs include, but are not limited to endometrial, cervical, ovarian, vaginal, and vulvar cancer, as well as sarcoma of the uterus.
[0062] Tumors of the digestive tract include, but are not limited to anal, colon, colorectal, esophageal, gallbladder, gastric, pancreatic, rectal, small-intestine, and salivary gland cancers.
[0063] Tumors of the urinary tract include, but are not limited to bladder, penile, kidney, renal pelvis, ureter, urethral and human papillary renal cancers.
[0064] Eye cancers include, but are not limited to intraocular melanoma and retinoblastoma. [0065] Examples of liver cancers include, but are not limited to hepatocellular carcinoma (liver cell carcinomas with or without fibrolamellar variant), cholangiocarcinoma (intrahepatic bile duct carcinoma), and mixed hepatocellular cholangiocarcinoma.
[0066] Skin cancers include, but are not limited to squamous cell carcinoma, Kaposi's sarcoma, malignant melanoma, Merkel cell skin cancer, and non-melanoma skin cancer.
[0067] Head-and-neck cancers include, but are not limited to laryngeal, hypopharyngeal, nasopharyngeal, oropharyngeal cancer, lip and oral cavity cancer and squamous cell. Lymphomas include, but are not limited to AIDS-related lymphoma, non-Hodgkin's lymphoma, cutaneous T-cell lymphoma, Burkitt lymphoma, Hodgkin' s disease, and lymphoma of the central nervous system.
[0068] Sarcomas include, but are not limited to sarcoma of the soft tissue, osteosarcoma, malignant fibrous histiocytoma, lymphosarcoma, and rhabdomyosarcoma.
[0069] Leukemias include, but are not limited to acute myeloid leukemia, acute lymphoblastic leukemia, chronic lymphocytic leukemia, chronic myelogenous leukemia, and hairy cell leukemia.
Methods of treating Ros, Insulin-Like Growth Factor 1 Receptor (IGFlR), Insulin Receptor (InsR), Anaplastic Lymphoma Kinase (ALK), Hepatocyte growth factor receptor tyrosine kinase (c-Met), and Nucleophosmin-ALK disorders
[0070] The present invention also provides methods for the treatment of disorders associated with aberrant expression of Ros, IGFlR, InsR, ALK, Hepatocyte growth factor receptor tyrosine kinase (c-Met), and/or Nucleophosmin-ALK.
[0071] The compounds described in this application are ATP-competitive kinase inhibitors. As such they competitively block ATP from binding to the kinase active site and thereby prevent phosphorylation of downstream substrates. This effectively blocks signal transduction from the targeted kinases. Compounds in this application have the potential to interact with any kinase in the human kinome and have been tested for their ability to bind to a panel of 320 distinct protein kinases. This panel includes the following kinases: AAKl, ABLl, ABL1(E255K), ABL1(H396P), ABL1(M351T), ABL1(Q252H), ABL1(T315I), ABL1(Y253F), AB L2, ACVRl, ACVRlB, ACVR2A, ACVR2B, ACVRLl, ADCK3,ADCK4,AKT1,AKT2, AKT3,ALK,AMPK-alphal,AMPK-alpha2, ANKKl, ARK5, AURKA, AURKB, AURKC, AXL, BIKE, BLK, BMPRlA, BMPR2, BMX, BRAF, BRAF(V600E), BRSKl, BRS K2, BTK, CAMKl, CAMKlD, CAMKlG, CAMK2A, CAMK2B, CAMK2D, CAMK2G, CAMK4, CAMKKl, CAMKK2, CDC2L1, CDC2L2, CDKI l, CDK2, CDK3, CDK5, CDK7, CDK8, CDK9, CHEKl, CIT, CLKl, CLK2, CLK3, CLK4, CSFlR, CSK, CSNKlAlL, CSNKlD, CSNKlE, CSNKlGl, CSNK1G2, CSNK1G3, CSNK2A1, CSNK2A2, DAPKl, DAPK2D, APK3, DCAMKLl, DCAMKL2, DCAMKL3, DDRl, DDR2, DLK, DMPK, DMPK2, DRAKl, DRAK2, DYRKlB, EGFR, EGFR(E746-A750del), EGFR(G719C), EGFR(G719S), EGFR(L747-E749del, A750P), EGFR(L747-S752del, P753S), EGFR(L747, T751del, Sins), EGFR(L858R), EGFR(L861Q), EGFR(S752-I759del), EPHAl, EPHA2, EPHA3, EPHA4, EPHA5, EPHA6, EPHA7, EPHA8, EPHBl, EPHB2, EPHB3, EPHB4, ERBB2, ERBB4, ERKl, ERK2, ERK3, ERK4, ERK5, ERK8, FER, FES, FGFRl, FGFR2, FGFR3, FGFR3(G697C), FGFR4, FGR, FLTl, FLT3, FLT3(D835H), FLT3(D835Y), FLT3(ITD), FLT3(N841I), FLT4, FRK, FYN, GAK, GCN2(Kin.Dom.2, S808G), GSK3A, GSK3B, HCK, IGFlR, IKKepsilon, INSR, INSRR, IRAK3, ITK, JAKl(Kin.Dom.l), JAK2(Kin.Dom.2), JAK3(Kin.Dom.2), JNKl, JNK2, JNK3, KIT, KIT(D816V), KIT(V559D), KIT(V559D, T670I), KIT(V559D, V654A), LATSl, LATS2, LCK, LIMKl, LIMK2, LKBl, LOK, LTK, LYN, MAP3K4, MAP3K5, MAP4K1, MAP4K3, MAP4K4, MAP4K5, MAPKAPK2, MAPKAPK5, MARKl, MARK2, MARK3, MARK4, MEKl, MEK2, MEK3, MEK4, MEK6, MELK, MERTK, MET, MKNKl, MKNK2, MLCK, MLKl, MLK2, MLK3, MRCKA, MRCKB, MSTl, MST2, MST3, MST4, MUSK, MYLK, MYLK2, MY03A, MYO3B, NDR2, NEKl, NEK2, NEK5, NEK6, NEK7, NEK9, NLK, p38-alpha, p38-beta, p38-gamma, PAKl, PAK2, PAK3, PAK4, PAK6, PAK7/PAK5, PCTKl, PCTK2, PCTK3, PDGFRA, PDGFRB, PDPKl, PFTKl, PHKGl, PHKG2, PIK3CA, PIK3CA(E545K), PIMl, PIM2, PIM3, PIP5K1A, PIP5K2B, PKACalpha, PKACbeta, PKMYTl, PKNl, PKN2, PLKl, PLK3, PLK4, PRKCD, PRKCE, PRKCH, PRKCQ, PRKDl, PRKD2, PRKD3, PRKGl, PRKG2, PRKR, PRKX, PTK2, PTK2B, PTK6, RAFl, RET, RET(M918T), RIOKl, RIOK3, RIPKl, RIPK2, ROSl, RPSόKAl(Kin.Dom.l), RPS6KAl(Kin.Dom.2), RPS6KA2(Kin.Dom.l), RPS6KA2(Kin.Dom.2), RPS6KA3(Kin.Dom.l), RPS6KA4(Kin.Dom.l), RPS6KA4(Kin.Dom.2), RPS6KA5(Kin.Dom.l), RPS6KA5(Kin.Dom.2), RPS6KA6(Kin.Dom.l), RPS6KA6(Kin.Dom.2), SgK085, SLK, SNARK, SNFlLK, SNF1LK2, SRC, SRMS, SRPKl, SRPK2, STK16, STK33, STK36, SYK, TEC, TESKl, TGFBRl, TGFBR2, TIEl, TIE2, TLKl, TLK2, TNIK, TNKl, TNK2, TNNI3K, TRKA, TRKB, TRKC, TSSKl, TTK, TXK, TYK2(Kin.Dom.2), TYRO3, VEGFR2, WEEl, YANK2, YANK3, YES, YSKl, ZAK. This panel includes both the wild-type kinase and mutants that have been discovered to occur naturally or are predicted to appear based upon clinical experience with other kinases.
[0072] In particular, the compounds of the invention demonstrate inhibitory activity against: ADCK3, ADCK4, ALK, CLKl, CLK4, EGFR, EGFR(E746-A750del), EGFR(L747-E749del, A750P), EGFR(L747-S752del, P753S), EGFR(L747-T751del,Sins), EGFR(L858R), EGFR(L861Q), EGFR(S752-I759del), ERBB4, FER, FES, GAK, IGFlR, INSR, INSRR, LTK, PTK2, PTK2B, ROSl, RPS6KAl(Kin.Dom.2), TNKl, TNK2, TTK.
[0073] The ALK inhibitory activity and inhibitory activity against ALK-containing gene fusions of the compounds described herein make them useful pharmaceutical agents for the treatment of proliferative diseases. The inventive compounds are particularly useful for treating a tumor which is a breast cancer, genitourinary cancer, lung cancer, gastrointestinal cancer, epidermoid cancer, melanoma, ovarian cancer, pancreas cancer, neuroblastoma, head and/or neck cancer or bladder cancer, or in a broader sense renal, brain or gastric cancer; in particular (i) a breast tumor; an epidermoid tumor, such as an epidermoid head and/or neck tumor or a mouth tumor; a lung tumor, for example a small cell or non-small cell lung tumor; a gastrointestinal tumor, for example, a colorectal tumor; or a genitourinary tumor, for example, a prostate tumor (especially a hormone-refractory prostate tumor); or (ii) a proliferative disease that is refractory to the treatment with other chemotherapeutics; or (iii) a tumor that is refractory to treatment with other chemotherapeutics due to multidrug resistance.
[0074] In a broader sense of the invention, a proliferative disease may furthermore be a hyperproliferative condition such as leukemias, hyperplasias, fibrosis (especially pulmonary, but also other types of fibrosis, such as renal fibrosis), angiogenesis, psoriasis, atherosclerosis and smooth muscle proliferation in the blood vessels, such as stenosis or restenosis following angioplasty. Proliferative diseases treated according to the present method include tumors of blood and lymphatic system (e.g. Hodgkin's disease, Non-Hodgkin's lymphoma, Burkitt's lymphoma, AIDS-related lymphomas, malignant immunoproliferative diseases, multiple myeloma and malignant plasma cell neoplasms, lymphoid leukemia, acute or chronic myeloid leukemia, acute or chronic lymphocytic leukemia, monocytic leukemia, other leukemias of specified cell type, leukemia of unspecified cell type, other and unspecified malignant neoplasms of lymphoid, haematopoietic and related tissues, for example diffuse large cell lymphoma, T-cell lymphoma or cutaneous T-cell lymphoma). Myeloid cancer includes e.g. acute or chronic myeloid leukaemia. Methods of Treating Neurotrophic Growth Factor Receptor Tyrosine Kinase A, B and C Disorders
[0075] The present invention also provides methods of treating disorders and diseases associated with aberrant expression of the neurotrophic growth factor receptor tyrosine kinase A, B and C (Trk A, B, C also known as NTRKl, 2, and 3).
[0076] The compounds in the present invention exhibit inhibition of the neurotrophic growth factor receptor tyrosine kinase A, B and C (Trk A, B, C also known as NTRKl, 2, and 3). Several lines of evidence have implicated NTRKs in the development and progression of cancer through deregulation of tyrosine kinase activity by mutations, chromosomal rearrangements, upregulation of either the receptor, their ligand (Nerve Growth Factor, Brain Derived Neurotropic Factor, Neurotrophins) or both. Chromosomal translocations involving both NTRKl & 3 have been found in several different types of tumors. Gene rearrangements involving NTRKl and a set of different fusion partners (TPR, TPM3, TFG) are a hallmark of a subset of papillary thyroid cancers (PTC) (Neuro Endocrinol Lett. 2007 Jun 12;28(3):221-229; Cancer Treat Res. 2004;122:207-19). Rare cancers such as secretory breast cancer, infant fibrosarcoma and congenital mesoblastic nephroma have been shown to be associated with a chromosomal rearrangement t(12;15) generating a ETV6-NTRK3 fusion gene that has been shown to have constitutive kinase activity and transforming potential in several different cell lines including fibroblasts, hematopoietic cells and breast epithelial cells (Nat Genet. 1998 Feb; 18(2): 184-7; Oncogene. 2000 Feb 17;19(7):906-15). TrkB is of central importance in preventing anoikis (detachment-induced apoptosis) which is believed to an important requirement in the metastatic process. Genetic abnormalities such as point mutations and chromosomal rearrangements of NTRK2 and NTRK3 have been found in a variety of cancer types (Nature, vol 446, p. 153, 8 March 2007). Hence agents that block TrkB kinase activity may serve as antimetastatic agents (Cell MoI Life Sci. 2006 Apr;63(7-8):755-9).
Methods of treating angiogenic disorders
[0077] The present invention also provides methods of treating disorders and diseases associated with excessive and/or abnormal angiogenesis.
[0078] Inappropriate and ectopic expression of angiogenesis can be deleterious to an organism. A number of pathological conditions are associated with the growth of extraneous blood vessels. These include, e.g., diabetic retinopathy, ischemic retinal-vein occlusion, and retinopathy of prematurity (Aiello et al. New Engl. J. Med. 1994, 331, 1480; Peer et al. Lab. Invest. 1995, 72, 638), age-related macular degeneration (AMD; see, Lopez et al. Invest. Opththalmol. Vis. Sci. 1996, 37, 855), neovascular glaucoma, psoriasis, retrolental fibroplasias, angiofibroma, inflammation, rheumatoid arthritis (RA), restenosis, in-stent restenosis, vascular graft restenosis, etc. In addition, the increased blood supply associated with cancerous and neoplastic tissue, encourages growth, leading to rapid tumor enlargement and metastasis. Moreover, the growth of new blood and lymph vessels in a tumor provides an escape route for renegade cells, encouraging metastasis and the consequence spread of the cancer. Thus, compounds of the present invention can be utilized to treat and/or prevent any of the aforementioned angiogenesis disorders, e.g., by inhibiting and/or reducing blood vessel formation; by inhibiting, blocking, reducing, decreasing, etc. endothelial cell proliferation or other types involved in angiogenesis, as well as causing cell death or apoptosis of such cell types.
Dose and administration
[0079] Based upon standard laboratory techniques known to evaluate compounds useful for the treatment of hyper-proliferative disorders and angiogenic disorders, by standard toxicity tests and by standard pharmacological assays for the determination of treatment of the conditions identified above in mammals, and by comparison of these results with the results of known medicaments that are used to treat these conditions, the effective dosage of the compounds of this invention can readily be determined for treatment of each desired indication. The amount of the active ingredient to be administered in the treatment of one of these conditions can vary widely according to such considerations as the particular compound and dosage unit employed, the mode of administration, the period of treatment, the age and sex of the patient treated, and the nature and extent of the condition treated.
[0080] The total amount of the active ingredient to be administered will generally range from about 0.001 mg/kg to about 200 mg/kg body weight per day, and preferably from about 0.01 mg/kg to about 20 mg/kg body weight per day. In certain applications, the average daily oral dosing will be from about 200 mg/day to about 600 mg/day which corresponds to 2.8-8.6 mg/kg for a person with an average weight of 70 kgs. Clinically useful dosing schedules will range from one to three times a day dosing to once every four weeks dosing. In addition, "drug holidays" in which a patient is not dosed with a drug for a certain period of time, may be beneficial to the overall balance between pharmacological effect and tolerability. A unit dosage may contain from about 0.5 mg to about 1500 mg of active ingredient, and can be administered one or more times per day or less than once a day. The average daily dosage for administration by injection, including intravenous, intramuscular, subcutaneous and parenteral injections, and use of infusion techniques will preferably be from 0.01 to 200 mg/kg of total body weight. The average daily rectal dosage regimen will preferably be from 0.01 to 200 mg/kg of total body weight. The average daily vaginal dosage regimen will preferably be from 0.01 to 200 mg/kg of total body weight. The average daily topical dosage regimen will preferably be from 0.1 to 200 mg administered between one to four times daily. The transdermal concentration will preferably be that required to maintain a daily dose of from 0.01 to 200 mg/kg. The average daily inhalation dosage regimen will preferably be from 0.01 to 100 mg/kg of total body weight.
[0081] Of course the specific initial and continuing dosage regimen for each patient will vary according to the nature and severity of the condition as determined by the attending diagnostician, the activity of the specific compound employed, the age and general condition of the patient, time of administration, route of administration, rate of excretion of the drug, drug combinations, and the like. The desired mode of treatment and number of doses of a compound of the present invention or a pharmaceutically acceptable salt or ester or composition thereof can be ascertained by those skilled in the art using conventional treatment tests.
Combination therapies
