US20070191401A1 - Indolylmaleimide derivatives - Google Patents

Indolylmaleimide derivatives Download PDF

Info

Publication number
US20070191401A1
US20070191401A1 US11/708,843 US70884307A US2007191401A1 US 20070191401 A1 US20070191401 A1 US 20070191401A1 US 70884307 A US70884307 A US 70884307A US 2007191401 A1 US2007191401 A1 US 2007191401A1
Authority
US
United States
Prior art keywords
alkyl
methyl
mmol
formula
compound
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/708,843
Inventor
Rainer Albert
Nigel Cooke
Sylvain Cottens
Claus Ehrhardt
Jean-Pierre Evenou
Richard Sedrani
Peter Von Matt
Jurgen Wagner
Gerhard Zenke
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Individual
Original Assignee
Individual
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 Individual filed Critical Individual
Priority to US11/708,843 priority Critical patent/US20070191401A1/en
Publication of US20070191401A1 publication Critical patent/US20070191401A1/en
Priority to US12/498,486 priority patent/US7786123B2/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/02Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings
    • C07D231/10Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D231/12Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with only hydrogen atoms, hydrocarbon or substituted hydrocarbon radicals, directly attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D233/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings
    • C07D233/54Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members
    • C07D233/56Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members with only hydrogen atoms or radicals containing only hydrogen and carbon atoms, attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D249/00Heterocyclic compounds containing five-membered rings having three nitrogen atoms as the only ring hetero atoms
    • C07D249/02Heterocyclic compounds containing five-membered rings having three nitrogen atoms as the only ring hetero atoms not condensed with other rings
    • C07D249/081,2,4-Triazoles; Hydrogenated 1,2,4-triazoles
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/10Spiro-condensed systems

Definitions

  • the present invention relates to indolylmaleimide derivatives, process for their production and pharmaceutical compositions containing them.
  • alkyl or alkyl moiety in e.g. alkoxy may be linear or branched.
  • Halogen may be F, Cl, Br or I, preferably F or Cl.
  • Any aryl may be phenyl or naphthyl, preferably phenyl.
  • heterocyclic residue as R 1 , R 4 , R 7 , R 8 , R 11 , R 14 or Y or formed, respectively, by NR 16 R 17 or NR 19 R 20 is meant a three to eight, preferably five to eight, membered saturated, unsaturated or aromatic heterocyclic ring comprising 1 or 2 heteroatoms, preferably selected from N, O and S, and optionally substituted.
  • Suitable examples include e.g. pyridyl, e.g. 3- or 4-pyridyl, piperidyl, e.g.
  • heterocyclic residue when substituted, this may be on one or more ring carbon atoms and/or on a ring nitrogen atom when present.
  • substituent on a ring carbon atom include e.g. C 1-4 alkyl e.g. CH 3 ;
  • C 3-6 cycloalkyl e.g. cyclopropyl, optionally further substituted by C 1-4 alkyl; wherein p is 1, 2 or 3, preferably 1; CF 3 ; halogen; OH; NH 2 ; —CH 2 -NH 2 ; —CH 2 —OH; piperidin-1-yl; or pyrrolidinyl.
  • a substituent on a ring nitrogen atom are e.g. C 1-6 alkyl; acyl, e.g.
  • R′ x is H, C 1-6 alkyl or phenyl optionally substituted by C 1-4 alkyl, C 1-4 alkoxy or amino, e.g formyl; C 3-6 cycloalkyl; C 3-6 cycloalkyl-C 1-4 alkyl; phenyl; phenyl-C 1-4 alkyl e.g. benzyl; a heterocyclic residue, e.g. as disclosed above, e.g. an aromatic heterocyclic residue comprising 1 or 2 nitrogen atoms; or a residue of formula ⁇ —R 21 —Y′ ( ⁇ )
  • R 21 is C 1-4 alkylene or C 2-4 alkylene interrupted by O and Y′ is OH, NH 2 , NH(C 1-4 alkyl) or N(C 1-4 alkyl) 2 .
  • C 2-4 alkylene interrupted by O may be e.g. —CH 2 —CH 2 —O—CH 2 —CH 2 —.
  • the substituent on a cyclic nitrogen is a heterocyclic residue, it may be a five or six membered saturated, unsaturated or aromatic heterocyclic ring comprising 1 or 2 heteroatoms, preferably selected from N, O and S.
  • Examples include e.g. 3- or 4-pyridyl, piperidyl, e.g. piperidin-1-yl, 3- or 4-piperidyl, homopiperidyl, piperazinyl, homopiperazinyl, pyrimidinyl, morpholin-4-yl, imidazolyl, imidazolidinyl, pyrrolyl or pyrrolidinyl,
  • R a is substituted C 1-4 alkyl
  • the substituent is preferably on the terminal carbon atom.
  • ring A When ring A is substituted, it may be mono- or polysubstituted, preferably monosubstituted, the substituent(s) being selected from the group consisting of e.g. halogen, OH, C 1-4 alkoxy, e.g. OCH 3 , C 1-4 alkyl, e.g. CH 3 , NO 2 , CF 3 , NH 2 , NHC 1-4 alkyl, N(C 1-4 alkyl) 2 and CN.
  • substituent(s) being selected from the group consisting of e.g. halogen, OH, C 1-4 alkoxy, e.g. OCH 3 , C 1-4 alkyl, e.g. CH 3 , NO 2 , CF 3 , NH 2 , NHC 1-4 alkyl, N(C 1-4 alkyl) 2 and CN.
  • ring A may be a residue of formula
  • R d is in position 1; preferably Re is in position 3.
  • R c has a CH 2 replaced by CR x R y , it is preferably the CH 2 bearing Y.
  • heterocyclic residue examples include e.g. a residue of formula ( ⁇ )
  • a preferred residue of formula ( ⁇ ) is one wherein the ring P forms a 1,4-piperazinyl ring optionally C- and/or N-substituted as indicated.
  • a residue of formula ( ⁇ ) are e.g. 3- or 4- pyridyl; piperidin-1-yl; 1—N-(C 1-4 alkyl)— or (co-hydroxy-C 1-4 alkyl)-3-piperidyl; morpholin-4-yl; imidazolyl; pyrrolidinyl; 1-piperazinyl; 2-C 1-4 alkyl- or -C 3-6 cycloalkyl-1-piperazinyl ;3-C 1-4 alkyl- or -C 3-6 cycloalkyl-1-piperazinyl; 2,2- or 3,5- or 2,5- or 2,6-di(C 1-4 alkyl)-1-piperazinyl; 3,4,5-tri-(C 1-4 alkyl)-1-piperazinyl; 4-N-(C 1-4 alkyl)- or -( ⁇ -hydroxy-C 1-4 alkyl)- or -( ⁇ -dimethylamino-C 1-4 alkyl)
  • the compounds of formula I may exist in free form or in salt form, e.g. addition salts with e.g. organic or inorganic acids, for example, hydrochloric acid, acetic acid, when R 1 , R 4 , R 7 , R 8 , R 11 or R 14 and/or R 2 , R 3 , R 5 , R 6 , R 9 , R 10 , R 12 , R 13 or R 15 comprises an optionally substituted amino group or a heterocyclic residue which can form acid addition salts.
  • addition salts with e.g. organic or inorganic acids, for example, hydrochloric acid, acetic acid
  • the compounds of formula I may exist in the form of optical isomers, racemates or diastereoisomers.
  • a ring carbon atom bearing a substituent in the heterocyclic residue as R 1 , R 4 , R 7 , R 8 , R 11 , R 14 or Y or formed, respectively, by NR 16 R 17 or NR 19 R 20 is asymmetric and may have the D- or L-configuration.
  • the present invention embraces all enantiomers and their mixtures. Similar considerations apply in relation to starting materials exhibiting asymetric carbon atoms as mentioned.
  • the present invention also includes a process for the preparation of a compound of formula I which process comprises
  • Process steps a) and (b) may conveniently be effected in the presence of a strong base, e.g. t-BuOK.
  • a strong base e.g. t-BuOK.
  • compounds of formula III or V comprising an OH group which should not participate to the reaction are used, such OH group is in protected form.
  • the OH-protecting group may be removed according to methods known in the art at the end of condensation step a) or b).
  • Process step c) may be carried out according to known methods: for example when R 1 , R 4 , R 7 , R 8 , R 11 or R 14 comprises a final OH group, this OH group may be replaced by the desired —NR 16 R 17 or —NR 19 R 20 .
  • Compounds of formula 11 may be prepared by reacting the corresponding indol compound with an oxalyl halogenide, e.g. chloride, or with a monoalkyl oxalyl chloride under basic conditions, e.g. as disclosed in Example 28.
  • an oxalyl halogenide e.g. chloride
  • a monoalkyl oxalyl chloride under basic conditions, e.g. as disclosed in Example 28.
  • compounds of formula III wherein R is a radical of formula (a), (b) or (c), R 1 , R 4 or R 7 being a radical of formula ( ⁇ ), may be prepared in accordance with known methods by reacting a compound of formula III′ wherein R′ is respectively a radical of formula (a), (b) or (c), each of which comprising OH in place of R 1 , R 4 or R 7 , with a compound of formula X a —X—R c —Y wherein X a is a leaving group, e.g. Cl, and X, R c or Y are as defined above.
  • R 12 and R 13 are as defined above and X a is a leaving group, e.g. halogen, and with a compound of formula VI R′ 11 H (VI)
  • R′ 11 is —O—R c —Y or —S—R c —Y. This reaction may be carded out in accordance with know methods, e.g. as disclosed in Example 28 below.
  • R 9 , R 10 , R 15 and R′ 15 are as defined above and X a is a leaving group, e.g. halogen, and with a compound of formula VI′ A—H (VI)
  • A is —O—R c —Y or —S—R c —Y. This reaction may be carried out in accordance with know methods.
  • 3-(1.H.-Indol-3-yl)4-[3-(2-methanesulfonyloxy-ethoxy)-5-hydroxy-phenyl]-pyrrole-2,5-dione, used as starting material, may be prepared as follows:
  • R 1 and R 2 are as defined in Table 1, may be prepared by following the procedure of Example 1 but using the appropriate starting materials. Starting materials which do not comprise a OH substituent will be prepared without the protecting steps as indicated in Example 1. TABLE 1 Example R 1 R 2 R 3 R a M.S.
  • 3-Indol-3-yl)-4-[3-(2-methanesulfonyloxy-ethoxy)-naphthalen-1-yl]-pyrrole-2,5-dione, used as starting materials may be prepared as follows:
  • R 3 is as defined in Table 2, may be prepared by following the procedure of Example 28 but using the appropriate starting materials.
  • TABLE 2 Ex. R 3 R M.S. Data 29 —O—CH 2 —CH 2 -(pyrrolidin-1- H MH + 452 yl) 30 H MH + 456 31 —O—CH 3 H (M ⁇ H) ⁇ 367 32 OH H (M ⁇ H) ⁇ 353 33 —O—(CH 2 ) 3 —N(CH 3 ) 2 H MH + 440 34 —O—(CH 2 ) 2 —N(CH 3 )- H MH + 452 cyclopropyl 35 —O—(CH 2 ) 2 —N(CH 3 )- CH 3 MH + 466 cyclopropyl 36 —O—(CH 2 ) 2 —NH-cyclopropyl CH 3 (M + H) + 452 37 H (M + H) + 438.2 (M ⁇ H) ⁇ 436.3 38 CH
  • Examples 48 and 52 which are racemates may also be prepared in the form of pure cis or trans enantiomers using the corresponding cis or trans starting materials.
  • step a) (7.28 g, 28.75 mmol) is dissolved in CH 2 Cl 2 (60 ml). 2 equivalents m-chloroperbenzoic acid (11.7 g, FLUKA 25800, 70%) is added at RT and after addition of sodium sulfate (5 g) the reaction mixture is treated with trifluoromethanesulfonic acid (250 ⁇ l, 2.88 mmol). The reaction is kept at RT for 16 hours. After thin layer control, m-chloroperbenzoic acid (2.25 g, 11 mmol), sodium sulfate (2 g) and trifluoromethanesulfonic acid (50 ⁇ l, 0.57 mmol) is added to the reaction (2 times within 6 hours).
  • the organic layer is filtered and extracted 3 times with an aqueous sodium thiosulfite solution.
  • the compound is purified on silica gel using a mixture of cyclo-hexane/ethyl acetate 2/1 to 1/1 obtaining the product as colorless oil.
  • step b) Compound of step b) (6.1 g, 22.66 mmol) is added to MeOH (200 ml) at RT. 1N sodium methoxide in methanol (22.7 ml) is stirred for 15 min. Amberlite IR-120 (H + -form) is added to the mixture until the reaction mixture becomes neutral. The ion exchanger is filtered off, the solvent is evaporated and the crude product is isolated without any further purification as a pale yellow solid.
  • step c) Compound of step c) (2.8 g, 12.33 mmol) is added to THF (100 ml) at RT. Triphenyl-phosphine (16.82 g, 64.13 mmol) and (after 10 minutes) diisopropylazodicarboxylate (11.66 ml, 59.2 mmol) are added and the mixture is stirred for 14 hours. The solvent is removed and the residue is purified on silica gel using cyclohexane (100%) to cyclohexane/-methylene chloride (1/1) as mobile phase, obtaining the product as a pale yellow solid.
  • step b) (3-Hydroxy-5,6,7,8-tetrahydro-naphthalen-1-yl)-acetic acid ethyl ester DL-Methionine (1.11 g, 7.5 mmol) and compound of step b) is dissolved in methanesulfonic acid (9 ml) and kept at RT over night. The reaction is poured into a ice-cold saturated NaCl solution and extracted 3 times with methylene chloride. The organic layer is dried, concentrated and purified on silica gel (methylene chloride/methanol 95/5) affording the pure compound.
  • R 1 , R 5 , R 6 and R 7 are as defined in Table 3 below, may be prepared by following the procedure of Example 56 but using the appropriate starting materials. The compounds may also be converted into the acetate salt as disclosed for the compound of Example 56, where appropriate. TABLE 3 R 6 (position 7 R 7 (position 5 Ex. R 5 R 1 or 8) or 6) M.S.
  • Example 77 which is a racemate may also be prepared in the form of pure cis or trans enantiomer using the corresponding cis or trans starting materials.
  • Compounds of Examples 116 and 117 may also be prepared as a racemate or in the corresponding S or R isomer form.
  • R, R 1 and R 2 are as defined in Table 4 below, may be prepared by following the procedure of Example 56 but using the appropriately substituted (1.H.-indol-3-yl)-oxo-acetic acid ethyl ester. TABLE 4 M.S. Data (ESI- Ex.
  • the compounds are prepared by following the procedure of Example 56 but using the appropriately substituted (1.H.-indol-3-yl)-oxo-acetic acid methyl ester.
  • R, R 1 and R 2 are as defined in Table 5 below, may be prepared by following the procedure of Example 56 but using the appropriately substituted (1.H.-indol-3-yl)-oxo-acetic acid methyl ester. TABLE 5 Ex. R 14 R 15 R′ 15 R 1 M.S.
  • 2-(2-chloro-quinolin-4-yl)-acetic acid methyl ester may be prepared as disclosed in EP-A1-0 364 327 and Indian J. Chem. 1994, 33B, 747-751.
  • 2-(2-chloro-quinolin-4-yl)-acetamide (500 mg, 2.27 mmol) is dissolved in 1-methyl-2-pyrrolidinone (3.0 mL). N-methylpiperazin (1.3 mL, 5 eq.) is added and the reaction is stirred at 80° C. for 48 h. AcOEt (20 mL) is added and the precipitate is purified by FCC (AcOEt/EtOH/28% NH 4 OH 80:18:1) to afford 2-[2-(4-methyl-piperazin-1-yl)-quinolin-4-yl]-acetamide as a solid.
  • R 8 and R 9 are as defined in Table 6 below, may be prepared by following the procedure of Example 162 or 163 but using the appropriate starting materials. TABLE 6 Ex. R 8 Position-R 9 R a M.S Data 164 Cl H H 374 (M + H) + 165 —OCH 3 6-CH 3 H 384 (M + H) + 166 —O—CH2—CH 2 —N(CH 3 )2 6-CH 3 H 441 (M + H) + 167 —O—(CH 2 ) 3 —N(CH 3 ) 2 6-CH 3 H 455 (M + H) + 168 —OCH 3 H H H 370 (M + H) + 169 —OCH 3 7-CH 3 H 384 (M + H) + 170 —O—CH 2 —CH 2 —N(CH 3 ) 2 7-CH 3 H 441 (M + H) + 171 -(4-N-methyl)-1-piperazinyl 7-CH 3 H 452 (M + H) + 172
  • 3-(3-Chloro-isoquinolin-1-yl)-4-(1.H.-indol-3-yl)-pyrrole-2,5-dione (110 mg, 0.30 mmol) is suspended in N-methylpiperazine (2.5 ml) and kept at 130° C. for 24 hours. The excess of N-methylpiperazine is removed at high vacuo at 60° C. and the residue is dissolved. in ethyl acetate and extracted with 0.5N aqueous H 2 O. The deep orange aqueous phase is adjusted to pH 9 with 1N NaOH and extracted with ethyl acetate.
  • the organic phase is separated, dried with Na 2 SO 4 , concentrated and purified on silica gel using methylene chloride/methanol/acetic acid 50% (9/1/0.25) as mobile phase.
  • the fractions with the title compound are collected and extracted with aqueous NaHCO 3 solution (6%).
  • the organic phase is separated, dried with Na 2 SO 4 , and concentrated to give pure title compound as orange powder.
  • 3-(3-Chloro-isoquinolin-1-yl)-4-(1.H.-indol-3-yl)-pyrrole-2,5-dione, used as starting material, may be prepared as follows:
  • a) (3-chloro-isoquinolin-1-yl) acetic acid ethyl ester may be prepared as disclosed by T. Kametani et al. in Chem. Pharm. Bull., 15(5), 704 (1967).
  • 2-[3-(4-Methyl-piperazin-1-yl)-isoquinolin-1-yl]-acetamide,- used as starting material, may be prepared as follows:
  • the aqueous phase is adjusted to pH 8.5 with solid sodium bicarbonate and extracted with methylene chloride (3 ⁇ ).
  • the combined organic phases are dried (Na 2 SO 4 ) and concentrated.
  • the resulting crude is purified on silica gel using methylene chloride (100%) ⁇ methylene chloride/methanol (90/10) as eluent obtaining the product as a pale yellow solid.
  • R a , R c , R 8 and R 9 are as defined in Table 7 below, may be prepared by following the procedure of Example 181 but using the appropriate starting materials.
  • the compounds of formula I in free form or in pharmaceutically acceptable salt form exhibit valuable pharmacological properties, e.g. inhibiting Protein Kinase C (PKC), e.g. PKC isoforms like ⁇ , ⁇ , ⁇ , ⁇ , ⁇ or ⁇ activity, inhibiting T-cell activation and proliferation, e.g. by inhibiting production by T-cells or cytokines, e,g, IL-2, by inhibiting the proliferative response of T-cells to cytokines, e.g. IL-2, e.g. as indicated in in vitro and in vivo tests and are therefore indicated for therapy.
  • PKC Protein Kinase C
  • the compounds of formula I are tested for their activity on different PKC isoforms according to a published method (D. Geiges et al. Biochem. Pharmacol. 1997;53:865-875)
  • the assay is performed in a 96-well polypropylene microtiterplate (Costar 3794) that has been previously siliconized with Sigmacote (Sigma SL-2).
  • the reaction mixture (50 ⁇ l) contains 10 ⁇ l of the relevant PKC isozyme together with 25 ⁇ l of the test compound and 15 ⁇ l of a mix solution that contains 200 ⁇ g/ml protamine sulfate, 10 mM Mg(NO 3 ) 2 , 10 ⁇ M ATP (Boehringer 519987) and 3750 Bq of 33 P-ATP (Hartmann Analytic SFC301 110 TBq/mmol) in 20 mM Tris-buffer pH 7.4+0.1% BSA. Incubation is performed for 15 min at 32° C. in a microtiterplate shaking incubator (Biolabo Scientific Instruments). Reaction is stopped by adding 10 ⁇ l of 0.5 M Na 2 EDTA, pH 7.4.
  • IC 50 measurement is performed on a routine basis by incubating a serial dilution of inhibitor at concentrations ranging between 1-1000 ⁇ M according to the method described above. IC 50 value are calculated from the graph by sigmoidal curve fitting.
  • Human recombinant PKCO is used under the assay conditions as described above.
  • compounds of formula I inhibit PKC ⁇ with an IC 50 ⁇ 1 ⁇ M.
  • Compound of Example 56 inhibits PKC ⁇ in this assay with an IC 50 ⁇ 10 nM.
  • the assay is performed with Jurkat cells transfected with a human interleukin-2 promoter/reporter gene construct as described by Baumann G et al. in Transplant. Proc. 1992;24:43-8, the ⁇ -galactosidase reporter gene being replaced by the luciferase gene (de Wet J., et al., Mol. Cell Biol. 1987, 7(2), 725-737).
  • Cells are stimulated by solid phase-coupled antibodies or phorbol myristate acetate (PMA) and the Ca ++ ionophore ionomycin as follows.
  • Microlite TM1 microtiter plates (Dynatech) are coated with 3 ⁇ g/ml goat anti-mouse IgG Fc antibodies (Jackson) in 55 ⁇ l phosphate-buffered saline (PBS) per well for three hours at room temperature. Plates are blocked after removing the antibodies by incubation with 2% bovine serum albumin (BSA) in PBS (300 ⁇ l per well) for 2 hours at room temperature.
  • BSA bovine serum albumin
  • test compounds in duplicates in assay medium RPMI 1640/10% fetal calf serum (FCS) containing 50 ⁇ M 2-mercaptoethanol, 100 units/ml penicillin and 100 ⁇ g/ml streptomycin
  • FCS fetal calf serum
  • 100 ⁇ l of this mixture containing 1 ⁇ 10 5 cells are then transferred to the antibody-coated assay plates.
  • 100 ⁇ l are incubated with 40 ng/ml PMA and 2 82 M ionomycin. After incubation for 5.5 hours at 37° C.
  • the level of luciferase is determined by bioluminescence measurement.
  • the plates are centrifuged for 10 min at 500 g and the supernatant is removed by flicking.
  • Lysis buffer containing 25 mM Tris-phosphate, pH 7.8, 2 mM DTT, 2 mM 1.2-diaminocyclohexane-N,N,N′,N-tetraacetic acid, 10% (v/v) glycerol and 1% (v/v) Triton X-100 is added (20 ⁇ l per well). The plates are incubated at room temperature for 10 minutes under constant shaking.
  • Luciferase activity is assessed with a bioluminescence reader (Labsystem, Helsinki, Finland) after automatic addition of 50 ⁇ l per well luciferase reaction buffer containing 20 mM Tricine, 1.07 mM (MgCO 3 ) 4 Mg(OH) 2 ⁇ 5H 2 O, 2.67 mM MgSO 4 , 0.1 mM EDTA, 33.3 mM DTT, 270 ⁇ M coenzyme A, 470 ⁇ M luciferin (Chemie Brunschwig AG), 530 ⁇ M ATP, pH 7.8.
  • Lag time is 0.5 seconds, total measuring time is 1 or 2 seconds.
  • Low control values are light units from anti-T cell receptor- or PMA-stimulated cells
  • high controls are from anti-T cell receptor/anti-CD28- or PMA/ionomycin-stimulated cells without any test sample.
  • Low controls are subtracted from all values.
  • the inhibition obtained in the presence of a test compound is calculated as percent inhibition of the high control.
  • the concentration of test compounds resulting in 50% inhibition (IC 50 ) is determined from the dose-response curves.
  • compounds of formula I inhibit anti-T cell receptor/anti-CD28 and PMA/ionomycin stimulated Jurkat cells with an IC 50 ⁇ 1 ⁇ M.
  • Compound of Example 56 has an IC 50 of 42 ⁇ 12 nM in this assay.
  • the two-way MLR is performed according to standard procedures (J. Immunol. Methods, 1973, 2, 279 and Meo T. et al., Immunological Methods, New York, Academic Press, 1979, 227-39). Briefly, spleen cells from CBA and BALB/c mice (1.6 ⁇ 10 5 cells from each strain per well in flat bottom tissue culture microtiter plates, 3.2 ⁇ 10 5 in total) are incubated in RPMI medium containing 10% FCS, 100 U/ml penicillin, 100 ⁇ g/ml streptomycin (Gibco BRL, Basel, Switzerland), 50 ⁇ M 2-mercaptoethanol (Fluka, Buchs, Switzerland) and serially diluted compounds.
  • the strain combination used Male Lewis (RT 1 haplotype) and BN (RT 1 haplotype).
  • the animals are anaesthetised using inhalational isofluorane. Following heparinisation of the donor rat through the abdominal inferior vena cava with simultaneous exsanguination via the aorta, the chest is opened and the heart rapidly cooled. The aorta is ligated and divided distal to the first branch and the brachiocephalic trunk is divided at the first bifurcation. The left pulmonary artery is ligated and divided and the right side divided but left open. All other vessels are dissected free, ligated and divided and the donor heart is removed into iced saline.
  • the recipient is prepared by dissection and cross-clamping of the infra-renal abdominal aorta and vena cava.
  • the graft is implanted with end-to-side anastomoses, using 10/0 monofilament suture, between the donor brachiocephalic trunk and the recipient aorta and the donor right pulmonary artery to the recipient vena cava.
  • the clamps are removed, the graft tethered retroabdominally, the abdominal contents washed with warm saline and the animal is closed and allowed to recover under a heating lamp.
  • Graft survival is monitored by daily palpation of the beating donor heart through the abdominal wall. Rejection is considered to be complete when heart beat stops. Increases of graft survival are obtained in animals treated with a compound of formula I administered orally at a daily dose of 1 to 30 mg/kg. Thus compound of Example 100 significantly increases the graft survival when administered at a dose of 30 mg/kg/day.
  • the compounds of formula I are, therefore, useful in the treatment and/or prevention of diseases or disorders mediated by T lymphocytes and/or PKC, e.g. acute or chronic rejection of organ or tissue allo- or xenografts, atherosclerosis, vascular occlusion due to vacular injury such as angioplasty, restenosis, hypertension, heart failure, chronic obstructive pulmonary disease, CNS diseases such as Alzheimer disease or amyotrophic lateral sclerosis, cancer, infectious diseases such as AIDS, septic shock or adult respiratory distress syndrome, ischemia/reperfusion injury e.g. myocardial infarction, stroke, gut ischemia, renal failure or hemorrhage shock, or traumatic shock.
  • diseases or disorders mediated by T lymphocytes and/or PKC e.g. acute or chronic rejection of organ or tissue allo- or xenografts, atherosclerosis, vascular occlusion due to vacular injury such as angioplasty, restenosis, hypertension
  • the compounds of formula I are also useful in the treatment and/or prevention of T-cell mediated acute or chronic inflammatory diseases or disorders or autoimmune diseases e.g. rheumatoid arthritis, osteoarthritis, systemic lupus erythematosus, Hashimoto's thyroidis, multiple sclerosis, myasthenia gravis, diabetes type I or II and the disorders associated therewith, respiratory diseases such as asthma or inflammatory lung injury, inflammatory liver injury, inflammatory glomerular injury, cutaneous manifestations of immunologically-mediated disorders or illnesses, inflammatory and hyperproliferative skin diseases (such as psoriasis, atopic dermatitis, allergic contact dermatitis, irritant contact dermatitis and further eczematous dermatitises, seborrhoeic dermatitis), inflammatory eye diseases, e.g.
  • Sjoegren's syndrome, keratoconjunctivitis or uveitis, inflammatory bowel disease, Crohn's disease or ulcerative colitis For the above uses the required dosage will of course vary depending on the mode of administration, the particular condition to be treated and the effect desired. In general, satisfactory results are indicated to be obtained systemically at daily dosages of from about 0.1 to about 100 mg/kg body weight.
  • An indicated daily dosage in the larger mammal, e.g. humans, is in the range from about 0.5 mg to about 2000 mg, conveniently administered, for example, in divided doses up to four times a day or in retard form.
  • the compounds of formula I may be administered by any conventional route, in particular enterally, e.g. orally, e.g. in the form of tablets or capsules, or parenterally, e.g. in the form of injectable solutions or suspensions, topically, e.g. in the form of lotions, gels, ointments or creams, or in a nasal or a suppository form.
  • Pharmaceutical compositions comprising a compound of formula I in free form or in pharmaceutically acceptable salt form in association with at least one pharmaceutical acceptable carrier or diluent may be manufactured in conventional manner by mixing with a pharmaceutically acceptable carrier or diluent.
  • Unit dosage forms for oral administration contain, for example, from about 0.1 mg to about 500 mg of active substance.
  • Topical administration is e.g. to the skin.
  • a further form of topical administration is to the eye.
  • the compounds of formula I may be administered in free form or in pharmaceutically acceptable salt form e.g. as indicated above.
  • Such salts may be prepared in conventional manner and exhibit the same order of activity as the free compounds.
  • the present invention further provides:
  • a method for preventing or treating disorders or diseases mediated by T lymphocytes and/or PKC, e.g. such as indicated above, in a subject in need of such treatment comprises administering to said subject an effective amount of a compound of formula I or a pharmaceutically acceptable salt thereof;
  • a method for preventing or treating acute or chronic transplant rejection or T-cell mediated inflammatory or autoimmune diseases, e.g. as indicated above, in a subject in need of such treatment comprises administering to said subject an effective amount of a compound of formula I or a pharmaceutically acceptable salt thereof;
  • a compound of formula I in free form or in a pharmaceutically acceptable salt form for use as a pharmaceutical, e.g. in any of the methods as indicated under 1.1 and 1.2 above.
  • a pharmaceutical composition e.g. for use in any of the methods as in 1.1 and 1.2 above comprising a compound of formula I in free form or pharmaceutically acceptable salt form in association with a pharmaceutically acceptable diluent or carrier therefor.
  • Compounds of formula I may be administered as the sole active ingredient or together with other drugs in immunomodulating regimens or other anti-inflammatory agents e.g. for the treatment or prevention of allo- or xenograft acute or chronic rejection or inflammatory or autoimmune disorders.
  • they may be used in combination with cyclosporines, or ascomycines or their immunosuppressive analogs or derivatives, e.g. cyclosporin A, cyclosporin G, FK-506, ABT-281, ASM 981; an mTOR inhibitor, e.g.
  • rapamycin 40-O-(2-hydroxy)ethyl-rapamycin etc.
  • corticosteroids cyclophosphamide
  • azathioprene methotrexate
  • an accelerating lymphocyte homing agent e.g. FTY 720
  • leflunomide or analogs thereof mizoribine
  • mycophenolic acid mycophenolate mofetil
  • 15-deoxysperguaiine or analogs thereof immunosuppressive monoclonal antibodies, e.g., monoclonal antibodies.
  • leukocyte receptors e.g., MHC, CD2, CD3, CD4, CD 11a/CD18, CD7, CD25, CD 27, B7, CD40, CD45, CD58, CD 137, ICOS, CD150 (SLAM), OX40, 4-1BB or their ligands, e.g. CD154; or other immunomodulatory compounds, e.g. a recombinant binding molecule having at least a portion of the extracellular domain of CTLA4 or a mutant thereof, e.g. an at least extracellular portion of CTLA4 or a mutant thereof joined to a non-CTLA4 protein sequence, e.g. CTLA4Ig (for ex. designated ATCC 68629) or a mutant thereof, e.g.
  • CTLA4Ig for ex. designated ATCC 68629
  • LEA29Y or other adhesion molecule inhibitors, e.g. mAbs or low molecular weight inhibitors including LFA-1 antagonists, Selectin antagonists and VLA-4 antagonists.
  • Compounds of formula I may also be administered together with an antiproliferative drug, e.g. a chemotherapeutic drug, e.g. in cancer treatment, or with an anti-diabetic drug in diabetes therapy.
  • a method as defined above comprising co-administration, e.g. concomitantly or in sequence, of a therapeutically effective amount of an inhibitor of PKC and of T-cell activation and proliferation, e.g. a compound of formula I in free form or in pharmaceutically acceptable salt form, and a second drug substance, said second drug substance being an immunosuppressant, immunomodulatory, anti-inflammatory, antiproliferative or anti-diabetic drug, e.g. as indicated above.
  • a therapeutic combination e.g. a kit, comprising a) an inhibitor of PKC and of T-cell activation and proliferation, e.g. a compound of formula I in free form or in pharma-ceutically acceptable salt form, and b) at least one second agent selected from an immunosuppressant, immunomodulatory, anti-inflammatory, antiproliferative and anti-diabetic drug.
  • Component a) and component b) may be used concomitantly or in sequence.
  • the kit may comprise instructions for its administration.
  • an inhibitor of PKC and of T-cell activation and proliferation e.g. a compound of formula I
  • other immunosuppressive/immunomodulatory, anti-inflammatory, antiproliferative or anti-diabetic therapy e.g. for preventing or treating acute or chronic graft rejection or inflammatory or autoimmune disorders as hereinabove specified
  • dosages of the co-administered immunosuppressant, immunomodulatory, anti-inflammatory, antiproliferative or anti-diabetic compound will of course vary depending on the type of co-drug employed, e.g. whether it is a steroid or a cyclosporine, on the specific drug employed, on the condition being treated and so forth.
  • a preferred compound according to the invention is e.g. the compound of Example 56.

