WO2006133006A2 - 1-methyl-1h-pyrazole-4-carboxamides useful as cancer chemotherapeutic agents - Google Patents

1-methyl-1h-pyrazole-4-carboxamides useful as cancer chemotherapeutic agents Download PDF

Info

Publication number
WO2006133006A2
WO2006133006A2 PCT/US2006/021550 US2006021550W WO2006133006A2 WO 2006133006 A2 WO2006133006 A2 WO 2006133006A2 US 2006021550 W US2006021550 W US 2006021550W WO 2006133006 A2 WO2006133006 A2 WO 2006133006A2
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
substituted
groups
alkoxy
independently
Prior art date
Application number
PCT/US2006/021550
Other languages
French (fr)
Other versions
WO2006133006A3 (en
Inventor
Philip Wickens
Harold C. E. Kluender
Zhenqiu Hong
Ellalahewage Sathyajith Kumarasinghe
Charles Kreiman
Mingbao Zhang
Istvan Enyedy
Chih-Yuan Chuang
Original Assignee
Bayer Healthcare Ag
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 Bayer Healthcare Ag filed Critical Bayer Healthcare Ag
Priority to JP2008514922A priority Critical patent/JP2008545756A/en
Priority to CA002610509A priority patent/CA2610509A1/en
Priority to EP06772019A priority patent/EP1891047A4/en
Publication of WO2006133006A2 publication Critical patent/WO2006133006A2/en
Publication of WO2006133006A3 publication Critical patent/WO2006133006A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • 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
    • 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

Definitions

  • This invention relates to novel 1 -Methyl- lH-pyrazole-4-carboxamide compounds, pharmaceutical compositions containing such compounds, and the use of those compounds or compositions as cancer chemotherapeutic agents.
  • retinopathies Many disease conditions are known to be associated with deregulated angiogenesis.
  • chronic inflammatory disorders including arthritis; arteriosclerosis; atherosclerosis; macular degeneration; and neoplastic diseases such as cancer.
  • arteriosclerosis including arthritis
  • atherosclerosis including atherosclerosis
  • macular degeneration including macular degeneration
  • neoplastic diseases such as cancer.
  • much work has been carried out to find inhibitors of angiogenesis, in hopes of developing treatments for such disorders.
  • WO 2004/063330 discloses (2-carboxamido)(3-amino)thiophene compounds for the treatment of cancer.
  • Published PCT application WO 02/066470 broadly discloses heterocycles containing amido and amino substituent groups, for prophylaxis and treatment of angiogenesis-mediated diseases.
  • Published PCT application WO 2004/005279 discloses certain substituted anthranilic amide derivatives for the prophylaxis and treatment of angiogenesis-mediated diseases.
  • Published PCT application WO 2004/007458 (Amgen) relates to substituted 2-alkylamine nicotinic amide derivatives and their uses in treatment of cancer and other disorders.
  • EP-B-832 061 discloses benzamide derivatives and their use as vasopressin antagonists.
  • the present invention relates to a compound of Formula (I)
  • Ar is selected from the group consisting of
  • X is CH or N
  • R 1 is selected from the group consisting of H, halogen,
  • R 1"2 is selected from the group consisting of
  • R 1"3 is H or (Ci-COalkyl
  • R 1"4 , R 1"5 and R 1'6 are independently selected from the group consisting of
  • R 1"3 and R 1"4 , R 1'3 and R 1"5 , and R 1'3 and R 1"6 when attached to the same nitrogen atom, may form, together with the N atom to which they are attached, a 5- or 6-membered saturated heterocyclic ring selected from pyrrolidinyl, morpholinyl, thiomorpholinyl and piperizinyl optionally substituted on N with (Q-C 4 )alkyl;
  • the invention also relates to pharmaceutical compositions which comprise a compound of Formula (I) as defined above plus a pharmaceutically acceptable carrier.
  • the invention relates to a method of treating cancer comprising administering to a subject in need thereof an effective amount of a compound of Formula (I) as defined above.
  • Pharmaceutically acceptable salts of the compounds (I) include acid addition salts of mineral acids, carboxylic acids and sulphonic acids, for example salts of hydrochloric acid, hydrobromic acid, sulphuric acid, phosphoric acid, methanesulphonic acid, ethanesulphonic acid, toluenesulphonic acid, benzenesulphonic acid, naphthalenedisulphonic acid, acetic acid, propionic acid, lactic acid, tartaric acid, malic acid, citric acid, fumaric acid, maleic acid and benzoic acid.
  • hydrochloric acid hydrobromic acid, sulphuric acid, phosphoric acid, methanesulphonic acid, ethanesulphonic acid, toluenesulphonic acid, benzenesulphonic acid, naphthalenedisulphonic acid, acetic acid, propionic acid, lactic acid, tartaric acid, malic acid, citric acid, fumaric acid, maleic acid and benzo
  • Pharmaceutically acceptable salts of the compounds (T) also include salts of customary bases, such as for example and preferably alkali metal salts (for example sodium and potassium salts, alkaline earth metal salts (for example calcium and magnesium salts) and ammonium salts derived from ammonia or organic amines having 1 to 16 carbon atoms, such as illustratively and preferably ethylamine, diethylamine, triethylamine, ethyldiisopropylamine, monoethanolamine, diethanolamine, triethanolamine, dicyclohexylamine, dimethylaminoethanol, procaine, dibenzylamine, N-methylmorpholine, dihydroabietylamine, arginine, lysine, ethylenediamine and methylpiperidine.
  • alkali metal salts for example sodium and potassium salts, alkaline earth metal salts (for example calcium and magnesium salts)
  • Solvates for the striges of the invention are those forms of the compounds that coordinate with solvent molecules to form a complex in the solid or liquid state. Hydrates are a specific form of solvates, where the coordination is with water. For the purposes of the present invention, the substituents have the following meanings, unless otherwise specified:
  • (C 1 -C 4 )alkyl and “(CrC 6 )alkyl” mean a linear or branched saturated carbon group having from about 1 to about 4 C atoms or from about 1 to about 6 C atoms, respectively. Such groups include but are not limited to methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, and the like.
  • (C 3 -C 6 )cycloalkyl means a saturated carbocyclic ring group having from about 3 to about 6 C atoms. Such groups include but are not limited to cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and the like.
  • the O atom is the point of attachment of the alkoxy substituent to the rest of the molecule.
  • Such groups include but are not limited to methoxy, ethoxy, n-propoxy, isopropoxy, and the like.
  • (Q-GOalkylamino) means an amino group having from one or two (independently selected) (Q-GOalkyl substituents, illustratively representing methylamino, ethylamino, n-propylamino, isopropylamino, teit-butylamino, n-pentylamino, n-hexylamino, N,N-dimethylamino, N,N-diethylamino, N-ethyl-N-methylamino, N-methyl-N- n-propylamino, N-isopropyl-N-n-propylamino, N-t-butyl-N-methylamino, iV-ethyl-iV- n-pentylamino, N-n-hexyl-N-methylamino and the like.
  • (d-C ⁇ alkylsulfonyl) means a sulfonyl group having a (Q-C ⁇ alkyl substituent, illustratively representing methylsulfonyl, ethyl sulfonyl, isopropylsulfonyl, t- butylsulfonyl, and the like.
  • (CrC 4 )alkoxycarbonyl means a (Ci-C 4 )alkoxygroup bound to the C atom of a carbonyl group [ -C(O) - ]said group being bound to the rest of the molecule, illustratively representing methoxycarbonyl, ethoxycarbonyl, /z-propoxycarbonyol, i- propoxycarbonyl, t-butoxycarbonyl, and the like
  • (CrC 4 )acyloxy means a (CrC 4 )group bound to the C atom of a carboxyl group [ -C(O)O- ], said group being bound by the oxygen atom to the rest of the molecule illustratively representing formyloxy, acetyloxy (acetoxy), propanoyloxy, butanoyloxy, t-butanoyloxy and the like
  • an aromatic ring made of 5 atoms and having 1, 2, 3 or 4 heteroatom(s) each selected independently from O, N, and S, the rest being C atoms, with the proviso that there can be no more than 1 O or S atom in the heteroaryl.
  • This heteroaryl is attached to the core molecule at any available C or N atom and is optionally substituted at any available C or N atom with the recited substituents.
  • groups include pyrrole, furan, thiophene, imidazole, pyrazole, thiazole, oxazole, isoxazole, isothiazole, triazole, oxadiazole, thiadiazole, and tetrazole in all their possible isomeric forms; or
  • an aromatic ring made of 6 atoms, 1, 2, or 3 of which are N atoms, the rest being C, where the heterocycle is attached to the core molecule at any available C atom and is optionally substituted at any available C atom with the recited substituents.
  • Such groups include pyridine, pyrimidine, pyridazine and triazine in all their possible isomeric forms.
  • heterocyclyl means a 5-or 6- membered saturated or partially saturated heterocyclic ring containing 1-2 heteroatoms selected from O, S or N, the remaining atoms being made up of C atoms, with the proviso that when there are 2 O atoms they must be nonadjacent.
  • This heterocycle is attached to the core molecule at any available C or N atom and is optionally substituted at any available C or N atom with the recited substituents.
  • Such groups include pyrrolidine, tetrahydrofuryl, tetrahydrothienyl, piperidinyl, tetrahydropyranyl, tetrahydrothiopyrano, piperizinyl, imidazolinyl, pyrazolinyl, morpholinyl, thiomorpholinyl and the like in all their possible isomeric forms.
  • a * symbol next to a bond denotes the point of attachment in the molecule.
  • 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. It is intended that all possible stereoisomers (including enantiomers and diastereomers) are included within the scope of the present invention.
  • Preferred compounds are those with the absolute configuration of the compound of this invention which exhibits the more desirable biological activity.
  • Separated, pure or partially purified stereoisomers or racemic mixtures of the compounds of this invention are also included within the scope of the present invention. The purification of said isomers and the separation of said stereoisomeric mixtures can be accomplished by standard techniques known in the art.
  • the invention relates to a compound of Formula (I), wherein
  • Ar is selected from the group consisting of
  • X is CH or N
  • R 1 is selected from the group consisting of O
  • R " is selected from the group consisting of
  • R 1 - 3 is H or (C r C 4 )alkyl
  • R 1"4 and R 1"5 are independently selected from the group consisting of
  • - phenyl substituted with 0, 1, or 2 groups independently selected from (Q-GOalkyl, halo, nitro, (C 1 -C ⁇ aIkOXy and cyano,
  • R 1"3 and R 1"4 , R 1"3 and R 1"5 , and R 1"3 and R 1"6 when attached to the same nitrogen atom, may form, together with the N atom to which they are attached, a 5- or 6-membered saturated heterocyclic ring selected from pyrrolidinyl, morpholinyl, thiomorpholinyl and piperizinyl optionally substituted on N with (Q-GOalkyl;
  • the invention relates to a compound of Formula (I), wherein
  • Ar is selected from the group consisting of
  • X is CH
  • R 1 is selected from the group consisting of
  • R 1"3 is H or (Ci-GOalkyl, R " is selected from the group consisting of indan-5-yl, phenyl substituted with 1 or 2 groups independently selected from
  • R 1"6 is selected from the group consisting of H, indan-5-yl, phenyl substituted with 0, 1, or 2 groups independently selected from (Q-GOalkyl, halo, nitro, (Q-GOalkoxy and cyano, 5- or 6-membered heteroaryl substituted with 0, 1 or 2 groups selected from cyano, halo, nitro, (C 1 -C 4 )alkyl, wherein said (CrC 4 )alkyl is optionally substituted with 0, 1, or 2 groups selected from (C 1 -C 4 )alkylamino, (C 1 -C 4 )acyloxy, (Ci-COalkoxy, and (C 2 -C 4 )alkoxy substituted with up to 0, 1 or 2 (C 1 -C 4 )alkoxy groups; (C 3 -C 6 )cycloalkyl
  • R 1"3 and R 1"5 , and R 1'3 and R 1"6 when attached to the same nitrogen atom, may form, together with the N atom to which they are attached, a 5- or 6-membered saturated heterocyclic ring selected from pyrrolidinyl, morpholinyl, thiomorpholinyl and piperizinyl optionally substituted on N with (C 1 -C 4 )alkyl;
  • the invention relates to a compound of Formula (I)
  • X is CH
  • R 1 is selected from the group consisting of
  • R 1 - 3 is H, wherein said (Ci-C 6 )alkyl is independently substituted with 1 group selected from (C r C 4 )alkoxy,
  • R 1"6 is selected from the group H, and (Ci-QOalkyl, wherein said (C 1 -C 6 )alkyl is independently substituted with 0 or 1 group selected from (Ci-C 4 )alkoxy, (C 2 -C 4 )alkoxy independently substituted with
  • the invention relates to a compound of Formula (I)
  • X is CH
  • R is selected from the group consisting of
  • R 1"3 is H
  • R 1"6 is selected from the group
  • (C 1 -C 6 )alkyl wherein said (C 1 -C 6 )alkyl is independently substituted with 0 or 1 group selected from (C r C 4 )alkoxy, (C 2 -C 4 )alkoxy independently substituted with
  • the invention relates to a process for making a compound of formula (I), comprising (A) reacting, with or without first hydrolizing the ester group -COOR', a compound of formula (VI)
  • Ar has the meaning described above, for example in the presence of a coupling agent such as PyBOP; or
  • Ig represents a leaving group, such as halo, OTs or OMs;
  • R 1 and X have the same meanings as defined hereinabove.
  • n 1 or 2
  • the amino group of the compound of Formula (DI) is subjected to either reductive amination using a pyridine or pyrimidine carboxaldehyde of Formula (IV) and a reducing agent, such as sodium triacetoxyborohydride, or to direct N-alkylation using a pyridine or pyrimidine methyl halide, tosylate or mesylate of Formula (V) and a optional base such as pyridine or K 2 CO 3 , or a catalyst such as sodium iodide.
  • a reducing agent such as sodium triacetoxyborohydride
  • the compound of Formula (III) is converted to the aminoamide of Formula (IX) either directly by reaction with an aromatic amine of Formula (VIII) as described above, or by first protecting the amino function, e.g., as a BOC derivative (VII), and subsequent coupling with (VIII), either directly with (R') 3 A1, or via hydrolysis, and then coupling in the presence of PyBOP, followed by deprotection.
  • the Formula (IX) compound is then converted to the Formula (I) compound using either the reductive amination method or direct N-alkylation as described above for preparation of (VI).
  • the amino group of the compound of formula (III) can be acylated using an acylating agent such as acetic anhydride and formic acid. N-alkylation of the acylated product Formula (II) using a pyridine or pyrimidine methyl halide, tosylate or mesylate of Formula (V) and a base such as DBU or K 2 CO 3 .
  • pg protecting group, e.g., BOC
  • Ig leaving group, e.g., halo, MsO, etc.
  • Ig leaving group, e.g., halo, MsO
  • the intermediate of Formula (X) is allowed to react with a carbamoyl chloride Formula R 1"6 R 1-3 NCOCl in an aprotic solvent such as dichloromethane in the presence of a base such as triethylamine or potassium carbonate.
  • a starting material of Formula (X) in which the R 1"3 on the left is
  • alkyl results in the preparation of a urea of structure (Vf) where R 1 is R R 1"3 in which the R 1"3 group on the left is alkyl.
  • R 1"6 is aryl or heteroaryl
  • phosgene diphosgene or triphosgene
  • a suitable solvent such as ethyl acetate
  • R 1"6 is alkyl or substituted alkyl
  • the preferred method is to treat the corresponding alkyl halide or dialkyl sulfate with inorganic cyanates.
  • the carbamoyl chloride of Formula R 1"6 R 1-3 NCOCl is not commercially available, it can conveniently be prepared by treatment of the amine of Formula R " R " NH with phosgene, diphosgene or triphosgene in a suitable solvent such as dichloromethane at 0-4 °C.
  • a suitable solvent such as dichloromethane at 0-4 °C.
  • the N-benzyl protected amine of Formula R 1"6 R 1-3 NCH 2 (C 5 H 6 ) can be reacted with triphosgene as described by M.G. Banwell, et al, J. Org. Chem. 2003, 68, 613-616.
  • the dimethyl pyrrole compound of Formula (Ij) is made by coupling [2-(2,5-dimethyl-lH-pyrrol-l-yl)pyridin-4-yl]methyl methanesulfonate with 5-amino-N- (2,2-difluoro-l,3-benzodioxol-5-yl)-l -methyl- lH-pyrazole-4-carboxamide as shown in Scheme 1.
  • [2-(2,5-dimethyl-lH-pyrrol-l-yl)pyridin-4-yl]methyl methanesulfonate is made by coupling (2-aminopyridin-4-yl)methanol with hexane-2,5-dione using catalytic acid and then mesylation of the alcohol produced.
  • the dimethyl pyrrole compound of Formula (Ij) can be deprotected to give a compound of Formula (Ia).
  • the amine can then be converted to the amide compound of Formula (Ib), the sulfonamide of Formula (Ic) or the urea of Formula (Id) as shown in Reaction Scheme 10, by reaction with an acid chloride, sulfonyl chloride or isocyanate, respectively.
  • chloro compound of Formula (Ie) can be converted to the substituted amino compound of Formula (If) by reaction with an amine and a base such as pyridine in a sealed tube at elevated temperatures.
  • Esters of Formula (Ih) and substituted amides of Formula (Ii) may be prepared from the unsubstituted amide of Formula (Ig) by the sequence illustrated in Reaction Scheme 12. Reaction of the amide (Ig) with dimethylformamide-dimethylacetal (DMF- DMA) in methanol provides the ester of Formula (Di); reaction of the ester with a substituted amine gives the amide of Formula (Ii).
  • a desired salt of a compound of this invention can be prepared in situ during the final isolation and purification of a compound by means well known in the art. Or, a desired salt can be prepared by separately reacting the purified compound in its free base form with a suitable organic or inorganic acid and isolating the salt thus formed. These methods are conventional and would be readily apparent to one skilled in the art.
  • protective groups on the compound of this invention may need to be protected and deprotected during any of the above methods.
  • Protecting groups in general may be added and removed by conventional methods well known in the art (see, for example, T. W. Greene and P.G.M. Wuts, Protective Groups in Organic Synthesis; Wiley: New York, (1999).
  • Electron impact mass spectra were obtained with a Hewlett Packard 5989A mass spectrometer equipped with a Hewlett Packard 5890 Gas Chromatograph with a J & W DB-5 column (0.25 ⁇ M coating; 30 m x 0.25 mm). The ion source is maintained at 250 0 C and spectra were scanned from 50-800 amu at 2 sec per scan.
  • Step 1 Preparation of ethyl 5- Fbis(tert-butoxycarbonyl)aminol-l -methyl- lH-pyrazole-
  • Step 3 Preparation of 5-amino-N-(2,2-difluoro-l,3-benzodioxol-5-yl)-l -methyl- IH- pvrazole-4-carboxamide
  • This intermediate can be prepared by using the method described above for the preparation of Intermediate A but using 2,2,3,3-tetrafluoro-2,3-dihydro-l,4-benzodioxin- 6-amine rather than 2,2-difluoro-l,3-benzodioxol-5-amine in Step 3.
  • the pure product is characterized by NMR and LCMS spectroscopy.
  • This intermediate can be prepared by using the method described above for the preparation of Intermediate A but using 2,2,4,4-tetrafluoro-4H-l,3-benzodioxin-6-amine rather than 2,2-difluoro-l,3 ⁇ benzodioxol-5-amine in Step 3.
  • This intermediate can be prepared by using the method described above for the preparation of Intermediate A but using 4-(trifluoromethoxy)aniline rather than 2,2- difluoro-l,3-benzodioxol-5-amine in Step 3.
  • This intermediate can be prepared by using the method described above for the preparation of Intermediate A but using 3-(trifluorometlioxy)aniline rather than 2,2- difluoro-l,3-benzodioxol-5-amine in Step 3.
  • This intermediate can be prepared by using the method described above for the preparation of Intermediate A but using 4-[(trifluoromethyl)thio] rather than 2,2-difluoro-l,3- benzodioxol-5-amine in Step 4.
  • Step 1 Preparation of ethyl 2-(aminocarbonyl)isonicotinate
  • This compound was prepared by using the method described above for Intermediate C but starting with methyl formamide rather than formamide in step 1 and methanesulfonic anhydride rather than methanesulfonyl chloride in step 3.
  • Step 2 Preparation of 2- ⁇ r4-(chloromethyl)pyridin-2-yl1amino
  • Intermediate F was prepared from 2.30 g of 4-(chloromethyl)pyridin-2-amine and proportional amounts of other reagents.
  • the yield of title compound was 2.0 g (67%) after silica gel chromatography. Even though examination of this material by NMR spectroscopy indicated that it was a mixture of the desired compound and the diacylated product N- acetyl-N-[4-(chloromethyl)pyridin-2-yl]acetamide (about 45:55), it was used as is in the next reaction and side products were separated by chromatography after the subsequent reaction.
  • Intermediate G was prepared from 731 mg of 4-(chloromethyl)pyridin-2-amine and proportional amounts of other reagents. The yield of pure title compound was 397 mg (45%) after silica gel chromatography using a gradient from 0-40% ethyl acetate in hexane.
  • Intermediate H was prepared from 599 mg of 4-(chloromethyl)pyridin-2-amine and proportional amounts of other reagents.
  • the yield of pure title compound was 314 mg (29%) after silica gel chromatography twice, first using a gradient from 2-3% methanol in dichloromethane, and then a second chromatography of the best fractions using a gradient from 0-40% ethyl acetate in hexane.
  • Step 2 By using the methods described for preparation of Intermediate E (Step 2) and by substituting 2-methoxypropanoyl chloride instead of acetoxyacetyl chloride, Intermediate I was prepared from 352 mg of 4-(chloromethyl)pyridin-2-amine and proportional amounts of other reagents. The yield of pure title compound was 341 mg (60%) after silica gel chromatography using a gradient from 0-30% ethyl acetate in hexane.
  • Step 2 By using the methods described for preparation of Intermediate E (Step 2) and by substituting 2-methoxy-2-methylpropanoyl chloride instead of acetoxyacetyl chloride, Intermediate J was prepared from 1.04 g of 4-(chloromethyl)pyridin-2-amine and proportional amounts of other reagents. The yield of title compound was 1.23 g (69%) after silica gel chromatography using 30% ethyl acetate in hexane.
  • Step 1 Preparation of N-r4-(chloromethyl)pyridin-2-yll-N-(methylsulfonyl) methanesulfonamide