[0082] The compounds of this invention can be administered as the sole pharmaceutical agent or in combination with one or more other pharmaceutical agents where the combination causes no unacceptable adverse effects. For example, the compounds of this invention can be combined with known anti-hyper-proliferative or other indication agents, and the like, as well as with admixtures and combinations thereof.
[0083] The additional pharmaceutical agent can be aldesleukin, alendronic acid, alfaferone, alitretinoin, allopurinol, aloprim, aloxi, altretamine, aminoglutethimide, amifostine, amrubicin, amsacrine, anastrozole, anzmet, aranesp, arglabin, arsenic trioxide, aromasin, 5-azacytidine, azathioprine, BCG or tice BCG, bestatin, betamethasone acetate, betamethasone sodium phosphate, bexarotene, bleomycin sulfate, broxuridine , bortezomib, busulfan, calcitonin, campath, capecitabine, carboplatin, casodex, cefesone, celmoleukin, cerubidine, chlorambucil, cisplatin, cladribine, cladribine, clodronic acid, cyclophosphamide, cytarabine, dacarbazine, dactinomycin, DaunoXome, decadron, decadron phosphate, delestrogen, denileukin diftitox, depo-medrol, deslorelin, dexrazoxane, diethylstilbestrol, diflucan, docetaxel, doxifluridine, doxorubicin, dronabinol, DW- 166HC, eligard, elitek, ellence, emend, epirubicin, epoetin alfa, epogen, eptaplatin, ergamisol, estrace, estradiol, estramustine phosphate sodium, ethinyl estradiol, ethyol, etidronic acid, etopophos, etoposide, fadrozole, farston, filgrastim, finasteride, filgrastim, floxuridine, fluconazole, fludarabine, 5-fluorodeoxyuridine monophosphate, 5- fluorouracil (5-FU), fluoxymesterone, flutamide, formestane, fosteabine, fotemustine, fulvestrant, gammagard, gemcitabine, gemtuzumab, gleevec, gliadel, goserelin, granisetron HCl, histrelin, hycamtin, hydrocortone, eyrthro-hydroxynonyladenine, hydroxyurea, ibritumomab tiuxetan, idarubicin, ifosfamide, interferon alpha, interferon- alpha 2, interferon alfa-2A, interferon alfa-2B, interferon alfa-nl, interferon alfa-n3, interferon beta, interferon gamma- Ia, interleukin-2, intron A, iressa, irinotecan, kytril, lentinan sulphate, letrozole, leucovorin, leuprolide, leuprolide acetate, levamisole, levofolinic acid calcium salt, levothroid, levoxyl, lomustine, lonidamine, marinol, mechlorethamine, mecobalamin, medroxyprogesterone acetate, megestrol acetate, melphalan, menest, 6-mercaptopurine, Mesna, methotrexate, metvix, miltefosine, minocycline, mitomycin C, mitotane, mitoxantrone, Modrenal, Myocet, nedaplatin, neulasta, neumega, neupogen, nilutamide, nolvadex, NSC-631570, OCT-43, octreotide, ondansetron HCl, orapred, oxaliplatin, paclitaxel, pediapred, pegaspargase, Pegasys, pentostatin, picibanil, pilocarpine HCl, pirarubicin, plicamycin, porfimer sodium, prednimustine, prednisolone, prednisone, premarin, procarbazine, procrit, raltitrexed, rebif, rhenium- 186 etidronate, rituximab, roferon-A, romurtide, salagen, sandostatin, sargramostim, semustine, sizofiran, sobuzoxane, solu-medrol, sparfosic acid, stem-cell therapy, streptozocin, strontium-89 chloride, synthroid, tamoxifen, tamsulosin, tasonermin, tastolactone, taxotere, teceleukin, temozolomide, teniposide, testosterone propionate, testred, thioguanine, thiotepa, thyrotropin, tiludronic acid, topotecan, toremifene, tositumomab, trastuzumab, treosulfan, tretinoin, trexall, trimethylmelamine, trimetrexate, triptorelin acetate, triptorelin pamoate, UFT, uridine, valrubicin, vesnarinone, vinblastine, vincristine, vindesine, vinorelbine, virulizin, zinecard, zinostatin stimalamer, zofran, ABI-007, acolbifene, actimmune, affinitak, aminopterin, arzoxifene, asoprisnil, atamestane, atrasentan, BAY 43-9006 (sorafenib), avastin, CCI-779, CDC-501, celebrex, cetuximab, crisnatol, cyproterone acetate, decitabine, DN-101, doxorubicin- MTC, dSLIM, dutasteride, edotecarin, eflornithine, exatecan, fenretinide, histamine dihydrochloride, histrelin hydrogel implant, holmium-166 DOTMP, ibandronic acid, interferon gamma, intron-PEG, ixabepilone, keyhole limpet hemocyanin, L-651582, lanreotide, lasofoxifene, libra, lonafarnib, miproxifene, minodronate, MS-209, liposomal MTP-PE, MX-6, nafarelin, nemorubicin, neovastat, nolatrexed, oblimersen, onco-TCS, osidem, paclitaxel polyglutamate, pamidronate disodium, PN-401, QS-21, quazepam, R- 1549, raloxifene, ranpirnase, 13-cis -retinoic acid, satraplatin, seocalcitol, T- 138067, tarceva, taxoprexin, thymosin alpha 1, tiazofurine, tipifarnib, tirapazamine, TLK-286, toremifene, TransMID-107R, valspodar, vapreotide, vatalanib, verteporfin, vinflunine, Z-100, zoledronic acid or combinations thereof.
[0084] Optional anti-hyper-proliferative agents which can be added to the composition include but are not limited to compounds listed on the cancer chemotherapy drug regimens in the 11th Edition of the Merck Index, (1996), which is hereby incorporated by reference, such as asparaginase, bleomycin, carboplatin, carmustine, chlorambucil, cisplatin, colaspase, cyclophosphamide, cytarabine, dacarbazine, dactinomycin, daunorubicin, doxorubicin (adriamycine), epirubicin, etoposide, 5-fluorouracil, hexamethylmelamine, hydroxyurea, ifosfamide, irinotecan, leucovorin, lomustine, mechlorethamine, 6-mercaptopurine, mesna, methotrexate, mitomycin C, mitoxantrone, prednisolone, prednisone, procarbazine, raloxifen, streptozocin, tamoxifen, thioguanine, topotecan, vinblastine, vincristine, and vindesine.
[0085] Other anti-hyper-proliferative agents suitable for use with the composition of the invention include but are not limited to those compounds acknowledged to be used in the treatment of neoplastic diseases in Goodman and Gilman's The Pharmacological Basis of Therapeutics (Ninth Edition), editor Molinoff et al., publ. by McGraw-Hill, pages 1225-1287, (1996), which is hereby incorporated by reference, such as aminoglutethimide, L-asparaginase, azathioprine, 5-azacytidine cladribine, busulfan, diethylstilbestrol, 2',2'-difluorodeoxycytidine, docetaxel, erythrohydroxynonyl adenine, ethinyl estradiol, 5-fluorodeoxyuridine, 5- fluorodeoxyuridine monophosphate, fludarabine phosphate, fluoxymesterone, flutamide, hydroxyprogesterone caproate, idarubicin, interferon, medroxyprogesterone acetate, megestrol acetate, melphalan, mitotane, paclitaxel, pentostatin, N-phosphonoacetyl-L-aspartate (PALA), plicamycin, semustine, teniposide, testosterone propionate, thiotepa, trimethylmelamine, uridine, and vinorelbine.
[0086] Other anti-hyper-proliferative agents suitable for use with the composition of the invention include but are not limited to other anti-cancer agents such as epothilone and its derivatives, irinotecan, raloxifen and topotecan. Dosage Forms and Modes of Administration
[0087] In general, compounds of the invention will be administered in therapeutically effective amounts via any of the usual and acceptable modes known in the art, either singly or in combination with one or more therapeutic agents. A therapeutically effective amount may vary widely depending on the severity of the disease, the age and relative health of the subject, the potency of the compound used and other factors known to those of ordinary skill in the art. For example, for the treatment of neoplastic diseases and immune system disorders, the required dosage will also vary depending on the mode of administration, the particular condition to be treated and the effect desired.
[0088] Oral Dosage Forms. Compounds of the invention and compositions comprising them that are suitable for oral administration can be presented as discrete dosage forms, such as, but are not limited to, tablets (e.g., chewable tablets), caplets, capsules, and liquids (e.g., flavored syrups). Such dosage forms contain predetermined amounts of active ingredients, and may be prepared by methods of pharmacy well known to those skilled in the art. See generally, Remington's Pharmaceutical Sciences, 18th ed., Mack Publishing, Easton Pa. (1990).
[0089] Typical oral dosage forms of the invention are prepared by combining the active ingredient(s) in an intimate admixture with at least one excipient according to conventional pharmaceutical compounding techniques. Excipients can take a wide variety of forms depending on the form of preparation desired for administration. For example, excipients suitable for use in oral liquid or aerosol dosage forms include, but are not limited to, water, glycols, oils, alcohols, flavoring agents, preservatives, and coloring agents. Examples of excipients suitable for use in solid oral dosage forms (e.g., powders, tablets, capsules, and caplets) include, but are not limited to, starches, sugars, micro-crystalline cellulose, diluents, granulating agents, lubricants, binders, and disintegrating agents.
[0090] Because of their ease of administration, tablets and capsules represent very advantageous oral dosage unit forms, in which case solid excipients are employed. If desired, tablets can be coated by standard aqueous or nonaqueous techniques. Such dosage forms can be prepared by any of the methods of pharmacy. In general, pharmaceutical compositions and dosage forms are prepared by uniformly and intimately admixing the active ingredients with liquid carriers, finely divided solid carriers, or both, and then shaping the product into the desired presentation if necessary. [0091] For example, a tablet can be prepared by compression or molding. Compressed tablets can be prepared by compressing in a suitable machine the active ingredients in a free- flowing form such as powder or granules, optionally mixed with an excipient. Molded tablets can be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
[0092] Examples of excipients that can be used in oral dosage forms of the invention include, but are not limited to, binders, fillers, disintegrants, and lubricants. Binders suitable for use in pharmaceutical compositions and dosage forms include, but are not limited to, corn starch, potato starch, or other starches, gelatin, natural and synthetic gums such as acacia, sodium alginate, alginic acid, other alginates, powdered tragacanth, guar gum, cellulose and its derivatives (e.g., ethyl cellulose, cellulose acetate, carboxymethyl cellulose calcium, sodium carboxymethyl cellulose), polyvinyl pyrrolidone, methyl cellulose, pre-gelatinized starch, hydroxypropyl methyl cellulose, (e.g., nos. 2208, 2906, 2910), microcrystalline cellulose, and mixtures thereof.
[0093] Examples of fillers suitable for use in the pharmaceutical compositions and dosage forms disclosed herein include, but are not limited to, talc, calcium carbonate (e.g., granules or powder), microcrystalline cellulose, powdered cellulose, dextrates, kaolin, mannitol, silicic acid, sorbitol, starch, pre-gelatinized starch, and mixtures thereof. The binder or filler in pharmaceutical compositions of the invention is typically present in from about 50 to about 99 weight percent of the pharmaceutical composition or dosage form.
[0094] Suitable forms of microcrystalline cellulose include, but are not limited to, the materials sold as AVICEL-PH- 101, AVICEL-PH- 103 AVICEL RC-581, AVICEL-PH- 105 (available from FMC Corporation, American Viscose Division, Avicel Sales, Marcus Hook, Pa.), and mixtures thereof. An specific binder is a mixture of microcrystalline cellulose and sodium carboxymethyl cellulose sold as AVICEL RC-581. Suitable anhydrous or low moisture excipients or additives include AVICEL-PH- 103.TM and Starch 1500 LM.
[0095] Disintegrants are used in the compositions of the invention to provide tablets that disintegrate when exposed to an aqueous environment. Tablets that contain too much disintegrant may disintegrate in storage, while those that contain too little may not disintegrate at a desired rate or under the desired conditions. Thus, a sufficient amount of disintegrant that is neither too much nor too little to detrimentally alter the release of the active ingredients should be used to form solid oral dosage forms of the invention. The amount of disintegrant used varies based upon the type of formulation, and is readily discernible to those of ordinary skill in the art. Typical pharmaceutical compositions comprise from about 0.5 to about 15 weight percent of disintegrant, specifically from about 1 to about 5 weight percent of disintegrant.
[0096] Disintegrants that can be used in pharmaceutical compositions and dosage forms of the invention include, but are not limited to, agar-agar, alginic acid, calcium carbonate, microcrystalline cellulose, croscarmellose sodium, crospovidone, polacrilin potassium, sodium starch glycolate, potato or tapioca starch, pre-gelatinized starch, other starches, clays, other algins, other celluloses, gums, and mixtures thereof.
[0097] Lubricants that can be used in pharmaceutical compositions and dosage forms of the invention include, but are not limited to, calcium stearate, magnesium stearate, mineral oil, light mineral oil, glycerin, sorbitol, mannitol, polyethylene glycol, other glycols, stearic acid, sodium lauryl sulfate, talc, hydrogenated vegetable oil (e.g., peanut oil, cottonseed oil, sunflower oil, sesame oil, olive oil, corn oil, and soybean oil), zinc stearate, ethyl oleate, ethyl laureate, agar, and mixtures thereof. Additional lubricants include, for example, a syloid silica gel (AEROSIL 200, manufactured by W. R. Grace Co. of Baltimore, Md.), a coagulated aerosol of synthetic silica (marketed by Degussa Co. of Piano, Tex.), CAB-O-SIL (a pyrogenic silicon dioxide product sold by Cabot Co. of Boston, Mass.), and mixtures thereof. If used at all, lubricants are typically used in an amount of less than about 1 weight percent of the pharmaceutical compositions or dosage forms into which they are incorporated.
[0098] Parenteral Dosage Forms. Parenteral dosage forms can be administered to patients by various routes including, but not limited to, subcutaneous, intravenous (including bolus injection and constant infusion), intramuscular, and intraarterial. Because their administration typically bypasses patients' natural defenses against contaminants, parenteral dosage forms are preferably sterile or capable of being sterilized prior to administration to a patient. Examples of parenteral dosage forms include, but are not limited to, solutions ready for injection, dry products (including, but not limited to lyophilized powders, pellets, and tablets) ready to be dissolved or suspended in a pharmaceutically acceptable vehicle for injection, suspensions ready for injection, and emulsions.
[0099] Suitable vehicles that can be used to provide parenteral dosage forms of the invention are well known to those skilled in the art. Examples include, but are not limited to: Water for Injection USP; aqueous vehicles such as, but not limited to, Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, and Lactated Ringer's Injection; water-miscible vehicles such as, but not limited to, ethyl alcohol, polyethylene glycol, and polypropylene glycol; and non-aqueous vehicles such as, but not limited to, corn oil, cottonseed oil, peanut oil, sesame oil, ethyl oleate, isopropyl myristate, and benzyl benzoate. Compounds that increase the solubility of one or more of the active ingredients disclosed herein can also be incorporated into the parenteral dosage forms of the invention.