Abstract

Indolylmaleimide derivatives comprising either a substituted phenyl, naphthyl, tetrahydronaphthyl, quinazolinyl, quinolyl, isoquinolyl or pyrimidinyl residue have interesting pharmaceutical properties, e.g. in the treatment and/or prevention of T-cell mediated acute or chronic inflammatory diseases or disorders, autoimmune diseases, graft rejection or cancer.

Description

  • This is a divisional of application Ser. No. 10/660,442 filed on Sep. 11, 2003, which is a continuation of application Ser. No. 10/007,368, filed on Nov. 5, 2001, which claims benefit of U.S. provisional Application No. 60/246,400, filed on Nov. 7, 2000, and U.S. provisonal Application No. 60/283,705, filed on Apr. 13, 2001, the entire disclosures of which are hereby incorporated by reference.
  • The present invention relates to indolylmaleimide derivatives, process for their production and pharmaceutical compositions containing them.
  • More particularly the present invention provides a compound of formula I
    Figure US20070191401A1-20070816-C00001
  • wherein
    • Ra is H; C1-4alkyl; or C1-4alkyl substituted by OH, NH2, NHC1-4alkyl or N(C1-4alkyl)2;
    • Rb is H; or C1-4alkyl;
    • R is a radical of formula (a), (b), (c), (d), (e) or (f)
      Figure US20070191401A1-20070816-C00002
  • wherein
    • each of R1, R4, R7, R8, R11 and R14 is OH; SH; a heterocyclic residue; NR16R17 wherein each of R16 and R17, independently, is H or C1-4alkyl or R16 and R17 form together with the nitrogen atom to which they are bound a heterocyclic residue; or a radical of formula α
      —X—Rc—Y  (α)
    • wherein X is a direct bond, O, S or NR18 wherein RIB is H or C1-4alkyl,
    • Rc is C1-4alkylene or C1-4alkylene wherein one CH2 is replaced by CRxRy wherein one of
    • Rx and Ry is H and the other is CH3, each of Rx and Ry is CH3 or Rx and Ry form together —CH2—CH2—, and
    • Y is bound to the terminal carbon atom and is selected from OH, a heterocyclic residue and —NR19R20 wherein each of R19 and R20 independently is H, C3-6cycloalkyl, C3-6cycloalkyl-C1-4alkyl, aryl-C1-4alkyl or C1-4alkyl optionally substituted on the terminal carbon atom by OH, or R19 and R20 form together with the nitrogen atom to which they are bound a heterocyclic residue;
    • each of R2, R3, R5, R6, R9, R10, R12, R13, R15 and R′15, independently, is H, halogen, C1-4alkyl, CF3, OH, SH, NH2, C1-4alkoxy, C1-4alkylthio, NHC1-4alkyl, N(C1-4alkyl)2 or CN;
    • either E is —N═ and G is —CH═ or E is —CH═ and G is —N═; and
    • ring A is optionally substituted.
  • Any alkyl or alkyl moiety in e.g. alkoxy may be linear or branched. Halogen may be F, Cl, Br or I, preferably F or Cl. Any aryl may be phenyl or naphthyl, preferably phenyl.
  • By heterocyclic residue as R1, R4, R7, R8, R11, R14 or Y or formed, respectively, by NR16R17 or NR19R20, is meant a three to eight, preferably five to eight, membered saturated, unsaturated or aromatic heterocyclic ring comprising 1 or 2 heteroatoms, preferably selected from N, O and S, and optionally substituted. Suitable examples include e.g. pyridyl, e.g. 3- or 4-pyridyl, piperidyl, e.g. piperidin-1-yl, 3- or 4-piperidyl, homopiperidyl, piperazinyl, homopiperazinyl, morpholin-4-yl, imidazolyl, imidazolidinyl, pyrrolyl or pyrrolidinyl, optionally substituted, e.g. mono- or polysubstituted. When the heterocyclic residue is substituted, this may be on one or more ring carbon atoms and/or on a ring nitrogen atom when present. Examples of a substituent on a ring carbon atom include e.g. C1-4alkyl e.g. CH3;
  • C3-6cycloalkyl e.g. cyclopropyl, optionally further substituted by C1-4alkyl;
    Figure US20070191401A1-20070816-C00003