  • Step 1 Preparation of r2-(2,5-dimethyl-l H-pyrrol- l-yl)pyridin-4-ynmethanol
  • Step 2 Preparation of r2-(2,5-dimemyl-lH-pyrrol-l-yl)pyridin-4-yllmethyl methanesulfonate
  • Step 1 Preparation of 4-( ⁇ rt g rt-butvirdimethyl)silylloxy ⁇ methvDpyridin-2-amine
  • Step 2 Preparation of N-( ⁇ r4-((rtg/t-butyl(dimethyl)silylloxy ⁇ methyl)pyridin-2- yliamino ⁇ carbonothioyl)benzamide
  • Step 3 Preparation of N-F4-( ' (rtert-butylfdimethyl ' )silyl1oxylmethyl)pyridin-2-yllthiourea
  • Step 4 Preparation of ⁇ 2-r(4-methyl-l,3-thiazol-2-yl)amino1pyridin-4-yl ⁇ methanol
  • the resulting solution was stirred at 60 0 C in a foil wrapped flask for 2Oh.
  • the resulting solution was evaporated and then diluted with 1 mL methanol and injected in two portions on a 150/20 mm Cl 8 HPLC column using a gradient from 10-50 % acetonitrile in water (plus 0.05% ti ⁇ fmoroacetic acid).
  • the best fractions containing the desired material, as identified by LCMS, were combined, mixed with saturated NaHCO 3 , and extracted three times with dichloromethane.
  • the combined extracts were dried (Na 2 SO 4 ) and evaporated in vacuo to yield 90 mg (21%) of pure title compound.
  • Step 1 Preparation of ethyl 5-(formylamino)-l -methyl- lH-pyrazole-4-carboxylate
  • Step 2 Preparation of ethyl 5 ⁇ rr(2-chloropyridin-4-yl)methyll(formyl)aminol-l- methyl- 1 H-pyrazole-4-carboxylate
  • Step 4 5-( Ff 2-chloropyridin-4-yl)methyH amino ⁇ -N-(2.2-difluoro- 1 ,3-benzodioxol-5-yl)-
  • reaction mixture was then diluted with EtOAc, and the organics were washed with concentrated NaHCO 3 solution, followed by water, and then brine. The organics were then dried over Na 2 SO 4 and concentrated in vacuo. The crude residue was triturated with hot hexanes yielding 55.0mg (68.6%) product.
  • Step 1 Preparation of 4- ⁇ f(4- ⁇ F(2,2-difluoro- 1 ,3-benzodioxol-5-yl)aminolcarbonyl I - 1 -methyl- 1 H-pyrazol-5-yl)aminolmethyl ⁇ pyridine-2-carboxamide
  • the title compound can be prepared by the following method.
  • a solution of 5-amino-N- (2,2-difluoro- 1 ,3-benzodioxol-5-yl)- 1 -methyl- lH-pyrazole-4-carboxamide (Intermediate A), sodium iodide, and 2,6-di(te/t)butyl ⁇ 4-methylphenol in dry dimethylformamide can be stirred under nitrogen as [2-(aminocarbonyl)pyridin-4-yl]methyl methanesulfonate (Intermediate C) added.
  • the resulting solution isstirred at 60 °C in a foil wrapped flask for 2Oh.
  • reaction solution is purified by HPLC using direct injection, in three portions, on a YMC-Pack Pro C18 column (150 x 20 mm) and is eluted at 20 mL/min with a gradient from 10-50 % acetonitrile in water plus 0.05% TFA. Pure fractions from each injection are combined, made basic by addition of sodium bicarbonate and extracted with ethyl acetate. Combined extracts are dried (Na 2 SO 4 ) and evaporated in vacuo to yield pure title compound
  • This material is prepared using the same method described for Example 1 but starting with 4-amino-7V-(2,2-difluoro- 1 ,3-benzodioxol-5-yl) ⁇ 1 ,3-thiazole-5-carboxamide (Intermediate A) and proportionate amounts of Intermediate G rather than Intermediate D and also proportional amounts of the other reaction components.
  • the reaction mixture is heated to 60 °C in a foil wrapped flask overnight before crude product isolated.
  • This material is purified by chromatography on silica gel using a gradient from 0-60 % ethyl acetate in hexane to yield final product.
  • the title compound is prepared using the same method described for Example 1 but starting with 5-amino-l-methyl-N-(2,2,3,3-tetrafluoro-2,3-dihydiO-l,4-benzodioxin-6- yl)-lH-pyrazole-4-carboxamide (Intermediate B) rather than 5-amino-N-(2,2-difluoro-l,3- benzodioxol-5-yl)-l -methyl- lH-pyrazole-4-carboxamide (Intermediate A) and proportionate amounts of N-[4-(chloromethyl)pyridin-2-yl]methanesulfonamide (Intermediate K) rather than Intermediate D and also proportional amounts of the other reaction components.
  • reaction mixture is heated to 60 0 C in a foil wrapped flask for 16 h and then cooled.
  • the reaction mixture is diluted with ethyl acetate, washed with water, dried (Na 2 SO 4 ) and evaporated in vacuo.
  • the crude product is purified by preparative HPLC to yield pure title compound.
  • Step 1 Preparation of 5-( ⁇ r2-(2,5-dimethyl-lH-pyrrol-l-yl)pyridin-4-yl1methyl ⁇ amino)- 1- methyl-N-(2,2,3,3-tetrafluoro-2,3-dihydro-l,4-benzodioxin-6-yl)-lH-pyrazole-4- carboxamide
  • Step 2 intermediate R was used in place of 2-Chloro-4- chloromethylpyridine.
  • Step 3 the hydrolysis also removes the benzoyl group along with the formyl group and hydrolysis of the ester.
  • Example 16 as shown in Table B was made according to this method substituting the appropriate starting materials.
  • the title compound was prepared using the same method described for Example 1.
  • the intermediate L was used in place of intermediate D and the concentrated HPLC fractions were analyzed without free-basing with NaHCO 3 .
  • Step 1 Preparation of ethyl 5-rr(2-aminopyridin-4-yl)methyl1(formyl)amino1-l-methyl- lH-pyrazole-4-carboxylate
  • Step 2 Preparation of ethyl 5-r( ⁇ 2-r(fert-butoxycarbonvDaminolpyridin-4- yl ⁇ methyl)(formyl)amino1-l-methyl-lH-pyrazole-4-carboxylate
  • reaction mixture was then diluted with ethyl acetate and washed with concentrated NH 4 Cl solution, followed by water, then brine.
  • the organic layer was dried over Na 2 SO 4 and concentrated in vacuo.
  • the crude residue was triturated with ether, and the solvent was again evaporated yielding 600mg (90.1%, impure) product. The product was used without purification.
  • Step 4 Preparation of tert-butyl (4-(r(4- ⁇ r(2,2-difruoro-l,3-benzodioxol-
  • Step 5 Preparation of 5- ⁇ r(2-aminopyridin-4-yl)methyllamino ⁇ -N-f2,2-difluoro-l,3- benzodioxol-5-yl)-l-methyl-lH-pyrazole-4-carboxamide
  • Step 6 Preparation of N-(2,2-difluoro-13-benzodioxol-5-yl)-l-methyl-5-( r(2-
  • Example 8 step 3 The title compound was prepared using the same method described for Example 8 step 3. In Step 3, methyl isocyanate was used in place of ethyl isocyanate. The workup for the final step was different in that the concentrated HPLC fractions were analyzed without free-basing with NaHCO 3 .
  • Step 2 The step is carried out using the method described for the preparation of Intermediate L but using the product of Step 2 above rather than 4-(chloromethyl)pyridin- 2-amine and methyl isocyanate rather than ethyl isocyanate.
  • Step 4 Preparation of N-(2,2-difluoro-l,3-benzodioxol-5-yl)-l-methyl-5-(r(2- ⁇ methyl r(methylamino)carbonyll amino
  • the title compound can be prepared using the same method described for Example 1 but starting with 5-amino-N-(2,2-difluoro- 1 ,3-benzodioxol-5-yl)- 1-methyl- IH- pyrazole-4-carboxamide (Intermediate A) and a proportionate amount of N- [4- (chloromethyl)pyridin-2-yl]-N,N'-dimethylurea (from Step 3) rather than Intermediate D.
  • the reaction mixture is heated at 60 °C in a foil wrapped flask under nitrogen for between 2 and 24 h until an LCMS analysis of the reaction mixture shows substantial conversion to products.
  • the resulting final crude mixture is diluted with saturated aqueous sodium bicarbonate and extracted 3 times with ethyl acetate.
  • the combined extracts are dried (Na 2 SO 4 ) and evaporated in vacuo to yield a residue that is purified by preparative C18 HPLC using water to acetonitrile gradient (usually 10-50%) with added 0.05-0.1 % TFA.
  • the free base is prepared from the TFA salt by addition of saturated aqueous NaHCO 3 to the fractions containing the product and extraction with dichloromethane followed by drying of the extract (Na 2 SO 4 ) and concentration in vacuo to yield pure title compound.
  • Step 1 Preparation of various ⁇ r -r4-(chloromethyl)pyridin-2-vnureas with other ISF- substituents Cl H R 1'3
  • the title compounds can be prepared using the same method described for Example 1 but starting with 5-amino-N-(2,2-difluoro-l,3-benzodioxol-5-yl)-l-methyl-lH- pyrazole-4-carboxamide (Intermediate A) or another material from the list of Intermediates B, B-2, B-3, B-4 or B-5 instead of Intermediate A and proportionate amounts of the appropriate Intermediate from Step 1 above rather than Intermediate D and also proportional amounts of the other reaction components.
  • the reaction mixture is heated at 60 °C in a foil wrapped flask for between 2 and 24 h until an LCMS analysis of the reaction mixture shows substantial conversion to products.
  • the resulting final crude mixture is diluted with saturated aqueous sodium bicarbonate and extracted 3 times with ethyl acetate.
  • the combined extracts are dried (Na 2 SO 4 ) and evaporated in vacuo to yield a residue that is purified by preparative C18 HPLC using water to acetonitrile gradient (usually 10-50%) with added 0.05-0.1 % TFA.
  • the free base is prepared from the TFA salt by addition of saturated aqueous NaHCO 3 to the fractions containing the product and extraction with dichloromethane followed by drying of the extract (Na 2 SO 4 ) and concentration in vacuo to yield pure title compound.
  • Step 2 Preparation of 5-( ⁇ r6-chloro-2-(methylamino)pyrimidin-4-yllmethyl ⁇ amino)- N-(2,2-difluoro-13-benzodioxol-5-yl)-l-methyl-lH-pyrazole-4-carboxamide and 5-( ⁇ ⁇ 2- chloro-6-(methyla ⁇ uno) ⁇ yrirrndin-4-yl1methyl ⁇ amino)-N-(2,2-difluoro- 1 ,3-benzodioxol-
  • the residue can be purified by preparative C18 HPLC using a gradient from 5 to 45% acetonitrile in water plus 0.1% TFA. Evaporation of product containing fractions can yield pure compounds as TFA salts.
  • the fractions containing the product can be mixed with saturated aqueous NaHCO 3 and extracted with dichloromethane. The extracts are dried (Na 2 SO 4 ) and evaporated in vacuo to yield pure free base title compound.
  • the utility of the compounds of the present invention can be illustrated, for example, by their activity in the P-AKT/PKB Cytoblot Assay described below.
  • the involvement of the P-AKT/PKB [ PDK/AKt] pathway as a target for cancer chemotherapy has been recognized in the art.
  • P. Chang et al Involvement of PI3K/Akt pathway in cell cycle progression, apoptosis, and neoplastic transformation: a target for cancer chemotherapy, Leukemia, 2003, 17: p. 590-603; K. A. West et al, Activation of the PI3K/Akt pathway and chemotherapeutic resistance, Drug Resistance Updates, 2002, 5: p. 234-248; and P. Sen et al, Involvement of the Akt/PKB signaling pathway with disease processes, Molecular and Cellular Biochemistry, 2003, 253: p. 241- 246.
  • H209 small cell lung carcinoma cells in log phase were plated at 50,000 cells/well in 96- well poly-lysine coated, clear bottom/ black-sided plates (Becton-Dickinson, USA Cat # 354640) in 100 ⁇ l RPMI medium containing 0.1% (w/v) BSA, and incubated overnight at 37 0 C in 5% CO 2 incubator. The following day, compounds (10 mM stock solutions in DMSO) were added to the plates to generate final concentrations of 0.0, 0.01, 0.03, 0.1, 0.3, 1.0, 3.0 and 10 ⁇ M for IC 50 determinations and incubated for 1 hour at 37 0 C.
  • Cells were then left untreated or stimulated with Stem Cell Factor (SCF: Biosource Cat # PHC2116) at a final concentration of 25 ng/mL for 5 minutes at 37 0 C in 5% CO 2 incubator. The media was then removed using a vacuum manifold and the cells were washed once with Tris Buffered Saline (TBS). Cells were then fixed by adding 200 ⁇ l of cold 3.7% (w/v) formaldehyde in TBS to each well for 15 minutes at 4 0 C. After removal of the formaldehyde, the cells were treated with the addition of 50 ⁇ l of methanol (at - 20 0 C) to each well for 5 minutes. After removal of the methanol, 200 ⁇ l of 1% (w/v) BSA in TBS was added to each well to block non-specific antibody binding sites and the plate was incubated at room temperature for 30 minutes.
  • SCF Stem Cell Factor
  • p-(S473) AKT rabbit polyclonal antibody (Cell Signaling, USA Cat # 9277S) was added at a dilution of 1:250 in 0.1% (w/v) BSA in TBS, and the plate was incubated at room temperature for 1 hour. Plates were then washed 3 times with cold TBS containing 0.05% (v/v) Tween 20 (TBS-T) and 100 ⁇ l of Horseradish peroxidase (HRP)-conjugated goat-anti-rabbit antibody (Amersham, USA Cat # NA934V) at a dilution of 1:250 in TBS-T was added and the plate was incubated at room temperature for Ih.
  • HRP Horseradish peroxidase
  • ECL Enhanced Chemiluminescence
  • MDA-MB-231 cells in log phase were plated at 25,000 cells/well in 96-well opaque plates (Falcon, USA Cat # 353296) in 100 ⁇ L RPMI medium containing 10% (w/v) FBS, and incubated overnight at 37 0 C in 5% CO 2 incubator. The following day, the growth medium was removed from the plate by aspiration and replaced with RPMI medium containing 0.1% BSA and example compounds diluted to generate final concentrations of 0.0, 0.001, 0.003, 0.01, 0.03, 0.1, 0.3, 1 and 3 ⁇ M. Cells were incubated with compound for 1 hour at 37 0 C in a 5% CO 2 incubator.
  • the media was then removed from the plate by aspiration and the cells were washed once with 180 ⁇ L/well cold Tris Buffered Saline (TBS). After removal of the wash buffer, the cells were fixed by adding 180 ⁇ L of cold 3.7% (v/v) formaldehyde in TBS to each well for 1 hour at 4 0 C. After removal of the formaldehyde, the cells were treated with the addition of 60 ⁇ L of -2O 0 C methanol to each well for 5 minutes at 4 0 C. The methanol was removed and the cells were washed with 180 ⁇ L/well of 5% (w/v) BSA in TBS.
  • TBS Tris Buffered Saline
  • each well was treated with 180 ⁇ L/well 5% BSA (w/v) in TBS for thirty minutes at room temperature. After removal of the blocking buffer, 50 ⁇ L of an anti-phospho-p44/42 MAP kinase (Thr202/Tyr204) rabbit polyclonal antibody (Cell Signaling, USA Cat # 9101) was added to each well at a dilution of 1:1000 in 5% (w/v) BSA in TBS, and the plate was incubated at 4 0 C overnight. Plates were then washed three times with 300 ⁇ L/well TBS at room temperature.
  • HRP Horseradish peroxidase
  • Amersham, USA Cat. # NA934V Horseradish peroxidase
  • ECL Enhanced Chemiluminescence
  • the present invention relates to a compound which exhibits an IC 50 value of less than 3 ⁇ M.
  • the utility of the compounds of the present invention can also be illustrated, for example, by their activity in the flk-1 (murine VEGFR2) Assay described below.
  • the VEGF- VEGFR2 signaling pathway has been extensively characterized as an important regulator of angiogenesis and tumor angiogeneisis (See G. Yancopoulos et al, Vascular-specific growth factors and blood vessel formation, Nature, 2000, 407: p.. 242- 248; D. Shweiki et al, Induction of vascular endothelial growth factor expression by hypoxia and by glucose deficiency inmulticell spheroids: Implications for tumor angiogenesis, Proc. Natl. Acad. Sci, 1995, 92: p. 768-772).
  • VEGFR2 receptors inhibit the growth of a wide variety of tumors (See C. Brans et al, Vascular endothelial growth factor is an in vivo survival factor for tumor endothelium in a murine model of colorectal liver metastases, Cancer, 2000, 89: p. 495-499; B. Millauer et al, Glioblastoma growth inhibited in vivo by a dominant-negative FLK-I mutant, Nature, 1994, 367: p. 576-579).
  • Neutralizing antibodies to VEGF or VEGFR2 and VEGF antisense suppress tumor growth in vivo (See K.
  • This assay was performed in 96-well opaque plates (Costar, USA Cat #3915) in the TR- FRET format. Reaction conditions were as follows: 10 ⁇ M ATP, 25 nM poly (Glu,Tyr)- biotin (CIS BIO International, USA Cat#61 GTOBLD), 2 nM Eu-labelled phospho-Tyr Ab (Perkin Elmer, USA Cat#AD0067), 10 nM Strepavidin-APC (Perkin Elmer, USA Cat#CR130-100), 7 nM FIk-I (kinase domain), 1% DMSO, 50 mM HEPES pH 7.5, 10 mM MgCl 2 , 0.1 mM EDTA, 0.015% BRIJ, 0.1 mg/mL BSA, 0.1% mercapto-ethanol.
  • the present invention relates to a compound which exhibits an IC 50 value of less than 500 nM.
  • Another embodiment of the present invention thus relates to a method of using the compounds described above, including salts thereof and corresponding compositions thereof, as cancer chemotherapeutic agents .
  • This method comprises administering to a patient an amount of a compound of this invention, or a pharmaceutically acceptable salt thereof, which is effective to treat the patient's cancer.
  • a patient for the purpose of this invention, is a mammal, including a human, in need of treatment for a particular cancer.
  • Cancers include but are not limited to 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.
  • 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, and urethral 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, and lip and oral cavity cancer.
  • Lymphomas include, but are not limited to ADDS-related lymphoma, non- Hodgkin's lymphoma, cutaneous T-cell 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 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, chemotherapeutic, or other indication agents, and the like, as well as with admixtures and combinations thereof.
  • 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), such as cisplatin.
  • anti-hyper-proliferative agents suitable for use with this 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) such as idarubicin. C. Operative examples relating to pharmaceutical compositions
  • the active compound can act systemically, locally or both.
  • it can be administered in a suitable manner, such as for example by oral, parenteral, pulmonary, nasal, sublingual, lingual, buccal, rectal, dermal, transdermal, conjunctival or aural administration or in the form of an implant or stent.
  • the active compound can be administered in forms suitable for these modes of administration.
  • Suitable forms of oral administration are those according to the prior art which function by releasing the active compound rapidly or in a modified or controlled manner and which contain the active compound in a crystalline, amorphous, or dissolved form, for example tablets (which can be uncoated or coated, for example with enteric coatings or coatings which dissolve after a delay in time or insoluble coatings which control the release of the active compound), tablets or films (wafers), which disintegrate rapidly in the oral cavity, films/lyophilisates, capsules (e.g. hard or soft gelatin capsules), dragees, pellets, powders, emulsions, suspensions and solutions.
  • An overview of application forms is given in Remington's Pharmaceutical Sciences, 18 th ed. 1990, Mack Publishing Group, Enolo.
  • Parenteral administration can be carried out by avoiding an absorption step (e.g. by intravenous, intraarterial, intracardial, intraspinal or intralumbar administration) or by including absoiption (e.g. by intramuscular, subcutaneous, intracutaneous or intraperitoneal administration).
  • Suitable parenteral administration forms are for example injection and infusion formulations in the form of solutions, suspensions, emulsions, lyophilisates and sterile powders.
  • Such parenteral pharmaceutical compositions are described in Part 8, Chapter 84 of Remington's Pharmaceutical Sciences, 18 l ed. 1990, Mack Publishing Group, Enolo.
  • Suitable forms of administration for the other modes of administration are for example inhalation devices (such as for example powder inhalers, nebulizers), nasal drops, solutions and sprays; tablets or films/wafers for lingual, sublingual or buccal administration or capsules, suppositories, ear and eye preparations, vaginal capsules, aqueous suspensions (lotions or shaking mixtures), lipophilic suspensions, ointments, creams, transdermal therapeutic systems, milky lotions, pastes, foams, dusting powders, implants or stents.
  • inhalation devices such as for example powder inhalers, nebulizers
  • nasal drops solutions and sprays
  • tablets or films/wafers for lingual, sublingual or buccal administration or capsules, suppositories, ear and eye preparations, vaginal capsules, aqueous suspensions (lotions or shaking mixtures), lipophilic suspensions, ointments, creams, transdermal therapeutic systems, milky lotion
  • the active compounds can be converted into the abovementioned forms of administration in a manner known to the skilled man and in accordance with the prior art using inert, non-toxic, pharmaceutically suitable auxiliaries.
  • the latter include for example excipients (e.g. macrocrystalline cellulose, lactose, mannitol, etc.), solvents (e.g. liquid polyethylene glycols), emulsifiers and dispersants or wetting agents (e.g. sodium dodecyl sulfate, polyoxysorbitan oleate etc.), binders (e.g. polyvinyl pyrrolidone), synthetic and/or natural polymers (e.g. albumin), stabilizers (e.g. antioxidants, such as, for example, ascorbic acid), dyes (e.g. inorganic pigments such as iron oxides) or taste- and/or odour-corrective agents.
  • excipients e.g. macrocrystalline cellulose, lactose, manni
  • the total amount of the active ingredient to be administered will generally range from about 0.01 mg/kg to about 200 mg/kg, and preferably from about 0.1 mg/kg to about 20 mg/kg body weight per day.
  • 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.
  • the 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 daily oral dosage regimen will preferably be from 0.01 to 200 mg/kg of total body weight.
  • the compounds according to the invention are preferably isolated in more or less pure form, that is more or less free from residues from the synthetic procedure.
  • the degree of purity can be determined by methods known to the chemist or pharmacist (see Remington's Pharmaceutical Sciences, 18 th ed. 1990, Mack Publishing Group, Enolo).
  • the compounds are greater than 99% pure (w/w), while purities of greater than 95%, 90% or 85% can be employed if necessary.
  • the compounds according to the invention can be converted into pharmaceutical preparations as follows:
  • the mixture of active component, lactose and starch is granulated with a 5% solution (m/m) of the PVP in water. After drying, the granules are mixed with magnesium stearate for 5 min. This mixture is moulded using a customary tablet press (tablet format, see above). The moulding force applied is typically 15 kN.
  • a single dose of 100 mg of the compound according to the invention is provided by 10 ml of oral suspension.
  • Rhodigel is suspended in ethanol and the active component is added to the suspension.
  • the water is added with stirring. Stirring is continued for about 6h until the swelling of the Rhodigel is complete.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)