[0100] Transdermal, Topical, And Mucosal Dosage Forms. Transdermal, topical, and mucosal dosage forms of the invention include, but are not limited to, ophthalmic solutions, sprays, aerosols, creams, lotions, ointments, gels, solutions, emulsions, suspensions, or other forms known to one of skill in the art. See, e.g., Remington's Pharmaceutical Sciences, 16th and 18th eds., Mack Publishing, Easton Pa. (1980 & 1990); and Introduction to Pharmaceutical Dosage Forms, 4th ed., Lea & Febiger, Philadelphia (1985). Dosage forms suitable for treating mucosal tissues within the oral cavity can be formulated as mouthwashes or as oral gels. Further, transdermal dosage forms include "reservoir type" or "matrix type" patches, which can be applied to the skin and worn for a specific period of time to permit the penetration of a desired amount of active ingredients.
[0101] Suitable excipients (e.g., carriers and diluents) and other materials that can be used to provide transdermal, topical, and mucosal dosage forms encompassed by this invention are well known to those skilled in the pharmaceutical arts, and depend on the particular tissue to which a given pharmaceutical composition or dosage form will be applied. With that fact in mind, typical excipients include, but are not limited to, water, acetone, ethanol, ethylene glycol, propylene glycol, butane-l,3-diol, isopropyl myristate, isopropyl palmitate, mineral oil, and mixtures thereof to form lotions, tinctures, creams, emulsions, gels or ointments, which are nontoxic and pharmaceutically acceptable. Moisturizers or humectants can also be added to pharmaceutical compositions and dosage forms if desired. Examples of such additional ingredients are well known in the art. See, e.g., Remington's Pharmaceutical Sciences, 16th and 18th eds., Mack Publishing, Easton Pa. (1980 & 1990).
[0102] Depending on the specific tissue to be treated, additional components may be used prior to, in conjunction with, or subsequent to treatment with active ingredients of the invention. For example, penetration enhancers can be used to assist in delivering the active ingredients to the tissue. Suitable penetration enhancers include, but are not limited to: acetone; various alcohols such as ethanol, oleyl, and tetrahydrofuryl; alkyl sulfoxides such as dimethyl sulfoxide; dimethyl acetamide; dimethyl formamide; polyethylene glycol; pyrrolidones such as polyvinylpyrrolidone; Kollidon grades (Povidone, Polyvidone); urea; and various water-soluble or insoluble sugar esters such as Tween 80 (polysorbate 80) and Span 60 (sorbitan monostearate).
[0103] The pH of a pharmaceutical composition or dosage form, or of the tissue to which the pharmaceutical composition or dosage form is applied, may also be adjusted to improve delivery of one or more active ingredients. Similarly, the polarity of a solvent carrier, its ionic strength, or tonicity can be adjusted to improve delivery. Compounds such as stearates can also be added to pharmaceutical compositions or dosage forms to advantageously alter the hydrophilicity or lipophilicity of one or more active ingredients so as to improve delivery. In this regard, stearates can serve as a lipid vehicle for the formulation, as an emulsifying agent or surfactant, and as a delivery-enhancing or penetration-enhancing agent. Different salts, hydrates or solvates of the active ingredients can be used to further adjust the properties of the resulting composition.
Kits
[0104] This invention encompasses kits which, when used by the medical practitioner, can simplify the administration of appropriate amounts of active ingredients to a patient.
[0105] A typical kit of the invention comprises one or more unit dosage forms of a compound of the invention or one or more compositions comprising a compound of the invention, or physiologically acceptable salts thereof, and instructions for use.
[0106] Kits of the invention can further comprise devices that are used to administer a compound of the invention to a patient. Examples of such devices include, but are not limited to, intravenous cannulation devices, syringes, drip bags, patches, topical gels, pumps, tubing, containers that provide protection from photodegredation, and inhalers.
[0107] Kits of the invention can further comprise pharmaceutically acceptable vehicles that can be used to administer one or more active ingredients. For example, if an active ingredient is provided in a solid form that must be reconstituted for parenteral administration, the kit can comprise a sealed container of a suitable vehicle in which the active ingredient can be dissolved to form a particulate-free sterile solution that is suitable for parenteral administration. Examples of pharmaceutically acceptable vehicles include, but are not limited to: Water for Injection USP; aqueous vehicles such as, but not limited to, Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, and Lactated Ringer's Injection; water-miscible vehicles such as, but not limited to, ethyl alcohol, polyethylene glycol, and polypropylene glycol; and non-aqueous vehicles such as, but not limited to, corn oil, cottonseed oil, peanut oil, sesame oil, ethyl oleate, isopropyl myristate, and benzyl benzoate.
Examples
[0108] The invention will now be further described by way of the following non-limiting examples. In general, the compounds of Formula I, particularly Formula IA can be prepared through the preparation shown in the Reaction Scheme 1 below.
mCPBA (4 0 eqiv) MC, 0 0C
Figure imgf000041_0001
Figure imgf000041_0002
aniline (2 0 eqiv), 4Λ/ HCI Dioxane (2 0 eqiv) H O2S. 2-Buthanol, 120 0C, 24 h
Figure imgf000041_0003
Scheme 1
Example 1-0 l-(4-(4-amino-5-chloro-6-(2-(methylsulfonyl)phenylamino)pyrimidin-2-ylamino)-3- methoxvphenyl)piperidin-4-ol
Figure imgf000042_0001
2,5,6-trichloro-N-(2-(methylthio)phenyl)pyrimidin-4-amine
Figure imgf000042_0002
[0109] To a solution of 2,3,5,6-tetrachloropyrimidine (5.0 g, 22.94 mmol) in dioxane (100 mL) was added 2-(methylthio)benzenamine (2.9 mL, 22.94 mmol) and diisopropylethylamine (4.2 mL, 25.23 mmol). The reaction mixture was stirred at 55 0C for 24 hours after which it was poured into ice water and the resulting solid was collected by filtration. The solid was dried (6.7 g, 91% yield) and used without further purification
2,5,6-trichloro-N-(2-(methylsulfonyl)phenyl)pyrimidin-4-amine
Figure imgf000042_0003
[0110] To a solution of 2,5,6-trichloro-N-(2-(methylthio)phenyl)pyrimidin-4-amine (6.7 g, 20.89 mmol) in dichloromethane (100 mL) was added mCPBA (14.3 g, 83.58 mmol). The reaction mixture was stirred for 4 hours after which it was diluted with dichloromethane (100 mL). The organic layer was washed with satd. ΝaHCθ3 solution and washed with brine. The organic layer was dried over MgSO4, filtered and concentrated. The crude product was purified by flash column chromatography using a 95:5 v/v Hexane:Ethyl acetate as solvent to afford title compound (5.2 g, 71 %) as a white solid.
2,5-dichloro-N4-(2-(methylsulfonyl)phenyl)pyrimidine-4,6-diamine
Figure imgf000043_0001
[0111] A sealed tube was charged with 2,5,6-trichloro-N-(2-
(methylsulfonyl)phenyl)pyrimidin-4-amine (500 mg, 1.42 mmol), 2.0 M ΝH3 in isopropyl alcohol (7.0 mL) and isopropyl alcohol (7.0 mL). The tube was sealed and the reaction mixture was stirred at 80 0C for 30 hours. The reaction mixture was poured into ice water and the resulting white solid was filtered and dried. Two isomers (380 mg, 80% yield), i.e. 2,5-dichloro- N4-(2-(methylsulfonyl)phenyl)pyrimidine-4,6-diamine and 5,6-dichloro-Ν4-(2- (methylsulfonyl)phenyl)pyrimidine-2,4-diamine were produced in a 3 : 1 ratio.
2,5-dichloro-N4-(2-(methylsulfonyl)phenyl)pyrimidine-4,6-diamine: 1H ΝMR 600 MHz (DMSO-J6) δ 9.16 (s, IH), 8.21 (d, J = 7.8 Hz, IH), 7.87 (dd, J = 1.2 Hz, J = 7.8Hz, IH), 7.73 (dt, J = 1.2 Hz, J = 8.4 Hz, IH), 7Λ5(bs, 2H), 7.33 (dt, J = 1.2 Hz, J = 8.4 Hz, IH), 3.22 (s, 3H), MS m/z : 333.12 (M + 1).
5,6-dichloro-Ν4-(2-(methylsulfonyl)phenyl)pyrimidine-2,4-diamine: 1H NMR 600 MHz (DMSO-J6) δ 9.35 (s, IH), 8.52 (J, J = 8.4 Hz, IH), 7.88 (dd, J = 1.2 Hz, J = 8.4 Hz, IH), 7.71 (dt, J = 1.2 Hz, J = 8.4 Hz, IH), 7.35 (dt, J = 1.2 Hz, J = 8.4 Hz, IH), 7.01 (bs, 2H), 3.24 (s, 3H), MS m/z : 333.12 (M + 1).
l-(4-(4-amino-5-chloro-6-(2-(methylsulfonyl)phenylamino)pyrimidin-2-ylamino)-3- methoxyphenyl)piperidin-4-ol
Figure imgf000044_0001
[0112] A sealed tube was charged with the isomers of 2,5-dichloro-N4-(2-
(methylsulfonyl)phenyl)pyrimidine-4,6-diamine and 5 ,6-dichloro-N4-(2- (methylsulfonyl)phenyl)pyrimidine-2,4-diamine (100 mg, 0.30 mmol), l-(4-amino-3- methoxyphenyl)piperidin-4-ol (134 mg, 0.60 mmol), 4N HC1 dioxane (0.15 mL) and 2-butanol (1.0 mL). The tube was sealed and the reaction mixture was stirred at 120 0C for 24 hours. The reaction mixture was then partitioned between ethyl acetate and water. The organic layer was separated and the aqueous layer was neutralized with satd. ΝaHCθ3 solution and extracted with ethyl acetate. The combined organic extracts were washed with brine, dried over MgSO4, filtered and concentrated. The residue was dissolved in DMSO (3 mL). The resulting solution was subjected to purification by reverse-phase HPLC to yield the title compound as a TFA salt. The product containing fraction was neutralized with satd. NaHCθ3 solution and extracted with ethyl acetate dried over MgSO4, filtered and concentrated. Two isomers, i.e. l-(4-(4-amino-5- chloro-6-(2-(methylsulfonyl)phenylamino)pyrimidin-2-ylamino)-3-methoxyphenyl)piperidin-4- ol and l-(4-(2-amino-5-chloro-6-(2-(methylsulfonyl)phenylamino)pyrimidin-4-ylamino)-3- methoxyphenyl)piperidin-4-ol were produced.
l-(4-(4-amino-5-chloro-6-(2-(methylsulfonyl)phenylamino)pyrimidin-2-ylamino)-3- methoxyphenyl)piperidin-4-ol: 1H NMR 600 MHz (CDCl3) δ 8.93 (s, IH), 8.40 (d, J = 8.4 Hz, IH), 8.00 (d, J = 9.0 Hz, IH), 7.89 (dd, J = 1.2 Hz, J = 7.8 Hz, IH), 7.54 (dt, J = 1.8 Hz, J = 8.4 Hz, IH), 7.15 (dt, J = 1.2 Hz, J = 7.8 Hz, IH), 7.06 (s, IH), 6.50 (m, IH), 6.41 {dd, J = 1.8 Hz, J = 8.4 Hz, IH), 5.02 (5, 2H), 3.78 (m, 4H), 3.41 (m, 2H), 3.00 (s, 3H), 2.84(m, 2H), 1.98(m, 2H), 1.69 (m, 2H), MS m/z : 519.47 (M + 1). l-(4-(2-amino-5-chloro-6-(2-(methylsulfonyl)phenylamino)pyrimidin-4-ylamino)-3- methoxyphenyl)piperidin-4-ol: 1H NMR 600 MHz (CDCl3) δ 8.87 (5, IH), 8.36 (d, J = 8.4 Hz, IH), 8.12 (d, J = 9.0 Hz, IH), 7.87 (dd, J = 1.8 Hz, J = 7.8 Hz, IH), 7.53 (dt, J = 1.8 Hz, J = 8.4 Hz, IH), 7.36 (s, IH), 7.11 (dt, J = 1.2 Hz, J = 7.8 Hz, IH), 6.50 (m, 2H), 4.65 (s, 2H), 3.83 (s, 3H), 3.77 (m, IH), 3.43 (m, 2H), 3.00 (5, 3H), 2.85(m, 2H), 1.96(m, 2H), 1.69 (m, 2H), MS m/z : 519.47 (M + 1).
[0113] By repeating the procedures described in the above examples, using appropriate amines, the following compounds in Table 1 are prepared.
Table 1
Figure imgf000045_0001
Figure imgf000046_0001
Figure imgf000047_0001
(s, (d, J J = 8.4 Hz, (m,
Figure imgf000048_0001
Figure imgf000049_0001
Figure imgf000050_0002
[0114] Alternatively, the compounds of Formula I, particularly Formula IA can be prepared through the preparation shown in the Reaction Scheme 2 below.
Figure imgf000050_0001
Scheme 2
Example 2-0 l-(4-(4-amino-5-chloro-6-(2-(isopropylsulfonyl)phenylamino)pyrimidin-2-ylamino)-3- methoxvphenyl)piperidin-4-ol
Figure imgf000051_0001
2,5,6-trichloro-N-(2-(isopropylsulfonyl)phenyl)pyrimidin-4-amine
Figure imgf000051_0002
[0115] A sealed tube was charged with 2,3,5,6-tetrachloropyrimidine (2.18 g, 10.03 mmol), 2-(isopropylsulfonyl)benzenamine (2.0 mg, 10.03 mmol), 4N HC1 dioxane (2.5 mL) and dioxane (50.0 mL). The tube was sealed and the reaction mixture was stirred at 120 0C for 24 hours. The reaction mixture was then partitioned between ethyl acetate and water. The organic layer was separated and the aqueous layer was neutralized with satd. ΝaHCθ3 solution and extracted with ethyl acetate. The combined organic extracts were washed with brine, dried over MgSO4, filtered and concentrated. The crude product was purified by flash column chromatography using a 95:5 v/v hexane:ethyl acetate as solvent to afford title compound (2.6 g, 68% yield) as a white solid. 1H NMR 600 MHz (CDCl3) δ 10.18 (5, IH), 8.53 (dd, J = 1.2, J = 8.4 Hz, IH), 7.92 (dd, J = 1.2 Hz, J = 8.4 Hz, IH), 7.38 (dt, J = 1.8 Hz, J = 8.4 Hz, IH), 7.35 (dt, J = 1.2 Hz, J = 7.2 Hz, IH), 3.21 (m, IH), 1.31 (d, J = 6.6 Hz, 6H). 2,5-dichloro-N4-(2-(isopropylsulfonyl)phenyl)pyrimidine-4,6-diamine
Figure imgf000052_0001
[0116] A sealed tube was charged with 2,5,6-trichloro-N-(2-
(methylsulfonyl)phenyl)pyrimidin-4-amine (900 mg, 1.42 mmol), 2.0 M NH3 in isopropyl alcohol (10 mL) and isopropyl alcohol (10.0 mL). The tube was sealed and the reaction mixture was stirred at 80 0C for 30 hours. The reaction mixture was poured into ice water and the resulting white solid was filtered and dried, yielding (560 mg) mixture of two isomers 2,5- dichloro-N4-(2-(isopropylsulfonyl)phenyl)pyrimidine-4,6-diamine and 5,6-dichloro-N4-(2- (isopropylsulfonyl)phenyl)pyrimidine-2,4-diamine.
l-(4-(4-amino-5-chloro-6-(2-(isopropylsulfonyl)phenylamino)pyrimidin-2-ylamino)-3- methoxyphenyl)piperidin-4-ol
Figure imgf000052_0002
[0117] A sealed tube was charged with the isomers of 2,5-dichloro-N4-(2-
(isopropylsulfonyl)phenyl)pyrimidine-4,6-diamine and 5,6-dichloro-N4-(2- (isopropylsulfonyl)phenyl)pyrimidine-2,4-diamine (100 mg, 0.28 mmol), l-(4-amino-3- methoxyphenyl)piperidin-4-ol (123 mg, 0.56 mmol), 4N HC1 dioxane (0.14 mL) and 2-butanol (1.0 mL). The tube was sealed and the reaction mixture was stirred at 120 0C for 24 hours. The reaction mixture was then partitioned between ethyl acetate and water. The organic layer was separated and the aqueous layer was neutralized with satd. ΝaHCθ3 solution and extracted with ethyl acetate. The combined organic extracts were washed with brine, dried over MgSO4, filtered and concentrated. The residue was dissolved in DMSO (3mL). The resulting solution was subjected to purification by reverse-phase HPLC to yield the title compound as a TFA salt. The product containing fraction was neutralized with satd. NaHCθ3 solution and extracted with ethyl acetate dried over MgSO4, filtered and concentrated. 1H NMR 600 MHz (CDCl3) δ 9.19 (s, IH), 8.47 (d, J = 8.4 Hz, IH), 8.00 (d, J = 3.0 Hz, IH), 7.81 (dd, J = 1.2 Hz, J = 7.8 Hz, IH), 7.52 (dt, J = 1.8 Hz, J = 8.4 Hz, IH), 7.13 (dt, J = 1.2 Hz, J = 7.8 Hz, IH), 7.01 (s, IH), 6.48 (d, J = 2.4 Hz, IH), 6.41 (dd, J = 2.4 Hz, J = 8.4 Hz, IH), 4.90 (s, 2H), 3.79 (m, 4H), 3.41 (m, 2H), 3.20 (m, IH), 2.84 (m, 2H), 1.97 (m, 2H), 1.69(m, 2H), 1.24 (d, J = 6.6 Hz, 6H), MS m/z : 547.50 (M + 1)
[0118] By repeating the procedures described in the above examples, using appropriate amines, the following compounds in Table 2 are prepared.
Figure imgf000053_0001
J =
7 =
(m, (m, (d, J 1).
IH), = 1.8 7 7.8 (m, (d, J + 1).
J =
IH), 6.45 (s, (m, (m,
Figure imgf000054_0001
Figure imgf000055_0001
Figure imgf000056_0001
Figure imgf000057_0001
Figure imgf000058_0001
Figure imgf000059_0001
J = (s, (d, J 2H), 4H), MS
Figure imgf000060_0001
[0119] Alternatively, the compounds of Formula I, particularly Formula IA can be prepared through the preparation shown in the Reaction Scheme 3 below.
