US20130172360A1 - Derivatives of azaindazole or diazaindazole type as medicament - Google Patents

Derivatives of azaindazole or diazaindazole type as medicament Download PDF

Info

Publication number
US20130172360A1
US20130172360A1 US13/823,658 US201213823658A US2013172360A1 US 20130172360 A1 US20130172360 A1 US 20130172360A1 US 201213823658 A US201213823658 A US 201213823658A US 2013172360 A1 US2013172360 A1 US 2013172360A1
Authority
US
United States
Prior art keywords
group
compound
formula
yield
mmol
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US13/823,658
Other languages
English (en)
Inventor
El Bachir Kaloun
Karim Bedjeguelal
Rémi Rabot
Anna Kruczynski
Philippe Schmitt
Michel Perez
Nicolas Rahier
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Pierre Fabre Medicament SA
Original Assignee
Pierre Fabre Medicament SA
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 Pierre Fabre Medicament SA filed Critical Pierre Fabre Medicament SA
Assigned to PIERRE FABRE MEDICAMENT reassignment PIERRE FABRE MEDICAMENT ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BEDJEGUELAL, KARIM, KALOUN, EL BACHIR, KRUCZYNSKI, ANNA, PEREZ, MICHEL, RABOT, REMI, RAHIER, NICOLAS, SCHMITT, PHILIPPE
Publication of US20130172360A1 publication Critical patent/US20130172360A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/445Non condensed piperidines, e.g. piperocaine
    • A61K31/4523Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems
    • A61K31/4545Non condensed piperidines, e.g. piperocaine containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring hetero atom, e.g. pipamperone, anabasine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/4985Pyrazines or piperazines ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • 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