Abstract

This invention relates to novel 1 -Methyl- lH-pyrazole-4-carboxamide compounds, pharmaceutical compositions containing such compounds, and the use of those compounds or compositions as cancer chemotherapeutic agents.

Description

l-MethvI-lH-Pyrazole-4-Carboxamides Useful as Cancer Chemotherapeutic Agents
This invention relates to novel 1 -Methyl- lH-pyrazole-4-carboxamide compounds, pharmaceutical compositions containing such compounds, and the use of those compounds or compositions as cancer chemotherapeutic agents.
Many disease conditions are known to be associated with deregulated angiogenesis. Among these are retinopathies; chronic inflammatory disorders including arthritis; arteriosclerosis; atherosclerosis; macular degeneration; and neoplastic diseases such as cancer. In recent years, much work has been carried out to find inhibitors of angiogenesis, in hopes of developing treatments for such disorders.
WO 2004/063330 discloses (2-carboxamido)(3-amino)thiophene compounds for the treatment of cancer.
US patent 6,448,277 (Novartis) discloses and claims certain benzamide derivatives for inhibition of VEGF receptor tyrosine kinase, tumor growth, and VEGF-dependent cell proliferation.
Published PCT application WO 02/066470 (Amgen) broadly discloses heterocycles containing amido and amino substituent groups, for prophylaxis and treatment of angiogenesis-mediated diseases. Published PCT application WO 2004/005279 (Amgen) discloses certain substituted anthranilic amide derivatives for the prophylaxis and treatment of angiogenesis-mediated diseases. Published PCT application WO 2004/007458 (Amgen) relates to substituted 2-alkylamine nicotinic amide derivatives and their uses in treatment of cancer and other disorders.
Published PCT application WO 00/27819 (Schering) discloses certain anthranilic acid amides for treatment of diseases that are triggered by angiogenesis. Published PCT application WO 02/090352 (Schering) relates to selective anthranilamide pyridine amides as inhibitors of VEGFR-2 and VEGFR-3. Published PCT application WO 01/81311 (Schering) relates to substituted benzoic acid amides and use thereof for the inhibition of angiogenesis. Anthranilamides as angiogenesis inhibitors have been discussed in a series of research papers by scientists at Novartis and Schering. See Manley, et al., J. Med. Chem., 45, 5687-5693 (2002); Furet, et al., Bioorganic & Medicinal Chemistry Letters, 13., 2967- 2971 (2003); Manley, et al., Cell. MoI. Biol. Lett., 8, 532-533 (2003); and Manley, et al., Biochimica et Biophysica Acta, 1697, 17-27 (2004).
EP-B-832 061 discloses benzamide derivatives and their use as vasopressin antagonists.
The present invention relates to a compound of Formula (I)
Figure imgf000003_0001
wherein
Ar is selected from the group consisting of
Figure imgf000003_0002
Figure imgf000004_0001
and
Figure imgf000004_0002
X is CH or N;
R1 is selected from the group consisting of H, halogen,
Figure imgf000004_0003
O
R1-3
M 1-5
N'
I 1-3
R and O
R1 "3 R1-3 wherein
R1"2 is selected from the group consisting of
• H,
• (Ci-C4)alkoxy,
• (d-GOalkyl, wherein said (Q-C^alkyl can be substituted with O, 1, or 2 groups independently selected from
- hydroxy,
- (Ci-C4)alkylamino,
- (Q-GOacyloxy,
- (C1-C^aIkOXy, and - (C2-C4)alkoxy substituted with 0,1 or 2 (C!-C4)alkoxy groups,
- wherein said (Q-C-Oalkyl is independently optionally substituted with F up to the perfluoro level,
• 5- or 6-membered heteroaryl, or
• phenyl substituted with 0, 1, or 2 groups independently selected from (Q-GOalkyl, halo, nitro, (C1-C^aIkOXy and cyano;
R1"3 is H or (Ci-COalkyl;
R1"4, R1"5 and R1'6 are independently selected from the group consisting of
• H,
• indan-5-yl,
• phenyl substituted with 0, 1, or 2 groups independently selected from (C1-C4) kVl, halo, nitro, (C1-C4)alkoxy and cyano,
• 5- or 6-membered heteroaryl substituted with 0,1 or 2 groups selected from
- cyano,
- halo,
- nitro,
- (Ci-GOalkyl, wherein said (C1-C4)alkyl is optionally substituted with 0, 1, or 2 groups selected from
(Ci-GOalkylamino,
(Ci-C4)acyloxy,
(C1-C^aIkOXy, and
(C2-C4)alkoxy substituted with up to 0, 1 or 2 (C1-
C4)alkoxy groups,
• (C3-C6)cycloalkyl substituted with 0,1 or 2 groups selected from (C1-C4)alkyl, (Q-Gøalkoxy, cyano, and halo, and
• (Ci-QOalkyl, wherein said (CrC6)alkyl is independently substituted with 0 or 1 group selected from
- NH2,
Figure imgf000006_0001
- (C2-C4)alkoxy independently substituted with 0,1, 2 or 3 (Ci-C4)alkoxy and OH groups, and independently optionally substituted with fluorine up to the perfluoro level,
- carboxyl,
- (C1-C4)alkoxycarbonyl
- (Q-C^alkylamino,
- aminocarbonyl,
- (C1-C4)alkylsulfonyl, phenyl substituted with 0, 1, or 2 groups independently selected from (C1-C4)alkyl, halo, nitro, (Q-GOalkoxy and cyano,
- 5- or 6-membered heteroaryl independently substituted with 0, 1, 2 or 3 groups selected from (Ci-C4)alkyl, (C1- C4)alkoxy, cyano, halo, and nitro and
- heterocyclyl independently substituted with 0, 1, 2 or 3 groups selected from (Q-C^alkyl, (Q-GOalkoxy, cyano, and halo,
- and wherein said (CrC6)alkyl is independently substituted with 0, 1 or 2 OH or halo groups,
- and wherein said (Ci-C6)alkyl is independently optionally substituted with F up to the perfluoro level; and
R1"3 and R1"4, R1'3 and R1"5, and R1'3 and R1"6, when attached to the same nitrogen atom, may form, together with the N atom to which they are attached, a 5- or 6-membered saturated heterocyclic ring selected from pyrrolidinyl, morpholinyl, thiomorpholinyl and piperizinyl optionally substituted on N with (Q-C4)alkyl;
or a pharmaceutically acceptable salt thereof.
The invention also relates to pharmaceutical compositions which comprise a compound of Formula (I) as defined above plus a pharmaceutically acceptable carrier.
In addition, the invention relates to a method of treating cancer comprising administering to a subject in need thereof an effective amount of a compound of Formula (I) as defined above.
Pharmaceutically acceptable salts of the compounds (I) include acid addition salts of mineral acids, carboxylic acids and sulphonic acids, for example salts of hydrochloric acid, hydrobromic acid, sulphuric acid, phosphoric acid, methanesulphonic acid, ethanesulphonic acid, toluenesulphonic acid, benzenesulphonic acid, naphthalenedisulphonic acid, acetic acid, propionic acid, lactic acid, tartaric acid, malic acid, citric acid, fumaric acid, maleic acid and benzoic acid.
Pharmaceutically acceptable salts of the compounds (T) also include salts of customary bases, such as for example and preferably alkali metal salts (for example sodium and potassium salts, alkaline earth metal salts (for example calcium and magnesium salts) and ammonium salts derived from ammonia or organic amines having 1 to 16 carbon atoms, such as illustratively and preferably ethylamine, diethylamine, triethylamine, ethyldiisopropylamine, monoethanolamine, diethanolamine, triethanolamine, dicyclohexylamine, dimethylaminoethanol, procaine, dibenzylamine, N-methylmorpholine, dihydroabietylamine, arginine, lysine, ethylenediamine and methylpiperidine.
Solvates for the puiposes of the invention are those forms of the compounds that coordinate with solvent molecules to form a complex in the solid or liquid state. Hydrates are a specific form of solvates, where the coordination is with water. For the purposes of the present invention, the substituents have the following meanings, unless otherwise specified:
The terms "halogen" and "halo" mean Cl, Br, F and I, where Cl, Br and F are preferred.
The terms "(C1-C4)alkyl" and "(CrC6)alkyl" mean a linear or branched saturated carbon group having from about 1 to about 4 C atoms or from about 1 to about 6 C atoms, respectively. Such groups include but are not limited to methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, and the like.
The term "(C3-C6)cycloalkyl" means a saturated carbocyclic ring group having from about 3 to about 6 C atoms. Such groups include but are not limited to cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and the like.
The term "
Figure imgf000008_0001
means a linear or branched saturated carbon group having from about 1 to about 4 C atoms, said carbon group being attached to an O atom. The O atom is the point of attachment of the alkoxy substituent to the rest of the molecule. Such groups include but are not limited to methoxy, ethoxy, n-propoxy, isopropoxy, and the like.
The term "(Q-GOalkylamino" means an amino group having from one or two (independently selected) (Q-GOalkyl substituents, illustratively representing methylamino, ethylamino, n-propylamino, isopropylamino, teit-butylamino, n-pentylamino, n-hexylamino, N,N-dimethylamino, N,N-diethylamino, N-ethyl-N-methylamino, N-methyl-N- n-propylamino, N-isopropyl-N-n-propylamino, N-t-butyl-N-methylamino, iV-ethyl-iV- n-pentylamino, N-n-hexyl-N-methylamino and the like.
The term "(d-C^alkylsulfonyl" means a sulfonyl group having a (Q-C^alkyl substituent, illustratively representing methylsulfonyl, ethyl sulfonyl, isopropylsulfonyl, t- butylsulfonyl, and the like. The term "(CrC4)alkoxycarbonyl" means a (Ci-C4)alkoxygroup bound to the C atom of a carbonyl group [ -C(O) - ]said group being bound to the rest of the molecule, illustratively representing methoxycarbonyl, ethoxycarbonyl, /z-propoxycarbonyol, i- propoxycarbonyl, t-butoxycarbonyl, and the like
The term "(CrC4)acyloxy" means a (CrC4)group bound to the C atom of a carboxyl group [ -C(O)O- ], said group being bound by the oxygen atom to the rest of the molecule illustratively representing formyloxy, acetyloxy (acetoxy), propanoyloxy, butanoyloxy, t-butanoyloxy and the like
The term "5- or 6-membered heteroaryl" means, respectively,
(1) an aromatic ring made of 5 atoms and having 1, 2, 3 or 4 heteroatom(s) each selected independently from O, N, and S, the rest being C atoms, with the proviso that there can be no more than 1 O or S atom in the heteroaryl. This heteroaryl is attached to the core molecule at any available C or N atom and is optionally substituted at any available C or N atom with the recited substituents. Such groups include pyrrole, furan, thiophene, imidazole, pyrazole, thiazole, oxazole, isoxazole, isothiazole, triazole, oxadiazole, thiadiazole, and tetrazole in all their possible isomeric forms; or
(2) an aromatic ring made of 6 atoms, 1, 2, or 3 of which are N atoms, the rest being C, where the heterocycle is attached to the core molecule at any available C atom and is optionally substituted at any available C atom with the recited substituents. Such groups include pyridine, pyrimidine, pyridazine and triazine in all their possible isomeric forms.
The term "heterocyclyl" means a 5-or 6- membered saturated or partially saturated heterocyclic ring containing 1-2 heteroatoms selected from O, S or N, the remaining atoms being made up of C atoms, with the proviso that when there are 2 O atoms they must be nonadjacent. This heterocycle is attached to the core molecule at any available C or N atom and is optionally substituted at any available C or N atom with the recited substituents. Such groups include pyrrolidine, tetrahydrofuryl, tetrahydrothienyl, piperidinyl, tetrahydropyranyl, tetrahydrothiopyrano, piperizinyl, imidazolinyl, pyrazolinyl, morpholinyl, thiomorpholinyl and the like in all their possible isomeric forms. A * symbol next to a bond denotes the point of attachment in the molecule.
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. It is intended that all possible stereoisomers (including enantiomers and diastereomers) are included within the scope of the present invention. Preferred compounds are those with the absolute configuration of the compound of this invention which exhibits the more desirable biological activity. Separated, pure or partially purified stereoisomers or racemic mixtures of the compounds of this invention are also included within the scope of the present invention. The purification of said isomers and the separation of said stereoisomeric mixtures can be accomplished by standard techniques known in the art.
In another embodiment, the invention relates to a compound of Formula (I), wherein
Ar is selected from the group consisting of
Figure imgf000010_0001
and
Figure imgf000011_0001
X is CH or N;
R1 is selected from the group consisting of O
R1"3
Figure imgf000011_0002
and
R1-3 R1-3 wherein
R " is selected from the group consisting of
• (CrC4)alkyl, wherein said (Q-C-Oalkyl is substituted with 1 or 2 groups independently selected from
- hydroxy,
- (CrC4)alkylamino,
- (CrC4)acyloxy,
- (C1-C4OaIkOXy, and
- (C2-C4)alkoxy substituted with 0, 1 or 2 (Q-C4)alkoxy groups,
- wherein said (Q-C4)alkyl is independently optionally substituted with F up to the perfluoro level,
• 5- or 6-membered heteroaryl, or
• phenyl substituted with 1 or 2 groups independently selected from (Q-C-Oalkyl, halo, nitro, (C1-C4)EIkOXy and cyano;
R1-3 is H or (CrC4)alkyl;
R1"4 and R1"5 are independently selected from the group consisting of
• indan-5-yl,
• phenyl substituted with 1 or 2 groups independently selected from (Q-GOalkyl, halo, nitro, (C1-C4)alkoxy and cyano,
• 5- or 6-membered heteroaryl substituted with 1 or 2 groups selected from cyano,
- halo,
- nitro,
- (C1-C4)alkyl, wherein said (Q-GOalkyl is optionally substituted with 0, 1, or 2 groups selected from
(C1-C4)alkylamino,
(Ci-C4)acyloxy,
Figure imgf000012_0001
and
(C2-C4)alkoxy substituted with up to 0, 1 or 2 (C1-
C4)alkoxy groups,
• (C3-C6)cycloalkyl substituted with 1 or 2 groups selected from (C1-C4)alkyl, (C1-C4JaIkOXy, cyano, and halo, and . (C1-C6)BIkYl, wherein said (Q-C^alkyl is independently substituted with 1 group selected from
- NH2, (CrC4)alkoxy,
(C2-C4)alkoxy independently substituted with 0,1, 2 or 3 (d-C4)alkoxy and OH groups, and independently optionally substituted with fluorine up to the perfluoro level,
- carboxyl, (Q-GOalkoxycarbonyl (Q-C^alkylamino, aminocarbonyl,
- (Ci-COalkylsulfonyl,
- phenyl substituted with 0, 1, or 2 groups independently selected from (Q-GOalkyl, halo, nitro, (C1-C^aIkOXy and cyano,
5- or 6-membered heteroaryl independently substituted with 0, 1, 2 or 3 groups selected from (Q-G^alkyl, (C1- C4)alkoxy, cyano, halo, and nitro and
- heterocyclyl independently substituted with 0, 1, 2 or 3 groups selected from (Q-C^alkyl, (CrC4)alkoxy, cyano, and halo, and wherein said (Q-C^alkyl is independently substituted with 0, 1 or 2 OH or halo groups, and wherein said (CrC6)alkyl is independently optionally substituted with F up to the perfluoro level; R1" is selected from the group consisting of
• H
• indan-5-yl,
• phenyl substituted with 0, 1, or 2 groups independently selected from (Ci-C-Oalkyl, halo, nitro, (Q-C-Oalkoxy and cyano,
• 5- or 6-membered heteroaryl substituted with 0, 1 or 2 groups selected from
- cyano,
- halo, nitro,
- (C1-C^aIkVl, wherein said (Q-C4)alkyl is optionally substituted with 0, 1, or 2 groups selected from
(Q-C^alkylamino,
(Q-GOacyloxy,
(C1-C4)alkoxy, and
(C2-C4)alkoxy substituted with up to 0, 1 or 2 (C1-
C4)alkoxy groups,
• (C3-C6)cycloalkyl substituted with 0,1 or 2 groups selected from (Q-GOalkyl, (C1-C4)alkoxy, cyano, and halo, and
• (CrC6)alkyl, wherein said (Q-C^alkyl is independently substituted with 0 or 1 group selected from
- NH2,
- (CrC4)alkoxy,
- (C2-C4) alkoxy independently substituted with 0,1, 2 or 3 (Ci-C4)alkoxy and OH groups, and independently optionally substituted with fluorine up to the perfluoro level,
- carboxyl,
- (Q-GOalkoxycarbonyl
- (Q-C^alkylarnino,
- aminocarbonyl,
- (Q-GOalkylsulfonyl,
- phenyl substituted with 0, 1, or 2 groups independently selected from (C1-C4^aIkVl, halo, nitro, (CrC4)alkoxy and cyano,
- 5- or 6-membered heteroaryl independently substituted with 0, 1, 2 or 3 groups selected from
Figure imgf000014_0001
(C1- C4)alkoxy, cyano, halo, and nitro and - heterocyclyl independently substituted with 0, 1, 2 or 3 groups selected from (Q-GOalkyl, (Q-C4)alkoxy, cyano, and halo, and wherein said (Ci-C6)alkyl is independently substituted with 0, 1 or 2 OH or halo groups, and wherein said (C1-C6)alkyl is independently optionally substituted with F up to the perfluoro level; and
R1"3 and R1"4, R1"3 and R1"5, and R1"3 and R1"6, when attached to the same nitrogen atom, may form, together with the N atom to which they are attached, a 5- or 6-membered saturated heterocyclic ring selected from pyrrolidinyl, morpholinyl, thiomorpholinyl and piperizinyl optionally substituted on N with (Q-GOalkyl;
or a pharmaceutically acceptable salt thereof.
In another embodiment, the invention relates to a compound of Formula (I), wherein
Ar is selected from the group consisting of
Figure imgf000015_0001
and
Figure imgf000016_0001
X is CH;
R1 is selected from the group consisting of
Figure imgf000016_0002
and
Figure imgf000016_0003
wherein
R1"3 is H or (Ci-GOalkyl, R " is selected from the group consisting of indan-5-yl, phenyl substituted with 1 or 2 groups independently selected from
(CrC4)alkyl, halo, nitro, (Ci-C4)alkoxy and cyano, 5- or 6-membered heteroaryl substituted with 1 or 2 groups selected from cyano, halo, nitro, (C1-C4)alkyl, wherein said (Q-C-Oalkyl is optionally substituted with 0, 1, or 2 groups selected from (C1-C4)alkylamino, (CrC^acyloxy, (d-C4)alkoxy, and (C2-C4)alkoxy substituted with up to 0, 1 or 2 (Q-GOalkoxy groups;
(C3-C6)cycloalkyl substituted with 1 or 2 groups selected from (C1- C-øalkyl, (CrC4)alkoxy, cyano, and halo; and
(Ci-QOalkyl, wherein said (C1-C6)alkyl is independently substituted with lgroup selected from NH2,
(Q-GOalkoxy, (C2-C4)alkoxy independently substituted with
0,1, 2 or 3 (C1-C^aIkOXy and OH groups, and independently optionally substituted with fluorine up to the perfluoro level, carboxyl,
(Ci-C^alkoxycarbonyl (Q-C^alkylamino, aminocarbonyl, (Ci-C4)alkylsulfonyl, phenyl substituted with 0, 1, or 2 groups independently selected from (CrC4)alkyl, halo, nitro, (Ci-C4)alkoxy and cyano,
5- or 6-membered heteroaryl independently substituted with 0, 1, 2 or 3 groups selected from (Ci-C4)alkyl, (C1- C4)alkoxy, cyano, halo, and nitro and heterocyclyl is independently substituted with 0, 1, 2 or 3 groups selected from (CrC4)alkyl, (Q-C^alkoxy, cyano, and halo, and wherein said (C1-C6)alkyl is independently substituted with 0, 1 or 2
OH or halo groups, and wherein said (Q-C^alkyl is independently optionally substituted with F up to the perfluoro level; R1"6 is selected from the group consisting of H, indan-5-yl, phenyl substituted with 0, 1, or 2 groups independently selected from (Q-GOalkyl, halo, nitro, (Q-GOalkoxy and cyano, 5- or 6-membered heteroaryl substituted with 0, 1 or 2 groups selected from cyano, halo, nitro, (C1-C4)alkyl, wherein said (CrC4)alkyl is optionally substituted with 0, 1, or 2 groups selected from (C1-C4)alkylamino, (C1-C4)acyloxy, (Ci-COalkoxy, and (C2-C4)alkoxy substituted with up to 0, 1 or 2 (C1-C4)alkoxy groups; (C3-C6)cycloalkyl substituted with 0,1 or 2 groups selected from
(C1-C4)alkyl, (C1-C^aIkOXy, cyano, and halo; and
(Ci-QOalkyl, wherein said (C1-C6^kVl is independently substituted with 0 or 1 group selected from NH2,
(Ci-C4)alkoxy, (C2-C4)alkoxy independently substituted with
0,1, 2 or 3 (C1-C4)alkoxy and OH groups, and independently optionally substituted with fluorine up to the perfluoro level, carboxyl,
(C i -C4)alkoxycarbonyl (C1 -C4)alkylamino, aminocarbonyl, (Q-GOalkylsulfonyl, phenyl substituted with 0, 1, or 2 groups independently selected from (Ci-C4)alkyl, halo, nitro, (CrC4)alkoxy and cyano,
5- or 6-membered heteroaryl independently substituted with 0, 1, 2 or 3 groups selected from (C1-C4)alkyl, (C1- C4)alkoxy, cyano, halo, and nitro and heterocyclyl is independently substituted with 0, 1, 2 or 3 groups selected from (C1-C4)alkyl, (Q-C^alkoxy, cyano, and halo, and wherein said (C1-C6)alkyl is independently substituted with 0, 1 or 2
OH or halo groups, and wherein said (CrC6)alkyl is independently optionally substituted with F up to the perfluoro level; and
R1"3 and R1"5, and R1'3 and R1"6, when attached to the same nitrogen atom, may form, together with the N atom to which they are attached, a 5- or 6-membered saturated heterocyclic ring selected from pyrrolidinyl, morpholinyl, thiomorpholinyl and piperizinyl optionally substituted on N with (C1-C4)alkyl;
or a pharmaceutically acceptable salt thereof.
In another embodiment, the invention relates to a compound of Formula (I)
lected from the group consisting of
Figure imgf000020_0001
and
Figure imgf000020_0002
X is CH;
R1 is selected from the group consisting of
Figure imgf000020_0003
and
Figure imgf000020_0004
wherein
R1-3 is H,
Figure imgf000020_0005
wherein said (Ci-C6)alkyl is independently substituted with 1 group selected from (CrC4)alkoxy,
(C2-Q)alkoxy independently substituted with 0,1, or 2 (Q-GOalkoxy and OH groups, and independently optionally substituted with fluorine up to the perfluoro level, and wherein said (Q-C^alkyl is independently substituted with 0, 1 or 2
OH or halo groups, and wherein said (Q-C^alkyl is independently optionally substituted with F up to the perfluoro level; R1"6 is selected from the group H, and (Ci-QOalkyl, wherein said (C1-C6)alkyl is independently substituted with 0 or 1 group selected from (Ci-C4)alkoxy, (C2-C4)alkoxy independently substituted with
0,1, 2 or 3 (Ci-C4)alkoxy and OH groups, and independently optionally substituted with fluorine up to the perfluoro level, and wherein said (C1-Ce)alkyl is independently substituted with 0, 1 or 2
OH or halo groups, and wherein said (Q-C^alkyl is independently optionally substituted with F up to the perfluoro level;
or a pharmaceutically acceptable salt thereof.
In another embodiment, the invention relates to a compound of Formula (I)
lected from the group consisting of
Figure imgf000021_0001
Figure imgf000022_0001
and
Figure imgf000022_0002
X is CH;
R is selected from the group consisting of
Figure imgf000022_0003
wherein
R1"3 is H,
R1"6 is selected from the group
H, and (C1-C6)alkyl, wherein said (C1-C6)alkyl is independently substituted with 0 or 1 group selected from (CrC4)alkoxy, (C2-C4)alkoxy independently substituted with
0,1, 2 or 3 (C1-C^aIkOXy and OH groups, and independently optionally substituted with fluorine up to the perfluoro level, and wherein said (CrC6)alkyl is independently substituted with 0, 1 or 2
OH or halo groups, and wherein said (Q-C^alkyl is independently optionally substituted with F up to the perfluoro level; or a pharmaceutically acceptable salt thereof.
In another embodiment, the invention relates to a process for making a compound of formula (I), comprising (A) reacting, with or without first hydrolizing the ester group -COOR', a compound of formula (VI)
Figure imgf000023_0001
wherein X and R1 have the meaning described above, and R' is lower alkyl,
with a compound of formula Ar-NH2 (VIH),
wherein Ar has the meaning described above, for example in the presence of a coupling agent such as PyBOP; or
(B) reacting a compound of formula (EX)
Figure imgf000023_0002
wherein Ar has the meaning described above, with a compound of formula (IV)
Figure imgf000024_0001
wherein X and R1 have the meaning described above, and subsequently reducing the resulting compound; or
(C) reacting a compound of formula (IX), wherein Ar has the meaning described above, with a compound of formula (V)
Figure imgf000024_0002
wherein Ig represents a leaving group, such as halo, OTs or OMs; or
(D) reacting a compound of formula (Ha)
Figure imgf000024_0003
wherein X and R1 have the meaning described above, and R' is lower alkyl, with a compound of formula (VIII)
ArNH2 (Vm),
wherein Ar has the meaning described above, in the presence of (R')3A1, wherein R' is lower alkyl, or