Figure imgf000061_0001
Scheme 3
Example 3-0
4-(4-amino-5-chloro-6-(2-(isopropylsulfonyl)phenylamino)pyrimidin-2-ylamino)-3-methoxy-N-
(4-(4-methylpiperazin- 1 -vDphenyDbenzamide
Figure imgf000061_0002
[0120] To a stirred solution of the isomers of methyl 4-(4-amino-5-chloro-6-(2- (isopropylsulfonyl)phenylamino)pyrimidin-2-ylamino)-3-methoxybenzoate and methyl 4-(2- amino-5-chloro-6-(2-(isopropylsulfonyl)phenylamino)pyrimidin-4-ylamino)benzoate (779 mg, 1.55 mmol, prepared by the above procedure) in THF/MeOH (1:1, v/v, 50 mL) was added 10 mL of LiOH ( 350 mg, 8.33 mmol) aqueous solution. The resulting mixture was stirred at room temperature until the reaction was completed. Then the reaction was diluted with water and extracted with ethyl acetate. The water layer was acidified with 0.5 N HCl solution until PH = 5 and the solid was precipitated. The solid was collected by filtration and dried by air to give the isomers of 4-(2-amino-5-chloro-6-(2-(isopropylsulfonyl)phenylamino)pyrimidin-4- ylamino)benzoic acid and 4-(4-amino-5-chloro-6-(2-(isopropylsulfonyl)phenylamino)pyrimidin- 2-ylamino)-3-methoxybenzoic acid (210 mg).
[0121] To a solution of the isomers of 4-(2-amino-5-chloro-6-(2-
(isopropylsulfonyl)phenylamino)pyrimidin-4-ylamino)benzoic acid and 4-(4-amino-5-chloro-6- (2-(isopropylsulfonyl)phenylamino)pyrimidin-2-ylamino)-3-methoxybenzoic acid (15 mg, 0.03 mmol), 4-(4-methylpiperazin-l-yl)benzenamine (19 mg, 0.1 mmol) and DIEA (27 μL, 0.15 mmol) in 1.0 mL of DMSO was added HATU (23 mg, 0.06 mmol). The mixture was stirred at room temperature until the reaction was completed determined by LC-MS. The resulting solution was subjected to purification by reverse-phase HPLC to yield the title compound as a TFA salt (17 mg).
[0122] By repeating the procedures described in the above examples, using appropriate amines, the following compounds in Table 3 are prepared.
Table 3
Figure imgf000062_0001
Figure imgf000063_0001
Figure imgf000064_0001
(dd, Hz, Hz, (m,
Figure imgf000065_0001
Figure imgf000066_0001
[0123] Alternatively, the compounds of Formula I may be prepared through the preparation of intermediates shown in the Reaction Scheme 4 below aq.)
Figure imgf000067_0001
0C _ RT
Figure imgf000067_0002
Figure imgf000067_0003
Scheme 4
Example 4-1 2-isopropoxy-5-methyl-4-(l-methylpiperidin-4-yl)benzenamine
Figure imgf000067_0004
5-chloro-4-methyl-2-nitrophenol
Figure imgf000067_0005
[0124] To a stirred solution of 3-chloro-4-methylphenol (4.9 g, 34 mmol) in 35 mL of glacial acetic acid was added 90% aqueous nitric acid (1.7 mL) in 20 min at 8 ~ 10 C. The resulting mixture was stirred at 10 - 15 C for 5 h. Then the mixture poured over ice water and the solid precipitated. The solid was collected by filtration and washed with water. Further purification by flash chromatography gave the title compound as bright yellow solid (2.67 g, 42%). 1H NMR (600 M, CDCl3) δ 10.42 (s, IH), 7.97 (s, IH), 7.19 (s, IH), 2.35 (s, 3H). l-chloro-5-isopropoxy-2-methyl-4-nitrobenzene
Figure imgf000068_0001
[0125] To a stirred solution of 5-chloro-4-methyl-2-nitrophenol (1.6 g, 8.5 mmol) in 30 mL of acetonitrile was added K2CO3 (2.37 g, 17 mmol) at room temperature. After 15 min, 2- iodopropane (1.7 mL, 17 mmol) was added and the resulting mixture was stirred at 85 C for 20 h. Then the reaction was cooled down to room temperature and filtered. The filtrate was diluted with ethyl acetate and washed with water and brine. The organic layer was dried over anhydrous Na2SO4 and concentrated to give title compound (1.02 g, 52%), which was used without further purification. 1H NMR (600 M, CDCl3) δ 7.69 (s, IH), 7.06 (s, IH), 4.62-4.57 (m, IH), 2.33 (s, 3H), 1.38 (d, J = 6.0 Hz, 6H).
4-(5-isopropoxy-2-methyl-4-nitrophenyl)pyridine
Figure imgf000068_0002
[0126] A mixture of l-chloro-5-isopropoxy-2-methyl-4-nitrobenzene (460 mg, 2.0 mmol), pyridin-4-ylboronic acid (300 mg, 2.4 mmol) and Pd(PPh3)2Cl2 (140 mg, 0.2 mmol) in dioxane/1.0 N Na2CO3 aqueous solution (1:1, v/v, 20 mL) was heated at 85 C under an argon atmosphere. After 1 h, the solution was cooled to room temperature and filtered through a pad of celite and eluted with ethyl acetate. The combined filtrate was washed with brine and dried over anhydrous Na2SO4. The resulting crude product was concentrated and purified by flash chromatography to yield the title compound (458 mg, 84%). 1H NMR (600 M, CDCl3) δ 8.70 (dd, J = 1.8, 4.8 Hz, 2H), 7.70 (s, IH), 7.22 (dd, J = 1.8, 4.8 Hz, 2H), 6.89 (s, IH), 4.65-4.59 (m, IH), 2.20 (s, 3H), 1.38 (d, J = 6.0 Hz, 6H). 2-isopropoxy-5-methyl-4-(l-methylpiperidin-4-yl)benzenamine
Figure imgf000069_0001
[0127] A sealed tube was charged with 4-(5-isopropoxy-2-methyl-4-nitrophenyl)pyridine (445 mg, 1.64 mmol), iodomethane (0.2 mL, 3.28 mmol) and acetonitrile (10 mL). The reaction mixture was stirred at 85 C for 12 h, then it was cooled down to room temperature. The reaction was concentrated and the residue was used in next step without further purification.
[0128] The residue was dissolved in 20 mL of methanol. After hydrogenation on platinum oxide, one atmosphere of hydrogen for 20 h, the reaction mixture was filtered through a pad of celite and eluted with methanol. The combined filtrate was concentrated to afford the title compound, which was used without further purification (430 mg, 99%). 1H NMR (600 M, CDCl3) δ 6.75 (s, IH), 6.50 (s, IH), 4.55-4.49 (m, IH), 3.68 (s, br, 2H), 3.53 (d, J = 12.0 Hz, 2H), 2.94-2.85 (m, 3H), 2.74 (s, 3H), 2.47-2.41 (m, 2H), 2.16 (s, 3H), 1.90 (d, J = 14.4 Hz, 2H), 1.31 (d, J = 6.0 Hz, 6H).
[0129] The compounds of Formula II, can be prepared through the preparation shown in the Reaction Scheme 5 below.
α> mCPBA
EA, 0 0C
Figure imgf000070_0001
0C
Figure imgf000070_0002
Scheme 5 Example 5-0
(4-(4-methylpiperazin-l-yl)piperidin-l-yl)(6-(4-(2-(methylsulfonyl)phenylamino)-lH- pyrrolor2,3-blpyridin-6-ylamino)pyridin-3-yl)methanone
Figure imgf000071_0001
Figure imgf000071_0002
[0130] To a solution of lH-pyrrolo[2,3-£]pyridine(20 g, 169.4 mmol) in ethyl acetate (250 mL) was added a solution of m-chloroperbenzoic acid (40.8 g, 237.0 mmol) in ethyl acetate(40 mL) at 0 0C for 30 min, followed by the addition of an additional 30 mL of ethyl acetate. After stirring at room temperature overnight, the reaction mixture was then cooled down to 0 0C. The resulting white solid was collected, washed with ethyl acetate, and dried. This white solid was suspended in water (100 mL) and treated with sat. K2CO3 slowly until a substantial amount of white solid was precipitated. This slurry mixture was slowly stirred overnight, cooled down to 0 0C. The resulting white solid was collected, washed with water and then dried to give 12.3 g (54% yield) of the title product.
4-chloro-lH-pyrrolor2,3-^lpyridine
Figure imgf000071_0003
[0131] To a solution of lH-pyrrolo[2,3-£]pyridine 7-oxide (12.3 g, 91.7 mmol) in NN- dimethylformamide (40 mL) was added methanesulfonyl chloride (10.8 mL, 137.6 mmol) at 50 0C. The orange reaction mixture was stirred at 70 0C for 2 h and cooled down to 40 0C and then treated with 40 mL of water. The resulting suspension was cooled to 0 0C and treated with 10.0 N ΝaOΗ solution to reach a pΗ of approximately 7. The resulting solid was collected, washed with water and dried to give 11.9 g (78% yield) of the title product.
[0132] 1H ΝMR 600 MHz (DMSO-J6) δ 12.01 (s, IH), 8.15(J, J = 5.4 Hz, IH), 7.56 (dd, J = 2.4 Hz, J = 3.0 Hz, IH), 7.16 (J, J = 4.8 Hz, IH), 6.48 (dd, J = 2.4 Hz, J = 3.6 Hz, 1 H), MS m/z 153.05 (M + 1).
4-chloro-lH-pyrrolor2,3-blpyridine 7-oxide
Figure imgf000072_0001
[0133] To a solution of 4-chloro-lH-pyrrolo[2,3-&]pyridine (8.0 g, 52.6 mmol) in ethyl acetate (80 mL) was added a solution of m-chloroperbenzoic acid (12.6 g, 52.63 mmol) in ethyl acetate(13 mL) at 0 0C for 30 min, followed by the addition of an additional 10 mL of ethyl acetate. After stirring at room temperature overnight, the reaction mixture was then cooled down to 0 0C. The resulting white solid was collected, washed with ethyl acetate, and dried. This white solid was suspended in water (35 mL) and treated with sat. K2CO3 slowly until a substantial amount of white solid precipitated. This slurry mixture was slowly stirred overnight and then cooled down to 0 0C. The resulting white solid was collected, washed with water and then dried to give 5.2 g (58% yield) of the title product.
4,6-dichloro-lH-pyrrolor2,3-^lpyridine
Figure imgf000072_0002
[0134] To a solution of 4-chloro-lH-pyrrolo[2,3-&]pyridine 7-oxide (5.2 g, 30.9 mmol) in NN-dimethylformamide (13.5 mL) was added methanesulfonyl chloride (3.42 mL, 43.3 mmol) at 50 0C. The orange reaction mixture was stirred at 80 0C for 3 h and cooled down to 40 0C and then treated with 13.5 mL of water. The resulting suspension was cooled to 0 0C and treated with 10 N ΝaOH solution to reach a pH of approximately 7. The resulting solid was collected, washed with water and dried to give 4.8 g (84% yield) of the title product.
[0135] 1H ΝMR 600 MHz (DMSO-J6) δ 12.24 (5, IH), 7.61(J, J = 3.6 Hz, IH), 7.32 (5, IH), 6.53 (d, J = 3.6 Hz, 1 H), MS m/z 187.01 (M + 1).
4,6-dichloro-l-((2-(trimethylsilyl)ethoxy)methyl)-lH-pyrrolor2,3-^lpyridine
Figure imgf000073_0001
[0136] To a solution of 4,6-dichloro-lH-pyrrolo[2,3-£]pyridine (1.50 g, 8.0 mmol) in N,N- dimethylformamide (26 mL) was added sodium hydride (481 mg, 12.0 mmol) at 0 0C. The reaction mixture was allowed to stir for 20 min. after which time 2-(trimethylsilyl)ethoxymethyl chloride (1.7 mL, 9.6 mmol) was added. The reaction mixture was further stirred at room temperature for a period of 3 hours after which time water and then ethyl acetate were added. The organic layer was separated and the aqueous layer was extracted with ethyl acetate. The combined organic extracts were washed with brine, dried over MgSO4, filtered and concentrated. The crude product was purified by flash column chromatography using a 9:1 v/v Ηexane:Ethyl acetate as solvent to afford title compound (2.1 g, 82% yield) as a yellow oil. 1H ΝMR 600 MHz (CDCl3) δ 7.41(J, J = 4.2 Hz, IH), 7.21 (5, IH), 6.66 (d, J = 4.2 Hz, 1 H), 5.67 (5, 2H), 3.58 (t, 2H), 0.96 (t, 2H), 0.00(5, 9H).
6-chloro-Ν-(2-(methylsulfonyl)phenyl)-l-((2-(trimethylsilyl)ethoxy)methyl)-lH- pyrrolor2,3-blpyridin-4-amine
Figure imgf000073_0002
[0137] To a solution of 4,6-dichloro-l-((2-(trimethylsilyl)ethoxy)methyl)-lH-pyrrolo[2,3- &]pyridine (1.13 g, 3.57 mmol) in t-BuOH (18 mL) was added 2-(methylsulfonyl)benzenamine hydrochloride (742 mg, 3.57 mmol) and K2CO3 (1.97 g, 14.28 mmol). The reaction mixture was degassed using Argon for 10 min and then Pd2(dba)2 (196 mg, 0.214 mmol) and 2- dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (153 mg, 0.32 mmol) were added. The reaction flask was placed into the preheated oil-bath at 100 0C. The reaction mixture was further stirred at 100 0C for a period of 4 hours after which, it was filtered and partitioned between ethyl acetate and water. The organic layer was separated and the aqueous layer was extracted with ethyl acetate. The combined organic extracts were washed with brine, dried over MgSO4, filtered and concentrated. The crude product was purified by flash column chromatography using a 9:1 v/v Hexane:Ethyl acetate as solvent to afford title compound (850 mg, 53% yield) as a yellow solid. 1H NMR 600 MHz (CDCl3) δ 8.38 (s, IH), 8.07 (d, J = 7.8 Hz, IH), 7.73 (d, J = 8.4 Hz, IH), 7.70 (t, IH), 7.32 (m, 2H), 7.00 (s, IH), 6.52 (d, J = 3.0 Hz, IH), 5.67 (5, 2H), 3.65 (m, 2H), 3.12 (5, 3H), 1.08 (m, 2H), 0.02 (5, 9H), MS m/z : 451.98 (M + 1).
Methyl 6-(4-(2-(methylsulfonyl)phenylamino)-l-((2-(trimethylsilyl)ethoxy)methyl)- IH- pyrrolor2,3-blpyridin-6-ylamino)nicotinate
Figure imgf000074_0001
[0138] To a solution of 6-chloro-N-(2-(methylsulfonyl)phenyl)-l-((2- (trimethylsilyl)ethoxy)methyl)-lH-pyrrolo[2,3-b]pyridin-4-amine (215 mg, 0.48 mmol) in t- BuOH (2 mL) was added methyl 6-aminonicotinate (80 mg, 0.52 mmol) and K2CO3 (217 mg, 1.57 mmol). The reaction mixture was degassed using Argon for 10 min after which Pd2(dba)2 (26 mg, 0.03 mmol) and 2-dicyclohexylphosphino-2',4',6'-triisopropylbiphenyl (22 mg, 0.05 mmol) were added. The reaction flask was put into the preheated oil-bath at 100 0C. The reaction mixture was further stirred at 100 0C for a period of 6 hours after which, it was filtered and partitioned between ethyl acetate and water. The organic layer was separated and the aqueous layer was extracted with ethyl acetate. The combined organic extracts were washed with brine, dried over MgSO4, filtered and concentrated. The crude product was purified by flash column chromatography using a 9:1 v/v Hexane:Ethyl acetate as solvent to afford title compound (185 mg, 69% yield). 1H NMR 600 MHz (CDCl3) δ 8.92 (d, J = 3.6 Hz, IH), 3.33 (5, IH), 8.27 (dd, J = 2.4 Hz, J = 9.0 Hz, IH), 8.09 (d, J = 9.0 Hz, IH), 8.06 {dd, J = 1.8 Hz, J = 7.8 Hz, IH), 7.86 (5, IH), 7.77 (d, J = 8.4 Hz, IH), 7.66 (dt, J = 1.2 Hz, J = 8.4 Hz, IH), 7.26 (dt, J = 0.6 Hz, J = 7.8 Hz, IH), 7.18 (J, J = 3.6 Hz, IH), 7.11 (5, IH), 6.46 (d, J = 4.2 Hz, IH), 5.67 (5, 2H), 3.97 (5, 3H), 3.69 (m, 2H), 3.35 (5, 3H), 1.02 (m, 2H), 0.00 (5, 9H), MS m/z : 568.13 (M + 1).
6-(4-(2-(methylsulfonyl)phenylamino)-lH-pyrrolor2,3-blpyridin-6-ylamino)nicotinic acid
Figure imgf000075_0001
[0139] To a solution of methyl 6-(4-(2-(methylsulfonyl)phenylamino)-l-((2-
(trimethylsilyl)ethoxy)methyl) -lH-pyrrolo[2,3-b]pyridin-6-ylamino)nicotinate (185 mg, 0.33 mmol) in methylene chloride (1.5 mL) was added trifluoroacetic Acid (0.13 mL, 1.63 mmol). The reaction mixture was stirred for 5h after which, the solvent was removed in vacuo. The crude product was redissolved in a mixture of methanol (0.5 mL) and THF (0.5 mL) followed by the addition of lithium monohydroxide (68 mg, 1.63 mmol) in water (0.5 mL). The reaction mixture was stirred for 6h after which, it was neutralized with IN HCl solution and partitioned between ethyl acetate and water. The organic layer was separated and the aqueous layer was extracted with ethyl acetate. The combined organic extracts were washed with brine, dried over MgSO4, filtered and concentrated. The crude product was used without further purification.
(4-(4-methylpiperazin-l-yl)piperidin-l-yl)(6-(4-(2-(methylsulfonyl)phenylamino)-lH- pyrrolor2,3-blpyridin-6-ylamino)pyridin-3-yl)methanone
Figure imgf000076_0001
[0140] To a solution of 6-(4-(2-(methylsulfonyl)phenylamino)- lH-pyrrolo[2,3- b]pyridin-6-ylamino)nicotinic acid (50 mg, 0.12 mmol) in N,N-dimethylformamide (0.6 mL) was added HATU (67 mg, 0.18 mmol), DIEA (60 uL, 0.3 mmol) and l-methyl-4-(piperidin-4- yl)piperazine (49 mg, 0.18 mmol). The reaction mixture was further stirred for a period of 6 hours after which, it was partitioned between ethyl acetate and water. The organic layer was separated and the aqueous layer was extracted with ethyl acetate. The combined organic extracts were washed with brine, dried over MgSO4, filtered and concentrated. The crude product was purified by reverse-phase HPLC to yield the title compound as a TFA salt (63 mg, 78% yield). 1H ΝMR 600 MHz (DMSO-J6) δ 12.09 (s, IH), 11.19 (s, IH), 9.61 (s, IH), 8.40 (s, IH), 8.10 (d, J = 7.8 Hz, IH), 7.90 (m, 2H), 7.69 (m, 2H), 7.22 (s, IH), 7.15 (m, IH), 6.54 (s, IH), 5.98 (s, IH), 3.42 (m, 2H), 3.23 (m, 5H), 3.10 (m, 2H), 2.81 (s, 3H), 2.52 (m, 2H), 2.48 (m, 4H), 1.95 (m, 2H), 1.54 (m, 2H), MS m/z : 589.48 (M + 1).
