Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
NOVEL INDOLINONE DERIVATIVES
Document Type and Number:
WIPO Patent Application WO/2006/105796
Kind Code:
A1
Abstract:
The invention relates to novel indolinone derivatives of formula (I), wherein X, R1, R2, R3, R4, R5, Re, Ri', Ri , FV/ R4'/ FV and R6' are as shown herein, as well as their use in the treatment of inflammatory autoimmune diseases, in particular by administration via the oral route.

Inventors:
DUVOLD LAETITIA MAUD ELYSA (DK)
HAVEZ SOPHIE ELISBETH (DK)
BINDERUP ERNST TORNDAL (DK)
ANDERSEN GRITH KROEYER (DK)
Application Number:
PCT/DK2006/000199
Publication Date:
October 12, 2006
Filing Date:
April 07, 2006
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
LEO PHARMA AS (DK)
DUVOLD LAETITIA MAUD ELYSA (DK)
HAVEZ SOPHIE ELISBETH (DK)
BINDERUP ERNST TORNDAL (DK)
ANDERSEN GRITH KROEYER (DK)
International Classes:
C07D209/34; A61K31/40; A61P29/00; C07D401/12; C07D401/14; C07D405/14
Domestic Patent References:
WO2003002107A22003-01-09
WO2005058309A12005-06-30
Foreign References:
US2533206A1950-12-12
EP0337628A21989-10-18
Other References:
LAI ET AL.: "Potent Small Inhibitors of Spleen Tyrosine Kinase (Syk)", BIOORG. & MED. CHEM. LETT., vol. 13, 2003, pages 3111 - 3114, XP002382305
ANDREANI A ET AL: "Synthesis and potential coanthracyclinic activity of pyridylmethylene and indolylmethylene lactams", EUROPEAN JOURNAL OF MEDICINAL CHEMISTRY, EDITIONS SCIENTIFIQUE ELSEVIER, PARIS, FR, vol. 33, no. 11, November 1998 (1998-11-01), pages 905 - 909, XP004160923, ISSN: 0223-5234
ANDREANI ET AL.: "Synthesis and Antitumor Activity of 1,5,6-Substituted E-3-(2-Chloro-3-indolylmethylene)-1,3-dihydroindol-2-ones", J.. MED. CHEM., vol. 45, 2002, pages 2666 - 2669, XP002382306
ANDREANI ET AL.: "Substituted E-3-(2-chloro-3-indolylmethylene)1,3-dihydroindol-2-ones with antitumor activity", BIOORG. & MED. CHEM., vol. 12, no. 5, 1 March 2004 (2004-03-01), pages 1121 - 1128, XP002382307
Attorney, Agent or Firm:
Thalsø-madsen, Birgit (Leo Pharma A/S Industriparken 55, Ballerup, DK)
Download PDF:
Claims:
CLAIMS
1. A compound of general formula I wherein X is O or S; R1 is hydrogen, OR9, OC(O)R9, NRi0Rn, C(O)NRi0RiI, 0C(0)NRκ>Rn, NHC(O)Ri0, NHC(O)ORi0, NHC(O)NRI0RU, S(O)R9, S(O)2R9, S(O)2OR9, S(O)2NRi0Rn, C(O)R12, C(O)ORi2, OC(O)ORi2, P(O)(ORI0)(ORU), OP(0)(ORiθ)(ORu), polyoxyethylene, C110 alkyl, C210 alkenyl, C210 alkynyl, aryl, carbocyclyl, heteroaryl or heterocyclyl, said C110 alkyl, C210 alkenyl, C210 alkynyl, aryl, carbocyclyl, heteroaryl or heterocyclyl being substituted with one or more substituents selected from the group consisting of OR10, C(O)R10, C(O)OR10, OC(O)R10, OC(O)OR10, NR10R11, C(O)NRi0Rn, OC(O)NR10R11, OP(O)(OR10)(OR11), P(O)(OR10)(OR11), NHC(O)R10, NHC(O)ORi0, NHC(O)NRi0Rn, SR10, S(O)R10, S(O)2R10, S(O)2NR10Rn, S(O)2OR10, polyoxyethylene, aryl, heteroaryl, carbocyclyl and heterocyclyl, said aryl, heteroaryl, carbocyclyl or heterocyclyl being optionally substituted with OR10, C(O)R10, C(O)OR10, OC(O)R10, OC(O)OR10, P(O)(OR10)(ORn), OP(O)(OR10)(OR11), NRi0Rn, C(O)NRi0Rn, NHC(O)R10, NHC(O)ORi0, NHC(O)NR10Rn, SRi0, S(O)Ri0, S(O)2Ri0, S(O)2NRi0Rn, S(O)2ORi0 or polyoxyethylene, and Ci6 alkyl substituted with polyoxyethylene, ORi0, C(O)Ri0, C(O)ORi0, OC(O)Ri0, OC(O)ORi0, NRi0Rn, C(O)NRi0Rn, P(O)(ORi0)(ORn), OP(O)(ORi0)(ORiI), NHC(O)R10, NHC(O)OR10, NHC(O)NR10Rn, SR10, S(O)R10, S(O)2Ri0, "S(O)2NR10R11 or S(O)2OR10, wherein R9 is Ci10 alkyl, C2I0 alkenyl, C2I0 alkynyl, aryl, heteroaryl, carbocyclyl or heterocyclyl, said Ci10 alkyl, C210 alkenyl, C210 alkynyl, aryl, heteroaryl, carbocyclyl or heterocyclyl being optionally substituted with one or more substituents selected from the group consisting of halogen, trihalomethyl, C16 alkyl, C26 alkenyl, C26 alkynyl, hydroxy, carboxy, formyl, aryl, heteroaryl, carbocyclyl, heterocyclyl, amino, carbamoyl, cyano, guanidino, carbamido, polyoxyethylene, OR13, C(O)R13, C(O)OR13, OC(O)Ri3, OC(O)OR13, NR13R14, C(O)NR13Ri4, OC(O)NR13Ri4, NHC(O)R13, NHC(O)OR13, NHC(O)NR13R14, P(O)(OR13)(OR14), OP(O)(OR13)(OR14), SR13, S(O)R13, S(O)2R13, S(O)2NR13R14 and S(O)2OR13; R10 and R11 are the same or different and independently selected from the group consisting of hydrogen, polyoxyethylene, C1I0 alkyl, C210 alkenyl, C210 alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl, or wherein R10 and R11, together with the nitrogen atom to which they are attached form a heterocyclic or heteroaryl ring, each of C110 alkyl, C210 alkenyl, C210 alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl being optionally substituted with one or more, same or different substituents selected from the group consisting of hydrogen, halogen, trihalomethyl, C16 alkyl, C26 alkenyl, C26 alkynyl, hydroxy, carboxy, formyl, aryl, heteroaryl, carbocyclyl, heterocyclyl, amino, carbamoyl, cyano, guanidino, carbamido, polyoxyethylene, OR13, C(O)R13, C(O)OR13, OC(O)R13, OC(O)OR13, N3, NR13R14, C(O)NR13R14, OC(O)NR13Ri4, NHC(O)Ri3, NHC(O)ORi3, NHC(O)NR13Ri4, P(O)(OR13)(OR14), OP(O)(OR13)(OR14), SR13, S(O)R13, S(O)2R13, S(O)2NR13R14 and S(O)2OR14, said aryl, heteroaryl, carbocyclyl or heterocyclyl being optionally substituted with one or more substituents selected from the group consisting of OR13, C(O)Ri3, C(O)ORi3, C(O)NRi3Ri4 and polyoxyethylene, provided that Ri0 and Ru are not both hydrogen; Ri2 is C110 alkyl, C210 alkenyl, C210 alkynyl, aryl or carbocyclyl substituted with hydroxy, amino, cyano, halogen, carboxy, nitro, amido, polyoxyethylene, OR13, C(O)R13, C(O)OR13, OC(O)R13, OC(O)OR13, NR13R14, C(O)NR13R14, OC(O)NR13Ri4, NHC(O)R13, NHC(O)ORi3, NHC(O)NRi3R14, P(O)(OR13)(ORi4), OP(O)(ORi3)(ORi4), SR13, S(O)R13, S(O)2R13, S(O)2NR13R14 and S(O)2OR14, or heteroaryl or heterocyclyl optionally substituted with hydroxy, amino, cyano, halogen, carboxy, nitro, amido, polyoxyethylene, OR13, C(O)R13, C(O)OR13, OC(O)R13, OC(O)OR13, NR13R14, C(O)NR13R14, OC(O)NR13R14, P(O)(OR13)(OR14), OP(O)(OR13)(OR14), NHC(O)R13, NHC(O)OR10, NHC(O)NR10R11, SR13, S(O)R13, S(O)2R13, S(O)2NR13R14 and S(O)2OR14; R13 and R14 are the same or different and independently selected from the group consisting of hydrogen, C16alkyl, C26alkenyl, C46alkadienyl, C26alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl, or wherein R13 and R14, together with the nitrogen atom to which they are attached form a heterocyclic or heteroaryl ring, each C1 6 alkyl, C26 alkenyl, C26 alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl substituent being optionally substituted with one or more, same or different substituents selected from the group consisting of hydrogen, hydroxy, C14 alkyl, Ci4 alkoxy, nitro, cyano, amino, oxo, halogen, trihalomethyl, Ci4 alkylthio, C14 alkylamino, Ci4 alkoxycarbonyl, carboxy, CONH2 or S(O)2NH2; R2, R3, R4 and R5 are the same or different and independently selected from the group consisting of hydrogen, halogen, trihalomethyl, CiI0 alkyl/ C2I0 alkenyl, C2I0 alkynyl, hydroxy, carboxy, formyl, aryl, heteroaryl, carbocyclyl, heterocyclyl, amino, carbamoyl, cyano, guanidino, carbamido, ORi5, C(O)Ri5, C(O)ORi5, OC(O)Ri5, OC(O)ORi5, NRi5Ri6, C(O)NRi5Ri6, OC(O)NRi5Ri6, NHC(O)R15, NHC(O)ORi5, NHC(O)NRi5Ri6, SRi5, S(O)Ri5, S(O)2Ri5, S(O)2NR15R16 and S(O)2OR16, wherein R15 and R16 are the same or different and independently selected from the group consisting of hydrogen, C1 I0 alkyl, C210 alkenyl, C210 alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl, or wherein R15 and R16, together with the nitrogen atom to which they are attached form a heterocyclic or heteroaryl ring, said C110 alkyl, C210 alkenyl, C210 alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl being optionally substituted with one or more, same or different substituents selected from the group consisting of hydrogen, halogen, trihalomethyl, C16alkyl, C26alkenyl, C26alkynyl, hydroxy, carboxy, formyl, aryl, heteroaryl, carbocyclyl, heterocyclyl, amino, carbamoyl, cyano, guanidino, carbamido, OR17, C(O)R17, C(O)OR17, OC(O)OR17, OC(O)R18, NR17R18, C(O)NR17R18, OC(O)NR17R18, NHC(O)R17, NHC(O)OR17, NHC(O)NR17R18, SR17, S(O)R17, S(O)2R17, S(O)2NR17Ri8 and S(O)2ORi7, wherein R17 and R18 are the same or different and independently selected from the group consisting of hydrogen, C16alkyl, C2.6alkenyl, C26alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl, or wherein R17 and R18, together with the nitrogen atom to which they are attached form a heterocyclic or heteroaryl ring, each C16alkyl, C26alkenyl, C26alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl substituent being optionally substituted with one or more, same or different substituents selected from the group consisting of hydrogen, hydroxy, C14 alkyl, C14 alkoxy, nitro, cyano, amino, oxo, halogen, trihalomethyl, C14 alkylthio, C14 alkylamino, C14 alkoxycarbonyl, carboxy, CONH2 or S(O)2NH2; R6 is hydrogen, C16 alkyl, cycloalkyl, aryl, heteroaryl, heterocyclyl, halogen, OR7, C(O)R7, C(O)OR7, NR7R8, S(O)2NR7R8, wherein R7 and R8 are independently hydrogen, C16 alkyl, aryl or heterocyclyl, said C16 alkyl or heterocyclyl being optionally substituted by heterocyclyl, OR7, C(O)R7 or C(O)OR7; Ri', R3', R4', R5' and R5' are the same or different and independently selected from the group consisting of hydrogen, halogen, trihalomethyl, Ci10 alkyl, C2I0 alkenyl, C2I0 alkynyl, hydroxy, carboxy, formyl, aryl, heteroaryl, carbocyclyl, heterocyclyl, amino, carbamoyl, cyano, guanidino, carbamido, ORi5, C(O)Ri5, C(O)ORi5, OC(O)Ri5, OC(O)ORi5, NRi5Ri6, C(O)NRi5Ri6, OC(O)NR15Ri6, NHC(O)Ri5, NHC(O)ORi5, NHC(O)NRi5Ri6, SR15, S(O)R15, S(O)2Ri5, S(O)2NRi5R16 and S(O)2ORi6, wherein R15 and Ri6 are the same or different and independently selected from the group consisting of hydrogen, CiI0 alkyl, C2io alkenyl, C2I0 alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl, or wherein Ri5 and R16, together with the nitrogen atom to which they are attached form a heterocyclic or heteroaryl ring, said CiI0 alkyl, C2I0 alkenyl, C2I0 alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl being optionally substituted with one or more, same or different substituents selected from the group consisting of hydrogen, halogen, trihalomethyl, Ci6alkyl, C26alkenyl, C26alkynyl, hydroxy, carboxy, formyl, aryl, heteroaryl, carbocyclyl, heterocyclyl, amino, carbamoyl, cyano, guanidino, carbamido, ORi7, C(O)Ri7, C(O)ORi7, OC(O)Ri8, OC(O)ORi7, NRi7Ri8, C(O)NRi7Ri8, OC(O)NRi7Ri8, NHC(O)Ri7, NHC(O)ORi7, NHC(O)NRi7Ri8, SR17, S(O)Ri7, S(O)2Ri7, S(O)2NR17R18 and S(O)2OR17, wherein R17 and Ri8 are the same or different and independently selected from the group consisting of hydrogen, Ci6alkyl, C26alkenyl, C26alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl, or wherein Ri7 and Ri8, together with the nitrogen atom to which they are attached form a heterocyclic or heteroaryl ring, each C16alkyl, C26alkenyl, C26alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl substituent being optionally substituted with one or more, same or different substituents selected from the group consisting of hydrogen, hydroxy, C14 alkyl, C14 alkoxy, nitro, cyano, amino, oxo, halogen, trihalomethyl, C14 alkylthio, C14 alkylamino, C14 alkoxycarbonyl, carboxy, CONH2 or S(O)NH2; and R2' is hydrogen, , OR9, OC(O)R9, NR10R11, C(O)NR10R11, OC(O)NR10Ru, NHC(O)R10, NHC(O)OR10, NHC(O)NR10R11, S(O)R9, S(O)2R9, S(O)2OR9, S(O)2NR10R11, C(O)R19, C(O)OR19, OC(O)OR19, P(O)(OR10)(OR11), OP(O)(OR10)(OR11), polyoxyethylene, C110 alkyl, C210 alkenyl, C210 alkynyl, aryl, carbocyclyl, heteroaryl or heterocyclyl, said CiI0 alkyl, C2I0 alkenyl, C2I0 alkynyl, aryl, carbocyclyl, heteroaryl or heterocyclyl being substituted with one or more substituents selected from the group consisting of OR10, C(O)R10, C(O)OR10, OC(O)R10, OC(O)OR10, NR10Rn, C(O)NR10R11, OC(O)NR10R11, OP(0)(ORio)(ORu), P(O)(OR10)(OR11), NHC(O)R10, NHC(O)OR10, NHC(O)NR10R11, SR10, S(O)R10, S(O)2R10, S(O)2NR10R11, S(O)2OR10, polyoxyethylene, aryl, heteroaryl, carbocyclyl and heterocyclyl, said aryl, heteroaryl, carbocyclyl or heterocyclyl being optionally substituted with ORi0, C(O)Ri0, C(O)ORi0, OC(O)Ri0, OC(O)OR10, P(O)(OR10)(OR11), OP(O)(ORi0)(ORu), NRi0Ru, C(O)NRi0Rn, NHC(O)Ri0, NHC(O)ORi0, NHC(O)NRi0R1I, SRi0, S(O)Ri0, S(O)2Ri0, S(O)2NR10Ru, S(O)2OR10 or polyoxyethylene, and C16 alkyl substituted with polyoxyethylene, OR10, C(O)R10, C(O)OR10, OC(O)R10, OC(O)OR10, NR10R11, C(O)NR10R11, P(O)(OR10)(OR11), OP(O)(OR10)(OR11), NHC(O)R10, NHC(O)OR10, NHC(O)NR10R11, SR10, S(O)R10, S(O)2R10, S(O)2NR10R11 or S(O)2OR10, wherein R9 is C110 alkyl, C210 alkenyl, C210 alkynyl, aryl, heteroaryl, carbocyclyl or heterocyclyl, said C110 alkyl, C210 alkenyl, C210 alkynyl, aryl, heteroaryl, carbocyclyl or heterocyclyl being optionally substituted with one or more substituents selected from the group consisting of halogen, trihalomethyl, C16 alkyl, C26 alkenyl, C26 alkynyl, hydroxy, carboxy, formyl, aryl, heteroaryl, carbocyclyl, heterocyclyl, amino, carbamoyl, cyano, guanidino, carbamido, polyoxyethylene, OR13, C(O)R13, C(O)OR13, OC(O)R13, OC(O)OR13, NR13R14, C(O)NR13R14, OC(O)NR13R14, NHC(O)R13, NHC(O)OR13, NHC(O)NR13R14, P(O)(OR13)(OR14), OP(O)(OR13)(OR14), SR13, S(O)R13, S(O)2R13, S(O)2NR13R14 and S(O)2OR13; R10 and R11 are the same or different and independently selected from the group consisting of hydrogen, polyoxyethylene, C110 alkyl, C210 alkenyl, C210 alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl, or wherein R10 and Rn, together with the nitrogen atom to which they are attached form a heterocyclic or heteroaryl ring, each of C110 alkyl, C210 alkenyl, C210 alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl being optionally substituted with one or more, same or different substituents selected from the group consisting of hydrogen, halogen, trihalomethyl, C16 alkyl, C26 alkenyl, C26 alkynyl, hydroxy, carboxy, formyl, aryl, heteroaryl, carbocyclyl, heterocyclyl, amino, carbamoyl, cyano, guanidino, carbamido, polyoxyethylene, OR13, C(O)R13, C(O)OR13, OC(O)R13, OC(O)OR13, N3, NR13R14, C(O)NR13R14, OC(O)NR13R14, NHC(O)R13, NHC(O)OR13, NHC(O)NR13R14, P(O)(OR13)(OR14), OP(O)(OR13)(OR14), SR13, S(O)R13, S(O)2R13, S(O)2NR13R14 and S(O)2OR14, said aryl, heteroaryl, carbocyclyl or heterocyclyl being optionally substituted with one or more substituents selected from the group consisting of OR13, C(O)R13, C(O)OR13, C(O)NR13R14 and polyoxyethylene, provided that R10 and R11 are not both hydrogen; R19 is C110 alkyl, C210 alkenyl, C210 alkynyl, aryl, heteroaryl, carbocyclyl or heterocyclyl, said C110 alkyl, C210 alkenyl, C210 alkynyl, aryl, heteroaryl, carbocyclyl or heterocyclyl being substituted with one or more substituents selected from the group consisting of substituted with one or more, same or different substituents selected from the group consisting of hydroxy, amino, cyano, halogen, carbocy, nitro, amido, polyoxyethylene, OR13, C(O)R13, C(O)OR13, OC(O)R13, OC(O)ORi3, NR13R14, C(O)NR13R14, OC(O)NR13R14, NHC(O)R13, NHC(O)OR13, NHC(O)NR13R14, SR13, S(O)R13, S(O)2R13, S(O)2NR13R14 and S(O)2OR14; R13 and R14 are the same or different and independently selected from the group consisting of hydrogen, C16alkyl, C25alkenyl, C46alkadienyl, C26alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl, or wherein R13 and R14, together with the nitrogen atom to which they are attached form a heterocyclic or heteroaryl ring, each C1. 6 alkyl, C26 alkenyl, C26 alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl substituent being optionally substituted with one or more, same or different substituents selected from the group consisting of hydrogen, hydroxy, C14 alkyl, Ci4 alkoxy, nitro, cyano, amino, oxo, halogen, trihalomethyl, C14 alkylthio, C14 alkylamino, C14 alkoxycarbonyl, carboxy, CONH2 or S(O)2NH2; provided that R1 and R2' are not both hydrogen and that, when R1 is other than hydrogen, R2' may also be heterocyclyl, heteroaryl, C(O)R20, C(O)OR20, S(O)2R20 or C16 alkyl optionally substituted with heterocyclyl, heteroaryl or C(O)OR20, wherein R20 is hydrogen, C16 alkyl, aryl, heteroaryl or heterocyclyl, and that, when R2' is other than hydrogen, R1 may also be heterocyclyl, heteroaryl, C(O)R20, C(O)OR20, S(O)2R20 or C1 6 alkyl optionally substituted with heterocyclyl, heteroaryl or C(O)OR20, wherein R20 is hydrogen, Ci6 alkyl, aryl, heteroaryl or heterocyclyl; or pharmaceutically acceptable salts thereof.
2. A compound according to claim 1, wherein R2, R3, R4 and R5 are all hydrogen.
3. A compound according to claim 1, wherein R1 is OR9, OC(O)R9, C(O)NR10R11, OC(O)NR10R11, S(O)2R9, C(O)R12, C(O)OR12, OC(O)OR12, P(O)(OR10)(OR11), OP(O)(ORiO)(OR11), polyoxyethylene, C110 alkyl, C210 alkenyl, C210 alkynyl or aryl, said C110 alkyl, C210 alkenyl, C210 alkynyl or aryl being substituted with one or more substituents selected from the group consisting of OR10, C(O)R10, C(O)OR10, OC(O)R10, OC(O)OR10, NR10R11, C(O)NR10Rn, OC(O)NR10R1^OP(O)(OR10)(OR11), P(O)(OR10)(OR11), NHC(O)R10, NHC(O)OR10, NHC(O)NR10Rn, SR10, S(O)R10, S(O)2R10, S(O)2NR10Rn, S(O)2OR10, polyoxyethylene, heteroaryl and heterocyclyl, said heteroaryl and heterocyclyl being optionally substituted with OR10, C(O)R10, C(O)OR10, OC(O)R10, OC(O)OR10, P(O)(OR10)(ORu), OP(O)(OR10)(OR11), NR10R11, C(O)NR10Rn, NHC(O)R10, NHC(O)OR10, NHC(O)NR10Rii, "SRi0, S(O)R10, S(O)2R10, S(O)2NR10R11, S(O)2OR10 or polyoxyethylene, and Cx6 alkyl substituted with OR10, C(O)R10, C(O)OR10, OC(O)R10, OC(O)OR10, NR10R11, C(O)NR10Rn, P(O)(OR10)(OR11), OP(O)(OR10)(OR11), NHC(O)R10, NHC(O)OR10, NHC(O)NR10R11, SR10, S(O)Ri0, S(O)2Ri0, S(0)2NRioR.ii or S(O)2ORi0, wherein R9, Ri0, Ru and R12 are as indicated in claim 1.
4. A compound according to claim 3, wherein R2' is hydrogen.
5. A compound according to claim 3 or 4, wherein Ri is OR9, OC(O)R9, C(O)Ri2 or C1I0 alkyl, said C1I0 alkyl being substituted with one or more substituents selected from the group consisting of polyoxyethylene, OR10, C(O)R10, C(O)OR10, OC(O)R10, OC(O)OR10, NR10R11, C(O)NR10R11, OC(O)NR10RiIrOP(O)(OR10)(OR11), P(O)(OR10)(OR11), NHC(O)R10, NHC(O)OR10, NHC(O)NR10R11, SR10, S(O)R10, S(O)2R10, S(O)2NR10R11, S(O)2OR10 wherein R9, R10, R11 and R12 are as indicated in claim 1.
6. A compound according to any one of claims 35, wherein R1 is wherein R10, R10' and R11 are the same or different and independently selected from the group consisting of hydrogen, C110 alkyl, C210 alkenyl, C210 alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl, or wherein R10 and R11, together with the nitrogen atom to which they are attached form a heterocyclic or heteroaryl ring, each of C110 alkyl, C210 alkenyl, C210 alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl being optionally substituted with one or more, same or different substituents selected from the group consisting of hydrogen, halogen, trihalomethyl, C16 alkyl, C26 alkenyl, C26 alkynyl, hydroxy, carboxy, formyl, aryl, heteroaryl, carbocyclyl, heterocyclyl, amino, carbamoyl, cyano, guanidino, carbamido, OR13, C(O)R13, C(O)OR13, OC(O)R13, OC(O)OR13, NR13R14, C(O)NR13R14, OC(O)NR13R14, NHC(O)R13, NHC(O)OR13, NHC(O)NR13R14, P(O)(OR13)(OR14), OP(O)(OR13)(OR14), SR13, S(O)R13, S(O)2R13, S(O)2NR13R14 and S(O)2ORi4, wherein R13 and R14 are as indicated in claim 1, provided that R10 and R11 are not both hydrogen.
7. A compound according to any one of claims 35, wherein R1 is wherein R10 and Ri0' are the same or different and independently selected from the group consisting of hydrogen, C110 alkyl, C210 alkenyl, C210 alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl, each of C110 alkyl, C210 alkenyl, C210 alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl being optionally substituted with one or more, same or different substituents selected from the group consisting of hydrogen, halogen, trihalomethyl, C16 alkyl, C26 alkenyl, C26 alkynyl, hydroxy, carboxy, formyl, aryl, heteroaryl, carbocyclyl, heterocyclyl, amino, carbamoyl, cyano, guanidino, carbamido, OR13, C(O)R13, C(O)OR13, OC(O)R13, OC(O)OR13, NR13R14, C(O)NR13R14, OC(O)NR13R14, NHC(O)R13, NHC(O)OR13, NHC(O)NR13R14, P(O)(OR13)(OR14), OP(O)(OR13)(OR14), SR13, S(O)R13, S(O)2R13, S(O)2NR13R14 and S(O)2OR14, wherein R13 and R14 are as indicated in claim 1.
8. A compound according to any one of claims 37, wherein R2' is S(O)2R9, C(O)R19, C(O)OR19, C110 alkyl, C210 alkenyl, C210 alkynyl or aryl, said C110 alkyl, C210 alkenyl, C2. io alkynyl or aryl being substituted with one or more substituents selected from the group consisting of OR10, C(O)R10, C(O)OR10, OC(O)R10, OC(O)OR10, NR10R11, P(O)(OR10)(OR11), OP(O)(OR10)(OR11), C(O)NR10R11, OC(O)NR10R11, NHC(O)R10, NHC(O)OR10, NHC(O)NR10R11, SR10, S(O)R10, S(O)2R10, S(O)2NR10R11, S(O)2OR10, polyoxyethylene, aryl, heteroaryl, carbocyclyl and heterocyclyl, said aryl, heteroaryl, carbocyclyl and heterocyclyl being optionally substituted with OR10, C(O)R10, C(O)OR10, OC(O)R10, OC(O)OR10, NR10R11, C(O)NR10R11, NHC(O)R10, NHC(O)OR10, NHC(O)NR10R11, SR10, S(O)R10, S(O)2R10, S(O)2NR10R11, S(O)2OR10 or polyoxyethylene, and C16 alkyl substituted with OR10, C(O)R10, C(O)OR10, OC(O)R10, OC(O)OR10, NR10R11, C(O)NR10R11, P(O)(OR10)(OR11), OP(O)(OR10)(OR11), NHC(O)R10, NHC(O)OR10, NHC(O)NR10R11, SR10, S(O)R10, S(O)2R10, S(O)2NR10R11 or S(O)2OR10, wherein R9, R19, R10 and R11 are as indicated in claim 1.
9. A compound according to claim 1, wherein R2' is S(O)2R9, C(O)R19, C110 alkyl, said C110 alkyl being substituted with one or more substituents selected from the group consisting of OR10, C(O)R10, C(O)OR10, OC(O)R10, OC(O)OR10, NR10R11, C(O)NR10R11, OC(O)NR10R11, NHC(O)R10, NHC(O)OR10, NHC(O)NR10R11, SR10, S(O)R10, S(O)2R10, S(O)2NR10Rn, S(O)2OR10, aryl, heteroaryl, carbocyclyl and heterocyclyl optionally substituted with OR10, C(O)R10, C(O)OR10, NR10R11, C(O)NR10R11, NHC(O)R10, NHC(O)OR10, NHC(O)NR10R11, SR10, S(O)R10, S(O)2R10, S(O)2NR10R11 or S(O)2OR10, and C16 alkyl substituted with OR10, C(O)R10, C(O)OR10, OC(O)R10, OC(O)OR10, NR10R11, C(O)NR10R11, OC(O)NR10R11, NHC(O)R10, NHC(O)OR10, NHC(O)NRIORII, SR10, S(O)R10, S(O)2R10, S(O)2NR10R11 and S(O)2OR10, wherein R9, R19, R10 and R11 are as indicated in claim 1.
10. A compound according to claim 9, wherein R1 is hydrogen.
11. A compound according to claim 9 or 10, wherein R2' is wherein R10, R10' and R11 are the same or different and independently selected from the group consisting of hydrogen, C110 alkyl, C210 alkenyl, C210 alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl, or wherein R10 and R11, together with the nitrogen atom to which they are attached form a heterocyclic or heteroaryl ring, each of C110 alkyl, C210 alkenyl, C210 alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl being optionally substituted with one or more, same or different substituents selected from the group consisting of hydrogen, halogen, trihalomethyl, C16 alkyl, C26 alkenyl, C26 alkynyl, hydroxy, carboxy, formyl, aryl, heteroaryl, carbocyclyl, heterocyclyl, amino, carbamoyl, cyano, guanidino, carbamido, OR13, C(O)R13, C(O)OR13, OC(O)R13, OC(O)OR13, NR13R14, C(O)NR13R14, OC(O)NR13R14, NHC(O)R13, NHC(O)OR13, NHC(O)NR13R14, P(O)(OR13)(OR14), OP(O)(OR13)(OR14), SR13, S(O)R13, S(O)2R13, S(O)2NR13R14 and S(O)2OR14, wherein R13 and R14 are as indicated in claim 1, provided that R10 and R11 are not both hydrogen.
12. A compound according to claim 9 or 10, wherein R2' is wherein R10 and R10' are the same or different and independently selected from the group consisting of hydrogen, C110 alkyl, C210 alkenyl, C210 alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl, each of C110 alkyl, C210 alkenyl, C210 alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl being optionally substituted with one or more, same or different substituents selected from the group consisting of hydrogen, halogen, trihalomethyl, C16 alkyl, C26 alkenyl, C26 alkynyl, hydroxy, carboxy, formyl, aryl, heteroaryl, carbocyclyl, heterocyclyl, amino, carbamoyl, cyano, guanidino, carbamido, OR13, C(O)R13, C(O)OR13, OC(O)R13, OC(O)OR13, NR13R14, C(O)NR13R14, OC(O)NR13R14, NHC(O)R13, NHC(O)OR13, NHC(O)NR13R14, P(O)(OR13)(OR14), OP(O)(OR13)(OR14), SR13, S(O)R13, S(O)2R13, S(O)2NR13R14 and S(O)2OR14, wherein R13 and R14 are as indicated in claim 1.
13. A compound according to any one of claims 911, wherein R2' is wherein R10, R10' and R11 are the same or different and independently selected from the group consisting of hydrogen, C110 alkyl, heteroaryl, carbocyclyl and heterocyclyl, or wherein R10 and R11, together with the nitrogen atom to which they are attached form a heterocyclic or heteroaryl ring, each of C110 alkyl, heteroaryl, carbocyclyl and heterocyclyl being optionally substituted with one or more, same or different substituents selected from the group consisting of hydrogen, halogen, trihalomethyl, C1, alkyl, hydroxy, carboxy, formyl, aryl, heteroaryl, carbocyclyl, heterocyclyl, amino, carbamoyl, cyano, guanidino, carbamido, OR13, C(O)R13, C(O)OR13, NR13R14, C(O)NR13R14, NHC(O)R13, NHC(O)OR13, NHC(O)NR13R14, P(O)(OR13)(OR14), OP(O)(OR13)(OR14), SR13, S(O)2R13, S(O)2NR13R14 and S(O)2OR14, wherein R13 and R14 are as indicated in claim 1, provided that R10 and R11 are not both hydrogen.
14. A compound according to any one of claims 9, 10, and 12, wherein R2' is wherein R10 and R10' are the same or different and independently selected from the group consisting of hydrogen, C110 alkyl, heteroaryl, carbocyclyl and heterocyclyl, each of C110 alkyl, heteroaryl, carbocyclyl and heterocyclyl being optionally substituted with one or more, same or different substituents selected from the group consisting of hydrogen, halogen, trihalomethyl, C16 alkyl, hydroxy, carboxy, formyl, aryl, heteroaryl, carbocyclyl, heterocyclyl, amino, carbamoyl, cyano, guanidino, carbamido, OR13, C(O)R13, C(O)OR13, NR13R14, C(O)NR13Ri4, NHC(O)R13, NHC(O)OR13, NHC(O)NR13R14, P(O)(OR13)(OR14), OP(O)(OR13)(OR14), SR13, S(O)2R13, S(O)2NR13R14 and S(O)2ORi4, wherein Ri3 and Ri4 are as indicated in claim 1, provided that Ri0 and Rn are not both hydrogen.
15. A compound according to any one of claims 9, 11, 12, 13 and 14, wherein Ri is wherein Ri0, Rio' and Ru are the same or different and independently selected from the group consisting of hydrogen, CiI0 alkyl, C210 alkenyl, C2io alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl, or wherein Ri0 and Rn, together with the nitrogen atom to which they are attached form a heterocyclic or heteroaryl ring, each of Ciio alkyl, aryl, heteroaryl, carbocyclyl and heterocyclyl being optionally substituted with one or more, same or different substituents selected from the group consisting of hydrogen, halogen, trihalomethyl, Ci6 alkyl, C26 alkenyl, C26 alkynyl, hydroxy, carboxy, formyl, aryl, heteroaryl, carbocyclyl, heterocyclyl, amino, carbamoyl, cyano, guanidino, carbamido, ORi3, C(O)Ri3, C(O)ORi3, OC(O)R13, OC(O)ORi3, NRi3Ri4, C(O)NR13R14, OC(O)NRi3Ri4, NHC(O)Ri3, NHC(O)OR13, NHC(O)NRi3Ri4, P(O)(ORi3)(ORi4), OP(O)(OR13)(ORi4), SRi3, S(O)R13, S(O)2R13, S(O)2NR13R14 and S(O)2OR14, wherein Ri3 and Ri4 are as indicated in claim 1, provided that Ri0 and Rn are not both hydrogen.
16. A compound according to any one of claims 9, 11, 12, 13 and 14, wherein Ri is wherein R10 and Ri0' are the same or different and independently selected from the group consisting of hydrogen, Cx10 alkyl, aryl, heteroaryl, carbocyclyl and heterocyclyl, each of C110 alkyl, aryl, heteroaryl, carbocyclyl and heterocyclyl being optionally substituted with one or more, same or different substituents selected from the group consisting of hydrogen, halogen, trihalomethyl, C16 alkyl, hydroxy, carboxy, formyl, aryl, heteroaryl, carbocyclyl, heterocyclyl, amino, carbamoyl, cyano, guanidino, carbamido, OR13, C(O)R13, C(O)OR13, OC(O)R13, OC(O)OR13, NR13R14, C(O)NR13R14, OC(O)NR13R14, NHC(O)R13, NHC(O)OR13, NHC(O)NR13R14, P(O)(OR13)(OR14), OP(O)(OR13)(OR14), SR13, S(O)R13, S(O)2R13, S(O)2NR13R14 and S(O)2OR14, wherein R13 and R14 are as indicated in claim 1, provided that R10 and R10' are not both hydrogen.
17. A compound according to claim 1, wherein R6 is hydrogen.
18. A compound according to claim 1, wherein R1', R3', R4', R5' and R6' are the same or different and independently selected from the group consisting of hydrogen, Cx10 alkyl, C110 alkoxy, aryl, heteroaryl, aryloxy, C110 alkylaryl, C110 alkylaryloxy, halogen, trihalomethyl, S(O)R20, S(O)2R20, S(O)2NR20R21, S(O)3R20, SR20, NO2, NR20R21, OR20, CN, CH2OH, C(O)R20, C(O)OR20, OC(O)R20, NHC(O)R20, (CH2)nC(O)2R20 and C(O)NR20R21, wherein R20 is hydrogen, C16 alkyl, heteroaryl or aryl, said C16 alkyl, heteroaryl or aryl being optionally substituted with hydroxy or NR22R23, wherein R22 and R23 are independently hydrogen or C16 alkyl or, together with the nitrogen atom to which they are attached, form a heteroaryl or heterocyclic ring, R21 is hydrogen, C16 alkyl or aryl, and n is 03.
19. A compound according to claim 1 and 18, wherein R1', R3', R4', R5' and R6' are the same or different and independently selected from the group consisting of hydrogen, C16 alkyl, C16 alkoxy and halogen.
20. A compound according to claim 1 selected from the group consisting of carbonic acid 6azidohexyl ester 3(2oxol,2dihydroindol3ylidenemethyl)indoll yl methyl ester carbonic acid 6azidohexyl ester 3(2oxolpiperidinlylmethyll,2dihydroindol3 ylidenemethyl)indollylmethyl ester 4{2[3(2oxol,2dihydroindol3ylidenemethyl)indollylmethoxycarbonyloxy] ethyl}piperidinelcarboxylic acid tertbutyl ester 4{2[3(2oxolpiperidinlylmethyll,2dihydroindol3ylidenemethyl)indoll ylmethoxycarbonyloxyjethy^piperidinelcarboxylic acid te/tbutyl ester carbonic acid 2[2(2methoxyethoxy)ethoxy]ethyl ester 3(2oxol,2dihydroindol 3ylidenemethyl)indollylmethyl ester acetic acid 3(2oxol,2dihydroindol3ylidenemethyl)indollylmethyl ester 2,2dimethylpropionic acid 3(2oxol,2dihydroindol3ylidenemethyl)indoll yl methyl ester 2,2dimethylpropionic acid 3(2oxolpiperidinlylmethyll,2dihydroindol3 ylidenemethyl)indollylmethyl ester cyclohexylacetic acid 3(2oxol,2dihydroindol3ylidenemethyl)indollylmethyl ester of ^/VdiethylZtZmethylSCZoxol^dihydroindolSylidenemethyOindollyl] acetamide 2[3(2oxol,2dihydroindol3ylidenemethyl)indollyl]/V(tetrahydrofuran2 ylmethyl)acetamide of Λ/benzo[l,3]dioxol5yl2[3(2oxol,2dihydroindol3ylidenemethyl)indollyl] acetamide /V(2methoxyethyl)2[3(2oxol,2dihydroindol3ylidenemethyl)indollyl] acetamide 3[l(2morpholin4yl2oxoethyl)lWindol3ylmethylene]l,3dihydroindol2one Λ/furan2ylmethyl2[3(2Oxol,2dihydroindol3ylidenemethyl)indollyl3 acetamide /Vcyclohexyl2[2methyl3(2Oxol,2dihydroindol3ylidenemethyl)indollyl] acetamide 3[2methyll(2oxo2piperidinlylethyl)lHindol3ylmethylene]l,3dihydro indol2one 2[2methyl3(2oxol,2dihydroindol3ylidenemethyl)indollyl]/V(tetrahydro furan2ylmethyl)acetamide Λ/cyclohexylmethyl2[3(2oxol,2dihydroindol3ylidenemethyl)indollyl] acetamide 4{2[3(2oxol,2dihydroindol3ylidenemethyl)indoIlyl]acetamino}butyric acid methyl ester 6{2[3(2oxol,2dihydroindol3ylidenemethyl)indollyl]acetamino}hexanoic acid ethyl ester 2[3(2oxol,2dihydroindol3ylidenemethyl)indollyl]Λ^(tetrahydrofuran2 ylmethyl)acetamide /V[2(lWindol3yl)ethyl]2[3(2oxol,2dihydroindol3ylidenemethyl)indollyl] /V(tetrahydrofuran2ylmethyl)acetamide 2[3(2oxol,2dihydroindol3ylidenemethyl)indollyl]Λ/(3phenylpropyl) acetamide 2[3(2oxol,2dihydroindol3ylidenemethyl)indollyl]/V(4phenylbutyl) acetamide Λ/[3(lformylpiperidin4yl)propyl]2[3(2oxol,2dihydroindol3ylidenemethyl) indollyl]acetamide Λ/(4hydroxybutyl)2[3(2oxol/2dihydroindol3ylidenemethyl)indollyl]~ acetamide 4{2[3(2oxol,2dihydroindol3ylidenemethyl)indollyl]acetamino}butyric acid ethyl ester 3{l[2(4methylpiperazinlyl)2oxoethyl]lWindol3ylmethylene}l,3dihydro indol2one 3{l[3(4methylpiperazinlyI)2oxo— propyl]l/yindol3yImethylene}l,3dihydro indol2one /V(2hydroxyethyl)3[3(2oxol,2dihydroindol3ylidenemethyl)indollyl] propionamide 3(lbenzenesulfonyll/Vindol3ylmethylene)l,3dihydroindol2one 3(lbenzyllWindol3ylmethylene)l,3dihydroindol2one lpiperidinlylmethyl3(lpiperidinlylmethyll/yindol3ylmethylene)l,3dihydro indol2one l^methylpiperazinlylmethyQSCl^methylpiperazinlylmethyOl/yindolS ylmethylene]l,3dihydroindol2one 3(lf/Indol3ylmethylene)lpiperidinlylmethyll,3dihydroindol2one 3(lWIndol3ylmethylene)lmorpholin4ylmethyll,3dihydroindol2one l(3,3dimethylpiperidinlylmethyl)3(l/yindol3ylmethylene)l,3dihydroindol2 one l[(te/tbutylmethylamino)methyl]3(lWindol3ylmethylene)l,3dihydroindol2 one 3{l[(diisobutylamino)methyl]lf/indol3ylmethylene}l,3dihydroindol2one 3(lmethyllWindol3ylmethylene)lpiperidinlylmethyll,3dihydroindol2one 3(lmethyl2phenyllWindol3ylmethylene)lpiperidinlylmethyll,3dihydro indol2one 3(5fluorolmethyllWindol3ylmethylene)lpiperidinlylmethyll,3dihydroindol 2one 3(6methoxylmethyllHindol3ylmethylene)lpiperidinlylmethyll,3dihydro indol2one 3(lbenzenesulfonyllWindol3ylmethylene)lpiperidinlylmethyll,3dihydroindol 2one lpiperidinlylmethyl3(4,5,6,7tetrafluorol/yindol3ylmethylene)l,3dihydro indol2one 3(7methyllWindol3ylmethylene)lpiperidinlylmethyll,3dihydroindol2one Λ/cyclohexyl2[2methyl3(2oxolpiperidinlylπnethyll,2dihydroindol3 ylidenemethyl)indollyl]acetamide /V(2methoxyethyl)2[3(2oxolpiperidinlylmethyll/2dihydroindol3 ylidenemethyl)indollyl]acetamide 3[l(2morpholin4yl2oxoethyl)l/Vindol3ylmethylene]lpiperidinlylmethyl l,3dihydroindol2one 5bromo3(lWindol3ylmethylene)lpiperidinlylmethyIl,3dihydroindol2one 7bromo3(ltfindol3ylmethylene)lpiperidinlylmethyll,3dihydroindol2one 6bromo3(lHindol3ylmethylene)lpiperidinlylmethyll,3dihydroindol2one 6fluorolpiperidinlylmethyl3(lpiperidinlylmethyllWindol3ylmethylene)l,3 dihydroindol2one 6chloro3(lWindol3yImethylene)lpiperidinlylmethyll,3dihydroindol2one acetic acid 3(2oxolpiperidinlylmethyll,2dihydroindol3ylidenemethyl)indoll y I methyl ester lhydroxymethyl3(lHindol3ylmethylene)l,3dihydroindol2one acetic acid l[3(lacetyllHindol3ylmethylene)2oxo2,3dihydroindollyl]ethyl ester 3(lHindol3yImethylene)l(tetrahydropyran2yloxy)l,3dihydroindol2one 3(lWindol3ylmethylene)l(2methoxyethoxy)l,3dihydroindol2one 3[l(2hydroxyehtyl)lWindole3ylmethylene]l,3dihydroindol2one 3(l/yindol3ylmethylene)2oxo2,3dihydroindolelcarboxylic acid benzyl ester 3(lacetoxymethyllWindol3ylmethylene)2oxo2,3dihydroindolelcarboxylic acid te/tbutyl ester .
21. A pharmaceutical composition comprising one or more compounds of formula I together with a pharmaceutically acceptable excipient or vehicle.
22. Use of a compound of general formula I for the preparation of a medicament for preventing, treating or ameliorating multiple sclerosis, or delaying the onset of or reducing the relapse rate in multiple sclerosis.
23. A method of preventing, treating or ameliorating multiple sclerosis, or delaying the onset of or reducing the relapse rate in multiple sclerosis, the method comprising administering to a patient in need thereof an effective amount of a compound of general formula I.
Description:
INDOLINONE DERIVATIVES FOR THE TREATMENT OF INFLAMMATORY AUTOIMMUNE DISEASES