  • the present invention relates to azaindazole and diazaindazole fused bicyclic derivatives, as well as to the therapeutic use of same, notably in the treatment of cancer, inflammation and neurodegenerative diseases such as Alzheimer's disease, as well as to methods for synthesizing same.
  • Protein kinases are enzymes that play a key role in cell signal transduction. They are involved in physiological processes such as cell proliferation, mitosis, differentiation, cell invasion and mobility, and apoptosis, for example.
  • Tyrosine kinase receptors form a particular class of protein kinases among which, among others, mention may be made of ALK, EGFR, Her2, PDGFR, Kit, VEGFR, IGFR, FGFR, Trk, Axl, Mer, Met, Ron and Ret.
  • ALK is regarded as a particularly relevant target because it is genetically modified in certain tumor pathologies and thus acquires an oncogenic nature. More precisely, chromosomal translocations leading to the production of fused protein kinases (ALK-X) which are then constitutively activated cause the development of certain cancers.
  • ALK in oncogenic form is expressed by various tumor pathologies of different histological types.
  • ALK-dependent pathologies are thus ALK-dependent.
  • ALK in oncogenic form exists only in tumor cells and is not expressed by normal cells.
  • this protein kinase provides the opportunity to specifically target ALK-dependent tumor tissues while saving healthy tissues from significant toxic effects (Ott G. R. et al., Anticancer Agents Med. Chem., 2010, 10(3), 236-49).
  • the fusion protein NPM-ALK is associated with anaplastic large-cell lymphoma (ALCL) for which an optimal treatment remains to be developed.
  • the fusion protein EML4-ALK is associated with tumor development in a subpopulation of patients suffering from non-small cell lung cancer. Mutated forms of ALK have also been observed in neuroblastoma.
  • c-Src is also a protein kinase whose activation state proved to be negatively correlated with the survival of patients suffering from various forms of cancer, including non-small cell lung cancer (Byers L. A. et al., Clin. Cancer Res. 2009, 15(22), 6852-6861).
  • this protein is also regarded as a target of interest in oncology.
  • inhibitors with a composite mode of action that are capable of intervening at several targets, in particular at several targets of the same signaling pathway, proposed as being more effective, with an improved therapeutic index and less likely to give rise to phenomena of compensation, resistance or therapeutic escape.
  • the compounds of the present invention thus have the property of inhibiting or modulating the enzymatic activity of protein kinases in general and ALK and c-Src in particular. Consequently, said compounds can be used as drug in the treatment of proliferative diseases such as cancer.
  • the present invention thus has as an object a compound of following general formula (I):
  • halogen refers to fluorine, chlorine, bromine or iodine.
  • (C 1 -C 6 ) alkyl refers to saturated linear or branched hydrocarbon chains comprising 1 to 6 carbon atoms. It may be a methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, pentyl or hexyl group.
  • (C 1 -C 6 )alkoxy refers to a (C 1 -C 6 ) alkyl chain linked to the rest of the molecule via an oxygen atom.
  • (C 1 -C 6 )thioalkoxy refers to a (C 1 -C 6 ) alkyl chain linked to the rest of the molecule via a sulfur atom.
  • (C 1 -C 6 )haloalkyl refers to a (C 1 -C 6 ) alkyl chain such as defined above wherein one or more hydrogen atoms are replaced by a halogen atom such as defined above. It may be in particular a trifluoromethyl group.
  • (C 1 -C 6 )haloalkoxy refers to a (C 1 -C 6 )alkoxy chain such as defined above wherein one or more hydrogen atoms are replaced by a halogen atom such as defined above. It may be in particular a trifluoromethoxy group.
  • (C 1 -C 6 )halothioalkoxy refers to a (C 1 -C 6 )thioalkoxy chain such as defined above wherein one or more hydrogen atoms are replaced by a halogen atom such as defined above. It may be in particular a trifluorothiomethoxy group.
  • (C 3 -C 12 )cycloalkyl refers to cyclic hydrocarbon systems comprising from 3 to 12 carbon atoms and comprising one or more rings, in particular fused rings. As an example, mention may be made of an adamantyl or cyclohexyl group.
  • aryl refers to an aromatic hydrocarbon group preferably comprising from 6 to 14 carbon atoms and comprising one or more fused rings, such as, for example, a phenyl or naphthyl group.
  • aryl is a phenyl group.
  • heteroaryl refers to a cyclic aromatic group comprising 5 to 7 atoms included in the ring or a bicyclic aromatic group comprising 8 to 11 atoms included in the rings, wherein 1 to 4 of the atoms included in the rings are a heteroatom selected independently from sulfur, nitrogen and oxygen atoms, and wherein the other atoms included in the rings are carbon atoms.
  • heteroaryl groups include furyl, thienyl, pyridinyl, and benzothienyl groups.
  • heterocycle refers either to a stable monocycle containing from 4 to 7 cyclic atoms, or to a stable bicycle containing from 8 to 11 cyclic atoms, which may be either saturated or unsaturated, wherein 1 to 4 of the cyclic atoms are a heteroatom selected independently from sulfur, nitrogen and oxygen atoms, and wherein the other cyclic atoms are carbon atoms.
  • “optionally substituted” means that the group in question is optionally substituted by one or more substituents which may be selected in particular from a halogen atom, (C 1 -C 6 )alkyl, (C 1 -C 6 )haloalkyl, (C 1 -C 6 )haloalkoxy, (C 1 -C 6 )halothioalkoxy, CN, NO 2 , OR 11 , SR 12 , NR 13 R 14 , CO 2 R 15 , CONR 16 R 17 , SO 2 R 18 , SO 2 NR 19 R 20 , COR 21 , NR 22 COR 23 , NR 24 SO 2 R 25 , and R 26 NR 27 R 28 , wherein R 11 to R 28 are such as defined above.
  • “pharmaceutically acceptable” refers to that which is useful in the preparation of a pharmaceutical composition that is generally safe, nontoxic and neither biologically nor otherwise undesirable and that is acceptable for veterinary and human pharmaceutical use.
  • “Pharmaceutically acceptable salt or solvate” of a compound refers to salts and solvates which are pharmaceutically acceptable, as defined herein, and which has the desired pharmacological activity of the parent compound.
  • Acceptable salts for the therapeutic use of the compounds of the present invention include the conventional nontoxic salts of the compounds of the invention such as those formed from pharmaceutically acceptable organic or inorganic acids or from pharmaceutically acceptable organic or inorganic bases.
  • salts derived from inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid and sulfuric acid, and those derived from organic acids such as acetic acid, trifluoroacetic acid, propionic acid, succinic acid, fumaric acid, malic acid, tartaric acid, citric acid, ascorbic acid, maleic acid, glutamic acid, benzoic acid, salicylic acid, toluenesulfonic acid, methanesulfonic acid, stearic acid and lactic acid.
  • salts derived from inorganic bases such as soda, potash or calcium hydroxide and salts derived from organic bases such as lysine or arginine.
  • salts may be synthesized from the compounds of the invention containing a basic or acidic part and the corresponding acids or bases according to conventional chemical methods well known to the person skilled in the art.
  • Acceptable solvates for the therapeutic use of the compounds of the present invention include conventional solvates such as those formed during the last step of the preparation of the compounds of the invention due to the presence of solvents. As an example, mention may be made of solvates due to the presence of water or ethanol.
  • stereoisomer refers to a geometric isomer or an optical isomer.
  • Geometric isomers result from the different position of substituents on a double bond which can then have a Z or E configuration.
  • Optical isomers result notably from the different position in space of substituents on a carbon atom comprising four different substituents. This carbon atom thus constitutes a chiral or asymmetrical center.
  • Optical isomers include diastereoisomers and enantiomers.
  • Optical isomers that are mirror images of each other but are non-superimposable are enantiomers.
  • Optical isomers that are not mirror images of each other are diastereoisomers.
  • tautomer refers to a constitutional isomer of the compound obtained by prototropy, i.e., by migration of a hydrogen atom and a change in location of a double bond.
  • the different tautomers of a compound are generally interconvertible and are in equilibrium in solution in proportions which may vary according to the solvent used, the temperature or the pH.
  • Y 4 N.
  • Y 2 C—X—Ar and Y 3 preferably represents a C—W group.
  • Y 1 CH or N, and advantageously CH,
  • Y 1 and/or Y 4 represent a nitrogen atom.
  • Y 2 and Y 3 preferably do not represent a nitrogen atom.
  • Y 2 CH or C—X—Ar
  • Y 3 C—W or C—X—Ar.
  • X represents a divalent group selected from O, S, S(O), S(O) 2 , NR 4 , CH 2 , CH 2 S, CH 2 S(O), CH 2 S(O) 2 , NHS(O) 2 , SCH 2 , S(O)CH 2 , S(O) 2 CH 2 , S(O) 2 NH, CH 2 CH 2 , CH ⁇ CH, C ⁇ C, CH 2 O, OCH 2 , NR 4 CH 2 , and CH 2 NR 4 .
  • X represents a divalent group selected from S, S(O), S(O) 2 , NR 4 , CH 2 , CH 2 S, CH 2 S(O), CH 2 S(O) 2 , NHS(O) 2 , SCH 2 , S(O)CH 2 , S(O) 2 CH 2 , S(O) 2 NH, CH 2 CH 2 , C ⁇ C, CH 2 O, OCH 2 , NR 4 CH 2 , and CH 2 NR 4 .
  • X may be selected from S, S(O), S(O) 2 , CH 2 , CH 2 S, CH 2 S(O), CH 2 S(O) 2 , NHS(O) 2 , SCH 2 , S(O)CH 2 , S(O) 2 CH 2 , S(O) 2 NH, CH 2 CH 2 , CH ⁇ CH, and C ⁇ C.
  • X may be selected from S, S(O) 2 , CH 2 , SCH 2 , S(O) 2 CH 2 , S(O) 2 NH, CH 2 S, CH 2 S(O) 2 , NHS(O) 2 , CH 2 CH 2 , and C ⁇ C.
  • X may notably be selected from S, S(O), S(O) 2 , NR 4 , CH 2 , SCH 2 , S(O)CH 2 , S(O) 2 CH 2 , S(O) 2 NH, CH 2 CH 2 , C ⁇ C, OCH 2 , and NR 4 CH 2 ; notably from S, S(O) 2 , CH 2 , SCH 2 , S(O) 2 CH 2 , S(O) 2 NH, CH 2 CH 2 , and C ⁇ C, wherein the first atom of these groups is bound to atom C of the C—X—Ar chain.
  • X may be in particular S, S(O) 2 , SCH 2 , S(O) 2 CH 2 , S(O) 2 NH, CH 2 S, CH 2 S(O) 2 , or NHS(O) 2 , and notably S, S(O) 2 , SCH 2 , S(O) 2 CH 2 , or S(O) 2 NH, wherein the first atom of these groups is bound to atom C of the C—X—Ar chain.
  • Ar represents a heteroaryl group, such as pyridine, or an aryl group, such as phenyl, optionally substituted by one or more groups selected from a halogen atom, (C 1 -C 6 )alkyl, (C 1 -C 6 )haloalkyl, (C 1 -C 6 )haloalkoxy, (C 1 -C 6 )halothioalkoxy, CN, NO 2 , OR 11 , SR 12 , NR 13 R 14 , CO 2 R 15 , CONR 16 R 17 , SO 2 R 18 , SO 2 NR 19 R 20 , COR 21 , NR 22 COR 23 , and NR 24 SO 2 R 25 ; and/or optionally fused to a heterocycle.
  • a halogen atom C 1 -C 6 )alkyl, (C 1 -C 6 )haloalkyl, (C 1 -C 6 )haloalkoxy, (C 1 -C 6
  • Ar may represent an aryl group, such as phenyl, optionally substituted by one or more groups selected from a halogen atom, (C 1 -C 6 )alkyl, (C 1 -C 6 )haloalkyl, (C 1 -C 6 )haloalkoxy, (C 1 -C 6 )halothioalkoxy, CN, NO 2 , OR 11 , SR 12 , NR 13 R 14 , CO 2 R 15 , CONR 16 R 17 , SO 2 R 18 , SO 2 NR 19 R 20 , COR 21 , NR 22 COR 23 , and NR 24 SO 2 R 25 .
  • a halogen atom C 1 -C 6 )alkyl, (C 1 -C 6 )haloalkyl, (C 1 -C 6 )haloalkoxy, (C 1 -C 6 )halothioalkoxy, CN, NO 2 , OR 11 , SR 12 ,
  • Ar may notably represent an aryl group, such as phenyl, optionally substituted by one or more groups selected from a halogen atom, (C 1 -C 6 )alkyl, (C 1 -C 6 )haloalkyl, and CONR 16 R 17 , and in particular from a halogen atom such as fluorine, (C 1 -C 6 )alkyl such as methyl, and CONR 16 R 17 such as CONH 2 .
  • a halogen atom such as fluorine
  • C 1 -C 6 )alkyl such as methyl
  • CONR 16 R 17 such as CONH 2 .
  • Ar can also represent a pyridine group.
  • Ar may notably be selected from the following groups:
  • W may advantageously represent an R 5 , SR 5 , OR 5 or NR 5 R 6 group, and preferably R 5 , OR 5 or NR 5 R 6 , with R 5 and R 6 representing, independently of each other, a hydrogen atom or a (C 1 -C 6 )alkyl group.
  • W may represent in particular H, OMe, Me, OH or NH 2 , and notably H.
  • R 3 represents a hydrogen atom.
  • U may represent more particularly a CH 2 or NH group.
  • n may represent 0.
  • V may represent more particularly a C(O) or C(S) group, and advantageously a C(O) group.
  • V C(O) or C(S), and notably C(O), and
  • n 0 or 1, and notably 0.
  • V C(O) or C(S), and notably C(O), and
  • n 0.
  • V C(O) or C(S), and notably C(O), and
  • n 0.
  • R 1 may represent more particularly a hydrogen atom or an NR 7 R 8 group, with R 7 notably representing a hydrogen atom and R 8 notably representing an optionally substituted (C 3 -C 12 )cycloalkyl group or an optionally substituted heterocycle.