(E) first hydrolizing the ester group -COOR' of a compound of formula (Ha) as described above, and subsequently reacting the resulting compound with a compound of formula ( VIH) as described above, for example in the presence of a coupling agent such as PyBOP.
General Methods of Preparation
Compounds of Formula (I) may be prepared by synthetic procedures known to those skilled in the art or by methods analogous thereto. These methods are summarized below in Reaction Scheme 1.
Unless otherwise specifically defined, R1 and X have the same meanings as defined hereinabove.
Reaction Scheme 1
Figure imgf000026_0001
n=1 or 2
Figure imgf000026_0002
(D As illustrated in Reaction Scheme 1, two general synthetic routes can be used to prepare the compounds of Formula (I).
In one route, the amino group of the compound of Formula (DI), is subjected to either reductive amination using a pyridine or pyrimidine carboxaldehyde of Formula (IV) and a reducing agent, such as sodium triacetoxyborohydride, or to direct N-alkylation using a pyridine or pyrimidine methyl halide, tosylate or mesylate of Formula (V) and a optional base such as pyridine or K2CO3, or a catalyst such as sodium iodide. The product formed, Formula (VI), is then allowed to react with an aromatic amine of Formula (VIII) in the presence of a coupling agent such as (R')3A1 (where R' = lower alkyl) giving the compound of Formula (I), or alternatively, the ester of Formula (VI) is hydrolyzed to the acid which is then coupled to the amine (VIII) using a coupling agent such as PyBOP.
In the second route, the compound of Formula (III) is converted to the aminoamide of Formula (IX) either directly by reaction with an aromatic amine of Formula (VIII) as described above, or by first protecting the amino function, e.g., as a BOC derivative (VII), and subsequent coupling with (VIII), either directly with (R')3A1, or via hydrolysis, and then coupling in the presence of PyBOP, followed by deprotection. The Formula (IX) compound is then converted to the Formula (I) compound using either the reductive amination method or direct N-alkylation as described above for preparation of (VI).
An additional route shown below in Scheme Ia is also possible. The amino group of the compound of formula (III) can be acylated using an acylating agent such as acetic anhydride and formic acid. N-alkylation of the acylated product Formula (II) using a pyridine or pyrimidine methyl halide, tosylate or mesylate of Formula (V) and a base such as DBU or K2CO3. The product formed, Formula (Ila), is then allowed to react with an aromatic amine of Formula (VIII) in the presence of a coupling agent such as (R')3A1 (where R' = lower alkyl) giving the compound of Formula (I) after a basic work up, or alternatively, the ester of Formula (Ila) is hydrolyzed to the acid in which the N-formyl group is also removed which is then coupled to the amine (VIII) using a coupling agent such as PyBOP. Reaction Scheme Ia
up, OT
Figure imgf000028_0001
PG removal (OH-) , e.g.,
Figure imgf000028_0002
Starting materials of Formulae (III), (IV), (V) and (VIE) are commercially available (e.g., Lanxess, Germany) or may be prepared by standard means well known in the art, or as described in Reaction Schemes 2-8.
Reaction Scheme 2
Figure imgf000028_0003
Compounds of Formula (Va),
O
[Formula (V) where R is H and Ig is Cl], may be prepared as shown in Reaction
Scheme 2 by reaction of an acid chloride with a chloromethyl heteroarylamine of Formula (X), generally in the presence of a base such as triethylamine.
Reaction Scheme 3
Figure imgf000029_0001
pg = protecting group, e.g., BOC
Ig = leaving group, e.g., halo, MsO, etc.
R1"5
Compounds of Formula (Vb) [Formula (V) where R1 is H ], can be prepared as shown in Reaction Scheme 3 from hydoxymethylheteroaryl amines of Formula (XI). Protection of the alcohol and conversion to the BOC-derivative of Formula (XIII) is followed by N-alkylation to give the intermediate of Formula (XIV). Deprotection of the alcohol and amine, followed by conversion of the hydroxy group to a leaving group, (for example, using SOCl2, when Ig is Cl) gives the intermediate of Formula (Vb). Reaction Scheme 4
Figure imgf000030_0001
(XVI) (XVII)
Figure imgf000030_0002
R' = lower alky I
Ig = leaving group, e.g., halo, MsO
R1-5 1 1 *?
Compounds of Formula (Vc) [Formula (V) where R is R " ] can be prepared by the route illustrated in Reaction Scheme 4. The chloroheteroarylcarboxylic acid derivative of Formula (XVI) is reduced to the chloroheteroaryl alcohol of Formula (XVII) with a standard reagent such as lithium borohydride. Reaction of the chloro compound with an amine of Formula (R1^)(R1 ~5)NH gives the intermediate alcohol of Formula (XViπ). Conversion of this alcohol to a leaving group, e.g. mesylate, completes the synthesis of the compound of Formula (Vc)
Reaction Scheme 5
Figure imgf000030_0003
O
Compounds of Formula (Vd) [Formula (V) where R1 is * NH2], can be prepared as shown in Reaction Scheme 5 from the dicarboxylic acid of Formula (XIX) by conversion through the half acid ester (XX) to the acid amide of Formula (XXI). Esterification of (XXI) provides (XXII) which can be reduced with sodium borohydride to the alcohol (XXHI) and then converted to the Formula (Vd) compound, using for example MsCl and a base such as triethylamine.
Reaction Scheme 6
Figure imgf000031_0001
(XXiV) (XXIi)
An alternate method of preparing the pyridine amide ester of Formula (XXII) is via the Minisci reaction shown in Reaction Scheme 6 in which the pyridine carboxylic acid ester is stirred in formamide with cooling to 10 °C in the presence of an equivalent of concentrated H2SO4, FeSO4 and H2O2.
Reaction Scheme 8
Figure imgf000031_0002
(X) (Vf)
O
Compounds of Formula (Vf) [Formula (V) where R1 is R R1"3 ] can be prepared by the route shown in Reaction Scheme 8. In the case that the R1"3 on the right is H, the intermediate of Formula (X) is allowed to react with an isocyanate of Formula R1-6NCO in an aprotic solvent such as dichloromethane. In the case that the R1'3 on the right is alkyl, or that R1"3 and R1"6 are combined in a cyclic structure, the intermediate of Formula (X) is allowed to react with a carbamoyl chloride Formula R1"6 R1-3NCOCl in an aprotic solvent such as dichloromethane in the presence of a base such as triethylamine or potassium carbonate. The use of a starting material of Formula (X) in which the R1"3 on the left is
O
alkyl results in the preparation of a urea of structure (Vf) where R1 is R R1"3 in which the R1"3 group on the left is alkyl.
In the cases that the isocyanate of Formula R1-6NCO is not commercially available
(and R " is H on the right side), it can conveniently be prepared by treatment of the amine of Formula R1-6NH2, wherein R1"6 is aryl or heteroaryl, with phosgene, diphosgene or triphosgene in a suitable solvent such as ethyl acetate. When R1"6 is alkyl or substituted alkyl, the preferred method is to treat the corresponding alkyl halide or dialkyl sulfate with inorganic cyanates. These methods, as well as others, are well known to those skilled in the art and examples are described in S. R. Sandler and W. Karo "Organic Functional Group Preparations," vol 12, 2nd ed., p 364-375, 1983, Academic Press and references cited therein.
In the cases that the carbamoyl chloride of Formula R1"6 R1-3NCOCl is not commercially available, it can conveniently be prepared by treatment of the amine of Formula R " R " NH with phosgene, diphosgene or triphosgene in a suitable solvent such as dichloromethane at 0-4 °C. Optionally, the N-benzyl protected amine of Formula R1"6 R1-3NCH2(C5H6) can be reacted with triphosgene as described by M.G. Banwell, et al, J. Org. Chem. 2003, 68, 613-616.
A variety of compounds of Formula (I) can be prepared by elaboration of compounds, also of Formula (I), prepared by the above schemes. These elaboration methods are illustrated below in Reaction Schemes 10-13.
Reaction Scheme 10
Figure imgf000033_0001
For example, the dimethyl pyrrole compound of Formula (Ij) is made by coupling [2-(2,5-dimethyl-lH-pyrrol-l-yl)pyridin-4-yl]methyl methanesulfonate with 5-amino-N- (2,2-difluoro-l,3-benzodioxol-5-yl)-l -methyl- lH-pyrazole-4-carboxamide as shown in Scheme 1. [2-(2,5-dimethyl-lH-pyrrol-l-yl)pyridin-4-yl]methyl methanesulfonate is made by coupling (2-aminopyridin-4-yl)methanol with hexane-2,5-dione using catalytic acid and then mesylation of the alcohol produced. The dimethyl pyrrole compound of Formula (Ij), can be deprotected to give a compound of Formula (Ia).
The amine can then be converted to the amide compound of Formula (Ib), the sulfonamide of Formula (Ic) or the urea of Formula (Id) as shown in Reaction Scheme 10, by reaction with an acid chloride, sulfonyl chloride or isocyanate, respectively.
Reaction Scheme 11
Figure imgf000034_0001
Additionally, the chloro compound of Formula (Ie) can be converted to the substituted amino compound of Formula (If) by reaction with an amine and a base such as pyridine in a sealed tube at elevated temperatures.
Reaction Scheme 12
Figure imgf000034_0002
(ig) (ih) do
Esters of Formula (Ih) and substituted amides of Formula (Ii) may be prepared from the unsubstituted amide of Formula (Ig) by the sequence illustrated in Reaction Scheme 12. Reaction of the amide (Ig) with dimethylformamide-dimethylacetal (DMF- DMA) in methanol provides the ester of Formula (Di); reaction of the ester with a substituted amine gives the amide of Formula (Ii). Generally, a desired salt of a compound of this invention can be prepared in situ during the final isolation and purification of a compound by means well known in the art. Or, a desired salt can be prepared by separately reacting the purified compound in its free base form with a suitable organic or inorganic acid and isolating the salt thus formed. These methods are conventional and would be readily apparent to one skilled in the art.
Additionally, sensitive or reactive groups on the compound of this invention may need to be protected and deprotected during any of the above methods. Protecting groups in general may be added and removed by conventional methods well known in the art (see, for example, T. W. Greene and P.G.M. Wuts, Protective Groups in Organic Synthesis; Wiley: New York, (1999).
By using the general schemes illustrated above and choosing the appropriate starting materials the compounds of the invention may be prepared. To further illustrate the invention, the following specific examples are provided, but are not meant to limit the scope of the invention in any way.
A. Examples
Abbreviations and Acronyms
When the following abbreviations are used throughout the disclosure, they have the following meaning: bm broad multiplet
BOC t-butoxycarbonyl bp boiling point bs broad singlet bt broad triplet
CD3CN acetonitrile-ύ?3
CD3OD methanol-^
Celite® registered trademark of Celite Corp. brand of diatomaceous earth d doublet
DMSO-J5 dimethylsulfoxide-6?(5
DMF ΛξN-dimethylformamide
EtOAc ethyl acetate h hour(s)
1H NMR proton nuclear magnetic resonance
HPLC high performance liquid chromatography
LCMS liquid chromatography / mass spectroscopy min minute(s) mL milliliter(s)
Ms methanesulfonyl m/z mass to charge ratio
PyBOP benzotriazole- 1 -yl-oxy-tris-pyrrolidino-phosphonium hexafluorophosphate rt room temperature
RT retention time (HPLC or LCMS)
S singlet t triplet
TFA trifluoroacetic acid
THF tetrahydrofuran TLC thin layer chromatography
Ts /7-toluenesulfonyl
General Analytical Procedures
The structure of representative compounds of this invention were confirmed using the following procedures.
Electron impact mass spectra (EI-MS) were obtained with a Hewlett Packard 5989A mass spectrometer equipped with a Hewlett Packard 5890 Gas Chromatograph with a J & W DB-5 column (0.25 μM coating; 30 m x 0.25 mm). The ion source is maintained at 250 0C and spectra were scanned from 50-800 amu at 2 sec per scan.
High pressure liquid chromatography-electrospray mass spectra (LC-MS) were obtained using either a:
(A) Hewlett-Packard 1100 HPLC equipped with a quaternary pump, a variable wavelength detector set at 254 nm, a YMC pro C-18 column (2 x 23 mm, 120A), and a Finnigan LCQ ion trap mass spectrometer with electrospray ionization. Spectra were scanned from 120-1200 amu using a variable ion time according to the number of ions in the source. The eluents were A: 2% acetonitrile in water with 0.02% TFA and B: 2% water in acetonitrile with 0.018% TFA. Gradient elution from 10% B to 95% over 3.5 min at a flowrate of 1.0 mL/min is used with an initial hold of 0.5 min and a final hold at 95% B of 0.5 min. Total run time is 6.5 min. or
(B) Gilson HPLC system equipped with two Gilson 306 pumps, a Gilson 215 Autosampler, a Gilson diode array detector, a YMC Pro C-18 column (2 x 23 mm, 120 A), and a Micromass LCZ single quadrupole mass spectrometer with z-spray electrospray ionization. Spectra were scanned from 120-800 amu over 1.5 seconds. ELSD (Evaporative Light Scattering Detector) data is also acquired as an analog channel. The eluents were A: 2% acetonitrile in water with 0.02% TFA and B: 2% water in acetonitrile with 0.018% TFA. Gradient elution from 10% B to 90% over 3.5 min at a flowrate of 1.5 mL/min is used with an initial hold of 0.5 min and a final hold at 90% B of 0.5 min. Total run time is 4.8 min. An extra switching valve is used for column switching and regeneration.
Routine one-dimensional NMR spectroscopy is performed on 400 MHz Varian
Mercury-plus spectrometers. The samples were dissolved in deuterated solvents obtained from Cambridge Isotope Labs, and transferred to 5 mm ID Wilmad NMR tubes. The spectra were acquired at 293 K. The chemical shifts were recorded on the ppm scale and were referenced to the appropriate solvent signals, such as 2.49 ppm for DMSO-<i<j, 1.93 ppm for CD3CN, 3.30 ppm for CD3OD 5.32 ppm for CD2Cl2 and 7.26 ppm for CDCl3 for 1H spectra.
Preparation of Intermediates
Intermediate A
Preparation of 5-amino-N-(2,2-difluoro-l.,3-benzodioxol-5-yl)-l-methvI-lH- pvrazoIe-4-carboxamide
Figure imgf000038_0001
Step 1: Preparation of ethyl 5- Fbis(tert-butoxycarbonyl)aminol-l -methyl- lH-pyrazole-
4-carboxylate
Figure imgf000038_0002
Ethyl S-amino-l-methyl-lH-pyrazole^-carboxylate (4.7 g, 27.78) in dichloromethane (100 mL) was stirred under nitrogen in a 500 mL flask as di-tert-butyl dicarbonate (7.88 g, 36.11 mmol) was added followed by N,N-dimethylpyridin-4-amine (339 mg, 2.78 mmol). The solution was stirred for 16 h. An additonal 5 g of di-tert-butyl dicarbonate was added and the solution was stirred for an additional 2 h. Very little starting material remained at this point and the solvents were evaporated to give a residue that was purified using silica gel. Products were eluted with a gradient from 0-80 % ethyl acetate in hexane to yield 8.1 g (79%) of the title pure material.
1H NMR (300 MHz, CD2C12-4) δ 7.85 (s, IH), 4.25 (q, 2H), 2.68 (s, 3H), 1.40 (s, 18H), 1.30 (t, 3H); ES-MS m/z 370.0 [M+H]+, LCMS RT (min) 3.17. Step 2: Preparation of 5-r(tert-butoxycarbonyl)aminol-l-methyl-lH-pyrazole-
4-carboxylic acid
Figure imgf000039_0001
A solution of ethyl 5~[bis(tert-butoxycarbonyl)armno]-l-methyl-lH-pyrazole- 4-carboxylate (8.0 g, 21.66 mmol) in ethanol (100 mL) was stirred under nitrogen as aqueous 1 N sodium hydroxide (108 mL) was added. The resulting mixture was stirred at 50 0C for 64 h, cooled to ambient temperature and then evaporated in vacuo. The pH was adjusted to 5-6 by slow addition of 2 N aqueous HCl. The product was extracted with ethyl acetate and then a 20% mixture of isopropanol in dichloromethane. The combined extracts were dried (Na2SO4) and evaporated in vacuo, and then toluene was added to the residue which was evaporated again to yield 3.5 g (67%) of pure dry product ready for the next step.
1H NMR (300 MHz, CD3OD-J4) δ 7.82 (s, IH), 3.75 (s, 3H), 1.5 (s, 9H); ES-MS m/z 241.9 [M+H]+, LCMS RT (min) 1.81.
Step 3: Preparation of 5-amino-N-(2,2-difluoro-l,3-benzodioxol-5-yl)-l -methyl- IH- pvrazole-4-carboxamide
Figure imgf000039_0002
A solution of 5-[(tert-butoxycarbonyl)amino]-l-methyl-lH-pyrazole- 4-carboxylic acid (1 g , 4.15 mmol), 2,2-difluoro-l,3-benzodioxol-5-amine (861 mg, 4.97 mmol), triethylamine (1.19 mL, 8.55 mmol) and (lH-l,2,3-benzotriazol-l- yloxy)(tripyrrolidin-l-yl)phosphonium hexafluorophosphate (PyBOP, 2.15 g, 4.15 mmol) in anhydrous dimethylformamide (20 niL) and dichloromethane (1 niL) was stirred at 60 °C for 12 h under nitrogen and then cooled. The resulting solution was diluted with ethyl acetate and washed with water and then saturated brine. The organic layer was dried (Na2SO4) and evaporated in vacuo. The resulting residue was chromatographed using silica gel with 0 - 100% ethyl acetate in hexane to yield tert-butyl (4-{ [(2,2-difluoro-l,3- benzodioxol-5-yl)amino]carbonyl}-l-methyl-lH-pyrazol-5-yl)carbamate (500 mg, >60% in purity) which was dissolved in dichloromethane (5 mL) and trifluoroacetic acid (3 niL) and was stirred under nitrogen for 2 h and evaporated in vacuo. The residue was dissolved in ethyl acetate and washed with saturated aqueous NaHCO3. The organic phase was dried (Na2SO4), evaporated in vacuo and purified using silica gel with 0-80 % ethyl acetate in hexanes to yield 250 mg of the title compound.
1H NMR (300 MHz, CD3OD-d4) δ 7.85 (bs, IH), 7.70 (s, IH), 7.25 (dd, IH), 7.10(dd, IH), 3.60 (s, 3H); ES-MS m/z 297.2 [M+H]+, LCMS RT (min) 3.03.
Intermediate B
Preparation of 5-amino-l-niethyI-N-(2,2,3,3-tetrafluoro-2.,3-dihydro-l,4- benzodioxin-6-vI)-lH-pyrazoIe-4-carboxamide
Figure imgf000040_0001
This intermediate can be prepared by using the method described above for the preparation of Intermediate A but using 2,2,3,3-tetrafluoro-2,3-dihydro-l,4-benzodioxin- 6-amine rather than 2,2-difluoro-l,3-benzodioxol-5-amine in Step 3. The pure product is characterized by NMR and LCMS spectroscopy.
Intermediate B -2
Preparation of 5-amino-l-methyI-N-(2,2,4,4-tetrafluoro-4H-l,3-benzodioxin-
6-vI)-lH-pyrazole-4-carboxamide
Figure imgf000041_0001
This intermediate can be prepared by using the method described above for the preparation of Intermediate A but using 2,2,4,4-tetrafluoro-4H-l,3-benzodioxin-6-amine rather than 2,2-difluoro-l,3~benzodioxol-5-amine in Step 3.
Intermediate B -3 Preparation of 5-amino-l-methyI-N-r4-(trifluoromethoxy)phenyn-4,5-dihydro- lH-pyrazole-4-carboxamide
Figure imgf000041_0002
This intermediate can be prepared by using the method described above for the preparation of Intermediate A but using 4-(trifluoromethoxy)aniline rather than 2,2- difluoro-l,3-benzodioxol-5-amine in Step 3.
Intermediate B -4
Preparation of 5-amino-l-methyI-N-r3-(trifluoromethoxy)phenyn-4,5-dihvdro- lH-pyrazoIe-4-carboxamide
Figure imgf000041_0003
This intermediate can be prepared by using the method described above for the preparation of Intermediate A but using 3-(trifluorometlioxy)aniline rather than 2,2- difluoro-l,3-benzodioxol-5-amine in Step 3.
Intermediate B -5
Preparation of 5-amino-l-methyl-N-{4-r(trifluoromethyl)thiolphenyl}-lH- pyrazoIe-4-carboxamide
Figure imgf000042_0001
This intermediate can be prepared by using the method described above for the preparation of Intermediate A but using 4-[(trifluoromethyl)thio] rather than 2,2-difluoro-l,3- benzodioxol-5-amine in Step 4.
Intermediate C Preparation of r2-(amϊnocarbonyI)pyridin-4-vnmethyI methanesulfonate
Figure imgf000042_0002
Step 1: Preparation of ethyl 2-(aminocarbonyl)isonicotinate
Figure imgf000042_0003
A solution of ethyl isonicotinate (25.2 mL, 165 mmol) in formamide (200 mL) was stirred with ice/methanol bath cooling as concentrated sulfuric acid (8.80 mL, 165 mmol) was added. Ferrous sulfate heptahydrate (69 g, 248 mmol) and hydrogen peroxide (25.6 mL of 30% in water) were added slowly over 25 min in alternating portions such that the temperature of the mixture was kept between 8-10.5 °C. During this addition small pieces of dry ice were added to the bath to keep the reaction temperature in the desired range. After the addition was complete, the ice bath was removed and the dark mixture was stirred for 2 h without cooling. The mixture was then poured into a solution of trisodium citrate dihydrate (80.6 g) in water (700 mL) and then residues left in the reaction flask were washed out with a little methanol and water. The resulting mixture was rapidly stirred in a large flask as solid NaHCO3 was added slowly, portion- wise, until the mixture was basic. Some saturated aqueous NaHCO3 was added to make the mixture more basic and then it was vacuum filtered through Celite® and the solids were washed down with three 200 mL portions of dichloromethane. The phases of the filtrate were separated and the aqueous layer was extracted twice with dichloromethane. The combined extract was dried
(Na2SO4) and evaporated in vacuo. The resulting solid residue was washed with ether/hexane (200 niL, 1:30) twice with warming and sonication followed by cooling and filtration to yield 13.9 g (44%) of pure title compound. The wash solutions, which contained some highly contaminated desired product, were discarded.
1H NMR (300 MHz, DMSO-J6) δ 8.83 (d, IH), 8.39 (d, IH, meta coupling), 8.24 (bs, IH), 8.00 (d, IH), 7.81 (bs, IH), 4.39 (q, 2H) and 1.37 ppm (t, 3H); ES-MS m/z 195.0 [M+H]+, HPLC RT (min) 1.83.
Step 2: Preparation of 4-(hvdroxymethyl)pyridine-2-carboxamide
A slurry of ethyl 2-(aminocarbonyl)isonicotinate (5.00 g, 25.8 mmol) in absolute ethanol (150 mL) was stirred under nitrogen as sodium borohydride (2.92 g, 77.2 mmol) was added. After 22 h stirring at ambient temperature, the reaction was carefully quenched by addition of 17 mL of saturated aqueous ammonium chloride followed by stirring until the bubbling stopped and then evaporation in vacuo to leave a white solid residue. Saturated aqueous sodium chloride (80 mL) was added followed by five extractions with 200 mL portions of ethyl acetate. Combined extracts were dried (Na2SO4) and evaporated in vacuo to yield 3.85 g (98%) of pure title compound as a white solid.
1H NMR (300 MHz, DMSO-J6) δ 8.52 (d, IH), 8.00 (s, IH), 8.07 (bs, IH), 7.46 (d, IH), 7.60 (bs, IH), 5.54 (t, IH) and 4.60 ppm (d, 2H); ES-MS m/z 154.0 [M+H, weak signal]"1", HPLC RT (min) 1.05. Step 3: Preparation of r2-(aminocarbonyl)pyridin-4-vnmethyl methanesulfonate
Figure imgf000043_0002
4-(hydroxymethyl)pyridine-2-carboxamide (1.00 g, 6.57 mmol) was dissolved in ethyl acetate (80 mL) and then cooled to 0 0C with stirring under nitrogen in an ice bath before triethylamine (1.37 mL, 9.86 mmol) was added, followed by methanesulfonyl chloride (0.66 mL, 8.54 mmol, added dropwise over 7 min). The ice bath was removed and the resulting suspension was stirred 2 h, and then the reaction mixture was poured into 60 mL water and stirred rapidly for 10 min. The phases were separated and the aqueous was extracted twice more with ethyl acetate. Each extract was washed with brine and the combined extracts were dried (Na2SO4) and evaporated in vacuo to yield 1.50 g (99%) of pure product as a fine white solid which turned pink on storage. Re-assay by NMR after such color change did not show significant decomposition.
1H NMR (300 MHz, DMSO-J6) δ 8.64 (d, IH), 8.06 (s, IH), 8.14 (bs, IH), 7.6 (d, IH), 7.70 (bs, IH), 5.41 (s, 2H) and 3.33 ppm (s, overlaps with water in solvent).
Intermediate D Preparation of {2-r(methyIamino)carbonγπpyridin-4-yl}methvI methanesulfonate
Figure imgf000044_0001
This compound was prepared by using the method described above for Intermediate C but starting with methyl formamide rather than formamide in step 1 and methanesulfonic anhydride rather than methanesulfonyl chloride in step 3.
1H NMR (300 MHz, DMSO-J6) 6 8.80 (bs, IH), 8.64 (d, IH), 8.03 (s, IH), 7.58 (d, IH), 5.41 (s, 2H), 3.29 (s, 3H) and 2.80 ppm (d, 3H); ES-MS m/z 145.1 [M+H]+, HPLC RT (min) 1.43.
Intermediate E Preparation of 2-{r4-(chloromethyI)pyridin-2-yllamino>-2-oxoethvI acetate
