Substituted indolines as tyrosine kinase inhibitors

Abstract
The present invention encompasses compounds of general Formula (1) wherein R2, R3, Q, W, X, Y and Z are defined as in claim 1, which are suitable for the treatment of diseases characterized by excessive or abnormal cell proliferation, and the use thereof for preparing a medicament having the above-mentioned properties.
Description

The present invention relates to new indolinones of general formula (1)




embedded image



wherein the groups R2, R3, Q, W, X, Y and Z have the meanings given in the claims and specification, the isomers thereof, processes for preparing these indolinones and their use as medicaments.


The aim of the present invention is to discover new active substances which can be used for the prevention and/or treatment of diseases characterised by excessive or abnormal cell proliferation.


BACKGROUND TO THE INVENTION

Indolinones are described for example as receptor tyrosinekinases and cyclin/CDK-complex inhibiting compounds (WO02/081445, WO01/27081 or WO 2004/026829).







DETAILED DESCRIPTION OF THE INVENTION

It has now been found that, surprisingly, compounds of general formula (1), wherein the groups R2, R3, Q, W, X, Y and Z have the meanings given hereinafter act as inhibitors of specific cell cycle kinases. Moreover, certain forms also inhibit kinases that play an important part in intracellular signal transduction pathways. Thus, the compounds according to the invention may be used for example for the treatment of diseases connected with the activity of specific cell cycle kinases and characterised by excessive or abnormal cell proliferation. They may also be used to treat diseases characterised by abnormal activation of the corresponding signal transduction pathways.


The present invention relates to compounds of general formula (1)




embedded image



wherein


Q denotes C6-10aryl or 5-12 membered heteroaryl, optionally substituted by one or more identical or different R1, and


W denotes a bond, —O—, —NR6—, —CR6R6, and


X denotes —O—, —S, —S(O), —S(O2), —NR4—, —CR4R4— or —C(O)—; and


Y denotes —O—, —NR5—, —CR5R5— or —C(O)—; while optionally an R4 may form, with an R5, a 4-7 membered ring, optionally substituted by one or more R8; or the substituents R4 and R4, R5 and R5 or R6 and R6 bound to the carbon atom may together form a 4-7-membered ring, which may optionally contain one or more heteroatoms selected from among N, O and S; and


Z denotes —O—, —NR7— or —CR7R7, or


Y denotes a bond, X denotes —NR4— and Z denotes —NR7—, and at least one substituent selected from among X, Y and Z denotes a heteroatom; and


R1, R2, R3, R4, R5, R6, R7 und R8 each independently of one another denote a group selected from among Ra, Rb and Ra substituted by one or more identical or different Rb and/or Rc, wherein R2 and/or R3 is not hydrogen; and


each Ra independently of one another denotes hydrogen or is selected from among C1-6alkyl, C3-10cycloalkyl, C4-16cycloalkylalkyl, C6-10aryl, C7-16arylalkyl, 2-6 membered heteroalkyl, 3-8 membered heterocycloalkyl, 4-14 membered heterocycloalkylalkyl, 5-12 membered heteroaryl and 6-18 membered heteroarylalkyl; each Rb is a suitable group and each is independently selected from among ═O, —ORc, C1-3haloalkyloxy, —OCF3, ═S, —SRc, ═NRc, ═NORc, ═NNRcRc, ═NN(Rg)C(O)NRcRc, —NRcRc, —ONRcRc, —N(ORc)Rc, —N(Rg)NRcRc, halogen, —CF3, —CN, —NC, —OCN, —SCN, —NO, —NO2, ═N2, —N3, —S(O)Rc, —S(O)ORc, —S(O)2Rc, —S(O)2ORc, —S(O)NRcRc, —S(O)2NRcRc, —OS(O)Rc, —OS(O)2Rc, —OS(O)2ORc, —OS(O)NRcRc, —OS(O)2NRcRc, —C(O)Rc, —C(O)ORc, —C(O)SRc, —C(O)NRcRc, —C(O)N(Rg)NRcRc, —C(O)N(Rg)ORc, —C(NRg)NRcRc, —C(NOH)Rc, —C(NOH)NRcRc, —OC(O)Rc, —OC(O)ORc, —OC(O)SRc, —OC(O)NRcRc, —OC(NRg)NRcRc, —SC(O)Rc, —SC(O)ORc, —SC(O)NRcRc, —SC(NRg)NRcRc, —N(Rg)C(O)Rc, —N[C(O)Rc]2, —N(ORg)C(O)Rc, —N(Rg)C(NRg)Rc, —N(Rg)N(Rg)C(O)Rc, —N[C(O)Rc]NRcRc, —N(Rg)C(S)Rc, —N(Rg)S(O)Rc, —N(Rg)S(O)ORc, —N(Rg)S(O)2Rc, N[S(O)2Rc]2, —N(Rg)S(O)2ORc, —N(Rg)S(O)2NRcRc, —N(Rg)[S(O)2]2Rc, N(Rg)C(O)ORc, —N(Rg)C(O)SRc, —N(Rg)C(O)NRcRc, —N(Rg)C(O)NRgNRcRc, N(Rg)N(Rg)C(O)NRcRc, —N(Rg)C(S)NRcRc, —[N(Rg)C(O)]2Rc, —N(Rg)[C(O)]2Rc, N{[C(O)]2Rc}2, —N(Rg)[C(O)]2ORc, —N(Rg)[C(O)]2NRcRc, —N{[C(O)]2ORc}2, —N{[C(O)]2NRcRc}2, —[N(Rg)C(O)]2ORc, —N(Rg)C(NRg)ORc, —N(Rg)C(NOH)Rc, N(Rg)C(NRg)SRc and —N(Rg)C(NRg)NRcRc,


each Re independently of one another denotes hydrogen or a group optionally substituted by one or more identical or different Rd and/or Re selected from among C1-6alkyl, C3-10cycloalkyl, C4-11cycloalkylalkyl, C6-10aryl, C7-16arylalkyl, 2-6 membered heteroalkyl, 3-8 membered heterocycloalkyl, 4-14 membered heterocycloalkylalkyl, 5-12 membered heteroaryl and 6-18 membered heteroarylalkyl;


each Rd is a suitable group and each is independently selected from among ═O, —ORe, C1-3haloalkyloxy, —OCF3, ═S, —SRe, ═NRe, ═NORe, ═NNReRe, ═NN(Rg)C(O)NReRe, —NReRe, —ONReRe, —N(Rg)NReRe, halogen, —CF3, —CN, —NC, —OCN, —SCN, —NO, —NO2, ═N2, —N3, —S(O)Re, —S(O)ORe, —S(O)2Re, —S(O)2ORe, —S(O)NReRe, —S(O)2NReRe, —OS(O)Re, —OS(O)2Re, —OS(O)2ORe, —OS(O)NReRe, —OS(O)2NReRe, —C(O)Re, —C(O)ORe, —C(O)SRe, —C(O)NReRe, —C(O)N(Rg)NReRe, —C(O)N(Rg)ORe, —C(NRg)NReRe, —C(NOH)Re, —C(NOH)NReRe, —OC(O)Re, —OC(O)ORe, —OC(O)SRe, —OC(O)NReRe, —OC(NRg)NReRe, —SC(O)Re, —SC(O)ORe, —SC(O)NReRe, —SC(NRg)NReRe, —N(Rg)C(O)Re, —N[C(O)Re]2, —N(ORg)C(O)Re, —N(Rg)C(NRg)Re, —N(Rg)N(Rg)C(O)Re, —N[C(O)Re]NReRe, —N(Rg)C(S)Re, —N(Rg)S(O)Re, —N(Rg)S(O)ORe—N(Rg)S(O)2Re, —N[S(O)2Rc]2, N(Rg)S(O)2ORe, N(Rg)S(O)2NReRe, N(Rg)[S(O)2]2Re, —N(Rg)C(O)ORe, —N(Rg)C(O)SRe, N(Rg)C(O)NReRe, N(Rg)C(O)NRgNReRe, —N(Rg)N(Rg)C(O)NReRe, —N(Rg)C(S)NReRe, —[N(Rg)C(O)]2Re, —N(Rg)[C(O)]2Re, —N{[C(O)]2Re}2, —N(Rg)[C(O)]2ORe, —N(Rg)[C(O)]2NReRe, —N{[C(O)]2ORe}2, —N{[C(O)]2NReRe}2, —[N(Rg)C(O)]2ORe, —N(Rg)C(NRg)ORe, N(Rg)C(NOH)Re, —N(Rg)C(NRg)SRe and —N(Rg)C(NRg)NReRe,


each Re independently of one another denotes hydrogen or a group optionally substituted by one or more identical or different Rf and/or Rg selected from among C1-6alkyl, C3-8cycloalkyl, C4-11cycloalkylalkyl, C6-10aryl, C7-16arylalkyl, 2-6 membered heteroalkyl, 3-8 membered heterocycloalkyl, 4-14 membered heterocycloalkylalkyl, 5-12 membered heteroaryl and 6-18 membered heteroarylalkyl;


each Rf is a suitable group and each is independently selected from among halogen and —CF3; and


each Rg independently of one another denotes hydrogen, C1-6alkyl, C3-8cycloalkyl, C4-11cycloalkylalkyl, C6-10aryl, C7-16arylalkyl, 2-6 membered heteroalkyl, 3-8 membered heterocycloalkyl, 4-14 membered heterocycloalkyl, 5-12 membered heteroaryl or 6-18 membered heteroarylalkyl, optionally in the form of the prodrugs, the tautomers, the racemates, the enantiomers, the diastereomers and the mixtures thereof, and optionally the pharmacologically acceptable acid addition salts thereof.