    wherein p is 1, 2 or 3, preferably 1; CF3; halogen; OH; NH2; —CH2-NH2; —CH2—OH; piperidin-1-yl; or pyrrolidinyl. Examples of a substituent on a ring nitrogen atom are e.g. C1-6alkyl; acyl, e.g. R′x—CO wherein R′x is H, C1-6alkyl or phenyl optionally substituted by C1-4alkyl, C1-4alkoxy or amino, e.g formyl; C3-6cycloalkyl; C3-6cycloalkyl-C1-4alkyl; phenyl; phenyl-C1-4alkyl e.g. benzyl; a heterocyclic residue, e.g. as disclosed above, e.g. an aromatic heterocyclic residue comprising 1 or 2 nitrogen atoms; or a residue of formula β
    —R21—Y′  (β)
  • wherein R21 is C1-4alkylene or C2-4alkylene interrupted by O and Y′ is OH, NH2, NH(C1-4alkyl) or N(C1-4alkyl)2.
  • C2-4alkylene interrupted by O may be e.g. —CH2—CH2—O—CH2—CH2—.
  • When the substituent on a cyclic nitrogen is a heterocyclic residue, it may be a five or six membered saturated, unsaturated or aromatic heterocyclic ring comprising 1 or 2 heteroatoms, preferably selected from N, O and S. Examples include e.g. 3- or 4-pyridyl, piperidyl, e.g. piperidin-1-yl, 3- or 4-piperidyl, homopiperidyl, piperazinyl, homopiperazinyl, pyrimidinyl, morpholin-4-yl, imidazolyl, imidazolidinyl, pyrrolyl or pyrrolidinyl,
  • When Ra is substituted C1-4alkyl, the substituent is preferably on the terminal carbon atom.
  • When ring A is substituted, it may be mono- or polysubstituted, preferably monosubstituted, the substituent(s) being selected from the group consisting of e.g. halogen, OH, C1-4alkoxy, e.g. OCH3, C1-4alkyl, e.g. CH3, NO2, CF3, NH2, NHC1-4alkyl, N(C1-4alkyl)2 and CN. For example, ring A may be a residue of formula
    Figure US20070191401A1-20070816-C00004
  • wherein
    • Rd is H; C1-4alkyl; or halogen; and
    • Re is OH; NO2; NH2; NHC1-4alkyl; or N(C1-4alkyl)2.
  • Preferably Rd is in position 1; preferably Re is in position 3.
  • When Rc has a CH2 replaced by CRxRy, it is preferably the CH2 bearing Y.
  • Examples of heterocyclic residue as R1, R4, R7, R8, R11, R14 or Y or formed, respectively, by NR16R17 or NR19R20, include e.g. a residue of formula (γ)
    Figure US20070191401A1-20070816-C00005
  • wherein
    • the ring D is a 5, 6 or 7 membered saturated, unsaturated or aromatic ring;
    • Xb is —N—, —C═ or —H—;
    • Xc is —N═, —NRf—, —CRf′═ or —CHRf′— wherein Rf is a substituent as indicated above for a ring nitrogen atom, and Rf′ is a substituent as indicated above for a ring carbon atom;
    • the bond between C1 and C2 is either saturated or unsaturated;
    • each of C1 and C2, independently, is a carbon atom which is optionally substituted by one or two substituents selected among those indicated above for a ring carbon atom; and
    • the line between C3 and Xb and between C1 and Xb, respectively, represents the number of carbon atoms as required to obtain a 5, 6 or 7 membered ring D.
  • A preferred residue of formula (γ) is one wherein the ring P forms a 1,4-piperazinyl ring optionally C- and/or N-substituted as indicated.
  • Representative examples of a residue of formula (γ) are e.g. 3- or 4- pyridyl; piperidin-1-yl; 1—N-(C1-4alkyl)— or (co-hydroxy-C1-4alkyl)-3-piperidyl; morpholin-4-yl; imidazolyl; pyrrolidinyl; 1-piperazinyl; 2-C1-4alkyl- or -C3-6cycloalkyl-1-piperazinyl ;3-C1-4alkyl- or -C3-6cycloalkyl-1-piperazinyl; 2,2- or 3,5- or 2,5- or 2,6-di(C1-4alkyl)-1-piperazinyl; 3,4,5-tri-(C1-4alkyl)-1-piperazinyl; 4-N-(C1-4alkyl)- or -(ω-hydroxy-C1-4alkyl)- or -(ω-dimethylamino-C1-4alkyl)-1-piperazinyl; 4-N-pyridin-4-yl-1-piperazinyl; 4-N-phenyl- or —C3-6cycloalkyl-1-piperazinyl; 4-N-(C1-4alkyl)- or -(ω-hydroxy-C1-4alkyl)-3-C1-4alkyl- or -3,3-di(C1-4alkyl)-1-piperazinyl; 4-N-(1-C1-4alkyl-C3-6cycloalkyl)-1-piperazinyl; 4-N-formyl-1-piperazinyl; 4-N-pyrimidin-2-yl-1-piperazinyl; or 4-N-C1-4alkyl-1-homopiperazinyl.
  • The compounds of formula I may exist in free form or in salt form, e.g. addition salts with e.g. organic or inorganic acids, for example, hydrochloric acid, acetic acid, when R1, R4, R7, R8, R11 or R14 and/or R2, R3, R5, R6, R9, R10, R12, R13 or R15 comprises an optionally substituted amino group or a heterocyclic residue which can form acid addition salts.
  • It will be appreciated that the compounds of formula I may exist in the form of optical isomers, racemates or diastereoisomers. For example, a ring carbon atom bearing a substituent in the heterocyclic residue as R1, R4, R7, R8, R11, R14 or Y or formed, respectively, by NR16R17 or NR19R20, is asymmetric and may have the D- or L-configuration. It is to be understood that the present invention embraces all enantiomers and their mixtures. Similar considerations apply in relation to starting materials exhibiting asymetric carbon atoms as mentioned.
  • In the compounds of formula I, the following significances are preferred individually or in any sub-combination:
    • 1. Ra is H or CH3;
    • 2. Rb is H;
    • 3. Ring A is unsubstituted; or is substituted by methyl in position 7;
    • 4. Preferred heterocyclic residue as formed by NR16R17 is e.g. piperazin-1-yl optionally N-substituted, e.g. by C1-4alkyl, ω-hydroxy-C1-4alkyl, ω-dimethylamino-C1-4alkyl, C5-6cycloalkyl, C1-4alkyl-C5-6cycloalkyl, an aromatic heterocyclic residue comprising 1 or 2 nitrogen atoms, e.g. pyridyl or pyrimidin-2-yl, or a residue of formula β as defined above and/or optionally C-substituted, e.g. by CH3 e.g. in positions 2, and/or 3 and/or 5 and/or 6 and/or 2, 2 or 3, 3 or by
      Figure US20070191401A1-20070816-C00006