  • the (C 3 -C 12 )cycloalkyl group may be in particular a cyclohexyl. It may be substituted by one or more halogen atoms. It may be in particular the group:
  • the heterocyclic group may be in particular a tetrahydropyran, notably unsubstituted. It may thus be the following group:
  • R 1 may thus represent more particularly one of the following groups: H,
  • R 2 may represent in particular an optionally substituted heterocycle, notably substituted by (C 1 -C 6 )alkyl or NH 2 .
  • the heterocycle may be in particular a heterocycle with 5 or 6 members comprising at least one nitrogen atom, and in particular one or two.
  • the heterocycle may thus be selected from piperazine, piperidine and pyrrolidine.
  • R 2 may notably represent one of the following groups: NH 2 , NH(CH 2 ) 3 NMe 2 , NMe(CH 2 ) 3 NMe 2 , NO 2 ,
  • the compounds of the present invention may be selected from the compounds cited in the following table:
  • the present invention also has as an object a compound according to the invention of formula (I) such as defined above, to be used as a drug, notably intended for the treatment of cancer, inflammation and neurodegenerative diseases such as Alzheimer's disease, in particular cancer.
  • the present invention also relates to the use of a compound of formula (I) such as defined above, for the manufacture of a drug, notably intended for the treatment of cancer, inflammation and neurodegenerative diseases such as Alzheimer's disease, in particular cancer.
  • the present invention also relates to a method for the treatment of cancer, inflammation and neurodegenerative diseases such as Alzheimer's disease, in particular cancer, comprising the administration to a person in need thereof of an effective dose of a compound of formula (I) such as defined above.
  • the cancer may be more particularly in this case colon cancer, breast cancer, kidney cancer, liver cancer, pancreatic cancer, prostate cancer, glioblastoma, non-small cell lung cancer, neuroblastoma, inflammatory myofibroblastic tumor, diffuse B-cell lymphoma or anaplastic large-cell lymphoma.
  • the present invention also relates to a compound according to the invention of formula (I) such as defined above, to be used as a drug intended for the treatment of a disease associated with a kinase, and in particular a tyrosine kinase such as the kinases ALK, Abl and/or c-Src, and in particular ALK.
  • a disease associated with a kinase, and in particular a tyrosine kinase such as the kinases ALK, Abl and/or c-Src, and in particular ALK.
  • the disease may be in particular associated with ALK and at least one other kinase, for example Abl or c-Src, in particular ALK and c-Src.
  • the present invention also has as an object a compound according to the invention of formula (I) such as defined above, to be used as a kinase inhibitor, and in particular an inhibitor of tyrosine kinases such as ALK, Abl and/or c-Src, and in particular ALK.
  • the compounds according to the invention may notably be used as an inhibitor of ALK and at least one other kinase, for example Abl or c-Src.
  • the compounds according to the invention are inhibitors of ALK and c-Src.
  • “disease associated with a kinase” or “kinase-associated disease” refers to any diseases, and in particular diseases related to deregulation of cell proliferation, in particular cancers, due to deregulation of the expression or activity of said kinase compared to its normal state of expression or activity. Deregulation of the expression of said kinase may be modification of the sequence expressed or modification of the quantity of protein expressed. These deregulations may lead to changes in cells which may, in particular, result in proliferative disorders including cancers.
  • kinase-associated diseases may be cancers related to deregulation of ALK and/or c-Src activity such as, for example, colon cancer, breast cancer, kidney cancer, liver cancer, pancreatic cancer, prostate cancer, glioblastoma, non-small cell lung cancer, neuroblastoma, inflammatory myofibroblastic tumors, diffuse B-cell lymphoma and anaplastic large-cell lymphoma.
  • cancers related to deregulation of ALK and/or c-Src activity such as, for example, colon cancer, breast cancer, kidney cancer, liver cancer, pancreatic cancer, prostate cancer, glioblastoma, non-small cell lung cancer, neuroblastoma, inflammatory myofibroblastic tumors, diffuse B-cell lymphoma and anaplastic large-cell lymphoma.
  • the expression “inhibitor of kinases” or “kinase inhibitor” refers to molecules that are able to interact with the kinase and to reduce its activity.
  • a kinase inhibitor according to the invention makes it possible to suppress the activity of said kinase.
  • the present invention also relates to a pharmaceutical composition
  • a pharmaceutical composition comprising at least one compound of formula (I) such as defined above, and at least one pharmaceutically acceptable excipient.
  • compositions according to the invention may be formulated notably for oral administration or for injection, wherein said compositions are intended for mammals, including humans.
  • the active ingredient may be administered in unit dosage forms of administration, in mixture with standard pharmaceutical carriers, to animals or to humans.
  • the compounds of the invention as active ingredients may be used in doses ranging between 0.01 mg and 1000 mg per day, given in a single dose once per day or administered in several doses throughout the day, for example twice a day in equal doses.
  • the dose administered per day advantageously is between 5 mg and 500 mg, even more advantageously between 10 mg and 200 mg. It may be necessary to use doses outside these ranges as determined by the person skilled in the art.
  • compositions according to the invention may further comprise at least one other active ingredient, such as an anticancer agent.
  • the present invention also has as an object a pharmaceutical composition comprising:
  • the present invention also relates to a pharmaceutical composition such as defined above to be used as a drug, notably intended for the treatment of cancer, inflammation and neurodegenerative diseases such as Alzheimer's disease, in particular cancer.
  • the present invention also has as an object method for the preparation of the compounds of formula (I) according to the invention.
  • N-protecting group refers to any substituent that protects the NH or NH 2 group against undesirable reactions such as the N-protecting groups described in Greene, “Protective Groups in Organic Synthesis” (John Wiley & Sons, New York (1981)) and Harrison et al., “Compendium of Synthetic Organic Methods”, Vols. 1 to 8 (J. Wiley & Sons, 1971 to 1996).
  • N-protecting groups include carbamates, amides, N-alkylated derivatives, amino acetal derivatives, N-benzylated derivatives, imine derivatives, enamine derivatives and N-heteroatom derivatives.
  • the N-protecting group consists of formyl, acetyl, benzoyl, pivaloyl, phenylsulfonyl, trityl (triphenylmethyl), tert-butyl, benzyl (Bn), t-butyloxycarbonyl (BOC), benzyloxycarbonyl (Cbz), p-methoxybenzyloxycarbonyl, p-nitrobenzyl-oxycarbonyl, trichloroethoxycarbonyl (TROC), allyloxycarbonyl (Alloc), 9-fluorenylmethyloxycarbonyl (Fmoc), trifluoro-acetyl, benzyl carbamates (substituted or not) and the like. It may be in particular a trityl, tert-butyl or BOC group.
  • “leaving group” refers to a chemical group which may be easily displaced by a nucleophile during a nucleophilic substitution reaction, wherein the nucleophile is more particularly an amine, and notably a primary or secondary amine.
  • a leaving group may be more particularly a halogen atom such as a chlorine atom, a mesylate (CH 3 —S(O 2 )O—), a triflate (CF 3 —S(O) 2 O—) or a tosylate (p-Me-C 6 H 4 —S(O) 2 O—).
  • Coupling between (A) and (B) may be carried out by techniques well known to the person skilled in the art.
  • the coupling may be carried out under peptide coupling conditions. It may thus be carried out in the presence of a coupling agent such as diisopropylcarbodiimide (DIC), dicyclohexylcarbodiimide (DCC), 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (EDC), carbonyldiimidazole (CDI), 2-(1H-benzotriazole-1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate (HBTU), 2-(1H-benzotriazole-1-yl)-1,1,3,3-tetramethyluronium tetrafluoroborate (TBTU) or O-(7-azobenzotriazol-1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate (HATU); optionally combined with a secondary coupling agent such as N-hydroxysuccin, DIC,
  • R 30 is a leaving group such as Cl
  • coupling may be carried out in the presence of a base such as pyridine, triethylamine or diisopropylethylamine (DIPEA).
  • a base such as pyridine, triethylamine or diisopropylethylamine (DIPEA).
  • the reaction may be carried out in an aprotic solvent such as tetrahydrofuran, toluene or dichloromethane, or in a base solvent such as pyridine.
  • the compounds of formula (A) and (B) can be prepared by the methods described in further detail below.
  • deprotection refers to the process by which a protecting group is eliminated once the selective reaction is completed. Certain protecting groups may be preferred over others due to their convenience or their relative ease of elimination.
  • the deprotection step may be carried out under conditions well known to the person skilled in the art.
  • substitution step may also be carried out by techniques well known to the person skilled in the art. If necessary, functionalities that may be sensitive to the reaction conditions of the substitution step may be protected beforehand and then deprotected once substitution is carried out.
  • This step may be carried out in the presence of a pharmaceutically acceptable organic or inorganic acid or a pharmaceutically acceptable organic or inorganic base such as defined above.
  • This step is carried out in the presence of a reducing agent such as a borohydride and in particular NaBH 4 , NaBH(OAc) 3 or NaBH 3 CN.
  • a reducing agent such as a borohydride and in particular NaBH 4 , NaBH(OAc) 3 or NaBH 3 CN.
  • This reaction is more particularly carried out at room temperature, i.e., at a temperature ranging between 15° C. and 40° C., in particular between 20° C. and 30° C.
  • the reaction may be typically carried out in a solvent such as dichloroethane (DCE), tetrahydrofuran (THF) or acetonitrile, optionally in the presence of water, acetic acid or trifluoroacetic acid.
  • a solvent such as dichloroethane (DCE), tetrahydrofuran (THF) or acetonitrile, optionally in the presence of water, acetic acid or trifluoroacetic acid.
  • the compounds of formula (A) and (D) can be prepared by the methods described in further detail below.
  • Step (b2) see step (b1)
  • Step (c2) see step (c1)
  • This step may be carried out under conditions well known to the person skilled in the art.
  • a polar or non-polar protic solvent may be more particularly used such as dichloromethane, acetone, acetonitrile, tetrahydrofuran or dioxane.
  • the compounds of formula (A) and (F) can be prepared by the methods described in further detail below.
  • Step (b3) see step (b1)
  • Step (c3) see step (c1)
  • the compound of formula (I) may be separated from the reaction medium by techniques well known to the person skilled in the art, and notably by evaporation of the solvent, crystallization and filtration, etc.
  • the compound obtained may be purified if necessary by techniques well known to the person skilled in the art, and notably by high-performance liquid chromatography (HPLC), silica gel chromatography, recrystallization when the compound is crystalline, etc.
  • HPLC high-performance liquid chromatography
  • silica gel chromatography silica gel chromatography
  • recrystallization when the compound is crystalline etc.
  • compounds of formula (I) are obtained by the preliminary synthesis of compounds of general formula (V) characterized by a halogenated heterobicyclic ring having an exocyclic primary amine. These compounds are obtained via the synthesis of intermediates of general formula (II) or (III).
  • the optionally substituted 2-chloro-5-iodonicotinonitrile (IIa) is obtained from the corresponding hydroxynicotinonitrile by the successive use of an iodination agent such as N-iodosuccinimide (NIS), or molecular iodine with an inorganic base such as, for example, K 2 CO 3 or Na 2 CO 3 , notably in a polar solvent such as hot DMF, followed by treatment with phosphorus oxychloride, pure or diluted in a high boiling-point non-polar solvent, or any other equivalent chlorination agent well known to the person skilled in the art. Reaction temperatures are between ⁇ 20° C. and 200° C.
  • the compound (IIa) thus obtained is then transformed into optionally substituted 5-iodo-pyrazolo[3,4-b]pyridine-3-amine (Va) by its reaction, preferably under heat, in the presence of a hydrazine optionally carrying an N-protecting group such as trityl, tert-butyl or BOC.