In one aspect the invention relates to compounds of general formula (1), wherein Z denotes —NR7—.


In another aspect the invention relates to compounds of general formula (1), wherein R7 denotes hydrogen or C1-6alkyl.


In another aspect the invention relates to compounds of general formula (1), wherein Q denotes phenyl.


In another aspect the invention relates to compounds of general formula (1), wherein X denotes —NR4—.


In another aspect the invention relates to compounds of general formula (1), wherein R4 denotes hydrogen.


In another aspect the invention relates to compounds of general formula (1), wherein Y denotes —CR5R5—.


In another aspect the invention relates to compounds of general formula (1), wherein R5 in each case denotes hydrogen.


In another aspect the invention relates to compounds of general formula (1) as pharmaceutical compositions.


In another aspect the invention relates to compounds of general formula (1) for preparing a pharmaceutical composition with an antiproliferative or signal transduction inhibiting activity.


In another aspect the invention relates to a pharmaceutical preparation, containing as active substance one or more compounds of general formula (1) or the physiologically acceptable salts thereof, optionally in combination with conventional excipients and/or carriers.


In another aspect the invention relates to the use of compounds of general formula (1) for preparing a pharmaceutical composition for the treatment and/or prevention of cancer, infections, inflammations and autoimmune diseases.


In another aspect the invention relates to a pharmaceutical preparation comprising a compound of general formula (1) and at least one further cytostatic or cytotoxic active substance, different from formula (1), optionally in the form of the prodrugs, the tautomers, the racemates, the enantiomers, the diastereomers and the mixtures thereof, and optionally the pharmacologically acceptable acid addition salts thereof.


DEFINITIONS

As used herein, the following definitions apply, unless stated otherwise.


Alkyl is made up of the sub-groups saturated hydrocarbon chains and unsaturated hydrocarbon chains, while the latter may be further subdivided into hydrocarbon chains with a double bond (alkenyl) and hydrocarbon chains with a triple bond (alkynyl). Alkenyl contains at least one double bond, alkynyl at least one triple bond. If a hydrocarbon chain should have both at least one double bond and at least one triple bond, by definition it belongs to the alkynyl sub-group. All the above-mentioned sub-groups may be further subdivided into straight-chain (unbranched) and branched. If an alkyl is substituted, it may be mono- or polysubstituted independently of one another at all the hydrogen-carrying carbon atoms.


Examples of individual sub-groups are listed below.


Straight-Chain (Unbranched) or Branched, Saturated Hydrocarbon Chains:


methyl; ethyl; n-propyl; isopropyl (1-methylethyl); n-butyl; 1-methylpropyl; isobutyl (2-methylpropyl); sec.-butyl (1-methylpropyl); tert.-butyl (1,1-dimethylethyl); n-pentyl; 1-methylbutyl; 1-ethylpropyl; isopentyl (3-methylbutyl); neopentyl (2,2-dimethyl-propyl); n-hexyl; 2,3-dimethylbutyl; 2,2-dimethylbutyl; 3,3-dimethylbutyl; 2-methyl-pentyl; 3-methylpentyl; n-heptyl; 2-methylhexyl; 3-methylhexyl; 2,2-dimethylpentyl; 2,3-dimethylpentyl; 2,4-dimethylpentyl; 3,3-dimethylpentyl; 2,2,3-trimethylbutyl; 3-ethylpentyl; n-octyl; n-nonyl; n-decyl etc.


Straight-Chained (Unbranched) or Branched Alkenyl:


vinyl (ethenyl); prop-1-enyl; allyl (prop-2-enyl); isopropenyl; but-1-enyl; but-2-enyl; but-3-enyl; 2-methyl-prop-2-enyl; 2-methyl-prop-1-enyl; 1-methyl-prop-2-enyl; 1-methyl-prop-1-enyl; 1-methylidenepropyl; pent-1-enyl; pent-2-enyl; pent-3-enyl; pent-4-enyl; 3-methyl-but-3-enyl; 3-methyl-but-2-enyl; 3-methyl-but-1-enyl; hex-1-enyl; hex-2-enyl; hex-3-enyl; hex-4-enyl; hex-5-enyl; 2,3-dimethyl-but-3-enyl; 2,3-dimethyl-but-2-enyl; 2-methylidene-3-methylbutyl; 2,3-dimethyl-but-1-enyl; hexa-1,3-dienyl; hexa-1,4-dienyl; penta-1,4-dienyl; penta-1,3-dienyl; buta-1,3-dienyl; 2,3-dimethylbuta-1,3-diene etc.


Straight-Chain (Unbranched) or Branched Alkynyl:


ethynyl; prop-1-ynyl; prop-2-ynyl; but-1-ynyl; but-2-ynyl; but-3-ynyl; 1-methyl-prop-2-ynyl etc.


By the terms propyl, butyl, pentyl, hexyl, heptyl, octyl, nonyl, decyl etc. unless otherwise stated are meant saturated hydrocarbon groups with the corresponding number of carbon atoms, including all the isomeric forms.


By the terms propenyl, butenyl, pentenyl, hexenyl, heptenyl, octenyl, nonenyl, decenyl etc. unless otherwise stated are meant unsaturated hydrocarbon groups with the corresponding number of carbon atoms and a double bond, including all the isomeric forms, also (Z)/(E)-isomers, where applicable.


By the terms butadienyl, pentadienyl, hexadienyl, heptadienyl, octadienyl, nonadienyl, decadienyl etc. unless otherwise stated are meant unsaturated hydrocarbon groups with the corresponding number of carbon atoms and two double bonds, including all the isomeric forms, also (Z)/(E)-isomers, where applicable.


By the terms propynyl, butynyl, pentynyl, hexynyl, heptynyl, octynyl, nonynyl, decynyl etc. unless otherwise stated are meant unsaturated hydrocarbon groups with the corresponding number of carbon atoms and a triple bond, including all the isomeric forms.


By the term heteroalkyl are meant groups which are derived from the alkyl as hereinbefore defined in its widest sense by replacing, in the hydrocarbon chains, one or more of the groups —CH3 independently of one another by the groups —OH, —SH or —NH2, one or more of the groups —CH2— independently of one another by the groups —O—, —S— or —NH—, one or more of the groups




embedded image



by the group




embedded image



one or more of the groups ═CH— by the group ═N—, one or more of the groups ═CH2 by the group ═NH or one or more of the groups ≡CH by the group ≡N, while a total of not more than three heteroatoms may be present in one heteroalkyl, there must be at least one carbon atom between two oxygen atoms and between two sulphur atoms or between one oxygen and one sulphur atom and the group as a whole must have chemical stability.


A direct result of the indirect definition/derivation from alkyl is that heteroalkyl is made up of the sub-groups saturated hydrocarbon chains with heteroatom(s), heteroalkenyl and heteroalkynyl, and it may be further subdivided into straight-chain (unbranched) and branched. If a heteroalkyl is substituted, it may be mono- or polysubstituted independently of one another at all the hydrogen-carrying oxygen, sulphur, nitrogen and/or carbon atoms. Heteroalkyl itself as a substituent may be attached to the molecule both through a carbon atom and through a heteroatom.


The following are listed by way of example:


dimethylaminomethyl; dimethylamino ethyl (1-dimethylamino ethyl; 2-dimethyl-aminoethyl); dimethylaminopropyl (1-dimethylaminopropyl, 2-dimethylaminopropyl, 3-dimethylaminopropyl); diethylamino methyl; diethylamino ethyl (1-diethylamino ethyl, 2-diethylamino ethyl); diethylaminopropyl (1-diethylaminopropyl, 2-diethylamino-propyl, 3-diethylaminopropyl); diisopropylaminoethyl (1-diisopropylaminoethyl, 2-di-isopropylaminoethyl); bis-2-methoxyethylamino; [2-(dimethylamino-ethyl)-ethyl-amino]-methyl; 3-[2-(dimethylamino-ethyl)-ethyl-amino]-propyl; hydroxymethyl; 2-hydroxy-ethyl; 3-hydroxypropyl; methoxy; ethoxy; propoxy; methoxymethyl; 2-methoxyethyl etc.