Example 5-20
Ν6-(2-isopropoxy-5-methyl-4-(l-methylpiperidin-4-yl)phenyl)-Ν4-(2- (isopropylsulfonyl)phenyl)-lH-pyrrolor2,3-blpyridine-4,6-diamine
Figure imgf000077_0001
Figure imgf000077_0002
Scheme 6
4-chloro-lH-pyrrolor2,3-blpyridine 7-oxide (2)
[0141] To a solution of 3.13 g of 4-chloro-lH-pyrrolo[2,3-b]pyridine (1) (0.02 mol) in 300 mL of Et20 was slowly added 6.72 g of 77% m-chloroperoxybenzoic acid. The resulting mixture was stirred at room temperature for 2 h and the precipitate which formed during this time was isolated by filtration, washed with cold Et2O to give the title compound as yellow solid which was used directly for the next step without further purification.
Methyl 4,6-dichloro-lH-pyrrolor2,3-blpyridine-l-carboxylate (3) [0142] To a solution of 504 mg of 4-chloro-lH-pyrrolo[2,3-b]pyridine 7-oxide (2) (3.0 mmol) and 577 uL of HMDS (7.5 mmol) in 30 mL of THF was drop- wise added 626 uL of methyl carbonochloridate (3.0 mmol) under N2 at r.t. After stirring for 2 hours the reaction mixture was partitioned between 100 mL of EtOAc and 100 mL of saturated aqueous NaHCθ3 solution. The organic layer was separated, washed with brine, concentrated and purified by column chromatography (silica gel with 0 to 25% EtOAc in Hexanes) to give the titled compound.
4-chloro-N-(2-isopropoxy-5-methyl-4-(l-methylpiperidin-4-yl)phenyl)-lH-pyrrolor2,3- blpyridin-6-amine and Methyl 4-chloro-6-(2-isopropoxy-5-methyl-4-(l-methylpiperidin-4- yl)phenylamino)-lH-pyrrolor2,3-blpyridine-l-carboxylate (4 and 5)
[0143] To a solution of 53.7 mg of methyl 4,6-dichloro-lH-pyrrolo[2,3-b]pyridine-l- carboxylate (3) (0.22 mmol) and 52.4 mg of 2-isopropoxy-5-methyl-4-(l-methylpiperidin-4- yl)aniline (0.2 mmol) in 4 mL of THF was added 8.8 mg Pd2(OAc)2 (0.01 mmol), 28.8 mg 4,5- Bis(diphenylphosphino)9,9-dimethylxanthene (0.02 mmol) and 130.4 mg Cs2CO3 (0.4 mmol). The resulting mixture was heated under microwave irradiation at 150 0C for 40 minutes. After cooled down to r.t, the mixture was filtered through celite, and the filtrate was concentrated and purified by column chromatography (silica gel with 0 to 10% MeOH in DCM) to give an inseparable mixture of titled compounds 4 and 5.
N6-(2-isopropoxy-5-methyl-4-(l-methylpiperidin-4-yl)phenyl)-N4-(2- (isopropylsulfonvDphenyl)- lH-pyrrolor2,3-blpyridine-4,6-diamine and N4-(2-isopropoxy-5- methyl-4-(l-methylpiperidin-4-yl)phenyl)-N6-(2-(isopropylsulfonyl)phenyl)-lH-pyrrolor2,3- blpyridine-4,6-diamine (6 and 7)
[0144] To a solution of 25mg of the mixture of 4-chloro-N-(2-isopropoxy-5-methyl-4-(l- methylpiperidin-4-yl)phenyl)-lH-pyrrolo[2,3-b]pyridin-6-amine and methyl 4-chloro-6-(2- isopropoxy-5-methyl-4-(l-methylpiperidin-4-yl)phenylamino)-lH-pyrrolo[2,3-b]pyridine-l- carboxylate (4 and 5) (0.053 mmol) and 12.7 mg of 2-(isopropylsulfonyl)aniline (0.061 mmol) in 1 mL of THF was added 2.4 mg Pd2(dba)3 (0.0027 mmol), 2.6 mg dicyclohexyl(2,4,6- triisopropylphenyl)phosphine (0.0053 mmol) and 7.7 mg NaOBu-t (0.08 mmol). The resulting mixture was heated under microwave irradiation at 150 0C for 40 minutes. After cooled down to r.t., 0.2 mL of NaOMe solution (0.5 M in MeOH) was added and the mixture was heated at 50 0C for 30 minutes and then filtered through celite. The filtrate was concentrated and purified by mass-triggered HPLC to give the titled compounds 6 and 7, respectively.
[0145] The following compounds were synthesized by following the procedures above with the appropriate anilines. Table 3 (m/z)
Figure imgf000079_0001
Figure imgf000080_0001
(m/z)
Figure imgf000081_0001
(m/z)
Figure imgf000082_0001
(m/z)
Figure imgf000083_0001
(m/z)
Figure imgf000084_0001
Figure imgf000085_0001
[0146] Similarly, the compounds of Formula III, can be prepared through the preparation shown in the Reaction Scheme 6 below. Scheme 6
Figure imgf000086_0001
Aniline (3.0 eq), 4N HCI Dioxane (3.0 eq) ethylene glycol, 180 0C, 30mιn, microwave
Figure imgf000086_0002
Example 6-0
N6-(2-isopropoxy-5-methyl-4-(l-methylpiperidin-4-yl)phenyl)-N4-(2- (isopropylsulfonyl)phenyl)-lH-pyrazolor3,4-dlpyrimidine-4,6-diamine
Figure imgf000086_0003
isopropyl(2-nitrophenyl)sulfane
Figure imgf000086_0004
[0147] To a solution of l-fluoro-2-nitrobenzene (3.0 g, 21.2 mmol) in DMF (60 mL) was added 2-propane thiol (2.2 mL, 23.4 mmol) and K2CO3 (6.8g, 42.4 mmol). The reaction was heated to 45 0C for 16 hours. The mixture was then diluted with 30OmL of ethyl acetate, filtrated, washed five times with 6OmL of water and brine. The organic phase was dried over sodium sulfate, filtered, concentrated. The resulting yellow solid (4.0 g, 95% yield) was used without further purification.
(Isopropylthio)benzenamine
Figure imgf000087_0001
[0148] To a solution of isopropyl (2-nitrophenyl)sulfane( 4.0 g, 20.3 mmol) in methanol (200 mL) was added Pd/C (0.40 g). The 50OmL of flask was evacuated and charged with high purity hydrogen gas. The reaction was stirred for 20 hours. The mixture was filtrated through celite. After concentration, the residue was purified by flash chromatography using 10:1 methylene chloride -ethyl acetate as solvent to afford title compound (3.36 g, 20.1 mmol) as brown oil.
4,6-dichloro- 1 H-pyrazolo [3 ,4-dlpyrimidine
Figure imgf000087_0002
[0149] To a 10OmL flask was added 4,6-dihydroxylpyrazolo(3,4-d)pyrimidine (2.0 g, 13.15 mmol), phosphorus oxychloride (12 mL, 131.5 mmol) and N, N-diethylaniline ( 4.OmL, 26.3 mmol). The reaction mixture was heated to 110 0C for 2 hours. After removal of phosphorus oxychloride, the dark residue was poured onto crushed ice water. The cold aqueous solution was extracted with diethyl ether (3 x 10OmL). The organic layer was washed with water and brine, dried over sodium sulfate, concentrated to afford 1.30 g of a crude tan product, which was used without further purification.
4,6-dichloro-l-(tetrahydro-2H-pyran-2-yl)-lH-pyrazolor3,4-dlpyrimidine
Figure imgf000088_0001
[0150] To a solution of 4,6-dichloro-lH-pyrazolo[3,4-d]pyrimidine (1.30 g, 6.88 mmol) in methylene chloride ( 20 niL) and THF (20 niL) was added TsOH ( 0.13 g, 0.688 mmol) and 3,4 -dihydro-2H-pyran (0.93 mL, 10.32 mmol). The solution was stirred for 4 hours. After removal of solvent, the residue was dissolved in methylene chloride (100 mL), washed with twice with a satd. aqueous solution of Na2CC^, water and brine. The methylene chloride solution was dried over sodium sulfate and concentrated. The resulting yellow oil was treated with 1 mL of ethyl acetate and 10 mL of hexane to induce solidification. The resulting 1.20 grams of white solid was used without further purification.
6-chloro-N-(2-(isopropylthio)phenyl)-l-(tetrahvdro-2H-pyran-2-yl)-lH-pyrazolor3,4- dlpyrimidin-4-amine
Figure imgf000088_0002
[0151] To a 25 mL of flask charged with 4,6-dichloro-l-(tetrahydro-2H-pyran-2-yl)-lH- pyrazolo[3,4-d]pyrimidine (0.955 g, 3.50 mmol) and (isopropylthio)benzenamine (1.168 g,7.0 mmol) in n-butanol (3 mL) was added DIEA (0.69 mL, 4.2 mmol). The reaction mixture was heated to 110 0C for 2 hours. After removal of solvent, the residue was purified by flash chromatography using 3: 1 hexane-ethyl acetate as solvent to afford title compound (1.20 g, 2.97 mmol) as white solid. 6-chloro-N-(2-(isopropylsulfonyl)phenyl)-l-(tetrahydro-2H-pyran-2-yl)-lH- pyrazolor3,4-dlpyrimidin-4-amine
Figure imgf000089_0001
[0152] To a solution of 6-chloro-N-(2-(isopropylthio)phenyl)-l-(tetrahydro-2H-pyran-2-yl)- lH-pyrazolo[3,4-d]pyrimidin-4-amine (1.20 g, 2.97 mmol) in dichloromethane (80 mL) was added mCPBA (2.00 g, 8.92 mmol). The reaction mixture was stirred for 4 hours after which, it was diluted with dichloromethane (100 mL). The organic layer was washed with satd. NaHCθ3/Na2S2θ3, satd. NaHCO3 solution and washed with brine. The organic layer was dried over Na2SO4, filtered and concentrated. The resulting light yellow solid (1.20 g, 2.75 mmol) was used without further purification.
N6-(2-isopropoxy-5-methyl-4-(l-methylpiperidin-4-yl)phenyl)-N4-(2- (isopropylsulfonyl)phenyl)-lH-pyrazolor3,4-dlpyrimidine-4,6-diamine
Figure imgf000089_0002
[0153] A microwave tube was charged with 6-chloro-N-(2-(isopropylsulfonyl)pheny I)-I- (tetrahydro-2H-pyran-2-yl)-lH-pyrazolo[3,4-d]pyrimidin-4-amine (40 mg, 0.0917 mmol), 2- isopropoxy-5-methyl-4-(l-methylpiperidin-4-yl)benzenamine (72.3 mg, 0.275 mmol) 4N HCl in Dioxane (0.069 mL, 0.276 mmol) and ethylene glycol (1.5 mL). The tube was sealed and the reaction mixture was stirred at 180 0C for 30 min in a microwave reactor. The reaction mixture was purified by reverse-phase preparative HPLC. The crude TFA salt of product was neutralized with a satd. aqueous solution of NaHCθ3 and extracted with ethyl acetate. The organic layer was dried over sodium sulfate, filtered and concentrated. The residue was purified by flash chromatography using 30:1:0.3 methylene chloride-methanol-triethylamine as solvent to afford the title compound (20.0 mg, 0.037 mmol). 1H NMR 600 MHz (CD3OD) δ 8.46 (d, J = 8.4 Hz, IH), 8.07 (s, IH), 7.92 (d, J = 7.8 Hz, IH), 7.84 (s, IH), 7.70 (t, J = 8.4 Hz, IH), 7.38 (m, IH), 6.78 (s, IH), 4.58 (m,lH), 3.49 (d, J = 12 Hz, 2H), 3.30 (m, IH), 3.00 (m, 3H), 2.80 (s,3H), 2.21 (s, 3H), 1.94 (m, 4H), 1.35 (d, J = 6.6 Hz, 6H), 1.20 (d, J = 6.6 Hz, 6H). MS m/z : 536.60 (M +
1).
[0154] By repeating the procedures described in the above examples, using appropriate amines, the following compounds in Table 4 are prepared.
Table 4
Figure imgf000090_0001
Figure imgf000091_0001
(m/z)
Figure imgf000092_0001
(m/z)
Figure imgf000093_0001
(m/z)
Figure imgf000094_0001
(m/z)
Figure imgf000095_0001
Figure imgf000096_0001
Biological Examples
Inhibition of ALK activity
[0155] Cell Lines. Murine pro-B cell line Ba/F3, the human t(2,5)-positive Karpas-299 and TEL-ALK transformed Ba/F3 are maintained in RPMI medium 1640 supplemented with 10% FBS (Sigma-Aldrich, St. Louis, MO). Ba/F3 cells are grown in the presence of 10% of WEHI media. Cell lines expressing luciferase alone or in combination with TEL-kinase fusion constructs are generated by retroviral transduction of cells with pMSCV-IRES puro/Luc vector.
[0156] Cell Proliferation Assays. Luciferase-expressing Ba/F3 cells, Karpas-299, Tel_ALK transformed Ba/F3 stably expressing NPM-ALK and TEL-ALK, are plated in 384-well plates (5,000 cells per well) and incubated with serial dilutions of ALK inhibitors or DMSO for 48 hours. Luciferase expression is used as a measure of cell proliferation/ survival and was evaluated with the Bright-Glo Luciferase Assay System (Promega, Madison, WI). Fifty percent inhibition values (IC50) are generated by using XLFit software. In order to monitor inhibition of NPM-ALK biochemically, STAT-3 and 5 phosphorylation are monitored using phosphorylation specific antibodies (Cell Signaling). To monitor the ability of the compounds to inhibit EML4- ALK, an activating ALK translocation identified in lung cancer, a proliferation assay using an established cell line (NCI-H3122) containing the fusion kinase is employed.
[0157] In Vivo Experiments. For compound efficacy studies in a murine model, treatment is initiated 72 h after tail vein injection of IxIO6 Ba/F3 NPM-ALK (a murine pre-B cell line engineered to stably express NPM-ALK) or Karpas-299 (a human-derived NPM-ALK cell line), or expressing cells into female Fox Chase SCIDBeige mice. Mice (10 animals per group) are administered either the test compound dissolved in 90% PEG 300 /10% l-methyl-2- pyrrolidinone (Sigma) at 1, 3, and 10 mg/kg once daily for three weeks or the vehicle solution at the same dosing schedule. Disease progression and compound efficacy is monitored weekly with bioluminescence using a xenogen imaging system. To determine the efficacy of the test compound on established disease, dosing is initiated on day 10, at which time the disease confirmed to be widespread by bioluminescence xenogen imaging. For monitoring pharmacodynamics, mice with established lymphomas are administered vehicle solution or test compound (typically 10 mg/kg) for 3 days. At the end of treatment, mice are sacrificed, and lymph nodes are extracted for immunoblotting and histological analysis.
[0158] The inhibition of ALK tyrosine kinase activity may also be measured using known methods, for example using the recombinant kinase domain of the ALK in analogy to the VEGF-R kinase assay described in J. Wood et al. Cancer Res. 60, 2178-2189 (2000).
[0159] The antiproliferative action of the compounds of the invention can also be determined in the human KARPAS-299 lymphoma cell line (described in WG Dirks et al. Int. J. Cancer 100, 49-56 (2002) using the same methodology described above).
[0160] Compounds 1-3, 1-13, 1-14, 2-1, 2-4, 2-7, 2-10, 2-14, 2-15, 3-14, 5-1, 5-7, 5-8, 5-9, 5-10, 5-11, 5-14, 6-1, 6-2, 6-5, 6-6, 6-9, 6-16, 6-18 are representative examples of inhibitors of NPM-ALK dependent cellular proliferation EC50's of 1 uM or less.
Inhibition of NTRK 3 activity
[0161] A cell-based assay consists of using a ETV6-NTRK3 transformed Ba/F3 cell line. This cell line may be used to discover compounds that are differentially cytotoxic as compared to parental Ba/F3 cells grown in the presence of IL-3. Compounds that are selectively cytotoxic to Ba/F3 ETV6-NTRK3 are confirmed using a biochemical NTRK3 kinase assay. Cellular inhibition of NTRK3 is confirmed using phosphospecific antibodies.
* * *
[0162] It is understood that the examples and embodiments described herein are for illustrative purposes only and that various modifications or changes in light thereof will be suggested to persons skilled in the art and are to be included within the spirit and purview of this application and scope of the appended claims. All publications, patents, and patent applications cited herein are hereby incorporated by reference for all purposes. References
WO 2006021457 - Kawahara, Eiji; Miyake, Takahiro; Roesel, Johannes. Preparation of pyrimidine compounds as FAK and/or ALK inhibitors. PCT Int. Appl. (2006), 83 pp.
WO 2006021454 - Imbach, Patricia; Kawahara, Eiji; Konishi, Kazuhide; Matsuura, Naoko; Miyake, Takahiro; Ohmori, Osamu; Roesel, Johannes; Teno, Naoki; Umemura, Ichiro. Preparation of bis(arylamino)pyrimidine derivatives as antitumor agents. PCT Int. Appl. (2006), 118 pp.
WO 2005016894 - Garcia-echeverria, Carlos; Kanazawa, Takanori; Kawahara, Eiji; Masuya, Keiichi; Matsuura, Naoko; Miyake, Takahiro; Ohmori, Osamu; Umemura, Ichiro; Steensma, Ruo; Chopiuk, Greg; Jiang, Jiqing; Wan, Yongqin; Ding, Qiang; Zhang, Qiong; Gray, Nathanael Schiander; Karanewsky, Donald. Preparation of 2,4-pyrimidinediamines useful in the treatment of neoplastic diseases, inflammatory and immune system disorders. PCT Int. Appl. (2005), 285 pp.
WO 2005097765 - Leahy, James William; Lewis, Gary Lee; Nuss, John M.; Ridgway, Brian Hugh; Sangalang, Joan C. Preparation of thiazoles and analogs as anaplastic lymphoma kinase modulators. PCT Int. Appl. (2005), 346 pp.