FIELD OF INVENTION

The present invention relates to derivatives of indolinone compounds and their inclusion in pharmaceutical compositions intended for oral administration as well as their use in the treatment of inflammatory autoimmune diseases such as multiple sclerosis.

BACKGROUND OF THE INVENTION

Multiple sclerosis is an autoimmune inflammatory disease of the central nervous system characterised by T-cell infiltration, demyelination of white matter and axonal injury. The disease mostly affects young adults with an onset at 20-40 years of age and affects twice as many women as men (A. Compton and A. Coles, The Lancet 359, 6 April 2002, pp. 1221-1231). Multiple sclerosis is more common in temperate climate zones and thus has a prevalence of 50-130 out of 100,000 in northern Europe and North America (N. Hellings et al., Immunologic Research 25(1), 2002, pp. 27-51). While the higher incidence and prevalence of multiple sclerosis in certain European populations has not been adequately explained, it is believed that increased genetic susceptibility in these populations is partly responsible. The presence of a genetic element in the etiology of the disease is supported by family studies showing that first-degree relatives of multiple sclerosis patients have a 20-40 times increased risk of developing the disease relative to the general population (J. H. Noseworthy et al., New England Journal of Medicine 343(13), 2000, pp. 938-952). Furthermore, it has been recognised that populations with a high frequency of for instance the HLA-DR2 allele have a significantly higher risk of developing multiple sclerosis (Hellings et al., supra; Noseworthy et al., supra). However, no single major susceptibility gene for multiple sclerosis has been identified so far, and the results of genome screens conducted to identify susceptibility genes rather point to multiple genes exerting a moderate effect (Hellings et al., supra).

Based on these studies, it would appear that genetic susceptibility is not enough in itself to provoke multiple sclerosis. This theory is given credence by the fact that the rate of prevalence of multiple sclerosis among people of European descent living outside Europe is half of that persisting in parts of northern Europe and that the low frequency of multiple sclerosis in Africans increases significantly among first-generation decendants living in Europe (Compton and Coles, supra). Environmental factors have therefore also been proposed as contributing to the development of multiple sclerosis. In particular, it

is believed that certain antigens present on pathogenic organisms such as viral or bacterial epitopes which structurally resemble autoantigenic epitopes of, for instance, myelin basic protein, proteolipid protein, myelin-associated glycoprotein or oligodendrocyte glycoprotein, which are all components of the myelin sheath, may lead to activation of T-cells that are reactive with such antigenic epitopes and initiating the inflammatory process eventually resulting in clinical manifestations of multiple sclerosis. This phenomenon is generally referred to as molecular mimicry (Hellings et al., supra; A. Bar-Or et al., J. Neuroimmunol. 100, 1999, pp. 252-259; A. Kami and H. L. Weiner, "Organ-Specific Inflammatory Diseases" Chapter 77 in Clinical Immunology; Principles and Practice, 2 nd Ed. (R.R. Rich et al., Eds.), Mosby, London, 2001).

Multiple sclerosis is usually defined as either a relapsing-remitting or a progressive disease. The relapsing-remitting form with which 80% of the patients are initially afflicted (Compton and Coles, supra) is characterised by discrete attacks with full or partial recovery between relapses. In 40-50% of the patients, the disease eventually becomes progressive (secondary progressive stage). The disease may also be progressive from the outset (primary progressive form) characterised by a gradual decline in neurological function with no periods of remission. The clinical symptoms of the relapsing-remitting form of multiple sclerosis may vary widely from one patient to the other, but commonly affected individuals initially experience some degree of visual and sensory impairment, limb paresthesias, limb weakness, clumsiness, fatigue and gait ataxia, while in the later stages cognitive impairment, progressive quadriparesis, sensory loss, ataxic tremors, pain and spasticity are more common (Noseworthy et al., supra). The primary progressive form may initially manifest as one or more of these symptoms, gradually declining into quadriparesis, cognitive decline, visual loss, brainstem syndromes and cerebellar, bowel and bladder dysfunction (Noseworthy et al., supra).

Pathologically, multiple sclerosis is characterised by the presence of demyelinated plaques or sclerotic lesions where the myelin sheath surrounding the axons is destroyed. The inflammatory infiltrate in the lesions is composed of T-cells, B-cells, microglia and macrophages which interact with the myelin sheath and participate in the demyelinating process by local production of immune-related molecules such as adhesion molecules, cytokines and chemokines as well as demyelinating antibodies, oxygen free radicals and nitric oxide (Kami and Weiner, supra). While axonal destruction is not pronounced in the early stages of the disease (although more pronounced in patients suffering from the primary progressing form), demyelination of the axons results in slowing and blocking conductivity (Noseworthy et al., supra). Regression of the symptoms may be associated

with partial remyelination after the initial inflammation has subsided showing that oligodendrocytes (myelin-producing cells) are present in the lesions (Kami and Weiner, supra). In later stages, irreversible axonal injury, gliotic scarring and gradual loss of oligodendrocyte progenitor cells may result from repeated episodes of inflammatory attack and lead to permanent loss of neurological function (Noseworthy et al., supra).

While the immunopathogenesis of multiple sclerosis is still largely unknown, it has been shown that autoreactive T-cells specific for myelin basic protein and other antigens of the central nervous system exist in the periphery of healthy individuals as wells as individuals who later develop multiple sclerosis (Bar-Or et al., supra; O'Connor et al., J. Clin. Immunol. 21(2), 2001, pp. 81-93). Thus, the presence of myelin-reactive T-cells in the periphery is not enough in itself to explain the development of multiple sclerosis. In multiple sclerosis patients, these T-cells become activated, possibly by cross-reactivity with bacterial or viral antigens that structurally resemble myelin antigens (i.e. the phenomenon known as molecular mimicry) and/or by bacterial superantigens, and persist in an enhanced state of activation (Hellings et al., supra). It has been found that the autoreactive T-cells are predominantly CD4+ T helper cells type 1 (ThI) producing interleukin-2 (IL-2), interferon-γ (IFN-γ) and tumour necrosis factor (TNF-α) (B. Gran and A. Rostami, Current Neurology and Neuroscience Reports 1, 2001, pp. 263-270). In order for such proinflammatory T-cells to migrate to the central neurvous system, they express chemokine receptors, adhesion molecules and matrix metalloproteinases that enable them to cross the blood-brain barrier. Thus, it has been found that expression levels of the chemokines which are chemotactic for ThI cells, IP-10 and RANTES, and their corresponding receptors, CXCR3 and CCR5, are elevated in sclerotic lesions and cerebrospinal fluid of multiple sclerosis patients (Bar-Or et al., supra). Altered levels of the adhesion molecules ICAM-I and VCAM-I have been identified on endothelial cells of multiple sclerosis lesions (O'Connor et al., supra). ICAM-I and VCAM-I are important for endothelial-leukocyte interactions and leukocyte extravasation. Matrix metalloproteinases expressed by activated T-cells, monocytes and astrocytes may disrupt the basement membrane of the blood-brain barrier and facilitate transmigration of T-cells and breakdown of the extracellular matrix (O'Connor et al., supra).

Once the T-cells have entered the central nervous system they become reactivated on encountering the autoantigen, e.g. myelin basic protein, presented by MHC class II expressing antigen presenting cells (microglia and dendritic cells), and the ThI cells respond by producing proinflammatory cytokines such as TNF-α, IFN-γ and IL-2, while the Th2 cells produce anti-inflammatory cytokines such as IL-4, IL-5 and IL-10 (Bar-Or

et al., supra). In turn, the inflammatory process leads to up-regulation of MHC class II expression and adhesion molecules on the blood-brain barrier endothelium, facilitating a further influx of T-cells, B-cells and macrophages and hence an amplification of the inflammatory response (Hellings et al., supra). This theory is supported by the finding that myelin basic protein reactive T-cell clones from multiple sclerosis patients were found to secrete increased amounts of different cytokines such as TNF-α, IL-2 and IL-IO (Hellings et al., supra). Demyelination (myelin destruction) is believed to be brought about by the combined effects of cytotoxic cells (macrophages and T-cells), oxygen free radicals, demyelinating autoantibodies and cytokine-induced toxicity (Hellings et al., supra).

Traditionally, corticosteroids such as prednisolone have been administered intravenously to multiple sclerosis patients during acute relapses in order to attenuate the inflammatory response. It has been found that treatment with corticosteroids during relapses reduces the duration of relapses and their short-term morbidity, but not the permanent disabilities resulting from repeated relapses (Compton and Coles, supra). Furthermore, treatment with potent corticosteroids at high doses has serious side effects, notably osteoporosis, aseptic bone necrosis, skin atrophy, striae cutis, insomnia, myopathy, posterior and capsular cataract and glaucoma as well as reactivation of the disease upon cessation of treatment. More recently, interferon-β (IFN-β) has been introduced as a treatment of relapsing-remitting multiple sclerosis and has been found to decrease the rate of relapse, increase the proportion of patients who were relapse free and reduce the number of patients who had moderate to severe relapses. On the other hand, INF-β treatment is extremely costly and its long-term efficacy has not been established. There is concern that the treatment may induce the formation of neutralising antibodies that may reduce the activity of IFN-β (Noseworthy et al., supra). Most of the patients initially experience flu-like symptoms when treated with IFN-β. Glatiramer acetate is another recent treatment based on a mixture of random synthetic peptides intended to mimic myelin basic protein. In a double-blind trial of relapsing- remitting multiple sclerosis, glatiramer acetate was found to decrease the rate of relapse. Glatiramer acetate is believed to be most effective for mildly disabled patients with a recent diagnosis of multiple sclerosis. Fewer treatment options exist for patients in the progressive phase of the disease. Immunosuppressive therapy, e.g. with cyclophosphamide or methotrexate, is frequently attempted, but it is generally recognised that once the disease enters the progressive stage treatment is very difficult. IFN-β has been in clinical trials for secondary progressive multiple sclerosis but the

results did not show that the treatment slowed progression of disability and the benefits of this treatment in secondary progressive disease are controversial.

Moreover, IFN-β and glatiramer acetate are peptidic in nature and as such must be administered parenterally, e.g. by injection. This is clearly less convenient to patients than an oral treatment. It would therefore constitute a significant improvement compared to existing multiple sclerosis therapies if a medicament suitable for oral administration were to be developed.

SUMMARY OF THE INVENTION

In the course of research leading to the present invention, certain compounds within the class of indolinones have been found to exhibit a surprisingly high level of activity in experimentally induced autoimmune encephalomyelitis (EAE), making them potentially useful as immunomodulatory agents in the treatment of autoimmune inflammatory diseases such as multiple sclerosis. However, these compounds are generally highly lipophilic and are therefore less suitable for administration by the oral route. Further experimental work has led to the development of derivatives of indolinone compounds showing a high activity in vivo when administered orally.

Accordingly, the present invention relates to a compound of formula I

wherein X is O or S;

R 1 is hydrogen, -OR 9 , -OC(O)R 9 , -NRi 0 Rn, -C(O)NRi 0 Rn, -OC(O)NR 10 R II , -NHC(O)R 10 , - NHC(O)ORi 0 , -NHC(O)NRi 0 Ru, -S(O)R 9 , -S(O) 2 R 9 , -S(O) 2 OR 9 , -S(O) 2 NR I0 RU, -C(O)RI 2 , - C(O)ORi 2 , -OC(O)ORi 2 , -P(O)(OR I0 )(OR U ), -OP(O)(OR I0 )(OR U ), polyoxyethylene, Ci-I 0 alkyl, C 2-I0 alkenyl, C 2-I0 alkynyl, aryl, carbocyclyl, heteroaryl or heterocyclyl, said Ci-I 0 alkyl, C 2-I0 alkenyl, C 2-I0 alkynyl, aryl, carbocyclyl, heteroaryl or heterocyclyl being substituted with one or more substituents selected from the group consisting of -ORi 0 , - C(O)Ri 0 , -C(O)OR 10 , -OC(O)R 10 , -OC(O)OR 10 , -NR 10 R 11 , -C(O)NR 10 R 11 , -OC(O)NR 10 R 11 , - OP(O)(OR 10 )(OR 11 ), -P(O)(OR 10 )(OR 11 ), -NHC(O)R 10 , -NHC(O)OR 10 , -NHC(O)NR 10 R 11 , - SR 10 , -S(O)R 10 , -S(O) 2 R 10 , -S(O) 2 NR 10 R 11 , -S(O) 2 OR 10 , polyoxyethylene, aryl, heteroaryl, carbocyclyl and heterocyclyl, said aryl, heteroaryl, carbocyclyl or heterocyclyl being optionally substituted with -OR 10 , -C(O)R 10 , -C(O)OR 10 , OC(O)R 10 , -OC(O)OR 10 , - P(O)(OR 10 )(OR 11 ), -OP(O)(OR 10 )(OR 11 ), -NR 10 R 11 , -C(O)NR 10 R 11 , -NHC(O)R 10 , - NHC(O)OR 10 , -NHC(O)NR 10 R 11 , -SR 10 , -S(O)R 10 , -S(O) 2 R 10 , -S(O) 2 NR 10 R 11 , -S(O) 2 OR 10 or polyoxyethylene, and C 1-6 alkyl substituted with polyoxyethylene, -OR 10 , -C(O)R 10 , - C(O)OR 10 , OC(O)R 10 , -OC(O)OR 10 , -NR 10 R 11 , -C(O)NR 10 R 11 , -P(O)(OR 10 )(OR 11 ), - OP(O)(OR 10 )(OR 11 ), -NHC(O)R 10 , -NHC(O)OR 10 , -NHC(O)NR 10 R 11 , -SR 10 , -S(O)R 10 , - S(O) 2 R 10 , -S(O) 2 NR 10 R 11 or -S(O) 2 OR 10 , wherein R 9 is C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl, aryl, heteroaryl, carbocyclyl or heterocyclyl, said C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl, aryl, heteroaryl, carbocyclyl or heterocyclyl being optionally substituted with one or more substituents selected from the group consisting of halogen, trihalomethyl, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, hydroxy, carboxy, formyl, aryl, heteroaryl, carbocyclyl, heterocyclyl, amino, carbamoyl, cyano, guanidino, carbamido, polyoxyethylene, -OR 13 , -C(O)R 13 , -C(O)OR i3 , -OC(O)R 13 , - OC(O)OR 13 , -NR 13 R 14 , -C(O)NR 13 R 14 , -OC(O)NR 13 R 14 , -NHC(O)R 13 , -NHC(O)OR 13 , - NHC(O)NRi 3 R 14 , -P(O)(OR 13 )(OR 14 ), -OP(O)(OR 13 )(OR 14 ), -SR 13 , -S(O)R 13 , -S(O) 2 R 13 , - S(O) 2 NR 13 R 14 and -S(O) 2 OR 13 ; R 10 and R 11 are the same or different and independently selected from the group consisting of hydrogen, polyoxyethylene, C x-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl, or wherein R 10 and R 11 , together with the nitrogen atom to which they are attached form a heterocyclic or heteroaryl ring, each of C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl being optionally substituted with one or more, same or different substituents selected from the group consisting of hydrogen, halogen, trihalomethyl, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, hydroxy, carboxy, formyl, aryl, heteroaryl, carbocyclyl, heterocyclyl, amino, carbamoyl, cyano, guanidino, carbamido, polyoxyethylene, -OR 13 , -C(O)R 13 , -C(O)OR 13 , -

OC(O)R 13 , -OC(O)OR 13 , -N 3 , -NR 13 R 14 , -C(O)NR 13 R 14 , -OC(O)NR 13 R 14 , -NHC(O)R 13 , - NHC(O)OR 13 , -NHC(O)NR 13 R 14 , -P(O)(OR 13 )(OR 14 ), -OP(O)(OR 13 )(OR 14 ), -SR 13 , -S(O)R 13 , -S(O) 2 R 13 , -S(O) 2 NR 13 R 14 and -S(O) 2 OR 14 , said aryl, heteroaryl, carbocyclyl or heterocyclyl being optionally substituted with one or more substituents selected from the group consisting of -OR 13 , -C(O)R 13 , -C(O)OR 13 , C(O)NR 13 R 14 and polyoxyethylene, provided that R 10 and R 11 are not both hydrogen;