Halogen encompasses fluorine, chlorine, bromine and/or iodine atoms.


Haloalkyl is derived from alkyl as hereinbefore defined in its broadest sense, by replacing one or more hydrogen atoms of the hydrocarbon chain independently of one another by halogen atoms, which may be identical or different. A direct result of the indirect definition/derivation from alkyl is that haloalkyl is made up of the sub-groups saturated hydrohalogen chains, haloalkenyl and haloalkynyl, and it may be further subdivided into straight-chain (unbranched) and branched. If a haloalkyl is substituted, it may be mono- or polysubstituted independently of one another at all the hydrogen-carrying carbon atoms. Typical examples include, for example:


—CF3; —CHF2; —CH2F; —CF2CF3; —CHFCF3; —CH2CF3; —CF2CH3; —CHFCH3; —CF2CF2CF3; —CF2CH2CH3; —CF═CF2; —CCl═CH2; —CBr═CH2; —CI═CH2; —C≡C—CF3; —CHFCH2CH3; and —CHFCH2CF3.


Cycloalkyl is made up of the sub-groups monocyclic hydrocarbon rings, bicyclic hydrocarbon rings and spirohydrocarbon rings, while each sub-group may be further subdivided into saturated and unsaturated (cycloalkenyl). By unsaturated is meant that there is at least one double bond in the ring system, but no aromatic system is formed. In bicyclic hydrocarbon rings two rings are linked such that they share at least two carbon atoms. In spirohydrocarbon rings one carbon atom (spiroatom) is shared by two rings. If a cycloalkyl is substituted, it may be mono- or polysubstituted independently of one another at all the hydrogen-carrying carbon atoms. Cycloalkyl itself as a substituent may be attached to the molecule through any suitable position of the ring system. The following individual sub-groups are listed by way of example:


Monocyclic Saturated Hydrocarbon Rings:


cyclopropyl; cyclobutyl; cyclopentyl; cyclohexyl; cycloheptyl etc.


Monocyclic Unsaturated Hydrocarbon Rings:


cycloprop-1-enyl; cycloprop-2-enyl; cyclobut-1-enyl; cyclobut-2-enyl; cyclopent-1-enyl; cyclopent-2-enyl; cyclopent-3-enyl; cyclohex-1-enyl; cyclohex-2-enyl; cyclohex-3-enyl; cyclohept-1-enyl; cyclohept-2-enyl; cyclohept-3-enyl; cyclohept-4-enyl; cyclobuta-1,3-dienyl; cyclopenta-1,4-dienyl; cyclopenta-1,3-dienyl; cyclopenta-2,4-dienyl; cyclohexa-1,3-dienyl; cyclohexa-1,5-dienyl; cyclohexa-2,4-dienyl; cyclohexa-1,4-dienyl; cyclohexa-2,5-dienyl etc.


Saturated and Unsaturated Bicyclic Hydrocarbon Rings:


bicyclo[2.2.0]hexyl; bicyclo[3.2.0]heptyl; bicyclo[3.2.1]octyl; bicyclo[2.2.2]octyl; bicyclo[4.3.0]nonyl (octahydroindenyl); bicyclo[4.4.0]decyl (decahydronaphthalene); bicyclo[2.2.1]heptyl (norbornyl); (bicyclo[2.2.1]hepta-2,5-dienyl (norborna-2,5-dienyl); bicyclo[2.2.1]hept-2-enyl (norbornenyl); bicyclo[4.1.0]heptyl (norcaranyl); bicyclo-[3.1.1]heptyl (pinanyl) etc.


Saturated and Unsaturated Spirohydrocarbon Rings:


spiro[2.5]octyl, spiro[3.3]heptyl, spiro[4.5]dec-2-ene, etc.


Cycloalkylalkyl denotes the combination of the alkyl and cycloalkyl groups defined hereinbefore, in each case in their broadest sense. The alkyl group as substituent is directly linked to the molecule and is in turn substituted by a cycloalkyl group. The linking of alkyl and cycloalkyl in both groups may be effected by means of any suitable carbon atoms. The sub-groups of alkyl and cycloalkyl are also included in the combination of the two groups.


Aryl denotes mono-, bi- or tricyclic carbon rings with at least one aromatic ring. If an aryl is substituted, the substitution may be mono- or polysubstitution in each case, at all the hydrogen-carrying carbon atoms, independently of one another. Aryl itself may be linked to the molecule as substituent via any suitable position of the ring system. Typical examples include phenyl, naphthyl, indanyl (2,3-dihydroindenyl), 1,2,3,4-tetrahydronaphthyl and fluorenyl.


Arylalkyl denotes the combination of the groups alkyl and aryl as hereinbefore defined, in each case in their broadest sense. The alkyl group as substituent is directly linked to the molecule and is in turn substituted by an aryl group. The alkyl and aryl may be linked in both groups via any carbon atoms suitable for this purpose. The respective sub-groups of alkyl and aryl are also included in the combination of the two groups.


Typical examples include benzyl; 1-phenylethyl; 2-phenylethyl; phenylvinyl; phenylallyl etc.


Heteroaryl denotes monocyclic aromatic rings or polycyclic rings with at least one aromatic ring, which, compared with corresponding aryl or cycloalkyl, contain instead of one or more carbon atoms one or more identical or different heteroatoms, selected independently of one another from among nitrogen, sulphur and oxygen, while the resulting group must be chemically stable. If a heteroaryl is substituted, the substitution may be mono- or polysubstitution in each case, at all the hydrogen-carrying carbon and/or nitrogen atoms, independently of one another. Heteroaryl itself as substituent may be linked to the molecule via any suitable position of the ring system, both carbon and nitrogen.


Typical examples are listed below.


Monocyclic Heteroaryls:


furyl; thienyl; pyrrolyl; oxazolyl; thiazolyl; isoxazolyl; isothiazolyl; pyrazolyl; imidazolyl; triazolyl; tetrazolyl; oxadiazolyl; thiadiazolyl; pyridyl; pyrimidyl; pyridazinyl; pyrazinyl; triazinyl; pyridyl-N-oxide; pyrrolyl-N-oxide; pyrimidinyl-N-oxide; pyridazinyl-N-oxide; pyrazinyl-N-oxide; imidazolyl-N-oxide; isoxazolyl-N-oxide; oxazolyl-N-oxide; thiazolyl-N-oxide; oxadiazolyl-N-oxide; thiadiazolyl-N-oxide; triazolyl-N-oxide; tetrazolyl-N-oxide etc.


Polycyclic Heteroaryls:


indolyl; isoindolyl; benzofuryl; benzothienyl; benzoxazolyl; benzothiazolyl; benzisoxazolyl; benzisothiazolyl; benzimidazolyl; indazolyl; isoquinolinyl; quinolinyl; quinoxalinyl; cinnolinyl; phthalazinyl; quinazolinyl; benzotriazinyl; indolizinyl; oxazolopyridyl; imidazopyridyl; naphthyridinyl; indolinyl; isochromanyl; chromanyl; tetrahydroisoquinolinyl; isoindolinyl; isobenzotetrahydrofuryl; isobenzotetrahydrothienyl; isobenzothienyl; benzoxazolyl; pyridopyridyl; benzotetrahydrofuryl; benzotetrahydro-thienyl; purinyl; benzodioxolyl; phenoxazinyl; phenothiazinyl; pteridinyl; benzothiazolyl; imidazopyridyl; imidazothiazolyl; dihydrobenzisoxazinyl; benzisoxazinyl; benzoxazinyl; dihydrobenzisothiazinyl; benzopyranyl; benzothiopyranyl; cumarinyl; isocumarinyl; chromonyl; chromanonyl; tetrahydroquinolinyl; dihydroquinolinyl; dihydroquinolinonyl; dihydroisoquinolinonyl; dihydrocumarinyl; dihydroisocumarinyl; isoindolinonyl; benzodioxanyl; benzoxazolinonyl; quinolinyl-N-oxide; indolyl-N-oxide; indolinyl-N-oxide; isoquinolyl-N-oxide; quinazolinyl-N-oxide; quinoxalinyl-N-oxide; phthalazinyl-N-oxide; indolizinyl-N-oxide; indazolyl-N-oxide; benzothiazolyl-N-oxide; benzimidazolyl-N-oxide; benzo-thiopyranyl-S-oxide and benzothiopyranyl-S,S-dioxide etc.