  • the brominated analogues of general formula (V) as described in diagram 1a may be obtained by the use of the method described in the following references: Witherington et al., Bioorg. Med. Chem. Lett., 2003, 13, 1577-1580 and Lijuan Chen et al., Bioorg. Med. Chem. Lett., 2010, 20, 4273-4278. For reasons of convenience, these molecules were obtained by the use of the reaction sequence presented in following diagram 3.
  • the optionally functionalized 2-methoxy-nicotinotrile is obtained, for example, by reaction of sodium methanolate in methanol at a temperature between ⁇ 20° C. and the boiling point of the mixture.
  • this compound may be obtained by methylation of 2-hydroxynicotinonitrile or other methods described above.
  • Bromination of 2-methoxy-nicotinonitrile is typically carried out with dibromine in acetic acid at a temperature varying between 20° C. and 110° C.
  • Formation of the pyrazole is typically carried out by reaction of an excess of hydrazine, functionalized or not, at a temperature varying between 20° C. and 100° C.
  • a polar solvent such as water, ethanol, tetrahydrofuran (THF) or any other solvent with comparable properties.
  • a polar solvent such as water, ethanol, tetrahydrofuran (THF) or any other solvent with comparable properties.
  • THF tetrahydrofuran
  • hydrazine in a saline or hydrated form, without solvent is also possible.
  • the optionally functionalized 3-amino-6-iodopyrazine-2-carboxamides are typically obtained in two steps from the corresponding methyl 3-aminopyrazine-2-carboxylates by iodination in the presence of N-iodosuccinimide or molecular iodine optionally in the presence of a cofactor such as KIO 3 , AgCO 2 CF 3 , Ag 2 SO 4 , AlCl 3 , CuCl 2 or HgO, followed by a conversion reaction of the methyl ester function into carboxamide, notably by the use of ammonia in a polar solvent such as water, methanol or THF at temperatures varying between 0° C. and 100° C.
  • a cofactor such as KIO 3 , AgCO 2 CF 3 , Ag 2 SO 4 , AlCl 3 , CuCl 2 or HgO
  • the carboxamide function of the optionally functionalized 3-amino-6-iodopyrazine-2-carboxamide is then converted into nitrile by the use of dehydration agents such as, in particular, CCl 4 /PPh 3 , SOCl 2 , PhSO 2 Cl, P 2 O 5 , TsCl, COCl 2 , DCC/py (N,N′-dicyclohexylcarbodiimide/pyridine) or (COCl) 2 used as the case may be in the presence of an organic base such as pyridine.
  • dehydration agents such as, in particular, CCl 4 /PPh 3 , SOCl 2 , PhSO 2 Cl, P 2 O 5 , TsCl, COCl 2 , DCC/py (N,N′-dicyclohexylcarbodiimide/pyridine) or (COCl) 2 used as the case may be in the presence of an organic base such as pyridine.
  • dehydration agents
  • Deprotection of the dimethylformimidamide function is carried out by treatment with acid such as aqueous hydrochloric acid or any other reagent with equivalent properties. Formation of the pyrazole ring is carried out by a Sandmeyer reaction, well known to the person skilled in the art, followed by a reaction in the presence of a hydrazine, functionalized or not, under conditions as described in the methods above.
  • the diazonium salt, an intermediate of the Sandmeyer reaction may be reduced by the use, for example, of tin chloride in an acid medium or any other equivalent agent, in order to form a hydrazine function that can undergo intramolecular cyclization under the effect of heat.
  • Method A3 aims at obtaining derivatives of general formula (V) featuring a variable function in position 6 of the pyrazolopyridine bicycle. It is detailed in diagram 5 below.
  • the second step of intramolecular cyclization and of alkylation is typically carried out by the heating to a temperature ranging between 20° C. and 100° C. of a solution of the intermediate thioamidate in a polar solvent, for example ethanol in the presence of a suitable alkylating agent such as alkyl halide or dialkyl sulfate.
  • a suitable alkylating agent such as alkyl halide or dialkyl sulfate.
  • the 5-cyano-6-(methylthio)nicotinic acids substituted in position 2 are typically obtained by saponification of the corresponding ethyl esters according to methods well known to the person skilled in the art, notably by the use of hot lithium hydroxide. Decarboxylation of these compounds is carried out by heat treatment in a high boiling-point solvent such as diphenylether at a temperature ranging between 150° C. and 250° C.
  • Halogenation reactions principally aim at obtaining iodinated, brominated or chlorinated derivatives, more particularly iodinated derivatives.
  • the latter are typically obtained by a molecular iodine treatment in the presence of a silver salt such as, for example, Ag 2 SO 4 in a polar solvent such as ethanol at a temperature ranging between 0° C. and 70° C.
  • a silver salt such as, for example, Ag 2 SO 4
  • a polar solvent such as ethanol
  • Alternative methods notably those based on other salts such as KIO 3 , AgCO 2 CF 3 , AlCl 3 , CuCl 2 or HgO, or other iodination agents such as N-iodosuccinimide, are also considered.
  • the conceivable bromination methods typically rely on agents such as N-bromosuccinimide or dibromine according to methods well known to the person skilled in the art.
  • the 5-halo-2-(methylthio) nicotinonitriles obtained are subjected to oxidation of their thiomethoxy function, typically by the use of m-CPBA (m-chloroperbenzoic acid), oxone or any other equivalent agent, to lead to the formation of the corresponding sulfoxide.
  • m-CPBA m-chloroperbenzoic acid
  • oxone oxone
  • These compounds which may contain variable quantities of the corresponding sulfone, are engaged in a reaction in the presence of an optionally substituted hydrazine to form the corresponding 5-halogeno-pyrazolo[3,4-b]pyridin-3-amine carrying a variable functionality in position 6.
  • Method A4 aims at obtaining derivatives of general formula (V) from the compounds of general formula (III) via intermediate formation of compounds of formula (IV). These compounds are typically obtained by the pathway presented in diagram 6.
  • the following references illustrate the method used: Gueiffier et al. Heterocycles, 1999, 51(7), 1661-1667; Gui-Dong Zhu et al. Bioorg. Med. Chem., 2007, 15, 2441-2452.
  • the compounds of general formula (IIIa), acetylated beforehand by one or another of the methods well known to the person skilled in the art, are subjected to the action of isoamyl nitrite, sodium nitrite or any other equivalent organic or inorganic nitrite, in water or acetic acid, for periods typically varying from 1 to 3 days at temperatures varying between 0° C. and 40° C.
  • the compounds of general formula (IVa) thus obtained are deprotected in acidic conditions, for example by the use of hydrochloric acid, before being subjected to the action of nitration agents such as concentrated nitric acid or potassium nitrate in sulfuric acid at temperatures varying between 0° C. and 25° C.
  • nitropyrazoles thus obtained are typically reduced into aminopyrazoles of general formula (Ve) by the use of SnCl 2 in hydrochloric acid.
  • Alternative methods include the use of iron, zinc or tin in acidic conditions and methods of catalytic hydrogenation in the presence of complexes of platinum, nickel or Pd/C under an atmosphere of hydrogen or in the presence of equivalent agents such as cyclohexadiene, cyclohexene, sodium borohydride or hydrazine.
  • the compounds of formula (I) are obtained by the preliminary synthesis of compounds of general formula (VI) characterized by a functionalized heterobicyclic ring possessing an exocyclic amine. These compounds are obtained via the synthesis of intermediates of general formula (VI).
  • Method B1 is represented in diagram 7 below, with W notably representing H, (C 1 -C 6 )alkyl, aryl or benzyl.
  • the 3-nitro-6-thioxo-1,6-dihydropyridin-2-carbonitrile and 3-nitro-6-thioxo-1,6-dihydropyrazine-2-carbonitrile derivatives, optionally functionalized in position 5, are typically obtained from the corresponding 2,6-dichloro-3-nitropyridines or 2,6-dichloro-3-nitropyrazines by the successive reactions of a cyanide salt, such as copper cyanide, in a high boiling-point polar solvent such as N-methylpyrrolidone at temperatures ranging between 100° C. and 200° C.; followed by the reaction of aqueous sodium hydrosulfite in a polar solvent.
  • a cyanide salt such as copper cyanide
  • a high boiling-point polar solvent such as N-methylpyrrolidone
  • a substituted benzyl bromide in basic medium, according to methods well known to the person skilled in the art.
  • the preferred protocol includes the use of an aprotic and anhydrous polar solvent such as acetone carrid at its boiling point and an organic base such as pyridine, triethylamine or DIPEA, or an inorganic base such as sodium, potassium or calcium carbonate. Reactions for reducing the nitro function in amine are preferentially carried out by the use of SnCl 2 in hydrochloric acid.
  • Alternative methods include the use of iron, zinc or tin in acidic conditions and methods of catalytic hydrogenation in the presence of complexes of platinum, nickel or Pd/C under an atmosphere of hydrogen or in the presence of equivalent agents such as cyclohexadiene, cyclohexene, sodium borohydride or hydrazine.
  • the product of the reduction reaction in addition to having a primary amine, has a carboxamide function resulting from hydrolysis of the nitrile function.
  • isolation of the corresponding 3-aminopicolinonitriles or 3-aminopyrazine-2-carbonitriles may be carried out by dehydration of the carboxamide into nitrile via the use of phosphorus oxychloride in the presence of DMF or any other method well known to the person skilled in the art.
  • aminopyrazole ring is carried out preferentially by the formation of a diazonium, obtained by the successive reaction at low temperature of isoamyl nitrite, sodium nitrite or any other equivalent organic or inorganic nitrite, in water, hydrochloric acid, acetic acid or sulfuric acid, at temperatures varying between 0° C. and 20° C., followed by its reduction into hydrazine and intramolecular cyclization activated by heating of the reaction medium.
  • the reduction reaction is preferentially carried out with tin chloride in acidic conditions but may also be carried out by catalytic hydrogenation or any other method well known to the person skilled in the art.
  • the intermediate diazonium undergoes a Sandmeyer reaction during which this functional group is substituted by a halogen atom, such as iodine, by the reaction of an adequate salt, such as NaI.
  • a halogen atom such as iodine
  • an adequate salt such as NaI.
  • formation of the aminopyrazole ring is carried out by the use of a hydrazine, functionalized or not, in a polar solvent such as ethanol at temperatures varying between 25° C. and 150° C.
  • nucleophiles used are phenols, thiophenols, benzyl alcohols or thiobenzyl alcohols as well as anilines or benzylamines, functionalized or not.
  • the 6-chloro-3-nitropicolinonitriles and 6-chloro-3-nitropyrazine-2-carbonitriles, optionally substituted in position 5, are reacted in the presence of the suitable nucleophile, alcohol or thiol, in a polar solvent such as acetonitrile in the presence of an inorganic base such as potassium or sodium carbonate.
  • Solvents such as DMSO (dimethylsulfoxide), DMF (dimethylformamide), acetone, THF (tetrahydrofuran) or pyridine may also be considered. If necessary, these reactions may be catalyzed by the action of copper and may also be carried out without solvent.
  • the preferred protocol involves temperatures ranging between 20° C. and 150° C.
  • bases such as pyridine, DIPEA, diisopropylamine, triethylamine, DBU, potassium tert-butylate, NEt 3 or NaH is also possible.
  • X N
  • toluene is a preferred solvent and triethylamine (NEt3) the base of choice.
  • Method B3 is a variant of method B2 characterized by a first step resulting from a catalytic coupling reaction between a benzyl boronate, in acid or ester form, and a 6-chloro-3-nitropicolinonitrile or 6-chloro-3-nitropyrazine-2-carbonitrile derivative. It is also well known to the person skilled in the art that catalytic coupling reactions using alternative catalysts and benzyl derivatives are also possible. Among these, the Stille reaction, based on tin complexes, or those based on organozinc compounds may be considered.
  • This compound is brought together with a 6-chloro-3-nitropicolinonitrile, a 6-chloro-3-nitropyrazine-2-carbonitrile optionally substituted in position 5 or a 5-chloro-2-nitronicotinonitrile optionally substituted in position 6 and a palladium catalyst such as Pd(dppf)Cl 2 or Pd(PPh 3 ) 4 , an organic base such as triethylamine or an alcoholate, or an inorganic base such as sodium, potassium or cesium carbonate in a solvent such as toluene, benzene, THF or dioxane.
  • the preferred reaction temperatures are between 20° C. and 100° C.