      e.g. in position 2 or 3; piperidin-1-yl optionally C-substituted, e.g. in position 4, by NH2, —CH2—NH2 or piperidin-1-yl, or in position 3, e.g. by OH or NH2; or pyrrolidinyl optionally C-substituted in position 3 by OH or NH2;
    • 5. R18 is H or CH3;
    • 6. Rc is C1-4alkylene or C1-4alkylene wherein the terminal CH2 is replaced by CRxRy wherein Rx and Ry form together —CH2—CH2—;
    • 7. X is O;
    • 8. The radical of formula (α) is —O—CH2—CH2—Y;
    • 9. Each of R19 and R20 is H, C1-4alkyl, e.g. methyl, C1-4alkyl substituted on the terminal carbon atom by OH, e.g. —CH2—CH2—OH, or cyclopropyl;
    • 10. Preferred heterocyclic residue as formed by NR19R20 is e.g. piperazin-1-yl optionally N-substituted by C1-4alkyl or a residue of formula β; piperidin-1-yl; 1-(C1-4alkyl)-piperidin-3-yl; 3- or 4-pyridyl; imidazolyl; pyrrolidinyl; or morpholin-4-yl;
    • 11. Each of R1, R4, R7, R8, R11 or R14, independently, is 1-N-methyl-piperidin-4-yl; 4-methyl-piperazin-1-yl; 4-methyl-1-homopiperazinyl; 4-(2-hydroxyethyl)-piperazin-1-yl; or —X′—C1, 2 or 3-alkylene-NR19R20 wherein X′ is a direct bond, O or NH;
    • 12. In the residue of formula (a) either each of R2 and R3 is H or one of R2 and R3 is H and the other is F, Cl, CH3, OH, OCH3 or CF3;
    • 13. In the residue of formula (a) R2 is OH;
    • 14. In the residue of formula (b) either each of R5 and R6 is H or one of R5 and R6 is H and the other is F, Cl, CH3, OCH3 or CF3;
    • 15. In the residue of formula (b) R4 is a radical of formula (α) or NR16R17;
    • 16. In the residue of formula (d) either each of R9 and R10 is H or one of R9 and R10 is H and the other is F, Cl, CH3, OCH3 or CF3; preferably R10 is H and R9 is in position 5, 6, 7 or 8, preferably in position 6;
    • 17. In the residue of formula (e) each of R12 and R13 is H;
    • 18. In the residue of formula (e) one of R12 and R13 is H and the other is F, Cl, CH3, OCH3 or CF3;
      • when E is —N═ and G is —CH═, preferably R13 is H and R12 is in position 6 or 7;
      • when E is —CH═ and G is —N═, preferably R13 is H and R12 is in position 7;
    • 19. In the residue of formula (f) R15 is H, CH3 or Cl, e.g. in position 5 or 6;
    • 20. In the residue of formula (f) R′15 is H or CH3, e.g. in position 5, preferably H;
    • 21. R is a radical of formula (d), (e) or (f).
  • The present invention also includes a process for the preparation of a compound of formula I which process comprises
  • a) reacting a compound of formula II
    Figure US20070191401A1-20070816-C00007
    •  wherein Ra, Rb and ring A are as defined above,
    •  with a compound of formula III
      R—CH2—CO—NH2  (III)
    •  wherein R is as defined above,
  • b) reacting a compound of formula IV
    Figure US20070191401A1-20070816-C00008
    •  wherein Ra, Rb and ring A are as defined above,
    •  with a compound of formula V
      R—CO—CO—OCH3  (V)
    •  wherein R is as defined above; or
  • c) converting in a compound of formula I a substituent R1, R4, R7, R8, R11 or R14 into another substituent R1, R4, R7, R8, R11 or R14
  • and, where required, converting the resulting compound of formula I obtained in free form to a salt form or vice versa, as appropriate.
  • Process steps a) and (b) may conveniently be effected in the presence of a strong base, e.g. t-BuOK. When compounds of formula III or V comprising an OH group which should not participate to the reaction are used, such OH group is in protected form. The OH-protecting group may be removed according to methods known in the art at the end of condensation step a) or b). Process step c) may be carried out according to known methods: for example when R1, R4, R7, R8, R11 or R14 comprises a final OH group, this OH group may be replaced by the desired —NR16R17 or —NR19R20.
  • Compounds of formula 11 may be prepared by reacting the corresponding indol compound with an oxalyl halogenide, e.g. chloride, or with a monoalkyl oxalyl chloride under basic conditions, e.g. as disclosed in Example 28.
  • Compounds of formula III or V, used as starting materials, may be prepared in accordance with known methods, e.g. by introducing the desired substituent R1, R4, R7, R8, R11 or R14, respectively, in a compound of formula III′ or V′
    R′—CH2—CO—NH2  (III′)
    R″—CO—CO—OCH3  (V′)
    wherein each of R′ or R″ is respectively a radical of formula (a), (b), (c), (d), (e) or (f), each of which comprising a leaving group, e.g. halogen, in place of R1, R4, R7, R8, R11 or R14.
  • Alternatively, compounds of formula III wherein R is a radical of formula (a), (b) or (c), R1, R4 or R7 being a radical of formula (α), may be prepared in accordance with known methods by reacting a compound of formula III′ wherein R′ is respectively a radical of formula (a), (b) or (c), each of which comprising OH in place of R1, R4 or R7, with a compound of formula Xa—X—Rc—Y wherein Xa is a leaving group, e.g. Cl, and X, Rc or Y are as defined above.
  • Compounds of formula I wherein R is a radical of formula (e) wherein E is —N═, G is —CH═ and R11 is —O—Rc—Y or —S—Rc—Y may also be prepared by reacting together a compound of formula II as defined above with a compound of formula III′ wherein R′ is a radical of formula (e′)
    Figure US20070191401A1-20070816-C00009
  • wherein R12 and R13 are as defined above and Xa is a leaving group, e.g. halogen, and with a compound of formula VI
    R′11H  (VI)
  • wherein R′11 is —O—Rc—Y or —S—Rc—Y. This reaction may be carded out in accordance with know methods, e.g. as disclosed in Example 28 below.
  • Compounds of formula I wherein R is a radical of formula (d) or (f) wherein R8 or R14 is —O—Rc—Y or —S—Rc—Y may also be prepared by reacting together a compound of formula II as defined above with a compound of formula III′ wherein R″ is a radical of formula (d′) or (f′)
    Figure US20070191401A1-20070816-C00010
  • wherein R9, R10, R15 and R′15 are as defined above and Xa is a leaving group, e.g. halogen, and with a compound of formula VI′
    A—H  (VI)
  • wherein A is —O—Rc—Y or —S—Rc—Y. This reaction may be carried out in accordance with know methods.
  • Insofar as the production of the starting materials is not particularly described, the compounds are known or may be prepared analogously to methods known in the art or as described hereafter.
  • The following Examples are illustrative of the invention.
    • RT=room temperature
    • THF=tetrahydrofuran
    • FCC=flash column chromatography
    • TBAF=tetrabutyl ammonium fluoride
    • BINAP=2,2′-bis(diphenylphosphino)-1,1′-binaphthyl
    EXAMPLE 1 3-(1.H.-Indol-3-yl)4-[3-(2-dimethylamino-ethoxy)-5-hydroxy-phenyl]-pyrrole-2,5-dione
  • Figure US20070191401A1-20070816-C00011
  • A solution of 400 mg (0.58 mmol) of 3-(1.H.-indol-3-yl)-4-[3-(2-methanesulfonyloxy-ethoxy)-5-triphenylmethoxy-phenyl]-pyrrole-2,5-dione in 5 mL of 33% dimethylamine in ethanol is stirred overnight at RT. The reaction mixture is diluted with ethyl acetate. The resulting mixture is washed with saturated aqueous sodium bicarbonate. The layers are separated and the aqueous layer is extracted with three portions of ethyl acetate. The combined organic solution is washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue is filtered through a plug of silica gel (70:30 ethyl acetate/methanol) to afford 3-(1.H.-Indol-3-yl)-4-[3-(2-dimethylamino-ethoxy)-5-triphenylmethoxy-phenyl]-pyrrole-2,5-dione, which is immediately. used in the next step without further purification.
  • To a solution of 370 mg (0.58 mmol) of 3-(1.H.-Indol-3-yl)4-[3-(2-dimethylamino-ethoxy)-5-triphenylmethoxy-phenyl]-pyrrole-2,5-dione in 5 mL of methanol is added 251 mg (1.46 mmol) of para-toluenesulfonic acid. After stirring for 2 h at room temperature, the mixture is diluted with ethyl acetate and washed with saturated aqueous sodium bicarbonate. The aqueous layer is extracted with three portions of ethyl acetate. The combined organic layers are washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue is purified by column chromatography on silica gel (7:3 ethyl acetate/methanol) to afford the title compound as an orange foam.
  • 1H NMR (DMSO-d6, 400 MHz) δ 11.89 (s, 1H), 11.00 (s, 1H), 9.45 (s, 1H), 7.98 (s, 1H), 7.43 (d, J=8.0 Hz, 1H), 7.08 (t, J=7.4 Hz, 1H), 6.78 (t, J=7.4 Hz, 1H), 6.50 (m, 2H), 6.34 (s, 1H), 6.30 (s, 1H), 3.69 (t, J=5.9 Hz, 2H), 2.35 (t, J=5.9 Hz, 2H), 2.06 (s, 6H); MS.(EI, negative ionization) m/z 390 [M−H], (EI, positive ionization) m/z 392 [M+H]+
  • 3-(1.H.-Indol-3-yl)4-[3-(2-methanesulfonyloxy-ethoxy)-5-hydroxy-phenyl]-pyrrole-2,5-dione, used as starting material, may be prepared as follows:
  • a) [3-(2-Triisopropylsilyloxy-ethoxy)-5-hydroxy-phenyl]-acetic acid methyl ester
  • A mixture of 9.39 g (51.5 mmol) of (3,5-dihydroxy-phenyl)-acetic acid methyl ester (prepared according to U. Eder, G. Sauer, G. Haffer, G. Neef, R. Wiechert, U.S. Pat. No. 4,066,674), 11.38 g (61.8 mmol) of 1-bromo-2-triisopropylsilyloxy-ethane and 14.50 g (51.5 mmol) of cesium carbonate is stirred at RT for 1 hour and at 60° C. for another hour. The reaction mixture is then treated with saturated aqueous sodium carbonate and extracted with ethyl acetate. The layers. are separated and the organic layer is washed three times with saturated aqueous sodium carbonate. The aqueous layers are combined and extracted three times with ethyl acetate. The combined organic solutions are then washed with saturated brine, dried, filtered and concentrated under reduced pressure. The residue is purified by column chromatography on silica gel (85:15 hexane/ethyl acetate then 70:30 hexane/ethyl acetate and finally pure ethyl acetate) to afford above title compound as a yellow oil.
  • MS (EI, negative ionization) m/z 381 [M−H], (EI, positive ionization) m/z 405 [M+Na]+
  • b) 2-[3-(2-Triisopropylsilyloxy-ethoxy)-5-hydroxy-phenyl]-acetamide
  • A mixture of 3.9 g (10.2 mmol) of the compound of step a) and 40 mL of concentrated aqueous ammonia is stirred at RT for 2 days and the solvents are removed under reduced pressure. The residue is dissolved in ethyl acetate and filtered through a plug of silica gel. The filtrate is reduced under reduced pressure. The residue is dissolved in a minimum of ethyl acetate and n-hexane is added upon which the desired product crystallized, affording above title compound after filtration and drying.
  • MS (EI, negative ionization) m/z 366 [M−H], (EI, positive ionization), m/z 390 [M+Na]+
  • c) 2-[3-(2-Triisopropylsilyloxy-ethoxy)-5-triphenylmethoxy-phenyl]-acetamide
  • A solution of 1.6 g (4.38 mmol) of compound b), 3.7 g (13.27 mmol) of triphenyl chloromethane, 3.7 mL (26.69 mmol) of triethylamine and 535 mg (4.38 mmol) of dimethylaminopyridine in 50 mL of dichloromethane is stirred at room temperature for 2 hours. Saturated aqueous sodium bicarbonate is added and the mixture is extracted with three portions of ethyl acetate. The combined organic layers are washed twice with saturated aqueous sodium bicarbonate and once with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue is purified by column chromatography on silica gel (1:2 n-hexane/ethyl acetate followed by 100% ethyl acetate) to afford the title compound c) as a white foam. 1H NMR (DMSO-d6, 400 MHz) δ 7.46-7.20 (m, 16H), 6.81 (s, 1H), 6.38 (s, 1H), 6.28 (s, 1H), 5.92 (s, 1H), 3.81 (dd, J=4.5, 4.7 Hz, 2H), 3.68 (dd, J=4.5, 4.7 Hz, 2H), 3.13 (s, 2H), 1.11-0.91 (m, 21H); MS (EI, negative ionization) m/z 608 [M−H], (EI, positive ionization) m/z 632 [M+Na]+
  • d) 3-(1.H.-Indol-3-yl)-4-[3-(2-triisopropylsilyloxy-ethoxy)-5-triphenylmethoxy-phenyl]-pyrrole-2,5-dione
  • To a stirred solution of 2.5 g (4.12 mmol) of compound c) and 1.3 g (6.40 mmol) of (1.H.-indol-3-yl)-oxo-acetic acid methyl ester in 18 mL of THF is added 20.6 mL (20.6 mmol) of a 1M solution of t-BuOK in THF at room temperature. The reaction mixture is heated to 60° C. for 45 minutes and then allowed to cool to RT. Saturated aqueous sodium bicarbonate is added and the resulting mixture is diluted with ethyl acetate. The layers are separated. The aqueous layer is extracted three times with ethyl acetate. The combined organic solutions are washed twice with saturated aqueous sodium bicarbonate and once with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue is purified by column chromatography on silica gel (2:1 n-hexane/ethyl acetate followed by 100% ethyl acetate) to afford the title compound d) as an orange foam. 1H NMR (DMSO-d6, 400 MHz) δ 11.90 (s,1H), 10.95 (s,1H), 7.93 (s, 1H), 7.43 (d, J=,8.0 Hz, 1H), 7.37-7.20 (m, 15H), 7.11 (dd, J=7.4, 7.6 Hz, 1H), 6.74 (t, J=7.6 Hz, 1H), 6.56 (s, 1H), 6.33 (s, 1H), 6.16 (d, J=8.0 Hz, 1H), 6.07 (t, J=2.1 Hz, 1H), 3.65 (dd, J=4.1, 5.1 Hz, 2H), 3.39 (m, 2H), 1.04-0.87 (m, 21H); MS (EI, negative ionization) m/z 761 [M−H], 518 [M−Ph3C], (EI, positive ionization) m/z 785 [M +Na]+
  • e) 3-(1.H.-Indol-3-yl)-4-[3-(2-hydroxy-ethoxy)-5-triphenylmethoxy-phenyl]-pyrrole-2,5-dione
  • To a stirred, cooled (0° C.) solution of 1.8 compound d) in 15 mL of THF is added 7.1 mL (7.1 mmol) of a 1M solution of TBAF in THF. After 45 minutes, saturated aqueous sodium bicarbonate is added, and the resulting mixture is diluted with ethyl acetate. The layers are separated. The aqueous layer is extracted with three portions of ethyl acetate. The combined organic layers are washed twice with saturated aqueous sodium bicarbonate and once with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue is purified by column chromatography on silica gel (1:1 n-hexane/ethyl acetate) to afford the title compound e) as an orange foam.
  • 1H NMR (DMSO-d6, 400 MHz) δ 11.89 (s, 1H), 10.95(s, 1H), 7.91 (d, J=2.1 Hz, 1H), 7.45 (d, J=8.0 Hz, 1H), 7.40-7.20 (m, 15H), 7.13 (dd, J=7.4,,7.6 Hz, 1H), 6.77 (dd, J=7.2, 7.8 Hz, 1H), 6.51 (s, 1H), 6.34 (s, 1H), 6.20 (d, J=8.0 Hz, 1H), 6.08 (t, J=2.1 Hz, 1H), 4.66 (s,1H), 3.37 (s, 4H); MS (EI, negative ionization) m/z 605 [M−H], 362 [M−Ph3C], (EI, positive ionization) m/z 629 [M+Na]+, 645 [M+K]+
  • f) 3-(1.H.-Indol-3-yl)-4-[3-(2-methanesulfonyloxy-ethoxy)-5-triphenylmethoxy-phenyl]-pyrrole-2,5-dione
  • A mixture of compound e), 1.0 g (5.74 mmol) of methanesulfonic anhydride and 0.76 mL (9.42 mmol) of pyridine in 20 mL of THF is stirred at room temperature for 1 hour. Saturated aqueous sodium bicarbonate is added, and the resulting mixture is diluted with ethyl acetate. The layers are separated. The aqueous layer is extracted with three portions of ethyl acetate. The combined organic layers are washed twice with saturated aqueous sodium bicarbonate and once with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure. The residue is purified by column chromatography on silica gel (1:2 n-hexane/ethyl acetate followed by 100% ethyl acetate) to afford the title compound f) as an orange oil.
  • The compounds of formula X1
    Figure US20070191401A1-20070816-C00012
  • wherein R1 and R2 are as defined in Table 1, may be prepared by following the procedure of Example 1 but using the appropriate starting materials. Starting materials which do not comprise a OH substituent will be prepared without the protecting steps as indicated in Example 1.
    TABLE 1
    Example R1 R2 R3 Ra M.S. Data
    2 OH H H H M+ 304
    3 —O—(CH2)3—N(CH3)2 H H H MH+ 390
    4 —O—CH2-4-pyridyl H H H MH+ 396
    5 —O—CH2-3-pyridyl H H H MH+ 396
    6 —O—CH2—CH2—OH H H H M+ 348
    7 —O—CH2—CH2-piperidin-1-yl H H H MH+ 416
    8 —O—CH2—CH2-(4-methyl-piperazin-1- H H H MH+ 431
    yl)
    9 —O—CH2—CH2-(morpholin-4-yl) H H H MH+ 418
    10 —O—CH2—CH2-[4-(2-hydroxyethyl)- H H H MH+ 461
    piperazin-1-yl]
    11 —O—CH2—CH2-(imidazol-1-yl) H H H MH+ 399
    12 —O—CH2—CH2—N(CH3)2 H H H MH+ 376
    13
    Figure US20070191401A1-20070816-C00013
    H H H MH+ 406
    14
    Figure US20070191401A1-20070816-C00014
    H H H MH+ 482
    15 —O—CH2—CH2—N(CH2—CH2—OH)2 H H H MH+ 436
    16 —O—CH2—CH2—(pyrrolidin-1-yl) H H H MH+ 402
    17 —O—CH2-(1-methyl-piperidin-3-yl) H H H MH+ 416
    18 —CH2—N(CH3)2 H H H MH+ 346
    19 —O—CH2—CH2—N(CH3)2 —O—CH2—CH3 H H M+ 419
    20 —O—CH2—CH2—N(CH3)2 —O—CH3 H H MH+ 406
    21 -(4-methyl-piperazin-1-yl) H —O—CH3 H (M + H)+ 417
    22 -(4-methyl-piperazin-1-yl) H H H (M + H)+ 387
    23 -(4-methyl-piperazin-1-yl) H H CH3 (M + H)+ 401
    24 -(4-methyl-piperazin-1-yl) H CH3 H (M + H)+ 401
    25 -(4-methyl-piperazin-1-yl) H CH3 CH3 (M + H)+ 415
    26 -(4-methyl-piperazin-1-yl) H Cl H (M + H)+ 421
    27 -(4-methyl-piperazin-1-yl) H Cl CH3 (M + H)+ 435
  • EXAMPLE 28 3-(1.H.-Indol-3-yl)-4-[3-4-(2-dimethylamino-ethoxy)-naphthalen-1-yl]-pyrrole-2,5-dione
  • Figure US20070191401A1-20070816-C00015
  • A suspension of 2.10 g (4.41 mmol) of 3-(1.H.-indol-3-yl)4-[3-(2-methanesulfonyloxy-ethoxy)-naphthalen-1-yl]-pyrrole-2,5-dione in 20 mL of a 33% solution of dimethylamine in ethanol is stirred at room temperature overnight. The solvents are removed under reduced pressure. The product is crystallized from 1:1 acetonitrile/water, filtered and washed with 1:1 acetonitrile/water, diethlylether and n-hexane. This procedure affords the title compound as a red-orange crystalline solid.
  • 1H NMR (DMSO-d6, 400 MHz) δ 11.84 (s, 1H), 11.13 (s, 1H), 7.97 (s, 1H), 7.84 (d, J=8.2 Hz, 1H), 7.62 (d, J=8.4 Hz, 1H), 7.47 (s, 1H), 7.39 (t, J=7.4 Hz, 1H), 7.31 (d, J=8.2 Hz, 1H), 7.16 (t, J=7.4 Hz, 1H), 7.12 (s, 1H), 6.92 (dd, J=7.4, 7.6 Hz, 1H), 6.46 (dd, J=7.4, 7.6 Hz, 1H), 6.25 (d, J=8.2 Hz, 1H), 4.16 (m, 2H), 2.63 (dd, J=5.5, 5.7 Hz, 2H), 2.20 (s, 6H); 13C NMR (DMSO-d6, 100 MHz) δ 173.3, 173.2, 156.3, 137.1, 136.6, 135.3, 132.2, 131.6, 128.6, 128.1, 127.7, 127.3, 126.4, 125.6, 124.7, 122.8, 121.9, 121.3, 120.7, 112.9, 109.2, 105.9, 66.8, 58.4, 46.4; IR (KBr) 3244,1698, 1629, 1597, 1220, 1039.
  • 3-Indol-3-yl)-4-[3-(2-methanesulfonyloxy-ethoxy)-naphthalen-1-yl]-pyrrole-2,5-dione, used as starting materials may be prepared as follows:
  • a) 1-Bromo-3-(2-triisopropylsilyloxy-ethoxy)-naphtalene
  • A stirred mixture of 4.38 g (19.6 mmol) of 1-bromo-naphtalen-3-ol (prepared according to the procedure of M. S. Newman, V. Sankaran, D. R. Olson, J. Am. Chem. Soc. 1976, 98, 3237-3242), 5.52 g (19.6 mmol) of 1-bromo-2-triisopropylsilyloxy-ethane, 13.56 g (98.1 mmol) of potassium carbonate and 1.45 g (3.9 mmol) of tetrabutylammonium iodide in 50 mL of dimethylformamide is heated to 60° C. for 4 hours. Then an additional 0.55 g (2.0 mmol) of 1-bromo-2-triisopropylsilyloxy-ethane are added and stirring is continued for another hour at 60° C., after which TLC analysis indicated complete consumption of 1-bromo-naphtalen-3-ol. The mixture is allowed to cool to room temperature and brine is added. The resulting solution is extracted with ethyl acetate. The organic solution is washed twice with brine and the combined aqueous layers are back extracted with ethyl acetate. The organic layers are combined, dried, filtered and concentrated under reduced pressure. The oily brown residue is purified by column chromatography on silica gel (97.5:2.5 n-hexane/diethylether) to afford above title compound as a brown solid.
  • b) Oxo-[3-(2-triisopropylsilyloxy-ethoxy)-naphthalen-1-yl]-ace tic acid methyl ester
  • To a stirred, cooled (−78° C.) solution of 1.59 g the compound of step a) in 15 mL of THF is added 2.6 mL (4.13 mmol) of a 1.6 M solution of n-BuLi in n-hexane. The resulting mixture is stirred for 1 h and a solution of 886 mg (7.50 mmol) of dimethyloxalate in 5 mL of THF is. added dropwise. After stirring the reaction mixture for 30 minutes at −78° C., it is warmed to 0° C., and stirring is continued at that temperature for 3 hours. Thereafter an additional 132 mg (1.12 mmol) of dimethyloxalate in 1 mL of THF is added. Stirring is continued for 1 h at 0° C., and the reaction is quenched by the addition of saturated aqueous ammonium chloride. Ethyl acetate is added and the layers are separated. The organic layer is washed twice with saturated brine. The aqueous layers are combined and extracted one more time with ethyl acetate. The combined organic solutions are dried, filtered and concentrated under reduced pressure. The residue is purified by column chromatography on silica gel (95:5 n-hexane/diethylether) to afford above title compound as a yellow oil.
  • c) 3-(1.H.-Indol-3-yl)4-[3-(2-triisopropylsilanyloxy-ethoxy)-naphthalen-1-yl]-pyrrole-2,5-dione
  • To a solution of 1.070 g the compound of step b) and 0.436 9 (2.50 mmol) of 2-(1.H.-Indol-3-yl)-acetamide in 10 mL of THF is added at RT 12.5 mL (12.5 mmol) of a 1M solution of t-BuOK in THF. After the addition is complete the mixture is heated to 60° C. for 4 hours and then allowed to cool to room temperature. Saturated aqueous sodium bicarbonate is added and the resulting mixture is extracted with ethyl acetate. The organic layer is washed twice with saturated brine. The aqueous layers are combined and extracted with ethyl acetate. The combined organic extracts are dried, filtered and concentrated under reduced pressure. The residue is purified by column chromatography on silica gel (60:40 n-hexane/ethyl acetate) to afford above title compound as a red-orange solid.
  • 1H NMR (DMSO-d6, 400 MHz) δ 11.85 (s, 1H), 11.15 (s, 1H), 7.98 (s, 1H), 7.83 (d, J=8.2 Hz, 1H), 7.60 (d, J=8.4 Hz, 1H), 7.47 (s, 1H), 7.39 (dd, J=7.4, 7.6 Hz, 1H), 7.31 (d, J=8.0 Hz, 1H), 7.15 (m, 2H),6.92 (dd, J=7.4, 7.6Hz, 1H), 6.44 (dd, J=7.4,7.8Hz, 1H), 6.26 (d, J=8.0 Hz, 1H), 4.18 (m, 2H), 4.03 (dd, J=4.3, 4.5 Hz, 2H), 1.16- 0.92 (m, 21 H); IR (KBr) 3346, 1710 cm−1; MS (EI, negative ionization) m/z 553 [M−H], (EI, positive ionization) m/z 574 [2M+K+H]2+, 577 [M+Na]+
  • d) 3-(1.H.-Indol-3-yl)-4-[3-(2-hydroxy-ethoxy)-naphthalen-1-yl]-pyrrole-2,5-dione
  • To a stirred, cooled (0° C.) solution of 807 mg (1.45 mmol) of the compound of step c) in 10 mL of THF is added 4.4 mL (4.40 mmol) of a 1 M solution of TBAF in THF. After 1 hour, saturated aqueous sodium bicarbonate is added, and the resulting mixture is extracted with ethyl acetate. The organic layer is washed twice with saturated brine. The aqueous layers are combined and extracted with ethyl acetate. The combined ethyl acetate solutions are dried, filtered and concentrated under reduced pressure. The residue is purified by column chromatography on silica gel (ethyl acetate) to afford above title compound as a red-orange solid.
  • 1H NMR (DMSO-d6, 400 MHz) δ 11.85 (s, 1H), 11.15 (s, 1H), 7.97 (s, 1H), 7.84 (d, J=8.2 Hz, 1H), 7.60 (d, J=8.4 Hz, 1H), 7.45 (d, J=1.8 Hz, 1H), 7.38 (dd, J=7.2, 7.6 Hz, 1H), 7.31 (d, J=8.0 Hz, 1H), 7.15 (m, 2H), 6.92 (dd, J=7.4, 7.6 Hz, 1H), 6.47 (dd, J=7.4, 7.8 Hz, 1H), 6.29 (d, J=8.2 Hz, 1H), 4.92 (t, J=5.5 Hz, 1H), 4.11 (m, 2H), 3.76 (m, 2H); IR,(KBr) 1705 cm−1; MS (EI, negative ionization) m/z 397 [M−H], (EI, positive ionization) m/z 418 [2M+K+H]2+, 421 [M+Na]+
  • e) 3-(1.H.-Indol-3-yl)-4-[3-(2-methanesulfonyloxy-ethoxy)-naphthalen-1-yl]-pyrrole-2,5-dione
  • A mixture of 1.99 g (5.00 mmol) of the compound of step d), 2.18 g (12.50 mmol) of methanesulfonic anhydride and 1.6 mL (19.80 mmol) of pyridine in 25 mL of THF is heated to 60° C. for 1 hour. The reaction mixture is allowed to cool to RT and is filtered. The filtrate is concentrated under reduced pressure. The product is then crystallized from diethylether, filtered and washed with one portion of diethylether, two portions of water and another portion of diethylether. This procedure affords above title compound as red-orange crystals.
  • 1H NMR (DMSO-d6, 400 MHz) δ 11.83 (s, 1H), 11.14 (s, 1H), 7.96,(d, J=2.5 Hz, 1H), 7.84 (d, J=8.2 Hz, 1H), 7.61 (d, J=8.2 Hz, 1H), 7.50 (d, J=2.1 Hz, 1H), 7.40 (dd, J=7.4, 7.6 Hz, 1H),7.30 (d, J=8.2Hz, 1H), 7.16 (m, 2H), 6.92 (dd, J=7.4, 7.8.Hz, 1H), 6.46 (t, J=7.6 Hz, 1H), 6.28 (d, J=8.2 Hz, 1H), 4.57 (dd, J=3.9, 4.1 Hz, 2H), 4.39 (m, .2H), 3.21 (s, 3H); MS (EI, negative ionization) m/z 475 [M−H], (EI, positive ionization) m/z 499 [M+Na]+
  • The compounds of formula X2
    Figure US20070191401A1-20070816-C00016
  • wherein R3 is as defined in Table 2, may be prepared by following the procedure of Example 28 but using the appropriate starting materials.
    TABLE 2
    Ex. R3 R M.S. Data
    29 —O—CH2—CH2-(pyrrolidin-1- H MH+ 452
    yl)
    30
    Figure US20070191401A1-20070816-C00017
    H MH+ 456
    31 —O—CH3 H (M − H) 367
    32 OH H (M − H) 353
    33 —O—(CH2)3—N(CH3)2 H MH+ 440
    34 —O—(CH2)2—N(CH3)- H MH+ 452
    cyclopropyl
    35 —O—(CH2)2—N(CH3)- CH3 MH+ 466
    cyclopropyl
    36 —O—(CH2)2—NH-cyclopropyl CH3 (M + H)+ 452
    37
    Figure US20070191401A1-20070816-C00018
    H (M + H)+ 438.2 (M − H) 436.3
    38
    Figure US20070191401A1-20070816-C00019
    CH3 (M + H)+ 452.3
    39 (4-methyl-piperazin-1-yl) H (M + H)+ 437
    (M − H) 435
    40 (4-methyl-piperazin-1-yl CH3 (M + H)+ 451
    41 1-piperazinyl H (M + H)+ 423
    (M − H) 421
    42 1-piperazinyl CH3 (M + H)+ 437
    (M − H) 435
    43 -(4-formyl-piperazin-1-yl) CH3 (M + H)+ 465
    (M + Na)+ 487
    44 O—(CH2)2-(piperidin-1-yl) H (M + H)+ 466
    45 O—(CH2)2—N(CH3)(n-Bu) H (M + H)+ 468
    46 O—(CH2)2—NH-cyclohexyl H (M + H)+ 480
    47 O—(CH2)2-(4-methyl- H (M + H)+ 480
    piperidin-1-yl)
    48 O—(CH2)2-(rac-2-methyl- H (M + H)+ 466
    pyrrolidin-1-yl)
    49 O—(CH2)2-(4-hydroxy- H (M + H)+ 482
    piperidin-1-yl)
    50 O—(CH2)2-((S)-2- H (M + H)+ 482
    hydroxymethyl-pyrrolidin-1-yl)
    51 O—(CH2)2-[4-(piperidin-1- H (M + H)+ 549
    yl)-piperidin-1-yl]
    52 O—(CH2)2-(rac-3-hydroxy- H (M + H)+ 482
    piperidin-1-yl)