Heteroarylalkyl denotes the combination of the alkyl and heteroaryl groups defined hereinbefore, in each case in their broadest sense. The alkyl group as substituent is directly linked to the molecule and is in turn substituted by a heteroaryl group. The linking of the alkyl and heteroaryl may be achieved on the alkyl side via any carbon atoms suitable for this purpose and on the heteroaryl side by any carbon or nitrogen atoms suitable for this purpose. The respective sub-groups of alkyl and heteroaryl are also included in the combination of the two groups.


By the term heterocycloalkyl are meant groups which are derived from the cycloalkyl as hereinbefore defined if in the hydrocarbon rings one or more of the groups —CH2— are replaced independently of one another by the groups —O—, —S— or —NH— or one or more of the groups ═CH— are replaced by the group ═N—, while not more than five heteroatoms may be present in total, there must be at least one carbon atom between two oxygen atoms and between two sulphur atoms or between one oxygen and one sulphur atom and the group as a whole must be chemically stable. Heteroatoms may simultaneously be present in all the possible oxidation stages (sulphur→sulphoxide —SO—, sulphone —SO2—; nitrogen→N-oxide). It is immediately apparent from the indirect definition/derivation from cycloalkyl that heterocycloalkyl is made up of the sub-groups monocyclic hetero-rings, bicyclic hetero-rings and spirohetero-rings, while each sub-group can also be further subdivided into saturated and unsaturated (heterocycloalkenyl). The term unsaturated means that in the ring system in question there is at least one double bond, but no aromatic system is formed. In bicyclic hetero-rings two rings are linked such that they have at least two atoms in common. In spirohetero-rings one carbon atom (spiroatom) is shared by two rings. If a heterocycloalkyl is substituted, the substitution may be mono- or polysubstitution in each case, at all the hydrogen-carrying carbon and/or nitrogen atoms, independently of one another. Heterocycloalkyl itself as substituent may be linked to the molecule via any suitable position of the ring system.


Typical examples of individual sub-groups are listed below.


Monocyclic Heterorings (Saturated and Unsaturated):


tetrahydrofuryl; pyrrolidinyl; pyrrolinyl; imidazolidinyl; thiazolidinyl; imidazolinyl; pyrazolidinyl; pyrazolinyl; piperidinyl; piperazinyl; oxiranyl; aziridinyl; azetidinyl; 1,4-dioxanyl; azepanyl; diazepanyl; morpholinyl; thiomorpholinyl; homomorpholinyl; homopiperidinyl; homopiperazinyl; homothiomorpholinyl; thiomorpholinyl-5-oxide; thiomorpholinyl-S,S-dioxide; 1,3-dioxolanyl; tetrahydropyranyl; tetrahydrothiopyranyl; [1,4]-oxazepanyl; tetrahydrothienyl; homothiomorpholinyl-S,S-dioxide; oxazolidinonyl; dihydropyrazolyl; dihydropyrrolyl; dihydropyrazinyl; dihydropyridyl; dihydro-pyrimidinyl; dihydrofuryl; dihydropyranyl; tetrahydrothienyl-5-oxide; tetrahydrothienyl-S,S-dioxide; homothiomorpholinyl-5-oxide; 2,3-dihydroazet; 2H-pyrrolyl; 4H-pyranyl; 1,4-dihydropyridinyl etc.


Bicyclic Heterorings (Saturated and Unsaturated):


8-azabicyclo[3.2.1]octyl; 8-azabicyclo[5.1.0]octyl; 2-oxa-5-azabicyclo[2.2.1]heptyl; 8-oxa-3-aza-bicyclo[3.2.1]octyl; 3,8-diaza-bicyclo[3.2.1]octyl; 2,5-diaza-bicyclo-[2.2.1]heptyl; 1-aza-bicyclo[2.2.2]octyl; 3,8-diaza-bicyclo[3.2.1]octyl; 3,9-diaza-bicyclo[4.2.1]nonyl; 2,6-diaza-bicyclo[3.2.2]nonyl; hexahydro-furo[3,2-b]furyl; etc.


Spiro-Heterorings (Saturated and Unsaturated):


1,4-dioxa-spiro[4.5]decyl; 1-oxa-3,8-diaza-spiro[4.5]decyl; and 2,6-diaza-spiro[3.3]heptyl; 2,7-diaza-spiro[4.4]nonyl; 2,6-diaza-spiro[3.4]octyl; 3,9-diaza-spiro[5.5]undecyl; 2,8-diaza-spiro[4.5]decyl etc.


Heterocycloalkylalkyl denotes the combination of the alkyl and heterocycloalkyl groups defined hereinbefore, in each case in their broadest sense. The alkyl group as substituent is directly linked to the molecule and is in turn substituted by a heterocycloalkyl group. The linking of the alkyl and heterocycloalkyl may be achieved on the alkyl side via any carbon atoms suitable for this purpose and on the heterocycloalkyl side by any carbon or nitrogen atoms suitable for this purpose. The respective sub-groups of alkyl and heterocycloalkyl are also included in the combination of the two groups.


By the term “suitable substituent” is meant a substituent which on the one hand is suitable by virtue of its valency and on the other hand leads to a system which is chemically stable.


By “prodrug” is meant an active substance in the form of its precursor metabolite. A distinction may be made between partly multi-part carrier-prodrug systems and biotransformation systems. The latter contain the active substance in a form that requires chemical or biological metabolisation. The skilled man will be familiar with prodrug systems of this kind (Sloan, Kenneth B.; Wasdo, Scott C. The role of prodrugs in penetration enhancement. Percutaneous Penetration Enhancers (2nd Edition) (2006). 51-64; Lloyd, Andrew W. Prodrugs. Smith and Williams' Introduction to the Principles of Drug Design and Action (4th Edition) (2006), 211-232; Neervannan, Seshadri. Strategies to impact solubility and dissolution rate during drug lead optimization: salt selection and prodrug design approaches. American Pharmaceutical Review (2004), 7(5), 108.110-113). A suitable prodrug contains for example a substance of the general formulae which is linked via an enzymatically cleavable linker (e.g. carbamate, phosphate, N-glycoside or a disulphide group to a dissolution-improving substance (e.g. tetraethyleneglycol, saccharide, amino acids). Carrier-prodrug systems contain the active substance as such, bound to a masking group which can be cleaved by the simplest possible controllable mechanism. The function of masking groups according to the invention in the compounds according to the invention is to neutralise the charge for improving cell uptake. If the compounds according to the invention are used with a masking group, these may also additionally influence other pharmacological parameters, such as for example oral bioavailability, tissue distribution, pharmacokinetics and stability against non-specific phosphatases. The delayed release of the active substance may also involve a sustained-release effect. In addition, modified metabolisation may occur, thus resulting in a higher efficiency of the active substance or organic specificity. In the case of a prodrug formulation, the masking group or a linker that binds the masking group to the active substance is selected such that the prodrug is sufficiently hydrophilic to be dissolved in the blood serum, has sufficient chemical and enzymatic stability to reach the activity site and is also sufficiently hydrophilic to ensure that it is suitable for diffusion-controlled membrane transport. Furthermore, it should allow chemically or ensymatically induced release of the active substance within a reasonable period and, it goes without saying, the auxiliary components released should be non-toxic. Within the scope of the invention, however, the compound without a mask or linker, and a mask, may be regarded as a prodrug which first of all has to be prepared in the cell from the ingested compound by enzymatic and biochemical processes.