R 12 is C 1-10 alkyl, C 2-1O alkenyl, C 2-10 alkynyl, aryl or carbocyclyl substituted with hydroxy, amino, cyano, halogen, carboxy, nitro, amido, polyoxyethylene, -OR 13 , -C(O)R 13 , - C(O)OR 13 , -OC(O)R 13 , -OC(O)OR 13 , -NR 13 R 14 , -C(O)NR 13 R 14 , -OC(O)NR 13 Ri 4 , -NHC(O)R 13 , -NHC(O)OR 13 , -NHC(O)NR 13 R 14 , -P(O)(OR 13 )(OR 14 ), -OP(O)(OR 13 )(OR 14 ), -SR 13 , -S(O)R 13 , -S(O) 2 R 13 , -S(O) 2 NR 13 R 14 and -S(O) 2 OR 14 , or heteroaryl or heterocyclyl optionally substituted with hydroxy, amino, cyano, halogen, carboxy, nitro, amido, polyoxyethylene, -OR 13 , -C(O)R 13 , -C(O)OR 13 , -OC(O)R 13 , -OC(O)OR 13 , -NRi 3 R 14 , - C(O)NR 13 R 14 , -OC(O)NR 13 R 14 , -P(O)(OR 13 )(OR 14 ), -OP(O)(OR 13 )(OR 14 ), -NHC(O)R 13 , - NHC(O)OR 10 , -NHC(O)NR 10 R 11 , -SR 13 , -S(O)R 13 , -S(O) 2 R 13 , -S(O) 2 NR 13 R i4 and - S(O) 2 OR 14 ;

R 13 and R 14 are the same or different and independently selected from the group consisting of hydrogen, Ci -5 -alkyl, C 2-6 -alkenyl, C 4-6 -alkadienyl, C 2-6 -alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl, or wherein R 13 and R 14 , together with the nitrogen atom to which they are attached form a heterocyclic or heteroaryl ring, each C 1- 6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl substituent being optionally substituted with one or more, same or different substituents selected from the group consisting of hydrogen, hydroxy, C 1-4 alkyl, C 1-4 alkoxy, nitro, cyano, amino, oxo, halogen, trihalomethyl, C 1-4 alkylthio, C 1-4 alkylamino, C 1-4 alkoxycarbonyl, carboxy, -CONH 2 or -S(O) 2 NH 2 ;

R 2 , R 3 , R 4 and R 5 are the same or different and independently selected from the group consisting of hydrogen, halogen, trihalomethyl, C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl, hydroxy, carboxy, formyl, aryl, heteroaryl, carbocyclyl, heterocyclyl, amino, carbamoyl, cyano, guanidino, carbamido, -OR 15 , -C(O)R 15 , -C(O)OR 15 , -OC(O)R 15 , -OC(O)OR i5 , - NR 15 R 16 , -C(O)NR 15 R 16 , -OC(O)NR 15 R 16 , -NHC(O)R 15 , -NHC(O)OR 15 , -NHC(O)NRi 5 R 16 , - SR 15 , -S(O)R 15 , -S(O) 2 Ri 5 , -S(O) 2 NR 15 R 16 and -S(O) 2 OR 16 , wherein R 15 and R 16 are the same or different and independently selected from the group consisting of hydrogen, C 1- 10 alkyl, C 2- I 0 alkenyl, C 2-10 alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl, or wherein R 15 and R 16 , together with the nitrogen atom to which they are attached form a heterocyclic or heteroaryl ring, said C 1-10 alkyl, C 2- I 0 alkenyl, C 2-10 alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl being optionally substituted with one or more,

same or different substituents selected from the group consisting of hydrogen, halogen, trihalomethyl, Ci -6 -alkyl, C 2-6 -alkenyl, C 2 - 6 -alkynyl, hydroxy, carboxy, formyl, aryl, heteroaryl, carbocyclyl, heterocyclyl, amino, carbamoyl, cyano, guanidino, carbamido, - OR 17 , -C(O)R 17 , -C(O)OR 17 , -OC(O)OR 17 , -OC(O)R 18 , -NR 17 R 18 , -C(O)NR 17 R 18 , - OC(O)NR 17 R 18 , -NHC(O)R 17 , -NHC(O)OR 17 , -NHC(O)NR 17 R 18 , -SR 17 , -S(O)R 17 , -S(O) 2 R 17 , - S(O) 2 NR 17 R 18 and -S(O) 2 OR 17 , wherein R 17 and R 18 are the same or different and independently selected from the group consisting of hydrogen, C 1-6 -alkyl, C 2-6 -alkenyl, C 2-6 -alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl, or wherein R 17 and R 18 , together with the nitrogen atom to which they are attached form a heterocyclic or heteroaryl ring, each C 1-6 -alkyl, C 2-6 -alkenyl, C 2-6 -alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl substituent being optionally substituted with one or more, same or different substituents selected from the group consisting of hydrogen, hydroxy, C 1-4 alkyl, C 1-4 alkoxy, nitro, cyano, amino, oxo, halogen, trihalomethyl, C 1-4 alkylthio, C 1-4 alkylamino, C 1-4 alkoxycarbonyl, carboxy, -CONH 2 or -S(O) 2 NH 2 ;

R 6 is hydrogen, C 1-6 alkyl, cycloalkyl, aryl, heteroaryl, heterocyclyl, halogen, -OR 7 , - C(O)R 7 , -C(O)OR 7 , -NR 7 R 8 , S(O) 2 NR 7 R 8 , wherein R 7 and R 8 are independently hydrogen, Ci -6 alkyl, aryl or heterocyclyl, said C 1-6 alkyl or heterocyclyl being optionally substituted by heterocyclyl, -OR 7 , -C(O)R 7 or C(O)OR 7 ;

R 1 ', R 3 ', R 4 ', Rs' and R 6 ' are the same or different and independently selected from the group consisting of hydrogen, halogen, trihalomethyl, C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl, hydroxy, carboxy, formyl, aryl, heteroaryl, carbocyclyl, heterocyclyl, amino, carbamoyl, cyano, guanidino, carbamido, -OR 15 , -C(O)R 15 , -C(O)OR 15 , -OC(O)R 15 , - OC(O)OR 15 , -NR 15 R 16 , -C(O)NR 15 R 16 , -OC(O)NR 15 R 16 , -NHC(O)R 15 , -NHC(O)OR 15 , -

NHC(O)NR 15 R 16 , -SR 15 , -S(O)R 15 , -S(O) 2 R 15 , -S(O) 2 NR 15 R 16 and -S(O) 2 OR 16 , wherein R 15 and R 16 are the same or different and independently selected from the group consisting of hydrogen, C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl, or wherein R 15 and R 16 , together with the nitrogen atom to which they are attached form a heterocyclic or heteroaryl ring, said C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl being optionally substituted with one or more, same or different substituents selected from the group consisting of hydrogen, halogen, trihalomethyl, C 1-6 -alkyl, C 2-6 -alkenyl, C 2-6 -alkynyl, hydroxy, carboxy, formyl, aryl, heteroaryl, carbocyclyl, heterocyclyl, amino, carbamoyl, cyano, guanidino, carbamido, -OR 17 , -C(O)R 17 , -C(O)OR 17 , -OC(O)R 18 , -OC(O)OR 17 , -NR 17 R 18 , -C(O)NR 17 R 18 , -OC(O)NR 17 R 18 , -NHC(O)R 17 , -NHC(O)OR 17 , -NHC(O)NR 17 R 18 , -SR 17 , -S(O)R 17 , -S(O) 2 R 17 , -S(O) 2 NR 17 R 18 and -S(O) 2 OR 17 , wherein R 17 and R 18 are the same or different and

independently selected from the group consisting of hydrogen, Ci -6 -alkyl, C 2-6 -alkenyl, C 2-6 -alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl, or wherein R 17 and R 18 , together with the nitrogen atom to which they are attached form a heterocyclic or heteroaryl ring, each Ci -6 -alkyl, C 2-6 -alkenyl, C 2-6 -alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl substituent being optionally substituted with one or more, same or different substituents selected from the group consisting of hydrogen, hydroxy, C 1-4 alkyl, C 1-4 alkoxy, nitro, cyano, amino, oxo, halogen, trihalomethyl, C 1-4 alkylthio, C 1-4 alkylamino, C 1-4 alkoxycarbonyl, carboxy, -CONH 2 or -S(O)NH 2 ; and

R 2 ' is hydrogen, , -OR 9 , -OC(O)R 9 , -NR 10 R 11 , -C(O)NR 10 R 11 , -OC(O)NR 10 R 11 , -NHC(O)R 10 , - NHC(O)OR 10 , -NHC(O)NR 10 R 11 , -S(O)R 9 , -S(O) 2 R 9 , -S(O) 2 OR 9 , -S(O) 2 NR 10 R 11 , -C(O)R 19 , - C(O)OR 19 , -OC(O)OR 19 , -P(O)(OR 10 )(OR 11 ), -OP(O)(OR 10 )(OR 11 ), polyoxyethylene, C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl, aryl, carbocyclyl, heteroaryl or heterocyclyl, said C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl, aryl, carbocyclyl, heteroaryl or heterocyclyl being substituted with one or more substituents selected from the group consisting of -OR 10 , - C(O)R 10 , -C(O)OR 10 , -OC(O)R 10 , -OC(O)OR 10 , -NR 10 R 11 , -C(O)NR 10 R 11 , -OC(O)NR 10 R 11 , - OP(O)(OR 10 )(OR 11 ), -P(O)(OR 10 )(OR 11 ), -NHC(O)R 10 , -NHC(O)OR 10 , -NHC(O)NR 10 R 11 , - SR 10 , -S(O)R 10 , -S(O) 2 R 10 , -S(O) 2 NR 10 R 11 , -S(O) 2 OR 10 , polyoxyethylene, aryl, heteroaryl, carbocyclyl and heterocyclyl, said aryl, heteroaryl, carbocyclyl or heterocyclyl being optionally substituted with -OR 10 , -C(O)R 10 , -C(O)OR 10 , OC(O)R 10 , -OC(O)OR 10 , - P(O)(OR 10 )(OR 11 ), -OP(O)(OR 10 )(OR 11 ), -NR 10 R 11 , -C(O)NR 10 R 11 , -NHC(O)R 10 , - NHC(O)OR 10 , -NHC(O)NR 10 R 11 , -SR 10 , -S(O)R 10 , -S(O) 2 R 10 , -S(O) 2 NR 10 R 11 , -S(O) 2 OR 10 or polyoxyethylene, and C 1-6 alkyl substituted with polyoxyethylene, -OR 10 , -C(O)R 10 , - C(O)OR 10 , OC(O)R 10 , -OC(O)OR 10 , -NR 10 R 11 , -C(O)NR 10 R 11 , -P(O)(OR 10 )(OR 11 ), - OP(O)(OR 10 )(OR 11 ), -NHC(O)R 10 , -NHC(O)OR 10 , -NHC(O)NR 10 R 11 , -SR 10 , -S(O)R 10 , - S(O) 2 R 10 , -S(O) 2 NR 10 R 11 or -S(O) 2 OR 10 , wherein

R 9 is C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl, aryl, heteroaryl, carbocyclyl or heterocyclyl, said C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl, aryl, heteroaryl, carbocyclyl or heterocyclyl being optionally substituted with one or more substituents selected from the group consisting of halogen, trihalomethyl, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, hydroxy, carboxy, formyl, aryl, heteroaryl, carbocyclyl, heterocyclyl, amino, carbamoyl, cyano, guanidino, carbamido, polyoxyethylene, -OR 13 , -C(O)R 13 , -C(O)OR 13 , -OC(O)R 13 , - OC(O)OR 13 , -NR 13 R 14 , -C(O)NR 13 R 14 , -OC(O)NR 13 R 14 , -NHC(O)R 13 , -NHC(O)OR 13 , - NHC(O)NR 13 R 14 , -P(O)(OR 13 )(OR 14 ), -OP(O)(OR 13 )(OR 14 ), -SR 13 , -S(O)R 13 , -S(O) 2 R 13 , - S(O) 2 NR 13 R 14 and -S(O) 2 OR 13 ;

Rio and Ru are the same or different and independently selected from the group consisting of hydrogen, polyoxyethylene, Ci-I 0 alkyl, C 2-I0 alkenyl, C 2-I0 alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl, or wherein Ri 0 and Rn, together with the nitrogen atom to which they are attached form a heterocyclic or heteroaryl ring, each of Ci-io alkyl, C 2- io alkenyl, C 2-I0 alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl being optionally substituted with one or more, same or different substituents selected from the group consisting of hydrogen, halogen, trihalomethyl, C x-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, hydroxy, carboxy, formyl, aryl, heteroaryl, carbocyclyl, heterocyclyl, amino, carbamoyl, cyano, guanidino, carbamido, polyoxyethylene, -ORi 3 , -C(O)Ri 3 , -C(O)ORi 3 , - OC(O)Ri 3 , -OC(O)ORi 3 , -N 3 , -NRi 3 Ri 4 , -C(O)NRi 3 Ri 4 , -OC(O)NRi 3 Ri 4 , -NHC(O)Ri 3 , -

NHC(O)ORi 3 , -NHC(O)NRi 3 Ri 4 , -P(O)(ORi 3 )(ORi 4 ), -OP(O)(ORi 3 )(ORi 4 ), -SRi 3 , -S(O)R 13 , -S(O) 2 Ri 3 , -S(O) 2 NRi 3 Ri 4 and -S(O) 2 ORi 4 , said aryl, heteroaryl, carbocyclyl or heterocyclyl being optionally substituted with one or more substituents selected from the group consisting of -ORi 3 , -C(O)Ri 3 , -C(O)ORi 3 , C(O)NRi 3 Ri 4 and polyoxyethylene, provided that Ri 0 and Ru are not both hydrogen;

Ri 9 is Ci-io alkyl, C 2-10 alkenyl, C 2-I0 alkynyl, aryl, heteroaryl, carbocyclyl or heterocyclyl, said Ci-io alkyl, C 2-I0 alkenyl, C 2-I0 alkynyl, aryl, heteroaryl, carbocyclyl or heterocyclyl being substituted with one or more substituents selected from the group consisting of substituted with one or more, same or different substituents selected from the group consisting of hydroxy, amino, cyano, halogen, carbocy, nitro, amido, polyoxyethylene, - ORi 3 , -C(O)Ri 3 , -C(O)OR 13 , -OC(O)R i3 , -OC(O)ORi 3 , -NR 13 Rj 4 , -C(O)NRi 3 Ri 4 , - OC(O)NR 13 R 14 , -NHC(O)R 13 , -NHC(O)OR 13 , -NHC(O)NR 13 R 14 , -SR 13 , -S(O)R i3 , -S(O) 2 Ri 3 , - S(O) 2 NRi 3 R 14 and -S(O) 2 OR 14 ; R 13 and R 14 are the same or different and independently selected from the group consisting of hydrogen, C 1-6 -alkyl, C 2-6 -alkenyl, C 4-6 -alkadienyl, C 2-6 -alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl, or wherein R 13 and R 14 , together with the nitrogen atom to which they are attached form a heterocyclic or heteroaryl ring, each C 1- 6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl substituent being optionally substituted with one or more, same or different substituents selected from the group consisting of hydrogen, hydroxy, C 1-4 alkyl, C 1-4 alkoxy, nitro, cyano, amino, oxo, halogen, trihalomethyl, C 1-4 alkylthio, C 1-4 alkylamino, C 1-4 alkoxycarbonyl, carboxy, -CONH 2 or -S(O) 2 NH 2 ; provided that R 1 and R 2 ' are not both hydrogen and that, when R 1 is other than hydrogen, R 2 ' may also be heterocyclyl, heteroaryl, -C(O)R 20 , -C(O)OR 20 , -S(O) 2 R 20 or C 1-6 alkyl optionally substituted with heterocyclyl, heteroaryl or -C(O)OR 20 , wherein R 20 is hydrogen, Ci -6 alkyl, aryl, heteroaryl or heterocyclyl, and that, when R 2 ' is other than hydrogen, R 1 may also be heterocyclyl, heteroaryl, -C(O)R 20 , -C(O)OR 20 , -S(O) 2 R 20 or Ci-

6 alkyl optionally substituted with heterocyclyl, heteroaryl or -C(O)OR 20 , wherein R 20 is hydrogen, Ci -6 alkyl, aryl, heteroaryl or heterocyclyl; or pharmaceutically acceptable salts thereof.

In another aspect, the invention relates to a pharmaceutical composition comprising one or more compounds of formula I together with a pharmaceutically acceptable excipient or vehicle.

In a further aspect, the invention relates to the use of a compound of general formula I for the preparation of a medicament for preventing, treating or ameliorating multiple sclerosis, or delaying the onset of or reducing the relapse rate in multiple sclerosis.

In a still further aspect, the invention relates to a method of preventing, treating or ameliorating multiple sclerosis, or delaying the onset of or reducing the relapse rate in multiple sclerosis, the method comprising administering to a patient in need thereof an effective amount of a compound of formula I.

DETAILED DESCRIPTION OF THE INVENTION

Definitions

In the present context, the term "Ci_ 12 -alkyr is intended to mean a linear or branched hydrocarbon group having 1 to 12 carbon atoms, such as methyl, ethyl, propyl, iso- propyl, butyl, te/t-butyl, /so-butyl, pentyl, hexyl, etc. Analogously, the term "Ci -I0 alkyl" and "Ci -6 -alkyl" is intended to mean a linear or branched hydrocarbon group having 1 to 10 or 1 to 6 carbon atoms, respectively, such as methyl, ethyl, propyl, /so-propyl, pentyl or hexyl, and the term "Ci -4 -alkyl" is intended to cover linear or branched hydrocarbon groups having 1 to 4 carbon atoms, e.g. methyl, ethyl, propyl, /so-propyl, butyl, /so- butyl, te/t-butyl.

Similarly, the terms "C 2- i 2 -alkenyl" are intended to cover linear, cyclic or branched hydrocarbon groups having 2 to 12, 4 to 12 or 6 to 12, carbon atoms and comprising one, two or three unsaturated bonds. Examples of alkenyl groups are vinyl, allyl, butenyl, pentenyl, hexenyl, heptenyl, octenyl, heptadecaenyl, butadienyl, pentadienyl, hexadienyl, heptadienyl, heptadecadienyl, hexatrienyl, heptatrienyl, octatrienyl and heptadecatrienyl. Preferred examples of alkenyl are vinyl, allyl, butenyl, especially allyl.

Similarly, the term "C 2- i 2 -alkynyr is intended to mean a linear or branched hydrocarbon group having 2 to 12 carbon atoms and comprising a triple bond. Examples hereof are ethynyl, propynyl, butynyl, octynyl, and dodecaynyl.

The term "halogen" or "halo" includes fluoro, chloro, bromo, and iodo.

In the present context the term "aryl" is intended to mean a fully or partially aromatic carbocyclic ring or ring system, such as phenyl, naphthyl, 1,2,3,4-tetrahydronaphthyl, anthracyl, phenanthracyl, pyrenyl, benzopyrenyl, fluorenyl and xanthenyl, among which phenyl is a preferred example.

The term "heteroaryl" is intended to mean a fully or partially aromatic carbocyclic ring or ring system, usually a mono- or bicyclic ring system comprising 5-12 ring atoms, where one or more of the carbon atoms have been replaced with heteroatoms, e.g. nitrogen (=N- or -NH-), sulphur, and/or oxygen atoms. Examples of such heteroaryl groups are oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, pyrrolyl, imidazolyl, pyrazolyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, triazinyl, coumaryl, furyl, thienyl, quinolyl, benzothiazolyl, benzotriazolyl, benzodiazolyl, benzooxazolyl, phthalazinyl, phthalanyl, triazolyl, tetrazolyl, isoquinolyl, acridinyl, carbazolyl, dibenzazepinyl, indolyl, benzopyrazolyl, phenoxazonyl. Particularly interesting heteroaryl groups are oxazolyl, isoxazolyl, oxadiazolyl, oxatriazolyl, thiazolyl, isothiazolyl, thiadiazolyl, thiatriazolyl, pyrrolyl, imidazolyl, pyrazolyl, pyridinyl, pyrimidinyl, pyrazinyl, pyridazinyl, furyl, thienyl, quinolyl, triazolyl, tetrazolyl, isoquinolyl, indolyl in particular pyrrolyl, imidazolyl, pyridinyl, pyrimidinyl, thienyl, quinolyl, tetrazolyl, and isoquinolyl.

The term "carbocyclyl" is intended to indicate a cyclic hydrocarbon radical, which may be a saturated or unsaturated, non-aromatic, mono- or bicyclic ring comprising 5-12 ring atoms, such as C 3-8 cycloalkyl, e.g. cyclopropyl, cyclopentyl, cyclohexyl or cyclooctyl, or a C 3-8 cycloalkylene radical, e.g. cycloprop-2-enyl, cyclobut-2-enyl, cyclopent-2-enyl, cyclohex-3-enyl, cycloocta-4-enyl or cyclohex-3,5-dienyl.

The term "heterocyclyl" is intended to mean a non-aromatic ring or ring system, usually a mono- or bicyclic ring system comprising 5-12 ring atoms, where one or more of the carbon atoms have been replaced with heteroatoms, e.g. nitrogen (=N- or -NH-), sulphur, and/or oxygen atoms. Examples of such heterocyclyl groups are imidazolidine, piperazine, hexahydropyridazine, hexahydropyrimidine, diazepane, diazocane, pyrrolidine, piperidine, azepane, azocane, aziridine, azirine, azetidine, pyroline, tropane,

oxazinane (morpholine), azepine, dihydroazepine, tetrahydroazepine, hexahydroazepine, oxazolane, oxazepane, oxazocane, thiazolane, thiazinane, thiazepane, thiazocane, oxazetane, diazetane, thiazetane, tetrahydrofuran, tetrahydropyran, oxepane, tetrahydrothiophene, tetrahydrothiopyrane, thiepane, dithiane, dithiepane, dioxane, dioxepane, oxathiane, oxathiepane, dioxolane. The most interesting examples are imidazolidine, piperazine, hexahydropyridazine, hexahydropyrimidine, diazepane, diazocane, pyrrolidine, piperidine, azepane, azocane, azetidine, tropane, oxazinane (morpholine), oxazolane, oxazepane, thiazolane, thiazinane, and thiazepane, in particular imidazolidine, piperazine, hexahydropyridazine, hexahydropyrimidine, diazepane, pyrrolidine, piperidine, azepane, dioxolane, and thiazinane.

The term "alkoxy" is intended to indicate a radical of formula OR*, wherein R* is alkyl as defined above, e.g. methoxy, ethoxy, propoxy, butoxy, etc.

The term "polyoxyethylene" is intended to indicate a group of formula R*(OCH 2 CH 2 )s- / wherein R* is hydrogen or alkyl as defined above and s is an integer of 1-200, such as 1- 100, 1-80, 1-70, 1-60, 1-50, 1-40, 1-30, 1-20, 1-10, 1-8, 1-6, 1-5, 1-4 or 1-3.

The term "alkylaryl" is intended to indicate an alkyl group covalently joined to an aryl group.

The term "carbamido" is intended to indicate the group -NHC(O)NH 2 .

The term "carbamoyl" is intended to indicate the group -C(O)NH 2 .

The term "pharmaceutically acceptable salt" is intended to indicate salts prepared by reacting a compound of formula I with a suitable inorganic or organic acid, e.g. hydrochloric, hydrobromic, hydroiodic, sulfuric, nitric, acetic, phosphoric, lactic, maleic, phthalic, citric, propionic, benzoic, glutaric, gluconic, methanesulfonic, salicylic, succinic, tartaric, toluenesulfonic, sulfamic or fumaric acid.

The compounds of formula I may be in the form of E- or Z-isomers, i.e. they may adopt an E or Z configuration about the double bond connecting the 2-indolinone moiety to the indole moiety or the group denoted R 6 , or they may be a mixture of the E- and Z- isomers. The E- or Z-isomerism of the compounds of formula I is indicated by a zigzag line.

The term ΛΛ indolinone compound" (used synonymously with "oxindole compound" herein) is intended to include compounds of formula I and formula I' as shown herein as well as other, structurally related compounds, such as the compounds disclosed in WO 98/07695, WO 96/22976, WO 00/08202 and WO 98/50356 which are hereby incorporated by reference in their entirety. Methods of preparing the compounds are also disclosed in these publications.

The term "prodrug" is intended to indicate a derivative of an active compound of formula I' which does not, or does not necessarily, exhibit the physiological activity of the active compound, but which may be subjected to enzymatic or other type of cleavage such as hydrolysis in vivo so as to release the active compound of formula I' on administration of the prodrug. In this particular instance, the prodrug comprises the active compound which in itself is highly lipophilic provided with a side chain with predominantly hydrophilic properties imparting improved solubility characteristics to the prodrug, thereby making it more suitable for parenteral administration in the form of a solution or for oral administration to obtain an improved bioavailability, cf. H. Bundgaard, Design of Prodrugs, Elsevier, 1985; H. Bundgaard, Arch. Pharm. Chem. 86(1), 1979, pp. 1-39; D. Fleisher et a\., Adv. Drug Delivery Rev. 19(2), 1996, pp. 115-130; H, Bundgaard, Controlled Drug Delivery 17, 1987, pp. 179-196; Friis and Bundgaard, Eur. J. Pharm. Sci.4, 1996, pp. 49-59; P. Ettmayer et al., J. Med. Chem. 47(10), 2004, pp. 2393-2404.

The term "EAE" or "EAE model" is used herein to denote experimentally induced autoimmune encephalomyelitis (EAE) which is generally recognised as an animal model of multiple sclerosis. EAE may be induced by injection of antigenic peptides of myelin such as myelin basic protein, proteolipid protein and myelin oligodendrocyte glycoprotein. EAE is an inflammatory condition of the central nervous system characterised by T-cell infiltration and focal demyelination. EAE can also be induced by transfer of myelin reactive T-cells to normal individuals.

The term "ameliorate" is intended to mean reducing the severity of the neurological symptoms during relapses of multiple sclerosis by administering an effective amount of an active compound whereby it may be possible to reduce or delay permanent disability resulting from neurological damage sustained during relapse, in particular demyelination.

The term "delay the onset of multiple sclerosis" is used to indicate a prophylactic administration of an effective amount of an active compound to prolong the period

where no symptoms, or at least no severe symptoms, of multiple sclerosis are observed in susceptible individuals, e.g. in first-degree relatives of multiple sclerosis patients.

The term "reduce the relapse rate in multiple sclerosis" is intended to mean reducing the frequency with which relapses occur or, in other words, prolong the periods of remission. This may make it possible to reduce or delay the accumulation of disabilities resulting from the neurological damage sustained during each relapse, in particular demyelination which eventually leads to increasingly severe disability.

Embodiments of the invention

Currently favoured compounds of formula I are compounds wherein R 2 , R 3 , R 4 and R 5 are all hydrogen.

In favoured compounds of formula I, R 1 is -OR 9 , -OC(O)R 9 , -C(O)NR 10 R 11 , -

OC(O)NR 10 R 11 , -S(O) 2 R 9 , -C(O)R 12 , -C(O)OR 12 , -OC(O)OR 12 , -P(O)(OR 10 )(OR 11 ), - OP(O)(OR 10 )(OR 11 ), polyoxyethylene, C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl or aryl, said C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl or aryl being substituted with one or more substituents selected from the group consisting of -OR 10 , -C(O)R 10 , -C(O)OR 10 , - OC(O)R 10 , -OC(O)OR 10 , -NR 10 R 11 , -C(O)NR 10 R 11 , -OC(O)NR 10 R 1U -OP(O)(OR 10 )(OR 11 ),- P(O)(OR 10 )(OR 11 ), -NHC(O)R 10 , -NHC(O)OR 10 , -NHC(O)NR 10 R 11 , -SR 10 , -S(O)R 10 , - S(O) 2 R 10 , -S(O) 2 NR 10 R 11 , -S(O) 2 OR 10 , polyoxyethylene, heteroaryl and heterocyclyl, said heteroaryl and heterocyclyl being optionally substituted with -OR 10 , -C(O)R 10 , -C(O)OR 10 , OC(O)R 10 , -OC(O)OR 10 , -P(O)(OR 10 )(OR 11 ), -OP(O)(OR 10 )(OR 11 ), -NR 10 R 11 , -C(O)NR 10 R 11 , -NHC(O)R 10 , -NHC(O)OR 10 , -NHC(O)NR 10 R 11 , -SR 10 , -S(O)R 10 , -S(O) 2 R 10 , -S(O) 2 NR 10 R 11 , - S(O) 2 OR 10 or polyoxyethylene, and C 1-6 alkyl substituted with -OR 10 , -C(O)R 10 , - C(O)OR 10 , OC(O)R 10 , -OC(O)OR 10 , -NR 10 R 11 , -C(O)NR 10 R 11 , -P(O)(OR 10 )(OR 11 ), - OP(O)(OR 10 )(OR 11 ), -NHC(O)R 10 , -NHC(O)OR 10 , -NHC(O)NR 10 R 11 , -SR 10 , -S(O)R 10 , - S(O) 2 R 10 , -S(O) 2 NR 10 R 11 or -S(O) 2 OR 10 , wherein R 9 , R 10 . R 11 and R 12 are as indicated above.

In other favoured compounds of formula I, R 1 is -OR 9 , -OC(O)R 9 , -C(O)R 12 or C 1-10 alkyl, said C 1-10 alkyl being substituted with one or more substituents selected from the group consisting of polyoxyethylene, -OR 10 , -C(O)R 10 , -C(O)OR 10 , -OC(O)R 10 , -OC(O)OR 10 , - NR 10 R 11 , -C(O)NR 10 R 11 , -OC(O)NR 10 R 117 -OP(O) (OR 10 )(OR 11 )^P(O)(OR 10 )(OR 11 ),

NHC(O)R 10 , -NHC(O)OR 10 , -NHC(O)NR 10 R 11 , -SR 10 , -S(O)R 10 , -S(O) 2 R 10 , -S(O) 2 NR 10 R 11 , - S(O) 2 OR 10 wherein R 9 , R 10 , R 11 and R 12 are as indicated above.

The compounds of formula I may be prodrugs, i.e. converted in vivo to compounds of formula I'

r wherein X, R 2 , R3, R 4 , R5, R 6 , Ri', R 3 ', R 4 ', R5' and R 6 ' are as indicated above for formula I. Compounds of formula I' are disclosed in, i.a., WO 98/07695 and WO 96/22976, in which they are indicated to be inhibitors of tyrosine kinases and as such useful in the treatment of cancer and other proliferative disorders, as well as Parkinson's disease and Alzheimer's disease. It is anticipated that compounds of formula I may also be used for these indications and that they will exhibit improved oral bioavailability compared to the compounds of formula I'. In co-pending international patent application No. PCT2004/DK/000875, filed on 16 December 2004, we have shown that compounds of formula I' show inhibitory activity against EAE induced in mice and are therefore proposed for use in the treatment of multiple sclerosis. However, the activity of compounds of formula I' was low on oral administration when tested in the EAE model.

In currently favoured compounds of formula I (corresponding to compounds of formula lib in the general synthesis methods described in the following), R 1 is

wherein Ri 0 , Rio' and Ru are the same or different and independently selected from the group consisting of hydrogen, C x-10 alkyl, C 2-I0 alkenyl, C 2-I0 alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl, or wherein Ri 0 and Rn, together with the nitrogen atom to which they are attached form a heterocyclic or heteroaryl ring, each of C 1-10 alkyl, C 2-I0 alkenyl, C 2- I 0 alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl being optionally

substituted with one or more, same or different substituents selected from the group consisting of hydrogen, halogen, trihalomethyl, Ci -6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, hydroxy, carboxy, formyl, aryl, heteroaryl, carbocyclyl, heterocyclyl, amino, carbamoyl, cyano, guanidino, carbamido, -OR i3 , -C(O)Ri 3 , -C(O)ORi 3 , -OC(O)Ri 3 , -OC(O)ORi 3 , - NRi 3 Ri 4 , -C(O)NR 13 Ri 4 , -OC(O)NRi 3 R 14 , -NHC(O)R 13 , -NHC(O)ORi 3 , -NHC(O)NRi 3 R 14 , - P(O)(ORi 3 )(ORi 4 ), -OP(O)(OR 13 )(OR 14 ), -SR 13 , -S(O)R 13 , -S(O) 2 R 13 , -S(O) 2 NR 13 R 14 and - S(O) 2 OR 14 , wherein R 13 and R 14 are as indicated above, provided that R 10 and R 11 are not both hydrogen.