Figure imgf000044_0002
Step 1: Preparation of 4-(chloromethyl')pyridin-2~amine
Figure imgf000044_0003
(2-Aminopyridin-4-yl)methanol (11.2 g, 90 mmol) was stirred in a flask with ice bath cooling as thionyl chloride (65.8 mL, 902 mmol) was slowly added. After about 10 mL was added, the temperature increased suddenly to about 50 0C and addition was halted as the mixture was broken up so that stirring could continue as the rest of the thionyl chloride was added. The cooling bath was then removed and the reaction was stirred for 2 h at ambient temperature before it was evaporated in vacuo and then toluene was added twice and evaporated each time in vacuo to yield the hydrochloride salt of the title compound. A suspension of this material in dichloromethane (150 mL) was stirred with saturated aqueous sodium bicarbonate (150 mL) for 1.5 h. The phases were separated and the organic extract was washed twice with water, once with brine and then dried (Na2SO4) and evaporated in vacuo to yield 10.71 g (83%) of pure title compound.
1H NMR (300 MHz, DMSO-J6) 67.87 (d, IH), 6.48 (d, IH), 6.45 (s, IH), 6.04 (s, 2H) and 4.60 ppm (s, 2H); ES-MS m/z 143.2 [M+H]+, HPLC RT (min) 1.34.
Step 2: Preparation of 2-{r4-(chloromethyl)pyridin-2-yl1amino|-2-oxoethyl acetate
Figure imgf000045_0001
A suspension of 4-(chloromethyl)pyridin-2-amine (2.50 g, 10 mmol) and triethylamine (11.7 mL) in dichloroethane (10 mL) was stirred under nitrogen with ice bath cooling as acetoxyacetyl chloride (1.86 mL, 17 mmol) was added slowly over 10 min. After 2 h stirring with cooling, TLC showed no starting material but three major product spots. The mixture was diluted with dichloromethane and washed with water and then brine. It was dried (Na2SO4) and evaporated in vacuo. The residue was purified by chromatography on silica gel using a gradient from 0-3% methanol in dichloromethane to yield 0.62 g (18%) of the correct and pure title compound.
1H NMR (300 MHz, DMSO-J6) δ 10.75 (s, IH), 8.30 (d, IH), 8.10 (bs, IH), 7.17 (d, IH), 4.79 (s, 2H), 4.71 (s, 2H) and 2.13 ppm (s, 3H); ES-MS m/z 243.1 [M+H]+, HPLC RT (min) 1.87.
Intermediate F Preparation of N-r4-(chIoromethyI)pyridin-2-yllacetamide
Figure imgf000046_0001
By using the methods described for preparation of Intermediate E and by substituting acetyl chloride instead of acetoxyacetyl chloride in step 2, Intermediate F was prepared from 2.30 g of 4-(chloromethyl)pyridin-2-amine and proportional amounts of other reagents. The yield of title compound was 2.0 g (67%) after silica gel chromatography. Even though examination of this material by NMR spectroscopy indicated that it was a mixture of the desired compound and the diacylated product N- acetyl-N-[4-(chloromethyl)pyridin-2-yl]acetamide (about 45:55), it was used as is in the next reaction and side products were separated by chromatography after the subsequent reaction.
1H ΝMR (300 MHz, CD2Cl2) δ 8.33 (bs, IH), 7.41 (d, IH), 7.30 (s, IH), 7.10 (d, IH), 4.65 (s, 2H) and 2.20 ppm (s, 3H); ES-MS m/z 185.0 [M+H]+, HPLC RT (min) 1.16. Signals for the contaminating diacyl compound show at 1H ΝMR (300 MHz, CD2Cl2) δ 8.56 (d, IH), 8.18 (s, IH), 78.24 (d, IH), 4.75 (s, 2H) and 2.25 ppm (s, 6H); ES-MS m/z no significant M+H+ion, HPLC RT (min) 0.97. Because of the closeness of the % content of the two compounds, it is possible that some of the ΝMR peak assignments have been switched between the desired material and the contaminant.
Intermediate G Preparation of N- r4-(chloromethyI)pyridin-2-yIl -2-methoxyacetamide
Figure imgf000046_0002
By using the methods described for preparation of Intermediate E and by substituting 2-methoxyacetyl chloride instead of acetoxyacetyl chloride in step 2, Intermediate G was prepared from 731 mg of 4-(chloromethyl)pyridin-2-amine and proportional amounts of other reagents. The yield of pure title compound was 397 mg (45%) after silica gel chromatography using a gradient from 0-40% ethyl acetate in hexane.
1H NMR (300 MHz, CDCl3) δ 9.00 (bs, IH), 8.31 (d, IH), 8.30 (s, IH), 7.13 (d,
IH), 4.55 (s, 2H), 4.06 (s, 2H) and 3.51 ppm (s, 3H); ES-MS m/z 215.0 [M+H]+, HPLC RT (min) 0.71.
Intermediate H Preparation of iV-r4-(chloromethyl)pyridin-2-yll-2-(2-methoxyethoxy)acetamide
Figure imgf000047_0001
By using the methods described for preparation of Intermediate E and by substituting 2-(2-methoxyethoxy)acetyl chloride instead of acetoxyacetyl chloride in step 2, Intermediate H was prepared from 599 mg of 4-(chloromethyl)pyridin-2-amine and proportional amounts of other reagents. The yield of pure title compound was 314 mg (29%) after silica gel chromatography twice, first using a gradient from 2-3% methanol in dichloromethane, and then a second chromatography of the best fractions using a gradient from 0-40% ethyl acetate in hexane.
1H NMR (300 MHz, CD2Cl2) 6 9.39 (bs, IH), 8.30 (d, IH), 8.29 (s, IH), 7.13 (d, IH), 4.59 (s, 2H), 4.14 (s, 2H), 3.76 (t, 2H), 3.60 (t, 2H) and 3.44 ppm (s, 3H); ES-MS m/z 259.1 [M+H]+, HPLC RT (min) 1.46.
Intermediate I Preparation of N- r4-(chloromethyI)pγridin-2-yπ -2-methoxypropanamide
Figure imgf000047_0002
Step 1 : Preparation of 2-methoxypropanoic acid
Figure imgf000047_0003
Sodium methoxide in methanol (25%, 16 mL) was added to a stirred solution of 2- bromopropionic acid (19.6 mmol) in methanol (5 mL) under nitrogen. The reaction was heated at 50 °C under nitrogen overnight. The reaction was then concentrated under vacuum. The residue was brought to pH 1 by the addition of 1 N aqueous HCl and this solution was then extracted with ethyl acetate three times (70 mL, 25 mL, 10 mL). The combined organic layer was dried (Na2SO4) and then concentrated under vacuum to yield the title compound as a colorless oil 2.04 g (99%) which was of sufficient purity to be used without purification. 1H NMR (CD3OD) δ 3.67 (q, IH), 3.33 (s, 3H), and 1.33 ppm
(d, 3H).
Step 2: Preparation of 2-methoxypropanoyl chloride
Figure imgf000048_0001
2-Methoxyproρanoic acid (2.04 g, 19.2 mmol) was dissolved in dichloromethane (3 mL) which was stirred under nitrogen as a drop of dimethylformamide was added. Thionyl chloride was added dropwise into the reaction over 3 min and then the reaction was stirred at room temperature overnight. The reaction solution was concentrated in vacuo and the resulting pale yellow oil was placed under high vacuum to remove last traces of thionyl chloride. The yield of pure title compound was 303 mg (13%). 1HNMR (CDCl3) δ 4.10 (q, IH), 3.48 (s, 3H), and 1.56 ppm (d, 3H). Step 3: Preparation of N-r4-(chloromethyl)pyridin-2-yl1-2-methoxypropanamide
Figure imgf000048_0002
By using the methods described for preparation of Intermediate E (Step 2) and by substituting 2-methoxypropanoyl chloride instead of acetoxyacetyl chloride, Intermediate I was prepared from 352 mg of 4-(chloromethyl)pyridin-2-amine and proportional amounts of other reagents. The yield of pure title compound was 341 mg (60%) after silica gel chromatography using a gradient from 0-30% ethyl acetate in hexane.
1H NMR (300 MHz, DMSOd6) δ 10.2 (bs, IH), 8.30 (d, IH), 8.17 (s, IH), 7.16 (d, IH), 4.77 (s, 2H), 4.00 (q, IH), 3.26 (s, 3H), and 1.27 ppm (d, 6H).
Intermediate J
Preparation of iV-r4-(chIoromethyl)pyridin-2-vn-2-methoxy-2-methyIpropanamide
Figure imgf000048_0003
Step 1 : Preparation of 2-methoxy-2-methylpiOpanoic acid
Figure imgf000049_0001
The procedure of Weizmann, Sulzbacher, and Bergmann as written in JACS 70,1153 (1948), which is hereby incorporated by reference, was used as follows: A solution of potassium hydroxide (8.96 g, 159.7 mmol) in 5 mL of water and 20 niL of methanol was stirred with ice bath cooling under nitrogen as 1,1 , l-trichloro-2- methylpropan-2-ol (7.10 g, 40.0 mmol) was carefully added dropwise over ten min. Vigorous bubbling was observed as a white precipitate formed. The ice bath was removed after 15 min. The reaction was stirred at room temperature for 2 h then refluxed for 3 h. The reaction was cooled to room temperature and the solids were then removed by filtration and rinsed with methanol (350 mL). The filtrate was concentrated under vacuum to remove methanol and the remaining aqueous layer was brought to pH 0 by the addition of aqueous HCl then extracted with ethyl acetate (300 mL). The extract was dried (Na2SO4) and concentrated in vacuo to yield 4.11 g of crude product, which was purified by vacuum distillation to yield 2.28 g (48%) of the pure title compound as a colorless oil which was distilled at 105 0C (28 mm Hg). 1HNMR (CDCl3) δ 9.65 (s, IH), 3.20 (s, 3H) and 1.32 ppm (s, 6H). Step 2: Preparation of 2-methoxy-2-methylpropanoyl chloride
O CH3
By following the procedure of Intermediate I (Step 2) but using 2-methoxy-2- methylpropanoic acid (6.99 g, 59.2 mmol) rather than 2-methoxypropanoic acid and proportional amounts of other reagents the title compound was synthesized. The crude product was distilled in vacuo to yield 2.671 g (33%) of pure compound, bp 44-48 0C (38 mm Hg).
IHNMR (CDCl3) δ 3.33 (s, 3H) and 1.51 ppm (s, 6H). Step 3: Preparation of iV-r4-(chloromethyl)pyridin-2-vn-2-methoxy-2-methylpropanamide
Figure imgf000049_0002
By using the methods described for preparation of Intermediate E (Step 2) and by substituting 2-methoxy-2-methylpropanoyl chloride instead of acetoxyacetyl chloride, Intermediate J was prepared from 1.04 g of 4-(chloromethyl)pyridin-2-amine and proportional amounts of other reagents. The yield of title compound was 1.23 g (69%) after silica gel chromatography using 30% ethyl acetate in hexane.
1H NMR (300 MHz, DMSO-^6) δ 9.41 (bs, IH), 8.32 (d, IH), 8.16 (s, IH), 7.19 (d, IH), 4.78 (s, 2H), 3.28 (s, 3H) and 1.36 ppm (s, 6H); ES-MS m/z 243.1 [M+H]+, HPLC RT (min) 2.12.
Intermediate K Preparation of N- r4-(chloromethyl)pyridin-2-vπmethanesuIfonamide
Figure imgf000050_0001
Step 1: Preparation of N-r4-(chloromethyl)pyridin-2-yll-N-(methylsulfonyl) methanesulfonamide
Figure imgf000050_0002
A solution of 4-(chloromethyl)pyridin-2-amine (500 mg, 3.51 mmol) and triethylamine (1.47 mL, 10.5 mmol) in ethyl acetate (4 mL) was stirred under nitrogen in a flask with ice bath cooling as methanesulfonyl chloride (0.81 mL, 10.5 mmol) was added dropwise. The reaction was then allowed to stir without cooling for 1 h before it was diluted with additional ethyl acetate, washed with water, dried (Na2SO4) and evaporated in vacuo. The resulting residue was purified by chromatography on silica gel using an ethyl acetate/ hexane gradient to yield 860 mg (82%) of pure title compound. 1H NMR (300 Hz, CD2Cl2) δ 8.56 (d, IH), 7.50 (d, IH), 7.41 (s, IH), 4.66 (s, 2H), and 3.55 ppm (s, 6H); ES-MS m/z 299.0 [M+H]+, HPLC RT (min) 2.08. Step 2: Preparation of N-r4-(chloromethyl)pyridin-2-yl1methanesulfonamide
Figure imgf000050_0003
A suspension of N-[4-(chloromethyl)pyridin-2-yl]-N-(methylsulfonyl)- methanesulfonamide (700 mg, 2.34 mmol) in methanol (10 mL) and aqueous sodium hydroxide (1 N, 11.7 mL, 11.7 mmol) was stirred at ambient temperature as the starting material dissolved over 10 min. After another 10 min the reaction was adjusted to a pH between 3 and 6 by addition of aqueous hydrochloric acid (2 N) to precipitate the desired product as a white solid that was collected by filtration, washed with methanol and dried in vacuo. The yield of title compound was 250 mg (48%).
1H NMR (300 MHz, DMSO-J6) δ 10.93 (bs, IH), 8.21 (d, IH), 7.02 (m, 2H), 4.73 (s, 2H), and 3.23 ppm (s, 3H); ES-MS m/z 221.1 [M+H]+, HPLC RT (min) 1.45.
Intermediate L Preparation of N44-(chIoromethyI)pyridin-2-yl1-N'-ethyIurea
Figure imgf000051_0001
To 4-(chloromethyl)pyridin-2-amine (lOOmg, 0.70mmol) in 3 mL DMF was added ethyl isocyanate (59 mg, 0.84 mmol) and the resulting mixture was stirred under nitrogen for 16 h. The reaction was diluted with EtOAc (15 mL) and washed with H2O three times, dried (Na2SO4) and evaporated in vacuo. The crude residue was purified by column chromatography on silica gel using 25% EtOAc in hexane to give 110 mg of N- [4- (chloromethyl)pyridin-2-yl]-7V1-ethyrurea (73 %).
1H NMR (DMSO-J6) δ 9.22 (s, IH), 8.14-8.16(m, IH), 7.91-7.94 (m, IH), 7.45 (d, J=0.8Hz, IH), 6.93-6.95 (m, IH), 4.70 (s, 2H), 3.12-3.14(m, 2H), 1.01-1.09(m, 3H) ppm; LCMS: 214.1 [M+H]+, RT 0.47 min.
Intermediate M Preparation of N-r4-(chloromethyI)pyridin-2-yI1-iV'-phenylurea
Figure imgf000051_0002
By using the methods described for preparation of Intermediate L and by substituting phenyl isocyanate instead of ethyl isocyanate, Intermediate M was prepared. From 250 mg of 4-(chloromethyl)pyridin-2-amine and proportional amounts of other reagents the yield of title compound was 218 mg (47%) after silica gel chromatography using a gradient from 0-40 % ethyl acetate in hexane. Though there was evidence of contamination with the starting material 4-(chloromethyl)pyridin-2-amine in the NMR spectrum, this material was used without further purification and side products were separated by chromatography after the next step.
1H NMR (300 MHz, DMSO-J6) δ 10.25 (bs, IH), 9.50 (bs, IH), 8.29 (d, IH), 7.95 (s, IH), 7.52 (d, IH), 7.27-7.36 (m, 2H), 7.0-7.1 (m, 2H), and 4.79 ppm (s, 2H); LCMS: 262.2 [M+H]+, RT 2.65 min.
Intermediate N Preparation of N44-(chloromethyI)pyridin-2-yI1-iV'-methyIurea
Figure imgf000052_0001
By using the methods described for preparation of Intermediate L and by substituting methyl isocyanate instead of ethyl isocyanate, Intermediate N was prepared. From 180 mg of 4-(chloromethyl)pyridin-2-amine and proportional amounts of other reagents the yield of pure title compound was 42 mg (17%) after silica gel chromatography using a gradient from 0-50 % ethyl acetate in hexane followed by trituration of the residue with ether to remove a contaminant.
1H NMR (DMSO-J6) 6 9.31 (s, IH), 8.16 (d, IH), 7.92 (bm, IH), 7.40 (s, IH), 6.93 (d, IH), 4.69 (s, 2H) and 2.70 ppm (d, 3H); LCMS: 200.1 [M+H]+, RT 1.17 min.
Intermediate NN Preparation of N' 44-(ehloromethyl)pyridin-2-yI1-N,N-dimethyIurea
Figure imgf000052_0002
By using the methods described for preparation of Intermediate K and by substituting dimethylcarbamic chloride instead of methanesulfonyl chloride, Intermediate NN is prepared.
Intermediate O Preparation of 2,4-dichloro-6-(chIoromethvDpvrimidine
Figure imgf000053_0001
This product was prepared similarly to the 5-methyl substituted analog described in Biorg. Med. Chem. 2002, 10, 525. A stirred suspension of 6-(chloromethyl)pyrimidine- 2,4(lH,3H)-dione (5.2 g, 32.6 mmol) in POCl3 (9.1 mL, 97.9 mmol) was refluxed for 16 h under nitrogen. The mixture was cooled and evaporated to leave a dark colored oil. Ice water was slowly added and the product was extracted into dichloromethane. The organic layer was washed with brine, dried over MgSO4, and concentrated under reduced pressure to give 2,4-dichloro-6-(chloromethyl)pyrimidine (5 g) as a yellow oil. Though this product could be used in the next step with out purification, another batch prepared in the same way was further purified by chromatography to show the following NMR.
1H NMR (DMSO-J6) δ 7.90 (s, IH) and 4.78 ppm (s, 2H).
Intermediate P Preparation of 2-chIoro-4-(chIoromethγI)pyridine
Figure imgf000053_0002
Step 1: Preparation of (2-chloropyridin-4-yl)methanol
Figure imgf000053_0003
A sample of methyl 2-chloroisonicotinate (5.00 g, 29.14 mmol) was dissolved in 10 mL THF, treated with 10 drops of methanol, and cooled to 0 0C. The solution was treated with lithium borohydride solution (21.86 mL of 1 M in THF, 43.71 mmol) and then allowed to warm to room temp. After 4 h the solution was cooled to 0 0C and quenched with 1 N HCl solution. The pH was adjusted to pH 10 with 1 N NaOH solution, and the reaction mixture was extracted with EtOAc. The organic extracts were washed with brine and concentrated in vacuo yielding 2.96 g (70.8%) of product.
1H NMR (300 MHz, CD3CN) δ 8.32 (d, 1 H), 7.39 (s, 1 H), 7.29 (d, 1 H), 4.62 (s, 2 H) and 3.53 ppm (bs, 1 H). Step 2: Preparation of 2-chloro-4-(chloromethyl)pyridine
Figure imgf000054_0001
A sample of (2-chloropyridin-4-yl)methanol (110.0 mg, 0.77 mmol) was dissolved in anhydrous THF (1.5 mL), treated with N,iV-diisopropylethylamine (0.29 niL, 1.69 mmol) and cooled to -78 0C. Methanesulfonyl chloride was added (0.07 mL, 0.84 mmol), and the reaction mixture was allowed to slowly warm to room temperature overnight. The reaction mixture was then diluted with dichloromethane and washed with water. The organic layer was dried over Na2SO4 and concentrated in vacuo yielding the title compound (110.0 mg, 88.6%).
1H NMR (300 MHz, CD3CN) δ 8.40 (d, 1 H), 7.49 (s, 1 H), 7.39 (d, 1 H) and 4.63 ppm (s, 2 H); ES-MS m/z 183.2 [M+Na]+, HPLC RT (min) 2.30.
Intermediate PP
Preparation of [2-(2,5-dimethyl- 1 H-pyrrol- 1 -yl)pyridin-4-yl1methyl methanesulfonate
Figure imgf000054_0002
Step 1: Preparation of r2-(2,5-dimethyl-l H-pyrrol- l-yl)pyridin-4-ynmethanol
Figure imgf000054_0003
(2-aminopyridin-4-yl)methanol (7.5 g, 60.41 mmol), hexane-2,5-dione (7.58 g, 66.46 mmol) and p-toluenesulfonic acid monohydrate(1.14 g, 6.04) was dissolved in Toluene (10 mL) in a flask fitted with a Dean Stark trap. The solution was heated to reflux at 135 0C for 16 h. The solvents were evaporated and ethyl acetate was added. The organic extracts were washed with brine, dried (Na2SO4) and concentrated in vacuo. The residue was purified using silica gel with a gradient elution from 10-50% ethyl acetate in hexanes to yield 10.2 g (83%) of product.
1H NMR (300 MHz, CD2C12-J2) δ 8.52 (dd, IH), 7.30 (m, IH), 7.22 (m, IH), 5.85(s, 2H), 4.78 (s, 2H), 2.08 (s, 6H); ES-MS m/z 203.2[M+H]+, LCMS RT (min) 2.04.
Step 2: Preparation of r2-(2,5-dimemyl-lH-pyrrol-l-yl)pyridin-4-yllmethyl methanesulfonate
Figure imgf000055_0001
A sample of [2-(2,5-dimethyl-lH-ρyrrol-l-yl)pyridin-4-yl]methanol (6.80 g, 33.62 mmol) was dissolved in anhydrous dichloromethane (30 mL), treated with triethylamine (14.06 mL, 100.9 mmol) and cooled to 0 0C. Methanesulfonyl chloride was added (3.90 mL, 50.43 mmol), and the reaction mixture was allowed to slowly warm to room temperature overnight. The reaction mixture was then diluted with dichloromethane and washed with water. The organic layer was dried over Na2SO4 and concentrated in vacuo and the residue was purified over silica gel using 10-60% ethylacetate in hexanes as eluant to yield the title compound (7.5 g, 80%).
1H NMR (300 MHz, CD2C12-J2) δ 8.65 (dd, IH), 7.35 (m, IH), 7.25 (m, IH), 5.88(s, 2H), 5.30 (s, 2H), 3.10 (s, 3H), 2.12 (s, 6H); ES-MS m/z 281.1[M+H]+, LCMS RT (min) 2.67.
Intermediate Q Preparation of 4-(chloromethvI)-N-(4-methyl-l,3-thiaζoI-2-yI)pyridin-2-amine
Figure imgf000055_0002
Step 1: Preparation of 4-({rtgrt-butvirdimethyl)silylloxy}methvDpyridin-2-amine
Figure imgf000056_0001
A solution of (2-aminopyridin-4-yl)methanol (5.0 g, 40 mmol), tert- butyldimethylsilyl chloride (6.07 g, 40 mmol), N-ethyl-N-isopropylpropan-2-amine (7.0 mL, 40 mmol) and ΛζN-dimethylpyridin-4-amine (0.49 g, 4 mmol) in dichloromethane (50 mL) was stirred 2 days at ambient temperature under nitrogen. The resulting reaction mixture was washed in sequence with aqueous sodium hydroxide (1 N), water and brine. It was then dried (Na2SO4) and concentrated in vacuo. The residue was chromatographed on silica gel using 50 % ethyl acetate in hexane to yield pure title compound (5.47 g).
1H NMR (300 MHz, CD3CN) δ 7.75 (m, IH), 6.39 -6.48 (m, 2H), 4.70 (bs, IH), 4. 50 (s, 2H), 0.83 (s, 9H) and 0.03 ppm (s, 6H); ES-MS m/z 239.3 [M+H]+, HPLC RT (min) 2.35.
Step 2: Preparation of N-({r4-((rtg/t-butyl(dimethyl)silylloxy}methyl)pyridin-2- yliamino }carbonothioyl)benzamide
Figure imgf000056_0002
A solution of 4-({[tgrt-butyl(dimethyl)silyl]oxy}methyl)pyridin-2-amine (2.00 g, 8.39 mmol) and benzoyl isothiocyanate (1.51 g, 9.23 mmol) in toluene (20 mL) was heated to 85 0C for 12 h. The solvent was removed by evaporation in vacuo and the residue was purified by chromatography on silica gel using a gradient from 0-10% ethyl acetate in hexane to yield pure title compound as a yellow oil which solidified on standing (2.68 g, 79%). 1H NMR (300 MHz, CD3OD) δ 8.79 (bs, IH), 8.18 (d, IH), 7.83 (m, 2H), 7. 50 (m, IH), 7.40 (m, 2H), 7.04 (m, IH), 4.68 (s, 2H), 0.82 (s, 9H), and 0.03 ppm (s, 6H); ES- MS m/z 402.0 [M+H]+, HPLC RT (min) 4.24.
Step 3: Preparation of N-F4-('(rtert-butylfdimethyl')silyl1oxylmethyl)pyridin-2-yllthiourea
Figure imgf000057_0001
A solution of N-({ [4-({ [tert-butyl(dimethyl)silyl]oxy }methyl)pyridin-2- yl]amino}carbonothioyl)benzamide (1.00 g, 2.49 mmol) in absolute ethanol (15 mL) was stirred with potassium carbonate (0.344 g, 2.49 mmol) and heated to reflux under nitrogen for 16 h ,after which the reaction mixture was filtered and the filtrate was evaporated under vacuum to give crude title compound (670 mg, > 100%) as a white solid which was carried on to the next step without purification.
1H NMR (300 MHz, DMSO-J6) 6 10.55 (bs, 2H), 8.75 (bs, IH), 8.05 (d, IH), 7.10 (s, IH), 6.83 (d, IH), 4. 60 (s, 2H), 0.83 (s, 9H) and 0.03 ppm (s, 6H); ES-MS m/z 298.2 [M+H]+, HPLC RT (min) 3.25.
Step 4: Preparation of {2-r(4-methyl-l,3-thiazol-2-yl)amino1pyridin-4-yl}methanol
Figure imgf000057_0002
A solution of iV-[4-({ [te?t-butyl(dimethyl)silyl]oxy}methyl)pyridin-2-yl]thiourea (crude material, 650 mg) and 1-chloroacetone (0.18 mL, 2.18 mmol) in ethanol (10 mL) was refluxed under nitrogen for 16 h and cooled. A white/pink solid was collected by filtration and washed with ethanol. The filtrate was evaporated in vacuo to yield a second white/pink solid. Comparison of the NMR of the two solids indicated that they were both the title compound and were pure enough (about 90%) to carry on to the next step without further purification (combined residue yield 516 mg, >100 %).
1H NMR (300 MHz, DMSO-J6) δ 8.13 (d, IH), 7.05 (s, IH), 6.83 (d, IH), 6. 58 (s, IH), 4.42 (s, 2H) and 2.18 ppm (s, 3H); ES-MS m/z 222.2 [M+H]+, HPLC RT (min) 1.41.
Step 5: Preparation of 4-(chloromethyl)-N-(4-methyl-l,3-thiazol-2-yl)pyridin-2-amine
Figure imgf000058_0001
A mixture of {2-[(4-methyl-l,3-thiazol-2-yl)amino]pyridin-4-yl}methanol (200 mg, 0.9 mmol) and thionyl chloride (0.66 mL, 9.04 mmol) was stirred for 3 h and then evaporated in vacuo. The residue was dissolved in ethyl acetate and washed with saturated sodium bicarbonate. The aqueous layer was back extracted twice with ethyl acetate and then twice with a mixture of isopropanol, ethyl acetate and dichloromethane (1:8:1). The combined extracts were dried (Na2SO4) and concentrated in vacuo. The resulting residue was mixed with methanol, evaporated and then mixed with ethyl acetate and then evaporated again to yield the title compound as a light pink solid (200 mg, 92%) which was taken on to the next step as a crude solid.
1H NMR (300 MHz, CD2Cl2) δ 8.30 (m, IH), 6.98 (s, IH), 6.90 (m, IH), 6. 50 (s, IH), 4.55 (s, 2H) and 2.33 ppm (s, 3H); ES-MS m/z 240.2 [M+H]+, HPLC RT (min) 1.14.
Intermediate R Preparation of N-({r4-(chloromethyI)pyridin-2-ynamino}carbonyl)benzamide
Figure imgf000058_0002
By using the methods described for preparation of Intermediate L and by substituting benzoyl isocyanate instead of ethyl isocyanate and using dichloromethane rather than DMF as solvent, Intermediate R was prepared. The product, which separated from the reaction mixure as a solid, was collected by filtration and washed with dichloromethane.
1H NMR (DMSOd6) δ 11.01 (s, IH), 10.98 (bs, IH), 8.06 (d, IH), 7.82 (s, IH), 7.73 (d, 2H), 7.37 (t, IH), 7.25 (t, 2H), 6.90 (d, IH), and 4.52 (s, 2H).
Preparation of Compounds of the Invention
Example 1
Preparation of 4-{r(4-{r(2,2-difluoro-l,3-benzodioxoI-5-yI)aniino1carbonyl}- l-methyI-lH-pyrazol-5-yl)aniino1methyl}-N-methvIpyridine-
2-carboxamide
Figure imgf000059_0001