  • ethyl 2-chloro-5-(chlorosulfonyl)nicotinate derivatives required for this reaction sequence may be obtained according to the methods described by Levett P. C. et al., Org. Proc. Res. Dev., 2002, 6(6), 767-772; WO 01/98284 and WO 2008/010964.
  • sulfonamides is typically carried out by mixing the 2-chloro-5-(chlorosulfonyl)nicotinate of interest with a primary or secondary aniline, optionally functionalized, in an aprotic solvent such as dichloromethane, THF, acetone or acetonitrile in the presence of an organic base such as triethylamine (NEt 3 ), pyridine or DIPEA.
  • an organic base such as triethylamine (NEt 3 ), pyridine or DIPEA.
  • an inorganic base such as sodium or potassium carbonate may also be considered.
  • the optimal reaction temperatures are between 0° C. and 70° C.
  • the corresponding acid chlorides are prepared by treatment with thionyl chloride in toluene under reflux or by any other dehydrochlorination method well known to the person skilled in the art.
  • the reaction of these intermediates with aqueous ammonia makes it possible to form optionally functionalized 2-chloro-5-(N-phenylsulfamoyl)nicotinamides which are then engaged in a dehydration reaction, notably by the use of POCl 3 , at a temperature ranging between 75° C. and 150° C.
  • POCl 3 notably by the use of POCl 3
  • the alternative use of agents such as P 2 O 5 or trifluoroacetic anhydride and pyridine may also be considered.
  • the method described below is inspired by the work of J. Baldwin et al., J. Heterocyclic. Chem., 1980, 17(3), 445-448.
  • the 5-hydroxynicotinonitrile derivatives, optionally functionalized in position 6, are alkylated, typically by the use of an optionally functionalized benzyl halide in the presence of a base.
  • the preferred method requires the use of an aprotic polar solvent such as DMF and a base such as NaH.
  • the optimal reaction temperatures are between 20° C. and 100° C.
  • the solvents which may be used include, for example, THF, DMSO, dioxane, acetonitrile, dichloromethane or acetone and bases such as t BuOK, DIPEA, pyridine, triethylamine, DBU or sodium, potassium or cesium carbonate.
  • Oxidation of the pyridine ring into pyridine-N-oxide is typically carried out by use of m-CPBA in dichloromethane at room temperature. Nevertheless, many alternative methods are conceivable, notably those based on the use of sodium percarbonate in the presence of a rhenium catalyst, sodium perborate in the presence of acetic acid or the urea-hydrogen peroxide complex.
  • Le method described below consists in forming a sulfonamide function from an aromatic amine and an arylsulfonyl halide, or any other equivalent reagent, in the presence of a base, which can optionally be introduced as solvent or co-solvent.
  • a base which can optionally be introduced as solvent or co-solvent.
  • the arylsulfonyl halide or its equivalent can be generated in situ.
  • Method C aims at the preparation of compounds of general formula (XI) as described in diagram 1.
  • This method can also be used to carry out the synthesis of molecules comprising a diatomic X group corresponding notably to an ArX group representing: —ArCH 2 NH—, —ArCH 2 N(R 4 )—, —ArCH 2 O—, —ArCH 2 S—, —ArCH 2 CH 2 —, —ArCHCH—, or —ArCC—.
  • 6-hydroxy-2-(methylthio)nicotinonitriles or 5-hydroxy-3-(methylthio)pyrazine-2-carbonitriles are subjected to a dehydrochlorination reaction, typically in the presence of phosphorus oxychloride, with or without solvent, at temperatures varying between 70° C. and 180° C. If a solvent is used, a high boiling-point non-polar solvent such as toluene or xylene will be preferred.
  • the 6-chloro-2(methylthio)nicotinonitriles and 5-chloro-3-(methylthio)pyrazine-2-carbonitriles respectively obtained, or their sulfonic ester analogues if this option is preferred, are then reacted with a nucleophile such as a phenol, an aniline or a thiophenol in the context of aromatic nucleophilic substitution.
  • a nucleophile such as a phenol, an aniline or a thiophenol in the context of aromatic nucleophilic substitution.
  • the reaction is carried out in a polar solvent such as DMSO, DMF, acetone, THF or acetonitrile, in the presence of a base such as potassium tert-butylate or NaH. If necessary, these reactions may be catalyzed by the action of copper and may also be carried out without solvent.
  • the preferred protocol involves temperatures ranging between 20° C. and 150° C.
  • organic bases such as pyridine, diisopropylamine, triethylamine or DBU, or inorganic bases such as sodium or potassium carbonate is also possible.
  • the compounds of formula (IXb) may give rise to a catalytic coupling reaction such as a Suzuki reaction.
  • these compounds are brought together with an optionally substituted 2-benzyl-4,4,5,5-tetramethyl-1,3,2-dioxaborolane already described in preceding method B3, a palladium catalyst such as Pd(dppf)Cl 2 or Pd(PPh 3 ) 4 , an organic base such as triethylamine or an alcoholate, or an inorganic base such as sodium, potassium or cesium carbonate in a solvent such as toluene, benzene, THF or dioxane.
  • the preferred reaction temperatures are between 20° C. and 100° C.
  • the derivatives obtained by one or another of these methods are then oxidized, typically by the use of m-CPBA or oxone to form the corresponding methyl sulfoxides or methyl sulfones.
  • These compounds, sometimes obtained as mixtures, are used as-is in the aminopyrazole ring formation reaction by use of an optionally substituted hydrazine in a polar solvent such as ethanol at temperatures varying between 25° C. and 150° C.
  • reaction sequence notably by reversing the synthesis steps.
  • 6-hydroxy-4-(methylthio)nicotinonitrile or 6-hydroxy-4-(methylthio)pyridazin-3-carbonitrile derivatives are oxidized, typically by the use of m-CPBA or oxone to form the corresponding methyl sulfoxides or methyl sulfones.
  • These compounds, sometimes obtained as mixtures, are used as-is in the aminopyrazole ring formation reaction by use of an optionally substituted hydrazine in a polar solvent such as ethanol at temperatures varying between 25° C. and 150° C.
  • pyrazolopyridines and pyrazolopyridazines thus obtained are subjected to a dehydrochlorination reaction, typically in the presence of phosphorus oxychloride, with or without solvent, at temperatures varying between 70° C. and 180° C. If a solvent is used, a high boiling-point non-polar solvent such as toluene or xylene will be preferred.
  • the optionally substituted 6-chloro-pyrazolo[4,3-c]pyridin-3-amine and 6-chloro-pyrazolo[4,3-c]pyridazin-3-amine respectively obtained are then reacted with a nucleophile such as a phenol, an aniline or a thiophenol in the context of aromatic nucleophilic substitution.
  • a nucleophile such as a phenol, an aniline or a thiophenol in the context of aromatic nucleophilic substitution.
  • the reaction is carried out in a polar solvent such as DMSO, DMF, acetone, THF or acetonitrile, in the presence of a base such as potassium tert-butylate or NaH. If necessary, these reactions may be catalyzed by the action of copper and may also be carried out without solvent.
  • the preferred protocol involves temperatures ranging between 20° C. and 150° C.
  • organic bases such as pyridine, diisopropylamine, triethylamine or DBU, or inorganic bases such as sodium or potassium carbonate is also possible.
  • the compounds of formula (XIVa) may give rise to a catalytic coupling reaction such as a Suzuki reaction.
  • these compounds are brought together with an optionally substituted 2-benzyl-4,4,5,5-tetramethyl-1,3,2-dioxaborolane described above in preceding method B3, a palladium catalyst such as Pd(dppf)Cl 2 or Pd(PPh 3 ) 4 , an organic base such as triethylamine or an alcoholate, or an inorganic base such as sodium, potassium or cesium carbonate in a solvent such as toluene, benzene, THF or dioxane.
  • the preferred reaction temperatures are between 20° C. and 100° C.
  • Method C3 is a variant of method C1 based on the regioselective functionalization of 2,6-dichloronicotinonitrile either by an anionic nucleophile such as a phenate or a thiophenate, or by an organometallic such as a benzylzinc chloride.
  • an anionic nucleophile such as a phenate or a thiophenate
  • an organometallic such as a benzylzinc chloride.
  • the reaction is catalyzed for example with a palladium(II) complex.
  • Method D1 makes use of the coupling reaction as described in J.A.C.S., 1984, 106, 158 between an organozinc compound prepared in situ and an aryl bromide catalyzed by palladium complexes.
  • the optionally substituted 3-amino-diazaindazoles or 3-amino-azaindazoles are brought together with a zinc benzyl chloride, optionally substituted, in an aprotic polar solvent such as THF or dioxane, in the presence of a catalytic quantity of a palladium complex such as (dppf) 2 PdCl 2 .CH 2 Cl 2 .
  • the coupling reaction is carried out at temperatures ranging between 25° C. and 100° C.
  • Method D2 makes use of the coupling reaction as described by Gueiffier A. et al., Tetrahedron, 2006, 62, 6042-6049, between a thiol, in particular a thiophenol or a benzylthiol, and an aryl iodide catalyzed by copper complexes.
  • This reaction is typically carried out in a high boiling-point polar solvent such as 2-propanol in the presence of a catalytic quantity of polyethylene glycol, a metal salt such as copper iodide (CuI) and an excess of an inorganic base such as potassium carbonate, calcium carbonate or sodium carbonate.
  • a high boiling-point polar solvent such as 2-propanol
  • a metal salt such as copper iodide (CuI)
  • an inorganic base such as potassium carbonate, calcium carbonate or sodium carbonate.
  • Method D3 makes use of the coupling reaction as described by Sonogashira, K. et al. in Tetrahedron Lett., 1975, 16, 4467-4470 between an acetylene derivative and an aryl halide catalyzed by copper and palladium complexes.
  • Such a reaction is typically carried out by the reaction under an inert atmosphere of a heteroaryl halide with a stoichiometric quantity of an optionally substituted ethynylbenzene in the presence of a catalytic quantity of a palladium complex, for example PdCl 2 (PPh 3 ) 2 or Pd(PPh 3 ) 4 , a catalytic quantity of a copper salt, for example CuI, and an organic base such as triethylamine or DIPEA, or an inorganic base such as potassium or cesium carbonate.
  • the protocol generally involves reaction temperatures ranging between 20° C. and 45° C. in solvents including DMF, THF, dioxane or diethyl ether.
  • the protocols of method E aim at functionalizing the exocyclic amine of aminopyrazole rings by their reaction with an intermediate featuring an electrophile function, optionally generated in situ, such as acid chloride, an isocyanate, a isothiocyanate or an aldehyde.
  • Method E1 presented in diagram 16 below, aims at the transformation of the primary exocyclic amine function of aminopyrazole compounds into an amide function.
  • These compounds are synthesized via the corresponding 3-aminopyrazole by the addition of adequate acid chloride prepared beforehand by the use of oxalyl chloride and a catalytic quantity of DMF in a solvent such as tetrahydrofuran.
  • acid chlorides may be obtained by the use of alternative methods, such as those based on the use of thionyl chloride or phosphorus oxychloride, well known to the person skilled in the art.
  • the condensation of acid chlorides on aminopyrazoles is typically carried out in an aprotic solvent such as tetrahydrofuran, toluene or dichloromethane in the presence of a base such as DIPEA, pyridine or triethylamine.
  • a base as a solvent, in particular pyridine, is a possibility.
  • this type of reaction may be conducted in a biphasic system according to the well-known Schotten-Baumann method.
  • formation of the amide bond may be carried out from the corresponding 3-aminopyrazole and the acid of interest by the use of peptide coupling conditions using reagents such as HOBt (hydroxybenzotriazole), TBTU (O-(benzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium tetrafluoroborate), HATU (2-(1H-7-azabenzotriazol-1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate), EDCI (1-ethyl-3-(3-dimethylaminopropyl)carbodiimide) or carbonyldiimidazole at a temperature ranging between ⁇ 20° C. and 100° C. in an aprotic solvent such as tetrahydrofuran, dioxane, dichloromethane or any solvent with similar properties.
  • reagents such as HOBt (hydroxybenzotriazole
  • Reducing amination reactions are typically carried out by mixing adequate stoichiometric quantities of aminopyrazole and aldehyde in a solvent such as DCE (dichloroethane), THF or acetonitrile, optionally in the presence of a quantity of water, TFA (trifluoroacetic acid) or acetic acid, by adding successive fractions of a reducing agent such as NaBH 4 , NaBH(OAc) 3 or NaBH 3 CN. These reactions are typically carried out at room temperature.