In other, currently favoured compounds of formula I (corresponding to compounds formula HIa and HIb in the general synthesis methods described in the following), R 1 is

wherein R 10 and R 10 ' are the same or different and independently selected from the group consisting of hydrogen, C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl, each of C 1-10 alkyl, C 2-I0 alkenyl, C 2-I0 alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl being optionally substituted with one or more, same or different substituents selected from the group consisting of hydrogen, halogen, trihalomethyl, Ci -6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, hydroxy, carboxy, formyl, aryl, heteroaryl, carbocyclyl, heterocyclyl, amino, carbamoyl, cyano, guanidino, carbamido, - ORi 3 , -C(O)Ri 3 , -C(O)ORi 3 , -OC(O)Ri 3 , -OC(O)OR 13 , -NR 13 Ri 4 , -C(O)NRi 3 Ri 4 , - OC(O)NRi 3 Ri 4 , -NHC(O)Ri 3 , -NHC(O)ORi 3 , -NHC(O)NRi 3 Ri 4 , "P(O)(OR 13 )(OR 14 ), - OP(O)(OR 13 )(OR 14 ), -SR 13 , -S(O)Ri 3 , -S(O) 2 R 13 , -S(O) 2 NR 13 R 14 and -S(O) 2 ORi 4 , wherein Ri 3 and R i4 are as indicated above.

In these embodiments R 2 ' may preferably be hydrogen. Alternatively, R 2 ' may be - S(O) 2 R 9 , -C(O)Ri 9 , -C(O)ORi 9 , C 1-I0 alkyl, C 2-I0 alkenyl, C 2-I0 alkynyl or aryl, said Ci -I0 alkyl, C 2-I0 alkenyl, C 2-I0 alkynyl or aryl being substituted with one or more substituents selected from the group consisting of -OR 10 , -C(O)R 10 , -C(O)OR 10 , -OC(O)R 10 , - OC(O)OR 10 , -NR 10 Rn, -P(O)(OR I0 )(OR H ), -OP(O)(OR 10 )(OR 11 ), -C(O)NRi 0 Rn, - OC(0)NRioRn, -NHC(O)Ri 0 , -NHC(O)ORi 0 , -NHC(O)NR 10 R n , -SR 10 , -S(O)R 10 , -S(O) 2 R 10 , â–  S(O) 2 NRi 0 Rn, -S(O) 2 OR 10 , polyoxyethylene, aryl, heteroaryl, carbocyclyl and heterocyclyl, said aryl, heteroaryl, carbocyclyl and heterocyclyl being optionally substituted with -ORi 0 , -C(O)Ri 0 , -C(O)ORi 0 , OC(O)R 10 , -OC(O)ORi 0 , -NR 10 R 11 , -

C(O)NR 10 Ru, -NHC(O)R 10 , -NHC(O)OR 10 , -NHC(O)NR 10 Rn, -SR 10 , -S(O)R 10 , -S(O) 2 R 10 , - S(O) 2 NR 10 R 11 , -S(O) 2 OR 10 or polyoxyethylene, and C 1-6 alkyl substituted with -OR 10 , - C(O)R 10 , -C(O)OR 10 , OC(O)R 10 , -OC(O)OR 10 , -NR 10 R 11 , -C(O)NR 10 R 11 , -P(O)(OR 10 )(OR 11 ), -OP(O)(OR 10 )(OR 11 ), -NHC(O)R 10 , -NHC(O)OR 10 , -NHC(O)NR 10 R 11 , -SR 10 , -S(O)R 10 , - S(O) 2 R 10 , -S(O) 2 NR 10 R 11 or -S(O) 2 OR 10 , wherein R 9 , R 10 , R 11 and R 19 are as indicated above,

More specifically, R 2 ' may be -S(O) 2 R 9 , -C(O)R 19 , C 1-10 alkyl, said C 1-10 alkyl being substituted with one or more substituents selected from the group consisting of -OR 10 , - C(O)R 10 , -C(O)OR 10 , -OC(O)R 10 , -OC(O)OR 10 , -NR 10 R 11 , -C(O)NR 10 R 11 , -OC(O)NR 10 R 11 , - NHC(O)R 10 , -NHC(O)OR 10 , -NHC(O)NR 10 R 11 , -SR 10 , -S(O)R 10 , -S(O) 2 R 10 , -S(O) 2 NR 10 R 11 , - S(O) 2 OR 10 , aryl, heteroaryl, carbocyclyl and heterocyclyl optionally substituted with - OR 10 , -C(O)R 10 , -C(O)OR 10 , , -NR 10 R 11 , -C(O)NR 10 R 11 , -NHC(O)R 10 , -NHC(O)OR 10 , - NHC(O)NR 10 R 11 , -SR 10 , -S(O)R 10 , -S(O) 2 R 10 , -S(O) 2 NR 10 R 11 or -S(O) 2 OR 10 , and C 1-6 alkyl substituted with -OR 10 , -C(O)R 10 , -C(O)OR 10 , -OC(O)R 10 , -OC(O)OR 10 , -NR 10 R 11 , - C(O)NR 10 R 11 , -OC(O)NR 10 R 11 , -NHC(O)R 10 , -NHC(O)OR 10 , -NHC(O)NR 10 R 11 , -SR 10 , - S(O)R 10 , -S(O) 2 R 10 , -S(O) 2 NR 10 R 11 and -S(O) 2 OR 10 , wherein R 9 , R 19 , R 10 and R 11 are as indicated above.

In currently favoured embodiments where R 1 is hydrogen, R 2 ' may be

(corresponding to compounds of formula Hc in the general synthesis methods described in the following) wherein R 10 , R 10 ' and R 11 are the same or different and independently selected from the group consisting of hydrogen, C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl, or wherein R 10 and R 11 , together with the nitrogen atom to which they are attached form a heterocyclic or heteroaryl ring, each of C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl being optionally substituted with one or more, same or different substituents selected from the group consisting of hydrogen, halogen, trihalomethyl, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, hydroxy, carboxy, formyl, aryl, heteroaryl, carbocyclyl, heterocyclyl, amino, carbamoyl, cyano, guanidino, carbamido, -ORi 3 , -C(O)R 13 , -C(O)OR 13 , -OC(O)R 13 , - OC(O)OR 13 , -NR 13 R 14 , -C(O)NR 13 R 14 , -OC(O)NR 13 R 14 , -NHC(O)R 13 , -NHC(O)OR 13 , - NHC(O)NR 13 R 14 , -P(O)(OR 13 )(OR 14 ), -OP(O)(OR 13 )(OR 14 ), -SR 13 , -S(O)R 13 , -S(O) 2 R 13 , -

S(O) 2 NRi 3 Ri 4 and -S(O) 2 ORi 4 , wherein Ri 3 and R i4 are as indicated above, provided that Rio and Rn are not both hydrogen;

(corresponding to compounds of formula VIId, Vila, or VIIb, respectively, in the general synthesis methods described in the following) wherein Ri 0 and Ri 0 ' are the same or different and independently selected from the group consisting of hydrogen, Ci -I0 alkyl, C 2-IO alkenyl, C 2- io alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl, each of Ci -I0 alkyl, C 2-I0 alkenyl, C 2-I0 alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl being optionally substituted with one or more, same or different substituents selected from the group consisting of hydrogen, halogen, trihalomethyl, Ci -6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, hydroxy, carboxy, formyl, aryl, heteroaryl, carbocyclyl, heterocyclyl, amino, carbamoyl, cyano, guanidino, carbamido, -ORi 3 , -C(O)Ri 3 , -C(O)ORi 3 , -OC(O)Ri 3 , - OC(O)ORi 3 , -NRi 3 Ri 4 , -C(O)NRi 3 R 14 , -OC(O)NRi 3 Ri 4 , -NHC(O)Ri 3 , -NHC(O)ORi 3 , - NHC(O)NRi 3 Ri 4 , -P(O)(ORi 3 )(ORi 4 ), -OP(O)(ORi 3 )(ORi 4 ), -SR 13/ -S(O)Ri 3 , -S(O) 2 Ri 3 , - S(O) 2 NRi 3 Ri 4 and -S(O) 2 ORi 4 , wherein Ri 3 and R i4 are as indicated above.

More specifically, R' 2 may be

(corresponding to compounds of formula Hc in the general synthesis methods described in the following) wherein Ri 0 , Rio' and Ru are the same or different and independently selected from the group consisting of hydrogen, Ci -I0 alkyl, heteroaryl, carbocyclyl and heterocyclyl, or wherein R i0 and Rn, together with the nitrogen atom to which they are attached form a heterocyclic or heteroaryl ring, each of Ci -I0 alkyl, heteroaryl, carbocyclyl and heterocyclyl being optionally substituted with one or more, same or different substituents selected from the group consisting of hydrogen, halogen, trihalomethyl, C 1-6 alkyl, hydroxy, carboxy, formyl, aryl, heteroaryl, carbocyclyl, heterocyclyl, amino, carbamoyl, cyano, guanidino, carbamido, -ORi 3 , -C(O)Ri 3 , - C(O)ORi 3 , -NRi 3 Ri 4 , -C(O)NRi 3 R 14 , -NHC(O)Ri 3 , -NHC(O)OR 13 , -NHC(O)NR 13 Ri 4 , - P(O)(ORi 3 )(ORi 4 ), -OP(O)(OR 13 )(ORi 4 ), "SRi 3 , -S(O) 2 R 13 , -S(O) 2 NR 13 R 14 and -S(O) 2 ORi 4 , wherein R i3 and Ri 4 are as indicated above, provided that R 10 and R 11 are not both hydrogen;

or

(corresponding to compounds of formula Vila or VIIb, respectively, in the general synthesis methods described in the following) wherein Ri 0 and R i0 ' are the same or different and independently selected from the group consisting of hydrogen, Ci -I0 alkyl, heteroaryl, carbocyclyl and heterocyclyl, each of C 1-10 alkyl, heteroaryl, carbocyclyl and heterocyclyl being optionally substituted with one or more, same or different substituents selected from the group consisting of hydrogen, halogen, trihalomethyl, Ci -6 alkyl, hydroxy, carboxy, formyl, aryl, heteroaryl, carbocyclyl, heterocyclyl, amino, carbamoyl, cyano, guanidino, carbamido, -OR 13 , -C(O)R 13 , -C(O)OR 13 , -NR 13 Ri 4 , - C(O)NR 13 R 14 , -NHC(O)R 13 , -NHC(O)OR 13 , -NHC(O)NR 13 R 14 , -P(O)(OR 13 )(OR 14 ), - OP(O)(OR 13 )(ORi 4 ), -SR 13 , -S(O) 2 R 13 , -S(O) 2 NR 13 R 14 and -S(O) 2 OR 14 , wherein R 13 and R 14 are as indicated above.

In these embodiments (corresponding to compounds of formula Hc, Vila or VIIb, respectively) R 1 , apart from being hydrogen, may also be

wherein R 10 , R 10 ' and R 11 are the same or different and independently selected from the group consisting of hydrogen, C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl, aryl, heteroaryl, carbocyclyl and heterocyclyl, or wherein Ri 0 and Rn, together with the nitrogen atom to which they are attached form a heterocyclic or heteroaryl ring, each of Ci -I0 alkyl, aryl, heteroaryl, carbocyclyl and heterocyclyl being optionally substituted with one or more, same or different substituents selected from the group consisting of hydrogen, halogen, trihalomethyl, Ci -6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, hydroxy, carboxy, formyl, aryl, heteroaryl, carbocyclyl, heterocyclyl, amino, carbamoyl, cyano, guanidino, carbamido, - ORi 3 , -C(O)R 13 , -C(O)ORi 3 , "OC(O)Ri 3 , -OC(O)ORi 3 , -NRi 3 Ri 4 , -C(O)NR 13 R 14 , - OC(O)NR 13 R 14 , -NHC(O)R 13 , -NHC(O)OR 13 , -NHC(O)NR 13 R 14 , -P(O)(OR 13 )(OR 14 ), - OP(O)(OR 13 )(OR 14 ), -SR 13 , -S(O)R 13 , -S(O) 2 R 13 , -S(O) 2 NR 13 R 14 and -S(O) 2 OR 14 , wherein R 13 and R 14 are as indicated above, provided that R 10 and R 11 are not both hydrogen; or

wherein Ri 0 and Ri 0 ' are the same or different and independently selected from the group consisting of hydrogen, Ci -I0 alkyl, aryl, heteroaryl, carbocyclyl and heterocyclyl, each of Ci -I0 alkyl, aryl, heteroaryl, carbocyclyl and heterocyclyl being optionally substituted with one or more, same or different substituents selected from the group consisting of hydrogen, halogen, trihalomethyl, Ci -6 alkyl, hydroxy, carboxy, formyl, aryl, heteroaryl, carbocyclyl, heterocyclyl, amino, carbamoyl, cyano, guanidino, carbamido, - ORi 3 , -C(O)Ri 3 , -C(O)ORi 3 , -OC(O)Ri 3 , -OC(O)OR 13 , -NR 13 Ri 4 , -C(O)NRi 3 Ri 4 , - OC(O)NRi 3 Ri 4 , -NHC(O)Ri 3 , -NHC(O)OR i3 , -NHC(O)NRi 3 Ri 4 , -P(O)(ORi 3 )(ORi 4 ), -

OP(O)(ORi 3 )(OR 14 ), -SRi 3 , -S(O)R 13 , -S(O) 2 R 13 , -S(O) 2 NR 13 R 14 and -S(O) 2 OR 14 , wherein Ri 3 and R 14 are as indicated above.

In currently favoured compounds of formula I, R 6 is hydrogen.

In currently favoured compounds of formula I, Ri', R 3 ', R 4 ', R 5 ' and R 6 ' are the same or different and independently selected from the group consisting of hydrogen, Ci -I0 alkyl, Ci -I0 alkoxy, aryl, heteroaryl, aryloxy, C 1-10 alkylaryl, C 1-10 alkylaryloxy, halogen, trihalomethyl, -S(O)R 20 , -S(O) 2 R 20 , -S(O) 2 NR 20 R 21 , -S(O) 3 R 20 , -SR 20 , -NO 2 , -NR 20 R 21 , - OR 20 , -CN, -CH 2 OH, C(O)R 20 , -C(O)OR 20 , -OC(O)R 20 , -NHC(O)R 20 , -NHC(O)OR 10 , -

NHC(O)NR 10 R 11 , -(CH 2 )nC(O) 2 R 20 and -C(O)NR 20 R 21 , wherein R 20 is hydrogen, Ci -6 alkyl, heteroaryl or aryl, said Ci -6 alkyl, heteroaryl or aryl being optionally substituted with hydroxy or -NR 22 R 23 , wherein R 22 and R 23 are independently hydrogen or C x-6 alkyl or, together with the nitrogen atom to which they are attached, form a heteroaryl or heterocyclic ring, R n is hydrogen, Ci -6 alkyl or aryl, and n is 0-3. More specifically, R 1 ', R 3 ', R 4 ', R 5 ' and R 6 ' may be the same or different and independently selected from the group consisting of hydrogen, Ci -6 alkyl, C 1-6 alkoxy and halogen.

Examples of specific compounds of formula I are carbonic acid 6-azido-hexyl ester 3-(2-oxo-l,2-dihydro-indol-3-ylidenemethyl)-indol-l- y I methyl ester carbonic acid 6-azido-hexyl ester 3-(2-oxo-l-piperidin-l-ylmethyl-l,2-dihydro-indol-3- ylidenemethyl)-indol-l-ylmethyl ester 4-{2-[3-(2-oxo-l,2-dihydro-indol-3-ylidenemethyl)-indol-l-yl methoxycarbonyloxy]- ethyl}-piperidine-l-carboxylic acid te/t-butyl ester

4-{2-[3-(2-oxo-l-piperidin-l-ylmethyl-l,2-dihydro-indol-3-yl idenemethyl)-indol-l- ylmethoxycarbonyloxy]-ethyl}-piperidine-l-carboxylic acid te/t-butyl ester carbonic acid 2-[2-(2-methoxy-ethoxy)-ethoxy]-ethyl ester 3-(2-oxo-l,2-dihydro-indol-

3-ylidenemethyl)-indol-l-ylmethyl ester acetic acid 3-(2-oxo-l,2-dihydro-indol-3-ylidenemethyl)-indol-l-ylmethyl ester

2,2-dimethyl-propionic acid 3-(2-oxo-l,2-dihydro-indol-3-ylidenemethyl)-indol-l- ylmethyl ester

2,2-dimethyl-propionic acid 3-(2-oxo-l-piperidin-l-ylmethyl-l,2-dihydro-indol-3- ylidenemethyl)-indol-l-ylmethyl ester cyclohexyl-acetic acid 3-(2-oxo-l,2-dihydro-indol-3-ylidenemethyl)-indol-l-ylmethyl ester of /V / Λ/-diethyl-2-[2-methyl-3-(2-oxo-l,2-dihydro-indol-3-y lidenemethyl)-indol-l-yl]- acetamide

2-[3-(2-oxo-l,2-dihydro-indol-3-ylidenemethyl)-indol-l-yl ]-Λ/-(tetrahydro-furan-2- ylmethyl)-acetamide of Λ/-benzo[l,3]dioxol-5-yl-2-[3-(2-oxo-l,2-dihydro-indol-3-yl idenemethyl)-indol-l-yl]- acetamide

Λ/-(2-methoxy-ethyl)-2-[3-(2-oxo-l,2-dihydro-indol-3-yli denemethyl)-indol-l-yl]- acetamide 3-[l-(2-morpholin-4-yl-2-oxo-ethyl)-lH-indol-3-ylmethylene]- l,3-dihydro-indol-2-one

/V-furan-2-ylmethyl-2-[3-(2-Oxo-l,2-dihydro-indol-3-ylide nemethyl)-indol-l-yl]- acetamide

Λ/-cyclohexyl-2-[2-methyl-3-(2-Oxo-l,2-dihydro-indol-3-y lidenemethyl)-indol-l-yl]- acetamide 3-[2-methyl-l-(2-oxo-2-piperidin-l-yl-ethyl)-l/7-indol-3-ylm ethylene]-l,3-dihydro- indol-2-one

2-[2-methyl-3-(2-oxo-l,2-dihydro-indol-3-ylidenemethyl)-i ndol-l-yl]-Λ/-(tetrahydro- furan-2-ylmethyl)-acetamide

Λ/-cyclohexylmethyl-2-[3-(2-oxo-l,2-dihydro-indol-3-ylid enemethyl)-indol-l-yl]- acetamide

4-{2-[3-(2-oxo-l,2-dihydro-indol-3-ylidenemethyl)-indol-l -yl]-acetamino}-butyric acid methyl ester

6-{2-[3-(2-oxo-l,2-dihydro-indol-3-ylidenemethyl)-indol-l -yl]-acetamino}-hexanoic acid ethyl ester 2-[3-(2-oxo-l,2-dihydro-indol-3-ylidenemethyl)-indol-l-yl]-Î ›/-(tetrahydro-furan-2- ylmethyl)-acetamide

^-[Z-ClW-indol-B-yO-ethyll-Z-tS-CZ-oxo-l^-dihydro-indol-B-yl idenemethyO-indol-l-yl]-

/V-(tetrahydro-furan-2-ylmethyl)-acetamide

2-[3-(2-oxo-l, 2-dihydro-indol-3-ylidenemethyl)-indol-l-yl]-Λ/-(3-phenyl-p ropyl)- acetamide 2-[3-(2-oxo-l,2-dihydro-indol-3-ylidenemethyl)-indol-l-yl]-Î ›/-(4-phenyl-butyl)- acetamide

/V-[3-(l-formyl-piperidin-4-yl)-propyl]-2-[3-(2-oxo-l,2-d ihydro-indol-3-ylidenemethyl)- indol-l-yl]-acetamide

Λ/-(4-hydroxy-butyl)-2-[3-(2-oxo-l,2-dihydro-indol-3-yli denemethyl)-indol-l-yl]— acetamide

4-{2-[3-(2-oxo-l,2-dihydro-indol-3-ylidenemethyl)-indol-l -yl]-acetamino}-butyric acid ethyl ester

3-{l-[2-(4-methyl-piperazin-l-yl)-2-oxo-ethyl]-lH-indol-3 -ylmethylene}-l,3-dihydro- indol-2-one 3-{l-[3-(4-methyl-piperazin-l-yl)-2-oxo— propyl]-lW-indol-3-ylmethylene}-l,3-dihydro- indol-2-one

Λ/-(2-hydroxy-ethyl)-3-[3-(2-oxo-l,2-dihydro-indol-3-yli denemethyl)-indol-l-yl]- propionamide

3-(l-benzenesulfonyl-lA/-indol-3-ylmethylene)-l,3-dihydro -indol-2-one 3-(l-benzyl-lW-indol-3-ylmethylene)-l,3-dihydro-indol-2-one l-piperidin-l-ylmethyl-3-(l-piperidin-l-ylmethyl-lH-indol-3- ylmethylene)-l,3-dihydro- indol-2-one l-(4-methyl-piperazin-l-ylmethyl)-3-[l-(4-methyl-piperazin-l -ylmethyl)-lH-indol-3- ylmethylene]-l,3-dihydro-indol-2-one 3-(l/V-Indol-3-ylmethylene)-l-piperidin-l-ylmethyl-l,3-dihyd ro-indol-2-one

3-(lH-Indol-3-ylmethylene)-l-morpholin-4-ylmethyl-l,3-dih ydro-indol-2-one l-(3,3-dimethyl-piperidin-l-ylmethyl)-3-(lW-indol-3-ylmethyl ene)-l,3-dihydro-indol-2- one l-[(te/t-butyl-methyl-amino)-methyl]-3-(lW-indol-3-ylmethyle ne)-l,3-dihydro-indol-2- one

3-{l-[(diisobutylamino)-methyl]-lW-indol-3-ylmethylene}-l ,3-dihydro-indol-2-one

3-(l-methyl-lAy-indol-3-ylmethylene)-l-piperidin-l-ylmeth yl-l,3-dihydro-indol-2-one

3-(l-methyl-2-phenyl-lAy-indol-3-ylmethylene)-l-piperidin -l-ylmethyl-l,3-dihydro- indol-2-one 3-(5-fluoro-l-methyl-lW-indol-3-ylmethylene)-l-piperidin-l-y lmethyl-l,3-dihydro-indol-

2-one

3-(6-methoxy-l-methyl-lW-indol-3-ylmethylene)-l-piperidin-l- ylmethyl-l,3-dihydro- indol-2-one

3-(l-benzenesulfonyl-lW-indol-3-ylmethylene)-l-piperidin- l-ylmethyl-l,3-dihydro-indol-

2-one l-piperidin-l-ylmethyl-3-(4,5,6,7-tetrafluoro-lAy-indol-3-yl methylene)-l,3-dihydro- indol-2-one

3-(7-methyl-lAHndoI-3-ylmethylene)-l-piperidin-l-ylmethyl -l,3-dihydro-indol-2-one

/V-cyclohexyl-2-[2-methyl-3-(2-oxo-l-piperidin-l-ylmethyl -l,2-dihydro-indol-3- ylidenemethyl)-indol-l-yl]-acetamide Λ/-(2-methoxy-ethyl)-2-[3-(2-oxo-l-piperidin-l-ylmethyl-l,2 -dihydro-indol-3- ylidenemethyl)-indol-l-yl]-acetamide

3-[l-(2-morpholin-4-yl-2-oxo-ethyl)-lH-indol-3-ylmethylen e]-l-piperidin-l-ylmethyl- l,3-dihydro-indol-2-one

5-bromo-3-(l/V-indol-3-ylmethylene)-l-piperidin-l-ylmethy l-l,3-dihydro-indol-2-one 7-bromo-3-(lH-indol-3-ylmethylene)-l-piperidin-l-ylmethyl-l, 3-dihydro-indol-2-one

6-bromo-3-(lW-indol-3-ylmethylene)-l-piperidin-l-ylmethyl -l,3-dihydro-indol-2-one

6-fluoro-l-piperidin-l-ylmethyl-3-(l-piperidin-l-ylmethyl -lH-indol-3-ylmethylene)-l,3- dihydro-indol-2-one

6-chloro-3-(lH-indol-3-ylmethylene)-l-piperidin-l-ylmethy l-l,3-dihydro-indol-2-one acetic acid 3-(2-oxo-l-piperidin-l-ylmethyl-l,2-dihydro-indol-3-ylidenem ethyl)-indol-l- yl methyl ester l-hydroxymethyl-3-(lW-indol-3-ylmethylene)-l,3-dihydro-indol -2-one acetic acid l-[3-(l-acetyl-lH-indol-3-ylmethylene)-2-oxo-2,3-dihydro-ind ol-l-yl]-ethyl ester 3-(lW-indol-3-ylmethylene)-l-(tetrahydro-pyran-2-yloxy)-l,3- dihydro-indol-2-one

3-(lH-indol-3-ylmethylene)-l-(2-methoxy-ethoxy)-l,3-dihyd ro-indol-2-one

3-[l-(2-hydroxy-ehtyl)-l/V-indole-3-ylmethylene]-l,3-dihy dro-indol-2-one

3-(lW-indol-3-ylmethylene)-2-oxo-2,3-dihydro-indole-l-car boxylic acid benzyl ester

3-(l-acetoxymethyl-lAy-indol-3-ylmethylene)-2-oxo-2,3-dih ydro-indole-l-carboxylic acid te/t-butyl ester

It is well established that T-cells contribute to the development of several chronic inflammatory and autoimmune diseases. Initially in the disease process, naive T-cells are activated by antigens and produce the proinflammatory cytokine interleukin-2 (IL-2) leading to clonal expansion and production of other inflammatory cytokines involved in the generation of the inflammatory or autoimmune response. Excessive T-cell activity is involved in allergies and immunoinflammatory diseases such as asthma, psoriasis, rheumatoid arthritis and multiple sclerosis. IL-2 has been found to have an important

role in promoting the growth of T-cells in that it is a growth factor for both CD4+ and CD8+ T-cells as well as natural killer cells. Furthermore, IL-2 influences the differentiation of T helper cells into ThI and Th2 cells and potentiates the production of cytokines by each cell type. IL-2 appears to be initially produced by activated CD4+ T- cells, inducing proliferation of CD8+ T-cells and production of proinflammatory cytokines such as IL-I, IL-6 and TNF-α.

Proinflammatory cytokines produced by activated T-cells in the central nervous system are important factors in the demyelination process characteristic of multiple sclerosis (cf. B. Gran and A. Rostami, supra). Proinflammatory cytokines are believed to participate directly in myelin destruction and axonal damage (O'Connor et al., supra) and also to play a role in the upregulation of MHC class II molecules on astrocytes and microglia as well as adhesion molecules on the blood-brain barrier endothelium, facilitating the further influx of T-cells, B-cells and macrophages in the central nervous system (Hellings et al., supra). Such cytokines may also be attractive targets for therapeutic intervention. As shown in example 51 below, compounds of formula I have been found to inhibit IL-2, and this property is believed to be partly responsible for the activity of the compounds in the EAE model.

In a further embodiment, the invention therefore relates to the use of a compound of general formula I capable of inihibiting the production of proinflammatory cytokines, in particular IL-2, by T-cells or capable of blocking a cytokine receptor for the preparation of a medicament for the prevention, treatment or amelioration of multiple sclerosis, or to delay the onset of or reduce the relapse rate in multiple sclerosis.

It is further anticipated that compounds of formula I may be used in the prevention or treatment of other ThI mediated diseases and conditions apart from multiple sclerosis, such as inflammatory bowel disease, Crohn's disease, allergies, asthma, arthritis, e.g. gout or rheumatoid arthritis, inflammatory skin diseases, e.g. psoriasis or atopic dermatitis, neuroinflammatory diseases, systemic vasculitis, arteritis, glomerulonephritis, synovitis, osteomyelitis, autoimmune inner ear disease or sepsis and septic conditions.

It is also anticipated that compounds of formula I may be used as modifiers of angiogenesis, e.g. in the treatment pathological conditions and diseases associated with deregulated angiogenesis such as rosacea, atherosclerosis, hemangioma, warts, pyogenic granuloma, scarring, nasal polyps, transplantation, liver regeneration, bone

and cartilage destruction, pannus growth, osteophyte formation, endometriosis, dysfunctional uterine bleeding, follicular cysts, ovarian hyperstimulation, thyroiditis, thyroid enlargement, obstructive lung disease, diabetic or ischemic retinopathy, neovascular glaucoma, age-related macular degeneration, acute macular degeneration, retinitis, cytomegalovirus retinitis, macular edema, choroidal neovascularisation, or cerebral ischemia reperfusion injury.

Pharmaceutical compositions

For use in the present invention, the compound or compounds of formula I (the "active ingredient") may be formulated into a pharmaceutical composition together with a pharmaceutically acceptable vehicle and optionally one or more other therapeutic ingredients. Such other therapeutic ingredient may be selected from anti-inflammatory drugs such as corticosteroids or non-steroid anti-inflammatory drugs, or immunosuppressive drugs such as methotrexate or cyclophosphamide. For the treatment of multiple sclerosis, it is also envisaged to combine the administration of the compound of formula I with conventional therapeutic agents used in the treatment of multiple sclerosis such as IFN-β or glatiramer acetate.

The vehicle must be "pharmaceutically acceptable" in the sense of being compatible with the other ingredients of the formulation and not deleterious to the recipient thereof. The formulation may be in a form suitable for oral or parenteral (including subcutaneous, intramuscular, interperitoneal, intraarticular and intravenous) administration.

The formulations may conveniently be presented in dosage unit form and may be prepared by any of the methods well known in the art of pharmacy, e.g. as disclosed in Remington, The Science and Practise of Pharmacy , 20 th Ed., 2000. All methods include the step of bringing the active ingredient into association with the vehicle which constitutes one or more excipients. In general, the formulations are prepared by uniformly and intimately bringing the active ingredient into association with a liquid vehicle or a finely divided solid vehicle or both, and then, if necessary, shaping the product into the desired formulation.

The term "dosage unit" is understood to mean a unitary, i.e. a single dose which is capable of being administered to a patient, and which may be readily handled and packed, remaining as a physically and chemically stable unit dose comprising either the

active ingredient as such or a mixture of it with solid or liquid pharmaceutical vehicle materials.

Unlike IFN-β and glatiramer acetate which are peptidic in nature and only suitable for parenteral administration, such by injection, the compounds of formula I are small organic molecules and may therefore be administered orally. This represents a clear benefit for the patient as it permits self-medication and is less painful than for instance injections of IFN-β which often cause pain at the site of injection. Compounds of formula I have surprisingly exhibited an excellent oral bioavailability and EAE inihibitory activity, cf. Table 2 below, and may therefore be suitable for oral administration.