Claims

1. A compound having the formula:
Figure imgf000099_0001
or a physiologically acceptable salt, solvate, hydrate or stereoisomer thereof, wherein:
A and B are each independently aryl or heteroaryl, each of which may be optionally substituted with one or more R4 groups;
Y is -SO2-, -SO2NH-, -NH-SO2-, -NH-C(O)-, -C(O)-NH-, -O-, Or -NR2-;
X is NH, O or S;
Z is N or CH;
R1 and R2 are independently H, C1-C6 alkyl, halo-(CrC6 alkyl), C3-C7 cycloalkyl, halo- (C3-C7 cycloalkyl), heterocyclyl, heterocyclylCi_6alkyl, aryl, arylC^alkyl, heteroaryl or heteroarylC i _6alkyl ;
Figure imgf000099_0002
Ra is hydrogen and Rb is halogen, or Ra and Rb, taken together with the atoms to which they are bound form a pyrazolo or a pyrrolo ring fused to the pyrimidine ring, said pyrazolo or pyrrolo ring optionally bearing one or two R4 groups;
Each occurrence of R4 is independently halogen, C1-C6 alkyl, ImIo-(CrC6 alkyl), C3-C7 cycloalkyl, ImIo-(C3-C7 cycloalkyl), heterocyclyl, heterocyclylCi-δalkyl, aryl, arylCi_6alkyl, heteroaryl, heteroarylCi_6alkyl, Ci_6alkoxy, Ci_6alkylthio, hydroxyl, nitro, azido, cyano, acyloxy, carboxy, ester, carbamoyl, carboxamide, ureido, amidino, guanidine, sulfonyl, sulphonylamino or aminosulphonyl;
>5 M NH
Each occurrence of R is independently is Ci-C6 alkoxy, ^ — ' , hydroxy,
\— N >— R6 \— N N-(C1-C6 alkyl)
, dialkylamino, or -N-R7;
\— N \ \— -Nt N-R' Each occurrence of R6 is independently is hydroxy, \ — ' , ^ — / , or -
CONH2; and
Each occurrence of R7 is independently is hydrogen, Ci-C6 alkyl, Ci-C6 alkoxy, hydroxyl, or Ci-C6 hydroxyalkyl; with the provisos that: (i) when B is heteroaryl, -X-R2 and -R3 may each independently represent hydrogen; f— N >— R6 \— N O f— V N-R'
(ii) if B is phenyl and Z is N, R is not
Figure imgf000100_0001
alternatively, R3 or R4 if present in B are not nitro, azido, ureido, guanidine or sulphonylamino;
(iii) if B is phenyl, pyridyl or thiazolyl and Z is CH, R3 is
Figure imgf000100_0002
Figure imgf000100_0003
alternatively, R3 or R4 if present in B are independently halogen, Ci-C6 alkyl, 1IaIo-(C1- C6 alkyl), C3-C7 cycloalkyl, halo-(C3-C7 cycloalkyl), heterocyclyl, heterocyclylCi-όalkyl, aryl, arylC]_6alkyl, heteroaryl or heteroarylC]_6alkyl.
2. The compound of claim 1, having the formula (IA) or (III):
Figure imgf000101_0001
wherein RJ is -CO-R3, i—
Figure imgf000101_0002
Figure imgf000101_0003
alternatively, R3 or R4 if present in B are independently halogen, Ci-C6 alkyl, halo-(d- C6 alkyl), C3-C7 cycloalkyl, halo-(C3-C7 cycloalkyl), heterocyclyl, heterocyclylCi-όalkyl, aryl, arylCi_6alkyl, heteroaryl, heteroarylCi_6alkyl, Ci_6alkoxy, Ci_6alkylthio, hydroxyl, cyano, acyloxy, carboxy, ester, carbamoyl, carboxamide, amidino, sulfonyl, or aminosulphonyl; n is an integer from 0-3; m is an integer from 0-4; p is an integer from 0- 1 ; and
Y, X, R1, R2, R5, R7, Ra and Rb are as defined in claim 1.
3. The compound of claim 1, having the formula (II):
Figure imgf000102_0001
t— N N-R6 \— N O t — / I N-R' I— N N-R' wherein R3 is
Figure imgf000102_0002
alternatively, R3 or R4 if present in B are independently halogen, Ci-C6 alkyl, 1IaIo-(C1- Ce alkyl), C3-C7 cycloalkyl, halo-(C3-C7 cycloalkyl), heterocyclyl, heterocyclylCi_6alkyl, aryl, arylCi_6alkyl, heteroaryl or heteroarylCi_6alkyl; n is an integer from 0-3; m is an integer from 0-4; p is an integer from 0-2; and
Y, X, R1, R2, R5, R7 are as defined in claim 1.
4. The compound of any one of claims 1-3, wherein X is O.
5. The compound of any one of claims 1-4, wherein R2 is methyl, ethyl, propyl, isopropyl, cyclopropyl or cyclobutyl.
6. The compound of claim 5, wherein R2 is methyl.
7. The compound of any one of claims 1-6, wherein Y is SO2.
8. The compound of claims 1-7, wherein R1 is methyl, ethyl, propyl, isopropyl, cyclopropyl or cyclobutyl.
9. The compound of claim 8, wherein R1 is isopropyl.
10. The compound of any one of the above claims, wherein X is O, R is methyl, Y is SO2 and R1 is isopropyl.
11. The compound of claim 1 , wherein Z is N and said compound is selected from the group consisting of: methyl 3-(4-amino-5-chloro-6-(2-(methylsulfonyl)phenylamino)pyrimidin-2-ylamino)-4- methoxybenzoate ;
(3-(4-amino-5-chloro-6-(2-(methylsulfonyl)phenylamino)pyrimidin-2-ylamino)-4- methoxyphenyl)(piperazin-l-yl)methanone;
5-chloro-N2-(2-methoxy-5-(2-(pyrrolidin-l-yl)ethoxy)phenyl)-N4-(2- (methylsulfonyl)phenyl)pyrimidine-2,4,6-triamine; l,4'-bipiperidin-l'-yl(4-(4-amino-5-chloro-6-(2-(methylsulfonyl)phenylamino)pyrimidin- 2-ylamino)-3-isopropoxyphenyl)methanone;
(4-(4-amino-5-chloro-6-(2-(methylsulfonyl)phenylamino)pyrimidin-2-ylamino)-3- isopropoxyphenyl)(4-(4-methylpiperazin-l-yl)piperidin-l-yl)methanone; l,4'-bipiperidin-l'-yl(4-(4-amino-5-chloro-6-(2-(methylsulfonyl)phenylamino)pyrimidin- 2-ylamino)-3-methoxyphenyl)methanone;
5-chloro-N4-(2-(isopropylsulfonyl)phenyl)-N2-(2-methoxy-5-(2-(pyrrolidin-l- yl)ethoxy)phenyl)pyrimidine-2,4,6-triamine; l,4'-bipiperidin-l'-yl(4-(4-amino-5-chloro-6-(2-(isopropylsulfonyl)phenylamino) pyrimidin-2-ylamino)-3-isopropoxyphenyl)methanone;
5-chloro-N2-(2-isopropoxy-5-methyl-4-(l-methylpiperidin-4-yl)phenyl)-N4-(2- (methylsulfonyl)phenyl)pyrimidine-2,4,6-triamine;
4'-(4-amino-5-chloro-6-(2-(methylsulfonyl)phenylamino)pyrimidin-2-ylamino)-5'- methoxy-n,2'-dimethylbiphenyl-4-carboxamide;
5-chloro-N2-(2-isopropoxy-5-methyl-4-(l-methylpiperidin-4-yl)phenyl)-N4-(2- (isopropylsulfonyl)phenyl)pyrimidine-2,4,6-triamine;
5-chloro-N4-(2-(isopropylsulfonyl)phenyl)-N2-(2-methoxy-4-(l-methylpiperidin-3- yl)phenyl)pyrimidine-2,4,6-triamine;
5-chloro-N4-(2-(isopropylsulfonyl)phenyl)-N2-(2-methoxy-4-(l-methylpiperidin-4- yloxy)phenyl)pyrimidine-2,4,6-triamine; 5-chloro-N2-(6-(2,6-dimethylmorpholino)pyridin-3-yl)-N4-(2- (isopropylsulfonyl)phenyl)pyrimidine-2,4,6-triamine; l,4'-bipiperidin-l'-yl(4-(4-amino-5-chloro-6-(2- (isopropylsulfonyl)phenylamino)pyrimidin-2-ylamino)-3-methoxyphenyl)methanone;
5-chloro-N2-(2-isopropoxy-5-methyl-4-(l-methylpiperidin-3-yl)phenyl)-N4-(2- (isopropylsulfonyl)phenyl)pyrimidine-2,4,6-triamine;
5-chloro-N2-(lH-indol-6-yl)-N4-(2-(methylsulfonyl)phenyl)pyrimidine-2,4,6-triamine;
5-chloro-N2-(lH-indazol-6-yl)-N4-(2-(methylsulfonyl)phenyl)pyrimidine-2,4,6- triamine;
4-(4-amino-5-chloro-6-(2-(isopropylsulfonyl)phenylamino)pyrimidin-2-ylamino)-3- methoxy-N-(4-(4-methylpiperazin-l-yl)phenyl)benzamide;
4-{4-Amino-5-chloro-6-[2-(propane-2-sulfonyl)-phenylamino]-pyrimidin-2-ylamino}-3- methoxy-N-(4-morpholin-4-yl-phenyl)-benzamide;
4-{4-Amino-5-chloro-6-[2-(propane-2-sulfonyl)-phenylamino]-pyrimidin-2-ylamino}-3- methoxy-N- [4-(4-methyl-piperazin- 1 -yl)-phenyl]-benzamide;
(4- { 4-Amino-5-chloro-6- [2-(propane-2-sulfonyl)-phenylamino]-pyrimidin-2-ylamino } - 3-methoxy-phenyl)-(4-pyrrolidin-l-yl-piperidin-l-yl)-methanone;
(4- { 4-Amino-5-chloro-6- [2-(propane-2-sulfonyl)-phenylamino]-pyrimidin-2-ylamino } - 3 -methoxy-phenyl)- [4-(4-methyl-piperazin- 1 -yl)-piperidin- 1 -yl] -methanone ;
3-[4-(4-{4-Amino-5-chloro-6-[2-(propane-2-sulfonyl)-phenylamino]-pyrimidin-2- ylamino } -3 -methoxy-benzoyl)-piperazin- 1 -yl] -propionitrile;
(4- { 4- Amino-5-chloro-ό- [2-(propane-2-sulfonyl)-phenylamino]-pyrimidin-2-ylamino } - 3-methoxy-phenyl)-(2-methyl-morpholin-4-yl)-methanone;
(4-{4-Amino-5-chloro-6-[2-(propane-2-sulfonyl)-phenylamino]-pyrimidin-2-ylamino}- 3-methoxy-phenyl)-(3-dimethylamino-pyrrolidin-l-yl)-methanone;
4-{4-Amino-5-chloro-6-[2-(propane-2-sulfonyl)-phenylamino]-pyrimidin-2-ylamino}-N- (2-dimethylamino-ethyl)-3-methoxy-benzamide;
4-{4-Amino-5-chloro-6-[2-(propane-2-sulfonyl)-phenylamino]-pyrimidin-2-ylamino}-N- (4-amino-cyclohexyl)-3-methoxy-benzamide;
N-(4-Acetylamino-cyclohexyl)-4-{4-amino-5-chloro-6-[2-(propane-2-sulfonyl)- phenylamino]-pyrimidin-2-ylamino}-3-methoxy-benzamide; 4-{4-Amino-5-chloro-6-[2-(propane-2-sulfonyl)-phenylamino]-pyrimidin-2-ylamino}-N- (4-dimethylamino-cyclohexyl)-3-methoxy-benzamide;
4-{4-Amino-5-chloro-6-[2-(propane-2-sulfonyl)-phenylamino]-pyrimidin-2-ylamino}-N- (3-amino-cyclohexyl)-3-methoxy-benzamide;
N-(3-Acetylamino-cyclohexyl)-4-{4-amino-5-chloro-6-[2-(propane-2-sulfonyl)- phenylamino] -pyrimidin-2-ylamino } -3 -methoxy-benzamide; and
4-{4-Amino-5-chloro-6-[2-(propane-2-sulfonyl)-phenylamino]-pyrimidin-2-ylamino}-N- (S-dimethylamino-cyclohexy^-S-methoxy-benzamide; or a physiologically acceptable salt, solvate, hydrate or stereoisomer thereof.
12. The compound of claim 1, wherein Z is CH and said compound is selected from the group consisting of: l-(4-(4-(2-(isopropylsulfonyl)phenylamino)-lH-pyrrolo[2,3-b]pyridin-6-ylamino)-3- methoxyphenyl)piperidin-4-ol;
N4-(2-(isopropylsulfonyl)phenyl)-N6-(2-methoxy-4-(4-(4-methylpiperazin-l- yl)piperidin-l-yl)phenyl)-lH-pyrrolo[2,3-b]pyridine-4,6-diamine;
N6-(2-isopropoxy-5-methyl-4-(piperidin-4-yl)phenyl)-N4-(2-(isopropylsulfonyl)phenyl)- lH-pyrrolo[2,3-b]pyridine-4,6-diamine;
N6-(2-isopropoxy-5-methyl-4-(l-methylpiperidin-4-yl)phenyl)-N4-(2- (isopropylsulfonyl)phenyl)-lH-pyrrolo[2,3-b]pyridine-4,6-diamine;
N4-(2-(isopropylsulfonyl)phenyl)-N6-(2-methoxy-4-(4-methylpiperazin-l-yl)phenyl)- lH-pyrrolo[2,3-b]pyridine-4,6-diamine;
2-(4-(4-(4-(2-(isopropylsulfonyl)phenylamino)-lH-pyrrolo[2,3-b]pyridin-6-ylamino)-3- methoxyphenyl)piperazin- 1 -yl)ethanol;
N6-(4-(4-aminopiperidin-l-yl)-2-methoxyphenyl)-N4-(2-(isopropylsulfonyl)phenyl)-lH- pyrrolo[2,3-b]pyridine-4,6-diamine; l-(3-isopropoxy-4-(4-(2-(isopropylsulfonyl)phenylamino)-lH-pyrrolo[2,3-b]pyridin-6- ylamino)phenyl)piperidin-4-ol; l-(3-ethoxy-4-(4-(2-(isopropylsulfonyl)phenylamino)-lH-pyrrolo[2,3-b]pyridin-6- ylamino)phenyl)piperidin-4-ol; l-(4-(4-(2-(isopropylsulfonyl)phenylamino)-lH-pyrrolo[2,3-b]pyridin-6-ylamino)-3- methoxyphenyl)piperidin-3-ol; N4-(2-(isopropylsulfonyl)phenyl)-N6-(2-methoxy-4-morpholinophenyl)-lH-pyrrolo[2,3- b]pyridine-4,6-diamine; and
N6-(2-isopropoxy-5-methyl-4-(l-methylpiperidin-4-yl)phenyl)-N4-(2- (isopropylsulfonyl)phenyl)-lH-pyrrolo[2,3-b]pyridine-4,6-diamine; or a physiologically acceptable salt, solvate, hydrate or stereoisomer thereof.
13. The compound of claim 2, having Formula (III) and selected from the group consisting of:
N4-(2-(isopropylsulfonyl)phenyl)-N6-(2-methoxy-4-(l-methylpiperidin-4- yloxy)phenyl)-lH-pyrazolo[3,4-d]pyrimidine-4,6-diamine;
N4-(2-(isopropylsulfonyl)phenyl)-N6-(2-methoxy-4-(l-methylpiperidin-3-yl)phenyl)- lH-pyrazolo[3,4-d]pyrimidine-4,6-diamine;
N6-(2-isopropoxy-5-methyl-4-(l-methylpiperidin-4-yl)phenyl)-N4-(2- (isopropylsulfonyl)phenyl)-lH-pyrazolo[3,4-d]pyrimidine-4,6-diamine;
N6-(2-isopropoxy-5-methyl-4-(piperidin-4-yl)phenyl)-N4-(2-(isopropylsulfonyl)phenyl)- lH-pyrazolo[3,4-d]pyrimidine-4,6-diamine;
N6-(2-isopropoxy-5-methyl-4-(l-methylpiperidin-3-yl)phenyl)-N4-(2- (isopropylsulfonyl)phenyl)-lH-pyrazolo[3,4-d]pyrimidine-4,6-diamine; l,4'-bipiperidin-l'-yl(4-(4-(2-(isopropylsulfonyl)phenylamino)-lH-pyrazolo[3,4- d]pyrimidin-6-ylamino)-3-methoxyphenyl)methanone;
N6-(2-isopropoxy-5-methyl-4-(piperidin-4-yl)phenyl)-N4-(2-(methylsulfonyl)phenyl)- lH-pyrazolo[3,4-d]pyrimidine-4,6-diamine;
5'-methoxy-N,2'-dimethyl-4'-(4-(2-(methylsulfonyl)phenylamino)-lH-pyrazolo[3,4- d]pyrimidin-6-ylamino)biphenyl-4-carboxamide; and
N6-(2-isopropoxy-5-methyl-4-(l-methylpiperidin-4-yl)phenyl)-N4-(2- (methylsulfonyl)phenyl)-lH-pyrazolo[3,4-d]pyrimidine-4,6-diamine; or a physiologically acceptable salt, solvate, hydrate or stereoisomer thereof.
14. A pharmaceutical composition comprising a therapeutically effective amount of a compound according to any one of claims 1-13 and a pharmaceutically acceptable diluent or carrier, and optionally in combination with a second therapeutic agent.
15. The pharmaceutical composition of claim 14, wherein said second therapeutic agent is an anti-hyperproliferative agent.
16. A method of inhibiting kinase activity in a cell comprising contacting a cell with one or more compounds of any one of claims 1-13 or a pharmaceutical composition thereof.
17. The method of claim 16 wherein said kinase is Ros, IGF-IR, InsR, anaplastic lymphoma kinase, hepatocyte growth factor receptor tyrosine kinase (c-Met) or monopolar spindle (Mpsl) kinase.
18. The use of a compound of any one of claims 1-13 or a pharmaceutical composition thereof, and optionally in combination with a chemotherapeutic agent, for the manufacture of a medicament for treating a hyperproliferative or angiogenesis disorder mediated by Ros, IGF-IR, InsR, anaplastic lymphoma kinase, hepatocyte growth factor receptor tyrosine kinase (c-Met) or monopolar spindle (Mpsl) kinase.
19. The use of claim 18, wherein the hyperproliferative disorder is multiple myeloma, neuroblastoma, lymphoma, leukemia, melanoma, sarcoma, osteosarcoma, synovial sarcoma, Ewing's sarcoma, hepatoma, gastrointestinal stromal tumor or a solid tumor of breast, renal, prostate, colorectal, thyroid, ovarian, pancreas, lung, uterus, respiratory tract, brain, digestive tract, urinary tract, eye, liver, skin, head and neck, thyroid or parathyroid.
PCT/US2008/074490 2007-08-28 2008-08-27 2- (het) arylamino-6-aminopyridine derivatives and fused forms thereof as anaplastic lymphoma kinase inhibitors WO2009032703A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US96644907P 2007-08-28 2007-08-28
US60/966,449 2007-08-28

Publications (1)

Publication Number Publication Date
WO2009032703A1 true WO2009032703A1 (en) 2009-03-12

Family

ID=40111042

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/US2008/074490 WO2009032703A1 (en) 2007-08-28 2008-08-27 2- (het) arylamino-6-aminopyridine derivatives and fused forms thereof as anaplastic lymphoma kinase inhibitors
PCT/US2008/074472 WO2009032694A1 (en) 2007-08-28 2008-08-27 Amino substituted pyrimidine, pyrollopyridine and pyrazolopyrimidine derivatives useful as kinase inhibitors and in treating proliferative disorders and diseases associated with angiogenesis

Family Applications After (1)

Application Number Title Priority Date Filing Date
PCT/US2008/074472 WO2009032694A1 (en) 2007-08-28 2008-08-27 Amino substituted pyrimidine, pyrollopyridine and pyrazolopyrimidine derivatives useful as kinase inhibitors and in treating proliferative disorders and diseases associated with angiogenesis

Country Status (2)

Country Link
AU (1) AU2008296479A1 (en)
WO (2) WO2009032703A1 (en)