Preparation of 6-nitroindolinone



embedded image


Method A—tert. Butyl 2-chloro-4-nitrobenzenecarboxylate (A1)

2-Chloro-5-nitrobenzoic acid (22 g, 109.1 mmol) and DMF (500 μL) are refluxed in toluene (50 mL)/thionyl chloride (8.5 mL) for 1.5 h with stirring. The reaction mixture is evaporated down and the residue is taken up in anhydrous THF (200 mL). Potassium-tert.-butoxide (12.5 g, 111.4 mmol) is added at 0° C., then the cooling is removed and the mixture is stirred for 30 min. The solvent is distilled off and the residue is divided between water and EtOAc. The organic phase is washed with water and 0.1 N NaOH, dried, filtered and evaporated down. Yield: 24 g (85%)


Method B—Dimethyl 2-(2-carboxy-4-nitrophenyl)malonate (A2)

Potassium-tert.-butoxide (50.0 g, 446 mmol) is dissolved at 20° C. in anhydrous DMSO (300 mL), at this temperature dimethyl malonate (67.0 mL, 586 mmol) is added and the mixture is stirred for 20 min. A1 (45.7 g, 177 mmol) is added and the mixture is stirred for 30 min at 100° C. It is poured from water (800 mL), acidified with concentrated HCl (30 mL) and extracted exhaustively with CH2Cl2. The organic phase is washed with water, dried, filtered and evaporated down. The residue is stirred in formic acid (300 mL) for 1.5 h at 72° C. The mixture is evaporated down, the residue is taken up in EtOAc, washed with NaCl solution and exhaustively extracted with dilute NaHCO3 solution. The combined aqueous phase is acidified with concentrated HCl and exhaustively extracted with CH2Cl2. The combined organic phase is washed with water, dried, filtered and evaporated down. Yield: 38.4 g (73%)


Method C—Dimethyl 6-nitro-2-oxo-1,2-dihydroindol-3,3-dicarboxylate (A3)

Triethylamine (9.4 mL, 67.8 mmol) is added to A2 (20 g, 67.3 mmol) and DPPA (14.5 mL, 67.4 mmol) in anhydrous THF (40 mL) and the mixture is stirred for 1.25 h at boiling temperature. The reaction mixture is evaporated down, the residue is taken up in CH2Cl2 and washed with 1 N HCl. The organic phase is combined with ether and the precipitate is filtered off. Yield: 9.89 g (50%)


Method D—6-Nitro-1,3-dihydroindol-2-one (A4)

A3 (5.30 g, 10 mmol) is stirred in MeOH (10 mL)/2 N NaOH (10 mL) for 30 min at 80° C. The reaction mixture is acidified with 1 N HCl, the precipitate is filtered off and stirred in acetic acid (10 mL) for 1 h at boiling temperature. The mixture is cooled to RT, the precipitate is isolated by filtration and digested with water. Yield: 2.18 g (68%)


Synthesis of a Basic Unsubstituted Indolinone Structure



embedded image


Method E—3-(Bismethylsulphanylmethylene)-6-nitro-1,3-dihydro-indol-2-one (A5)

K2CO3 (3.88 g, 28.07 mmol) is added to A4 (1 g, 5.61 mmol) and CS2 (375 μL) in anhydrous DMF (30 mL) at 15° C. and stirred for 1 h. Methyl iodide (775 μL) in anhydrous DMF (5 mL) is added and the mixture is stirred for 6 h at RT. The reaction mixture is stirred into water (150 mL), the precipitate is suction filtered, washed repeatedly with water and digested once with MeCN. Yield: 1.17 g (74%)


Method F—Cyclic Vinylogous Amides/Analogue Compounds to A6

A5 (350 mg, 1.24 mmol) and 2-aminobenzylamine are stirred in n-butanol for 15 min at 150° C. in the microwave. The precipitate formed is filtered off, washed with ether and dried. Yield: 300 mg (78%)
















embedded image


















No.
R5
R5
X
Yield [%]






A6
H
H
NH
78



A7
H
H
O
59



A8
Me
Me
NH
49














No.
X
Yield [%]






A9
N—iPr
quant.



A10
NH
88



A11
O
90












embedded image
















No.
X
Yield [%]






A9
N—iPr
quant.



A10
NH
88



A11
O
90






A12


embedded image


38











embedded image


Analogously to Method E, starting from 5-methoxycarbonyl-1,3-dihydroindol-2-one, the corresponding compound 3-(bismethylsulphanylmethylene)-5-methoxycarbonyl-1,3-dihydro-indol-2-one (A13) is obtained, which is reacted analogously to Method F to form the corresponding cyclic vinylogous amides.
















embedded image

















No.
R5/R5
X
Yield [%]






A14
H/H
NH
99



A15
Me/Me
NH
87



A16
Me/Me
NH
99









Preparation of the Aniline Components



embedded image


Method G—5-(4-Methylpiperazin-1-yl)-2-nitrobenzonitrile (A17)

2-Fluoro-5-nitrobenzonitrile (6.60 g, 39.7 mmol), N-methylpiperazine (10.62 mL) and triethylamine (16.5 mL) are stirred in 2-propanol (50 mL) for 16 h at RT and for 3 h at reflux temperature. The product is obtained by filtration, digested with water, 2-propanol and ether and dried. Yield: 8.90 g (91%)
















embedded image














No.
R
Yield [%]





A17


embedded image


 91





A18


embedded image


 90





A19


embedded image


100









Method H—Reduction of the Nitro Group (A20)

A17 (500 mg, 2.03 mmol), 4-methyl-1-cyclohexene (1.22 mL) and Pd/C (100 mg) are stirred in THF (5 mL)/MeOH (5 mL) for 25 min. at 80° C. in the microwave. The catalyst is filtered off and the filtrate is evaporated down. Yield: 420 mg (96%)
















embedded image














No.
R
Yield [%]





A20


embedded image


 96





A21


embedded image


 81





A22


embedded image


100









Method I—Reduction to Form the Benzylamine

A20 (420 mg, 1.94 mmol) and Raney nickel (100 mg) are hydrogenated in methanolic ammonia (2 mol/L, 60 mL) at RT and 3 bar hydrogen pressure. The catalyst is filtered off and the filtrate is evaporated down. Yield: 343 mg (80%)
















embedded image














No.
R
Yield [%]





A23


embedded image


 80





A24


embedded image


quant.





A25


embedded image


100









Method J—5-(4-Methylpiperazin-1-yl)-2-nitrobenzaldehyde (A26)

2-Fluoro-5-nitrobenzaldehyde (2.50 g, 15 mmol), N-methylpiperazine (1.8 mL) and potassium carbonate (3.10 g, 22 mmol) are stirred in anhydrous DMSO (20 mL) for 2 h at 70° C. The reaction mixture is mixed with water, the product is obtained by filtration, digested with water and dried. Yield: 3.41 g (93%)














embedded image
















No.
R
Yield [%]






A26


embedded image


93






A27


embedded image


98






A28


embedded image


96






A29


embedded image


66











embedded image


Method K—Reductive Amination (A30)

A26 (1.50 g, 6.02 mmol) and methylamine (70% in water, 0.8 mL) are stirred in methanol for 15 min at RT. NaBH4 (175 mL 4.63 mmol) is added and the mixture is stirred for 15 min at RT. The reaction mixture is used in the next step without any further working up.


Method L—Reduction of the Nitro Group (A31)

The crude product obtained from A30 is stirred with palladium on activated charcoal (100 mg) in MeOH (5 mL)/DCM (20 mL) for 2.5 h at RT at a hydrogen pressure of 5 bar. The catalyst is filtered off and the filtrate is evaporated down. The residue is dissolved in DCM, washed with water and dilute potassium carbonate solution, dried, filtered and evaporated down. Yield: 1.01 g (72%).














embedded image















No.
Rx
Ry
Yield [%]













A31


embedded image




embedded image


72





A32


embedded image




embedded image


93





A33


embedded image




embedded image


95





A34


embedded image




embedded image


87





A35


embedded image




embedded image


96





A36


embedded image




embedded image


99





A37


embedded image




embedded image


92





A38


embedded image




embedded image


73





A39
H


embedded image


78





A40
H


embedded image


93





A41
H
Me
100


A42
H
Et
83





A43


embedded image


Me
71











embedded image


Method M—2-Amino-5-formylbenzenzonitrile (A44)

2-Fluoro-5-formylbenzonitrile (500 mg, 3.53 mmol) in THF (7.5 mL) and conc. aqueous ammonia (2.6 mL) are stirred for 900 s at 100° C. in the microwave. The reaction solution is evaporated down, the residue is combined with 1 N HCl, the precipitate is filtered off, washed with water until neutral and dried in vacuo. Yield: 390 mg (80%)


Method N—Reductive amination (A45)

NaBH(OAc)3 (2.32 g 10.9 mmol) and glacial acetic acid (0.31 mL) are added to A44 (800 mg, 5.47 mmol) and pyrrolidine (2.26 mL, 27.4 mmol) in anhydrous CH2Cl2 (20 mL) and stirred for 2 h at RT. Concentrated K2CO3 solution is added and the organic phase is extracted exhaustively with 1 N HCl. The combined aqueous phase is made basic with K2CO3 and exhaustively extracted with EtOAc. The combined organic phase is washed with NaCl solution, dried, filtered and evaporated down. Yield: 935 mg (85%).