A solution 5-amino-N-(2,2-difluoro- 1 ,3-benzodioxol-5-yl)- 1-methyl- IH- pyrazole-4-carboxamide (Intermediate A, 280 mg, 0.95 mmol), sodium iodide (283 mg, 1.89 mmol), and 2,6-di(te/t)butyl-4-methylphenol (10 mg) in dry dimethylformamide (1.5 mL) was stirred under nitrogen as {2-[(methylamino)carbonyl]pyridin-4-yl}methyl methanesulfonate (Intermediate D, 461 mg, 1.89 mmol) was added. The resulting solution was stirred at 60 0C in a foil wrapped flask for 2Oh. The resulting solution was evaporated and then diluted with 1 mL methanol and injected in two portions on a 150/20 mm Cl 8 HPLC column using a gradient from 10-50 % acetonitrile in water (plus 0.05% tiϊfmoroacetic acid). The best fractions containing the desired material, as identified by LCMS, were combined, mixed with saturated NaHCO3, and extracted three times with dichloromethane. The combined extracts were dried (Na2SO4) and evaporated in vacuo to yield 90 mg (21%) of pure title compound.
1H NMR (300 MHz, CD3OD-J4) δ 8.50 (d, IH), 8.10 (s, IH), 7.85 (s, IH), 7.70 (m, IH), 7.55 (d, IH), 7.25 (m, IH), 7.10 (m, IH), 4.65 (s, 2H), 3.70 (s, 3H), 2.95 (s,3H); ES-MS m/z 445.3 [M+H]+, LCMS RT (min) 2.87. Example 2
Preparation of 5-{r(2-chIoropyridin-4-yl)methynaminol-N-(2,2-dif[uoro- l,3-benzodioxoI-5-yI)-l-methvl-lH-pvrazole-4-carboxamide
Figure imgf000060_0001
Step 1: Preparation of ethyl 5-(formylamino)-l -methyl- lH-pyrazole-4-carboxylate
Figure imgf000060_0002
A solution of formic acid (4ml, 106.02mmol) and acetic anhydride (6ml, 63.59mmol) was added to a solution of ethyl 5-amino-l -methyl- lH-pyrazole-4- carboxylate (5.0Og, 29.55mmol) in dry THF (3OmL) and allowed to stir at room temperature for 16 h. The reaction mixture was then diluted with EtOAc and washed with concentrated NaHCO3 solution, followed by water, and then brine. The organic layer was dried over Na2SO4 and concentrated in vacuo yielding 4.16g (71.4%) crude product. This material was used without further purification.
1H NMR (300 MHz, CD3CN) δ 8.32 (bs, IH), 7.78 (s, IH), 4.23 (q, 2H), 3.68 (s, 3H), 1.25 (t, 3H); ES-MS m/z 198.0 [M+H]+, HPLC RT (min) 1.41.
Step 2: Preparation of ethyl 5~rr(2-chloropyridin-4-yl)methyll(formyl)aminol-l- methyl- 1 H-pyrazole-4-carboxylate
Figure imgf000060_0003
A solution of crude ethyl 5-(formylamino)-l -methyl- 1 H-pyrazole-4-carboxylate (3.0Og, 15.21mmol) from step 1 in dichloromethane (30ml) was treated with diazabicyclo(5.4.0)undec-7-ene (3.41ml, 22.82mmol) and allowed to stir at room temperature for 30 minutes. 2-Chloro-4-chloromethylpyridine (4.93g, 30.43mmol) was added, and the reaction mixture was allowed to stir at room temperature for 16 h. The reaction mixture was then diluted with DCM and washed with concentrated NaHCO3 solution, followed by water, then brine. The organic layer was dried over Na2SO4 and concentrated in vacuo. The crude residue was washed with ether, and the ether washings were concentrated and triturated with hexanes. The residue was again concentrated yielding 3.93g (80.1%) product as a thin oil.
1R NMR (300 MHz, CD3CN) δ 8.30 (d, IH), 8.23 (s, IH), 7.85 (s, IH), 7.33 (s, IH), 7.22 (d, IH), 4.80 (bs, 2H), 4.17 (q, 2H), 3.67 (s, 3H), 1.23 (t, 3H); ES-MS m/z 323.1 [M+H]+, HPLC RT (min) 2.31.
Step 3: Preparation of 5-{r(2-chloiOpyridin-4-yl)methyl1amino)-l-methyl-lH-pyrazole-
4-carboxylic acid
Figure imgf000061_0001
A solution of ethyl 5-[[(2-chloropyridin-4-yl)methyl](formyl)amino]-l- methyl-lH-pyrazole-4-carboxylate (3.93g, 12.18mmol) in THF (25mL), MeOH (5mL), and water (5mL) was treated with LiOH (2.9 Ig, 1221.77mmol) and allowed to stir at 800C overnight. The reaction mixture was then cooled to room temperature, and the pH was adjusted to three using IN HCl. The reaction mixture was then diluted with EtOAc and washed with water, brine, and then dried over Na2SO4. Concentration of the ethyl acetate solution in vacuo gave crude product, which was triturated with ether yielding 2.Og (61.6%) product.
1H NMR (SOO MHZ5 CD3CN) 6 8.32 (d, IH), 7.56 (s, IH), 7.38 (s, IH), 7.28 (d, IH), 6.18 (bm, IH), 4.59 (d, 2H), 3.64 (s, 3H); ES-MS m/z 267.1 [M+H]+, HPLC RT (min) 1.66.
Step 4: 5-( Ff 2-chloropyridin-4-yl)methyH amino } -N-(2.2-difluoro- 1 ,3-benzodioxol-5-yl)-
1 -methyl- 1 H-pyrazole-4-carboxamide
Figure imgf000062_0001
A solution of 5-{ [(2-chloropyridin-4-yl)methyl]amino}-l-methyl-lH-pyrazole- 4-carboxylic acid (50.0mg, 0.19mmol) in dry DMF (3mL) was treated with N5N- diisopropylethylamine (0.1OmL, 0.56mmol), followed by PyBOP (97.57mg, 0.19mmol) and allowed to stir for 30 minutes. 5-Amino-2, 2-difluorobenzo-l,3-dioxole (64.91mg, 0.37mmol) was added, and the reaction mixture was allowed to stir for 16 h at 6O0C. The reaction mixture was then diluted with EtOAc, and the organics were washed with concentrated NaHCO3 solution, followed by water, and then brine. The organics were then dried over Na2SO4 and concentrated in vacuo. The crude residue was triturated with hot hexanes yielding 55.0mg (68.6%) product.
1H NMR(300 MHz, CD3CN) δ 8.43 (bs, IH), 8.29 (d, IH), 7.75 (d, 2H), 7.40 (s, IH), 7.23-7.32 (m, 2H), 7.17 (d, IH), 6.76 (bs, IH), 4.53 (d, 2H), 3.68 (s, 3H); ES-MS m/z 422.0 [M+H]+, HPLC RT (min) 3.12.
Example 3
Preparation of N-(2<2-difluoro-l,3-benzodioxol-5-yl)-5-r({2-r(2-hvdroxyethvI) amϊno1pyridin-4-yl}methyI)amϊno1-l-methyl-lH-pyrazoIe-4- carboxamϊde
Figure imgf000062_0002
A solution of 5-{ [(2-chloiOpyridin-4-yl)methyl]amino}-N-(2,2-difluoro-l,3- benzodioxol-5-yl)-l-methyl-lH-pyrazole-4-carboxamide (lOO.Omg, 0.24mmol) in pyridine (ImL) was treated with ethanolamine (0.5OmL, 8.28mmol) and heated to 200°C in a sealed tube for 6 h. The reaction mixture was then allowed to cool to room temperature overnight. It was then diluted with water and extracted with EtOAc. The organic extracts were washed with water, dried over Na2SO4, and concentrated in vacuo. Purification of the crude residue by HPLC (10-90% MeCN in water gradient containing 0.1% TFA) gave 23.0 mg (17.3%) of the title compound as the TFA salt.
1H NMR (300 MHz, CD3CN) 6 8.40 (bs, IH), 7.67-7.78 (m, 3H), 7.27 (d, IH), 7.18 (d, IH), 7.02 (s, IH), 6.78 (d, IH), 4.57 (s, 2H), 3.6-3.71 (m, 4H), 3.38 (m, 2H); ES-MS m/z 447.2 [M+H]+, HPLC RT (min) 2.23.
Examples 3-a to 3-e in Table A were made using the procedure of Example 2 (step 4) and Example 3 by substituting the appropriate starting materials:
Table A
Figure imgf000064_0001
Example 4
Preparation of methyl 4-{r(4-{r(2<2-difluoro-l,3-benzodioxoI-5-vI)amino]carbonvII-l- methvI-lH-pyrazol-5-vDamϊno1methvI>pvridine-2-carboxvIate
Figure imgf000065_0001
Step 1 : Preparation of 4- { f(4- { F(2,2-difluoro- 1 ,3-benzodioxol-5-yl)aminolcarbonyl I - 1 -methyl- 1 H-pyrazol-5-yl)aminolmethyl } pyridine-2-carboxamide
Figure imgf000065_0002
The title compound can be prepared by the following method. A solution of 5-amino-N- (2,2-difluoro- 1 ,3-benzodioxol-5-yl)- 1 -methyl- lH-pyrazole-4-carboxamide (Intermediate A), sodium iodide, and 2,6-di(te/t)butyl~4-methylphenol in dry dimethylformamide can be stirred under nitrogen as [2-(aminocarbonyl)pyridin-4-yl]methyl methanesulfonate (Intermediate C) added. The resulting solution isstirred at 60 °C in a foil wrapped flask for 2Oh. The resulting solution evaporated and then diluted with methanol and injected on a 150/20 mm C18 HPLC column using a gradient from 10-50 % acetonitrile in water (plus 0.05% trifluoroacetic acid). The best fractions containing the desired material, as identified by LCMS, are combined, mixed with saturated NaHCO3, and extracted three times with dichloromethane. The combined extracts are dried (Na2SO4) and evaporated in vacuo to yield pure title compound. Step 2 Preparation of methyl 4-{rf4-{r(2.2-difluoro-L3-benzodioxol-5- yl)amino1carbonyljmethyl-lH-pyrazol-5-yl)aminolrnethyl}pyridme-2-carboxylate
Figure imgf000066_0001
A suspension of 4-{ [(4-{ [(2,2-difluoro-l,3-benzodioxol-5-yl)amino]carbonyl}- l-methyl-lH-pyrazol-5-yl)amino]methyl}pyridine-2-carboxamide in 1.2 mL methanol and N,N '-dimethylformamide dimethylacetal is heated with stirring in a sealed vial at 50 °C. After 2 h heating the solution is evaporated in vacuo and the residue is chromatographed on silica gel with a gradient from 0-1% methanol in dichloromethane to yield pure title compound.
Example 5
Preparation of 4-{r(4-{r(2,2-difluoro-l,3-benzodioxoI-5-yI)amino1carbonyI}-l-methyI- lH-pyrazoI-5-yI)amino1methyI}-N-(4-pyrroIidin-l-ylbutyl)pyridine-2-carboxamide
Figure imgf000066_0002
A slurry of 4-{ [(4-{ [(2,2-difluoro-l,3-benzodioxol-5-yl)amino]carbonyl}- l-methyl-lH-pyrazol-5-yl)amino]methyl}pyridine-2-carboxamide (Product from example 4 step 1) (95 mg, 0.21 mmol) in methanol (0.60 mL) is treated as in Example 4, step 2 to prepare a solution of methyl 4- { [(4- { [(2,2-difluoro- 1 ,3-benzodioxol-5- yl)amino]carbonyl}-l-methyl-lH-pyrazol-5-yl)amino]methyl}pyridine-2-carboxylate, which isused directly and addition of 4-pyrrolidin-l-ylbutan-l -amine (266 mg, 1.87 mmol) and stirring at 65 0C for 16 h . The reaction solution is purified by HPLC using direct injection, in three portions, on a YMC-Pack Pro C18 column (150 x 20 mm) and is eluted at 20 mL/min with a gradient from 10-50 % acetonitrile in water plus 0.05% TFA. Pure fractions from each injection are combined, made basic by addition of sodium bicarbonate and extracted with ethyl acetate. Combined extracts are dried (Na2SO4) and evaporated in vacuo to yield pure title compound
Example 6
Preparation of N-(2,2-difluoro-l,3-benzodioxoI-5-yl)-5-r({2r(methoxyacetyI)amino1 pyridin-4-yl}methyl)aminol-l-methyl-lH-pyrazole-4-carboxamϊde
Figure imgf000067_0001
This material is prepared using the same method described for Example 1 but starting with 4-amino-7V-(2,2-difluoro- 1 ,3-benzodioxol-5-yl)~ 1 ,3-thiazole-5-carboxamide (Intermediate A) and proportionate amounts of Intermediate G rather than Intermediate D and also proportional amounts of the other reaction components. The reaction mixture is heated to 60 °C in a foil wrapped flask overnight before crude product isolated. This material is purified by chromatography on silica gel using a gradient from 0-60 % ethyl acetate in hexane to yield final product.
Example 7 Preparation of l-methvI-5-r({2-r(methyIsulfonyl)amino1pyridin-4-yI}methyl)aminol-
N-(2,2,3,3-tetrafluoro-2,3-dihydro-l,4-benzodioxin-6-yl)-lH-pyrazoIe-4-carboxamide
Figure imgf000067_0002
The title compound is prepared using the same method described for Example 1 but starting with 5-amino-l-methyl-N-(2,2,3,3-tetrafluoro-2,3-dihydiO-l,4-benzodioxin-6- yl)-lH-pyrazole-4-carboxamide (Intermediate B) rather than 5-amino-N-(2,2-difluoro-l,3- benzodioxol-5-yl)-l -methyl- lH-pyrazole-4-carboxamide (Intermediate A) and proportionate amounts of N-[4-(chloromethyl)pyridin-2-yl]methanesulfonamide (Intermediate K) rather than Intermediate D and also proportional amounts of the other reaction components. The reaction mixture is heated to 60 0C in a foil wrapped flask for 16 h and then cooled. The reaction mixture is diluted with ethyl acetate, washed with water, dried (Na2SO4) and evaporated in vacuo. The crude product is purified by preparative HPLC to yield pure title compound.
Example 8
Preparation of 5-{r(2-{r(ethyIamino)carbonyIlamino}pyridin-4-vI)methvnamino}- l-methvI-N-(2,2,3.3-tetrafluoro-2,3-dihvdro-l,4-benzodioxin-
6-yl)-lH-pyrazole-4-carboxamide
Figure imgf000068_0001
Step 1 Preparation of 5-({ r2-(2,5-dimethyl-lH-pyrrol-l-yl)pyridin-4-yl1methyl} amino)- 1- methyl-N-(2,2,3,3-tetrafluoro-2,3-dihydro-l,4-benzodioxin-6-yl)-lH-pyrazole-4- carboxamide
Figure imgf000068_0002
The title compound was prepared using the same method described for Example 2 using steps 1, 2, 3, and 4. In Step 2, intermediate PP was used in place of intermediate P. 1H NMR (300 MHz, CD3OD-d4) δ 8.45 (d, IH), 7.85 (s, IH), 7.62 (m, 2H), 7.45 (m, IH), 7.35 (m, IH), 7.20 (m, 2H), 5.75 (s, IH), 4.70 (s, 2H), 3.75 (s, 3H), 1.93 (s, 6H); ES-MS m/z 485.1 [M+Hf, LCMS RT (min) 3.45. Step 2 Preparation of 5-{rf2-aminopyridin-4-vDmethyllaminoi-l-methyl-N-(2,2,3,3- tetrafluoro-2,3-dihydro- 1 ,4-benzodioxin-6-yl)- lH-pyrazole-
4-carboxamide
Figure imgf000069_0001
To a solution of 5-({[2-(2,5-dimethyl-lH-pyrrol-l-yl)pyridin-4-yl]methyl}amino)- l-methyl-N-(2,2,3,3-tetrafluoro-2,3-dihydro-l,4~benzodioxin-6-yl)-lH-pyrazole-4- carboxamide (340 mg, 0.64 mmol) in ethanol (3 mL) was added water (1 mL), hydroxylamine hydrochloride (445 mg, 6.41mmol), followed by triethyl amine (0.18 mL). The mixture was heated to reflux for 5 h and then cooled. The mixture was diluted with EtOAc and extracted with saturated NaHCO3. The organic layer was dried, evaporated and purified over silica gel using a 0-10% MeOH in DCM as a gradient to yield 222 mg (77%) of white solid.
1H NMR (300 MHz, CD3OD-d4) 67.85 (s, IH), 7.79 (m, 2H), 7.42 (m, IH), 7.20 (d, IH), 6.58 (m,2H), 4.44 (s,2H), 3.74 (s,3H); ES-MS m/z 453.0 [M+H]+, LCMS RT (min) 2.50.
Step 3 Preparation of 5-{ r(2-{r(ethylamino)carbonyllamino}pyridin-4-yl)methyllamino)-
1 -methyl-N-f 2.2.3 ,3 -tetrafluoro-2,3-dihvdro- 1 ,4-benzodioxin-
6-vD- 1 H-pyrazole-4-carboxamide
Figure imgf000069_0002
To a solution of 5-{[(2-aminopyridin-4-yl)methyl]amino}-l-methyl-N-(2,2,3,3- tetrafluoro-2,3-dihydro-l,4-benzodioxin-6-yl)-lH-pyrazole-4-carboxamide (100 mg, 0.22 mmol) in dichloroethane (1 mL) was added ethyl isocyanate (0.3 mL, 3.33 mmol). The mixture was stirred at room temperature for 72 h. The solid precipitated out as the reaction proceeded. Ether (2mL) was added to the reaction mixture and the solid was filtered. The solid was washed with methanol (1 mL to remove any remaining starting material) and again with ether (2 mL) before evaporation to give the desired product (62 mg, 53%).
1H NMR (300 MHz, DMSO-J6) δ 9.80 (s, IH), 9.12 (s, IH), 8.05 (m, 2H), 7.88 (m, IH), 7.82 (s, IH), 7.45 (m, IH), 7.40 (m, IH), 7.30 (bs, IH), 6.98 (t, IH), 6.80 (d, IH), 4.54 (d, 2H), 6.05 (s, 3H), 3.15 (m, 2H), 1.02 (t, 3H); ES-MS m/z 524.1 [M+H]+, LCMS RT (min) 2.71.
The compounds of examples 13, 14, and 15 as shown in Table B were made according to this method substituting the appropriate starting materials.
Example 9
Preparation of l-methyl-5-{ \(2-\ r(methylamino)carbonyllamino)pyridin-4- vDmethyll amino I -N-f 2,2 A4-tetrafluoro-4H- 1 ,3-benzodioxin-
6-vD- lH-pvrazole-4-carboxamide
Figure imgf000070_0001
The title compound was prepared using the same method described for Example 2 using steps 1, 2, 3, and 4. In Step 2, intermediate N was used in place of 2-Chloro-4- chloromethylpyridine. In Step 4, the appropriate aniline was also substituted to give the title compound.
1H NMR (300 MHz, CD3OD-J4) δ 8.25 (s, IH), 8.15 (m, IH), 7.90-7.97 (m, 2H), 7.32-7.37 (m, IH), 7.12 (s, IH), 6.94-6.97 (m, IH), 4.60 (s, 2H under the water peak), 3.75 (s, 3H), 2.88 (s, 3H), ES-MS m/z 510.0 [M+H]+, LCMS RT (min) 2.67.
Examples 17, 18, and 19 as shown in Table B were made according to this method substituting the appropriate starting materials. Example 10
Preparation of 5-r((2-r(aminocarbonyl)amino1pyridin-4-yllmethyl)amino1- N-f 2,2-difluoro- 1 ,3-benzodioxol-5-yl)- 1 -methyl- 1 H-pyrazole-
4-carboxamide
Figure imgf000071_0001
The title compound was prepared using the same method described for Example 2 using steps 1, 2, 3, and 4. In Step 2, intermediate R was used in place of 2-Chloro-4- chloromethylpyridine. In Step 3, the hydrolysis also removes the benzoyl group along with the formyl group and hydrolysis of the ester.
1H NMR (300 MHz, CD3OD-d4) δ 8.18 (d, IH), 7.85 (s, IH), 7.70 (m, IH), 7.38 (m, IH), 7.32 (m, IH), 7.20 (m, IH), 7.13 (m, IH), 4.75 (s, 2H), 3.74 (s, 3H); ES-MS m/z 446.0 [M+H]+, LCMS RT (min) 2.34.
Example 16 as shown in Table B was made according to this method substituting the appropriate starting materials.
Example 11
Preparation of N-(2,2-difluoro-l,3-benzodioxol-5-yl)-5-{ \(2-{ lϊethylamino) carbonyli amino } pyridin-4- vDmethyll amino } - 1 -methyl- 1 H-pyrazole- 4-carboxamide trifluoroacetate
Figure imgf000071_0002
The title compound was prepared using the same method described for Example 1. The intermediate L was used in place of intermediate D and the concentrated HPLC fractions were analyzed without free-basing with NaHCO3.
1H NMR (300 MHz, CD3CN) δ 8.45 (s, IH), 8.30 (s, IH), 8.02 (d, IH), 7.77 (s, IH), 7.73 (s, IH), 7.34 (s, IH), 7.26 (d, IH), 7.20 (d, IH), 7.15 (d, IH), 4.65 (s, 2H), 3.68 (s, 3H), 3.28 (q, 2H), 1.15 (t, 3H); ES-MS m/z 474.3 [MH-H]+, LCMS RT (min) 2.53.
Example 12
Preparation of N-(2,2-difluoro-l,3-benzodioxol-5-yl)-l-methyl-5-{ \(2-
I F(methylamino)carbonvπamino |pyridin-4-yl)methyllamino }-
1 H-pyrazole-4-carboxamide trifluoroacetate
Figure imgf000072_0001
Step 1: Preparation of ethyl 5-rr(2-aminopyridin-4-yl)methyl1(formyl)amino1-l-methyl- lH-pyrazole-4-carboxylate
Figure imgf000072_0002
A solution of crude ethyl 5-(formylamino)-l -methyl- lH-pyrazole-4-carboxylate (500.00mg, 2.54mmol) from Example 2, step 1, in THF (7.5ml) was treated with diazabicyclo(5.4.0)undec-7-ene (0.57ml, 3.80mmol) and allowed to stir at room temperature for 30 minutes. 2-Amino-4-chloromethylpyridine (723.09mg, 5.07mmol) was added, and the reaction mixture was heated to 6O0C for one hour, then allowed to stir at room temperature for 16 h. The reaction mixture was then diluted with ethyl acetate and washed with saturated NaHCO3 solution, followed by water, then brine. The organic layer was dried over Na2SO4 and concentrated in vacuo, yielding 440mg (57.1%, impure) product as a 2: 1 mixture of product to deformylated product. ES-MS m/z 304.1 [M+H]+, HPLC RT (min) 1.08.
Step 2: Preparation of ethyl 5-r({2-r(fert-butoxycarbonvDaminolpyridin-4- yl}methyl)(formyl)amino1-l-methyl-lH-pyrazole-4-carboxylate
Figure imgf000073_0001
A solution of crude ethyl 5-[[(2-aminopyridin-4-yl)methyl](formyl)amino]-l- methyl- lH-pyrazole-4-carboxylate (500.00mg, 1.65mmol) in THF (5ml) was treated with N5N- diisopropylethylamine (0.32ml, 1.81mmol) and 4-dimethylaminopyridine (20.14mg, O.lβmmol). Di-tert-butyl dicarbonate (1.81ml of IM solution in THF) was added to the reaction mixture and allowed to stir at room temperature for 16 h. The reaction mixture was then diluted with ethyl acetate and washed with concentrated NH4Cl solution, followed by water, then brine. The organic layer was dried over Na2SO4 and concentrated in vacuo. The crude residue was triturated with ether, and the solvent was again evaporated yielding 600mg (90.1%, impure) product. The product was used without purification.
Step 3: Preparation of 5-r({2-r(fert-butoxycarbonyl)amino1pyridin-4-yl}methyl)amino1-
1 -methyl- 1 H-pyrazole-4-carboxylic acid
Figure imgf000073_0002
The title compound (130.0mg, 25.1%) was prepared in a similar fashion to Example 2, step 3, using ethyl 5-[({2-[(tert-butoxycarbonyl)amino]pyridin-4- yl}methyl)(formyl)amino]-l-methyl-lH-pyrazole-4-carboxylate (600.0mg, 1.49mmol) as starting material.
1H NMR (300 MHz, CD3CN) δ 8.28 (bs, IH), 8.18 (d, IH), 7.89 (s, IH), 7.55 (d, IH), 6.98 (m, IH), 4.59 (m, 2H), 3.64 (s, 3H), 1.50 (s, 9H).
Step 4: Preparation of tert-butyl (4-(r(4-{r(2,2-difruoro-l,3-benzodioxol-
5-yl)aminolcarbonyl I - 1-methyl- lH-pyrazol-5-yl)aminol methyl I pyridin-2-yl)carbamate
Figure imgf000074_0001
The title compound (90.0mg, 48.4%) was prepared in a similar fashion to Example 2, step 4, using 5-[({2-[(tert-butoxycarbonyl)amino]pyridin-4-yl}methyl)amino]-l-methyl- lH-pyrazole-4-carboxylic acid (130.0mg, 0.37mmol) as starting material. This material was used without purification. ES-MS m/z 502.8 [M+H]+, HPLC RT (min) 2.83.
Step 5: Preparation of 5-{r(2-aminopyridin-4-yl)methyllamino}-N-f2,2-difluoro-l,3- benzodioxol-5-yl)-l-methyl-lH-pyrazole-4-carboxamide
Figure imgf000074_0002
A solution of tert-butyl (4-{[(4-{[(2,2-difluoro-l,3-benzodioxol-5- yl)amino]carbonyl}-l-methyl-lH-pyrazol-5-yl)amino]methyl}pyridin-2-yl)carbamate (66.15mg, 0.13mmol) in dichloromethane (1.5ml) was treated with trifluoroacetic acid (1.4ml, 18.12mmol) and allowed to stir at room temperature for 16 hours. The reaction mixture was then diluted with ethyl acetate and washed with IN NaOH solution. The aqueous layer was extracted with EtOAc, and the organic extracts were combined and dried over MgSO4. Concentration of the EtOAc solution in vacuo gave 60.0mg (>99%) crude product estimated 50% pure by 1H NMR. The product was used without purification. ES-MS m/z 403.1 [M+H]+, HPLC RT (min) 0.99.
Step 6: Preparation of N-(2,2-difluoro-13-benzodioxol-5-yl)-l-methyl-5-( r(2-
{ r(methylamino)carbonyll amino } pyridin-4-yl)methyll amino I -
1 H-pyrazole-4-carboxamide trifluoroacetate
Figure imgf000075_0001
The title compound was prepared using the same method described for Example 8 step 3. In Step 3, methyl isocyanate was used in place of ethyl isocyanate. The workup for the final step was different in that the concentrated HPLC fractions were analyzed without free-basing with NaHCO3.
1H NMR (300 MHz, CD3CN) 6 12.10 (s, IH), 8.40 (s, IH), 8.06 (bd, 2H), 7.72 (d, 2H), 7.32 (s, IH), 7.20-7.27 (m, 2H), 7.15 (d, IH), 4.63 (s, 2H), 3.63 (s, 3H), 2.80 (d, 3H); ES-MS m/z 460.3 [M+H]+, LCMS RT (min) 2.37.
Table B
Figure imgf000076_0001
Figure imgf000077_0002
Example 20-a
Preparation of N-(2,2-difluoro-l.,3-benzodioxol-5-yI)-l-methyl-5- {r(2{methyir(methyIamino)carbonyl1amino|pyridin-4-yl)methynamino}-lH-pyrazoIe
4-carboxamide
Figure imgf000077_0001
Step 1: Preparation of r2-(methylamino)pyridin-4-yllmethanol
OH CH3 NH
YY
A solution of (2-chloropyridine-4-yl)methanol (from the preparation of Intermediate P, Step 1) and methylamine hydrochloride in pyridine is heated at 200 °C in a sealed tube for about 16 h. The solvent is removed by evaporation in vacuo and the crude product residue is purified by chromatography on silica gel using a gradient from dichloromethane to about 10 % methanol in dichloromethane. Step 2 Preparation of 4-(chloromethvlVN-methvlpvridin-2-amine
Figure imgf000078_0001
By using the general method of preparation of Intermediate E, Step 1 but substituting [2-(methylamino)pyridin-4-yl]methanol for (2-aminopyridin-4-yl)methanol, the Title compound is prepared.
Step 3. Preparation of N-F4-(chloromethyl)pyridin-2-yl1-N,N'-dimethylurea
Figure imgf000078_0002
The step is carried out using the method described for the preparation of Intermediate L but using the product of Step 2 above rather than 4-(chloromethyl)pyridin- 2-amine and methyl isocyanate rather than ethyl isocyanate.
Step 4: Preparation of N-(2,2-difluoro-l,3-benzodioxol-5-yl)-l-methyl-5-(r(2- { methyl r(methylamino)carbonyll amino |pyridin-4-yl)methyl1amino } - lH-pyrazole-4- carboxamide
Figure imgf000078_0003
The title compound can be prepared using the same method described for Example 1 but starting with 5-amino-N-(2,2-difluoro- 1 ,3-benzodioxol-5-yl)- 1-methyl- IH- pyrazole-4-carboxamide (Intermediate A) and a proportionate amount of N- [4- (chloromethyl)pyridin-2-yl]-N,N'-dimethylurea (from Step 3) rather than Intermediate D. The reaction mixture is heated at 60 °C in a foil wrapped flask under nitrogen for between 2 and 24 h until an LCMS analysis of the reaction mixture shows substantial conversion to products. The resulting final crude mixture is diluted with saturated aqueous sodium bicarbonate and extracted 3 times with ethyl acetate. The combined extracts are dried (Na2SO4) and evaporated in vacuo to yield a residue that is purified by preparative C18 HPLC using water to acetonitrile gradient (usually 10-50%) with added 0.05-0.1 % TFA. The free base is prepared from the TFA salt by addition of saturated aqueous NaHCO3 to the fractions containing the product and extraction with dichloromethane followed by drying of the extract (Na2SO4) and concentration in vacuo to yield pure title compound.
Examples 20-b to 20-x
Figure imgf000079_0001
Step 1: Preparation of various Λr-r4-(chloromethyl)pyridin-2-vnureas with other ISF- substituents Cl H R1'3