  • The compounds of Examples 48 and 52 which are racemates may also be prepared in the form of pure cis or trans enantiomers using the corresponding cis or trans starting materials. The same applies to the cis isomer of Example 50: it may also be prepared as a racemate or in pure trans form.
  • EXAMPLE 53 3-[3-(2-hydroxy-ethoxy)-5,6,7,8-tetrahydro-naphthalen-1-yl]-4-(1.H.-indol-3-yl)-pyrrole-2,5-dione
  • Figure US20070191401A1-20070816-C00020
  • 3-[3-(2-t.butoxy-ethoxy)-5,6,7,8-tetrahydro-naphthalen-1-yl]4-(1. H.-indol-3-yl)-pyrrole-2,5-dione is treated with TFA/H2O (5 ml, 95/5) at RT for 15 min. The reaction mixture is poured into ice-cold saturated aqueous NaHCO3 solution and the resulting suspension is extracted with ethyl acetate. Pure title compound is obtained as orange powder after purification on silica gel using cyclo-hexane/ethyl acetate (2/1) as mobile phase. MH+: 403 (ES+)
  • 1H NMR (400 MHz, DMSO-d6) δ 11.84 (bs,1H), 10.96 (s, 1H), 7.95 (d, 1H, J=1.47 Hz), 7.37 (d, 1H, J=8.32 Hz), 7.03 (t, 1H, J=7.34Hz), 6.72 (d, 1H, J=2.44 Hz), 6.67 (t, 1H, J=7.33 Hz), 6.62 (d, 1H, J=2.69 Hz), 6.45 (d, 1H, J=8.07 Hz), 4.77 (t, 1H, J=5.38 Hz), 3.87 (m, 2H), 3.62 (q, 2H, J=5.38 Hz), 2.67 (m, 2H), 2.34 (m, 1H), 2.06 (m, 1H), 1.59 (m, 1H), 1.49 (m, 1H), 1.36 (m, 2H)
  • 3-[3-(2-t.butoxy-ethoxy)-5,6,7,8-tetrahydro-naphthalen-1-yl]4-(I.H.-indol-3-yl)-pyrrole-2,5-dione, used as starting materials, is prepared as follows:
  • a) 8-bromo-6-acetyltetralin
  • To AlCl3 (9.9 g, 75 mmol) is added dropwise 6-acetyltetralin (5.25 g, 30 mmol) under mechanical stirring at RT. After 20 minutes at 70° C. the reaction is cooled to RT and treated in small portions with Br2 (5.76 g [=1.86 ml], 36 mmol) over a period of 30 minutes. After that the mixture is stirred for further 60 minutes at 85° C. After cooling down to RT and addition of ice-water (450 ml) the compound is extracted with methylene chloride and purified on silica gel using a mixture of cyclo-hexane/ethyl acetate 2/1 to 1/1 obtaining the product as a pale yellow solid.
  • b) 8-bromo-tetralin-6-yl acetate
  • Compound of step a) (7.28 g, 28.75 mmol) is dissolved in CH2Cl2 (60 ml). 2 equivalents m-chloroperbenzoic acid (11.7 g, FLUKA 25800, 70%) is added at RT and after addition of sodium sulfate (5 g) the reaction mixture is treated with trifluoromethanesulfonic acid (250 μl, 2.88 mmol). The reaction is kept at RT for 16 hours. After thin layer control, m-chloroperbenzoic acid (2.25 g, 11 mmol), sodium sulfate (2 g) and trifluoromethanesulfonic acid (50 μl, 0.57 mmol) is added to the reaction (2 times within 6 hours). The organic layer is filtered and extracted 3 times with an aqueous sodium thiosulfite solution. The compound is purified on silica gel using a mixture of cyclo-hexane/ethyl acetate 2/1 to 1/1 obtaining the product as colorless oil.
  • c) 5,6,7,8-tetrahydro-4-bromo-2-naphthol
  • Compound of step b) (6.1 g, 22.66 mmol) is added to MeOH (200 ml) at RT. 1N sodium methoxide in methanol (22.7 ml) is stirred for 15 min. Amberlite IR-120 (H+-form) is added to the mixture until the reaction mixture becomes neutral. The ion exchanger is filtered off, the solvent is evaporated and the crude product is isolated without any further purification as a pale yellow solid.
  • d) 2-(2-t.butoxy-ethoxy)4-bromo-5,6,7,8-tetrahydro-naphthalene
  • Compound of step c) (2.8 g, 12.33 mmol) is added to THF (100 ml) at RT. Triphenyl-phosphine (16.82 g, 64.13 mmol) and (after 10 minutes) diisopropylazodicarboxylate (11.66 ml, 59.2 mmol) are added and the mixture is stirred for 14 hours. The solvent is removed and the residue is purified on silica gel using cyclohexane (100%) to cyclohexane/-methylene chloride (1/1) as mobile phase, obtaining the product as a pale yellow solid.
  • e) 3-(2-t.butoxy-ethoxy)-5,6,7,8-tetrahydro-naphthalen-1-yl-oxo-acetic acid methyl ester
  • Compound of step d) (2.61 g, 7.96 mmol) is dissolved in dry THF (70 ml) at RT and cooled down to −70° C. Butyllithium (5.5 ml, 8.76 mmol, 1.6 M in hexane) is added dropwise to the reaction mixture under inert atmosphere (argon). A solution of dimethyl oxalate (1.9 g, 15.92 mmol) in 5 ml dry THF is added to the reaction mixture and the reaction is warmed up to RT. The reaction mixture is poured into Titrisol buffer solution (pH 7) and extracted with ethyl acetate. The organic layer is dried and the compound is purified on silica gel using cyclohexane/methylene chloride 1/2 as mobile phase obtaining the product as white solid.
  • f) 3-[3-(2-t.butoxy-ethoxy)-5,6,7,8-tetrahydro-naphthalen-1-yl]4-(1.H.-indol-3-yl)-pyrrole-2,5-dione
  • Compound of step e) (1 g, 2.99 mmol) and indole-3-acetamide (270 mg, 1.49 mmol) is added to THF (10 ml) and heated up to reflux. t.-BuOK (6 ml, 6 mmol, 1 M in THF) is added dropwise under argon and the reaction is kept at reflux for 1 hour. The reaction mixture is diluted with ethyl acetate and extracted with saturated aqueous NaHCO3 solution. The organic layer is dried and the compound is isolated as an orange solid after removal of the solvent without any further purification.
  • EXAMPLE 54 3-[3-(2-Dimethylamino-ethoxy)-5,6,7,8-tetrahydro-naphthalen-1-yl]-4-(1.H-indol-3-yl)-pyrrole-2,5-dione
  • Figure US20070191401A1-20070816-C00021
  • The compound of example 53 (380 mg, 0.94 mmol) is suspended in methylene chloride (20 ml) containing 225 μl pyridine (2.8 mmol). After addition of methanesulfonic anhydride (393 mg, 2.26 mmol) the reaction mixture is kept at RT for 14 hours. The reaction is extracted with 1N HCl in water, the organic layer is dried over sodium sulfate and the solvent is removed under reduced pressure. The residue is dissolved in THF and then treated with an aqueous solution of dimethylamine (2.5 ml). After 96 hours the solvent is removed and pure title compound is obtained as an orange powder after silica gel chromatography (methanol/ethylacetate 1/1 as mobile phase). MH+: 430 (ES+)
  • 1H NMR (400 MHz, DMSO-d6)δ 11.87 (bs, 1H), 10.99 (s, 1H), 7.98 (t, 1H, J=0.40 Hz), 7.39 (d, 1H, J=8.07 Hz), 7.05 (t, 1H, J=7.34 Hz), 6.75 (d, 1H, J=2.44 Hz), 6.68 (t, 1H, J=7.33 Hz), 6.63 (d, 1H, J=2.69 Hz), 6.44 (d, 1H, J=8.07 Hz), 3.98 (m, 2H), 2.70 (m, 2H), 2.63:(m, 2H), 2.40 (m, 1H), 2.23 (s, 6H), 2.12 (m, 1H), 1.63 (m, 1H), 1.53 (m 1H), 1.41 (m, 2H)
  • EXAMPLE 55A 3-(1.H.-Indol-3-yl)-4-[3-(4-methyl-piperazin-1-yl)-5,6,7,8-tetrahydro-naphthalen-1-yl]-pyrrole-2,5-dione
  • The title compound is prepared by condensing 2-[3-(4-methyl-piperazin-1-yl)-5,6,7,8-tetrahydro-naphthalen-1-yl]-acetamide with (1.H.-indol-3-yl)-oxo-acetic acid methyl ester in analogy to example 53 (step f). MH+: 441 (ES+)
  • EXAMPLE 55B 3-(1-Methyl-1.H.-indol-3-yl)-4-[3-(4-methyl-piperazin-1-yl)-5,6,7,8-tetrahydro-naphthalen-1-yl]-pyrrole-2,5-dione
  • The title compound is prepared by condensing 2-[3-(4-methyl-piperazin-1-yl)-5,6,7,8-tetrahydro-naphthalen-1-yl]-acetamide with (1-methyl-1.H.-indol-3-yl)-oxo-acetic acid methyl ester in analogy to example 53 (step f). MH+: 455 (ES+)
    Figure US20070191401A1-20070816-C00022
  • 2-[3-(4-methyl-piperazin-1-yl)-5,6,7,8-tetrahydro-naphthalen-1-yl]-acetamide, used as starting material, is prepared as follows:
  • a) 5-Bromo-7-methoxy-1,2,3,4-tetrahydro-naphthalene
  • 5,6,7,8-tetrahydro-4-bromo-2-naphthol (7 g, 30.8 mmol; step c) in example 29) is dissolved in dry acetone (100 ml) and treated with K2CO3 (15 g, 0.11 mol) and methyliodide (7.25 ml, 0.13 mmol). The reaction mixture is kept at RT over night. After filtration and removal of the solvent the residue is purified over silica gel (methylene chloride/cyclohexane 1/1) affording the compound as a white solid.
  • b) (3-Methoxy-5,6,7,8-tetrahydro-naphthalen-1-yl)-acetic acid ethyl ester
  • To a solution of compound of step a) (2.75 g, 11.4 mmol) in DMF (110 ml) ZnBr2 (3.4 g, 14.8 mmol), are added α-(tributylstannyl)acetate (5.5 g, 14.8 mmol) and dichlorobis(tri-o-tolylphosphine)-palladium. The reaction is kept under argon at 80° C. over night. DMF is removed under reduced pressure and the crude residue is dissolved in ethyl acetate and extracted with aqueous NaHCO3 (6%). The organic layer is dried, concentrated and purified on silica gel (methylene chloride/cyclohexane 1/1) affording the pure compound.
  • c) (3-Hydroxy-5,6,7,8-tetrahydro-naphthalen-1-yl)-acetic acid ethyl ester DL-Methionine (1.11 g, 7.5 mmol) and compound of step b) is dissolved in methanesulfonic acid (9 ml) and kept at RT over night. The reaction is poured into a ice-cold saturated NaCl solution and extracted 3 times with methylene chloride. The organic layer is dried, concentrated and purified on silica gel (methylene chloride/methanol 95/5) affording the pure compound.
  • d) (3-Trifluoromethanesulfonyloxy-5,6,7,8-tetrahydro-naphthalen-1-yl)-acetic acid ethyl ester
  • To a solution of compound of step c) (940 mg, 4 mmol) in pyridine (2 ml) trifluoromethane-sulfonic anhydride (720 μl, 4.4 mmol) is added at 0° C. The reaction mixture is kept at RT over night. The reaction is poured into a ice-cold water and extracted 3 times with diethyl ether. The organic layer is dried, concentrated and purified on silica gel (methylene chloride) affording the compound.
  • e) [3-(4-Methyl-piperazin-1-yl)-5,6,7,8-tetrahydro-naphthalen-1-yl]-acetic acid ethyl ester
    Figure US20070191401A1-20070816-C00023
  • To a solution of compound of step d) (0.5 g, 1.36 mmol) in dry THF (10 ml) K3PO4 (405 mg, 1.90 mmol), N-methylpiperazine (180 μl, 163 mmol), tris(dibenzylideneacetoneacetone)-dipalladium(0) (60 mg, 0.07 mmol) and 2-(di-t-butylphosphino)biphenyl (21 mg, 0.07 mmol) are added under argon. The reaction is kept under argon at 80° C. for 24 hours. After filtration DMF is removed under reduced pressure and the crude residue is purified on silica gel (methylene chloride/methanol 95/5) affording the pure compound.
  • 1H NMR (400 MHz, CDCl3) δ 6.68 (d, 1H, J=2.44 Hz), 6.59 (d, 1H, J=2.20 Hz), 4.15 (q, 2H, J=7.34 Hz), 3.55 (s, 2H), 3.17 (t, 4H, J=5.13 Hz), 2.75 (t, 2H, J=5.87 Hz), 2.60 (t, 2H, J=6.11 Hz), 2.57 (t, 4H, J=4.90 Hz), 2.35 (s, 3H), 1.77 (m,.4H), 1.25 (t, 3H, J=7.10 Hz)
  • f) 2-[3-(4-Methyl-piperazin-1-yl)-5,6,7,8-tetrahydro-naphthalen-1-yl]-acetamide
  • A solution of compound of step e) (394 mg, 1.2 mmol) in methanol/NH3 (4 molar) is transferred into an autoclave and kept at 120° C. for 48 hours. After cooling the reaction is concentrated and the crude residue is purified on silica gel (methylene chloride/methanol 90/105) affording the pure title compound.
  • EXAMPLE 56 3-1.H.-indol-3-yl)-4-[2-(4-methyl-piperazin-1-yl)-quinazolin yl]-pyrrole-2,5-dione
  • Figure US20070191401A1-20070816-C00024
  • 2-[2-(4-methyl-piperazin-1-yl)-quinazolin-4-yl]-acetamide (213 mg, 0.75 mmol) and 3-indoleglyoxylic acid methyl ester (167 mg, 1.1 eq.) are dissolved in THF (15 mL). To-the suspension is added dropwise at 0° C. t-BuOK 1.0 M in THF (2.25 mL, 3.0 eq.). The mixture is stirred at RT overnight. A second portion of glyoxalate (30 mg, 0.2 eq.) and t-BuOK (0.5 mL) are added and the mixture is stirred at RT for 24 h. AcOEt is added and the organic phase is washed with aqueous NaHCO3 1.0 M and brine. The organic phase is dried over Na2SO4 and evaporated. The residue is purified by FCC (Et2O/MeOH/aqueous NH4OH 90:10:1) to afford the title compound as an orange-red powder. ESI-MS: 437 [M−H]+;
  • 1H NMR (DMSO, 400 MHz) δ 2.13 (s, 3H), 2.16 (m, 4H), 3.69 (m, 4H), 6.35 (d, J=8.0 Hz, 1H), 6.64 (dd, J=7.8, 7.4 Hz, 1H), 7.02 (dd, J=7.6, 7.4 Hz, 1H), 7.10 (dd, J=7.8, 7.2 Hz, 1H), 7.38 (d, J=8.2 Hz, 1H), 7.53 (d, J=8.4 Hz, 1H), 7.63-7.73. (m, 2H), 8.13 (s, 1H), 11.29 (br s, 1H), 12.01 (br s, 1H). The resulting compound is dissolved in ethanol and 1.2 equivalents of acetic acid are added. The solvent is evaporated under reduced pressure yielding the acetate salt.
  • 2-[2-(4-methyl-piperazin-1-yl)-quinazolin-4-yl]-acetamide
    Figure US20070191401A1-20070816-C00025
  • used as starting material may be prepared as follows:
  • a) To a suspension of 1H,3H-quinazolin-2,4-dione (10.0 g, 61.7 mmol) in POCl3 (37.0 mL) is added dropwise N,N-dimethylaniline (7.8 mL, 1.0 eq.). The mixture is heated to 110° C. and kept at reflux for 35 h. The solution is cooled to RT and poured onto an ice-water mixture. The precipitate is filtered off and washed with H2O. The solid is redissolved in AcOEt and washed with H2O and brine. The organic phase is dried over Na2SO4 and evaporated to afford crude 2,4-dichloro-quinazoline which can be recrystallized from toluene/pentane. EI-MS: 198 [M−H]+, 163 [M−Cl]+;
  • b) Ethyl acetoacetate (5.08 mL, 2.0 eq.) dissolved in THF (25 mL) is added dropwise to a suspension of NaH (60%, 1.04 g, 1.3 eq.) in THF (25 mL) at 0° C. The solution is stirred 30 min. at 0° C. and the solvent is evaporated. The residue is redissolved in toluene (125 mL) and 2,4-dicloro-quinazoline (4.0 g, 20.0 mmol) is added. The mixture is stirred 30 min. at reflux and the toluene is evaporated. The oily residue is redissolved in aqueous NH4OH 25% (80 mL) and stirred overnight at RT. All volatile materials are evaporated and the residue is taken up in AcOEt (80 mL). The suspension is heated at reflux for 15 min., cooled to 0° C. and filtered to afford 2-(2-chloro-quinazolin-4-yl)-acetamide as a white solid.
  • EI-MS: 221 [M]+178; IR (KBr) vmax 3302, 3138, 1681, 1542, 1388, 1287, 948, 771; 1H NMR (DMSO, 400 MHz) δ 4.21 (s, 2H), 7.24 (br s, 1H), 7.75-7.84 (m, 2H), 7.97 (d, J=8.4 Hz, 1H), 8.08 (dd, J=8.4, 7.5 Hz, 1H), 8.34 (d, J=8.4 Hz, 1H)
  • c) 2-(2-chloronuinazolin-4-yl)-acetamide (221 mg, 1.0 mmol) is dissolved in 1-methyl-2-pyrrolidinone (2.0 mL) and N-methylpiperazine (555 μL, 5.0 eq.) is added. The mixture is heated 45 min. at 50° C. AcOEt is added and the suspension is filtered to afford 2-[2-(4-methyl-piperazin-1-yl)-quinazolin-4-yl]-acetamide as a white solid.
  • ESI-MS: 284 [M−H]+, 241; 1H NMR (DMSO, 400 MHz) δ 2.24 (s, 3H), 2.40 (m, 4H), 3.86 (m, 4H), 3.98 (s, 2H), 7.12 (br s, 1H), 7.24 (dd, J=8.2, 7.5 Hz, 1H), 7.49 (d, J=8.4 Hz, 1H), 7.63-7.72 (m, 2H), 7.95 (d, J=8.2 Hz, 1H);
  • The compounds of formula X3
    Figure US20070191401A1-20070816-C00026
  • wherein R1, R5, R6 and R7 are as defined in Table 3 below, may be prepared by following the procedure of Example 56 but using the appropriate starting materials. The compounds may also be converted into the acetate salt as disclosed for the compound of Example 56, where appropriate.
    TABLE 3
    R6 (position 7 R7 (position 5
    Ex. R5 R1 or 8) or 6) M.S. Data
    57 —OCH3 H H H M+ 370 (EI)
    58 —N(CH3)2 H H H M+ 383 (EI)
    59 —O—CH2—CH2—N(CH3)2 H H (M − H) 426 (ES)
    60 —O—CH2—CH2—OH H H H (M − H) 399 (ES)
    61 —HN—CH2—CH2—N(CH3)2 H H H (M − H) 425 (ES)
    62 —O-(1-methyl-piperidin-4-yl) H H H (M − H) 452 (ES)
    63 —O—(CH2)3—N(CH3)2 H H H (M + H)+ 442 (ES)
    64 —N(CH3)—CH2—CH2—N(CH3)2 H H H (M − H) 439 (ES)
    65 -(4-methyl-piperazin-1-yl) H 7-OCH3 H (M − H) 467 (ES)
    66 -(4-methyl-piperazin-1-yl) H 8-CH3 H (M − H) 451 (ES)
    67 -(4-methyl-piperazin-1-yl) H 7-OCH3 6-OCH3 (M − H) 497 (ES)
    68 -(4-methyl-piperazin-1-yl) H H 6-Cl (M − H) 471 (ES)
    69 -(4-methyl-piperazin-1-yl) H 7-Cl H (M − H) 471 (ES)
    70 -1-piperazinyl H H H (M − H) 423 (ES)
    71 morpholin-4-yl H H H (M − H) 424 (ES)
    72 -(4-methyl-piperazin-1-yl) H H 6-CH3 (M − H) 451 (ES)
    73 -[4-(2-hydroxyethyl)- H H H (M − H) 467 (ES)
    piperazin-1-yl]
    74 -(4-methyl-piperazin-1-yl) H H 6-F M + 456 (EI)
    75 (cis)3,5-dimethyl-1-piperazinyl H H H (M − H) 451 (ES)
    76 -4-N-(pyridin-4-yl)-1- H H H (M − H) 500 (ES)
    piperazinyl
    77 (rac.) 3-methyl-piperazin-1-yl H H H (M − H) 437 (ES)
    78 -(4-methyl-piperazin-1-yl) H H 6-OCH3 (M − H) 467 (ES)
    79 -(4-methyl-piperazin-1-yl) H H 6-OH MH+ 455
    80 -(4-benzyl-piperazin-1-yl) H H H MH+ 515
    81 (trans) 2,5,-dimethyl-1- H H H MH+ 453
    piperazinyl
    82 -[4-(2-dimethylamino-ethyl)- H H H MH+ 496
    piperazin-1-yl]
    83 4-phenyl-piperazin-1-yl H H H (M − H) 499
    84 4-ethyl-piperazin-1-yl H H H MH+ 453
    85 4-isppropyl-piperazin-1-yl H H H MH+ 466
    86 -(4-methyl-piperazin-1-yl) H 7-F H MH+ 457
    87 -(4-methyl-piperazin-1-yl) CH3 H 6-Cl MH+ 487
    88 1-piperazinyl CH3 H 6-Cl MH+ 473
    89 (cis)3,5-dimethyl-piperazin-1- H H 6-Cl MH+ 487
    yl
    90 (cis)3,5-dimethy-1-piperazinyl CH3 H 6-Cl MH+ 501
    91 (cis)3,4,5-trimethyl-1- H H 6-Cl MH+ 501
    piperazinyl
    92 (cis)3,4,5-trimethyl-1- CH3 H 6-Cl MH+ 515
    piperazinyl
    93 -(4-methyl-piperazin-1-yl) H H 5-CH3 MH+ 453
    94 -(4-ethyl-piperazin-1-yl) H H 6-Cl MH+ 487
    95 -(4-isopropyl-1-piperazin-1-yl) H H 6-Cl MH+ 501
    96 -(4-ethyl-piperazin-1-yl) CH3 H 6-Cl MH+ 501
    97 -(4-isopropyl-piperazin-1-yl) CH3 H 6-Cl MH+ 515
    98 -(4-cyclopropyl-piperazin-1-yl) H H H MH+ 465
    99 -(4,7-diaza-spiro[2.5]oct-7-yl) H H H MH+ 451,
    MNa+ 473
    100 -(4,7-diaza-spiro[2.5]oct-7-yl) CH3 H H MH+ 465,
    MK+ 503
    101 -(4-cyclopropyl-piperazin-1-yl) CH3 H H MH+ 479
    102 -(4-methyl-4,7-diaza- H H H MH+ 465
    spiro[2.5]oct-7-yl)
    103 -(4-methyl-3,3-diethyl- H H H MH+ 495
    piperazin-1-yl)
    104 -(4-methyl-4,7-diaza- CH3 H H MH+ 479;
    spiro[2.5]oct-7-yl) MNa+ 501
    105 -(4-methyl-3,3-diethyl- CH3 H H MH+ 509
    piperazin-1-yl)
    106 -4-(1-methyl-cyclopropyl)-1- H H H MH+ 479
    piperazinyl
    107 -4-(1-methyl-cyclopropyl)-1- CH3 H H MH+ 493;
    piperazinyl MNa+ 515
    108 1-piperazinyl CH3 H H MH+ 439
    109 -(4-methyl-piperazin-1-yl) CH3 H H (M + H)+ 453
    110 -(4-methyl-piperazin-1-yl) 2-hydroxy- H 6-Cl (M + H)+ 517
    ethyl
    111 (4-N-methyl-1-homopiperazinyl) H H 6-Cl (M + H)+ 487
    112 -(4-t-butyl-piperazin-1-yl) H H 6-Cl (M + H)+ 515
    113 3-methyl-piperazin-1-yl CH3 H 6-Cl (M + H)+ 487
    114 -(4-t-butyl-piperazin-1-yl) CH3 H 6-Cl (M + H)+ 519
    115 -(4-methyl-4,7-diaza-spiro[2.5]oct- CH3 H 6-Cl (M + H)+ 499
    7-yl)
    116 3-R-methyl-piperazin-1-yl CH3 H 6-Cl (M + H)+ 487
    117 3-S-methyl-piperazin-1-yl CH3 H 6-Cl (M + H)+ 487
    118 3,3-dimethyl-1-piperazinyl CH3 H 6-Cl (M + H)+ 501
    119 3,3-dimethyl-1-piperazinyl H H 6-Cl (M + H)+ 487
    120 3,3-dimethyl-1-piperazinyl CH3 H H (M + H)+ 467
    121 3,3-dimethyl-1-piperazinyl H H H (M − H) 451
    122 -(4-methyl-piperazin-1-yl) 2-(CH3)2N- H 6-Cl (M + H)+ 544
    ethyl
  • The compound of Example 77 which is a racemate may also be prepared in the form of pure cis or trans enantiomer using the corresponding cis or trans starting materials. The same applies to the cis isomers of Examples 75, and 89 to 92 and to the trans isomer of Example 81: they may also be prepared as a racemate or in pure trans or cis form, respectively, using the corresponding starting materials. Compounds of Examples 116 and 117 may also be prepared as a racemate or in the corresponding S or R isomer form.
  • The compounds of formula X4
    Figure US20070191401A1-20070816-C00027
  • wherein R, R1 and R2 are as defined in Table 4 below, may be prepared by following the procedure of Example 56 but using the appropriately substituted (1.H.-indol-3-yl)-oxo-acetic acid ethyl ester.
    TABLE 4
    M.S. Data (ESI-
    Ex. R R1 R2 MS)
    123 4-F H H 457 [M + H]+
    124 4-OMe H H 469 [M + H]+
    125 5-F H H 457 [M + H]+
    126 5-OMe H H 469 [M + H]+
    127 5-Cl H H 473 [M + H]+
    128 6-F H H 457 [M + H]+
    129 7-F H H 457 [M + H]+
    130 6-OCH3 H H 469 [M + H]+
    131 6-Cl H H 473, 475 [M + H]+
    132 5-Br H H 517, 519 (M+)
    133 4-CH3 H H 453 [M + H]+
    134 4-Cl H H 473, 475 [M + H]+
    135 5-CH3 H H 453 [M + H]+
    136 7-Br H H 517, 519 (M+)
    137 7-Cl H H 473, 475 [M + H]+
    138 6-CH3 H H 453 [M + H]+
    139 7-CH3 H H 453 [M + H]+
    140 7-OCH3 H H 469 [M + H]+
    141 4-Br H H 517, 519 [M + H]+
    142 6-Br H H 517, 519 [M + H]+
    143 H CH3 CH3 MH+ 467
    144 H H CH3 MH+ 453
    145 5-OH H H MH+ 455
    146 5-NO2 H H MH+ 484
    147 5-NHCH3 H H MH+ 468
    148 5-NH2 H H MH+ 454
  • EXAMPLES 149A and 149B
  • The compounds are prepared by following the procedure of Example 56 but using the appropriately substituted (1.H.-indol-3-yl)-oxo-acetic acid methyl ester.
    Figure US20070191401A1-20070816-C00028
  • (7-Fluoro-1.H.-indol-3-yl)-oxo-acetic acid methyl ester, used as starting material may be prepared as follows:
  • 7-Fluoroindole (0.147 g, 1.09 mmol) is dissolved in dry THF (5 ml) and the solution cooled to 0° C. A solution of 2M oxalyl chloride in dichloromethane (0.65 mL, 1.31 mmol) is added and the mixture is stirred for a further 10 min at 0° C. and then 4 h at room temperature. The mixture is cooled to 0° C. and methanol (10 ml) is added. The mixture is stirred for a further 18 h at room temperature. The mixture is evaporated to dryness under reduced pressure, the solid residue is washed with a mixture of ethyl acetate:hexane (1:1) and dried under high vacuum to afford (7-fluoro-1.H.-indol-3-yl)-oxo-acetic acid methyl ester. The product is used without further purification.
  • ESI-MS: 220 [M−H]+
  • The corresponding substituted (1.H.-indol-3-yl)-oxo-acetic acid methyl ester, used as starting materials in the preparation of examples 123-148 were prepared in an a way from the corresponding indoles.
  • The compounds of formula X5
    Figure US20070191401A1-20070816-C00029
  • wherein R, R1 and R2 are as defined in Table 5 below, may be prepared by following the procedure of Example 56 but using the appropriately substituted (1.H.-indol-3-yl)-oxo-acetic acid methyl ester.
    TABLE 5
    Ex. R14 R15 R′15 R1 M.S. Data
    150 -(4-N-methyl)-1-piperazinyl H H H [M + H]+ 389
    151 -(4-N-methyl)-1-piperazinyl H H CH3 [M + H]+ 403
    152 -3,5-dimethyl-1-piperazinyl H H H [M + H]+ 403
    153 -3,5-dimethyl-1-piperazinyl H H CH3 [M + H]+ 417
    154 -(4-N-methyl)-1-piperazinyl 5-CH3 H H [M + H]+ 403
    155 -(4-N-methyl)-1-piperazinyl 5-CH3 H CH3 [M + H]+ 417
    156 -(4-N-methyl)-1-piperazinyl 6-CH3 H H [M + H]+ 403
    157 -(4-N-methyl)-1-piperazinyl 6-CH3 H CH3 [M + H]+ 417
    158 -(4-N-methyl)-1-piperazinyl 6-CH3 5-CH3 CH3 [M + H]+ 431
    159 -(4-N-methyl)-1-piperazinyl 6-CH3 5-CH3 H [M + H]+ 417
    160 -(4-N-methyl)-1-piperazinyl 5-Cl H CH3 [M + H]+ 437
    161 -(4-N-methyl)-1-piperazinyl 5-Cl H H [M + H]+ 423
  • EXAMPLE 162 3-[2-(2-dimethylamino-ethoxy)-quinolin-4-yl]-4-1.H.-indol-3-yl)-pyrrole-2,5-dione
  • Figure US20070191401A1-20070816-C00030
  • 2-(2-chloro-quinolin-4-yl)-acetamide (110 mg, 0.5 mmol),.methyl 3-indoleglyoxylate (102 mg, 0.5 mmol) and N,N-dimethylaminoethanol (508 μl, 10 eq.) are, dissolved in dry THF (5.0 mL) at 0° C. 1.0 M t.-BuOK (2.5 mL, 5 eq.) is added and the reaction is stirred at 80″C overnight. The mixture is allowed to cool to RT, diluted with AcOEt (20 mL) and washed with H20 (10 mL) and brine (5 mL). The organic phase is dried over Na2SO4 and evaporated. The residue is purified by FCC (AcOEt/EtOH/28% NH4OH 90:9:1) to afford a red powder, which can be recrystallized from CH2Cl2/Et2O to give the pure title compound.
  • 1H NMR (DMSO, 400 MHz) δ 2.20 (s, 6H), 2.67 (m, 2H), 4.51 (m, 2H), 6.38 (d, J=8.4 Hz, 1H), 6.53 (dd, J=8.4, 7.5 Hz, 1H), 6.95 (m, 2H), 7.21 (dd, J=8.4, 7.5 Hz, 11H), 7.34 (d, J=8.4 Hz, 1H), 7.57 (dd, J=8.4, 7.5 Hz, 1H), 7.64 (d, J=8.4 Hz, 1H), 7.78 (d, J=8.4 Hz, 1H), 8.00 (s, 1H), 11.20 (br s, 1H), 11.92 (br s, 1H);
  • ES-MS: 427 [M+H]+
  • 2-(2-chloro-quinolin4-yl)-acetamide
    Figure US20070191401A1-20070816-C00031
  • used as starting material may be prepared as follows:
  • a) 2-(2-chloro-quinolin-4-yl)-acetic acid methyl ester may be prepared as disclosed in EP-A1-0 364 327 and Indian J. Chem. 1994, 33B, 747-751.
  • b) 2-(2-chloro-uinolin-4-yl)-acetic acid methyl ester (100 mg, 0.42 mmol) is suspended in 28% NH4OH (2.0 mL). The mixture is stirred at RT overnight. The solvent is evaporated to afford 2-(2-chloroquinolin-4-yl)-acetamide as a solid.
  • 1H NMR (DMSO, 300 MHz) d 3.88 (s, 2H), 7.16 (br s, 1H), 7.30 (s, 1H), 7.65 (dd, J=8.4, 7.5 Hz, 1H), 7.70 (br s, 1H), 7.80 (dd, J=8.4, 7.5 Hz, 1H), 7.93 (d, J=8.4 Hz, 1H), 8.12 (d, J=8.4 Hz, 1H)
  • EXAMPLE 163 3-(1.H.-indol-3-yl)-4-[2-(4-methyl-piperazin-1i-yl)-quinolin-4-yl]-pyrrole-2,5-dione
  • Figure US20070191401A1-20070816-C00032
  • 2-[2-(4-methyl-piperazin-1-yl)-quinolin4-yl]-acetamide (200 mg, 0.70 mmol) and methyl 3-indoleglyoxylate (143 mg, 0.70 mmol) are dissolved in DMF (7.0 mL) at 0″C. 1.0 M t-BuOK (3.52 mL, 5 eq.) is added and the reaction is stirred at 80° C. overnight. The mixture is cooled to RT, diluted with CH2Cl2 (40 mL) and washed with H2O (2×10 mL) and brine (5 mL). The organic phase is dried over Na2SO4 and evaporated. The residue is purified by FCC (AcOEt/H2O/AcOH 7:1:1) to afford the acetate salt. The salt is redissolved in AcOEt (20 mL) and washed with saturated aqueous NaHCO3 (2×10 mL). The organic phase is dried over Na2SO4 and evaporated to afford the title compound as a red powder.
  • 1H NMR (DMSO, 400 MHz) δ 2.22 (s, 3H), 2.37 (m, 4H), 3.62 (m, 4H), 6.63 (dd, J=8.4, 7.5 Hz, 1H), 6.74 (d, J=8.4 Hz, 1H), 6.95-7.05 (m, 2H), 7.10 (s,1H), 7.34 (d, J=8.4 Hz, 1H), 7.41-7.50 (m, 2H), 7.60 (d, J=8.4 Hz, 1H), 7.85 (s, 1H), 10.77 (br s,1H), 11.48,(br s,1H);
  • ES-MS: 438 [M+H]+
  • 2-[2-(4-methyl-piperazin-1-yl)-quinolin4-yl]-acetamide
    Figure US20070191401A1-20070816-C00033
  • used as starting material may be prepared as follows:
  • 2-(2-chloro-quinolin-4-yl)-acetamide (500 mg, 2.27 mmol) is dissolved in 1-methyl-2-pyrrolidinone (3.0 mL). N-methylpiperazin (1.3 mL, 5 eq.) is added and the reaction is stirred at 80° C. for 48 h. AcOEt (20 mL) is added and the precipitate is purified by FCC (AcOEt/EtOH/28% NH4OH 80:18:1) to afford 2-[2-(4-methyl-piperazin-1-yl)-quinolin-4-yl]-acetamide as a solid.