Method O—Reduction to Form the Benzylamine (A46)

A45 (878 mg, 4.36 mmol) in anhydrous THF (10 mL) is added to a solution of LiAlH4 (1 M in THF, 12 mL) within 30 min at 0° C. and stirred for 16 h at RT. Water (1 mL) in THF (1 mL) is added to the mixture at 0° C., it is diluted with THF (20 mL) and 1 N NaOH (25 mL). The organic phase is separated off and the aqueous phase is exhaustively extracted with THF. The combined organic phase is dried, filtered and evaporated down. Yield: 676 mg (75%).




embedded image


Method R—Cleaving of the tert.-butyl ester (A47)

Tert-.butyl 3-formyl-4-nitrobenzoate (2 g, 7.96 mmol) is stirred in 4 N HCl in dioxane (20 mL) for 3 h at 55° C. and for 12 h at RT. The reaction mixture is evaporated down. Yield: 1.59 g (88%)


Method S—Amide Formation (A48 and A49)

A47 (1.59 g, 7.01 mmol) in anhydrous toluene (30 mL) is combined with thionyl chloride (350 μL) and DMF (100 μL) and stirred for 2 h at boiling temperature. The reaction mixture is evaporated down and the residue is taken up in anhydrous THF (10 mL). 2 M Me2NH in THF (7.7 mL) is added at 0° C. and stirred for 1 h at RT. The reaction mixture is evaporated down and the residue is divided between EtOAc and 1 N HCl. The organic phase is washed with saturated K2CO3 solution and NaCl solution, dried, filtered and evaporated down Yield 930 mg (54%)














embedded image
















No.
Rx
Yield [%]






A49


embedded image


54









The reductive amination with subsequent reduction of the nitro group is carried out according to Method K and Method L.














embedded image

















No.
Ry
Rx
Yield [%]






A50


embedded image




embedded image


98









The reduction to the benzylamine is carried out according to Method O.














embedded image

















No.
Ry
Rx
Yield [%]






A51


embedded image




embedded image


80











embedded image


Method P—5-(4-Methylpiperazin-1-yl)-2-nitrobenzoic acid (A52)

Methyl 5-chloro-2-nitrobenzoate acid (1 g, 4.50 mmol) and N-methylpiperazine (3 mL, 27 mmol) are stirred in the microwave for 30 min at 150° C. The reaction mixture is stirred in water (50 mL), the pH is adjusted to 5 with NH4Cl, and the mixture is washed with EtOAc. The aqueous phase is freeze-dried, the residue is stirred with THF (200 mL) and filtered. The filtrate is evaporated down and the residue is digested with MeCN. Yield: 434 mg (36%).


Method Q—Reduction to Form the benzylalcohol (A53)

LiAlH4 (248 mg, 38 mmol) is added at 0° C. to A52 (434 mg, 1.64 mmol) in anhydrous ether (20 mL) and stirred for 48 h at RT. EtOAc (10 mL) is added and the mixture is stirred for 1 h at RT. The precipitate is filtered off and extracted with THF, the combined filtrates are evaporated down. Yield: 257 mg (71%).


Synthesis of Substituted Indolinones



embedded image


The cyclic vinylogous amides are prepared according to Method F using isopropanol as solvent.














embedded image


















Yield


No.
R1
X
[%]













A54


embedded image


O
79





A55


embedded image


O
12





A56


embedded image


N
28





A57


embedded image


N
96





A58


embedded image




embedded image


55





A59


embedded image




embedded image


81





A60


embedded image




embedded image


quant.





A61


embedded image




embedded image


quant.





A62


embedded image




embedded image


92





A63


embedded image




embedded image


quant.





A64


embedded image




embedded image


quant.





A65


embedded image


H
75





A66


embedded image




embedded image


73





A67
H


embedded image


77





A68
H


embedded image


81





A69


embedded image




embedded image


48











embedded image














embedded image

















No.
R1
X
Yield [%]















A70


embedded image


N
100






A71


embedded image


N
99






A72
NH2
N
48






A73


embedded image


N
68






A74


embedded image


N—Me
72






A75


embedded image


N—Me
85






A76
H
N—Me
100



A77
H
N—Et
89



A78
H
N—iPr
87











embedded image


Reaction with Sulphonyl Chlorides (A80)

A79 (469 mg, 1.5 mmol) is dissolved in THF (50 mL) and combined with triethylamine (650 μL, 4.5 mmol) and methylsulphonyl chloride (350 μL, 4.5 mmol) and stirred for 3 days at 50° C. After elimination of the solvent the crude product is taken up in methanol and purified by preparative HPLC-MS. Yield: 162 mg (27%)




embedded image


Method T—Cleaving of the boc Protective Group (A82)

A81 (886 mg, 1.62 mmol) is stirred in CH2Cl2 (10 mL)/TFA (2.55 mL) for 1 h at ambient temperature. The reaction solution is diluted with CH2Cl2 and neutralised with K2CO3. The mixture is diluted with water and extracted exhaustively with EtOAc. The combined organic phases are dried, filtered and evaporated down. Yield: 700 mg (97%)


Method U—Reductive Amination (A83)

A82 (723 mg, 1.62 mmol) in CH2Cl2 (25 mL)/MeOH (5 mL) and 37% formaldehyde in water (5 mL) are stirred for 1 h at RT. NaBH(OAc)3 (1.06 g, 5.02 mmol) is added batchwise at 0° C., then the mixture is stirred for 3 h at RT. The reaction solution is divided between CH2Cl2 and saturated K2CO3 solution, the organic phase is washed with saturated K2CO3 solution, dried, filtered and evaporated down. Yield: 750 mg (quant.)
















No.
Structure
Educt
Method
Yield [%]







A84


embedded image




embedded image


U
quant.









The reduction of the nitro group to form the aniline is carried out using the General Working Method specified under Method L.


Method W—General Working Method for Amide Formation

Triethylamine (3 equiv.) and TBTU (1.2 equiv.) are added to a solution of the carboxylic acid (1 equiv.) in anhydrous DMSO (10-20 μL/mg amino compound) and shaken for 5 min at RT. The amino compound is added and the mixture is shaken for 30 min at RT. The reaction solution is filtered and purified by preparative HPLC.


Aniline Components:


The following components are prepared analogously to methods G, H and I. The reduction of the cyano group may be carried out both with Raney nickel as described in Method I and also with borane/THF complex.




embedded image


A88 (340 mg) is dissolved in 50 mL THF and mixed with borane/THF complex (4.3 mL, 1 mmol/mL). This reaction mixture is refluxed for 16 h. The reaction solution is combined with methanol and the solvent is eliminated in vacuo. The crude mixture is purified by preparative HPLC.














embedded image

















No.
R1
R1′
R1′






A87


embedded image


H
H






A88


embedded image


H
H






A89


embedded image


F
H









The following components are prepared analogously to methods J, K and I. For components wherein Ry′ does not denote H, 5-chloro-2-nitro-acetophenone is used as the starting material.














embedded image


















Nr.
Rx
Rx′
Ry
Ry′






A90


embedded image


H
Me
H






A91
H
H
Me
H






A92


embedded image


H
Me
H






A93


embedded image


F
Me
H






A94


embedded image


H
Me
Me






A95


embedded image


H
H
Me






A96
H
H
Me
Me






A97


embedded image


H
H
Me






A98


embedded image


H
Me
Me






A99


embedded image


H
Me
H






A100


embedded image


H
H
H









The following components are prepared analogously to methods M, N and O:














embedded image















No.
Rx






A101


embedded image








A102


embedded image













embedded image


Method Z

A100 (1 g) is dissolved in 25 mL ethanol and combined with sodium borohydride (166 mg). This reaction mixture is stirred for 16 h at 20° C. Then 25 mL saturated sodium hydrogen carbonate solution are added to the reaction solution, it is stirred for 1 h and the organic constituents are eliminated in vacuo. The aqueous phase is extracted with diethyl ether, the organic phase is dried and the solvent is eliminated in vacuo. The following components are prepared analogously to methods J, Z and I. For components wherein Ry′ is H, 5-fluoro-2-nitro-benzaldehyde is used as starting material.














embedded image














No.
Rx
Ry′





A107


embedded image


Me





A108


embedded image


H





A109


embedded image


H











embedded image


Method AA

A105 (500 mg), 1-methyl-(4-methylamino)piperidine (440 μL) and Hünig base (600 μL) are suspended in 1.5 mL butanol and stirred for 1 h at 150° C. in a Biotage microwave. The reaction mixture is combined with water and DCM, the organic phase is separated off and dried.


The following components are prepared analogously to methods AA, Y and I.














embedded image















No.
Rx






A114


embedded image








A115


embedded image








A116


embedded image








A117


embedded image













embedded image


Method AB: A119

NaOMe (0.185 g) is added to A118 (0.3 g) and CS2 (111 μL) in anhydrous DMSO (10 mL) at 15° C. and the mixture is stirred for 1 h. Ethylbromide (106 μL) is added and the mixture is stirred for 14 h at RT. The reaction mixture is stirred into saturated NH4Cl solution (20 mL) and extracted with CH2Cl2. The organic phase is washed with saturated NaCl solution (20 mL), dried and concentrated by rotary evaporation.