Formulations suitable for oral administration may be in the form of discrete units such as capsules, sachets, tablets or lozenges, each containing a therapeutically effective amount of the active ingredient; in the form of a powder or granules; in the form of a solution or a suspension in an aqueous liquid or non-aqueous liquid, such as ethanol or glycerol; or in the form of an oil-in-water emulsion or a water-in-oil emulsion. Such oils may be edible oils, such as e.g. cottonseed oil, sesame oil, coconut oil or peanut oil. Suitable dispersing or suspending agents for aqueous suspensions include synthetic or natural gums such as tragacanth, alginate, acacia, dextran, sodium carboxymethylcellulose, gelatin, methylcellulose and polyvinylpyrrolidone. The active ingredient may also be administered in the form of a bolus, electuary or paste.

A tablet may be prepared by compressing or moulding the active ingredient optionally with one or more excipients. Compressed tablets may be prepared by compressing, in a suitable machine, the active ingredient(s) in a free-flowing form such as a powder or granules, optionally mixed with a binder, such as lactose, glucose, starch, gelatine, acacia gum, tragacanth gum, sodium alginate, carboxymethylcellulose polyethylene glycol, waxes, hydroxypropylmethylcellulose, microcrystalline cellulose or the like; a lubricant such as sodium oleate, sodium stearate, magnesium stearate, calcium stearate, sodium benzoate, sodium acetate, sodium chloride or the like; a disintegrating agent such as starch, methyl cellulose, agar, bentonite, xanthan gum, croscarmellose, crospovidone, sodium starch glycolate, alginic acid, guar gum, carboxymethylcellulose or the like; or a diluent such as dicalcium phosphate, calcium sulphate, lactose, cellulose, kaolin, mannitol, sorbitol, sucrose or inositol, lactose, mannitol, sorbitol and sucrose also being useful as disintegrating agents. Moulded tablets may be made by moulding, in a suitable machine, a mixture of the powdered active ingredient and suitable carrier moistened with an inert liquid diluent.

Capsule formulations may be in the form of hard or soft capsules, e.g. gelatin capsules, containing the active ingredient in a suitable dry or liquid vehicle. Hard capsules typically contain the active ingredient in powder or crystalline form together with one or more dry excipients which may conveniently be selected from the excipients indicated above for use in tablet formulations. Soft capsules typically contain a solution, suspension or emulsion comprising the active ingredient. Suitable excipients for soft capsules include, for instance, suspension vehicles such as vegetable, animal or mineral oils, e.g. triglycerides of lauric, palmitic, stearic, oleic, caproic or linoleic acid, liquid hydrocarbons, or polyethylene glycol; one or more solvents such as ethanol, propylene glycol, dimethylacetamide, lactic acid, glycerol, butanediol or polyvinylpyrrolidone; one or more surfactants such as fatty alcohols, polyoxyethylene stearate, polysorbates, sorbitol esters or monoglycerides. The compounds of formula I may also be included as a solid solution or dispersion, e.g. prepared by dissolving the active ingredient in a molten carrier, filling the molten mixture into the capsules and letting it cool.

In addition to the aforementioned ingredients, the formulations comprising a compound of formula I may include one or more additional ingredients such as buffers, flavouring agents, colourants, thickeners, preservatives, e.g. methyl hydroxybenzoate (including anti-oxidants), emulsifying agents and the like.

Tablets or capsules may optionally be provided with a coating for modifying the release of the active ingredient therefrom. Suitable coating materials comprise, e.g. mixtures of wax with stearic acid, glyceryl monostearate, palmitic acid, glyceryl monopalmitate, cetyl alcohol, shellac, zein, ethylcellulose, acrylic resins or cellulose acetate or diacetate.

For systemic treatment according to the present invention, daily doses of from 0.001-100 mg/kg body weight, preferably from 0.002-15 mg/kg body weight, for example 0.003-10 mg/kg of a compound of formula I are administered, typically corresponding to a daily dose for an adult human of from 0.2 to 750 mg of the active ingredient. Oral compositions are formulated, preferably as tablets, capsules, or drops, containing from 0.05-250 mg, preferably from 0.1-125 mg, of a compound of formula I per dosage unit.

The invention is further described in the following examples which are not in any way intended to limit the scope of the invention as claimed.

EXPERIMENTAL

GENERAL SYNTHESIS

The compounds of general formula I can be prepared in a number of ways well known to those skilled in the art of organic synthesis. The compounds of formula I can be synthesised using the methods outlined below, together with methods known in the art of synthetic organic chemistry, or variations thereof as appreciated by those skilled in the art. Preferred methods include, but are not limited to, those described below.

The compounds of formula I can be prepared by techniques and procedures readily available to one of ordinary skill in the art, for example by following the procedures as set forth in the following schemes. The reactions are performed in solvents appropriate to the reagents and materials employed and suitable for the transformations being effected. Also, in the synthetic methods described below, it is to be understood that all proposed reaction conditions, including choice of solvent, reaction atmosphere, reaction temperature, duration of experiment and work-up procedures, are chosen to be conditions of standard for that reaction, which should be readily recognised by one skilled in the art. It is understood by one skilled in the art of organic synthesis that the functionalities present on various portions of the starting molecules in a reaction must be compatible with the reagents and reactions proposed. Not all compounds of formula I falling into a given class may be compatible with some of the reaction conditions required in some of the methods described. Such restrictions to the substituents which are compatible with the reaction conditions will be readily apparent to one skilled in the art and alternative methods can be used.

The schemes described in this section are not intended to limit the scope of the invention in any way. All substituents, unless otherwise indicated, are previously defined. The reagents and starting materials are either available from commercial suppliers or prepared by methods known to one of ordinary skilled in the art following procedures set forth in references such as Fieser and Fieser's Reagents for Organic Synthesis, Volumes 1-22 (John Wiley and Sons, 2004); Rodd's Chemistry of Carbon Compounds, Volumes 1-5 and Supplements (Elsevier Science Publishers, 2000); Organic Reactions, Volumes 1-64 (John Wiley and Sons, 2004); March's Advanced Organic Chemistry (John Wiley and Sons, 5 th Edition) and Larock's Comprehensive Organic Transformations (VCH Publishers Inc., 1999). These schemes are merely illustrative of some methods by which the compounds of this invention can be synthesised, and various modifications to these schemes can be made and will be suggested to one skilled

in the art having referred to this disclosure. The starting materials and the intermediates of the reactions may be isolated and purified if desired using conventional techniques, including but not limited to filtration, distillation, crystallisation, chromatography and the like. Such materials may be characterised using conventional means, including physical constants and spectral data.

The introduction of Ri (R 1 ≠ H) may occur at different stage of the synthesis. The precursor of R 1 may be purchased or synthesised as a reactive species. R 1 may be chemically modified after its introduction on the indolinone, before or after condensation to the indole-3-carbaldehyde / indole-3-ketone. Some examples for the introduction of R 1 are described in, but not limited to, the following section.

The introduction of R 2 ' (R 2 ' ≠ H) may occur at different stage of the synthesis. The precursor of R 2 ' may be purchased or synthesised as a reactive species. R 2 ' may be chemically modified after its introduction on the indole, before or after condensation to the indolinone. Some examples for the introduction of R 2 ' are described in, but not limited to, the following section.

Possibly R 1 and R 2 ' may be identical or may contain identical functions or may contain reactive functions under reaction conditions. Therefore modifications of R 1 or R 2 ' may also modify R 2 ' or R 1 respectively. These implications will be obvious to one skilled in the art.

In the same manner possible protecting groups may be added and removed at different step of the synthesis.

General method A: Some of the compounds of general formula I can be prepared according to the general method A described below.

Scheme 1

a. Base such as nitrogen base or inorganic base. "Nitrogen bases" are selected from acyclic and cyclic amines. Examples of nitrogen bases include, but are not limited to, ammonia, methylamine, trimethylamine, triethylamine, aniline, 1,8- diazabicyclo-[5.4.1]-undec-7-ene, diisopropyl ethylamine, pyrrolidine, piperidine, morpholine, and pyridine or substituted pyridine {e.g., 2,6-di-te/tbutylpyridine). "Inorganic bases" are bases that do not contain any carbon atoms. Examples of inorganic bases include, but are not limited to, hydroxide, phosphate, bisulfate, hydrosulfide (SH " ), and amide anions. Preferred nitrogen bases are piperidine and morpholine. Preferred inorganic bases are hydroxide anion, preferably used as its sodium or potassium salt. The reaction generally takes place in a protic solvent such as water or alcohols or in an aprotic solvent such as toluene, CH 2 CI 2 , THF, DMF. Most preferred solvents are alcohols such as ethanol, and CH 2 CI 2 .

General method B Ri and R 2 ' may be introduced by alkylation of the nitrogen of the parent compound where Ri = H and R 2 ' = H, respectively. In the following scheme, introduction of R x is depicted.

Scheme2

b. Alkylation by RiX in the presence of a base such as NEt 3 , NaH, NaOH, KOH, carbonates in an appropriate solvent such as DMF, pyridine, DMSO, CH 3 CN, acetone, toluene. X is a leaving group such as chloride, bromide, and iodide. Alternatively reaction with an alcohol RiOH may also be considered. Such reaction

is run in the presence of a phosphine such as PBu 3 , PPh 3 and the like, an azodicarboxylate or an azodicarboxamide in an aprotic solvent, typically THF.

General method C

Some of the compounds of the present disclosure have the general formula II. Scheme 3 illustrates the synthesis of II.

Scheme 3

H, and excess of reagents

lie if R 1 # H and R 2 ' = H or if R 1 = H and R 2 ' = H depending on the reactants

c. Reaction with an aldehyde such as formaldehyde, acetaldehylde, and the like, and a suitable amine affords a compound of general formula II. The solvent in which the reaction is carried out may be a protic or an aprotic solvent, preferably it is a protic solvent such as an alcohol e.g., methanol or ethanol, or an aqueous alcohol. The reaction may be carried out at temperatures greater than room temperature. The temperature is generally from about 20 0 C to about 100 0 C, preferably about 40 0 C to about 80 0 C. By "about" is meant that the temperature

range is preferably within 10 degrees Celsius of the indicated temperature, more preferably within 5 degrees Celsius of the indicated temperature, most preferably within 2 degrees Celsius of the indicated temperature. Suitable amines include acyclic and cyclic secondary amines such as diethyl amine, N- tert- butyl methylamine, diisobutylamine, piperidine, morpholine, /V-methyl piperazine, 3,3- dimethylpiperidine and the like.

Alternatively, compounds of general formula II can be prepared via reaction of compounds of general formula I where R 1 = H and a preformed Mannich reagent such as an aminal, an /V,O-acetal, a benzotriazole aminal or an iminium salt in the presence of a base such as NEt 3 , t-BuOK, n-BuLi.

Compounds of general formula II where Ri 0 and Ru combine to form a heteroaryl ring, may be prepared by reacting I where Ri = H with a suitable aldehyde Rio'CHO to yield a C-substituted Λ/-hydroxymethyl intermediate (see conditions d) and reacting the intermediate with phosphorus oxychloride and a suitable heteroaryl such as pyridine, pyrrole, oxazolyl, imidazolyl, and the like. The reaction may be carried out at temperature less than r.t..

General method D

Scheme 4 is depicting the synthesis of compounds of general formula III.

Scheme 4

~OR 1 1 0 0

IMc iiid

d. Reaction with an aldehyde of formula R i0 'CHO in the presence of a base, such as an organic base, preferably a tertiary nitrogen base such as NMe 3 , NEt 3 , pyridine, diisopropylethylamine, l,8-diazabicyclo-[5.4.1]-undec-7-ene, and the like, or alkali bases such as KOH, NaOH, NaH, BuLi, LDA in an aprotic solvent such as acetonitrile, DMF, DMSO, THF or pyridine.

e. To obtain Ilia reaction with an acylating agent such as acid anhydride (e.g. acetic anhydride, succinic anhydride), acid halides (e.g. acetyl chloride, propionyl chloride) and carboxylic acid active esters (e.g. p-nitrophenyl ester, pentafluorophenyl ester). The reaction is carried out in an organic base such as pyridine, DMAP and the like. In a similar fashion IHb can be obtained by reaction with chloroformates; IHc may be obtained by reaction with carbamoyl chlorides or icocyanates. Formation of IHd may be achieved by alkylation of the intermediate alcohol following b. f. To obtain Ilia reaction with an aldehyde and a suitable acylating agent as described in c. and d. respectively without isolating the intermediate alcohol. Alternatively R 1 = CHRi 0 'OCOR 10 may be introduced by alkylation of I where Ri = H by RiX following b. IHb and IHc may also be prepared by alkylation. A synthesis of RiCI is exemplified in scheme 5 (for R x = CHRi 0 'OCORi 0 , CHRio'OCOORio, CHRio'OCONRioRu).

Scheme 5 is an example but one skilled in the art may consider alternative method for the synthesis of such alkylating agent. Scheme 5 exemplifies the synthesis of carboxylic esters, carbonic esters, and carbamates. The chlorine atom may be interchanged with iodide with methods known to those skilled in the art such as NaI in acetone.

Scheme 5

X = leaving group

g. HClSO 3 in an aprotic solvent such as pentane, CH 2 CI 2 , CHCI 3 , THF at temperature below room temperature (Synth. Communic. 14(9), 857 (1984))

h. Carboxylic acid or carbonic acid or carbamic acid in the presence of a base such as alkali carbonate, alkali bicarbonate, alkali hydroxide and a phase transfer catalyst such as tetra n-butylammonium hydrogensulfate, tetra n- butylammonium chloride in a mixture of water and an aprotic solvent such as

CH 2 CI 2 , Et 2 O, CHCI 3 .

i. Reaction with RioOH in the neat alcohol or in a suitable solvent such as CH 2 CI 2 ,

CHCI 3 , THF, diethyl ether and DMF optionally in the presence of an acid such as hydrochloric acid, trifluoroacetic acid, and the like or a base such as pyridine, NEt 3 , and the like.

j. Reaction with HNR 10 Ru in a suitable solvent such as CH 2 CI 2 , CHCI 3 , THF, diethyl ether, dioxane, DMSO and DMF, eventually with water as a co-solvent, optionally in the presence of a non-nucleophilic base such as pyridine, NaHCO 3 , NEt 3 .

General method E

Compounds of general formula IV may be obtained by either direct acylation (k) or in two steps as shown in the following schemes.

Scheme 6

IVa IVb IVc

k. Acylation may be obtained via reaction with an acylating agent such as acid anhydride (e.g. acetic anhydride, succinic anhydride), acid halides (e.g. acetyl chloride, propionyl chloride), carboxylic acid active esters (e.g. p-nitrophenyl

ester, pentafluorophenyl ester), chloroformates and carbamoyl chlorides. The reaction is carried out in an organic base such as pyridine, DMAP and the like or in an aprotic solvent such as CH 3 CN, DMF in the presence of a base such as NEt 3 , DMAP, pyridine or NaH.

Scheme 7

IVc

I. For examples for LG = Cl, the intermediate can be prepared by treatment of I (Ri = H) with triphosgene in the presence of an organic amine such as NEt 3 , pyridine and the like. A compound where LG is imidazol-1-yl can be prepared by treatment of I (R 1 = H) with carbonyl diimidazole under conditions well known in the art.

General method F

If Ri = OR 9 , derivatisation of l-hydroxy-3-(lW-indol-3-ylmethylene)-l,3-dihydro-indol-

2-one may be used.

Scheme 8

m

V

The hydroxyl functionality may be alkylated following b using R 9 X (X = leaving group) instead of R 1 X in an appropriate solvent such as DMSO, toluene, DMF or CH 3 CN in the presence of a base such as NaOH, KOH, K 2 CO 3 , Cs 2 CO 3 and the like. Reaction with an alcohol R 9 OH (instead of RiOH) may also be considered. (See b)

m. Addition to double bonds may also be considered to afford I where R 1 = OR 9 . The reaction may be catalysed by bases or by acids such as p-toluene sulfonic acid, pyridinium p-toluenesulfonate, POCI 3 , trimethylsilyliodide and the like.

The hydroxyl functionality may be acylated following e.

General method G

If Rl contains a carboxylic functionality, it may be modified as described in the following scheme, wherein n is an integer of from 1 to 10.

Scheme 9

Vl

n. Standard hydrolysis conditions using first a base such as an aqueous solution of LiOH or NaOH followed by treatment with an acid such as an aqueous solution of HCI. o. Esterification using classical esterification conditions (see March's Advanced Organic Chemistry Reactions, mechanisms, and Structure, 5 th edition, by M. B. Smith and J. March chapter 10-23 p. 484, chapter 10-26 p. 488 and chapter 10- 28 p. 490) such as acid catalysed esterification (e.g. using sulphuric acid as a catalyst in MeOH), or such as base catalyzed esterification (e.g. using chlorosulfates as reagents under phase transfer conditions, see Synthetic Communications, 14 (9), 857-864, 1984) or such as using diazomethane in a suitable solvent such as Et 2 O or EtOH, or using trimethylsilyldiazomethane in a suitable solvent such as toluene, or using an electrophilic reactant (as for example benzyl bromide or methyliodide) in the presence of a base such as

K 2 CO 3 or Cs 2 CO 3 in a solvent such as DMF or acetonitrile. p. Formation of the amide using cyclic and acyclic amines in the presence of classical coupling agents. Preferred coupling agents include 1,1'- carbonyldiimidazole (CDI), diphenylphoshinic chloride (DPP-CI), benzotriazol- yloxy-tripyrolidinophosphonium hexafluorophosphate (PyBOP), benzotriazol-1- yloxy-tris(dimethylamino)-phosphonium hexafluorophosphate (BOP), N, N'- dicyclohexylcarbodiimide (DCC), or l-ethyl-3-(3-dimethylaminopropyl)- carbodiimide; hydrochloride (EDCI). Preferred bases include

diisopropylethylamine, triethylamine, 4-methylmorpholine, or pyridine or a substituted pyridine, for example 4-dimethylaminopyridine or 2,6- dimethylpyridine. Preferred solvents are solvents such as diethylether, dichloromethane, tetrahydrofuran, l-methyl-2-pyrrolidinone, dimethylsulfoxyde or dimethylformamide. The reactions are generally carried out in the presence of a base such as Et 3 N or Bu 3 N and in the presence of an activator such as HOBt (for example where HOBt is used to improve reactions rates, see Windridge, G. C; Jorgensen, E. C. JACS 1971, 93, 6318), at a temperature between about -78 0 C to about 60 0 C, and are normally complete within about 2 hours to about 5 days.

Most of the chemistry described in the above schemes may apply to the introduction of substituents on the indole moiety (R 2 ' # H) providing that the substituents are compatible with the chemistry in question, which will be obvious to one skilled in the art and providing that the group to be introduced enters into the definition of R 2 '. Some specific examples are described in the following section.

R 2 ' may be introduced by alkylation of the parent indole as depicted in scheme 10.

Scheme 10

Compounds of general formula VIId may be prepared as in the following scheme. Alkylation of the intermediate alcohol can be performed following b using R 10 X instead of RiX.

Scheme 11

Analogous to the general method G, scheme 12 exemplifies a modification of R 2 ' after condensation with the indolinone.

Scheme 12

Examples

For 1 H nuclear magnetic resonance (NMR) spectra (300 MHz) and 13 C NMR (75.6 MHz) chemical shift values (δ) (in ppm) are quoted for dimethyl-α^ sulfoxide (DMSO-c/ 6 ) or CDCI 3 solutions relative to internal tetramethylsilane (δ = 0) standard. The value of a multiplet, either defined (doublet (d), triplet (t), quartet (q)) or not (m) at the approximate mid point is given unless a range is quoted, (bs) indicates a broad singlet. The ES mass spectra were obtained on a VG Quattro II triple quadrapole mass spectrometer (Micromass, Manchester, UK) operating in either positive or negative electrospray mode with a cone voltage of 30V.

The compounds of the present invention can exist in two isomeric forms: the Z and the E isomeric forms. The NMR data characterize the isomer forms that are present in the solvent used to record the NMR spectrum and determine their molar ratio. For NMR solutions where both the E-isomer and the Z-isomer are present in equal or close to equal amounts, the chemical shifts of both forms are given. For NMR solutions where the equilibrium is shifted in favour of one form, the chemical shifts of the dominating form are given.

The organic solvents used were anhydrous unless otherwise specified. Flash chromatography was performed on silica gel or Sephadex LH-20 from Fluka Chemie

GmbH, Switzerland. Some of the starting materials have been described in international patent application number PCT/DK2004/000875.

The following abbreviations have been used throughout: Ac Acetate or acetyl aq. Aqueous

Brine Saturated aqueous sodium chloride

Boc te/t-Butoxycarbonyl

DMAP 4-Dimethylaminopyridine DMF /^/V-Dimethylformamide

DMSO Dimethylsulfoxide

EDAC l-[3-(dimethylamino)propy]]-3-ethylcarbodiimide hydrochloride

EDCI l-ethyl-3-(3-dimethylaminopropyl)-carbodiimide

EtOAc Ethyl acetate eq. Equivalent

HOBt 1-hydroxybenzotriazole

LDA Lithium diisopropylamine

LG Leaving group

M Molar (mol/L)

NMR Nuclear magnetic resonance

PE Petroleum ether (bp 40-60 0 C) r.t. Room temperature sat. Saturated

TBAF Tetrabutyl ammonium fluoride

THF Tetrahydrofuran

TLC Thin layer chromatography

Example 1: synthesis of carbonic acid 6-azido-hexyl ester 3-(2-oxo-l,2-dihydro-indol-3- ylidenemethyl)-indol-l-ylmethyl ester and carbonic acid 6-azido-hexyl ester 3-(2-oxo-l- piperidin-l-ylmethyl-l,2-dihydro-indol-3-ylidenemethyl)-indo l-l-ylmethyl ester

6-Azidohexan-l-ol (1) NaN 3 (10.8 g, 165.7 mmol) was added to a DMF (50 mL) solution of 6-bromohexan-l-ol (10 g, 55.2 mmol). The suspension was stirred at 80 0 C for 3 h and allowed to come to r.t. H 2 O (300 mL) was added and the mixture was extracted with Et 2 O (3 x 100 mL). The combined extracts were washed with H 2 O (2 x 100 mL) and brine (2 x 100 mL), dried

over Na 2 SO 4 , filtered and concentrated in vacuo to afford 6-azidohexan-l-ol (6.3 g,

80%).

13 C NMR (CDCI 3 ) δ 62.7, 51.4, 32.6, 28.8, 26.5, 25.4

Carbonic acid 6-azido-hexyl ester chloromethyl ester and carbonic acid 6-azido-hexyl ester iodomethyl ester (2)

A solution of 6-azidohexan-l-ol (2.9 g, 20 mmol) and pyridine (1.9 ml_, 24 mmol) in CH 2 CI 2 (15 mL) was cooled to 0 0 C. A solution of chloroformic acid chloromethyl ester (2.8 g, 22 mmol) in CH 2 CI 2 (8 mL) was added dropwise over 45 min at such a rate that the internal temperature did not raise over 10 0 C. A precipitate appeared. The ice-bath was removed and the mixture was stirred at r.t. overnight. The organic layer was washed with 1 M HCI (2 x 30 mL), sat. aq. NaHCO 3 and brine, dried over Na 2 SO 4 , filtered and concentrated in vacuo to afford carbonic acid 6-azido-hexyl ester chloromethyl ester (4 g) which was used without further purification. 1 H NMR (CDCI 3 ) δ 5.73 (s,2H), 4.23 (t,2H), 3.27 (t,2H), 1.72 (m,2H), 1.62 (m,2H), 1.42 (m,4H)

To a solution of carbonic acid 6-azido-hexyl ester chloromethyl ester (4 g, 17 mmol) in acetone (20 mL) was added NaI (10.1 g, 68 mmol). The mixture was stirred at 40 0 C for 3 h before being concentrated in vacuo. The residue was taken in CH 2 CI 2 (50 mL) and washed with H 2 O, sat. aq. NaHCO 3 , 0.1 M Na 2 S 2 O 3 and brine, dried over Na 2 SO 4 , filtered and concentrated in vacuo. Purification by flash chromatography (PE - EtOAc) afforded carbonic acid 6-azido-hexyl ester iodomethyl ester (4.5 g, 81%). 13 C NMR (CDCI 3 ) δ 153.2, 69.1, 51.3, 34.0, 28.7, 28.4, 26.3, 25.2

General procedure A

To a solution of indole-3-carboxaldehyde in DMF was added NaH (60% in oil, 1.1 eq.).

The mixture was stirred at r.t. for 30 min before cooling to 0 0 C and addition of halogenomethyl ester (1.1 eq.). The ice-bath was removed and the mixture was stirred at r.t. for 2 h. H 2 O was added. The solution was extracted with Et 2 O (3 x). The combined extracts were washed with water, brine, dried over Na 2 SO 4 , filtered, concentrated in vacuo and purified.

Carbonic acid 6-azido-hexyl ester 3-formyl-indol-l-ylmethyl ester (3)

General procedure A was followed using 0.93 g of indole-3-carboxaldehyde (6.4 mmol) in 20 mL of DMF, 0.28 g of NaH and 2.3 g of carbonic acid 6-azido-hexyl ester iodomethyl ester. Flash chromatography afforded carbonic acid 6-azido-hexyl ester 3- formyl-indol-1-ylmethyl ester (1.7 g, 77%).

1 H NMR (CDCI 3 ) δ 10.05 (s,lH), 8.31 (m,lH), 7.93 (s,lH), 7.55 (m,lH), 7.38 (m,2H), 6.13 (s,2H), 4.15 (t,2H), 3.22 (t,2H), 1.73 - 1.49 (m,4H), 1.36 (m,4H)

General procedure B

A solution of indolinone and /V-substituted indole-3-carboxaldehyde (1 eq.) in CH 2 CI 2 was stirred at r.t. overnight. Piperidine (3 eq.) was added and the mixture was heated to reflux for 1 h. The precipitate was filtered and washed with cold CH 2 CI 2 . If no precipitation occurred, the solvent was removed and the product was purified by flash chromatography.

Carbonic acid 6-azido-hexyl ester 3-(2-oxo-l,2-dihydro-indol-3-ylidenemethyl)-indol-l- ylmethyl ester (4) and carbonic acid 6-azido-hexyl ester 3-(2-oxo-l-piperidin-l- ylmethyl-l,2-dihydro-indol-3-ylidenemethyl)-indol-l-ylrnethy l ester (5)

General procedure B was followed using 0.7 g of indolinone (5 mmol), 1.7 g of carbonic acid 6-azido-hexyl ester 3-formyl-indol-l-ylmethyl ester and 1.5 ml_ of piperidine in 25 ml_ of CH 2 CI 2 . Flash chromatography on silica gel afforded 1.25 g of an impure product which crystallised upon treatment with Et 2 O. The crystals were filtered and further purified on a Sephadex column (150 g, eluent: CH 2 CI 2 - MeOH - hexane 78:2:20) to afford 0.30 g (11%) of carbonic acid 6-azido-hexyl ester 3-(2-oxo-l-piperidin-l- ylmethyl-l,2-dihydro-indol-3-ylidenemethyl)-indol-l-ylmethyl ester and 0.34 g (15%) of carbonic acid 6-azido-hexyl ester 3-(2-oxo-l,2-dihydro-indol-3-ylidenemethyl)-indol-l- ylmethyl ester.

13 C NMR (DMSO-CZ 6 ) δ 167.8, 153.8, 139.6, 135.9, 135.5, 128.8, 127.5, 125.5, 125.1, 123.3, 121.8, 121.6, 120.6, 119.2, 118.8, 112.0, 110.8, 109.0, 71.7, 68.0, 50.4, 27.9, 27.7, 25.6, 24.5 13 C NMR (DMSO-c/e) δ 166.8, 153.8, 140.9, 136.2, 135.5, 128.9, 127.4, 126.2, 123.9, 123.4, 121.9, 121.2, 120.2, 118.9, 118.8, 112.0, 110.9, 109.5, 71.8, 68.0, 61.8, 51.6, 50.4, 28.0, 27.7, 25.6, 25.3, 24.5, 23.6

Example 2: synthesis of 4-{2-[3-(2-oxo-l,2-dihydro-indol-3-ylidenemethyl)-indol-l- ylmethoxycarbonyloxy]-ethyl}-piperidine-l-carboxylic acid te/t-butyl ester, 4-{2-[3-(2- oxo-l-piperidin-l-ylmethyl-l,2-dihydro-indol-3-ylidenemethyl )-indol-l- ylmethoxycarbonyloxy]-ethyl}-piperidine-l-carboxylic acid tert-butyl ester

1. CH 2 CI 2 , pyridine bocv

4-(2-Hydroxy-ethyl)-piperidine-l-carboxylic acid te/t-butyl ester (6)

To an ice-cold solution of 4-(2-hydroxyethyI) piperidine (4.25 g, 33 mmol) in CH 2 CI 2 (5 ml_) was added a solution of di-te/t-butyl dicarbonate (7.2 g, 33 mmol) in CH 2 CI 2 (30 ml.) over 1 h. The mixture was stirred at r.t. overnight. The solvent was removed. The residue was dissolved in Et 2 O (5OmL), washed with 1 M aq. NaH 2 PO 4 , sat. aq. NaHCO 3 and brine, dried over Na 2 SO 4 , filtered and concentrated in vacuo to afford 6.9 g (91%) of the title compound.

13 C NMR (CDCI 3 ) δ 154.9, 79.3, 60.1, 44.0, 39.3, 32.5, 32.1, 28.5

4-(2-Chloromethoxycarbonyloxy-ethyl)-piperidine-l-carboxy lic acid te/t-butyl ester and 4-(2-iodomethoxycarbonyloxy-ethyl)-piperidine-l-carboxylic acid te/t-butyl ester (7) A solution of 4-(2-hydroxy-ethyl)-piperidine-l-carboxylic acid te/t-butyl ester (6.9 g, 30 mmol) and pyridine (2.9 mL, 36 mmol) in CH 2 CI 2 (20 mL) was cooled to O 0 C. A solution of chloroformic acid chloromethyl ester (4.26 g, 33 mmol) in CH 2 CI 2 (12 mL) was added dropwise over 1 h at such a rate that the internal temperature did not exceed 10 0 C. A precipitate appeared. The ice-bath was removed and the mixture was stirred at r.t. overnight. The organic layer was washed with 1 M HCI (2 x 50 mL), sat. aq. NaHCO 3 and brine, dried over Na 2 SO 4 , filtered and concentrated in vacuo to afford 8.6 g (89%) of 4-

(2-chloromethoxycarbonyloxy-ethyl)-piperidine-l-carboxylic acid te/t-butyl ester which was used without further purification.