  • a solvent such as DCE (dichloroethane), THF or acetonitrile
  • TFA trifluoroacetic acid
  • a reducing agent such as NaBH 4 , NaBH(OAc) 3 or NaBH 3 CN.
  • the reaction mixture is prepared in a polar or non-polar aprotic solvent such as dichloromethane, acetone, DMF, DMA, acetonitrile, THF or dioxane carried at temperatures varying between 20° C. and the boiling point of the chosen solvent.
  • a polar or non-polar aprotic solvent such as dichloromethane, acetone, DMF, DMA, acetonitrile, THF or dioxane carried at temperatures varying between 20° C. and the boiling point of the chosen solvent.
  • a weakly nucleophilic organic or inorganic base may prove to be necessary.
  • sodium hydride is a possible option.
  • the trifluoroacetate protecting groups are removed by the action of an organic base such as triethylamine or pyridine in a polar solvent such as methanol, ethanol or THF at the reflux temperatures of the solvents used.
  • an organic base such as triethylamine or pyridine
  • a polar solvent such as methanol, ethanol or THF
  • tert-butyl or trityl protecting groups carried by the pyrazole rings are displaced by the action of a strong acid, typically TFA, in a non-polar solvent such as dichloromethane or DCE.
  • Reactions for reducing diaryl alkynes into diaryl alkanes are typically carried out by catalytic hydrogenation, under hydrogen pressure, in the presence of catalysts such as PtO 2 , Pt, Pd/C, Ni or Rh.
  • catalysts such as PtO 2 , Pt, Pd/C, Ni or Rh.
  • DIBAL-H diisobutylaluminum hydride
  • Oxidation reactions of sulfides into sulfoxides are typically carried out via the use of oxone in a mixture of polar solvents such as THF/MeOH or DMF/water.
  • the optimal reaction temperatures are typically between 25° C. and 50° C.
  • H 2 O 2 and metal complexes such as Sc(OTf) 3 promotes partial oxidation derivatives.
  • N-iodosuccinimide 1.5 equivalents of N-iodosuccinimide are added at room temperature to 5 g (32.7 mmol) of a methyl 3-aminopyrazine-2-carboxylate solution in 25 ml of dimethylformamide.
  • the reaction medium is heated at 65° C. for 1 hour, added together with 0.5 equivalents of N-iodosuccinimide and maintained at 65° C. for 24 hours. After returning to room temperature, the solvent is evaporated and then the product is extracted several times with dichloromethane.
  • Ethyl 5-cyano-2-hydroxy-6-(methylthio)nicotinate is obtained by following the procedure of Ya. Yu. Yakunin et al., Russian Chemical Bulletin, 1999, 48(1), 195-6 with a total yield of 34%.
  • 4-methylmorpholinium (2,4)-ethyl-5-amino-2,4-dicyano-5-mercaptopenta-2,4-dienoate is prepared according to the procedure described by V. D. Dyachenko et al., Chemistry of Heterocyclic Compounds, 2005, 41(4), 503-10 with a yield of 50%.
  • 2,6-Dichloro-3-nitropyridine (5.18 mmol, 1 g) is mixed with 5 ml of N-methyl-2-pyrrolidinone in a microwave reactor. The reaction mixture is heated at 180° C. for 15 minutes (6 bars). The crude reaction product is dissolved in ethyl acetate, filtered and washed several times using an aqueous phase. The organic phase is collected, dried on magnesium sulfate and dry concentrated. The crude product thus obtained is purified by silica gel chromatography (heptane/AcOEt) to yield, after concentration, 0.62 g (65%) of a brown oil.
  • a solution cooled to 0° C. of NaNO 2 in 3 ml of water is added drop by drop to a solution at 0° C. of 3-amino-6-(3,5-difluorobenzylthio)picolinonitrile (1.587 mmol, 0.44 g) in 15 ml of 6 N HCl solution.
  • a solution cooled to 0° C. of SnCl 2 .2H 2 O diluted in 4 ml of 12 N HCl is added drop by drop.
  • the reaction medium is then stirred at 25° C. for 1 hour.
  • the solution is extracted with ethyl acetate and then washed using saturated NaHCO 3 solution and then saturated NaCl solution.
  • the organic phase is collected, dried on magnesium sulfate and then concentrated under reduced pressure.
  • the residue is purified by silica gel chromatography (AcOEt/heptane) to yield, after concentration of the organic phases, 0.07 g (15%) of black crystals.
  • a mixture of 6-chloro-3-nitropicolinonitrile (3.70 g, 0.02 mol), 3,5-dichlorobenzenethiol (3.60 g, 0.02 mol) and K 2 CO 3 (5.6 g, 0.04 mol) in 100 ml of acetonitrile is carried at 70° C. for 16 hours.
  • the crude reaction product is diluted in an ethyl acetate fraction and washed using an aqueous phase.
  • the organic phase is dried with sodium sulfate and the residue is purified by silica gel chromatography (AcOEt/petroleum ether) to yield 5.4 g (80%) of a yellow solid.
  • a solution of 350 mg of NaNO 2 (5.07 mmol) in water (2 ml) is added to a stirring solution of 1.5 g of 3-amino-6-(3,5-dichlorophenylthio)picolinonitrile (5.07 mmol) in 100 ml of 50% sulfuric acid at 0° C. The mixture is stirred for 20 minutes at 0-5° C.
  • a solution of 2.9 g of SnCl 2 .2H 2 O (12.7 mmol, 2.5 eq) in hydrochloric acid (12 N solution, 10 ml) is then added and the solution is stirred for 1 hour at room temperature. The solid formed is filtered and then washed twice with 20 ml of water.
  • the solid is suspended in 100 ml and the pH is adjusted to 10 by adding 30% soda solution.
  • the organic phase is separated and then dried on anhydrous sodium sulfate before being concentrated under vacuum.
  • a light yellow solid is obtained after recrystallization in ethyl acetate (470 mg, 34%).
  • a solution of 713 mg of NaNO 2 (10.3 mmol) in water (5 ml) is added, drop by drop, to a stirring solution of 2.3 g of 3-amino-6-(3,5-difluorophenylamino)picolinonitrile (9.4 mmol) in 100 ml of 6 N hydrochloric acid at 0° C.
  • the mixture is stirred for 20 minutes at 0-5° C.
  • a solution of 5.3 g of SnCl 2 .2H 2 O (23.5 mmol, 2.5 eq) in hydrochloric acid (12 N solution, 30 ml) is then added drop by drop and the solution is stirred for 1 hour at room temperature.
  • the reaction medium is then cooled at 0° C.
  • This compound can be prepared from the following intermediates, according to method B3.
  • 0.288 ml (3.87 mmol) of thionyl chloride and a drop of DMF are added successively to 0.450 g (1.29 mmol) of 2-chloro-5-(N-(3,5-difluorophenyl)sulfamoyl)nicotinic acid in 5 ml of anhydrous toluene.
  • the mixture is placed under stirring, at reflux of toluene, for 2 hours.
  • the acid chloride reaction mixture is then added drop by drop to an iced solution, under stirring, of 4.5 ml of 25% ammonium hydroxide. A release of gas is observed.
  • the reaction medium is left under stirring at room temperature for 30 minutes.
  • the reaction medium is extracted several times with ethyl acetate.
  • This compound can be prepared from the following intermediates, according to method B5.
  • a mixture of 1 g of 5-methoxynicotinonitrile (7.46 mmol) and 8.62 g of pyridine hydrochloride is heated at 200° C. for 2 hours.
  • the crude reaction product is diluted in a water fraction several times with diethyl ether.
  • the aqueous phase is basified by adding sodium bicarbonate and then extracted again with diethyl ether.
  • the organic phase is dried and then concentrated to yield 850 mg of 5-hydroxynicotinonitrile (95%) in the form of a beige solid.
  • This compound can be prepared from the following intermediates, according to method C1.
  • the product is purified on a silica gel column (eluent: cyclohexane/dichloromethane 5:5) to yield 5.31 g (85%) of 5-cyano-6-(methylthio)pyridin-2-yl trifluoromethanesulfonate in the form of a yellow solid.
  • the medium is degassed for 5 minutes under argon before adding 0.25 g (0.06 eq) of de 2,2′-bis(diphenylphosphino)-1,1′-binaphthyl and 0.08 g (0.04 eq) of (1E,4E)-1,5-diphenylpenta-1,4-dien-3-one, palladium(II) complex.
  • the reaction medium is stirred at 100° C. for 2 hours. After return to room temperature, ethyl acetate and brine are added. The organic phase is dried on anhydrous magnesium sulfate, filtered and evaporated.
  • a solution of 362 mg (1.05 eq) of potassium hydroxide in 10 mL of ethanol is added, under nitrogen, to a solution of 698 ⁇ L (6.16 mmol) of 2,4-difluorobenzenethiol in 30 mL of ethanol.
  • the reaction medium is stirred at room temperature for 15 minutes and then cooled in ice before adding a solution of 1.015 g (0.95 eq) of 2,6-dichloronicotinonitrile in 30 mL of ethanol.
  • the reaction medium is stirred for 2 hours at 0-5° C.
  • 63 mL of a 0.1N HCl solution is added to stop the reaction. Water is added and the producted is extracted with ethyl acetate.
  • the product thus obtained is dissolved in 10 mL of dichloromethane at 0° C. and 56 mg (0.5 mmol) of TFA is added.
  • the reaction medium is stirred for 4 hours. Water is added and the pH of the reaction medium is adjusted to 7 with a NaHCO 3 solution.
  • the aqueous phase is collected, basified (pH 9-10) with a concentrated K 2 CO 3 solution and extracted with dichloromethane.
  • the protocols comprising method E aim at functionalizing the exocyclic amine of the aminopyrazole rings by their reaction with an intermediate featuring an electrophilic function, optionally generated in situ, such as acid chloride, isocyanate, isothiocyanate or aldehyde.
  • This compound was obtained by reproducing example 16d using tert-butyl (S)-pyrrolidin-3-ylcarbamate.
  • This compound can be prepared from the following intermediates.
  • This compound can be prepared from the following intermediates.
  • This compound can be prepared from the following intermediates.
  • platine (IV) oxide 2.12 mmol
  • platine (IV) oxide 0.48 g
  • a solution of 26.8 g of 4-(4-(tert-butoxycarbonyl)-3-nitrophenyl)-1-methylpyridinium iodide 60.6 mmol
  • the reaction medium is brought under 5 bar of hydrogen for 24 h.
  • the catalyst is filtered and the filtrate is concentrated under reduced pressure to yield 24.8 g (98%) of white crystals.
  • This compound was prepared by adapting the method described in EP1215208.
  • This compound can be prepared from the following intermediates.
  • This compound can be prepared from the following intermediates.
  • the acid chloride is dissolved in 35 ml of anhydrous tetrahydrofuran at ⁇ 20° C. and then the solution formed is added to a solution containing 1.56 g (5.61 mmol) of 5-(3,5-difluorophenylthio)-1H-pyrazolo[3,4-b]pyridin-3-amine and 3.71 ml (21.30 mmol) of N-ethyl-N-isopropylpropan-2-amine in 30 ml of anhydrous tetrahydrofuran. The reaction mixture is stirred for 3 hours at ⁇ 20° C. and then overnight at room temperature.
  • 26-8 13.59 (1H, sl, NH), 11.05 (1H, sl, NH), 8.68 (1H, d, CH arom ), 8.57 (1H, d, CH arom ), 7.19 (2H, d, CH arom ), 6.99-7.08 (1H, m, CH arom ), 6.88 (2H, d, CH arom ), 6.75-6.79 (2H, m, CH arom ), 3.61 (2H, m, CH 2 ), 3.07-3,09 (4H, m, CH), 2.41-2.44 (4H, m, CH), 2.20 (3H, s, CH 3 ).
  • the major product of these reactions corresponds to the disubstituted product characterized by the additional functionalization of the pyrazole ring.
  • this product is isolated and transformed into a monosubstituted product by treatment with a base as described below.
  • the acid chloride is added at 0° C. in small fractions to 1 g (3.58 mmol) of 5-(3,5-difluorophenylthio)-1H-pyrazolo[3,4-b]pyrazine-3-amine dissolved in 15 ml of pyridine.
  • the reaction mixture is stirred at 25° C. overnight at room temperature.
  • reaction medium is heated at 65° C. for 4 hours, and then overnight at room temperature. After evaporation of the solvent, the product is extracted several times with ethyl acetate. The organic phases are combined, washed with saturated sodium bicarbonate solution, dried on magnesium sulfate and concentrated.
  • Reactions carried out in pyridine often make it possible to modify the regioisomer distribution of the products.
  • the following example is characteristic of a reaction of this type.
  • the acid chloride is dissolved in 5 ml of anhydrous pyridine and then the solution formed is added to a solution of 0.214 g (0.658 mmol) of 3-amino-N-(3,5-difluorophenyl)-1H-pyrazolo[3,4-b]pyridine-5-sulfonamide in 5 ml of pyridine at 0° C.
  • the reaction mixture is stirred for 3 hours at 0° C., and then overnight at room temperature.
  • the pyridine is evaporated and the crude reaction product is redissolved in toluene and then dry concentrated.
  • the reaction mixture is diluted with saturated NaHCO 3 solution and extracted with ethyl acetate.
  • the organic phase is dried on MgSO 4 , filtered and concentrated and the crude product is used directly in the deprotection reaction with no purification or characterization.
US13/823,658 2011-01-27 2012-01-27 Derivatives of azaindazole or diazaindazole type as medicament Abandoned US20130172360A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
FR1150651A FR2970967B1 (fr) 2011-01-27 2011-01-27 Derives de type azaindazole ou diazaindazole comme medicament
FR1150651 2011-01-27
PCT/EP2012/051283 WO2012101239A1 (fr) 2011-01-27 2012-01-27 Dérivés de type azaindazole ou diazaindazole utilisés comme médicaments