Method AC: A120

2-Aminobenzylalcohol (98.13 mg) is added to A119 (0.21 g) in 1 mL isopropanol and the mixture is stirred for 4 h at 100° C. The reaction mixture is stirred into water (20 mL) and extracted with CH2Cl2. The organic phase is washed with saturated NaCl solution (20 mL), dried and freed from the solvent in vacuo.


The following products are obtained analogously to methods AB and AC from the correspondingly substituted indolinones and the suitably configured 2-aminobenzyl-alcohols (e.g. prepared analogously to Method P/Q, J/Z/I or according to other general methods known to the skilled man).














embedded image

















No.
R5/R5
R1
X2






A121
H/H


embedded image


NO2






A122
H/H


embedded image


NO2






A123
H/H


embedded image


COOMe






A124
H/H


embedded image


COOMe






A125
CH3/H


embedded image


COOMe






A126
H/H
H
COOMe











embedded image


The following components are prepared analogously to Method G, Y and L.














embedded image
















No.
R5/R5
R1






A131
H/H


embedded image








A132
H/H


embedded image








A133
H/H
F






A134
H/H


embedded image








A135
H/H


embedded image








A136


embedded image




embedded image













embedded image


A137 is reacted analogously to Method Y to form A138.


Examples 1-51













embedded image

























tret

UVmax



Ex.
Rc
R1
R5/R5
X
[min]
[M + H]+
[nm]
HPLC


















 1


embedded image


H
H/H
NH
2.05
409
278
A





 2


embedded image


H
H/H
NH
1.97
424
348
A





 3


embedded image


H
H/H
NH
1.98
423
361
A





 4


embedded image


H
H/H
O
1.79
388
364
A





 5


embedded image


H
Me/Me
NH
2.12
451
366
A





 6


embedded image




embedded image


H/H
NH
1.48
521
360
A





 7


embedded image




embedded image


H/H
N—Me
0.12
499
311
A





 8


embedded image




embedded image


H/H
N—Me
0.12
535
307
A





 9


embedded image




embedded image


H/H
NH
1.52
535
367
A





10


embedded image




embedded image


H/H
NH
1.34
499
364
A





11


embedded image




embedded image


H/H
N—Et
1.23
513
312
A





12


embedded image




embedded image


H/H
N—Et
1.47
535
291
A





13


embedded image




embedded image


H/H
N—Et
1.47
549
306
A





14


embedded image




embedded image


H/H
N—Et
0.12
499
309
A





15


embedded image




embedded image


H/H
NH
1.52
506
365
A





16


embedded image




embedded image


H/H
N—iPr
0.13
527
312
A





17


embedded image




embedded image


H/H
N—iPr
1.55
549
289
A





18


embedded image




embedded image


H/H
N—iPr
1.52
563
305
A





19


embedded image




embedded image


H/H
N—iPr
0.12
513
376
A





20


embedded image




embedded image


H/H
O
1.94
522
382
B





21


embedded image




embedded image


H/H
O
2.51
562
391
B





22


embedded image




embedded image


H/H
N—iPr
1.69
564
383
B





23


embedded image




embedded image


H/H
N—Me
1.54
536
377
B





24


embedded image




embedded image


H/H
N—Et
1.61
550
381
B





25


embedded image




embedded image


H/H


embedded image


1.80
590
386
B





26


embedded image




embedded image


H/H


embedded image


2.07
589
384
B





27


embedded image




embedded image


H/H


embedded image


1.88
553
380
B





28


embedded image




embedded image


H/H


embedded image


2.14
575
387
B





29


embedded image




embedded image


H/H
NH
1.68
507
367
B





30


embedded image




embedded image


H/H
N—iPr
2.131
579
383
B





31


embedded image




embedded image


H/H
N—iPr
2.07
563
384
B





32


embedded image




embedded image


H/H
N—iPr
2.06
593
385
B





33


embedded image




embedded image


H/H
N—iPr
2.10
569
380
B





34


embedded image




embedded image


H/H
N—iPr
1.74
564
380
B





35


embedded image




embedded image


H/H
N—iPr
2.17
580
391
B





36


embedded image




embedded image


H/H
N—iPr
1.93
563
378
B





37


embedded image




embedded image


H/H
N—iPr
1.96
542
377
B





38


embedded image




embedded image


H/H
N—iPr
1.85
569
377
B





39


embedded image




embedded image


H/H
N—iPr
1.89
571
379
B





40


embedded image




embedded image


H/H
N—iPr
2.01
564
367
B





41


embedded image




embedded image


H/H


embedded image


1.72
578
385
B





42


embedded image




embedded image


H/H


embedded image


1.84
541
385
B





43


embedded image




embedded image


H/H


embedded image


2.04
577
385
B





44


embedded image




embedded image


H/H


embedded image


2.08
563
385
B





45


embedded image




embedded image


H/H
N—iPr
1.76
523
384
B





46


embedded image




embedded image


H/H
N—iPr
2.01
522
383
B





47


embedded image


H
H/H


embedded image


2.01
506
281
B





48


embedded image


H
H/H


embedded image


1.77
484
357
B





49


embedded image


H
H/H


embedded image


1.95
520
381
B





50


embedded image


H
H/H


embedded image


1.66
521
381
B





51


embedded image


H
H/H
N—iPr
1.98
429
374
B









Examples 52-54













embedded image





















tret

UVmax



Ex
Rc
X
[min]
[M + H]+
[nm]
HPLC





52


embedded image


N—iPr
1.80
451
311
A





53


embedded image


N—iPr
1.49
401
282
A





54


embedded image


N—iPr
1.55
415
282
A











embedded image


Method X—General Working Method for the Saponification of Esters

A suspension of the ester in methanol (20-50 μL/mg ester) is combined with six equivalents of 1N NaOH and stirred for 5-24 h at 50° C. After cooling the reaction mixture is added to 1N HCL and the precipitate formed is filtered, washed and dried.