1 H NMR (CDCI 3 ) δ 5.73 (s, 2H), 4.28 (t, 2H), 4.09 (d, 2H), 2.69 (t, 2H), 1.75-1.50 (m, 5H), 1.45 (s, 9H), 1.15 (m, 2H) To a solution of 4-(2-chloromethoxycarbonyloxy-ethyl)-piperidine-l-carboxylic acid tert- butyl ester (4.9 g, 15 mmol) in acetone (20 mL) was added NaI (9 g, 60 mmol). The mixture was stirred at 40 0 C for 3 h before being concentrated in vacuo. The residue was taken in CH 2 CI 2 (50 mL) and washed with sat. aq. NaHCO 3 , 0.1 M Na 2 S 2 O 3 and brine, dried over Na 2 SO 4 , filtered and concentrated in vacuo. Purification by flash chromatography (PE - EtOAc) afforded 4-(2-iodomethoxycarbonyloxy-ethyl)-piperidine- 1-carboxylic acid tert-butyl ester (5.3 g, 85.5%).

1 H NMR (CDCI 3 ) δ 5.95 (s, 2H), 4.28 (t, 2H), 4.09 (d, 2H), 2.69 (t, 2H), 1.75-1.50 (m, 5H), 1.45 (s, 9H), 1.13 (m, 2H)

4-[2-(3-Formyl-indol-l-ylmethoxycarbonyloxy)-ethyl]-piper idine-l-carboxylic acid tert- butyl ester (8)

General procedure A was followed using 0.3 g of indole-3-carboxaldehyde (2.0 mmol) in 6 mL of DMF, 88 mg of NaH and 0.9 g of 4-(2-iodomethoxycarbonyloxy-ethyl)- piperidine-1-carboxylic acid te/t-butyl ester. The product crystallised upon concentration.

Filtration afforded 0.65 g (75%) of the title compound.

1 H NMR (DMSO-Cf 6 ) δ 9.99 (s,lH), 8.43 (s,lH), 8.12 (d,lH), 7.73 (d,lH), 7.38 (t,lH),

7.32 (t,lH), 6.33 (s,2H), 4.16 (t,2H), 3.86 (d,2H), 2.60 (t,2H), 1.55 (d,2H), 1.50 (t,2H), 1.41 (m,lH), 1.37 (s,9H), 0.94 (m,2H)

4-{2-[3-(2-Oxo-l,2-dihydro-indol-3-ylidenemethyl)-indol-l -ylmethoxycarbonyloxy]- ethyl}-piperidine-l-carboxylic acid te/t-butyl ester (9), 4-{2-[3-(2-Oxo-l-piperidin-l- ylmethyl-l,2-dihydro-indoI-3-ylidenemethyl)-indol-l-ylmethox ycarbonyloxy]-ethyl}- piperidine-1-carboxylic acid tert-butyl ester (10)

General procedure B was followed using 0.8 g of indolinone (6 mmol), 2.6 g of 4-[2-(3- formyl-indol-l-ylmethoxycarbonyloxyj-ethyπ-piperidine-l-car boxylic acid tert-butyl ester and 1.8 mL of piperidine in 30 mL of CH 2 CI 2 . Flash chromatography on silica gel afforded 2.1 g of an impure product which crystallised upon treatment with Et 2 O. The crystals were filtered and further purified on a Sephadex column (150 g, eluent: CH 2 CI 2 - MeOH - hexane 78:2:20) to afford 0.47 g (14%) of 4-{2-[3-(2-oxo-l,2-dihydro-indol- 3-ylidenemethyl)-indol-l-ylmethoxycarbonyloxy]-ethyl}-piperi dine-l-carboxylic acid te/t-butyl ester. One of the fractions was concentrated and run on Sephadex column (150 g, eluent: CH 2 CI 2 - MeOH - hexane 78:2:20) to afford 0.2 g (5%) of 4-{2-[3-(2- Oxo-l-piperidin-l-ylrnethyl-l,2-dihydro-indol-3-ylidenemethy l)-indol-l- ylmethoxycarbonyloxyj-ethy^-piperidine-l-carboxylic acid te/t-butyl ester. 13 C NMR (DMSO-Cf 6 ) δ 167.8, 153.8, 153.7, 139.6, 135.9, 135.5, 128.8, 127.5, 125.5,

125.1, 123.3, 121.8, 121.6, 120.6, 119.2, 118.8, 112.0, 110.8, 109.0, 78.3, 71.8, 65.9, 34.3, 31.8, 31.3, 28.0

13 C NMR (DMSO-de) δ E/Z mixture 168.5, 166.8, 153.9, 153.8, 153.7, 143.3, 140.9,

136.2, 135.8, 135.5, 132.7, 128.9, 127.9, 127.4, 126.8, 126.2, 123.9, 123.6, 123.4, 122.5, 122.0, 121.9, 121.5, 121.2, 120.9, 120.2, 119.2, 118.9, 112.0, 111.3, 110.9,

109.9, 109.5, 79.1, 78.3, 71.8, 65.9, 61.8, 51.6, 51.5, 43.2, 34.3, 31.8, 31.3, 28.0, 25.3, 23.6

Example 3: synthesis of carbonic acid 2-[2-(2-methoxy-ethoxy)-ethoxy]-ethyl ester 3- (2-oxo-l,2-dihydro-indol-3-ylidenemethyl)-indol-l-ylmethyl ester

Carbonic acid 3-formyl-indol-l-ylmethyl ester 2-[2-(2-methoxy-ethoxy)-ethoxy]-ethyl ester

General procedure A was followed using 2.26 g of indole-3-carboxaldehyde (5.5 mmol) in 25 mL of DMF, 1.7 g of NaH (4.5 eq.) and 4 g of carbonic acid chloromethyl ester 2- [2-(2-methoxy-ethoxy)-ethoxy]-ethyl ester (prepared as described in the 2 precedent schemes). Purification by flash-chromatography using a gradient of eluent EtOAC - petroleum ether from 50:50 to 100:0, afforded 0.58 g (10%) of the title compound. 1 H NMR (DMSOd 6 ) δ 9.99 (s,lH), 8.43 (s,lH), 8.12 (d,lH), 7.74 (d,lH), 7.36 (m,2H), 6.34 (s,2H), 4.22 (m,2H), 3.59 (m,2H), 3.52 - 3.32 (m,8H), 3.20 (s,3H)

Carbonic acid 2-[2-(2-methoxy-ethoxy)-ethoxy]-ethyl ester 3-(2-oxo-l,2-dihydro-indol- 3-ylidenemethyl)-indol-l-ylmethyl ester

General procedure B was followed using 208 mg of indolinone (1.56 mmol), 570 mg of carbonic acid 3-formyl-indol-l-ylmethyl ester 2-[2-(2-methoxy-ethoxy)-ethoxy]-ethyl ester (1.56 mmol) and 0.7 mL of piperidine in 12 ml_ of CH 2 CI 2 . After 24h, the crude was concentrated and purified on a Sephadex column (eluent: CH 2 CI 2 - MeOH - hexane 78:2:20) to afford 0.19 g (25%) of the title compound.

13 C NMR (DMSO-c/e) E/Z mixture δ 169.1, 167.8, 154.0, 153.8, 142.1, 139.6, 135.8, 135.5, 132.4, 129.0, 128.8, 127.9, 127.5, 125.9, 125.5, 125.1, 123.7, 123.6, 123.3,

122.5, 121.9, 121.8, 121.6, 120.9, 120.6, 119.2, 118.9, 112.0, 111.4, 111.0, 110.8,

109.6, 109.0, 71.8, 71.7, 71.1, 69.6, 69.5, 69.4, 67.9, 67.3, 59.6, 57.9, 50.9

Example 4: synthesis of acetic acid 3-(2-oxo-l,2-dihydro-indol-3-ylidenemethyl)-indol- 1-ylmethyl ester

Acetic acid 3-formyl-indol-l-ylmethyl ester

General procedure A was followed using 1.95 g of indole-3-carboxaldehyde (13.4 mmol) in 25 mL of DMF, 0.84 g of NaH (1.5 eq.) and 1.43 mL acetic acid chloromethyl ester. Purification afforded 1.48 g (51%) of the title compound as off-white crystals. 13 C NMR (DMSO-d 6 ) δ 185.5, 170.1, 141.5, 136.8, 124.6, 124.3, 123.2, 121.2, 118.5, 111.2, 68.8, 20.5

Acetic acid 3-(2-oxo-l,2-dihydro-indol-3-ylidenemethyl)-indol-l-ylmethyl ester

General procedure B was followed using 700 mg of indolinone (5.25 mmol), 1.14 g of acetic acid 3-formyl-indol-l-ylmethyl ester and 1.5 mL of piperidine in 28 ml_ of CH 2 CI 2 . Filtration afforded 1.03 g (59%) of the title compound as a yellow solid. 13 C NMR (DMSO-de) δ 170.0, 167.9, 139.7, 136.1, 135.7, 128.9, 127.5, 125.7, 125.2, 123.3, 121.8, 121.5, 120.7, 119.3, 118.9, 111.9, 110.9, 109.2, 68.8, 20.6

Example 5: synthesis of 2,2-dimethyl-propionic acid 3-(2-oxo-l,2-dihydro-indol-3- ylidenemethyl)-indol-l-ylmethyl ester and 2,2-dimethyl-propionic acid 3-(2-oxo-l- piperidin-l-ylmethyl-l,2-dihydro-indol-3-ylidenemethyl)-indo l-l-ylmethyl ester

2,2-Dimethyl-propionic acid 3-formyl-indol-l-ylmethyl ester

General procedure A was followed using 0.8 g of indole-3-carboxaldehyde (5.5 mmol) in 20 ml_ of DMF, 0.2 g of NaH (1.5 eq.) and 0.79 mL of 2,2-dimethyl-propionic acid chloromethyl ester. Purification afforded 0.55 g (39%) of the title compound. 1 H NMR (DMSO-c/s) δ 9.98 (s,lH), 8.43 (s,lH), 8.12 (d,lH), 7.69 (d,lH), 7.38 (t,lH), 7.31 (t,lH), 6.30 (d,2H), 1.09 (s,9H)

2,2-dimethyl-propionic acid 3-(2-oxo-l,2-dihydro-indoI-3-ylidenemethyl)-indol-l- ylmethyl ester and 2,2-dimethyl-propionic acid 3-(2-oxo-l-piperidin-l-ylmethyl-l,2- dihydro-indol-3-ylidenemethyl)-indol-l-ylmethyl ester

General procedure B was followed using 216 mg of indolinone (1.62 mmol), 420 mg of 2,2-dimethyl-propionic acid 3-formyl-indol-l-ylmethyl ester (1.62 mmol) and 0.5 mL of piperidine in 12 mL of CH 2 CI 2 . Filtration afforded 0.19 g of 2,2-dimethyl-propionic acid 3- (2-oxo-l,2-dihydro-indol-3-ylidenemethyl)-indol-l-ylmethyl ester. The filtrate was concentrated and purified on a Sephadex column (eluent: CH 2 CI 2 - MeOH - hexane 78:2:20) to afford 0.28 g extra (total yield of 77%) of 2,2-dimethyl-propionic acid 3-(2- oxo-l,2-dihydro-indol-3-ylidenemethyl)-indol-l-ylmethyl ester and 0.02 g of 2,2- dimethyl-propionic acid 3-(2-oxo-l-piperidin-l-ylmethyl-l,2-dihydro-indol-3- ylidenemethyl)-indol-l-ylmethyl ester.

13 C NMR (DMSO-de) δ 176.9, 167.8, 139.5, 135.8, 135.5, 128.8, 127.4, 125.6, 125.1, 123.2, 121.6, 121.3, 120.6, 119.1, 118.8, 111.7, 110.7, 109.0, 69.2, 38.3, 26.5 E/Z mixture δ 177.2, 177.0, 168.6, 166.9, 143.4, 141.0, 136.3, 136.0, 135.6, 132.7, 128.9, 127.9, 127.4, 127.1, 126.4, 124.1, 123.7, 123.4, 122.4, 121.9, 121.5, 121.3, 120.0, 119.3, 119.0, 118.9, 111.7, 111.2, 111.0, 110.9, 110.0, 109.6, 69.4, 61.9, 51.7, 51.6, 38.4, 26.6, 25.4, 23.7

Example 6: synthesis of cyclohexyl-acetic acid 3-(2-oxo-l,2-dihydro-indol-3- ylidenemethyl)-indol-l-ylmethyl ester

Cyclohexyl-acetic acid 3-formyl-indol-l-ylmethyl ester

General procedure A was followed using 0.8 g of indole-3-carboxaldehyde (5.5 mmol) in 20 mL of DMF, 0.2 g of NaH (1.5 eq.) and 1.05 g of cyclohexyl-acetic acid chloromethyl ester. Purification afforded 0.89 g (54%) of the title compound.

1 H NMR (DMSOd 6 ) δ 9.98 (s,lH), 8.42 (s,lH), 8.11 (d,lH), 7.69 (d,lH), 7.37 (t,lH), 7.30 (t,lH), 6.30 (s,2H), 2.20 (d,2H), 1.68 - 1.45 (m,6H), 1.18 - 0.75 (m,5H)

Cyclohexyl-acetic acid 3-(2-oxo-l,2-dihydro-indol-3-ylidenemethyl)-indol-l-ylmethyl ester

General procedure B was followed using 244 mg of indolinone (1.83 mmol), 548 mg of cyclohexyl-acetic acid 3-formyl-indol-l-ylmethyl ester (1.83 mmol) and 0.5 mL of piperidine in 12 mL of CH 2 CI 2 . Filtration afforded 0.31 g of cyclohexyl-acetic acid 3-(2- oxo-l,2-dihydro-indol-3-ylidenemethyl)-indol-l-ylmethyl ester. The filtrate was concentrated and purified on a Sephadex column (eluent: CH 2 CI 2 - MeOH - hexane

78:2:20) to afford 0.12 g extra (total yield of 57%) of cyclohexyl-acetic acid 3-(2-oxo- l,2-dihydro-indol-3-ylidenemethyl)-indol-l-ylmethyl ester.

13 C NMR (DMSO-Gf 6 ) E/Z mixture δ 171.9, 171.6, 169.2, 167.8, 142.0, 139.5, 136.0, 135.9, 135.5, 132.4, 128.9, 128.7, 127.8, 127.4, 126.0, 125.6, 125.1, 123.4, 123.2, 122.4, 121.6, 121.3, 120.8, 120.6, 119.1, 118.8, 111.8, 111.1, 111.0, 110.8, 109.6, 109.0, 68.5, 40.8, 34.3, 34.2, 32.0, 25.4, 25.3, 22.4

General procedure C

A solution of aldehyde (1 eq.), l,3-dihydro-indol-2-one (1 eq.) and piperidine (catalytic amount) in EtOH were heated under reflux overnight. The crude mixture was allowed to come to room temperature, cooled in an ice bath. A precipitate was obtained which was filtered and washed with cold EtOH. When no precipitation occurred, the solvent was removed and the product was purified by flash chromatography.

Example 7: synthesis of /V / /V-diethyl-2-[2-methyl-3-(2-oxo-l,2-dihydro-indol-3- ylidenemethyl)-indol-l-yl]-acetamide

General procedure C was followed using l,3-dihydro-indol-2-one (0.24 g, 1.84 mmol), Λ/ / Λ/-diethyl-2-(3-formyl-2-methyl-indol-l-yl)-acetamide (0.50 g, 1.83 mmol) and piperidine (5 drops) as commercially available starting materials to afford 0.60 g (yield 83%) of the title compound. 1 H NMR (DMSO-tf 5 ) E-isomer δ 10.49 (s,lH), 7.83 (s,lH), 7.48 (d,lH), 7.24 - 7.03

(m,4H), 6.91 - 6.70 (m,3H), 5.26 (s,2H), 3.53 (q,2H), 3.33 (q,2H), 2.32 (s,3H), 1.29 (t,3H), 1.06 (t,3H)

Example 8: synthesis of 2-[3-(2-oxo-l,2-dihydro-indol-3-ylidenemethyl)-indol-l-yl]-Î ›/- (tetrahydro-furan-2-ylmethyl)-acetamide

General procedure C was followed using l,3-dihydro-indol-2-one (0.23 g, 1.75 mmol), 2-(3-formyl-indol-l-yl)-/V-(tetrahydro-furan-2-ylmethyl)-ace tamide (0.50 g, 1.75 mmol) and piperidine (5 drops) as commercially available starting materials to afford 0.60 g (yield 86%) of the title compound.

1 H NMR (DMSO-^ 6 ) Z-isomer δ 10.53 (S,1H), 9.41 (s,lH), 8.46 (t,lH), 8.20 (m,lH), 8.12 (s,lH), 7.89 (d,lH), 7.50 (m,lH), 7.28 (m,2H), 7.15 (t,lH), 6.99 (t,lH), 6.84 (Cl 7 IH), 5.03 (s,2H), 3.87 (m,lH), 3.77 (m,lH), 3.64 (m,lH), 3.21 (m,2H), 1.96 - 1.71 (m,3H), 1.48 (m,lH)

Example 9: synthesis of /V-benzo[l,3]dioxol-5-yl-2-[3-(2-oxo-l,2-dihydro-indol-3- ylidenemethyl)-indol-l-yl]-acetamide

General procedure C was followed using l,3-dihydro-indol-2-one (0.21 g, 1.55 mmol), /V-benzo[l,3]dioxol-5-yl-2-[3-formyl-indol-l-yl)-acetamide (0.50 g, 1.55 mmol) and piperidine (5 drops) as commercially available starting materials to afford 0.64 g (yield

94%) of the title compound.

1 H NMR (DMSO-c/s) δ 10.53 (s,lH), 10.43 (s,lH), 9.46 (s,lH), 8.22 (m,lH), 8.14 (s,lH),

7.90 (d,lH), 7.56 (m,lH), 7.34 - 7.25 (m,3H), 7.15 (t,lH), 7.00 (m,2H), 6.9 - 6.82 (m,2H), 5.98 (s,2H), 5.22 (s,2H)

Example 10: synthesis of Λ/-(2-methoxy-ethyl)-2-[3-(2-oxo-l,2-dihydro-indol-3- ylidenemethyl)-indol-l-yl]-acetamide

General procedure C was followed using l,3-dihydro-indol-2-one (0.26 g, 1.92 mmol), 2-(3-formyl-indol-l-yl)-Λ/-(2-methoxy-ethyl)-acetamide (0.50 g, 1.92 mmol) and piperidine (5 drops) as commercially available starting materials to afford the title compound (0.65 g, 90% yield). 1 H NMR (DMSOd 6 ) Z-isomer δ 10.52 (s,lH), 9.41 (s,lH), 8.49 (t,lH), 8.20 (m,lH), 8.12 (s,lH), 7.89 (d,lH), 7.50 (m,lH), 7.28 (m,2H), 7.15 (t,lH), 6.99 (t,lH), 6.84 (d,lH), 5.02 (s,2H), 3.40 (t,2H), 3.30 (t,2H), 3.28 (s,3H)

Example 11: synthesis of 3-[l-(2-morpholin-4-yl-2-oxo-ethyl)-lW-indol-3-ylmethylene]- l,3-dihydro-indol-2-one

General procedure C was followed using l,3-dihydro-indol-2-one (0.30 g, 2.26 mmol), l-(2-morpholin-4-yl-2-oxo-ethyl)-lH-indole-3-carbaldehyde (0.62 g, 2.26 mmol) and piperidine (5 drops) as commercially available starting materials and reagents to afford the title compound (0.60 g, 68% yield).

13 C NMR (DMSO-CZ 6 ) Z-isomer δ 168.0, 165.5, 139.2, 137.5, 136.8, 128.6, 126.8, 126.4,

125.6, 122.4, 120.9, 120.5, 119.3, 118.7, 118.4, 110.9, 110.7, 108.9, 65.9, 47.6, 44.8,

41.9

Exam p le 12: synthesis of /V-furan-2-ylmethyl-2-[3-(2-Oxo-l,2-dihydro-indol-3- ylidenemethyl)-indol-l-yl]-acetamide

General procedure C was followed using indolinone (0.19 g, 1.46 mmol), 2-(3-formyl- indol-l-yl)-/V-furan-2-ylmethyl-acetamide (0.41 g, 1.46 mmol) and piperidine (5 drops) as starting materials and reagents to afford the title compound (0.46 g, 79% yield).

13 C NMR (DMSO-de) Z-isomer δ 168.0, 166.6, 151.7, 142.2, 139.3, 137.2, 136.4, 128.7,

126.8, 126.3, 125.6, 122.6, 121.0, 120.5, 119.5, 118.7, 118.5, 110.8, 110.6, 110.4,

108.9, 107.1, 49.2, 35.7

Example 13: synthesis of Λ/-cyclohexyl-2-[2-methyl-3-(2-Oxo-l,2-dihydro-indol-3- ylidenemethyl)-indol-l-yl]-acetamide

General procedure C was followed using indolinone (0.18 g, 1.35 mmol), Λ/-cyclohexyl-2- (3-formyl-2-methyl-indol-l-yl)-acetamide (0.40 g, 1.35 mmol) and piperidine (5 drops) as starting materials and reagents to afford the title compound (0.48 g, 85% yield). 13 C NMR (DMSO-CZ 6 ) E-isomer δ 169.1, 165.8, 142.0, 141.2, 137.2, 128.1, 128.0, 125.3, 123.9, 123.8, 122.2, 121.6, 120.4, 120.2, 120.1, 109.9, 109.2, 108.5, 47.8, 46.1, 32.3, 25.1, 24.3, 11.7

Example 14: synthesis of 3-[2-methyl-l-(2-oxo-2-piperidin-l-yl-ethyl)-lH-indol-3- ylmethylene]-l,3-dihydro-indol-2-one

General procedure C was followed using indolinone (0.18 g, 1.39 mmol), 2-methyl-l-(2- oxo-2-piperidin-l-yl-ethyl)-l/y-indole-3-carbaldehyde (0.40 g, 1.39 mmol) and piperidine (5 drops) as starting materials and reagents to afford the title compound (0.53 g, 95% yield).

13 C NMR (DMSO-Cf 6 ) E-isomer δ 169.2, 164.9, 142.0, 141.7, 137.5, 128.2, 128.1, 125.3, 123.9, 123.7, 122.3, 121.5, 120.4, 120.1, 110.2, 109.2, 108.4, 45.4, 44.5, 42.7, 26.1, 25.3, 23.9, 11.6

Example 15: synthesis of 2-[2-methyl-3-(2-oxo-l,2-dihydro-indol-3-ylidenemethyl)- indol-l-yl]-Λ/-(tetrahydro-furan-2-ylmethyl)-acetamide

General procedure C was followed using indolinone (0.16 g, 1.21 mmol), 2-(3-formyl-2- methyl-indol-l-yl)-Λ/-(tetrahydro-furan-2-ylmethyl)-acetami de (0.40 g, 1.21 mmol) and piperidine (5 drops) as starting materials to afford the title compound (0.34 g, 64% yield). 13 C NMR (DMSO-Cf 6 ) E-isomer δ 169.1, 167.1, 142.0, 141.1, 137.1, 128.1, 127.9, 125.3, 124.0, 123.8, 122.2, 121.6, 120.4, 120.2, 120.1, 110.0, 109.2, 108.5, 77.0, 67.2, 46.0, 42.7, 28.3, 25.2, 11.7

Exam p le 16: synthesis of /V-cyclohexylmethyl-2-[3-(2-oxo-l,2-dihydro-indol-3- ylidenemethyl)-indol-l-yl]-acetamide, 4-{2-[3-(2-oxo-l,2-dihydro-indol-3-

ylidenemethyl)-indol-l-yl]-acetamino}-butyric acid methyl ester, 6-{2-[3-(2-oxo-l,2- dihydro-indol-3-ylidenemethyl)-indol-l-yl]-acetamino}-hexano ic acid ethyl ester, 2-[3- (2-oxo-l, 2-dihydro-indol-3-ylidenemethyl)-indol-l-yl]-/V-(tetrahydro- furan-2-ylmethyl)- acetamide, Λ/-[2-(lW-indol-3-yl)-ethyl]-2-[3-(2-oxo-l,2-dihydro-indol- 3-ylidenemethyl)- indol-l-yl]-/V-(tetrahydro-furan-2-ylmethyl)-acetamide, 2-[3-(2-oxo-l,2-dihydro-indol- 3-ylidenemethyl)-indol-l-yl]-Λ/-(3-phenyl-propyl)-acetamide , 2-[3-(2-oxo-l,2-dihydro- indol-3-ylidenemethyl)-indol-l-yl]-/V-(4-phenyl-butyl)-aceta mide, Λ/-[3-(l-formyl- piperidin-4-yl)-propyl]-2-[3-(2-oxo-l,2-dihydro-indol-3-ylid enemethyl)-indol-l-yl]- acetamide, /V-(4-hydroxy-butyl)-2-[3-(2-oxo-l,2-dihydro-indol-3-ylidene methyl)-indol- l-yl]~acetamide, 4-{2-[3-(2-oxo-l,2-dihydro-indol-3-ylidenemethyl)-indol-l-yl ]- acetamino}-butyric acid ethyl ester, 3-{l-[2-(4-methyl-piperazin-l-yl)-2-oxo-ethyl]-lW- indol-3-ylmethylene}-l,3-dihydro-indol-2-one

14

General procedure D

To a suspension of NaH (1.5 eq.) in DMF was added a solution of aldehyde (1 eq.) in DMF. After stirring at room temperature for 30 minutes, the mixture was cooled down in an ice bath and the chloroester (1 eq.) was added dropwise. The crude mixture was then allowed to come to room temperature and left under stirring. The reaction was followed by TLC. The reaction mixture was poured into ice and extracted with Et 2 O (2x20 mL). The combined organic phases were washed once with brine and concentrated in vacuo together with silica gel. The residue was purified by chromatography.

(3-Formyl-indol-l-yl)-acetic acid methyl ester (11)

General procedure D was followed using the following amounts of reagents and starting materials: NaH (0.49 g, 20.5 mmol) in DMF (5 ml_), lH-indole-3-carbaldehyde (1.99 g, 13.7 mmol) in DMF (15 ml_), and chloro-acetic acid methyl ester (1.05 ml_, 13.7 mmol). Flash chromatography on silica gel using a gradient of EtOAc/Petroleum ether from 25/75 to 75/25 furnished the title compound (2.1 g, 71%).

1 H NMR (DMSO-c/e) δ 9.95 (s,lH), 8.28 (s,lH), 8.12 (m,lH), 7.54 (m,lH), 7.29 (m,2H), 5.31 (s,2H), 3.71 (s,3H)

[3-(2-Oxo-l,2-dihydro-indol-3-ylidenemethyl)-indol-l-yl]- acetic acid methyl ester (12) and [3-(2-oxo-l,2-dihydro-indol-3-ylidenemethyl)-indol-l-yl]-ace tic acid ethyl ester (13)

General procedure C was followed using l,3-dihydro-indol-2-one (0.14 g, 1.84 mmol), (3-formyl-indol-l-yl)-acetic acid methyl ester (1.87 g, 1.83 mmol) and piperidine (10 drops). After filtration a mixture of [3-(2-oxo-l,2-dihydro-indol-3-ylidenemethyl)-indol- l-yl]-acetic acid methyl ester (45%) and [3-(2-oxo-l,2-dihydro-indol-3-ylidenemethyl)- indol-l-yl]-acetic acid ethyl ester (55%) was obtained as an orange solid (2.27 g, yield about 80%) and was directly used without further purification.

1 H NMR (DMSO-CZ 6 ) δ

methyl ester 10.55 (s,lH), 9.41 (s,lH), 8.21 (m,lH), 8.13 (S 7 IH), 7.91 (d,lH), 7.53 (m,lH), 7.29 (m,2H), 7.16 (t,lH), 7.00 (t,lH), 6.86 (d,lH), 5.37 (s,2H), 3.72 (s,3H) ethyl ester 10.55 (s,lH), 9.41 (s,lH), 8.21 (m,lH), 8.13 (s,lH), 7.91 (d,lH), 7.53 (m,lH), 7.29 (m,2H), 7.16 (t,lH), 7.00 (t,lH), 6.86 (d,lH), 5.35 (s,2H), 4.19 (q,2H), 1.23 (t,3H)

General procedure E

A solution of the ester (1 eq.) in a 1:1 mixture of THF or MeOH and aqueous solution of LiOH was heated under reflux. The reaction was followed by TLC. When TLC indicated full conversion the pH of the solution was adjusted to acidic pH (6-7) by addition of HCI (IN). A precipitate formed which was filtered and dried furnishing the desired product.

[3-(2-Oxo-l,2-dihydro-indol-3-ylidenemethyl)-indol-l-yl]- acetic acid (14)

General procedure E was followed using a 45/55 mixture of [3-(2-oxo-l,2-dihydro-indol- 3-ylidenemethyl)-indol-l-yl]-acetic acid methyl ester and [3-(2-oxo-l,2-dihydro-indol-3- ylidenemethyl)-indol-l-yl]-acetic acid ethyl ester (1.76 g, about 8 mmol) in a 1/1 mixture of an aqueous solution of LiOH IN (25 mL) and THF (25 mL). After heating under reflux for 6 hours, and addition of HCI (IN) the title compound was obtained as an orange solid (1.37 g, 53%).

1 H NMR (DMSO-CZ 6 ) δ 13.19 (br,lH), 10.55 (s,lH), 9.40 (s,lH), 8.21 (m,lH), 8.13 (s,lH), 7.90 (d,lH), 7.54 (m,lH), 7.29 (m,2H), 7.15 (t,lH), 7.00 (t,lH), 6.86 (d,lH), 5.24 (s,2H)

General procedure F

To a mixture of [3-(2-oxo-l,2-dihydro-indol-3-ylidenemethyl)-indol-l-yl]-ace tic acid or 3-[3-(2-Oxo-l,2-dihydro-indol-3-ylidenemethyl)-indol-l-yl]-p ropionic acid (1 eq.), EDCI (72 mg, 0.37 mmol, 1.5 eq.), HOBt (51 mg, 0.37 mmol, 1.5 eq.), Et 3 N (0.07 mL or 0.14 mL, 0.5 or 1 mmol, 2 or 4 eq.) in DMF (0.8 mL) was added the corresponding amine as a free base or a salt (2 eq.). The crude mixture was shaken or stirred at room

temperature overnight. After removal of DMF under high vacuum, the crude was purified by flash chromatography using MeOH/CH 2 CI 2 mixtures as eluent.

Λ/-Cyclohexylmethyl-2-[3-(2-Oxo-l,2-dihydro-indol-3-ylid enemethyl)-indol-l-yl]- acetamide

General procedure F was followed using [3-(2-oxo-l,2-dihydro-indol-3-ylidenemethyl)- indol-l-yl]-acetic acid (80 mg, 0.25 mmol), C-cyclohexyl-methylamine (0.065 ml_, 0.5 mmol) and Et 3 N (0.07ml_, 2 eq.). After purification by flash chromatography 15 mg of the title compound were obtained (14% yield).