Publications (1)

Publication Number Publication Date
US20130172360A1 true US20130172360A1 (en) 2013-07-04

Family

ID=44318089

Family Applications (2)

Application Number Title Priority Date Filing Date
US13/823,658 Abandoned US20130172360A1 (en) 2011-01-27 2012-01-27 Derivatives of azaindazole or diazaindazole type as medicament
US13/686,334 Active US8883821B2 (en) 2011-01-27 2012-11-27 Substituted pyrazolo[3,4-b]pyridines as medicaments

Family Applications After (1)

Application Number Title Priority Date Filing Date
US13/686,334 Active US8883821B2 (en) 2011-01-27 2012-11-27 Substituted pyrazolo[3,4-b]pyridines as medicaments

Country Status (30)

Country Link
US (2) US20130172360A1 (fr)
EP (1) EP2668184B1 (fr)
JP (1) JP5931926B2 (fr)
KR (2) KR20180122028A (fr)
CN (1) CN103339129B (fr)
AR (1) AR084935A1 (fr)
AU (1) AU2012210467B2 (fr)
CA (1) CA2823824C (fr)
CY (1) CY1120088T1 (fr)
DK (1) DK2668184T3 (fr)
ES (1) ES2661695T3 (fr)
FR (1) FR2970967B1 (fr)
HK (1) HK1186737A1 (fr)
HR (1) HRP20180524T1 (fr)
HU (1) HUE037153T2 (fr)
IL (2) IL227496B (fr)
LT (1) LT2668184T (fr)
MA (1) MA34903B1 (fr)
MX (1) MX356411B (fr)
MY (1) MY180666A (fr)
PL (1) PL2668184T3 (fr)
PT (1) PT2668184T (fr)
RS (1) RS57101B1 (fr)
RU (1) RU2600976C2 (fr)
SI (1) SI2668184T1 (fr)
TN (1) TN2013000293A1 (fr)
TR (1) TR201802944T4 (fr)
TW (1) TWI546303B (fr)
UA (1) UA109698C2 (fr)
WO (1) WO2012101239A1 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20170317082A1 (en) * 2016-04-27 2017-11-02 International Business Machines Corporation Field effect transistor structure for reducing contact resistance