By using the methods described for the preparation of Intermediate L but substituting the appropriate alkyl or aryl isocyanate rather than ethyl isocyanate, (using either DMF or dichloromethane) the intermediates which lead to examples 20-b to 20-g, 20-i, and 20-o to 20-w of Table C are prepared. By using the methods described for the preparation of Intermediate E, step 2 but using the appropriate carbamoyl chloride rather than acetoxy acetyl chloride, the intermediates which lead to Examples 21-k to 21-n of Table C can be prepared. In all cases the appropriate isocyanate or carbamoyl chloride is either commercially available or the synthesis is straightforward to one skilled in the art and is reported in the general literature.
Step 2: Preparation of the Title Compounds (Table C)
Figure imgf000080_0001
The title compounds can be prepared using the same method described for Example 1 but starting with 5-amino-N-(2,2-difluoro-l,3-benzodioxol-5-yl)-l-methyl-lH- pyrazole-4-carboxamide (Intermediate A) or another material from the list of Intermediates B, B-2, B-3, B-4 or B-5 instead of Intermediate A and proportionate amounts of the appropriate Intermediate from Step 1 above rather than Intermediate D and also proportional amounts of the other reaction components. The reaction mixture is heated at 60 °C in a foil wrapped flask for between 2 and 24 h until an LCMS analysis of the reaction mixture shows substantial conversion to products. The resulting final crude mixture is diluted with saturated aqueous sodium bicarbonate and extracted 3 times with ethyl acetate. The combined extracts are dried (Na2SO4) and evaporated in vacuo to yield a residue that is purified by preparative C18 HPLC using water to acetonitrile gradient (usually 10-50%) with added 0.05-0.1 % TFA. The free base is prepared from the TFA salt by addition of saturated aqueous NaHCO3 to the fractions containing the product and extraction with dichloromethane followed by drying of the extract (Na2SO4) and concentration in vacuo to yield pure title compound. In the case of Title compounds 20-h and 20-j, the initial blocked products 20-g and 20-i respectively are converted to the final Title compounds by treatment with potassium carbonate in methanol or ethanol. Structures and names of the Title Compounds 20-a to 20-x are shown in Table C
Table C
-
Figure imgf000081_0001
-
Figure imgf000082_0001
}
Figure imgf000083_0001
Figure imgf000084_0001
Figure imgf000085_0001
Figure imgf000086_0001
Example 21
Preparation of N-(2,2-difluoro-l,3-benzodioxoI-5-vD-l-methyl-5-αr2- (methylamino)pyrimidin-4-yI1methvI>ainino)-lH-pyrazole-4- carboxamide
Figure imgf000087_0001
Step 1. Preparation of 5-{r(2,6-dichloropyrimidin-4-yl)methyllamino}-N-(2,2-difluoro- 1 ,3 -benzodioxol-5- yl) - 1 -methyl- 1 H-pyrazole-4-carboxamide
Figure imgf000087_0002
A mixture of 2,4-dichloro-6-(chloromethyl)pyrimidine (Intermediate O, 99 mg, 0.5 mmol) and sodium iodide (75 mg, 0.5 mmol) in anhydrous DMF (0.5 mL) is stirred under nitrogen until a solution forms and then 5-amino-N-(2,2-difluoro-l,3-benzodioxol-5-yl)-l- methyl-1 H-pyrazole-4-carboxamide (Intermediate A, 100 mg, 0.33 mmol) is added. The resulting mixture is heated and stirred under nitrogen at 40 °C for 4 h and then at 59 °C overnight. If after TLC analysis some remaining pyrazole starting materials are present, additional 2,4-dichloro-6-(chloromethyl)pyrimidine (30 mg) is added and the reaction mixture is again heated at 59 °C for another 2 h. The resulting final crude mixture is diluted with saturated aqueous sodium bicarbonate and extracted 3 times with ethyl acetate. The combined extracts is dried (Na2SO4) and evaporated in vacuo to yield a residue that is purified by chromatography on silica gel using a gradient from 10-40 % ethyl acetate in hexane.
Step 2. Preparation of 5-({ r6-chloro-2-(methylamino)pyrimidin-4-yllmethyl}amino)- N-(2,2-difluoro-13-benzodioxol-5-yl)-l-methyl-lH-pyrazole-4-carboxamide and 5-({ \2- chloro-6-(methylaπuno)ρyrirrndin-4-yl1methyl } amino)-N-(2,2-difluoro- 1 ,3-benzodioxol-
5-yl)-l-methyl-lH-pyrazole-4-carboxamide
Figure imgf000088_0001
A solution of 5-{ [(2,6-dichloiOpyrimidin-4-yl)memyl]arnino}-N-(2,2-difluoro- l,3-benzodioxol-5-yl)-l-methyl-lH-pyrazole-4-carboxamide (the product of step 1, 0.2 minol) and methylamine (0.4 mmol) in methanol (1.2 niL) can be stirred in a sealed tube for ~5h. The products can be purified by direct injection on a preparative HPLC to yield purified isomers wherein the 6-(methylamino)pyrimidin-4-yl isomer is expected to be the major product. Step 3. Preparation of the Title Compound
A mixture of 5-({[6-chloro-2-(methylamino)pyrimidin-4-yl]methyl}amino)- N-(2,2-difluoro- 1 ,3-benzodioxol-5-yl)- 1 -methyl- 1 H-pyrazole-4-carboxamide (the minor product of step 2, 0.18 mmol) plus palladium (II) hydroxide (0.36 mmol) and ammonium formate (1.76 mmol) in ethyl acetate (15 mL) and methanol (15 mL) can be stirred with heating at reflux for 16 h. The product solution can be filtered using Celite® filter aid and evaporated in vacuo. The residue can be purified by preparative C18 HPLC using a gradient from 5 to 45% acetonitrile in water plus 0.1% TFA. Evaporation of product containing fractions can yield pure compounds as TFA salts. Alternatively, the fractions containing the product can be mixed with saturated aqueous NaHCO3 and extracted with dichloromethane. The extracts are dried (Na2SO4) and evaporated in vacuo to yield pure free base title compound.
Example 22
Preparation of 5-{r(2-aminopyridin-4-yI)methynamino|-l-methyI-N-(2,2,3,3- tetrafluoro-2,3-dihydro-l,4-benzodioxin-6-yI)-lH-pyrazoIe-
4-carboxamide
Figure imgf000089_0001
This compound was prepared as shown in Example 8 and is the product of Step 2.
Example 23
Preparation of fert-butyl (4-{IY4-{r(2,2-difluoro-l.,3-benzodioxol-
5-yl)amino1carbonyI>-l-methyl-lH-pyrazoI-5-yl)amino1 methyI|pyridin-2-yl)carbamate
Figure imgf000089_0002
This compound was prepared as shown in Example 12 and is the product of Step 4.
Example 24
Preparation of 5-{r(2-aminopyridin-4-yl)methynamino|-N-(2,2-difluoro-l,3- benzodioxoI-5-yl)-l-methyI-lH-pyrazole-4-carboxamide
Figure imgf000089_0003
This compound was prepared as shown in Example 12 and is the product of Step 5. B. Evaluation of physiological activity
The utility of the compounds of the present invention can be illustrated, for example, by their activity in the P-AKT/PKB Cytoblot Assay described below. The involvement of the P-AKT/PKB [ PDK/AKt] pathway as a target for cancer chemotherapy has been recognized in the art. For example, see F. Chang et al, Involvement of PI3K/Akt pathway in cell cycle progression, apoptosis, and neoplastic transformation: a target for cancer chemotherapy, Leukemia, 2003, 17: p. 590-603; K. A. West et al, Activation of the PI3K/Akt pathway and chemotherapeutic resistance, Drug Resistance Updates, 2002, 5: p. 234-248; and P. Sen et al, Involvement of the Akt/PKB signaling pathway with disease processes, Molecular and Cellular Biochemistry, 2003, 253: p. 241- 246.
P-AKT/PKB Cvtoblot Assay Protocol with H209 Cells
H209 small cell lung carcinoma cells in log phase were plated at 50,000 cells/well in 96- well poly-lysine coated, clear bottom/ black-sided plates (Becton-Dickinson, USA Cat # 354640) in 100 μl RPMI medium containing 0.1% (w/v) BSA, and incubated overnight at 370C in 5% CO2 incubator. The following day, compounds (10 mM stock solutions in DMSO) were added to the plates to generate final concentrations of 0.0, 0.01, 0.03, 0.1, 0.3, 1.0, 3.0 and 10 μM for IC50 determinations and incubated for 1 hour at 37 0C. Cells were then left untreated or stimulated with Stem Cell Factor (SCF: Biosource Cat # PHC2116) at a final concentration of 25 ng/mL for 5 minutes at 37 0C in 5% CO2 incubator. The media was then removed using a vacuum manifold and the cells were washed once with Tris Buffered Saline (TBS). Cells were then fixed by adding 200 μl of cold 3.7% (w/v) formaldehyde in TBS to each well for 15 minutes at 4 0C. After removal of the formaldehyde, the cells were treated with the addition of 50 μl of methanol (at - 20 0C) to each well for 5 minutes. After removal of the methanol, 200 μl of 1% (w/v) BSA in TBS was added to each well to block non-specific antibody binding sites and the plate was incubated at room temperature for 30 minutes.
After removal of the blocking buffer, 50 μl of p-(S473) AKT rabbit polyclonal antibody (Cell Signaling, USA Cat # 9277S) was added at a dilution of 1:250 in 0.1% (w/v) BSA in TBS, and the plate was incubated at room temperature for 1 hour. Plates were then washed 3 times with cold TBS containing 0.05% (v/v) Tween 20 (TBS-T) and 100 μl of Horseradish peroxidase (HRP)-conjugated goat-anti-rabbit antibody (Amersham, USA Cat # NA934V) at a dilution of 1:250 in TBS-T was added and the plate was incubated at room temperature for Ih. After washing with ice-cold TBS-T four times, 100 μl of Enhanced Chemiluminescence (ECL) reagent (Amersham, USA Cat# RPN2209) was added to each well and mixed on a mini-orbital shaker for 1 min. The plate was then read on a Perkin Elmer Victor 5 Multilabel Counter (#1420-0421).
Compounds of examples 1, 3-a, 3-b, 3-c, 3-d, 8, 9, 10,11, 12, 13, 14, 15, 16, 16, 17, 18, 19. were tested in the above P-AKT/PKB Cytoblot assay, with the result that these examples exhibited IC50 values of less than 500 nM. In one embodiment, the present invention relates to a compound which exhibits an IC50 value of less than 500 nM in this assay.
The utility of the compounds of the present invention can also be illustrated, for example, by their activity in the phosph-ERK Assay described below.
Growth-factor induction of the RAS/MEK/ERK signaling pathway leads to the induction of phosphorylation of a number of proteins including phospho-ERK (See C. J. Marshall, MAP kinase kinase kinase, MAP kinase kinase and MAP kinase, Current Opinions in Genetic Development, 1994, 4: p. 82-89). The importance of this pathway in cancer biology has been recognized in the art. Activation of the RAS signaling pathway is an important mechanism by which cancer develops (R. Herrera, et al, Unraveling the complexities of the Raf/MAP kinase pathway for pharmacological intervention, Trends MoI. Med., 2002, 8: p. S27-31). Mutational activation of RAS or downstream effectors as well as growth factor induction of this pathway leads to increased tumor cell proliferation and survival (A. A. Adjei, Blocking oncogenic RAS signaling for cancer therapy, J. Natl. Cancer Inst., 2001, 93(14): p. 1062-1074; J Schlessinger, Cell signaling by receptor tyrosine kinases, Cell, 2000, 103: p. 211-225).
Phospho-ERK Cvtoblot Assay Protocol with MDA-MB 231 Cells
MDA-MB-231 cells in log phase were plated at 25,000 cells/well in 96-well opaque plates (Falcon, USA Cat # 353296) in 100 μL RPMI medium containing 10% (w/v) FBS, and incubated overnight at 370C in 5% CO2 incubator. The following day, the growth medium was removed from the plate by aspiration and replaced with RPMI medium containing 0.1% BSA and example compounds diluted to generate final concentrations of 0.0, 0.001, 0.003, 0.01, 0.03, 0.1, 0.3, 1 and 3 μM. Cells were incubated with compound for 1 hour at 370C in a 5% CO2 incubator. The media was then removed from the plate by aspiration and the cells were washed once with 180 μL/well cold Tris Buffered Saline (TBS). After removal of the wash buffer, the cells were fixed by adding 180 μL of cold 3.7% (v/v) formaldehyde in TBS to each well for 1 hour at 40C. After removal of the formaldehyde, the cells were treated with the addition of 60 μL of -2O0C methanol to each well for 5 minutes at 40C. The methanol was removed and the cells were washed with 180 μL/well of 5% (w/v) BSA in TBS. To block non-specific antibody binding sites, each well was treated with 180 μL/well 5% BSA (w/v) in TBS for thirty minutes at room temperature. After removal of the blocking buffer, 50 μL of an anti-phospho-p44/42 MAP kinase (Thr202/Tyr204) rabbit polyclonal antibody (Cell Signaling, USA Cat # 9101) was added to each well at a dilution of 1:1000 in 5% (w/v) BSA in TBS, and the plate was incubated at 4 0C overnight. Plates were then washed three times with 300 μL/well TBS at room temperature. The plates were then incubated with 50 μL of Horseradish peroxidase (HRP)-conjugated goat-anti-rabbit antibody (Amersham, USA Cat. # NA934V) at a dilution of 1:1000 in 5% BSA-TBS at room temperature for 1 hr. After washing the plate three times with 300 μL/well TBS, 60 μL of Enhanced Chemiluminescence (ECL) reagent (Amersham, USA Cat# RPN2209) was added to each well and incubated at room temperature for five minutes. The plate was then read on a Perkin Elmer Victor 5 Multilabel Counter (#1420-0421).
The compounds of examples 1, 10, 11, 12, 13, 14, 15, 17, 18, 19, were tested and showed an IC50 value of less than 3 μM in this assay. In one embodiment, the present invention relates to a compound which exhibits an IC50 value of less than 3 μM.
The utility of the compounds of the present invention can also be illustrated, for example, by their activity in the flk-1 (murine VEGFR2) Assay described below. The VEGF- VEGFR2 signaling pathway has been extensively characterized as an important regulator of angiogenesis and tumor angiogeneisis (See G. Yancopoulos et al, Vascular-specific growth factors and blood vessel formation, Nature, 2000, 407: p.. 242- 248; D. Shweiki et al, Induction of vascular endothelial growth factor expression by hypoxia and by glucose deficiency inmulticell spheroids: Implications for tumor angiogenesis, Proc. Natl. Acad. Sci, 1995, 92: p. 768-772). Inhibition of tumor cell growth by blocking this pathway has been well documented in the art. Administration of soluble VEGFR2 receptors inhibits the growth of a wide variety of tumors (See C. Brans et al, Vascular endothelial growth factor is an in vivo survival factor for tumor endothelium in a murine model of colorectal liver metastases, Cancer, 2000, 89: p. 495-499; B. Millauer et al, Glioblastoma growth inhibited in vivo by a dominant-negative FLK-I mutant, Nature, 1994, 367: p. 576-579). Neutralizing antibodies to VEGF or VEGFR2 and VEGF antisense suppress tumor growth in vivo (See K. Kim et al, Inhibition of vascular endothelial growth factor-induced angiogenesis suppresses tumor growth in vivo, Nature, 1993, 362: p. 841-844; M. Prewett et al, Antivascular endothelial growth factor receptor (fetal liver kinase I) monoclonal antibody inhibits tumor angiogenesis and growth of several mouse and human tumors, Cancer Research, 1999, 59: p. 5209-5218; M. Saleh et al, Inhibition of growth of C6 glioma cells in vivo by expression of antisense vascular endothelial growth factor sequence, Cancer Research, 1996, 56: p. 393-401).
FIk-I (murine VEGFR-2) Biochemical Assay
This assay was performed in 96-well opaque plates (Costar, USA Cat #3915) in the TR- FRET format. Reaction conditions were as follows: 10 μM ATP, 25 nM poly (Glu,Tyr)- biotin (CIS BIO International, USA Cat#61 GTOBLD), 2 nM Eu-labelled phospho-Tyr Ab (Perkin Elmer, USA Cat#AD0067), 10 nM Strepavidin-APC (Perkin Elmer, USA Cat#CR130-100), 7 nM FIk-I (kinase domain), 1% DMSO, 50 mM HEPES pH 7.5, 10 mM MgCl2, 0.1 mM EDTA, 0.015% BRIJ, 0.1 mg/mL BSA, 0.1% mercapto-ethanol. Prior to the addition of enzyme, compounds were added to final concentrations ranging from 10 μM to 4.56 nM in 1% DMSO. The reaction was initiated upon addition of enzyme. Final reaction volume in each well was lOOμL. Time-resolved fluorescence was read after excitation at 340 nM. Emission readings were taken at both 665 and 615 nM on a Perkin Elmer Victor V Multilabel counter at 1.5 - 2.0 hrs after reaction initiation. Signal was calculated as follows: Emission 665 nm/ Emission 615 nM x 10000 for each well.
The compounds of examples 10,13, 16, 17, 18, 19, were tested and showed an IC50 value of less than 500 nM in this assay. In one embodiment, the present invention relates to a compound which exhibits an IC50 value of less than 500 nM. Method of Treating
Another embodiment of the present invention thus relates to a method of using the compounds described above, including salts thereof and corresponding compositions thereof, as cancer chemotherapeutic agents . This method comprises administering to a patient an amount of a compound of this invention, or a pharmaceutically acceptable salt thereof, which is effective to treat the patient's cancer. A patient, for the purpose of this invention, is a mammal, including a human, in need of treatment for a particular cancer. Cancers include but are not limited to 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.
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.
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.
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.
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, and urethral cancers.
Eye cancers include, but are not limited to intraocular melanoma and retinoblastoma. 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.
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, and lip and oral cavity cancer.
Lymphomas include, but are not limited to ADDS-related lymphoma, non- Hodgkin's lymphoma, cutaneous T-cell 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.
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.
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, chemotherapeutic, or other indication agents, and the like, as well as with admixtures and combinations thereof.
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), such as cisplatin.
Other anti-hyper-proliferative agents suitable for use with this 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) such as idarubicin. C. Operative examples relating to pharmaceutical compositions
The active compound can act systemically, locally or both. For this purpose it can be administered in a suitable manner, such as for example by oral, parenteral, pulmonary, nasal, sublingual, lingual, buccal, rectal, dermal, transdermal, conjunctival or aural administration or in the form of an implant or stent. The active compound can be administered in forms suitable for these modes of administration.
Suitable forms of oral administration are those according to the prior art which function by releasing the active compound rapidly or in a modified or controlled manner and which contain the active compound in a crystalline, amorphous, or dissolved form, for example tablets (which can be uncoated or coated, for example with enteric coatings or coatings which dissolve after a delay in time or insoluble coatings which control the release of the active compound), tablets or films (wafers), which disintegrate rapidly in the oral cavity, films/lyophilisates, capsules (e.g. hard or soft gelatin capsules), dragees, pellets, powders, emulsions, suspensions and solutions. An overview of application forms is given in Remington's Pharmaceutical Sciences, 18th ed. 1990, Mack Publishing Group, Enolo.
Parenteral administration can be carried out by avoiding an absorption step (e.g. by intravenous, intraarterial, intracardial, intraspinal or intralumbar administration) or by including absoiption (e.g. by intramuscular, subcutaneous, intracutaneous or intraperitoneal administration). Suitable parenteral administration forms are for example injection and infusion formulations in the form of solutions, suspensions, emulsions, lyophilisates and sterile powders. Such parenteral pharmaceutical compositions are described in Part 8, Chapter 84 of Remington's Pharmaceutical Sciences, 18l ed. 1990, Mack Publishing Group, Enolo.
Suitable forms of administration for the other modes of administration are for example inhalation devices (such as for example powder inhalers, nebulizers), nasal drops, solutions and sprays; tablets or films/wafers for lingual, sublingual or buccal administration or capsules, suppositories, ear and eye preparations, vaginal capsules, aqueous suspensions (lotions or shaking mixtures), lipophilic suspensions, ointments, creams, transdermal therapeutic systems, milky lotions, pastes, foams, dusting powders, implants or stents.
The active compounds can be converted into the abovementioned forms of administration in a manner known to the skilled man and in accordance with the prior art using inert, non-toxic, pharmaceutically suitable auxiliaries. The latter include for example excipients (e.g. macrocrystalline cellulose, lactose, mannitol, etc.), solvents (e.g. liquid polyethylene glycols), emulsifiers and dispersants or wetting agents (e.g. sodium dodecyl sulfate, polyoxysorbitan oleate etc.), binders (e.g. polyvinyl pyrrolidone), synthetic and/or natural polymers (e.g. albumin), stabilizers (e.g. antioxidants, such as, for example, ascorbic acid), dyes (e.g. inorganic pigments such as iron oxides) or taste- and/or odour-corrective agents.
The total amount of the active ingredient to be administered will generally range from about 0.01 mg/kg to about 200 mg/kg, and preferably from about 0.1 mg/kg to about 20 mg/kg body weight per day. 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. The 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 daily oral dosage regimen will preferably be from 0.01 to 200 mg/kg of total body weight.
It may however be necessary to deviate from the abovementioned quantities, depending on the body weight, mode of administration, the individual patient response to the active compound, the type of preparation and the time or interval of administration.
If used as active compounds, the compounds according to the invention are preferably isolated in more or less pure form, that is more or less free from residues from the synthetic procedure. The degree of purity can be determined by methods known to the chemist or pharmacist (see Remington's Pharmaceutical Sciences, 18th ed. 1990, Mack Publishing Group, Enolo). Preferably the compounds are greater than 99% pure (w/w), while purities of greater than 95%, 90% or 85% can be employed if necessary. The compounds according to the invention can be converted into pharmaceutical preparations as follows:
Tablet;
Composition:
100 mg of the compound of Example 1, 50 mg of lactose (monohydrate), 50 mg of maize starch (native), 10 mg of polyvinylpyrrolidone (PVP 25) (from BASF, Ludwigshafen,
Germany) and 2 mg of magnesium stearate.
Tablet weight 212 mg, diameter 8 mm, curvature radius 12 mm.
Preparation:
The mixture of active component, lactose and starch is granulated with a 5% solution (m/m) of the PVP in water. After drying, the granules are mixed with magnesium stearate for 5 min. This mixture is moulded using a customary tablet press (tablet format, see above). The moulding force applied is typically 15 kN.
Orally administrable suspension:
Composition:
1000 mg of the compound of Example 1, 1000 mg of ethanol (96%), 400 mg of Rhodigel
(xanthan gum from FMC, Pennsylvania, USA) and 99 g of water.
A single dose of 100 mg of the compound according to the invention is provided by 10 ml of oral suspension.
Preparation:
The Rhodigel is suspended in ethanol and the active component is added to the suspension. The water is added with stirring. Stirring is continued for about 6h until the swelling of the Rhodigel is complete.
It is believed that one skilled in the art, using the preceding information, can utilize the present invention to its fullest extent. It should be apparent to one of ordinary skill in the art that changes and modifications can be made to this invention without departing from the spirit or scope of the invention as it is set forth herein. Other embodiments of the invention will be apparent to the skilled in the art from a consideration of this specification or practice of the invention disclosed herein. It is intended that the specification and examples be considered as exemplary only, with the true scope and spirit of the invention being indicated by the following claims.