1 H NMR (DMSO-CZ 6 ) E/Z:50/50 δ 10.52 (s,0.5H), 10.48 (s,0.5H), 9.41 (s,0.5H), 8.33 (t,0.5H), 8.29 (t,0.5H), 8.27 (s,0.5H), 8.20 (m,0.5H), 8.12 (s,0.5H), 7.89 (d,lH), 7.86 (s,0.5H), 7.73 (d,0.5H), 7.50 (m,lH), 7.33 - 7.10 (m,3H), 7.03 - 6.81 (m,2H), 5.04 (s,lH), 5.00 (s,lH), 2.98 (t,2H), 1.75 - 1.50 (m,5H), 1.41 (m,lH), 1.16 (m,3H), 0.90 (m,2H)

4-{2-[3-(2-Oxo-l,2-dihydro-indol-3-ylidenemethyl)-indol-l -yl]-acetamino}-butyric acid methyl ester

General procedure F was followed using [3-(2-oxo-l,2-dihydro-indol-3-ylidenemethyl)- indol-l-yl]-acetic acid (80 mg, 0.25 mmol), 4-amino-butyric acid methyl ester, hydrochloride (77 mg, 0.5 mmol) and Et 3 N (0.14 ml_, 1 mmol, 4 eq.). After purification by flash chromatography 55 mg of the title compound were obtained (52% yield).

1 H NMR (DMSOd 6 ) E/Z:50/50 δ 10.52 (s,0.5H), 10.47 (s,0.5H), 9.40 (s,0.5H), 8.39 (t,0.5H), 8.33 (t,0.5H), 8.27 (s,0.5H), 8.20 (m,0.5H), 8.12 (s,0.5H), 7.89 (d,lH), 7.85

(s,0.5H), 7.73 (d,0.5H), 7.48 (m,lH), 7.35 - 7.10 (m,3H), 7.03 - 6.82 (m,2H), 5.03 (s,lH), 4.99 (s,lH), 3.59 (s,1.5H), 3.58 (s,1.5H), 3.14 (mq,2H), 2.35 (t,2H), 1.71 (m,2H)

6-{2-[3-(2-Oxo-l,2-dihydro-indol-3-ylidenemethyl)-indol-l -yl]-acetamino}-hexanoic acid ethyl ester

General procedure F was followed using [3-(2-oxo-l,2-dihydro-indol-3-ylidenemethyl)- indol-l-yl]-acetic acid (80 mg, 0.25 mmol), 6-amino-hexanoic acid ethyl ester, hydrochloride (98 mg, 0.5 mmol) and Et 3 N (0.14 ml_, 1 mmol, 4 eq.). After purification by flash chromatography 82 mg of the title compound were obtained (71% yield). 1 H NMR (DMSO-CZ 6 ) E/Z:10/90 δ 10.51 (s,lH), 9.41 (s,lH), 8.34 (t,lH), 8.20 (m,lH), 8.12 (s,lH), 7.89 (d,lH), 7.48 (m,lH), 7.28 (m,2H), 7.14 (t,lH), 7.00 (t,lH), 6.84 (d,lH), 4.98 (s,2H), 4.04 (q,2H), 3.10 (q,2H), 2.26 (t,2H), 1.6 - 1.2 (m,6H), 1.17 (t,3H)

2-[3-(2-Oxo-l,2-dihydro-indol-3-ylidenemethyl)-indol-l-yl ]-Λ/-(tetrahydro-furan-2- ylmethyl)-acetamide

General procedure F was followed using [3-(2-oxo-l,2-dihydro-indol-3-ylidenemethyl)- indol-l-yl]-acetic acid (80 mg, 0.25 mmol), tetrahydro-furfurylamine (51 mg, 0.5 mmol) and Et 3 N (0.07 ml_, 0.5 mmol, 2 eq.). After purification by flash chromatography 42 mg of the title compound were obtained (41% yield).

1 H NMR (DMSO-CZ 6 ) Z-isomer δ 10.53 (s,lH), 9.41 (s,lH), 8.46 (t,lH), 8.20 (m,lH), 8.12 (S,1H), 7.89 (d,lH), 7.50 (m,lH), 7.28 (m,2H), 7.15 (t,lH), 6.99 (t,lH), 6.84 (d,lH), 5.03 (s,2H), 3.87 (m,lH), 3.77 (m,lH), 3.64 (m,lH), 3.21 (m,2H), 1.96 - 1.71 (m,3H), 1.48 (m,lH)

Λ/-[2-(lW-Indol-3-yl)-ethyl]-2-[3-(2-oxo-l,2-dihydro-ind ol-3-ylidenemethyl)-indol-l-yl]- N-(tetrahydro-furan-2-ylmethyl)-acetamide

General procedure F was followed using [3-(2-oxo-l,2-dihydro-indol-3-ylidenemethyl)- indol-l-yl]-acetic acid (80 mg, 0.25 mmol), 2-(lW-indol-3-yl)-ethylamine (80 mg, 0.5 mmol) and Et 3 N (0.07 ml_, 0.5 mmol, 2 eq.). After purification by flash chromatography 77 mg of the title compound was obtained (66% yield).

1 H NMR (DMSO-CZ 6 ) Z-isomer δ 10.82 (s,lH), 10.53 (s,lH), 9.43 (s,lH), 8.48 (t,lH), 8.21 (m,lH), 8.13 (s,lH), 7.90 (d,lH), 7.55 (d,lH), 7.42 (m,lH), 7.35 (d,lH), 7.27 (m,2H), 7.20 - 6.93 (m,6H), 6.85 (d,lH), 4.99 (s,lH), 3.42 (q,2H), 2.88 (t,2H)

2-[3-(2-Oxo-l,2-dihydro-indol-3-ylidenemethyl)-indol-l-yl ]-/V-(3-phenyl-propyl)- acetamide

General procedure F was followed using [3-(2-oxo-l,2-dihydro-indol-3-ylidenemethyl)- indol-l-yl]-acetic acid (80 mg, 0.25 mmol), 3-phenyl-propylamine (68 mg, 0.5 mmol) and Et 3 N (0.07 ml_, 0.5 mmol, 2 eq.). After purification by flash chromatography 44 mg of the title compound were obtained (40% yield).

1 H NMR (DMSO-d 6 ) Z-isomer δ 10.52 (s,lH), 9.42 (S 7 IH), 8.41 (t,lH), 8.20 (m,lH), 8.12 (s,lH), 7.90 (d,lH), 7.51 (m,lH), 7.35 - 7.09 (m,8H), 6.99 (t,lH), 6.85 (d,lH), 5.01 (s,2H), 3.14 (q,2H), 2.60 (m,2H), 1.74 (m,2H)

2-[3-(2-Oxo-l,2-dihydro-indol-3-ylideneπnethyl)-indol-l- yl]-/V-(4-phenyl-butyl)- acetamide

General procedure F was followed using [3-(2-oxo-l,2-dihydro-indol-3-ylidenemethyl)- indol-l-yl]-acetic acid (80 mg, 0.25 mmol), 4-phenyl-butylamine (75 mg, 0.5 mmol) and Et 3 N (0.07 mL, 0.5 mmol, 2 eq.). After purification by flash chromatography 113 mg of the title compound were obtained (100% yield). 1 H NMR (DMSO-CZ 6 ) Z-isomer δ 10.52 (s,lH), 9.41 (s,lH), 8.37 (t,lH), 8.21 (m,lH), 8.12 (s,lH), 7.89 (d,lH), 7.48 (m,lH), 7.30 - 7.10 (m,8H), 6.99 (t,lH), 6.84 (d,lH), 4.99 (s,2H), 3.15 (q,2H), 2.57 (t,2H), 1.65 - 1.40 (m,4H)

Λ/-[3-(l-Formyl-piperidin-4-yl)-propyl]-2-[3-(2-oxo-l,2- dihydro-indol-3-ylidenemethyl)- indol-l-yl]-acetamide

General procedure F was followed using [3-(2-oxo-l,2-dihydro-indol-3-ylidenemethyl)- indol-l-yl]-acetic acid (80 mg, 0.25 mmol), /V-formyl-4-(3-aminopropyl)-piperidin, hydrochloride (104 mg, 0.5 mmol) and Et 3 N (0.14 mL, 1 mmol, 4 eq.). After purification by flash chromatography 76 mg of the title compound were obtained (64% yield). 1 H NMR (DMSO-CZ 6 ) δ 10.51 (s,lH), 9.41 (s,lH), 8.35 (s,lH), 8.20 (m,lH), 8.12 (s,lH), 7.95 (s,lH), 7.89 (d,lH), 7.49 (m,lH), 7.28 (m,2H), 7.15 (t,lH), 6.99 (t,lH), 6.85

(d,lH), 4.98 (s,2H), 4.12 (m,lH), 3.61 (m,lH), 3.11 (q,2H), 2.96 (m,lH), 2.54 (m,lH), 1.65 (m,2H), 1.46 (m,3H), 1.21 (m,2H), 0.95 (m,lH), 0.86 (m,lH)

Λ/-(4-Hydroxy-butyl)-2-[3-(2-oxo-l,2-dihydro-indol-3-yli denemethyl)-indol-l-yl]-- acetamide

General procedure F was followed using [3-(2-oxo-l,2-dihydro-indol-3-ylidenemethyl)- indol-l-yl]-acetic acid (80 mg, 0.25 mmol), 4-amino-l-butanol (45 mg, 0.5 mmol) and

Et 3 N (0.07 mL, 0.5 mmol, 2 eq.). After purification by flash chromatography 46 mg of the title compound were obtained (47% yield).

1 H NMR (DMSOd 6 ) δ 10.52 (s,lH), 9.41 (s,lH), 8.35 (t,lH), 8.20 (m,lH), 8.12 (s,lH),

7.89 (d,lH), 7.48 (m,lH), 7.28 (m,3H), 7.15 (t,lH), 6.99 (t,lH), 6.85 (d,lH), 4.98 (s,2H), 3.40 (m,2H), 3.12 (m,2H), 1.46 (m,4H)

4-{2-[3-(2-Oxo-l,2-dihydro-indol-3-ylidenemethyl)-indol-l -yl]-acetamino}-butyric acid ethyl ester

General procedure F was followed using [3-(2-oxo-l,2-dihydro-indol-3-ylidenemethyl)- indol-l-yl]-acetic acid (80 mg, 0.25 mmol), 4-amino-butyric acid ethyl ester, hydrochloride (84 mg, 0.5 mmol) and Et 3 N (0.14 mL, 1 mmol, 4 eq.). After purification by flash chromatography 108 mg of the title compound were obtained (100% yield). 1 H NMR (DMSO-CZ 6 ) Z-isomer δ 10.52 (s,lH), 9.40 (s,lH), 8.39 (t,lH), 8.20 (m,lH), 8.11 (S,1H), 7.89 (s,lH), 7.48 (m,lH), 7.28 (m,2H), 7.15 (t,lH), 6.99 (t,lH), 6.84 (d,lH), 4.99 (s,2H), 4.05 (q,2H), 3.14 (q,2H), 2.39 (t,2H), 1.70 (m,2H), 1.17 (t,3H)

General Procedure G:

To a mixture of the desired carboxylic acid (1 eq.), EDAC (1.3 eq.), HOBt (1 eq.), N- methyl-morpholine (2 eq.) in CH 2 CI 2 was added the desired amine (2 eq.). The crude mixture was stirred at room temperature and the reaction was followed by TLC. H 2 O (10 mL) was added and the crude mixture was extracted with CH 2 CI 2 (3x10 ml_). The combined organic phases were washed once with H 2 O, once with brine and concentrated in vacuo. The residue was purified by flash chromatography (silica gel, CH 2 CI 2 /Me0H mixtures as eluent), furnishing the title compound.

3-Cl-[2-(4-Methyl-piperazin-l-yl)-2-oxo— ethyl]-l/Y-indol-3-ylmethylene}-l,3-dihydro- indol-2-one

General procedure G was followed. To a mixture of [3-(2-oxo-l,2-dihydro-indol-3- ylidenemethyl)-indol-l-yl]-acetic acid (0.11 g, 0.36 mmol), EDAC (91 mg, 0.47 mmol, 1.3 eq.), HOBt (49 mg, 0.36 mmol, 1 eq.), /V-methyl-morpholine (0.08 mL, 0.73 mmol, 2 eq.) in CH 2 CI 2 (10 mL) was added 1-methyl-piperazine, hydrochloride (0.08 mL, 0.73 mmol, 2 eq.). Purification by flash chromatography (silica gel, CH 2 CI 2 /Me0H mixtures as eluent), furnished the title compound (0.12 g, 84%).

13 C NMR (DMSO-Cf 6 ) 25/75 E/Z-mixture δ 169.5, 167.9, 165.3, 165.2, 141.6, 139.1, 137.5, 137.0, 136.7, 133.8, 128.5, 128.3, 127.6, 126.7, 126.4, 125.5, 122.6, 122.3, 122.2, 122.0, 121.2, 120.9, 120.8, 120.4, 119.1, 118.7, 118.4, 111.0, 110.8, 110.6, 109.7, 109.4, 108.9, 56.7, 54.5, 54.1, 47.6, 47.4, 45.6, 45.1, 44.1, 41.4, 33.9, 27.9

Exam p le 17: synthesis of 3-{l-[3-(4-methyl-piperazin-l-yl)-2-oxo— propyl]-l/V-indol-3- ylmethylene}-l,3-dihydro-indol-2-one

3-(3-Formyl-indol-l-yl)-propionic acid ethyl ester (15)

General procedure D was followed using the following amounts of reagents and starting materials: NaH (60% in oil, 0.41 g, 10.3 mmol) in DMF (5 ml_), lH-indole-3- carbaldehyde (1.0 g, 6.9 mmol) in DMF (15 ml_), and 3-chloro-propionic acid ethyl ester (0.86 mL, 6.9 mmol). Flash chromatography on silica gel using a gradient of EtOAc/Petroleum ether from 25/75 to 75/25 furnished the title compound (1.49 g, 88%).

1 H NMR (DMSO-c/e) δ 9.91 (s,lH), 8.30 (s,lH), 8.11 (d,lH), 7.66 (d,lH), 7.30 (m,2H), 4.54 (t,2H), 4.03 (q,2H), 2.93 (t,2H), 1.11 (t,3H)

3-[3-(2-Oxo-l,2-dihydro-indol-3-ylidenemethyl)-indol-l-yl ]-propionic acid ethyl ester (16)

General procedure C was followed using l,3-dihydro-indol-2-one (0.46 g, 3.43 mmol), 3-(3-formyl-indol-l-yl)-propionic acid ethyl ester (0.84 g, 3.42 mmol) and piperidine (10 drops). After filtration the title compound was obtained as a yellow solid (1.08 g, 88% yield).

1 H NMR (DMSO-Cy 6 ) δ 10.52 (s,lH), 9.45 (s,lH), 8.20 (m,lH), 8.11 (s,lH), 7.88 (d,lH), 7.64 (m,lH), 7.29 (m,2H), 7.15 (t,lH), 6.99 (t,lH), 6.85 (d,lH), 4.58 (t,2H), 4.04 (q,2H), 2.92 (t,2H), 1.12 (t,3H)

3-[3-(2-Oxo-l,2-dihydro-indol-3-ylidenemethyl)-indol-l-yl ]-propionic acid (17)

General procedure E was followed starting from 3-[3-(2-oxo-l,2-dihydro-indol-3- ylidenemethyl)-indol-l-yl]-propionic acid ethyl ester (0.62 g, 1.73 mmol) in a 1:1 mixture of an aqueous solution of LiOH 0.2N (30 ml_) and MeOH (30 ml_). After heating under reflux for 4 hours, addition of HCI (IN), the title compound was obtained as orange crystals (0.57 g, 100%).

1 H NMR (DMSO-CZ 6 ) δ 12.46 (br,lH), 10.49 (s,lH), 9.47 (S 7 IH), 8.19 (m,lH), 8.10 (S,1H), 7.88 (d,lH), 7.64 (m,lH), 7.29 (m,2H), 7.15 (t,lH), 6.99 (t,lH), 6.85 (d,lH), 4.55 (t,2H), 2.86 (t,2H)

3-{l-[3-(4-Methyl-piperazin-l-yl)-2-oxo— propyl]-lH-indol-3-ylmethylene}-l,3-dihydro- indol-2-one

General procedure G was followed. To a mixture of 3-[3-(2-Oxo-l,2-dihydro-indol-3- ylidenemethyl)-indol-l-yl]-propionic acid (0.10 g, 0.30 mmol), EDAC (74 mg, 0.39 mmol, 1.3 eq.), HOBt (40 mg, 0.30 mmol, 1 eq.), /V-methyl-morpholine (0.07 ml_, 0.60 mmol, 2 eq.) in CH 2 CI 2 (10 mL) was added 1-methyl-piperazine, hydrochloride (0.07 ml_, 0.60 mmol, 2 eq.). Purification by flash chromatography (silica gel, CH 2 CI 2 /Me0H mixtures as eluent), furnished the title compound. MS [M+H] + = 415, [M-H] " = 413

Example 18: synthesis of /V-(2-hydroxy-ethyl)-3-[3-(2-oxo-l,2-dihydro-indol-3- ylidenemethyl)-indol-l-yl]-propionamide

A mixture of 3-[3-(2-oxo-l,2-dihydro-indol-3-ylidenemethyl)-indol-l-yl]-p ropionic acid ethyl ester (0.45 g, 1.25 mmol) K 2 CO 3 (0.28 g, 2.0 mmol) in ethanolamine (10 mL) was stirred at room temperature overnight. TLC showed that the reaction went to completion. H 2 O was added (100 mL) and the aqueous phase was extracted with EtOAc (2x100 mL). The combined organic phases were concentrated in vacuo together with silica gel. The residue was purified by 2 consecutive chromatographies using a gradient MeOH/CH 2 CI 2 from 0/100 to 10/90 as eluents, furnishing the title compound (66 mg, 14% yield). 1 H NMR (DMSO-c/e) δ 10.50 (s,lH), 9.41 (s,lH), 8.19 (d,lH), 8.10 (s,lH), 7.99 (t,lH), 7.88 (d,lH), 7.63 (d,lH), 7.28 (m,2H), 7.14 (t,lH), 6.99 (t,lH), 6.84 (d,lH), 4.59 (t,lH), 4.54 (t,2H), 3.32 (t,2H), 3.10 (q,2H), 2.69 (t,2H)

Example 19: synthesis of 3-(l-benzenesulfonyl-lW-indol-3-ylmethylene)-l,3-dihydro- indol-2-one

General procedure C was followed using indolinone (117 mg, 0.88 mmol), 1- benzenesulfonyl-lΛHndole-3-carbaldehyde (250 mg, 1 eq.) and piperidine (5 drops). Filtration afforded 227 mg (64%) of the title compound.

13 C NMR (DMSO-CZ 6 ) E/ Z mixture δ 168.3, 167.6, 142.8, 140.4, 136.6, 135.0, 133.5, 130.9, 130.2, 130.1, 128.8, 126.9, 126.6, 126.1, 125.7, 125.5, 124.3, 124.0, 123.2, 121.1, 120.9, 120.2, 119.9, 116.9, 115.9, 113.4, 113.1, 110.0, 109.4

General procedure H

Under dry conditions was prepared a solution of KOH (4 eq., 2-3 N) in DMSO. After stirring at room temperature for 5 minutes was added the starting indole (1 eq.) under argon atmosphere and the mixture was stirred an additional 45 minutes.

The alkylating agent (1.1 eq.) was then added dropwise at 0 0 C and the mixture was allowed to come to room temperature. The reaction was followed by TLC. H 2 O was added and the aqueous phase was extracted with Et 2 O (3χ). The combined organic phases was washed once with H 2 O, once with brine, dried over MgSO 4 . Removal of solvent under vacuo afforded the expected compound which could be directly used for the next step of the synthesis.

Example 20: synthesis of 3-(l-benzyl-lAY-indol-3-ylmethylene)-l,3-dihydro-indol-2-one

l-Benzyl-l-tf-indole-3-carbaldehyde

General procedure H was followed using KOH (1.60 g, 28 mmol) in DMSO (15 mL), IH- indole-3-carbaldehyde (1 g, 6.9 mmol) and bromomethyl-benzene (0.90 mL, 7.6 mmol). The title compound was obtained as an orange powder (1.42 g, 89% yield). 1 H NMR (DMSO-CZ 6 ) δ 9.95 (s,lH), 8.46 (s,lH), 8.12 (m,lH), 7.58 (m,lH), 7.40 - 7.20 (m,7H), 5.55 (s,2H)

3-(l-Benzyl-l/f-indol-3-ylmethylene)-l,3-dihydro-indol-2- one

The title compound was obtained as a yellow solid using l-benzyl-l-H-indole-3- carbaldehyde (268 mg, 1.14 mmol) and l,3-dihydro-indol-2-one (152 mg, 1.14 mmol) and following the general procedure C (161 mg, 40% yield after cristallization in EtOAc). 13 C NMR (DMSO-Cf 6 , Z isomer) δ 167.9, 139.2, 137.1, 136.1, 135.8, 128.8, 128.6, 127.6, 127.1, 126.8, 126.3, 125.5, 122.7, 121.0, 120.5, 119.5, 118.8, 118.7, 111.0, 110.9, 108.9, 49.8

General procedure I

To a suspension of substituted 3-(lH-indol-3-ylmethylene)-l,3-dihydro-indol-2-one in MeOH were added formaldehyde (13.1 M in water) and an amine. The mixture was heated to temperature T for a time t and allowed to cool to r.t.. The precipitate was filtered and washed with water. If no precipitation occurred, the solvent was removed and the product was purified by flash chromatography.

Example 21: synthesis of l-piperidin-l-ylmethyl-3-(l-piperidin-l-ylmethyl-lW-indol-3- ylmethylene)-l,3-dihydro-indol-2-one

General procedure I was followed using 1.0 g (3.8 mmol) of 3-(l/V-indol-3-ylmethylene)- l,3-dihydro-indol-2-one, 2.9 mL (10 eq.) formaldehyde solution and 3.8 ml_ (10 eq.) piperidine in 50 mL methanol. Heating at 60 0 C for 1 h. Filtration of the precipitate afforded 1.3 g (75%) of the title compound as yellow powder. 13 C NMR (CDCI 3 ) δ 168.0, 141.2, 137.3, 137.2, 129.3, 126.8, 126.7, 125.0, 122.8, 121.4, 121.3, 119.0, 117.9, 117.6, 111.4, 111.0, 109.5, 69.6, 62.8, 52.2, 51.8, 25.9, 25.8, 24.2, 23.8

Example 22: synthesis of l-(4-methyl-piperazin-l-ylmethyl)-3-[l-(4-methyl-piperazin- l-ylmethyl)-lH-indol-3-ylmethylene]-l,3-dihydro-indol-2-one

General procedure I was followed using 1 g (3.8 mmol) of 3-(ltf-indol-3-ylmethylene)- l,3-dihydro-indol-2-one, 2.9 mL (10 eq.) formaldehyde solution and 4.3 mL (10 eq.) N- methylpiperazine in 50 mL methanol. Heating at 60 0 C for 1 h. Filtration of the precipitate afforded 1.02 g (55%) of the title compound as yellow powder. 1 HNMR (CDCI 3 ) δ 9.44 (s,lH), 7.96 (s,lH), 7.90 (m,lH), 7.63 (d,lH), 7.55 (m,lH), 7.31 (m,2H), 7.23 (t,lH), 7.09 (t,lH), 7.04 (d,lH), 4.94 (s,2H), 4.58 (s,2H), 2.68 (m,8H), 2.42 (br,8H), 2.26 (s,3H), 2.24 (s,3H)

Exam p le 23: synthesis of 3-(l#-Indol-3-ylmethylene)-l-piperidin-l-ylmethyl-l,3- dihydro-indol-2-one

General procedure I was followed using 0.85 g (3.3 mmol) of 3-(lW-indol-3- ylmethylene)-l,3-dihydro-indol-2-one, 0.293 mL (1.17 eq.) formaldehyde solution and 0.380 mL (1.17 eq.) piperidine in 50 mL methanol. Heating at 60 0 C for 3 h. After standing in the fridge for 2h, filtration of the precipitate afforded 829 mg (71%) of the title compound as yellow powder.

13 C NMR (DMSO-Cy 6 ) δ 166.9, 140.4, 135.8, 133.7, 128.1, 127.7, 126.5, 124.4, 122.5, 121.0, 120.7, 118.4, 118.2, 117.6, 112.2, 111.2, 109.3, 61.7, 51.6, 25.3, 23.6

Example 24: synthesis of 3-(lW-Indol-3-ylmethylene)-l-morpholin-4-ylmethyl-l,3- dihydro-indol-2-one

General procedure I was followed using 596 mg (2.3 mmol) of 3-(lAy-indol-3- ylmethylene)-l,3-dihydro-indol-2-one, 0.174 mL (1.0 eq.) formaldehyde solution and 0.199 mL (1.0 eq.) morpholine in 50 mL methanol. Heating at 40 0 C for 20 h. After standing in the fridge for 2 h, filtration of the precipitate afforded an impure product. Flash chromatography (PE - EtOAc) afforded 203 mg (24%) of the title compound as yellow powder.

13 C NMR (DMSO-CZ 6 ) δ 166.9, 140.1, 135.8, 133.8, 128.1, 127.9, 126.5, 124.4, 122.5, 121.2, 120.8, 118.4, 118.3, 117.4, 112.2, 111.2, 109.2, 66.0, 61.1, 50.8

Example 25: synthesis of l-(3,3-dimethyl-piperidin-l-ylmethyl)-3-(l/7-indol-3- ylmethylene)-l,3-dihydro-indol-2-one

General procedure I was followed using 596 mg (2.3 mmol) of 3-(l/-/-indol-3- ylmethylene)-l,3-dihydro-indol-2-one, 0.204 mL (1.17 eq.) formaldehyde solution and 305 mg (1.17 eq.) of 3,3-dimethylpiperidine in 30 mL methanol. Heating at 60 0 C for 4 days. After standing in the fridge for 2 h, filtration of the precipitate afforded 328 mg (37%) of the title compound as yellow powder. 1 H NMR (DMSO-de) δ 12.07 (br,lH), 9.46 (s,lH), 8.22 (s,lH), 8.19 (m,lH), 7.93 (d,lH), 7.54 (m,lH), 7.25 (m,2H), 7.22 (t,lH), 7.12 (d,lH), 7.06 (t,lH), 4.51 (d,2H), 2.47 (m,2H), 2.22 (s,2H), 1.48 (m,2H), 1.14 (m,2H), 0.88 (s,6H)

Example 26: synthesis of l-[(tert-butyl-methyl-amino)-methyl]-3-(lW-indol-3- ylmethylene)-l,3-dihydro-indol-2-one

General procedure I was followed using 596 mg (2.3 mmol) of 3-(lH-indol-3- ylmethylene)-l,3-dihydro-indol-2-one, 0.204 mL (1.17 eq.) formaldehyde solution and 0.320 mL (1.17 eq.) Λ/-tert-butyl methylamine in 50 mL methanol. Heating at 60 0 C for 20 h. After standing in the fridge for 2 h, filtration of the precipitate afforded 516 mg (62%) of the title compound as yellow powder.

1 H NMR (DMSO-Cf 6 ) δ 12.07 (br,lH), 9.47 (s,lH), 8.21 (s,lH), 8.19 (m,lH), 7.94 ((.,1H), 7.53 (m,lH), 7.24 (m,2H), 7.21 (t,lH), 7.12 (d,lH), 7.06 (t,lH), 4.57 (s,2H), 2.12 (s,3H), 1.20 (s,9H)

Example 27: synthesis of 3-{l-[(diisobutylamino)-methyl]-l/7-indol-3-ylmethylene}- l,3-dihydro-indol-2-one

General procedure I was followed using 596 mg (2.3 mmol) of 3-(l#-indol-3- ylmethylene)-l,3-dihydro-indol-2-one, 0.204 ml_ (1.17 eq.) formaldehyde solution and 345 mg (1.17 eq.) of diisobutylamine in 30 ml_ methanol. Heating at 60 0 C for 4 days.

After standing in the fridge for 2 h, filtration of the precipitate afforded 502 mg (55%) of the title compound as yellow powder.

13 C NMR (DMSO-dβ) δ 168.0, 139.2, 136.8, 136.6, 128.4, 126.8, 126.5, 125.5, 122.5,

120.8, 120.4, 119.4, 118.7, 118.4, 111.0, 110.5, 108.8, 65.1, 60.8, 25.8, 20.5

Example 28: synthesis of 3-(l-methyl-l/y-indol-3-ylmethylene)-l-piperidin-l-ylmethyl- l,3-dihydro-indol-2-one

General procedure I was followed using 41 mg (0.15 mmol) of 3-(l-methyl-l/7-indol-3- ylmethylene)-l,3-dihydro-indol-2-one, 0.115 mL (10 eq.) formaldehyde solution and 0.148 mL (10 eq.) piperidine in 2 mL methanol. Heating at 60 0 C for 16 h. Filtration of the precipitate afforded 43 mg (77%) of the title compound as yellow powder. 13 C NMR (CDCI 3 ) δ 168.1, 141.1, 137.6, 136.9, 129.2, 126.8, 126.7, 125.1, 122.8, 121.4, 121.4, 118.3, 118.0, 117.5, 111.0, 110.0, 109.4, 62.8, 52.3, 33.6, 25.9, 24.1

Example 29: synthesis of 3-(l-methyl-2-phenyl-lW-indol-3-ylmethylene)-l-piperidin-l- ylmethyl-l,3-dihydro-indol-2-one

General procedure I was followed using 53 mg (0.15 mmol) of 3-(l-methyl-2-phenyl- lft-indol-3-ylmethylene)-l,3-dihydro-indol-2-one, 0.115 ml_ (10 eq.) formaldehyde solution and 0.148 mL (10 eq.) piperidine in 2 mL methanol. Heating at 60 0 C for 16 h. Filtration of the precipitate afforded 39 mg (58%) of the title compound as yellow powder. 13 C NMR (CDCI 3 ) δ 170.0, 144.6, 143.6, 138.1, 131.4, 131.0, 130.8, 130.3, 129.1, 128.6, 127.7, 125.1, 124.9, 123.7, 123.0, 122.8, 121.2, 120.9, 110.7, 110.2, 109.1, 62.9, 52.2, 31.6, 25.9, 24.1

Example 30: synthesis of 3-(5-fluoro-l-methyl-lW-indol-3-ylmethylene)-l-piperidin-l- ylmethyl-l,3-dihydro-indol-2-one

General procedure I was followed using 44 mg (0.15 mmol) of 3-(5-fluoro-l-methyl-l/f- indol-3-ylmethylene)-l,3-dihydro-indol-2-one, 0.115 mL (10 eq.) formaldehyde solution and 0.148 mL (10 eq.) piperidine in 2 mL methanol. Heating at 60 0 C for 16 h. Filtration of the precipitate afforded 40 mg (68%) of the title compound as pale-yellow powder. 13 C NMR (CDCI 3 ) δ 168.1, 159.2, 141.1, 138.7, 133.4, 130.0, 128.3, 126.9, 126.2, 124.9, 121.5, 118.6, 117.6, 110.9, 110.8, 109.5, 103.6, 62.8, 52.3, 33.8, 25.9, 24.1

Example 31: synthesis of 3-(6-methoxy-l-methyl-lW-indol-3-ylmethylene)-l-piperidin- l-ylmethyl-l,3-dihydro-indol-2-one

General procedure I was followed using 46 mg (0.15 mmol) of 3-(6-methoxy-l-methyl- lAy-indol-3-ylmethylene)-l,3-dihydro-indol-2-one, 0.115 mL (10 eq.) formaldehyde solution and 0.148 mL (10 eq.) piperidine in 2 mL methanol. Heating at 60 0 C for 16 h. Filtration of the precipitate afforded 51 mg (85%) of the title compound as orange powder.