Families Citing this family (28)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012088266A2 (fr) 2010-12-22 2012-06-28 Incyte Corporation Imidazopyridazines et benzimidazoles substitués en tant qu'inhibiteurs de fgfr3
FR2970967B1 (fr) * 2011-01-27 2013-02-15 Pf Medicament Derives de type azaindazole ou diazaindazole comme medicament
EP2817292B1 (fr) 2012-02-22 2019-12-18 Sanford-Burnham Medical Research Institute Composés sulfonamides et leurs utilisations en tant qu'inhibiteurs de tnap
LT3495367T (lt) 2012-06-13 2021-02-25 Incyte Holdings Corporation Pakeistieji tricikliniai junginiai, kaip fgfr inhibitoriai
EP2689779A1 (fr) * 2012-07-27 2014-01-29 Pierre Fabre Medicament Dérivés de type azaindazole ou diazaindazole pour traiter un cancer surexprimant la trk
EP2689778A1 (fr) * 2012-07-27 2014-01-29 Pierre Fabre Medicament Dérivés de type azaindazole ou diazaindazole pour le traitement de la douleur
WO2014026125A1 (fr) * 2012-08-10 2014-02-13 Incyte Corporation Dérivés de pyrazine en tant qu'inhibiteurs de fgfr
US9266892B2 (en) 2012-12-19 2016-02-23 Incyte Holdings Corporation Fused pyrazoles as FGFR inhibitors
MY181497A (en) 2013-04-19 2020-12-23 Incyte Holdings Corp Bicyclic heterocycles as fgfr inhibitors
US10851105B2 (en) 2014-10-22 2020-12-01 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
EP3233840B1 (fr) 2014-12-16 2018-11-21 Eudendron S.r.l. Dérivés hétérocycliques modulant l'activité de certaines protéines kinases
MA41551A (fr) 2015-02-20 2017-12-26 Incyte Corp Hétérocycles bicycliques utilisés en tant qu'inhibiteurs de fgfr4
WO2016134320A1 (fr) 2015-02-20 2016-08-25 Incyte Corporation Hétérocycles bicycliques utilisés en tant qu'inhibiteurs de fgfr
US9580423B2 (en) 2015-02-20 2017-02-28 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
CN108570052A (zh) * 2017-03-13 2018-09-25 中国科学院上海药物研究所 五元杂环并吡嗪化合物、制备方法、中间体、组合和应用
AR111960A1 (es) 2017-05-26 2019-09-04 Incyte Corp Formas cristalinas de un inhibidor de fgfr y procesos para su preparación
MA52494A (fr) 2018-05-04 2021-03-10 Incyte Corp Formes solides d'un inhibiteur de fgfr et leurs procédés de préparation
EP3788046A1 (fr) 2018-05-04 2021-03-10 Incyte Corporation Sels d'un inhibiteur de fgfr
US11628162B2 (en) 2019-03-08 2023-04-18 Incyte Corporation Methods of treating cancer with an FGFR inhibitor
EP3725777A1 (fr) * 2019-04-17 2020-10-21 Rottapharm Biotech S.r.l. Benzo- et pyrido-pyrazoles en tant qu`inhibiteurs de protéine kinase
WO2021007269A1 (fr) 2019-07-09 2021-01-14 Incyte Corporation Hétérocycles bicycliques en tant qu'inhibiteurs de fgfr
EP4045151A1 (fr) 2019-10-14 2022-08-24 Incyte Corporation Hétérocycles bicycliques utilisés en tant qu'inhibiteurs de fgfr
WO2021076728A1 (fr) 2019-10-16 2021-04-22 Incyte Corporation Hétérocycles bicycliques en tant qu'inhibiteurs de fgfr
WO2021113479A1 (fr) 2019-12-04 2021-06-10 Incyte Corporation Hétérocycles tricycliques en tant qu'inhibiteurs de fgfr
CA3162010A1 (fr) 2019-12-04 2021-06-10 Incyte Corporation Derives d'un inhibiteur de fgfr
WO2021197467A1 (fr) * 2020-04-02 2021-10-07 上海华汇拓医药科技有限公司 Composé antitumoral multicible, son procédé de préparation et son utilisation
CN116745268A (zh) 2020-11-20 2023-09-12 2692372安大略公司 苯磺酰胺衍生物及其用途
TW202313611A (zh) 2021-06-09 2023-04-01 美商英塞特公司 作為fgfr抑制劑之三環雜環

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TR199902301T2 (xx) * 1997-03-19 1999-12-21 Basf Aktiengesellschaft Pirilo $2,3D]pirimidinler ve onlar�n kullan�m�.
EP1215208B1 (fr) 1997-10-27 2006-07-12 Agouron Pharmaceuticals, Inc. Dérivées de 4-aminothiazole, leur préparation et leur utilisation comme inhibiteurs des kinases cycline-depenents
CN100338047C (zh) 2000-06-22 2007-09-19 辉瑞大药厂 吡唑并嘧啶酮类的制备方法
WO2003068773A1 (fr) * 2002-02-12 2003-08-21 Glaxo Group Limited Derives de pyrazolopyridine
DE102004061288A1 (de) * 2004-12-14 2006-06-29 Schering Ag 3-Amino-Pyrazolo[3,4b]pyridine als Inhibitoren von Proteintyrosinkinasen, deren Herstellung und Verwendung als Arzneimittel
CA2620254A1 (fr) * 2005-08-25 2007-03-01 F. Hoffmann-La Roche Ag Inhibiteurs de la map-kinase p38 et ses methodes d'utilisation
WO2008010964A1 (fr) 2006-07-17 2008-01-24 Merck & Co., Inc. Composés de 1-hydroxy naphtyridine en tant qu'agents anti-vih
PL2120932T3 (pl) 2006-12-20 2015-02-27 Nerviano Medical Sciences Srl Indazolowe pochodne jako inhibitory kinazy do leczenia raka
NZ583450A (en) 2007-07-20 2012-05-25 Nerviano Medical Sciences Srl Substituted indazole derivatives active as kinase inhibitors
ES2630026T3 (es) 2008-12-18 2017-08-17 Nerviano Medical Sciences S.R.L. Derivados indazólicos sustituidos activos como inhibidores de cinasa
FR2970967B1 (fr) * 2011-01-27 2013-02-15 Pf Medicament Derives de type azaindazole ou diazaindazole comme medicament

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20170317082A1 (en) * 2016-04-27 2017-11-02 International Business Machines Corporation Field effect transistor structure for reducing contact resistance
US10211205B2 (en) * 2016-04-27 2019-02-19 International Business Machines Corporation Field effect transistor structure for reducing contact resistance

Also Published As

Publication number Publication date
UA109698C2 (xx) 2015-09-25
JP2014503574A (ja) 2014-02-13
MY180666A (en) 2020-12-04
CN103339129B (zh) 2015-07-29
TW201247664A (en) 2012-12-01
NZ614432A (en) 2015-03-27
IL227496A0 (en) 2013-09-30
AU2012210467A1 (en) 2013-09-12
KR20140014147A (ko) 2014-02-05
RU2013138624A (ru) 2015-03-10
US20130085144A1 (en) 2013-04-04
MX2013008673A (es) 2013-12-02
TN2013000293A1 (en) 2015-01-20
AU2012210467B2 (en) 2016-07-07
IL257748A (en) 2018-04-30
PT2668184T (pt) 2018-03-13
DK2668184T3 (en) 2018-03-12
PL2668184T3 (pl) 2018-06-29
JP5931926B2 (ja) 2016-06-08
US8883821B2 (en) 2014-11-11
CN103339129A (zh) 2013-10-02
ES2661695T3 (es) 2018-04-03
KR20180122028A (ko) 2018-11-09
LT2668184T (lt) 2018-04-10
FR2970967B1 (fr) 2013-02-15
CY1120088T1 (el) 2018-12-12
EP2668184A1 (fr) 2013-12-04
MX356411B (es) 2018-05-24
CA2823824A1 (fr) 2012-08-02
KR101923751B1 (ko) 2018-11-29
RS57101B1 (sr) 2018-06-29
TR201802944T4 (tr) 2018-03-21
IL227496B (en) 2019-02-28
HRP20180524T1 (hr) 2018-05-04
EP2668184B1 (fr) 2018-01-10
HUE037153T2 (hu) 2018-08-28
CA2823824C (fr) 2018-12-11
FR2970967A1 (fr) 2012-08-03
IL257748B (en) 2020-06-30
WO2012101239A1 (fr) 2012-08-02
HK1186737A1 (en) 2014-03-21
SI2668184T1 (en) 2018-06-29
MA34903B1 (fr) 2014-02-01
AR084935A1 (es) 2013-07-10
TWI546303B (zh) 2016-08-21
RU2600976C2 (ru) 2016-10-27

Similar Documents

Publication Publication Date Title
US8883821B2 (en) Substituted pyrazolo[3,4-b]pyridines as medicaments
JP6732855B2 (ja) 疼痛を治療するためのアザインダゾールまたはジアザインダゾール型の誘導体
US9428510B2 (en) Derivatives of azaindazole or diazaindazole type for treating a cancer overexpressing Trk
OA16493A (en) Derivatives of azaindazole or diazaindazole type as medicament.
NZ614432B2 (en) Derivatives of azaindazole or diazaindazole type as medicament
BR112013018852B1 (pt) Derivados do tipo azaindazol ou diazaindazol, seus usos na preparação de fármacos, métodos de preparação dos mesmos, e composições farmacêuticas compreendendo os referidos compostos
OA17324A (en) Derivatives of azaindazole or diazaindazole type for treating pain.

Legal Events

Date Code Title Description
AS Assignment

Owner name: PIERRE FABRE MEDICAMENT, FRANCE

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:KALOUN, EL BACHIR;BEDJEGUELAL, KARIM;RABOT, REMI;AND OTHERS;REEL/FRAME:030363/0213

Effective date: 20130408

STCB Information on status: application discontinuation

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