  • 1H NMR (DMSO, 400 MHz) δ 2.25 (s, 3H), 2.47 (m, 4H), 3.67 (m, 4H), 3.77,(s, 2H), 7.00 (br s, 1H), 7.16 (s, 1H), 7.22 (dd, J=8.4, 7.5 Hz, 1H), 7.50 (dd, J=8.4, 7.5 Hz, 1H), 7.56 (m, 2H), 7.85 (d, J=8.4 Hz, 1H);
  • ES-MS: 285 [M+H]+
  • The compounds of formula X6
    Figure US20070191401A1-20070816-C00034
  • wherein R8 and R9 are as defined in Table 6 below, may be prepared by following the procedure of Example 162 or 163 but using the appropriate starting materials.
    TABLE 6
    Ex. R8 Position-R9 Ra M.S Data
    164 Cl H H 374 (M + H)+
    165 —OCH3 6-CH3 H 384 (M + H)+
    166 —O—CH2—CH2—N(CH3)2 6-CH3 H 441 (M + H)+
    167 —O—(CH2)3—N(CH3)2 6-CH3 H 455 (M + H)+
    168 —OCH3 H H 370 (M + H)+
    169 —OCH3 7-CH3 H 384 (M + H)+
    170 —O—CH2—CH2—N(CH3)2 7-CH3 H 441 (M + H)+
    171 -(4-N-methyl)-1-piperazinyl 7-CH3 H 452 (M + H)+
    172 -(4-N-methyl)-1-piperazinyl 7-CH3 CH3 466 (M + H)+
    173 -(4-N-methyl)-1-piperazinyl H CH3 438 (M + H)+
    174 -(4-N-methyl)-1-piperazinyl 6-CH3 H 452 (M + H)+
    175 -(4-N-methyl)-1-piperazinyl 6-CH3 CH3 466 (M + H)+
    176 -(4-N-cyclopentyl)-1-piperazinyl H CH3 506 (M + H)+
    177 -(4-N-cyclopentyl)-1-piperazinyl H H 492 (M + H)+
    178 -(4-N-3-hydroxypropyl)-1- H H 482 (M + H)+
    piperazinyl
    179 -(4-N-3-dimethylamino-propyl) H H 509 (M + H)+
    180 -(4-N-pyrimidin-2-yl)-1- CH3 H 516 (M + H)+
    piperazinyl
  • EXAMPLE 181 3-(1.H.-Indol-3-yl)-4-[3-(4-methyl-piperazin-1-yl)-isoquinolin-1-yl]-pyrrole-2,5-dione
  • Figure US20070191401A1-20070816-C00035
  • 3-(3-Chloro-isoquinolin-1-yl)-4-(1.H.-indol-3-yl)-pyrrole-2,5-dione (110 mg, 0.30 mmol) is suspended in N-methylpiperazine (2.5 ml) and kept at 130° C. for 24 hours. The excess of N-methylpiperazine is removed at high vacuo at 60° C. and the residue is dissolved. in ethyl acetate and extracted with 0.5N aqueous H2O. The deep orange aqueous phase is adjusted to pH 9 with 1N NaOH and extracted with ethyl acetate. The organic phase is separated, dried with Na2SO4, concentrated and purified on silica gel using methylene chloride/methanol/acetic acid50% (9/1/0.25) as mobile phase. The fractions with the title compound are collected and extracted with aqueous NaHCO3 solution (6%). The organic phase is separated, dried with Na2SO4, and concentrated to give pure title compound as orange powder. MH+: 438 (ES+) 1H NMR (400 MHz, DMSO-d6) δ 11.88 (s, 1H), 11.14 (broad, 1H), 8.04 (s, 1H), 7.68 (d, 1H, J=8.56 Hz), 7.65 (d, 1H, J=9.05 Hz), 7.45 (t, 1H, J=8.07 Hz), 7.31 (d, 1H, J=8.07 Hz), 7.08 (s, 1H, J=8.33 Hz), 7.08 (t, 1H, J=8.31 Hz), 6,94 (t, 1H, J=8.07 Hz), 6.50 (t, 1H, J=7.34 Hz), 6.16 (d, 1H, J=8.07 Hz), 3.53 (m, 4H), 2.30 (m, 4H), 2.14 (s, 3H)
  • 3-(3-Chloro-isoquinolin-1-yl)-4-(1.H.-indol-3-yl)-pyrrole-2,5-dione, used as starting material, may be prepared as follows:
  • a) (3-chloro-isoquinolin-1-yl) acetic acid ethyl ester may be prepared as disclosed by T. Kametani et al. in Chem. Pharm. Bull., 15(5), 704 (1967).
  • b)(3-chloro-isoquinolin-1-yl)-acetic acid ethyl ester (2.5 g, 10 mmol) is dissolved in 4N NH3/MeOH (50 ml). The solution is transferred into an autoclave and kept at 120° C. for 48 hours. After cooling to RT the solvent is removed and the resulting crude product is purified on silica gel using methylene chloride (100%)→methylene chloride/methanol (95/5) as eluent, affording 2-(3-chloro-isoquinolin-1-yl)-acetamide as a pale yellow solid.
  • c) 2-(3-chloro-isoquinolin-1-yl)-acetamide (440 mg, 2.5 mmol) and methyl 3-indoleglyoxalate (1.0 g, 5 mmol) are added to THF (10 ml) and heated up to reflux. t-BuOK (10 ml, 10 mmol, 1 M in THF) is added dropwise under argon and the reaction is kept at reflux for 1 hour. The reaction mixture is diluted with ethyl acetate and extracted with saturated. aqueous NaHCO3 solution. The organic layer is dried over sodium sulfate and the compound is isolated after treatment with diethylether and filtration as an orange solid. MH+: 375 (ES@).
  • 1H NMR (400 MHz, DMSO-d6) δ 12.04 (s, 1H), 11.30 (s, 1H), 8.14 (s, 1H), 8.11 (s, 1H), 7.99 (d, 1H, J=8.32 Hz), 7.95 (d, 1H, J=8.56 Hz), 7.75 (t, 1H, J=8.33 Hz), 7.50 (t, 1H, J=8.33 Hz), 7.32 (d, 1H, J=8.07 Hz), 6.92 (t, 1H, J=8.07 Hz), 6.48 (t, 1H, J=7.33 Hz), 5.96 (d, 1H, J=8.07 Hz).
  • EXAMPLE 182 3-(1-Methyl-1.H.-indol-3-yl)-4-[3-(4-methyl-piperazin-1-yl)-isoquinolin-1-yl]-pyrrole-2,5-dione
  • Figure US20070191401A1-20070816-C00036
  • 2-[3-(4-Methyl-piperazin-1-yl)-isoquinolin-1-yl]-acetamide (710 mg, 2.5 mmole) and methyl (1-methylindolyl)-3-glyoxalate (1.1 g, 5 mmol) are reacted in THF in the presence of t-BuOK at reflux according to the procedure disclosed in Example 181 c) above. The title compound is isolated as disclosed in Example 181 c). MH+: 452 (ES+)
  • 2-[3-(4-Methyl-piperazin-1-yl)-isoquinolin-1-yl]-acetamide,- used as starting material, may be prepared as follows:
  • a) (3-Chloro-isoquinolin-1-yl)-acetic acid ethyl ester (13 g, 50 mmol) is dissolved in dioxane (150 ml) under argon. To this solution BINAP (1.3 g, 2 mmol), palladium(II)acetate (1.3 g, 4 mmol), N-methylpiperazine (11 ml, 0.1 mol) and t.-BuONa (5.4 g, 55 mmol) is added (under argon) and the reaction is kept under reflux for 30 minutes. After cooling the. reaction is diluted with methylene chloride (300 ml) and extracted with 0.5 N aqueous HCl (300 ml). After filtration the aqueous phase is adjusted to pH 8.5 with solid sodium bicarbonate and extracted with methylene chloride (3×). The combined organic phases are dried (Na2SO4) and concentrated. The resulting crude is purified on silica gel using methylene chloride (100%)→methylene chloride/methanol (90/10) as eluent obtaining the product as a pale yellow solid.
  • b) To a solution of [3-(4-Methyl-piperazin-1-yl)-isoquinolin-1-yl]-acetic acid ethyl ester (1.7 g, 5.4 mmol) in DMF, formamide (0.72 ml, 18.1 mmol) is added under argon. After heating up to 100° C. 5.4N MeONa in methanol (1 ml) is added in 10 portions (0.1 ml each) over a. period of 45 minutes. After 60 minutes at 100° C. the reaction is cooled to RT and diluted with isopropanol (100 ml). The solvents are removed under reduced pressure, the residue is dissolved in ethyl acetate and extracted with aqueous sodium bicarbonate solution (5%). The organic phase is dried (Na2SO4) and concentrated. The resulting crude is purified on silica gel using methylene chloride/methanol (90/10→80/20) as eluent obtaining the product as a white solid.
  • The compounds of formula X7
    Figure US20070191401A1-20070816-C00037
  • wherein Ra, Rc, R8 and R9 are as defined in Table 7 below, may be prepared by following the procedure of Example 181 but using the appropriate starting materials.
    TABLE 7
    Ex. R8 Position-R9 Ra Rc M.S Data
    183 -(4-N-methyl)-1-piperazinyl H H CH3 452 (M + H)+
    184 -(4-N-methyl)-1-piperazinyl H H F 456 (M + H)+
    185 -1-piperazinyl H H F 442 (M + H)+
    186 -(4-N-methyl)-1- H H F 470 (M + H)+
    homopiperazinyl
    187 -(4-N-methyl)-1- H H H 452 (M + H)+
    homopiperazinyl
    188 (rac.)-(3-methyl-piperazin- H H H 438 (M + H)+
    1-yl)
    189 1-piperazinyl H H H 424 (M + H)+
    190 (4-N-isopropyl)-1- H H H 466 (M + H)+
    piperazinyl
    191 3-methyl-1-piperazinyl H H H 438 (M + H)+
    192 -(4-N-methyl)-1-piperazinyl H CH3 F 470 (M + H)+
    193 -(4-N-methyl)-1-piperazinyl H CH3 H 452 (M + H)+
    194 -(4-N-methyl)-1-piperazinyl H CH3 CH3 466 (M + H)+
    195 -(4-N-methyl)-1-piperazinyl 7-Cl H H 473 (M + H)+
    196 -(4-N-methyl)-1-piperazinyl 7-Cl CH3 H 487 (M + H)+
  • The compounds of formula I in free form or in pharmaceutically acceptable salt form exhibit valuable pharmacological properties, e.g. inhibiting Protein Kinase C (PKC), e.g. PKC isoforms like α, β, δ, ε, η or θ activity, inhibiting T-cell activation and proliferation, e.g. by inhibiting production by T-cells or cytokines, e,g, IL-2, by inhibiting the proliferative response of T-cells to cytokines, e.g. IL-2, e.g. as indicated in in vitro and in vivo tests and are therefore indicated for therapy.
  • A. In vitro
  • 1. Protein Kinase C assay
  • The compounds of formula I are tested for their activity on different PKC isoforms according to a published method (D. Geiges et al. Biochem. Pharmacol. 1997;53:865-875) The assay is performed in a 96-well polypropylene microtiterplate (Costar 3794) that has been previously siliconized with Sigmacote (Sigma SL-2). The reaction mixture (50 μl) contains 10 μl of the relevant PKC isozyme together with 25 μl of the test compound and 15 μl of a mix solution that contains 200 μg/ml protamine sulfate, 10 mM Mg(NO3)2, 10 μM ATP (Boehringer 519987) and 3750 Bq of 33P-ATP (Hartmann Analytic SFC301 110 TBq/mmol) in 20 mM Tris-buffer pH 7.4+0.1% BSA. Incubation is performed for 15 min at 32° C. in a microtiterplate shaking incubator (Biolabo Scientific Instruments). Reaction is stopped by adding 10 μl of 0.5 M Na2EDTA, pH 7.4. 50 μl of mixture are pipetted onto a pre-wetted phosphocellulose paper (Whatmann 3698-915) under gentle pressure. Non-incorporated ATP is washed away with 100 μl bi-dist H2O. The paper is washed twice in 0.5% H3PO4 for 15 min followed by 5 min in EtOH. Thereafter the paper is dryed and placed in an omnifilter (Packard 6005219), and overlayed with 10 μl/well of Microscint-O (Packard 6013611) before counting in a Topcount radioactivity counter (Packard). IC50 measurement is performed on a routine basis by incubating a serial dilution of inhibitor at concentrations ranging between 1-1000 μM according to the method described above. IC50 value are calculated from the graph by sigmoidal curve fitting.
  • 2. Protein Kinase C θ Assay
  • Human recombinant PKCO is used under the assay conditions as described above. In this assay, compounds of formula I inhibit PKC θ with an IC50≦1 μM. Compound of Example 56 inhibits PKCθ in this assay with an IC50<10 nM.
  • 3. Protein Kinase Cα Assay
  • Human recombinant PKCα was obtained from Oxford Biomedical Research and is used under the assay conditions as described under Section A.1 above: Compound of Example 100 inhibits PKCα in this assay with an IC50 of 39±15 nM.
  • 4. Protein Kinase Cβ1 Assay
  • Human recombinant PKCβ1 was obtained from Oxford Biomedical Research and is used under the assay conditions as described under Section A.1 above. Compound of Example 163 inhibits PKCβ1 in this assay with an IC50 of 8±2 nM.
  • 5. Protein Kinase Cδ Assay
  • Human recombinant PKCδ was obtained from Oxford Biomedical Research and is used under the assay conditions as described under Section A.1 above. Compound of Example 181 inhibits PKCδ in this assay with an IC50 of 18±8 nM.
  • 6. Protein Kinase Cε Assay
  • Human recombinant PKCε was obtained from Oxford Biomedical Research and is used under the assay conditions as described under Section A.1 above. Compound of Example 139 inhibits PKCε in this assay with an IC50 of 20±7 nM.
  • 7. Protein Kinase Cη Assay
  • Human recombinant PKCη was obtained from PanVera and is used under the assay conditions as described under Section A.1 above. Compound of Example 85 inhibits PKCη in this assay with an IC50 of 50±9 nM.
  • 8. CD28 Costimulation Assay
  • The assay is performed with Jurkat cells transfected with a human interleukin-2 promoter/reporter gene construct as described by Baumann G et al. in Transplant. Proc. 1992;24:43-8, the β-galactosidase reporter gene being replaced by the luciferase gene (de Wet J., et al., Mol. Cell Biol. 1987, 7(2), 725-737). Cells are stimulated by solid phase-coupled antibodies or phorbol myristate acetate (PMA) and the Ca++ ionophore ionomycin as follows. For antibody-mediated stimulation Microlite TM1 microtiter plates (Dynatech) are coated with 3 μg/ml goat anti-mouse IgG Fc antibodies (Jackson) in 55 μl phosphate-buffered saline (PBS) per well for three hours at room temperature. Plates are blocked after removing the antibodies by incubation with 2% bovine serum albumin (BSA) in PBS (300 μl per well) for 2 hours at room temperature. After washing three times with 300 μl PBS per well, 10 ng/ml anti-T cell receptor antibodies (WT31, Becton & Dickinson) and 300 ng/ml anti-CD28 antibodies (15E8) in 50 μl 2% BSA/PBS are added as stimulating antibodies and incubated overnight at 4° C. Finally the plates are washed three times with 300 μl PBS per well. Seven three-fold serial dilutions of test compounds in duplicates in assay medium (RPMI 1640/10% fetal calf serum (FCS) containing 50 μM 2-mercaptoethanol, 100 units/ml penicillin and 100 μg/ml streptomycin) are prepared in separate plates, mixed with transfected Jurkat cells (clone K22 290_H23) and incubated for 30 minutes at 37° C. in 5% CO2. 100 μl of this mixture containing 1×105 cells are then transferred to the antibody-coated assay plates. In parallel 100 μl are incubated with 40 ng/ml PMA and 2 82 M ionomycin. After incubation for 5.5 hours at 37° C. in 5% CO2, the level of luciferase is determined by bioluminescence measurement. The plates are centrifuged for 10 min at 500 g and the supernatant is removed by flicking. Lysis buffer containing 25 mM Tris-phosphate, pH 7.8, 2 mM DTT, 2 mM 1.2-diaminocyclohexane-N,N,N′,N-tetraacetic acid, 10% (v/v) glycerol and 1% (v/v) Triton X-100 is added (20 μl per well). The plates are incubated at room temperature for 10 minutes under constant shaking. Luciferase activity is assessed with a bioluminescence reader (Labsystem, Helsinki, Finland) after automatic addition of 50 μl per well luciferase reaction buffer containing 20 mM Tricine, 1.07 mM (MgCO3)4Mg(OH)2×5H2O, 2.67 mM MgSO4, 0.1 mM EDTA, 33.3 mM DTT, 270 μM coenzyme A, 470 μM luciferin (Chemie Brunschwig AG), 530 μM ATP, pH 7.8. Lag time is 0.5 seconds, total measuring time is 1 or 2 seconds. Low control values are light units from anti-T cell receptor- or PMA-stimulated cells, high controls are from anti-T cell receptor/anti-CD28- or PMA/ionomycin-stimulated cells without any test sample. Low controls are subtracted from all values. The inhibition obtained in the presence of a test compound is calculated as percent inhibition of the high control. The concentration of test compounds resulting in 50% inhibition (IC50) is determined from the dose-response curves. In this assay, compounds of formula I inhibit anti-T cell receptor/anti-CD28 and PMA/ionomycin stimulated Jurkat cells with an IC50≦1 μM. Compound of Example 56 has an IC50 of 42±12 nM in this assay.
  • 9. Allogeneic Mixed Lymphocyte Reaction (MLR)
  • The two-way MLR is performed according to standard procedures (J. Immunol. Methods, 1973, 2, 279 and Meo T. et al., Immunological Methods, New York, Academic Press, 1979, 227-39). Briefly, spleen cells from CBA and BALB/c mice (1.6×105 cells from each strain per well in flat bottom tissue culture microtiter plates, 3.2×105 in total) are incubated in RPMI medium containing 10% FCS, 100 U/ml penicillin, 100 μg/ml streptomycin (Gibco BRL, Basel, Switzerland), 50 μM 2-mercaptoethanol (Fluka, Buchs, Switzerland) and serially diluted compounds. Seven three-fold dilution steps in duplicates per test compound are performed. After four days of incubation 1 μCi 3H-thymidine is added. Cells are harvested after an additional five-hour incubation period, and incorporated 3H-thymidine is determined according to standard procedures. Background values (low control) of the MLR are the proliferation of BALB/c cells alone. Low controls are subtracted from all values. High controls without any sample are taken as 100% proliferation. Percent inhibition by the samples is calculated, and the concentrations required for 50% inhibition (IC50 values) are determined. In this assay, Compound of Example 56 has an IC50 of 168±20 nM.
  • B. In Vivo
  • Rat Heart Transplantation
  • The strain combination used: Male Lewis (RT1 haplotype) and BN (RT1 haplotype). The animals are anaesthetised using inhalational isofluorane. Following heparinisation of the donor rat through the abdominal inferior vena cava with simultaneous exsanguination via the aorta, the chest is opened and the heart rapidly cooled. The aorta is ligated and divided distal to the first branch and the brachiocephalic trunk is divided at the first bifurcation. The left pulmonary artery is ligated and divided and the right side divided but left open. All other vessels are dissected free, ligated and divided and the donor heart is removed into iced saline.
  • The recipient is prepared by dissection and cross-clamping of the infra-renal abdominal aorta and vena cava. The graft is implanted with end-to-side anastomoses, using 10/0 monofilament suture, between the donor brachiocephalic trunk and the recipient aorta and the donor right pulmonary artery to the recipient vena cava. The clamps are removed, the graft tethered retroabdominally, the abdominal contents washed with warm saline and the animal is closed and allowed to recover under a heating lamp. Graft survival is monitored by daily palpation of the beating donor heart through the abdominal wall. Rejection is considered to be complete when heart beat stops. Increases of graft survival are obtained in animals treated with a compound of formula I administered orally at a daily dose of 1 to 30 mg/kg. Thus compound of Example 100 significantly increases the graft survival when administered at a dose of 30 mg/kg/day.
  • The compounds of formula I are, therefore, useful in the treatment and/or prevention of diseases or disorders mediated by T lymphocytes and/or PKC, e.g. acute or chronic rejection of organ or tissue allo- or xenografts, atherosclerosis, vascular occlusion due to vacular injury such as angioplasty, restenosis, hypertension, heart failure, chronic obstructive pulmonary disease, CNS diseases such as Alzheimer disease or amyotrophic lateral sclerosis, cancer, infectious diseases such as AIDS, septic shock or adult respiratory distress syndrome, ischemia/reperfusion injury e.g. myocardial infarction, stroke, gut ischemia, renal failure or hemorrhage shock, or traumatic shock. The compounds of formula I are also useful in the treatment and/or prevention of T-cell mediated acute or chronic inflammatory diseases or disorders or autoimmune diseases e.g. rheumatoid arthritis, osteoarthritis, systemic lupus erythematosus, Hashimoto's thyroidis, multiple sclerosis, myasthenia gravis, diabetes type I or II and the disorders associated therewith, respiratory diseases such as asthma or inflammatory lung injury, inflammatory liver injury, inflammatory glomerular injury, cutaneous manifestations of immunologically-mediated disorders or illnesses, inflammatory and hyperproliferative skin diseases (such as psoriasis, atopic dermatitis, allergic contact dermatitis, irritant contact dermatitis and further eczematous dermatitises, seborrhoeic dermatitis), inflammatory eye diseases, e.g. Sjoegren's syndrome, keratoconjunctivitis or uveitis, inflammatory bowel disease, Crohn's disease or ulcerative colitis. For the above uses the required dosage will of course vary depending on the mode of administration, the particular condition to be treated and the effect desired. In general, satisfactory results are indicated to be obtained systemically at daily dosages of from about 0.1 to about 100 mg/kg body weight. An indicated daily dosage in the larger mammal, e.g. humans, is in the range from about 0.5 mg to about 2000 mg, conveniently administered, for example, in divided doses up to four times a day or in retard form.
  • The compounds of formula I may be administered by any conventional route, in particular enterally, e.g. orally, e.g. in the form of tablets or capsules, or parenterally, e.g. in the form of injectable solutions or suspensions, topically, e.g. in the form of lotions, gels, ointments or creams, or in a nasal or a suppository form. Pharmaceutical compositions comprising a compound of formula I in free form or in pharmaceutically acceptable salt form in association with at least one pharmaceutical acceptable carrier or diluent may be manufactured in conventional manner by mixing with a pharmaceutically acceptable carrier or diluent. Unit dosage forms for oral administration contain, for example, from about 0.1 mg to about 500 mg of active substance.
  • Topical administration is e.g. to the skin. A further form of topical administration is to the eye.
  • The compounds of formula I may be administered in free form or in pharmaceutically acceptable salt form e.g. as indicated above. Such salts may be prepared in conventional manner and exhibit the same order of activity as the free compounds.
  • In accordance with the foregoing the present invention further provides:
  • 1.1 A method for preventing or treating disorders or diseases mediated by T lymphocytes and/or PKC, e.g. such as indicated above, in a subject in need of such treatment, which method comprises administering to said subject an effective amount of a compound of formula I or a pharmaceutically acceptable salt thereof;
  • 1.2 A method for preventing or treating acute or chronic transplant rejection or T-cell mediated inflammatory or autoimmune diseases, e.g. as indicated above, in a subject in need of such treatment, which method comprises administering to said subject an effective amount of a compound of formula I or a pharmaceutically acceptable salt thereof;
  • 2. A compound of formula I, in free form or in a pharmaceutically acceptable salt form for use as a pharmaceutical, e.g. in any of the methods as indicated under 1.1 and 1.2 above.
  • 3. A pharmaceutical composition, e.g. for use in any of the methods as in 1.1 and 1.2 above comprising a compound of formula I in free form or pharmaceutically acceptable salt form in association with a pharmaceutically acceptable diluent or carrier therefor.
  • 4. A compound of formula I or a pharmaceutically acceptable salt thereof for use in the preparation of a pharmaceutical composition for use in any of the method as in 1.1 and 1.2 above.
  • Compounds of formula I may be administered as the sole active ingredient or together with other drugs in immunomodulating regimens or other anti-inflammatory agents e.g. for the treatment or prevention of allo- or xenograft acute or chronic rejection or inflammatory or autoimmune disorders. For example, they may be used in combination with cyclosporines, or ascomycines or their immunosuppressive analogs or derivatives, e.g. cyclosporin A, cyclosporin G, FK-506, ABT-281, ASM 981; an mTOR inhibitor, e.g. rapamycin, 40-O-(2-hydroxy)ethyl-rapamycin etc.; corticosteroids; cyclophosphamide; azathioprene; methotrexate; an accelerating lymphocyte homing agent, e.g. FTY 720; leflunomide or analogs thereof; mizoribine; mycophenolic acid; mycophenolate mofetil; 15-deoxysperguaiine or analogs thereof; immunosuppressive monoclonal antibodies, e.g., monoclonal antibodies. to leukocyte receptors, e.g., MHC, CD2, CD3, CD4, CD 11a/CD18, CD7, CD25, CD 27, B7, CD40, CD45, CD58, CD 137, ICOS, CD150 (SLAM), OX40, 4-1BB or their ligands, e.g. CD154; or other immunomodulatory compounds, e.g. a recombinant binding molecule having at least a portion of the extracellular domain of CTLA4 or a mutant thereof, e.g. an at least extracellular portion of CTLA4 or a mutant thereof joined to a non-CTLA4 protein sequence, e.g. CTLA4Ig (for ex. designated ATCC 68629) or a mutant thereof, e.g. LEA29Y, or other adhesion molecule inhibitors, e.g. mAbs or low molecular weight inhibitors including LFA-1 antagonists, Selectin antagonists and VLA-4 antagonists. Compounds of formula I may also be administered together with an antiproliferative drug, e.g. a chemotherapeutic drug, e.g. in cancer treatment, or with an anti-diabetic drug in diabetes therapy.
  • In accordance with the foregoing the present invention provides in a yet further aspect:
  • 5. A method as defined above comprising co-administration, e.g. concomitantly or in sequence, of a therapeutically effective amount of an inhibitor of PKC and of T-cell activation and proliferation, e.g. a compound of formula I in free form or in pharmaceutically acceptable salt form, and a second drug substance, said second drug substance being an immunosuppressant, immunomodulatory, anti-inflammatory, antiproliferative or anti-diabetic drug, e.g. as indicated above.
  • 6. A therapeutic combination, e.g. a kit, comprising a) an inhibitor of PKC and of T-cell activation and proliferation, e.g. a compound of formula I in free form or in pharma-ceutically acceptable salt form, and b) at least one second agent selected from an immunosuppressant, immunomodulatory, anti-inflammatory, antiproliferative and anti-diabetic drug. Component a) and component b) may be used concomitantly or in sequence. The kit may comprise instructions for its administration.
  • Where an inhibitor of PKC and of T-cell activation and proliferation, e.g. a compound of formula I, is administered in conjunction with other immunosuppressive/immunomodulatory, anti-inflammatory, antiproliferative or anti-diabetic therapy, e.g. for preventing or treating acute or chronic graft rejection or inflammatory or autoimmune disorders as hereinabove specified, dosages of the co-administered immunosuppressant, immunomodulatory, anti-inflammatory, antiproliferative or anti-diabetic compound will of course vary depending on the type of co-drug employed, e.g. whether it is a steroid or a cyclosporine, on the specific drug employed, on the condition being treated and so forth.
  • A preferred compound according to the invention is e.g. the compound of Example 56.