Cited By (50)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010007756A1 (en) * 2008-07-14 2010-01-21 塩野義製薬株式会社 Pyridine derivative having ttk inhibition activity
WO2010128659A1 (en) 2009-05-08 2010-11-11 アステラス製薬株式会社 Diamino heterocyclic carboxamide compound
US8039479B2 (en) 2006-12-08 2011-10-18 Irm Llc Compounds and compositions as protein kinase inhibitors
CN102746308A (en) * 2012-07-09 2012-10-24 四川大学 Allopurinol derivative and preparation method and application thereof
US8318702B2 (en) 2007-07-06 2012-11-27 Astellas Pharma Inc. Di(arylamino)aryl compounds
US8338439B2 (en) 2008-06-27 2012-12-25 Celgene Avilomics Research, Inc. 2,4-disubstituted pyrimidines useful as kinase inhibitors
US8450335B2 (en) 2008-06-27 2013-05-28 Celgene Avilomics Research, Inc. 2,4-disubstituted pyrimidines useful as kinase inhibitors
US8563568B2 (en) 2010-08-10 2013-10-22 Celgene Avilomics Research, Inc. Besylate salt of a BTK inhibitor
WO2014006554A1 (en) 2012-07-03 2014-01-09 Aurigene Discovery Technologies Limited 3-(PYRAZOLYL)-1H-PYRROLO[2,3-b]PYRIDINE DERIVATIVES AS KINASE INHIBITORS
US8685988B2 (en) 2012-08-06 2014-04-01 Acea Biosciences, Inc. EGFR modulators and uses thereof
US8796255B2 (en) 2010-11-10 2014-08-05 Celgene Avilomics Research, Inc Mutant-selective EGFR inhibitors and uses thereof
US8975249B2 (en) 2010-11-01 2015-03-10 Celgene Avilomics Research, Inc. Heterocyclic compounds and uses thereof
US9012462B2 (en) 2008-05-21 2015-04-21 Ariad Pharmaceuticals, Inc. Phosphorous derivatives as kinase inhibitors
US9056839B2 (en) 2012-03-15 2015-06-16 Celgene Avilomics Research, Inc. Solid forms of an epidermal growth factor receptor kinase inhibitor
US9108927B2 (en) 2012-03-15 2015-08-18 Celgene Avilomics Research, Inc. Salts of an epidermal growth factor receptor kinase inhibitor
US9126950B2 (en) 2012-12-21 2015-09-08 Celgene Avilomics Research, Inc. Heteroaryl compounds and uses thereof
US9145387B2 (en) 2013-02-08 2015-09-29 Celgene Avilomics Research, Inc. ERK inhibitors and uses thereof
US9206166B2 (en) 2012-11-06 2015-12-08 SHANGHAI iNSTITUTE OF MATERIA MEDICA ACADEMY OF SCIENCES Certain protein kinase inhibitors
US9238629B2 (en) 2010-11-01 2016-01-19 Celgene Avilomics Research, Inc. Heteroaryl compounds and uses thereof
US9273077B2 (en) 2008-05-21 2016-03-01 Ariad Pharmaceuticals, Inc. Phosphorus derivatives as kinase inhibitors
US9364476B2 (en) 2011-10-28 2016-06-14 Celgene Avilomics Research, Inc. Methods of treating a Bruton's Tyrosine Kinase disease or disorder
US9415049B2 (en) 2013-12-20 2016-08-16 Celgene Avilomics Research, Inc. Heteroaryl compounds and uses thereof
US9464089B2 (en) 2012-01-13 2016-10-11 Acea Biosciences Inc. Heterocyclic compounds and uses thereof
US9492471B2 (en) 2013-08-27 2016-11-15 Celgene Avilomics Research, Inc. Methods of treating a disease or disorder associated with Bruton'S Tyrosine Kinase
US9586965B2 (en) 2012-01-13 2017-03-07 Acea Biosciences Inc. Pyrrolo[2,3-d]pyrimidine compounds as inhibitors of protein kinases
US9611283B1 (en) 2013-04-10 2017-04-04 Ariad Pharmaceuticals, Inc. Methods for inhibiting cell proliferation in ALK-driven cancers
US9834571B2 (en) 2012-05-05 2017-12-05 Ariad Pharmaceuticals, Inc. Compounds for inhibiting cell proliferation in EGFR-driven cancers
US9834518B2 (en) 2011-05-04 2017-12-05 Ariad Pharmaceuticals, Inc. Compounds for inhibiting cell proliferation in EGFR-driven cancers
WO2018021826A1 (en) * 2016-07-26 2018-02-01 한국화학연구원 Novel pyrimidine-2,4-diamine derivative and pharmaceutical composition for prevention or treatment of cancer containing same as active ingredient
US9908884B2 (en) 2009-05-05 2018-03-06 Dana-Farber Cancer Institute, Inc. EGFR inhibitors and methods of treating disorders
US10005760B2 (en) 2014-08-13 2018-06-26 Celgene Car Llc Forms and compositions of an ERK inhibitor
US20180244676A1 (en) * 2015-02-13 2018-08-30 Dana-Farber Cancer Institute, Inc. Lrrk2 inhibitors and methods of making and using the same
CN108473490A (en) * 2015-12-21 2018-08-31 癌症研究科技有限公司 New pyrrole simultaneously [3,2-c] pyridine -6- aminoderivatives
KR101896568B1 (en) * 2017-03-23 2018-09-10 재단법인 대구경북첨단의료산업진흥재단 Pyrolo-pyridine derivatives, preparation method thereof, and pharmaceutical composition for use in preventing or treating protein kinase related disease as an active ingredient
WO2018183112A1 (en) * 2017-03-27 2018-10-04 Cardurion Pharmaceuticals, Llc Heterocyclic compound
US20190106422A1 (en) * 2014-11-06 2019-04-11 Ohio State Innovation Foundation Pyrrolopyrimidine derivatives as mps1/ttk kinase inhibitors
WO2019112344A1 (en) * 2017-12-07 2019-06-13 주식회사 온코빅스 Novel pyrimidine derivative having effect of inhibiting cancer cell growth and pharmaceutical composition containing same
US10533011B2 (en) 2015-10-09 2020-01-14 ACEA Therapeutics, Inc. Pharmaceutical salts, physical forms, and compositions of pyrrolopyrimidine kinase inhibitors, and methods of making same
US10562918B2 (en) 2013-07-11 2020-02-18 ACEA Therapeutics, Inc. Heterocyclic compounds and uses thereof
US10577344B2 (en) 2013-09-10 2020-03-03 The Board Of Regents Of The University Of Texas System Therapeutics targeting truncated adenomatous polyposis coli (APC) proteins
US10596174B2 (en) 2012-01-13 2020-03-24 ACEA Therapeutics, Inc. Pyrrolopyrimidine compounds as inhibitors of protein kinases
CN111386266A (en) * 2017-12-07 2020-07-07 昂科比克斯有限公司 Novel pyrimidine derivative having cancer cell growth inhibitory effect and pharmaceutical composition comprising same
CN111484484A (en) * 2020-04-13 2020-08-04 沈阳药科大学 2, 4-diaryl aminopyrimidine derivative containing aromatic heterocycle and preparation and application thereof
CN112125884A (en) * 2010-12-17 2020-12-25 诺华股份有限公司 Process for preparing pyrimidine-2, 4-diamine dihydrochloride
US11130752B2 (en) 2018-09-25 2021-09-28 Cardurion Pharmaceuticals, Llc Aminopyrimidine compound
US11208696B2 (en) 2015-04-17 2021-12-28 Netherlands Translational Research Center B.V. Prognostic biomarkers for TTK inhibitor chemotherapy
US11351168B1 (en) 2008-06-27 2022-06-07 Celgene Car Llc 2,4-disubstituted pyrimidines useful as kinase inhibitors
US11498922B2 (en) 2017-04-07 2022-11-15 ACEA Therapeutics, Inc. Pharmaceutical composition comprising N-(3-((2-((3-fluoro-4-(4-methylpiperazin-1-yl phenyl)amino)-7H-pyrrolo[2,3-d]pyrimidin-4-yl)oxy)phenylacrylamide
RU2811770C1 (en) * 2019-12-16 2024-01-17 Онкобикс Ко., Лтд. New pyrimidine derivative substituted with deuterium and pharmaceutical composition containing it
EP4079726A4 (en) * 2019-12-16 2024-01-24 Korea Res Inst Chemical Tech Novel pyrimidin derivative and use thereof

Families Citing this family (47)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8592432B2 (en) 2008-04-07 2013-11-26 Bei Chen Compounds and compositions as protein kinase inhibitors
WO2010111406A2 (en) * 2009-03-24 2010-09-30 Myriad Pharmaceuticals, Inc. Compounds and therapeutic uses thereof
WO2010144909A1 (en) 2009-06-12 2010-12-16 Novartis Ag Fused heterocyclic compounds and their uses
TW201107329A (en) 2009-07-30 2011-03-01 Oncotherapy Science Inc Fused imidazole derivative having ttk inhibitory action
US8629132B2 (en) * 2009-11-13 2014-01-14 Genosco Kinase inhibitors
ES2365960B1 (en) * 2010-03-31 2012-06-04 Palobiofarma, S.L NEW ANTAGONISTS OF ADENOSINE RECEPTORS.
EP2571361A4 (en) 2010-05-19 2013-11-13 Univ North Carolina Pyrazolopyrimidine compounds for the treatment of cancer
GB201104267D0 (en) * 2011-03-14 2011-04-27 Cancer Rec Tech Ltd Pyrrolopyridineamino derivatives
BR112014007788A2 (en) 2011-10-03 2017-04-18 Univ North Carolina Chapel Hill Pyrrolopyrimidine compounds for cancer treatment
JP6106685B2 (en) 2011-11-17 2017-04-05 ダナ−ファーバー キャンサー インスティテュート, インコーポレイテッド Inhibitors of C-JUN-N-terminal kinase (JNK)
CA2867469A1 (en) 2012-03-16 2013-09-19 Axikin Pharmaceuticals, Inc. 3,5-diaminopyrazole kinase inhibitors
WO2013142817A2 (en) 2012-03-23 2013-09-26 Dennis Brown Compositions and methods to improve the therapeutic benefit of indirubin and analogs thereof, including meisoindigo
IN2014DN09610A (en) 2012-05-22 2015-07-31 Univ North Carolina
GB201216017D0 (en) 2012-09-07 2012-10-24 Cancer Rec Tech Ltd Inhibitor compounds
GB201216018D0 (en) 2012-09-07 2012-10-24 Cancer Rec Tech Ltd Pharmacologically active compounds
EP2909211A4 (en) 2012-10-17 2016-06-22 Univ North Carolina Pyrazolopyrimidine compounds for the treatment of cancer
WO2014063068A1 (en) 2012-10-18 2014-04-24 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinase 7 (cdk7)
US10000483B2 (en) 2012-10-19 2018-06-19 Dana-Farber Cancer Institute, Inc. Bone marrow on X chromosome kinase (BMX) inhibitors and uses thereof
WO2014063061A1 (en) 2012-10-19 2014-04-24 Dana-Farber Cancer Institute, Inc. Hydrophobically tagged small molecules as inducers of protein degradation
EP2925752A4 (en) 2012-11-27 2016-06-01 Univ North Carolina Pyrimidine compounds for the treatment of cancer
NZ631142A (en) 2013-09-18 2016-03-31 Axikin Pharmaceuticals Inc Pharmaceutically acceptable salts of 3,5-diaminopyrazole kinase inhibitors
CA2927917C (en) 2013-10-18 2022-08-09 Syros Pharmaceuticals, Inc. Heteroaromatic compounds useful for the treatment of proliferative diseases
WO2015058140A1 (en) 2013-10-18 2015-04-23 Dana-Farber Cancer Institute, Inc. Polycyclic inhibitors of cyclin-dependent kinase 7 (cdk7)
JP6434506B2 (en) * 2013-10-21 2018-12-05 ジェノスコ Substituted pyrimidine compounds and their use as SYK inhibitors
CN104672214B (en) * 2013-12-03 2019-04-12 上海翰森生物医药科技有限公司 Compound and its preparation and purposes with ALK inhibitory activity
GB201403536D0 (en) 2014-02-28 2014-04-16 Cancer Rec Tech Ltd Inhibitor compounds
US9555031B2 (en) 2014-04-11 2017-01-31 The University Of North Carolina At Chapel Hill Therapeutic uses of selected pyrrolopyrimidine compounds with anti-mer tyrosine kinase activity
WO2015164614A1 (en) * 2014-04-23 2015-10-29 Dana-Farber Cancer Institute, Inc. Janus kinase inhibitors and uses thereof
US9862688B2 (en) 2014-04-23 2018-01-09 Dana-Farber Cancer Institute, Inc. Hydrophobically tagged janus kinase inhibitors and uses thereof
CN105272921A (en) * 2014-06-09 2016-01-27 江苏奥赛康药业股份有限公司 Method for preparing Ceritinib and intermediate compound of Ceritinib
HUE049801T2 (en) 2014-12-23 2020-10-28 Sma Therapeutics Inc 3,5-diaminopyrazole kinase inhibitors
AU2015371251B2 (en) 2014-12-23 2020-06-11 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinase 7 (CDK7)
BR112017017887A2 (en) * 2015-03-04 2018-04-10 Novartis Ag chemical process for preparing pyrimidine derivatives and intermediates
WO2016160617A2 (en) 2015-03-27 2016-10-06 Dana-Farber Cancer Institute, Inc. Inhibitors of cyclin-dependent kinases
CA2986441A1 (en) 2015-06-12 2016-12-15 Dana-Farber Cancer Institute, Inc. Combination therapy of transcription inhibitors and kinase inhibitors
WO2017035354A1 (en) 2015-08-26 2017-03-02 Blueprint Medicines Corporation Compounds and compositions useful for treating disorders related to ntrk
JP7028766B2 (en) 2015-09-09 2022-03-02 ダナ-ファーバー キャンサー インスティテュート, インコーポレイテッド Inhibitor of cyclin-dependent kinase
AR106756A1 (en) 2015-11-19 2018-02-14 Blueprint Medicines Corp USEFUL COMPOUNDS AND COMPOSITIONS FOR THE TREATMENT OF NTRK-RELATED DISORDERS
US11214565B2 (en) 2015-11-20 2022-01-04 Denali Therapeutics Inc. Compound, compositions, and methods
WO2017106771A1 (en) 2015-12-16 2017-06-22 Southern Research Institute Pyrrolopyrimidine compounds, use as inhibitors of the kinase lrrk2, and methods for preparation thereof
US11028080B2 (en) 2016-03-11 2021-06-08 Denali Therapeutics Inc. Substituted pyrimidines as LRKK2 inhibitors
US10709708B2 (en) 2016-03-17 2020-07-14 The University Of North Carolina At Chapel Hill Method of treating cancer with a combination of MER tyrosine kinase inhibitor and an epidermal growth factor receptor (EGFR) inhibitor
EP3472153B1 (en) 2016-06-16 2021-09-22 Denali Therapeutics Inc. Pyrimidin-2-ylamino-1h-pyrazols as lrrk2 inhibitors for use in the treatment of neurodegenerative disorders
GB201709840D0 (en) 2017-06-20 2017-08-02 Inst Of Cancer Research: Royal Cancer Hospital Methods and medical uses
JP2022518723A (en) * 2019-01-18 2022-03-16 ボロノイ・カンパニー・リミテッド Its use in the prevention or treatment of pyrrolopyridine derivatives and protein kinase related diseases
WO2021098883A1 (en) * 2019-11-21 2021-05-27 浙江同源康医药股份有限公司 Compound used as egfr kinase inhibitor and use thereof
CN115368366A (en) * 2022-08-02 2022-11-22 江苏省中医药研究院 Pyrimidopyrazole compound and application thereof

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005016894A1 (en) * 2003-08-15 2005-02-24 Novartis Ag 2, 4-pyrimidinediamines useful in the treatment of neoplastic diseases, inflammatory and immune system disorders
WO2007070872A1 (en) * 2005-12-15 2007-06-21 Rigel Pharmaceuticals, Inc. Kinase inhibitors and their uses
WO2008039359A2 (en) * 2006-09-25 2008-04-03 Janssen Pharmaceutica N.V. Bicyclic pyrimidine kinase inhibitors
WO2008073687A2 (en) * 2006-12-08 2008-06-19 Irm Llc Compounds and compositions as protein kinase inhibitors
WO2008081928A1 (en) * 2006-12-28 2008-07-10 Taisho Pharmaceutical Co., Ltd. Pyrazolopyrimidine compound

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2004259012C1 (en) * 2003-07-23 2012-08-02 Exelixis, Inc. Anaplastic lymphoma kinase modulators and methods of use
WO2005026129A1 (en) * 2003-09-15 2005-03-24 Gpc Biotech Ag Pharmaceutically active 4,6-disubstituted aminopyrimidine derivatives as modulators of protein kinases
EP1598343A1 (en) * 2004-05-19 2005-11-23 Boehringer Ingelheim International GmbH 2-Arylaminopyrimidine derivatives as PLK inhibitors

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005016894A1 (en) * 2003-08-15 2005-02-24 Novartis Ag 2, 4-pyrimidinediamines useful in the treatment of neoplastic diseases, inflammatory and immune system disorders
WO2007070872A1 (en) * 2005-12-15 2007-06-21 Rigel Pharmaceuticals, Inc. Kinase inhibitors and their uses
WO2008039359A2 (en) * 2006-09-25 2008-04-03 Janssen Pharmaceutica N.V. Bicyclic pyrimidine kinase inhibitors
WO2008073687A2 (en) * 2006-12-08 2008-06-19 Irm Llc Compounds and compositions as protein kinase inhibitors
WO2008081928A1 (en) * 2006-12-28 2008-07-10 Taisho Pharmaceutical Co., Ltd. Pyrazolopyrimidine compound