13 C NMR (CDCI 3 ) δ 168.1, 157.1, 141.0, 137.7, 136.7, 126.9, 126.6, 125.1, 123.3, 121.4, 118.7, 118.2, 117.5, 111.0, 110.8, 109.4, 93.8, 62.8, 55.8, 52.2, 33.6, 25.9, 24.1

Example 32: synthesis of 3-(l-benzenesulfonyl-lW-indol-3-ylmethylene)-l-piperidin-l- ylmethyl-l,3-dihydro-indol-2-one

General procedure I was followed using 60 mg (0.15 mmol) of 3-(l-benzenesulfonyl-l/-/- indol-3-ylmethylene)-l,3-dihydro-indol-2-one, 0.115 mL (10 eq.) formaldehyde solution and 0.148 mL (10 eq.) piperidine in 2 mL methanol. Heating at 60 0 C for 16 h. Filtration of the precipitate afforded 64 mg (86%) of the title compound as yellow powder.

13 C NMR (CDCI 3 ) δ 167.3, 142.4, 138.1, 134.5, 134.1, 132.4, 130.6, 129.4, 128.6, 127.1, 125.2, 125.0, 123.9, 123.8, 123.2, 121.7, 118.5, 118.5, 115.7, 113.8, 109.8, 62.8, 52.2, 25.9, 24.1

Example 33: synthesis of l-piperidin-l-ylmethyl-3-(4,5,6,7-tetrafluoro-lH-indol-3- ylmethylene)-l,3-dihydro-indol-2-one

General procedure I was followed using 48 mg (0.15 mmol) of 3-(4,5,6,7-tetrafluoro- l#-indol-3-ylmethylene)-l,3-dihydro-indol-2-one, 14 μL (1.17 eq.) formaldehyde solution and 17 μL (1.17 eq.) piperidine in 2 ml_ methanol. Heating at 60 0 C for 16 h.

Filtration of the precipitate afforded 39 mg (61%) of the title compound as yellow powder.

1 H NMR (DMSO-de) E/Z mixture δ 13.04 (br,lH), 9.43 (s,0.5H), 9.42 (s,0.5H), 7.95 (s,0.5H), 7.90 (s,0.5H), 7.56 (d,0.5H), 7.52 (d,0.5H), 7.30 - 6.95 (m,2.5H), 6.86

(d,0.5H), 4.50 (s,2H), 2.53 (m,4H), 1.47 (m,4H), 1.33 (m,2H)

Example 34: synthesis of 3-(7-methyl-l#-indol-3-ylmethylene)-l-piperidin-l-ylmethyl- l,3-dihydro-indol-2-one

General procedure I was followed using 41 mg (0.15 mmol) of 3-(7-methyl-lA/-jndol-3- ylmethylene)-l,3-dihydro-indol-2-one, 14 μL (1.17 eq.) formaldehyde solution and 17

μL (1.17 eq.) piperidine in 2 mL methanol. Heating at 60 0 C for 16 h. Filtration of the precipitate afforded 38 mg (68%) of the title compound as orange powder. 13 C NMR (CDCI 3 ) δ 168.1, 141.2, 135.3, 133.0, 128.0, 127.3, 127.0, 125.0, 123.7, 121.6, 121.5, 121.1, 119.4, 117.7, 115.6, 112.6, 109.5, 62.8, 52.3, 25.9, 24.1, 16.6

Example 35: synthesis of Λ/-cyclohexyl-2-[2-methyl-3-(2-oxo-l-piperidin-l-ylmethyl- l,2-dihydro-indol-3-ylidenemethyl)-indol-l-yl]-acetamide

General procedure I was followed using 62 mg (0.15 mmol) of Λ/-cyclohexyl-2-[2- methyl-3-(2-oxo-l,2-dihydro-indol-3-ylidenemethyl)-indol-l-y l]-acetamide, 14 μL (1.17 eq.) formaldehyde solution and 17 μL (1.17 eq.) piperidine in 2 mL methanol. Heating at 60 0 C for 16 h. Filtration of the precipitate afforded 53 mg (69%) of the title compound as yellow powder.

13 C NMR (CDCI 3 ) δ 169.6, 166.3, 144.0, 138.4, 136.6, 128.8, 128.5, 126.3, 125.2, 123.9, 123.1, 121.9, 121.5, 121.4, 111.1, 109.6, 109.2, 63.0, 52.2, 48.2, 47.4, 32.7, 32.5, 25.9, 25.2, 24.4, 24.1

Example 36: synthesis of Λ/-(2-methoxy-ethyl)-2-[3-(2-oxo-l-piperidin-l-ylmethyl-l,2 - dihydro-indol-3-ylidenemethyl)-indol-l-yl]-acetamide

General procedure I was followed using 56 mg (0.15 mmol) of Λ/-(2-methoxy-ethyl)-2- [3-(2-oxo-l,2-dihydro-indol-3-ylidenemethyl)-indol-l-yl]-ace tamide, 14 μL (1.17 eq.) formaldehyde solution and 17 μL (1.17 eq.) piperidine in 2 ml_ methanol. Heating at 60 0 C for 16 h. Filtration of the precipitate afforded 40 mg (56%) of the title compound as yellow powder.

13 C NMR (DMSO-dβ) δ 166.9, 166.6, 140.5, 137.5, 136.3, 128.7, 127.0, 126.7, 124.3, 122.6, 121.1, 118.6, 118.4, 117.9, 110.6, 109.3, 70.4, 61.7, 57.8, 51.6, 49.2, 25.3, 23.6

Example 37: synthesis of 3-[l-(2-morpholin-4-yl-2-oxo-ethyl)-lW-indol-3-ylmethylene]- l-piperidin-l-ylmethyl-l,3-dihydro-indol-2-one

General procedure I was followed using 58 mg (0.15 mmol) of 3-[l-(2-morpholin-4-yl-2- oxo-ethyl)-lH-indol-3-ylmethylene]-l,3-dihydro-indol-2-one, 14 μL (1.17 eq.) formaldehyde solution and 17 μL (1.17 eq.) piperidine in 2 mL methanol. Heating at 60 0 C for 16 h. Filtration of the precipitate afforded 59 mg (81%) of the title compound as yellow powder.

13 C NMR (CDCI 3 ) δ 168.0, 164.9, 141.2, 137.1, 136.8, 129.1, 127.0, 126.4, 124.9, 123.2, 121.6, 121.5, 119.4, 118.3, 117.8, 111.9, 110.2, 109.4, 66.8, 66.3, 62.7, 52.3, 48.7, 45.5, 42.5, 25.9, 24.1

Example 38: synthesis of 5-bromo-3-(l/y-indol-3-ylmethylene)-l-piperidin-l-ylmethyl- l,3-dihydro-indol-2-one

General procedure I was followed using 42 mg (0.12 mmol) of 5-bromo-3-(lA/-indol-3- ylmethylene)-l,3-dihydro-indol-2-one, 11 μl_ (1.17 eq.) formaldehyde solution and 13 μL (1.17 eq.) piperidine in 2 mL methanol. Heating at 60 0 C for 16 h. Filtration of the precipitate afforded 45 mg (86%) of the title compound as orange powder. 13 C NMR (DMSO-Gf 6 ) δ 166.5, 139.2, 135.8, 134.5, 129.8, 128.5, 128.3, 126.8, 122.6, 121.0, 120.9, 118.7, 116.1, 113.5, 112.3, 111.4, 111.1, 61.7, 51.5, 25.3, 23.6

Example 39: synthesis of 7-bromo-3-(lH-indol-3-ylmethylene)-l-piperidin-l-ylmethyl- l,3-dihydro-indol-2-one

General procedure I was followed using 42 mg (0.12 mmol) of 7-bromo-3-(l/f-indol-3- ylmethylene)-l,3-dihydro-indol-2-one, 11 μL (1.17 eq.) formaldehyde solution and 13 μL (1.17 eq.) piperidine in 2 mL methanol. Heating at 60 0 C for 16 h. Filtration of the precipitate afforded 48 mg (92%) of the title compound as orange powder. 1 H NMR (CDCI 3 ) δ 9.49 (s,lH), 7.98 (s,lH), 7.91 (m,lH), 7.72 (br,lH), 7.61 - 7.51 (m,2H), 7.37 - 7.28 (m,3H), 6.97 (t,lH), 5.00 (s,2H), 2.60 (m,4H), 1.59 (m,4H), 1.37 (m,2H)

Example 40: synthesis of 6-bromo-3-(l/f-indol-3-ylmethylene)-l-piperidin-l-ylmethyl- l,3-dihydro-indol-2-one

General procedure I was followed using 13 mg (0.04 mmol) of 6-bromo-3-(l#-indol-3- ylmethylene)-l,3-dihydro-indol-2-one, 3 μl_ (1.17 eq.) formaldehyde solution and 4 μl_ (1.17 eq.) piperidine in 2 mL methanol. Heating at 60 0 C for 16 h. Filtration of the precipitate afforded 11 mg (84%) of the title compound as brown powder. 13 C NMR (DMSO-Gf 6 ) δ 166.7, 141.4, 135.9, 134.2, 129.0, 128.2, 123.8, 123.6, 122.6, 120.9, 119.9, 118.9, 118.5, 116.3, 112.3, 112.1, 111.3, 61.6, 51.4, 25.3, 23.6

Example 41: synthesis of 6-fluoro-l-piperidin-l-ylmethyl-3-(l-piperidin-l-ylmethyl-lÎ »/- indol-3-ylmethylene)-l,3-dihydro-indol-2-one

General procedure I was followed using 42 mg (0.15 mmol) of 6-fluoro-3-(lH-indol-3- ylmethylene)-l,3-dihydro-indol-2-one, 13 μl_ (1.17 eq.) formaldehyde solution and 16 μl_ (1.17 eq.) piperidine in 2 mL methanol. Heating at 60 0 C for 16 h. Filtration of the precipitate afforded 26 mg (36%) of the title compound as brown powder. 1 H NMR (DMSO-CZ 6 ) δ 9.39 (s,lH), 8.23 (m,lH), 8.19 (s,lH), 7.98 (m,lH), 7.75 (m,lH), 7.27 (m,2H), 7.03 (d,lH), 6.88 (t,lH), 5.09 (s,2H), 4.52 (s,2H), 2.53 (m,8H), 1.47 (m,8H), 1.31 (m,4H)

Example 42: synthesis of 6-chloro-3-(l/Y-indol-3-ylmethylene)-l-piperidin-l-ylmethyl- l,3-dihydro-indol-2-one

General procedure I was followed using 35 mg (0.12 mmol) of 6-chloro-3-(lW-indol-3- ylmethylene)-l,3-dihydro-indol-2-one, 10 μl_ (1.17 eq.) formaldehyde solution and 12 μl_ (1.17 eq.) piperidine in 2 mL methanol. Heating at 60 0 C for 16 h. Filtration of the precipitate afforded 30 mg (64%) of the title compound as brown powder. 13 C NMR (DMSO-CZ 6 ) δ 166.8, 141.3, 135.8, 134.2, 130.6, 128.9, 128.2, 123.4, 122.6, 120.9, 120.7, 119.6, 118.4, 116.3, 112.3, 111.3, 109.4, 61.6, 51.4, 25.3, 23.6

Example 43: synthesis of acetic acid 3-(2-oxo-l-piperidin-l-ylmethyl-l,2-dihydro-indol- 3-ylidenemethyl)-indol-l-ylmethyl ester

To a solution of acetic acid 3-formyl-indol-l-ylmethyl ester (739 mg, 3.4 mmol) in CH 2 CI 2 (40 mL) in a pressure flask were added indolinone (453 mg, 1 eq.) and piperidine (1.0 mL, 3 eq.). The mixture was stirred at 50 0 C for 3 days. A white precipitate was filtered off. The mother liquor was concentrated and triturated with CH 2 CI 2 . A yellow solid was filtered off. The mother liquor was washed with water (3 x), dried over Na 2 SO 4 , filtered and purified by chromatography to afford 463 mg (32%) of the title compound. 13 C NMR (DMSO-Cf 6 ) δ 169.9, 166.8, 140.9, 136.2, 135.5, 128.8, 127.3, 126.2, 124.0, 123.3, 121.8, 121.2, 119.9, 118.8, 118.8, 111.8, 110.8, 109.5, 68.7, 61.7, 51.6, 25.3, 23.6, 20.5

Example 44: synthesis of l-hydroxymethyl-3-(ltf-indol-3-ylmethylene)-l,3-dihydro- indol-2-one

General procedure I was followed using 700 mg (2.29 mmol) of 6-chloro-3-(lH-indol-3- ylmethylene)-l,3-dihydro-indol-2-one, 204 μl_ (1.17 eq.) formaldehyde solution and 377 μl_ (1.17 eq.) diisopropylamine in 30 ml_ methanol. Heating at 60 0 C for 16 h. After cooling to r.t., the mixture was placed in the fridge for 1 day. Filtration of the precipitate afforded 408 mg (61%) of the title compound as yellow powder. 1 H NMR (DMSO-tf 6 ) δ 12.08 (br,lH), 9.49 (s,lH), 8.23 (s,lH), 8.20 (m,lH), 7.96 ((.,1H), 7.53 (m,lH), 7.25 (m,3H), 7.13 (d,lH), 7.08 (t,lH), 6.17 (t,lH), 5.25 (d,2H)

Example 45: synthesis of acetic acid l-[3-(l-acetyl-lA/-indol-3-ylmethylene)-2-oxo-2,3- dihydro-indol-l-yl]-ethyl ester

To a solution of 3-(lA/-indol-3-ylmethylene)-l,3-dihydro-indol-2-one (780 mg, 3.0 mmol) in DMF (25 mL) were added acetaldehyde (1.7 ml_, 10 eq.), acetic anhydride (1.13 mL, 4 eq.) and triethylamine (1.67 mL, 4 eq.). The solution was heated to 80 0 C for 4 h and concentrated in vacuo. Flash chromatography (PE - EtOAc) afforded 173 mg (15%) of the title compound as an orange foam.

13 C NMR (DMSO-de) δ 169.7, 168.9, 164.6, 137.6, 134.6, 132.5, 129.9, 128.5, 125.8, 125.5, 123.9, 123.9, 123.0, 122.0, 120.1, 119.1, 115.9, 115.0, 110.5, 72.7, 23.8, 20.4, 17.8

Example 46: synthesis of 3-(lW-indol-3-ylmethylene)-l-(tetrahydro-pyran-2-yloxy)-l,3- dihydro-indol-2-one

To a solution of l-hydroxy-3-(lA/-indol-3-ylmethylene)-l,3-dihydro-indol-2-on e (50 mg,

0.18 mmol) in dry THF (ImL) was added p-toluene sulfonic acid (12 mg) under argon.

The solution was heated to 50 0 C before slow addition of a solution of 3,4-dihydro-2A/- pyran (20 μl_, 0.22 mmol) in dry THF (0.5 ml_). The reaction mixture was shaken at 50

0 C overnight. After concentration in vacuo flash chromatography (PE - EtOAc) afforded

42 mg (65%) of the title compound.

1 H NMR (DMSO-d 6 ) δ 10.80 (br,lH), 9.68 (s,lH), 8.20 (m,2H), 7.95 (d,lH), 7.70

(m,lH), 7.36 - 7.19 (m,3H), 7.05 (t,lH), 6.96 (d,lH), 5.79 (t,lH), 3.99 (m,lH), 3.8 (m,lH), 2.17 - 1.97 (m,3H), 1.83 (m,lH), 1.63 (m,2H)

Example 47: synthesis of 3-(lH-indol-3-ylmethylene)-l-(2-methoxy-ethoxy)-l,3- dihydro-indol-2-one

To a solution of l-hydroxy-3-(l/7-indol-3-ylmethylene)-l,3-dihydro-indol-2-on e (100 mg, 0.36 mmol) in dry THF (ImL) under argon were added 2-methoxy ethanol (43 μL, 0.54 mmol) and tributylphosphine (0.40 mmol). The solution was cooled to 0 0 C. A

solution of l,l'-(azodicarbonyl)dipiperidine (103 mg, 0.40 mmol) in dry THF (1 ml_) was added dropwise. The ice-bath was removed and the reaction mixture was stirred at r.t. for 20 h. After filtration, the filtrate was concentrated in vacuo and purified by flash chromatography (PE-EtOAc) to afford 41 mg of the title compound as yellow crystals. 1 H NMR (CDCI 3 ) δ 9.64 (s,lH), 8.03 (s,lH), 7.93 (m,lH), 7.64 (d,lH), 7.51 (m,lH), 7.35 - 7.21 (m,4H), 7.17 - 7.08 (m,2H), 3.8 - 3.7 (m,4H), 3.45 (s,3H)

Example 48: synthesis of 3-[l-(2-hydroxy-ehtyl)-lH-indole-3-ylmethylene]-l,3-dihydro- indol-2-one

20 21

(2-Bromo-ethoxy)-te/t-butyl-dimethyl-silane (18)

To a solution of 2-bromoethanol (568 μL, 8.0 mmol) in CH 2 CI 2 (10 ml_) were added imidazole (545 mg, 8.0 mmol) and te/t-butyl-dimethyl-chlorosilane (1.4 g, 8.8 mmol).

Stirring was continued overnight. The solution was washed with water (3 times) and brine, it was dried over MgSO 4 , filtered and concentrated in vacuo to afford 1.5 g the title compound.

1 H NMR (CDCI 3 ) δ 3.88 (t,2H), 3.39 (t,2H), 0.90 (s,9H), 0.08 (s,6H)

l-[2-(tert-Butyl-dimethyl-silanyloxy)-ethyl]-l^-indole-3- carbaldehyde (19) Indole-3-carbaldehyde (580 mg, 4.0 mmol) in DMF (1OmL) was cooled to 0 0 C. NaH (60%, 240 mg, 6.0 mmol) was added. The suspension was stirred for 30 min before addition of (2-bromo-ethoxy)-£e/t-butyl-dimethyl silane. Stirring was continued at r.t.

for 2 h before addition of water. The mixture was extracted with diethyl ether (3 times). The combined extracts were washed with brine, dried over MgSO 4 , filtrated and concentrated in vacuo. Flash chromatography (PE - EtOAc) afforded 436 mg (36%) of the title compound as yellow crystals. 1 H NMR (CDCI 3 ) 10.00 (s,lH), 8.31 (m,lH), 7.78 (s,lH), 7.40 - 7.25 (m,3H), 4.28 (t,2H), 3.95 (t,2H), 0.80 (s,9H), -0.16 (s,6H)

3-{l-[2-(te/t-Butyl-dimethyl-silanyloxy)-ethyl]-lA/-indol e-3-ylmethylene}-l,3-dihydro- indol-2-one (20) General procedure C was followed using indolinone (133 mg, 1.4 mmol) and l-[2-(tert- butyl-dimethyl-silanyloxy)-ethyl]-lf/-indole-3-carbaldehyde (436 mg, 1.4 mmol). The reaction time was 3 h. Flash chromatography (PE - EtOAc) afforded 223 mg (53%) of the title compound as orange crystals.

1 H NMR (DMSO-de) Z isomer δ 10.48 (s,lH), 9.48 (s,lH), 8.18 (m,lH), 8.10 (s,lH), 7.87 (d,lH), 7.61 (m,lH), 7.26 (m,2H), 7.13 (t,lH), 6.98 (t,lH), 6.83 (d,lH), 4.43

(t,2H), 3.94 (t,2H), 0.70 (s,9H), -0.25 (s,6H)

3-[l-(2-hydroxy-ehtyl)-lW-indole-3-ylmethylene]-l,3-dihyd ro-indol-2-one (21)

3-{l-[2-(ϋe/t-Butyl-dimethyl-silanyloxy)-ethyl]-l/y-indo le-3-ylmethylene}-l,3-dihydro- indol-2-one ( 233 mg, 0.56 mmol) was dissolved in THF (5 ml_) and was stirred upon addition of TBAF (560 μl_, 1 eq.). Stirring was continued at r.t. for 20 min. The precipitate was filtered and washed with cold water (4 times). Drying in vacuo afforded 123 mg (72%) of the title compound as yellow solid.

1 H NMR (DMSO-Cy 6 ) δ 10.49 (s,lH), 9.47 (s,lH), 8.19 (m,lH), 8.11 (s,lH), 7.87 (d,lH), 7.62 (m,lH), 7.27 (m,2H), 7.13 (t,lH), 6.98 (t,lH), 6.84 (d,lH), 4.99 (br,lH), 4.36 (t,2H), 3.80 (t,2H)

Example 49: synthesis of 3-(ltf-indol-3-ylmethylene)-2-oxo-2,3-dihydro-indole-l- carboxylic acid benzyl ester

To a solution of 3-(lH-indol-3-ylmethylene)-l,3-dihydro-indol-2-one (0.13 g, 0.5 mmol) in DMF (5ml_) was added NaH (60% in oil, 20 mg, 0.5 mmol). The suspension was stirred for 10 min before cooling to 0 0 C. Chloroformic acid benzyl ester (71 μL, 0.5 mmol) was added and the mixture was stirred for 2 h. Water was added and the mixture was extracted with EtOAc. The organic phase was washed with water and brine, concentrated in vacuo and purified by flash chromatography to afford 50 mg (25%) of the title compound.

13 C NMR (DMSOd 6 ) E/Z mixture δ 168.6, 167.7, 150.1, 149.9, 142.8, 140.3, 135.2, 134.7, 134.4, 131.1, 130.1, 129.9, 128.7, 128.6, 128.6, 128.3, 127.8, 127.2, 125.6, 125.4, 125.3, 124.9, 124.6, 123.9, 123.7, 123.6, 122.9, 121.3, 121.0, 120.9, 120.1, 120.0, 119.4, 115.6, 115.1, 115.0, 114.8, 110.1, 109.4, 68.9

Example 50: synthesis of 3-(l-acetoxymethyl-lAHndol-3-ylmethylene)-2-oxo-2,3- dihydro-indole-1-carboxylic acid tert-butyl ester

2-Oxo-2,3-dihydro-indole-l-carboxylic acid te/t-butyl ester (22)

To a solution of indolinone (1.0 g, 7.57 mmol) in acetonitrile (50 mL) were added DMAP

(100 mg, 0.82 mmol) and di-terf-butyl dicarbonate (1.8 g, 8.33 mmol). The solution was stirred at r.t. for 3 days. After evaporation of the solvent, the residue was taken in

CH 2 CI 2 , washed with 10% aq. KHSO 4 , sat. aq. NaHCO 3 and water. Flash chromatography afforded 564 mg (32%) of the title compound.

1 H NMR (DMSO-Gf 5 ) δ 7.69 (d,lH), 7.30 (m,2H), 7.14 (t,lH), 3.73 (s,2H), 1.57 (s,9H)

3-(lW-Indol-3-ylmethylene)-2-oxo-2,3-dihydro-indole-l-car boxylic acid te/t-butyl ester (23)

2-0x0-2, 3-dihydro-indole-l-carboxylic acid te/t-butyl ester (564 mg, 2.42 mmol), indole -3-carbaldehyde (351 mg, 2.42 mmol), piperidine (3 drops) and ethanol (5 mL) were heated to reflux for 15 min. The reaction mixture was allowed to cool to r.t. After filtration the product was dried in vacuo to afford 782 mg (55%) of the title compound. 1 H NMR (DMSO-Of 6 ) δ 12.17 (br,lH), 9.36 (s,lH), 8.31 (s,lH), 8.24 (m,lH), 8.06 (d,lH), 7.75 (d,lH), 7.56 (m,lH), 7.34 - 7.16 (m,4H), 1.63 (s,9H)

3-(l-acetoxymethyl-l^-indol-3-ylmethylene)-2-oxo-2,3-dihy dro-indole-l-carboxylic acid te/t-butyl ester (24)

To a solution of 3-(lW-indol-3-ylmethylene)-2-oxo-2,3-dihydro-indole-l-carbox ylic acid te/t-butyl ester (151 mg, 0.42 mmol) in DMF (8 ml_) was added NaH (60% in oil, 25 mg, 0.63 mmol). The mixture was stirred at r.t. for 30 min before cooling to 0 0 C and addition of chloromethyl acetate (38 μl_, 0.42 mmol). The ice-bath was removed and the mixture was stirred at r.t. overnight. H 2 O was added. The solution was extracted with

Et 2 O (3 x). The combined extracts were washed with water, brine, dried over Na 2 SO 4 , filtered, concentrated in vacuo and purified by flash chromatography to afford 50 mg (27%) of the title compound.

1 H NMR (DMSO-Cy 6 ) E/Z mixture δ 9.41 (s,0.5H), 8.44 (s,0.5H), 8.29 (d,0.5H), 8.26 (s,0.5H), 8.11 (d,0.5H), 7.95 (s,0.5H), 7.94 (d,0.5H), 7.87 (d,0.5H), 7.80 - 7.66 (m,2H), 7.44 - 7.13 (m,4H), 6.37 (s,2H), 2.06 (s,3H), 1.63 (s,4.5H), 1.61 (s,4.5H)

Example 51

Compounds exhibiting IL- 2 inhibitory activity

Inhibition of IL- 2 by oxindole compounds

Peripheral blood mononuclear cells (PBMCs) were isolated from human blood. The lymphocytes, a fraction of the PBMCs, were activated to secrete IL-2 using the polyclonal mitogen phytohemagglutinin. The test compounds were added in concentrations from 10 "5 to 10 "10 , and the cells (10 6 /ml) were incubated in microtiter wells at 37°C for approximately 46 hours.

After 46 hours the cells were centrifuged down for 25 minutes at 1000 x g and the supernatants were transferred to transferred to microtiter wells precoated with a monoclonal antibody against human IL-2. The IL-2 concentration in the supernatants was determined by a sandwich ELISA. Microtiter plates were coated (1 μg/ml) with a monoclonal antibody (R&D, UK) against human IL-2, washed 4 times, blocked with 1 % casein buffer for 2 hours and washed 2 times. 100 μl sample was added to each well and

incubated overnight. All samples were tested in triplicate. 100 μl IL-2 standards (R&D, UK) at a concentration range of 10000 -0 pg/ml were tested in triplicate. After incubation the plates were washed and incubated with biotinylated polyclonal secondary antibody against human IL-2 (R&D, UK) for 45 minutes and thereafter washed 4 times. 100 μl enzyme conjugate, horseradish peroxidase conjugated streptavidin diluted 1:4000 (Zymed, USA), was added to all wells and incubated for 30 minutes. 100 μl OPD substrate (KEM EN TEC-DK) was added and the enzyme/substrate reaction stopped after 10 minutes at room temperature with 50 μl 1 M H 2 SO 4 . The colour development (optical density - OD) was determined at 492 nm on an ELISA reader and the background OD at 620 nm subtracted).

The results are shown in Table 1 below. The results are expressed as a percentage of the control (PHA + DMSO) and the potency of the test compound was expressed as the concentration resulting in 50 % inhibition of the response of the control stimulated cells (PIC 50 , corresponding to -log IC 50 ).

Table 1

Example 52

EAE activity of compounds of formula I on oral administration

Materials and Methods Compounds

Compound A: acetic acid 3-(2-oxo-l,2-dihydro-indol-3-ylidenemethyl)-indol-l-ylmethyl ester;

Compound B: acetic acid 3-(2-oxo-l-piperidin-l-ylmethyl-l,2-dihydro-indol-3- ylidenemethyl)-indol-l-ylmethyl ester; Compound C: l-[(te/t-butyl-methyl-amino)-methyl]-3-(lH-indol-3-ylmethyle ne)-l,3- dihydro-indol-2-one;

Compound D: 3-(lW-Indol-3-ylmethylene)-l-piperidin-l-ylmethyl-l,3-dihydr o-indol-2- one;

Compound E: l-piperidin-l-ylmethyl-3-(l-piperidin-l-ylmethyl-l/-/-indol- 3- ylmethylene)-l,3-dihydro-indol-2-one.

Compound F: 3-(lA/-indol-3-ylmethylene)-l,3-dihydro-indol-2-one (Compound 57 in PCT/DK2004/000875)

Peptide The following peptide from myelin proteolipid protein was used; PLP 139-153 H- HCLGKWLGHPDKFVG-OH. The peptide was synthesized by Fmoc chemistry (Schafer-N, Copenhagen, Denmark). Purity (>95%) was verified by reversed- phase HPLC and integrity by mass spectrometry.

Mice Female SJL/J (H-2 S ) inbred mice purchased from Charles River.

Immunization The SJL/J mice (about 8 weeks old) were immunized on day 0 with the

PLP 139-153 peptide (dissolved in sterile NaCI) emulsified 1: 1 (vol/vol) in Complete Freund's

Adjuvant (5 mg Mycobacterium tuberculosis/ m\) (SSI, Copenhagen, Denmark).

Intradermal injections corresponding to 100 μg peptide and 125 μg Mycobacterium tuberculosis were given at the base of the tail in a total volume of 50 μl. The mice were additionally given an Lv. injection with 100 ng pertussis toxin (Sigma) dissolved in sterile NaCI on day 0 and day 2, injection volume was 100 μl.

Compound treatment Groups of 10 mice were dosed daily with compounds (50 mg/kg i.p. or p.o.) in suspension vehicle (4 g Tween-80, 2 g Carboxy-methyl cellulose 7H4XF, 8 g NaCI, 1 liter H 2 O), starting on day 0 (i.p.) or day 1 (p.o.). Control groups were given either suspension vehicle or dexamethasone (Dexadreson Vet, Intervet, Holland) (1 mg/kg). The pH in the suspension vehicle used was in some experiments adjusted to pH=4.0.

Clinical evaluation Mice were weighed and assessed clinically daily from day 5 p.i. according to the following criteria: 0, no disease; 1, tail paralysis; 2, clumsy gait/poor

righting ability and limb weakness; 3, moderate or total hind limb paralysis; 4, moribund state or dead.

Statistics

Area-under-curve (AUC) of the disease score was calculated for all mice. The medians of AUC of all groups were compared using Kruskal-Wallis test. When P<0.05 in the Kruskal- Wallis test, the Mann-Whitney test was used to compare drug treated groups with the suspension vehicle treated control group (P<0.05). AUC were calculated from day 0 to the termination of the experiment (day 21).

Results

A series of compounds were tested per orally and i.p. in the EAE model. The results are shown in table 2. All the shown inhibitory effects are significant (p<0.05). The compounds inhibit disease with varying effect ranging from 58% to 64% inhibition p.o. and from 59% to 100% inhibition i.p.. In conclusion, several compounds have the potential to inhibit EAE disease when given orally and several compounds exhibit a very high activity in the EAE model when dosed i.p. alone or as a combination.

Table 2. Inhibition of EAE

* Area-under-curve of the disease score. A suspension vehicle group was included in all experiments. AUC of the disease score were calculated for all groups and the Mann- Withney test was used to compare the treated groups with the suspension vehicle group. Some compounds have been tested both Lp. and p.o., some compounds have been tested several times, and some compounds have been tested as a 1.1 mixture.

** PH in the suspension vehicle used for compound suspensions was adjusted to pH=4.0 as was the suspension vehicle used for the control group.




 
Previous Patent: AN ACTUATOR

Next Patent: TONGUE BASED CONTROL METHOD AND SYSTEM