Claims (4)

1-11. (canceled)
12. A method for preventing or treating disorders or diseases mediated by lymphocytes and or PKC in a subject in need of such treatment, which method comprises administering to said subject an effective amount of a compound of formula I:
Figure US20070191401A1-20070816-C00038
wherein
Ra is H; C1-4alkyl; or C1-4alkyl substituted by OH, NH2, NHC1-4alkyl or N(C1-4alkyl)2; Rb is H or C1-4alkyl;
R is a radical of formula (b) or (c),
Figure US20070191401A1-20070816-C00039
wherein
each of R4 and R7 is OH; SH; NR16R17 wherein each of R16 and R17, independently, is H or C1-4alkyl or a radical of formula α

—X—Rc—Y  (α)
wherein X is a direct bond, O, S or NR18 wherein R18 is H or C1-4alkyl,
Rc is C1-4alkylene or C1-4alkylene wherein one CH2 is replaced by CRxRy wherein one of Rx and Ry is H and the other is CH3 each of Rx and Ry is CH3 or Rx and Ry form together —CH2—CH2—, and
Y is bound to the terminal carbon atom and is selected from OH and —NR19R20 wherein each of R19 and R20 independently is H,
C3-6cycloalkyl, C3-6cycloalkyl-C1-4alkyl, aryl-C1-4alkyl or C, 4alkyl optionally substituted on the terminal carbon atom by OH;
each of R5and R6, independently, is H, halogen, C1-4alkyl, CF3, OH, SH, NH2, C1-4alkoxy, C1-4alkylthio, NHC1-4alkyl, N(C1-4alkyl)2 or CN;
and
ring A is optionally substituted,
or a pharmaceutically acceptable salt thereof.
13. A method according to claim 12 of treating T cell mediated acute or chronic autoimmune or inflammatory diseases.
14. A method for the prevention or treatment of acute or chronic transplant rejection of organ or tissue allo- or xenografts in a subject in need of such treatment which comprises administering to said subject an effective amount of a compound according to claim 12.
US11/708,843 2000-11-07 2007-02-20 Indolylmaleimide derivatives Abandoned US20070191401A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US11/708,843 US20070191401A1 (en) 2000-11-07 2007-02-20 Indolylmaleimide derivatives
US12/498,486 US7786123B2 (en) 2000-11-07 2009-07-07 Indolylmaleimide derivatives