Cited By (114)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8399450B2 (en) 2006-12-08 2013-03-19 Irm Llc Compounds and compositions as protein kinase inhibitors
US8039479B2 (en) 2006-12-08 2011-10-18 Irm Llc Compounds and compositions as protein kinase inhibitors
US8957081B2 (en) 2006-12-08 2015-02-17 Irm Llc Compounds and compositions as protein kinase inhibitors
US8372858B2 (en) 2006-12-08 2013-02-12 Irm Llc Compounds and compositions as protein kinase inhibitors
US8377921B2 (en) 2006-12-08 2013-02-19 Irm Llc Compounds and compositions as protein kinase inhibitors
US8318702B2 (en) 2007-07-06 2012-11-27 Astellas Pharma Inc. Di(arylamino)aryl compounds
US9273077B2 (en) 2008-05-21 2016-03-01 Ariad Pharmaceuticals, Inc. Phosphorus derivatives as kinase inhibitors
US9012462B2 (en) 2008-05-21 2015-04-21 Ariad Pharmaceuticals, Inc. Phosphorous derivatives as kinase inhibitors
US9212181B2 (en) 2008-06-27 2015-12-15 Celgene Avilomics Research, Inc. Substituted 2,4-diaminopyrimidines as kinase inhibitors
US10596172B2 (en) 2008-06-27 2020-03-24 Celgene Car Llc 2,4-disubstituted pyrimidines useful as kinase inhibitors
US9409921B2 (en) 2008-06-27 2016-08-09 Celgene Avilomics Research, Inc. 2,4-disubstituted pyrimidines as kinase inhibitors
US8609679B2 (en) 2008-06-27 2013-12-17 Celgene Avilomics Research, Inc. 2,4-diaminopyrimidines useful as kinase inhibitors
US9296737B2 (en) 2008-06-27 2016-03-29 Celgene Avilomics Research, Inc. Substituted 2,4-diaminopyrimidines as kinase inhibitors
US10828300B2 (en) 2008-06-27 2020-11-10 Celgene Car Llc Substituted 2,4-diaminopyrimidines as kinase inhibitors
US8710222B2 (en) 2008-06-27 2014-04-29 Celgene Avilomics Research, Inc. 2,4-disubstituted pyrimidines useful as kinase inhibitors
US10010548B2 (en) 2008-06-27 2018-07-03 Celgene Car Llc 2,4-disubstituted pyrimidines useful as kinase inhibitors
US8450335B2 (en) 2008-06-27 2013-05-28 Celgene Avilomics Research, Inc. 2,4-disubstituted pyrimidines useful as kinase inhibitors
US8338439B2 (en) 2008-06-27 2012-12-25 Celgene Avilomics Research, Inc. 2,4-disubstituted pyrimidines useful as kinase inhibitors
US9987276B2 (en) 2008-06-27 2018-06-05 Celgene Car Llc Substituted 2,4-diaminopyrimidines as kinase inhibitors
US11351168B1 (en) 2008-06-27 2022-06-07 Celgene Car Llc 2,4-disubstituted pyrimidines useful as kinase inhibitors
WO2010007756A1 (en) * 2008-07-14 2010-01-21 塩野義製薬株式会社 Pyridine derivative having ttk inhibition activity
US9908884B2 (en) 2009-05-05 2018-03-06 Dana-Farber Cancer Institute, Inc. EGFR inhibitors and methods of treating disorders
US9487491B2 (en) 2009-05-08 2016-11-08 Astellas Pharma Inc. Diamino heterocyclic carboxamide compound
WO2010128659A1 (en) 2009-05-08 2010-11-11 アステラス製薬株式会社 Diamino heterocyclic carboxamide compound
US8969336B2 (en) 2009-05-08 2015-03-03 Astellas Pharma Inc. Diamino heterocyclic carboxamide compound
EP3009428A1 (en) 2009-05-08 2016-04-20 Astellas Pharma Inc. Diamino heterocyclic carboxamide compound
US9604936B2 (en) 2010-08-10 2017-03-28 Celgene Car Llc Besylate salt of a BTK inhibitor
US8563568B2 (en) 2010-08-10 2013-10-22 Celgene Avilomics Research, Inc. Besylate salt of a BTK inhibitor
US9867824B2 (en) 2010-11-01 2018-01-16 Celgene Car Llc Heterocyclic compounds and uses thereof
US9238629B2 (en) 2010-11-01 2016-01-19 Celgene Avilomics Research, Inc. Heteroaryl compounds and uses thereof
US11096942B2 (en) 2010-11-01 2021-08-24 Celgene Car Llc Heterocyclic compounds and uses thereof
US10434101B2 (en) 2010-11-01 2019-10-08 Celgene Car Llc Heterocyclic compounds and uses thereof
US9765038B2 (en) 2010-11-01 2017-09-19 Celgene Car Llc Heteroaryl compounds and uses thereof
US9375431B2 (en) 2010-11-01 2016-06-28 Celgene Avilomics Research, Inc. 2,4-disubstituted pyrimidine compounds useful as kinase inhibtors
US10081606B2 (en) 2010-11-01 2018-09-25 Celgene Car Llc Heteroaryl compounds and uses thereof
US8975249B2 (en) 2010-11-01 2015-03-10 Celgene Avilomics Research, Inc. Heterocyclic compounds and uses thereof
US9868723B2 (en) 2010-11-10 2018-01-16 Celgene Car Llc Mutant-selective EGFR inhibitors and uses thereof
US9409887B2 (en) 2010-11-10 2016-08-09 Celgene Avilomics Research, Inc. Mutant-selective EGFR inhibitors and uses thereof
US8796255B2 (en) 2010-11-10 2014-08-05 Celgene Avilomics Research, Inc Mutant-selective EGFR inhibitors and uses thereof
CN112125884A (en) * 2010-12-17 2020-12-25 诺华股份有限公司 Process for preparing pyrimidine-2, 4-diamine dihydrochloride
US9834518B2 (en) 2011-05-04 2017-12-05 Ariad Pharmaceuticals, Inc. Compounds for inhibiting cell proliferation in EGFR-driven cancers
US9364476B2 (en) 2011-10-28 2016-06-14 Celgene Avilomics Research, Inc. Methods of treating a Bruton's Tyrosine Kinase disease or disorder
US9920074B2 (en) 2012-01-13 2018-03-20 Acea Biosciences Inc. Heterocyclic compounds and uses thereof
US10799504B2 (en) 2012-01-13 2020-10-13 ACEA Therapeutics, Inc. Heterocyclic compounds and uses as anticancer agents
US10596174B2 (en) 2012-01-13 2020-03-24 ACEA Therapeutics, Inc. Pyrrolopyrimidine compounds as inhibitors of protein kinases
US9464089B2 (en) 2012-01-13 2016-10-11 Acea Biosciences Inc. Heterocyclic compounds and uses thereof
US9586965B2 (en) 2012-01-13 2017-03-07 Acea Biosciences Inc. Pyrrolo[2,3-d]pyrimidine compounds as inhibitors of protein kinases
US9034885B2 (en) 2012-01-13 2015-05-19 Acea Biosciences Inc. EGFR modulators and uses thereof
US11612602B2 (en) 2012-01-13 2023-03-28 ACEA Therapeutics, Inc. Heterocyclic compounds and uses as anticancer agents
US9763949B2 (en) 2012-01-13 2017-09-19 Acea Biosciences Inc. EGFR modulators and uses thereof
US10005738B2 (en) 2012-03-15 2018-06-26 Celgene Car Llc Salts of an epidermal growth factor receptor kinase inhibitor
US9540335B2 (en) 2012-03-15 2017-01-10 Celgene Avilomics Research, Inc. Salts of an epidermal growth factor receptor kinase inhibitor
US10570099B2 (en) 2012-03-15 2020-02-25 Celgene Car Llc Salts of an epidermal growth factor receptor kinase inhibitor
US11292772B2 (en) 2012-03-15 2022-04-05 Celgene Car Llc Salts of an epidermal growth factor receptor kinase inhibitor
US9539255B2 (en) 2012-03-15 2017-01-10 Celgene Avilomics Research, Inc. Solid forms of an epidermal growth factor receptor kinase inhibitor
US9056839B2 (en) 2012-03-15 2015-06-16 Celgene Avilomics Research, Inc. Solid forms of an epidermal growth factor receptor kinase inhibitor
US10946016B2 (en) 2012-03-15 2021-03-16 Celgene Car Llc Solid forms of an epidermal growth factor receptor kinase inhibitor
US9108927B2 (en) 2012-03-15 2015-08-18 Celgene Avilomics Research, Inc. Salts of an epidermal growth factor receptor kinase inhibitor
US10004741B2 (en) 2012-03-15 2018-06-26 Celgene Car Llc Solid forms of an epidermal growth factor receptor kinase inhibitor
US9834571B2 (en) 2012-05-05 2017-12-05 Ariad Pharmaceuticals, Inc. Compounds for inhibiting cell proliferation in EGFR-driven cancers
WO2014006554A1 (en) 2012-07-03 2014-01-09 Aurigene Discovery Technologies Limited 3-(PYRAZOLYL)-1H-PYRROLO[2,3-b]PYRIDINE DERIVATIVES AS KINASE INHIBITORS
CN102746308A (en) * 2012-07-09 2012-10-24 四川大学 Allopurinol derivative and preparation method and application thereof
CN102746308B (en) * 2012-07-09 2014-12-31 四川大学 Allopurinol derivative and preparation method and application thereof
US8685988B2 (en) 2012-08-06 2014-04-01 Acea Biosciences, Inc. EGFR modulators and uses thereof
US11007197B2 (en) 2012-08-06 2021-05-18 ACEA Therapeutics, Inc. EGFR modulators and uses thereof
US10449196B2 (en) 2012-08-06 2019-10-22 ACEA Therapeutics, Inc. EGFR modulators and uses thereof
US9567342B2 (en) 2012-11-06 2017-02-14 Shanghai Fochon Pharmaceutical Co., Ltd. Certain protein kinase inhibitors
US9206166B2 (en) 2012-11-06 2015-12-08 SHANGHAI iNSTITUTE OF MATERIA MEDICA ACADEMY OF SCIENCES Certain protein kinase inhibitors
US9549927B2 (en) 2012-12-21 2017-01-24 Celgene Avilomics Research, Inc. Heteroaryl compounds and uses thereof
US9126950B2 (en) 2012-12-21 2015-09-08 Celgene Avilomics Research, Inc. Heteroaryl compounds and uses thereof
US9145387B2 (en) 2013-02-08 2015-09-29 Celgene Avilomics Research, Inc. ERK inhibitors and uses thereof
US9561228B2 (en) 2013-02-08 2017-02-07 Celgene Avilomics Research, Inc. ERK inhibitors and uses thereof
US9796700B2 (en) 2013-02-08 2017-10-24 Celgene Car Llc ERK inhibitors and uses thereof
US9504686B2 (en) 2013-02-08 2016-11-29 Celgene Avilomics Research, Inc. ERK inhibitors and uses thereof
US9980964B2 (en) 2013-02-08 2018-05-29 Celgene Car Llc ERK inhibitors and uses thereof
US9611283B1 (en) 2013-04-10 2017-04-04 Ariad Pharmaceuticals, Inc. Methods for inhibiting cell proliferation in ALK-driven cancers
US10562918B2 (en) 2013-07-11 2020-02-18 ACEA Therapeutics, Inc. Heterocyclic compounds and uses thereof
US9492471B2 (en) 2013-08-27 2016-11-15 Celgene Avilomics Research, Inc. Methods of treating a disease or disorder associated with Bruton'S Tyrosine Kinase
US10577344B2 (en) 2013-09-10 2020-03-03 The Board Of Regents Of The University Of Texas System Therapeutics targeting truncated adenomatous polyposis coli (APC) proteins
US9415049B2 (en) 2013-12-20 2016-08-16 Celgene Avilomics Research, Inc. Heteroaryl compounds and uses thereof
US10005760B2 (en) 2014-08-13 2018-06-26 Celgene Car Llc Forms and compositions of an ERK inhibitor
US10202364B2 (en) 2014-08-13 2019-02-12 Celgene Car Llc Forms and compositions of an ERK inhibitor
US10611765B2 (en) * 2014-11-06 2020-04-07 Ohio State Innovation Foundation Pyrrolopyrimidine derivatives as Mps1/TTK kinase inhibitors
US20190106422A1 (en) * 2014-11-06 2019-04-11 Ohio State Innovation Foundation Pyrrolopyrimidine derivatives as mps1/ttk kinase inhibitors
US10913744B2 (en) 2015-02-13 2021-02-09 Dana-Farber Cancer Institute, Inc. LRRK2 inhibitors and methods of making and using the same
US20180244676A1 (en) * 2015-02-13 2018-08-30 Dana-Farber Cancer Institute, Inc. Lrrk2 inhibitors and methods of making and using the same
US11208696B2 (en) 2015-04-17 2021-12-28 Netherlands Translational Research Center B.V. Prognostic biomarkers for TTK inhibitor chemotherapy
US10533011B2 (en) 2015-10-09 2020-01-14 ACEA Therapeutics, Inc. Pharmaceutical salts, physical forms, and compositions of pyrrolopyrimidine kinase inhibitors, and methods of making same
CN108473490A (en) * 2015-12-21 2018-08-31 癌症研究科技有限公司 New pyrrole simultaneously [3,2-c] pyridine -6- aminoderivatives
CN108473490B9 (en) * 2015-12-21 2020-12-25 癌症研究科技有限公司 Pyrrolo [3,2-c ] pyridin-6-amino derivatives
CN108473490B (en) * 2015-12-21 2020-11-13 癌症研究科技有限公司 Pyrrolo [3,2-c ] pyridin-6-amino derivatives
WO2018021826A1 (en) * 2016-07-26 2018-02-01 한국화학연구원 Novel pyrimidine-2,4-diamine derivative and pharmaceutical composition for prevention or treatment of cancer containing same as active ingredient
CN110662745B (en) * 2017-03-23 2021-08-03 大邱庆北尖端医疗产业振兴财团 Pyrrolopyridine derivatives, process for producing the same, and pharmaceutical composition for preventing or treating protein kinase-associated diseases
AU2018239798B2 (en) * 2017-03-23 2020-08-27 Daegu Gyeongbuk Institute Of Science And Technology Pyrrolo-pyridine derivative compound, method for preparing same, and pharmaceutical composition containing same as active ingredient for prevention or treatment of protein kinase-related diseases
WO2018174650A1 (en) * 2017-03-23 2018-09-27 재단법인 대구경북첨단의료산업진흥재단 Pyrrolo-pyridine derivative compound, method for preparing same, and pharmaceutical composition containing same as active ingredient for prevention or treatment of protein kinase-related diseases
CN110662745A (en) * 2017-03-23 2020-01-07 大邱庆北尖端医疗产业振兴财团 Pyrrolopyridine derivatives, process for preparing the same, and pharmaceutical composition for preventing or treating protein kinase-associated diseases comprising the same as active ingredient
US11117892B2 (en) 2017-03-23 2021-09-14 Daegu-Gyeongbuk Medical Innovation Foundation Pyrrolo-pyridine derivative compound, method for preparing same, and pharmaceutical composition containing same as active ingredient for prevention or treatment of protein kinase-related diseases
KR101896568B1 (en) * 2017-03-23 2018-09-10 재단법인 대구경북첨단의료산업진흥재단 Pyrolo-pyridine derivatives, preparation method thereof, and pharmaceutical composition for use in preventing or treating protein kinase related disease as an active ingredient
WO2018183112A1 (en) * 2017-03-27 2018-10-04 Cardurion Pharmaceuticals, Llc Heterocyclic compound
US11197858B2 (en) 2017-03-27 2021-12-14 Cardurion Pharmaceuticals, Llc Substituted amines for treating cardiac diseases
TWI789381B (en) * 2017-03-27 2023-01-11 美商卡都瑞恩醫藥有限責任公司 Heterocyclic compound
US10543212B2 (en) 2017-03-27 2020-01-28 Cardurion Pharmaceuticals, Llc Substituted amines for treating cardiac diseases
US11498922B2 (en) 2017-04-07 2022-11-15 ACEA Therapeutics, Inc. Pharmaceutical composition comprising N-(3-((2-((3-fluoro-4-(4-methylpiperazin-1-yl phenyl)amino)-7H-pyrrolo[2,3-d]pyrimidin-4-yl)oxy)phenylacrylamide
US11248003B2 (en) 2017-12-07 2022-02-15 Oncobix Co., Ltd. Pyrimidine derivative having effect of inhibiting cancer cell growth and pharmaceutical composition containing same
AU2018379499B2 (en) * 2017-12-07 2021-03-04 Oncobix Co., Ltd. Novel pyrimidine derivative having effect of inhibiting cancer cell growth and pharmaceutical composition containing same
RU2744168C1 (en) * 2017-12-07 2021-03-03 Онкобикс Ко., Лтд. New pyrimidine derivative with an effect of inhibiting growth of cancer cells and pharmaceutical composition containing it
CN111386266A (en) * 2017-12-07 2020-07-07 昂科比克斯有限公司 Novel pyrimidine derivative having cancer cell growth inhibitory effect and pharmaceutical composition comprising same
WO2019112344A1 (en) * 2017-12-07 2019-06-13 주식회사 온코빅스 Novel pyrimidine derivative having effect of inhibiting cancer cell growth and pharmaceutical composition containing same
CN111386266B (en) * 2017-12-07 2023-08-04 昂科比克斯有限公司 Pyrimidine derivatives having cancer cell growth inhibiting effect and pharmaceutical compositions containing the same
US11130752B2 (en) 2018-09-25 2021-09-28 Cardurion Pharmaceuticals, Llc Aminopyrimidine compound
RU2811770C1 (en) * 2019-12-16 2024-01-17 Онкобикс Ко., Лтд. New pyrimidine derivative substituted with deuterium and pharmaceutical composition containing it
EP4079726A4 (en) * 2019-12-16 2024-01-24 Korea Res Inst Chemical Tech Novel pyrimidin derivative and use thereof
CN111484484B (en) * 2020-04-13 2021-11-23 沈阳药科大学 2, 4-diaryl aminopyrimidine derivative containing aromatic heterocycle and preparation and application thereof
CN111484484A (en) * 2020-04-13 2020-08-04 沈阳药科大学 2, 4-diaryl aminopyrimidine derivative containing aromatic heterocycle and preparation and application thereof

Also Published As

Publication number Publication date
WO2009032694A1 (en) 2009-03-12
AU2008296479A1 (en) 2009-03-12

Similar Documents

Publication Publication Date Title
WO2009032703A1 (en) 2- (het) arylamino-6-aminopyridine derivatives and fused forms thereof as anaplastic lymphoma kinase inhibitors
KR102558066B1 (en) Pyrrolotriazine compounds as tam inhibitors
EP3399968B1 (en) Selective inhibitors of clinically important mutants of the egfr tyrosine kinase
KR101036394B1 (en) Diaryl Urea Derivatives Useful for the Treatment of Protein Kinase Dependent Diseases
CN102816162B (en) Pyrimido-pyrimidine ketone compounds and medicinal compositions thereof and application
CA2631775C (en) Substituted 4-amino-pyrrolotriazine derivatives useful for treating hyper-proliferative disorders and diseases associated with angiogenesis
JP5702390B2 (en) Pyrimidine derivatives as protein tyrosine kinase 2 inhibitors
US8129379B2 (en) Substituted 4-amino-pyrrolotriazine derivatives useful for treating hyper-proliferative disorders and diseases associated with angiogenesis
KR101432352B1 (en) Pyrimidine derivatives as kinase inhibitors
SG192686A1 (en) Compounds and methods for kinase modulation, and indications therefor
CN103012399A (en) 7-oxopyridinopyrimidine compound as well as medicinal composition and application thereof
JP2022522153A (en) Inhibitors of BTK and its variants
CA3037097C (en) Deuterated 3-(4,5-substituted aminopyrimidine) phenyl derivatives and use thereof
CN111356687B (en) Pyrimidine-containing tri-substituted imidazole compound and application thereof
TW201245176A (en) New 5-alkynyl-pyridines
CN103570731B (en) Pyrimido three encircles or pyrimido Fourth Ring compounds and Pharmaceutical composition and application
JP2013528635A (en) Biphenyl-substituted 1,3-dihydro-benzimidazol-2-ylideneamine derivatives
WO2022148317A1 (en) 2-aminopyrimidine compound and pharmaceutical composition thereof and application thereof
WO2020192302A1 (en) Pyrimidine-containing tri-substituted imidazole compound and application thereof
CN110437220B (en) Triazole compound and application thereof
TW201245175A (en) New 5-alkynyl-pyridines
JP2020531501A (en) Bcl-2 inhibitor

Legal Events

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

Ref document number: 08798818

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 08798818

Country of ref document: EP

Kind code of ref document: A1