Claims

CLAIMS:We claim:
1. A compound of formula (I)
Figure imgf000100_0001
wherein
Ar is selected from the group consisting of
Figure imgf000100_0002
and
Figure imgf000100_0003
X is CH or N;
R1 is selected from the group consisting of H, halogen,
Figure imgf000101_0001
,R1"5
-N'
R1"3 and
Figure imgf000101_0002
wherein
R , 1-"2 is selected from the group consisting of
• H,
• (C1-C^aIkOXy,
• (C1-C4)alkyl, wherein said (Q-GOalkyl can be substituted with 0, 1, or 2 groups independently selected from
- hydroxy,
- (C1-C4)alkylamino,
- (Ci-COacyloxy,
- (CrC4)alkoxy, and
- (C2-C4)alkoxy substituted with 0,1 or 2 (CrC4)alkoxy groups,
- wherein said (Q-C^alkyl is independently optionally substituted with F up to the perfluoro level,
• 5- or 6-membered heteroaryl, or
• phenyl substituted with 0, 1, or 2 groups independently selected from (Q-GOalkyl, halo, nitro, (Q-GOalkoxy and cyano; R1"3 is H or (Ci-GOalkyl;
R1"4, R1"5 and R1"6 are independently selected from the group consisting of
• H,
• indan-5-yl,
• phenyl substituted with 0, 1, or 2 groups independently selected from (Ci-GOalkyl, halo, nitro, (Q-GOalkoxy and cyano,
• 5- or 6-membered heteroaryl substituted with 0,1 or 2 groups selected from
- cyano,
- halo,
- nitro,
- (d-C4)alkyl, wherein said (Ci~C4)alkyl is optionally substituted with 0, 1, or 2 groups selected from
(Q-GOalkylamino,
(d-C4)acyloxy,
(CrC4)alkoxy, and
(C2-C4)alkoxy substituted with up to 0, 1 or 2 (C1-
C4)alkoxy groups,
• (C3-C6)cycloalkyl substituted with 0,1 or 2 groups selected from (Q-GOalkyl, (Ci-C4)alkoxy, cyano, and halo, and
• (Q-C6)alkyl, wherein said (C1-C6)alkyl is independently substituted with 0 or 1 group selected from
- NH2,
- (CrC4)alkoxy,
(C2-C4)alkoxy independently substituted with 0,1, 2 or 3 (Q-Gøalkoxy and OH groups, and independently optionally substituted with fluorine up to the perfluoro level,
- carboxyl, (Ci-GOalkoxycarbonyl
- (Q-C^alkylarnino,
- aminocarbonyl,
- (C1-C4)alkylsulfonyl,
- phenyl substituted with 0, 1, or 2 groups independently selected from (Q-Gøalkyl, halo, nitro, (Q-C4)alkoxy and cyano,
- 5- or 6-membered heteroaryl independently substituted with 0, 1, 2 or 3 groups selected from (C1-C4)alkyl, (C1- C4)alkoxy, cyano, halo, and nitro and
- heterocyclyl independently substituted with 0, 1, 2 or 3 groups selected from (Q-GOalkyl, (C1-C4)alkoxy, cyano, and halo, and wherein said (C1-C6)alkyl is independently substituted with 0, 1 or 2 OH or halo groups,
- and wherein said (C1-C6)alkyl is independently optionally substituted with F up to the perfluoro level; and
R1'3 and R1"4, R1"3 and R1"5, and R1"3 and R1"6, when attached to the same nitrogen atom, may form, together with the N atom to which they are attached, a 5- or 6-membered saturated heterocyclic ring selected from pyrrolidinyl, morpholinyl, thiomorpholinyl and piperizinyl optionally substituted on N with (Ci-C4)alkyl;
or a pharmaceutically acceptable salt thereof.
2. The compound of claim 1, wherein Ar is selected from the group consisting of
Figure imgf000104_0001
and
Figure imgf000104_0002
X is CH or N;
R1 is selected from the group consisting of O
R 1-3
O
R 1-4
^NT
R 1-3
,R 1-5
-N'
R 1-3 and
Figure imgf000105_0001
wherein
R1"2 is selected from the group consisting of
• (CrC4)alkyl, wherein said (Q-C^alkyl is substituted with 1 or 2 groups independently selected from
- hydroxy,
- (Q-GOalkylamino,
- (C1-C4)acyloxy,
- (C1-C4)EIkOXy, and
- (C2-C4)alkoxy substituted with 0,1 or 2 (Ci-C4)alkoxy groups,
- wherein said (Q-C^alkyl is independently optionally substituted with F up to the perfluoro level,
• 5- or 6-membered heteroaryl, or
• phenyl substituted with 1 or 2 groups independently selected from (Ci-C4)alkyl, halo, nitro, (Q-C4)alkoxy and cyano;
R1"3 is H or (CrC4)alkyl;
R1"4 and R1"5 are independently selected from the group consisting of
• indan-5-yl,
• phenyl substituted with 1 or 2 groups independently selected from (C1-C4)alkyl, halo, nitro, (CrC4)alkoxy and cyano,
• 5- or 6-membered heteroaryl substituted with 1 or 2 groups selected from
- cyano,
- halo,
- nitro,
- (Ci-GOalkyl, wherein said (Ci-C4)alkyl is optionally substituted with 0, 1, or 2 groups selected from
(Ci-C^alkylamino,
(Ci-C4)acyloxy,
(Q-GOalkoxy, and
(C2-C4)alkoxy substituted with up to 0, 1 or 2 (C1-
C4)alkoxy groups,
• (C3-C6)cycloalkyl substituted with 1 or 2 groups selected from (C1-C4OaIkVl, (Q-C^alkoxy, cyano, and halo, and
• (C1-C6)alkyl, wherein said (CrC6)alkyl is independently substituted with 1 group selected from
- NH2,
- (C1-C^aIkOXy,
(C2-C4)alkoxy independently substituted with
0,1, 2 or 3 (CrC4)alkoxy and OH groups, and independently optionally substituted with fluorine up to the perfluoro level,
- carboxyl, (C1-C4)alkoxycarbonyl (C1-C4)alkylamino,
- aminocarbonyl,
- (Q-GOalkylsulfonyl,
- phenyl substituted with 0, 1, or 2 groups independently selected from (C1-C4)alkyl, halo, nitro, (Ci-C4)alkoxy and cyano,
- 5- or 6-membered heteroaryl independently substituted with 0, 1, 2 or 3 groups selected from (Q-GOalkyl, (C1- C4)alkoxy, cyano, halo, and nitro and - heterocyclyl independently substituted with 0, 1, 2 or 3 groups selected from (CrC4)alkyl, (Q-GOalkoxy, cyano, and halo,
- and wherein said (C1-C6)alkyl is independently substituted with 0, 1 or 2 OH or halo groups,
- and wherein said (C1-C6)alkyl is independently optionally substituted with F up to the perfluoro level;
R1"6 is selected from the group consisting of
• H
• indan-5-yl,
• phenyl substituted with 0, 1, or 2 groups independently selected from (CrC4)alkyl, halo, nitro, (Q-GOalkoxy and cyano,
• 5- or 6-membered heteroaryl substituted with 0,1 or 2 groups selected from cyano, halo,
- nitro,
Figure imgf000107_0001
wherein said (Q-GOalkyl is optionally substituted with 0, 1, or 2 groups selected from
(Q-C^alkylamino,
(Q-GOacyloxy,
(C1-C^aIkOXy, and
(C2-C4)alkoxy substituted with up to 0, 1 or 2 (C1-
C4)alkoxy groups,
• (C3-C6)cycloalkyl substituted with 0,1 or 2 groups selected from (Ci-C4)alkyl, (C1-C4)alkoxy, cyano, and halo, and
• (CrC6)alkyl, wherein said (C1-C6)alkyl is independently substituted with 0 or 1 group selected from - NH2,
- (Ci-C4)alkoxy,
- (C2-C4)alkoxy independently substituted with 0,1, 2 or 3 (Ci-COalkoxy and OH groups, and independently optionally substituted with fluorine up to the perfluoro level, carboxyl,
- (Q-C^alkoxycarbonyl
- (C1-C4)alkylamino, aminocarbonyl,
- (C1-C4)alkylsulfonyl, phenyl substituted with 0, 1, or 2 groups independently selected from (Q-COalkyl, halo, nitro, (Q-GOalkoxy and cyano,
- 5- or 6-membered heteroaryl independently substituted with 0, 1, 2 or 3 groups selected from (C1-C-OaIkVl, (C1- C4)alkoxy, cyano, halo, and nitro and
- heterocyclyl independently substituted with 0, 1, 2 or 3 groups selected from (Ci-C4)alkyl, (Q-GOalkoxy, cyano, and halo,
- and wherein said (Q-C^alkyl is independently substituted with 0, 1 or 2 OH or halo groups,
- and wherein said (CrC6)alkyl is independently optionally substituted with F up to the perfluoro level; and
R1"3 and R1"4, R1"3 and R1"5, and R1'3 and R1"6, when attached to the same nitrogen atom, may form, together with the N atom to which they are attached, a 5- or 6-membered saturated heterocyclic ring selected from pyrrolidinyl, moipholinyl, thiomorpholinyl and piperizinyl optionally substituted on N with (CrC4)alkyl; or a pharmaceutically acceptable salt thereof.
3. The compound of claim 1, wherein
Ar is selected from the group consisting of
Figure imgf000109_0001
and
Figure imgf000109_0002
X is CH;
R1 is selected from the group consisting of
Figure imgf000109_0003
and
Figure imgf000109_0004
wherein
R1-3 is H or (Ci-COalkyl, R1"5 is selected from the group consisting of indan-5-yl, phenyl substituted with 1 or 2 groups independently selected from
(Q-C-Oalkyl, halo, nitro, (Q-GOalkoxy and cyano, 5- or 6-membered heteroaryl substituted with 1 or 2 groups selected from cyano, halo, nitro, (Ci-Gύalkyl, wherein said (Q-GOalkyl is optionally substituted with 0, 1, or 2 groups selected from (C i -C4) alkylamino , (Ci-C4)acyloxy, (Ci-C4)alkoxy, and (C2-C4)alkoxy substituted with up to 0, 1 or 2 (Q-C^alkoxy groups; (C3-C6)cycloalkyl substituted with 1 or 2 groups selected from (C1-
C4)alkyl, (Q-GOalkoxy, cyano, and halo; and
(Ci-QOalkyl, wherein said (C1-C6)alkyl is independently substituted with lgroup selected from NH2,
(Ci-C4)alkoxy, (C2-C4)alkoxy independently substituted with
0,1, 2 or 3 (Ci-C4)alkoxy and OH groups, and independently optionally substituted with fluorine up to the perfluoro level, carboxyl,
(C i -C4) alkoxycarbonyl (C i -C4) alkylamino , aminocarbonyl, (Q-GOalkylsulfonyl, phenyl substituted with O, 1, or 2 groups independently selected from (C1-C4)alkyl, halo, nitro, (d-C4)alkoxy and cyano,
5- or 6-membered heteroaryl independently substituted with 0, 1, 2 or 3 groups selected from (CrC4)alkyl, (C1- C4)alkoxy, cyano, halo, and nitro and heterocyclyl is independently substituted with 0, 1, 2 or 3 groups selected from (Q-GOalkyl, (CrC4)alkoxy, cyano, and halo, and wherein said (Ci-C6)alkyl is independently substituted with 0, 1 or 2
OH or halo groups, and wherein said (Ci-C6)alkyl is independently optionally substituted with F up to the perfluoro level; R1" is selected from the group consisting of H, indan-5-yl, phenyl substituted with 0, 1, or 2 groups independently selected from (C1-C4)alkyl, halo, nitro, (CrC4)alkoxy and cyano, 5- or 6-membered heteroaryl substituted with 0, 1 or 2 groups selected from cyano, halo, nitro, (CrC4)alkyl, wherein said (Q-G^alkyl is optionally substituted with 0, 1, or 2 groups selected from (C1-C4)alkylamino, (Q-GOacyloxy,
(CrGOalkoxy, and
(C2-C4)alkoxy substituted with up to 0, 1 or 2 (C1-C-OaIkOXy groups; (C3-C6)cycloalkyl substituted with 0,1 or 2 groups selected from
(Q-C-Oalkyl, (Q-GOalkoxy, cyano, and halo; and
(Ci-C6)alkyl, wherein said (C1-C6)alkyl is independently substituted with 0 or 1 group selected from NH2,
(C1-C^aIkOXy, (C2-C4)alkoxy independently substituted with
0,1, 2 or 3 (CrC4)alkoxy and OH groups, and independently optionally substituted with fluorine up to the perfluoro level, carboxyl,
(C1-C4)alkoxycarbonyl (C1-C4)alkylamino, aminocarbonyl, (Q-C^alkylsulfonyl, phenyl substituted with 0, 1, or 2 groups independently selected from (C1-C4)alkyl, halo, nitro,
(C1-C4)alkoxy and cyano, 5- or 6-membered heteroaryl independently substituted with
0, 1, 2 or 3 groups selected from (Q-GOalkyl, (C1-
C4)alkoxy, cyano, halo, and nitro and heterocyclyl is independently substituted with 0, 1, 2 or 3 groups selected from (Q-C4)alkyl, (CrC4)alkoxy, cyano, and halo, and wherein said (Q-C^alkyl is independently substituted with 0, 1 or 2
OH or halo groups, and wherein said (Ci-C6)alkyl is independently optionally substituted with F up to the perfluoro level; and
R1"3 and R1"5, and R1"3 and R1"6, when attached to the same nitrogen atom, may form, together with the N atom to which they are attached, a 5- or 6-membered saturated heterocyclic ring selected from pyrrolidinyl, morpholinyl, thiomorpholinyl and piperizinyl optionally substituted on N with (Q-GOalkyl; or a pharmaceutically acceptable salt thereof.
4. The compound of claim 1, wherein
Ar is selected from the group consisting of
Figure imgf000113_0001
and
Figure imgf000113_0002
X is CH;
R1 is selected from the group consisting of
Figure imgf000113_0003
and
1-6
R1"3 R1"3 wherein
Rl-3 is H,
Figure imgf000114_0001
wherein said (CrC6)alkyl is independently substituted with 1 group selected from (Ci-C4)alkoxy,
(C2-C4)alkoxy independently substituted with 0,1, or 2 (Q-GOalkoxy and OH groups, and independently optionally substituted with fluorine up to the perfluoro level, and wherein said (Ci-C6)alkyl is independently substituted with 0, 1 or 2
OH or halo groups, and wherein said (Ci-C6)alkyl is independently optionally substituted with F up to the perfluoro level; R1"6 is selected from the group H, and (Ci-C6)alkyl, wherein said (Ci-C6)alkyl is independently substituted with 0 or 1 group selected from (Ci-COalkoxy, (C2-C4)alkoxy independently substituted with
0,1, 2 or 3 (Ci-Q)alkoxy and OH groups, and independently optionally substituted with fluorine up to the perfluoro level, and wherein said (C1-C6)OIlCyI is independently substituted with 0, 1 or 2
OH or halo groups, and wherein said (Ci-C6)alkyl is independently optionally substituted with F up to the perfluoro level;
or a pharmaceutically acceptable salt thereof.
5. The compound of claim 1 , wherein
Ar is selected from the group consisting of
Figure imgf000115_0001
and
Figure imgf000115_0002
X is CH;
R1 is selected from the group consisting of O
R1"3 R1"3 wherein R1-3 is H,
R1'6 is selected from the group H, and
Figure imgf000116_0001
wherein said (CrC6)alkyl is independently substituted with O or 1 group selected from (C1-C^aIkOXy, (C2-C4) alkoxy independently substituted with
0,1, 2 or 3 (C1-C^aIkOXy and OH groups, and independently optionally substituted with fluorine up to the perfluoro level, and wherein said (CrC6)alkyl is independently substituted with 0, 1 or 2
OH or halo groups, and wherein said (C1-C6)alkyl is independently optionally substituted with F up to the perfluoro level;
or a pharmaceutically acceptable salt thereof.
6. A process for making a compound of claim 1 , comprising
(A) reacting, with or without first hydrolizing the ester group -COOR', a compound of formula (VI)
Figure imgf000116_0002
wherein X and R1 have the meaning described in claim 1, and R' is lower alkyl,
with a compound of formula Ar-NH2 (VIII),
wherein Ar has the meaning described in claim 1; or
(B) reacting a compound of formula (IX)
Figure imgf000117_0001
wherein Ar has the meaning described in claim 1, with a compound of formula (IV)
Figure imgf000117_0002
wherein X and R1 have the meaning described in claim 1, and subsequently reducing the resulting compound; or
(C) reacting a compound of formula (IX), wherein Ar has the meaning described in claim 1, with a compound of formula (V)
Figure imgf000117_0003
wherein Ig represents a leaving group; or
(D) reacting a compound of formula (Ha)
Figure imgf000118_0001
wherein X and R1 have the meaning described in claim 1, and R' is lower alkyl, with a compound of formula (VIH)
ArNH2 (Vm),
wherein Ar has the meaning described in claim 1, in the presence of (R')3A1, wherein R' is lower alkyl, or
(E) first hydrolizing the ester group -COOR' of a compound of formula (Ha) as described above, and subsequently reacting the resulting compound with a compound of formula (VIII) as described above.
7. A compound of claim 1 for the treatment or prevention of disorders.
8. A pharmaceutical composition comprising the compound of claim 1.
9. The pharmaceutical composition of claim 8, additionally comprising at least one pharmaceutically acceptable carrier or excipient.
10. A pharmaceutical composition of claim 8 or 9 for the treatment or prevention of cancer.
11. A process for preparing the pharmaceutical composition of claim 9, comprising combining at least one compound according to claim 1 with at least one pharmaceutically acceptable carrier or excipient and bringing the resulting combination into a form suitable for said pharmaceutical composition.
12. A use of a compound of claim 1 for manufacturing a pharmaceutical composition for the treatment or prevention of a disease.
13. The use of claim 12, wherein the disease is cancer.
14. A method of treating a disease or condition in a mammal, comprising administering to a mammal in need thereof an effective amount of a compound of claim 1.
15. The method of claim 14, wherein the disease or condition is cancer.
PCT/US2006/021550 2005-06-03 2006-06-02 1-methyl-1h-pyrazole-4-carboxamides useful as cancer chemotherapeutic agents WO2006133006A2 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
JP2008514922A JP2008545756A (en) 2005-06-03 2006-06-02 1-methyl-1H-pyrazole-4-carboxamides useful as cancer chemotherapeutic agents
CA002610509A CA2610509A1 (en) 2005-06-03 2006-06-02 1-methyl-1h-pyrazole-4-carboxamides useful as cancer chemotherapeutic agents
EP06772019A EP1891047A4 (en) 2005-06-03 2006-06-02 1-methyl-1h-pyrazole-4-carboxamides useful as cancer chemotherapeutic agents

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US68745405P 2005-06-03 2005-06-03
US60/687,454 2005-06-03

Publications (2)

Publication Number Publication Date
WO2006133006A2 true WO2006133006A2 (en) 2006-12-14
WO2006133006A3 WO2006133006A3 (en) 2007-02-22

Family

ID=37498955

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2006/021550 WO2006133006A2 (en) 2005-06-03 2006-06-02 1-methyl-1h-pyrazole-4-carboxamides useful as cancer chemotherapeutic agents

Country Status (4)

Country Link
EP (1) EP1891047A4 (en)
JP (1) JP2008545756A (en)
CA (1) CA2610509A1 (en)
WO (1) WO2006133006A2 (en)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011028995A1 (en) * 2009-09-03 2011-03-10 Allergan, Inc. Compounds as tyrosine kinase modulators
WO2013083604A1 (en) 2011-12-09 2013-06-13 Chiesi Farmaceutici S.P.A. Kinase inhibitors
WO2013083606A1 (en) 2011-12-09 2013-06-13 Chiesi Farmaceutici S.P.A. Kinase inhibitors
WO2014195402A1 (en) 2013-06-06 2014-12-11 Chiesi Farmaceutici S.P.A. Kinase inhibitors
US9546165B2 (en) 2009-10-06 2017-01-17 Sunesis Pharmaceuticals, Inc. Heterocyclic compounds useful as PDK1 inhibitors
US10221192B2 (en) 2009-09-03 2019-03-05 Allergan, Inc. Compounds as tyrosine kinase modulators
WO2019068572A1 (en) 2017-10-04 2019-04-11 Bayer Aktiengesellschaft Derivatives of heterocyclic compounds as pest control agents
WO2019175046A1 (en) 2018-03-12 2019-09-19 Bayer Aktiengesellschaft Condensed bicyclic heterocyclic derivatives as pest control agents
WO2019201921A1 (en) 2018-04-20 2019-10-24 Bayer Aktiengesellschaft Heterocyclene derivatives as pest control agents

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AR105955A1 (en) * 2015-09-08 2017-11-29 Santen Pharmaceutical Co Ltd METHOD FOR PRODUCING A PYRIDINACARBOXAMIDE

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6878714B2 (en) * 2001-01-12 2005-04-12 Amgen Inc. Substituted alkylamine derivatives and methods of use
WO2003037336A1 (en) * 2001-11-02 2003-05-08 Pfizer Products Inc. 1-(5-sulfonyl-pyridin-2-yl)-5-(methylidene-cycloalkylmethoxy)-1h-pyrazole-4-carbonitrile derivatives and other compounds as cyclooxygenase inhibitors for the treatment of arthritis, neurodegeneration and colon cancer

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of EP1891047A4 *

Cited By (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2010289353B2 (en) * 2009-09-03 2016-12-08 Allergan, Inc. Compounds as tyrosine kinase modulators
US9328103B2 (en) 2009-09-03 2016-05-03 Allergan, Inc. Compounds as tyrosine kinase modulators
US10221192B2 (en) 2009-09-03 2019-03-05 Allergan, Inc. Compounds as tyrosine kinase modulators
US9725433B2 (en) 2009-09-03 2017-08-08 Allergan, Inc. Compounds as tyrosine kinase modulators
WO2011028995A1 (en) * 2009-09-03 2011-03-10 Allergan, Inc. Compounds as tyrosine kinase modulators
US8614234B2 (en) 2009-09-03 2013-12-24 Allergan, Inc. Compounds as tyrosine kinase modulators
CN102686577A (en) * 2009-09-03 2012-09-19 阿勒根公司 Compounds as tyrosine kinase modulators
US9475801B2 (en) 2009-09-03 2016-10-25 Allergan, Inc. Compounds as tyrosine kinase modulators
US9546165B2 (en) 2009-10-06 2017-01-17 Sunesis Pharmaceuticals, Inc. Heterocyclic compounds useful as PDK1 inhibitors
US10030016B2 (en) 2009-10-06 2018-07-24 Sunesis Pharmaceuticals, Inc. Heterocyclic compounds useful as PDK1 inhibitors
WO2013083606A1 (en) 2011-12-09 2013-06-13 Chiesi Farmaceutici S.P.A. Kinase inhibitors
WO2013083604A1 (en) 2011-12-09 2013-06-13 Chiesi Farmaceutici S.P.A. Kinase inhibitors
WO2014195402A1 (en) 2013-06-06 2014-12-11 Chiesi Farmaceutici S.P.A. Kinase inhibitors
WO2019068572A1 (en) 2017-10-04 2019-04-11 Bayer Aktiengesellschaft Derivatives of heterocyclic compounds as pest control agents
WO2019175046A1 (en) 2018-03-12 2019-09-19 Bayer Aktiengesellschaft Condensed bicyclic heterocyclic derivatives as pest control agents
WO2019201921A1 (en) 2018-04-20 2019-10-24 Bayer Aktiengesellschaft Heterocyclene derivatives as pest control agents

Also Published As

Publication number Publication date
EP1891047A4 (en) 2008-07-09
JP2008545756A (en) 2008-12-18
WO2006133006A3 (en) 2007-02-22
EP1891047A2 (en) 2008-02-27
CA2610509A1 (en) 2006-12-14

Similar Documents

Publication Publication Date Title
EP1891047A2 (en) 1-methyl-1h-pyrazole-4-carboxamides useful as cancer chemotherapeutic agents
ES2935173T3 (en) Pharmaceutical compositions comprising imidazothiadiazole and imidazopyridazine derivatives as protease-activated receptor 4 (PAR4) inhibitors to treat platelet aggregation
CA2691507C (en) 5-membered heterocyclic amides and related compounds
US9518064B2 (en) Imidazothiadiazole and imidazopyridazine derivatives as protease activated receptor 4 (PAR4) inhibitors for treating platelet aggregation
EP2366699B1 (en) 8-substituted isoquinoline derivative and use thereof
CA3085561A1 (en) Cyclohexyl acid triazole azines as lpa antagonists
AU2003290744A1 (en) Indolyl pyrazinone derivatives useful for treating hyper-proliferative disorders and diseases associated with angiogenesis
US20150203479A1 (en) Androgen receptor modulating compounds
KR20070045227A (en) Condensed pyridines as kinase inhibitors
CA2480638A1 (en) Substituted benzazoles and use thereof as raf kinase inhibitors
US20080293696A1 (en) 2-Aminoarylcarboxamides Useful as Cancer Chemotherapeutic Agents
WO2019101086A1 (en) Halo-allylamine ssao/vap-1 inhibitor and use thereof
CA2530281A1 (en) Substituted tetrahydrobenzothienopyrimidinamine compounds useful for treating hyper-proliferative disorders
AU2012291041A1 (en) Uracil derivative and use thereof for medical purposes
WO2022112352A1 (en) Heterocycle derivatives for treating trpm3 mediated disorders
TW201348213A (en) Quinazolinedione derivative
CA2567352A1 (en) 5-anilino-4-heteroarylpyrazole derivatives useful for the treatment of diabetes
AU2019383845A1 (en) Novel tricyclic compound as IRAK4 inhibitor
EP1858893A1 (en) 1,3-thiazole-5-carboxamides useful as cancer chemotherapeutic agents
WO2006023707A2 (en) 2-aminothiophenecarboxamides useful as cancer chemotherapeutic agents
IL296632A (en) Phd inhibitor compounds, compositions, and use
KR20070120182A (en) Pyrimidine derivatives and their use in the treatment of cancer

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2006772019

Country of ref document: EP

ENP Entry into the national phase in:

Ref document number: 2610509

Country of ref document: CA

ENP Entry into the national phase in:

Ref document number: 2008514922

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase in:

Ref country code: DE