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US24640000P 2000-11-07 2000-11-07
US28370501P 2001-04-13 2001-04-13
US10/007,368 US6645970B2 (en) 2000-11-07 2001-11-05 Indolylmaleimide derivatives
US10/660,442 US7220774B2 (en) 2000-11-07 2003-09-11 Indolylmaleimide derivatives
US11/708,843 US20070191401A1 (en) 2000-11-07 2007-02-20 Indolylmaleimide derivatives

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/660,442 Division US7220774B2 (en) 2000-11-07 2003-09-11 Indolylmaleimide derivatives

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/498,486 Continuation US7786123B2 (en) 2000-11-07 2009-07-07 Indolylmaleimide derivatives

Publications (1)

Publication Number Publication Date
US20070191401A1 true US20070191401A1 (en) 2007-08-16

Family

ID=27358348

Family Applications (6)

Application Number Title Priority Date Filing Date
US10/007,368 Expired - Lifetime US6645970B2 (en) 2000-11-07 2001-11-05 Indolylmaleimide derivatives
US10/660,442 Expired - Lifetime US7220774B2 (en) 2000-11-07 2003-09-11 Indolylmaleimide derivatives
US11/708,843 Abandoned US20070191401A1 (en) 2000-11-07 2007-02-20 Indolylmaleimide derivatives
US11/708,840 Abandoned US20070155817A1 (en) 2000-11-07 2007-02-20 Indolylmaleimide derivatives
US11/708,841 Expired - Fee Related US7825124B2 (en) 2000-11-07 2007-02-20 Indolylmaleimide derivatives
US12/498,486 Expired - Fee Related US7786123B2 (en) 2000-11-07 2009-07-07 Indolylmaleimide derivatives

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US10/007,368 Expired - Lifetime US6645970B2 (en) 2000-11-07 2001-11-05 Indolylmaleimide derivatives
US10/660,442 Expired - Lifetime US7220774B2 (en) 2000-11-07 2003-09-11 Indolylmaleimide derivatives

Family Applications After (3)

Application Number Title Priority Date Filing Date
US11/708,840 Abandoned US20070155817A1 (en) 2000-11-07 2007-02-20 Indolylmaleimide derivatives
US11/708,841 Expired - Fee Related US7825124B2 (en) 2000-11-07 2007-02-20 Indolylmaleimide derivatives
US12/498,486 Expired - Fee Related US7786123B2 (en) 2000-11-07 2009-07-07 Indolylmaleimide derivatives

Country Status (1)

Country Link
US (6) US6645970B2 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7320643B1 (en) * 2006-12-04 2008-01-22 Harmonix Music Systems, Inc. Game controller simulating a musical instrument
US20080113797A1 (en) * 2006-11-15 2008-05-15 Harmonix Music Systems, Inc. Method and apparatus for facilitating group musical interaction over a network

Families Citing this family (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6645970B2 (en) * 2000-11-07 2003-11-11 Novartis Ag Indolylmaleimide derivatives
TWI324064B (en) * 2002-04-03 2010-05-01 Novartis Ag Indolylmaleimide derivatives
WO2005068455A1 (en) * 2004-01-19 2005-07-28 Novartis Ag Indolylmaleimde derivatives as pkc inhibitors
US20070232658A1 (en) * 2004-04-08 2007-10-04 Jurgen Wagner Protein Kinase C Inhibitors for the Treatment of Autoimmune Diseases and of Transplant Rejiction
GB0410713D0 (en) * 2004-05-13 2004-06-16 Novartis Ag Organic compounds
PL1904482T3 (en) * 2005-07-11 2011-06-30 Novartis Ag Indolylmaleimide derivatives
GB0613162D0 (en) * 2006-06-30 2006-08-09 Novartis Ag Organic compounds
US7820711B2 (en) * 2006-11-14 2010-10-26 Pharmacyclics Inc. Uses of selective inhibitors of HDAC8 for treatment of T-cell proliferative disorders
CA2671040A1 (en) * 2006-12-07 2008-06-19 Novartis Ag Use of pkc inhibitors in transplantation
MX2010003259A (en) * 2007-09-27 2010-04-12 Novartis Ag Drug monitoring assay.
BR112012024585A2 (en) * 2010-03-30 2016-05-31 Novartis Ag pkc inhibitors for treating b-cell lymphoma having chronic active b-cell receptor signaling
JP6437444B2 (en) 2012-11-29 2018-12-12 ノバルティス アーゲー Pharmaceutical combination
WO2014174478A1 (en) 2013-04-26 2014-10-30 Novartis Ag Pharmaceutical combinations of a pkc inhibitor and a c-met receptor tyrosine kinase inhibitor
JO3589B1 (en) 2014-08-06 2020-07-05 Novartis Ag Protein kinase c inhibitors and methods of their use
WO2018175302A1 (en) 2017-03-20 2018-09-27 Sienna Biopharmaceuticals, Inc. Polymer conjugates targeting c-src with reduced exposure
EP3600440A1 (en) 2017-03-20 2020-02-05 Sienna Biopharmaceuticals, Inc. Reduced exposure conjugates modulating therapeutic targets
CN110437092B (en) * 2019-07-11 2022-06-10 泓博智源(开原)药业有限公司 Preparation method of ticagrelor key intermediate aromatic cyclopropane amide

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5057614A (en) * 1988-02-10 1991-10-15 Hoffmann-La Roche Inc. Substituted pyrroles
US5399712A (en) * 1991-11-04 1995-03-21 Hoffmann-La Roche Inc. Process for the preparation of 2-substituted and 2,3-disubstituted maleimides
US5721245A (en) * 1989-02-23 1998-02-24 Hoffmann-La Roche Inc. 4- 3-indolyl!-1H-pyrrolone
US6040152A (en) * 1996-12-31 2000-03-21 National Jewish Medical And Research Center Method and assay for regulation of T cell proliferation
US6479490B2 (en) * 2000-07-27 2002-11-12 Syntex (U.S.A.) Llc 3-indolyl-4-phenyl-1H-pyrrole-2,5-dione derivatives as inhibitors of glycogen synthase kinase-3β
US6645970B2 (en) * 2000-11-07 2003-11-11 Novartis Ag Indolylmaleimide derivatives

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE4005969A1 (en) 1990-02-26 1991-08-29 Boehringer Mannheim Gmbh NEW TRISUBSTITUTED PYRROLE, METHOD FOR THE PRODUCTION THEREOF AND MEDICINAL PRODUCTS CONTAINING THESE COMPOUNDS
DE4005970A1 (en) 1990-02-26 1991-08-29 Boehringer Mannheim Gmbh NEW TRISUBSTITUTED MALEINIMIDES, METHOD FOR THE PRODUCTION THEREOF AND MEDICINAL PRODUCTS CONTAINING THESE COMPOUNDS
GB9828640D0 (en) 1998-12-23 1999-02-17 Smithkline Beecham Plc Novel method and compounds
AU6593600A (en) 1999-08-20 2001-03-19 Rei Asakai Drugs inhibiting cell death
TWI324064B (en) * 2002-04-03 2010-05-01 Novartis Ag Indolylmaleimide derivatives

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5057614A (en) * 1988-02-10 1991-10-15 Hoffmann-La Roche Inc. Substituted pyrroles
US5721245A (en) * 1989-02-23 1998-02-24 Hoffmann-La Roche Inc. 4- 3-indolyl!-1H-pyrrolone
US5399712A (en) * 1991-11-04 1995-03-21 Hoffmann-La Roche Inc. Process for the preparation of 2-substituted and 2,3-disubstituted maleimides
US6040152A (en) * 1996-12-31 2000-03-21 National Jewish Medical And Research Center Method and assay for regulation of T cell proliferation
US6479490B2 (en) * 2000-07-27 2002-11-12 Syntex (U.S.A.) Llc 3-indolyl-4-phenyl-1H-pyrrole-2,5-dione derivatives as inhibitors of glycogen synthase kinase-3β
US6645970B2 (en) * 2000-11-07 2003-11-11 Novartis Ag Indolylmaleimide derivatives

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080113797A1 (en) * 2006-11-15 2008-05-15 Harmonix Music Systems, Inc. Method and apparatus for facilitating group musical interaction over a network
US7758427B2 (en) * 2006-11-15 2010-07-20 Harmonix Music Systems, Inc. Facilitating group musical interaction over a network
US8079907B2 (en) 2006-11-15 2011-12-20 Harmonix Music Systems, Inc. Method and apparatus for facilitating group musical interaction over a network
US7320643B1 (en) * 2006-12-04 2008-01-22 Harmonix Music Systems, Inc. Game controller simulating a musical instrument

Also Published As

Publication number Publication date
US20030069424A1 (en) 2003-04-10
US20070184061A1 (en) 2007-08-09
US7786123B2 (en) 2010-08-31
US7220774B2 (en) 2007-05-22
US20040053949A1 (en) 2004-03-18
US6645970B2 (en) 2003-11-11
US20090318463A1 (en) 2009-12-24
US20070155817A1 (en) 2007-07-05
US7825124B2 (en) 2010-11-02

Similar Documents

Publication Publication Date Title
US7786123B2 (en) Indolylmaleimide derivatives
EP1337527B1 (en) Indolylmaleimide derivatives as protein kinase c inhibitors
AU2002221810A1 (en) Indolylmaleimide derivatives as protein kinase C inhibitors
US7781438B2 (en) Indolylmaleimide derivatives
EP1709030B1 (en) Indolylmaleimide derivatives
US8742132B2 (en) Indolylmaleimide derivatives processes for their production and pharmaceutical compositions
US7820672B2 (en) Indolylmaleimide derivatives
US7851475B2 (en) Substituted pyrrole-2, 5-diones as protein kinase C inhibitors

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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