Examples 55-218













embedded image

























tret

UVmax



Ex
Rc
R1
R5/R5
X
[min]
[M + H]+
[nm]
HPLC


















 55


embedded image




embedded image


H/H
N
1.57
448

D





 56


embedded image




embedded image


H/H
N
1.48
483
346
C





 57


embedded image




embedded image


H/H
N
1.44
483
346
C





 58


embedded image




embedded image


H/H
N
1.5
497
346
C





 59


embedded image




embedded image


H/H
N
1.71
472

B1





 60


embedded image




embedded image


H/H
N
1.47
483
346
C





 61


embedded image




embedded image


H/H
N
1.49
464
346
C





 62


embedded image




embedded image


H/H
N
1.47
450
346
C





 63


embedded image




embedded image


H/H
N
1.6
472
346
C





 64


embedded image




embedded image


H/H
N
1.58
497

B1





 65


embedded image




embedded image


H/H
N
1.43
497
346
C





 66


embedded image




embedded image


H/H
N
1.48
497
346
C





 67


embedded image




embedded image


H/H
N
1.46
497
346
C





 68


embedded image




embedded image


H/H
N
1.43
419

D





 69


embedded image




embedded image


H/H
N
1.54
461

D





 70


embedded image




embedded image


H/H
N
1.63
501

D





 71


embedded image




embedded image


H/H
N
1.64
487
350
C





 72


embedded image




embedded image


H/H
N
1.54
485
346
C





 73


embedded image




embedded image


H/H
N
1.58
496
346
B1





 74


embedded image




embedded image


H/H
N
1.55
496
346
B1





 75


embedded image




embedded image


H/H
N
1.52
463
346
B1





 76


embedded image




embedded image


H/H
N
1.5
496
346
B1





 77


embedded image




embedded image


H/H
N
1.55
477
346
B1





 78


embedded image




embedded image


H/H
N
1.45
485
346
D





 79


embedded image




embedded image


H/H
N
1.47
510
346
C





 80


embedded image




embedded image


H/H
N
1.49
511
346
C





 81


embedded image




embedded image


H/H
N
1.51
510
346
C





 82


embedded image




embedded image


H/H
N—Me
1.57
497

B1





 83


embedded image




embedded image


H/H
N—Me
1.47
462

B1





 84


embedded image




embedded image


H/H
N—Me
1.51
497

B1





 85


embedded image




embedded image


H/H
N—Me
1.53
497

B1





 86


embedded image




embedded image


H/H
N—Me
1.33
406

B1





 87


embedded image




embedded image


H/H
N—Me
1.57
511

B1





 88


embedded image




embedded image


H/H
N—Me
1.58
511

B1





 89


embedded image




embedded image


H/H
N—Me
1.58
511

B1





 90


embedded image




embedded image


H/H
N—Me
1.6
511

B1





 91


embedded image




embedded image


H/H
N—Me
1.68
486

B1





 92


embedded image




embedded image


H/H
N—Me
1.55
478

B1





 93


embedded image




embedded image


H/H
N—Me
1.49
464

B1





 94


embedded image




embedded image


H/H
N—Me
1.65
511

B1





 95


embedded image




embedded image


H/H
N—Me
1.35
419

B1





 96


embedded image




embedded image


H/H
N—Me
1.64
524

B1





 97


embedded image


H
H/H
N—Et
1.65
426

B1





 98


embedded image


H
H/H
N—Et
1.86
391

B1





 99


embedded image


H
H/H
N—Et
1.57
426

B1





100


embedded image


H
H/H
N—Et
1.60
426

B1





101


embedded image


H
H/H
N—Et
1.36
335

B1





102


embedded image


H
H/H
N
1.56
321

A





103


embedded image


H
H/H
N
1.68
335

A





104


embedded image


H
H/H
N
1.79
349

A





105


embedded image


H
H/H
N
1.89
363

A





106


embedded image


H
H/H
N
1.68
335

A





107


embedded image


H
H/H
N
2.01
377

A





108


embedded image


H
H/H
N
2.09
403

A





109


embedded image


H
H/H
N
1.79
349

A





110


embedded image


H
H/H
N
1.41
404

D





111


embedded image


H
H/H
N
1.88
401

A





112


embedded image


H
H/H
N
1.49
360
338
C





113


embedded image


H
H/H
N
1.53
415
342
C





114


embedded image


H
H/H
N
1.52
398
342
C





115


embedded image


H
H/H
N
1.5
398
338
C





116


embedded image


H
H/H
N
1.65
400
342
C





117


embedded image


H
H/H
N
1.40
413
338
C





118


embedded image


H
H/H
N
1.54
412
342
C





119


embedded image


H
H/H
N
1.53
412
342
C





120


embedded image


H
H/H
N
1.55
412
338
C





121


embedded image


H
H/H
N
1.71
389
342
C





122


embedded image


H
H/H
N
1.40
378
338
C





123


embedded image


H
H/H
N
1.43
365
340
C





124


embedded image


H
H/H
N
1.49
401
342
C





125


embedded image


H
H/H
N
1.46
387
338
C





126


embedded image


H
H/H
N
1.47
415
338
C





127


embedded image


H
H/H
N
1.62
387
342
C





128


embedded image


H
H/H
N
1.61
387
342
C





129


embedded image


H
H/H
N
1.49
398
338
C





130


embedded image


H
H/H
N
1.49
365
340
C





131


embedded image


H
H/H
N
1.37
392
338
D





132


embedded image


H
H/H
N
1.46
432
338
D





133


embedded image


H
H/H
N
1.52
379
342
C





134


embedded image


H
H/H
N
1.50
391
338
C





135


embedded image


H
H/H
N
1.53
391
342
C





136


embedded image


H
H/H
N
1.51
346
338
C





137


embedded image


H
H/H
N
1.73
377
342
C





138


embedded image


H
H/H
N
1.61
437
342
C





139


embedded image


H
H/H
N
1.70
401
342
C





140


embedded image


H
H/H
N
1.96
440

B1





141


embedded image


H
H/H
N
1.84
426

B1





142


embedded image


H
H/H
N
1.70
413

B1





143


embedded image


H
H/H
N
1.94
441

B1





144


embedded image


H
H/H
N
1.89
397

B1





145


embedded image


H
H/H
N
1.57
374
342
C





146


embedded image


H
H/H
N
1.85
515
342
C





147


embedded image


H
H/H
N
1.51
412
338
C





148


embedded image


H
H/H
N
1.51
412
338
C





149


embedded image


H
H/H
N
1.60
432
342
C





150


embedded image


H
H/H
N
1.51
412
338
C





151


embedded image


H
H/H
N
1.46
413
338
C





152


embedded image


H
H/H
N
1.58
426
342
C





153


embedded image


H
H/H
N
1.60
412
338
C





154


embedded image


H
H/H
N
1.55
412
340
C





155


embedded image


H
H/H
N
1.52
413
338
C





156


embedded image


H
H/H
N
1.47
413
338
C





157


embedded image


H
H/H
N
1.52
412
342
C





158


embedded image


H
H/H
N
1.47
399
342
C





159


embedded image


H
H/H
N
1.60
412
338
C





160


embedded image


H
H/H
N
2.04
425

B1





161


embedded image


H
H/H
N
1.98
411

B1





162


embedded image


H
H/H
N
1.99
411

B1





163


embedded image


H
H/H
N
1.95
427

B1





164


embedded image


H
H/H
N
1.97
411

B1





165


embedded image


H
H/H
N
2.00
431

B1





166


embedded image


H
H/H
N
2.02
431

B1





167


embedded image


H
H/H
N
2.02
431

B1





168


embedded image


H
H/H
N
1.70
416

B1





169


embedded image


H
H/H
N
1.92
414

B1





170


embedded image


H
H/H
N
1.73
393

B1





171


embedded image


H
H/H
N
1.86
413

B1





172


embedded image


H
H/H
N
1.68
379

B1





173


embedded image


H
H/H
N
1.66
413

B1





174


embedded image


H
H/H
N
1.67
379

B1





175


embedded image


H
H/H
N
1.91
411

B1





176


embedded image


H
H/H
N
1.69
393

B1





177


embedded image


H
H/H
N
1.64
426

B1





178


embedded image


H
H/H
N
1.56
432
342
C





179


embedded image


H
H/H
N
1.97
422

B1





180


embedded image


H
H/H
N
1.90
446

B1





181


embedded image


H
H/H
N
1.76
430

B1





182


embedded image


H
H/H
N
1.79
426

B1





183


embedded image


H
H/H
N
2.07
465

B1





184


embedded image


H
H/H
N
2.08
445

B1





185


embedded image


H
H/H
N
1.96
411

B1





186


embedded image


H
H/H
N
1.65
412

B1





187


embedded image


H
H/H
N
1.71
441

C





188


embedded image


H
H/H
N
1.82
471

B1





189


embedded image


H
H/H
N
1.98
441

B1





190


embedded image


H
H/H
N
1.63
412

B1





191


embedded image


H
H/H
N
1.64
412

B1





192


embedded image


H
H/H
N—iPr
1.74
440

B1





193


embedded image


H
H/H
N—iPr
1.94
405

B1





194


embedded image


H
H/H
N—iPr
1.66
440

B1





195


embedded image


H
H/H
N—iPr
1.67
440

B1





196


embedded image


H
H/H
N—iPr
1.50
349

B1





197


embedded image


H
H/H
N—Me
1.74
377

B1





198


embedded image


H
H/H
N—Me
1.53
412

B1





199


embedded image


H
H/H
N—Me
1.48
412

B1





200


embedded image


H
H/H
N—Me
1.67
401

B1





201


embedded image


H
H/H
N—Me
1.46
379

B1





202


embedded image


H
H/H
N—Me
1.58
426

B1





203


embedded image


H
Me/H
N
1.65
412

B1





204


embedded image


H
Me/H
N
1.66
406

B1





205


embedded image


H
Me/H
N
1.70
412

B1





206


embedded image


H
Me/H
N
1.66
412

B1





207


embedded image


H
Me/H
N
1.70
392

B1





208


embedded image


H
Me/H
N
1.66
335

B1





209


embedded image


H
Me/H
N
2.17
417

B1





210


embedded image


H
Me/H
N
1.97
377

B1





211


embedded image


H
Me/Me
N
1.96
391

B1





121


embedded image


H
Me/Me
N
1.56
335

B1





213


embedded image


H
Me/Me
N
1.78
426

B1





214


embedded image


H
Me/Me
N
1.69
426

B1





215


embedded image


H
Me/Me
N
1.73
426

B1





216


embedded image


H
Me/Me
N
1.82
440

B1





217


embedded image




embedded image


H/H
N
1.36
414

B1





218


embedded image




embedded image


H/H
N
1.43
477

B1











embedded image


A 85 (110 mg, 0.24 mmol) is dissolved in CH2Cl2 (7.5 mL), combined with TFA (3.8 mL) and stirred for 2 h at RT. After evaporation of the solvent and the TFA the free acid is reacted according to Method W with N,N-dimethylethylene-diamine (53 μL, 0.48 mmol) to obtain the desired compound 2-[5-butylcarbamoyl-2-oxo-1,2-dihydro-indol-(3Z)-ylidene]-1,2,3,4-tetrahydro-quinazoline-6-carboxylic acid (2-dimethylamino-ethyl)-amide. Yield: 10 mg.




embedded image


2-Oxo-2,3-dihydro-1H-indole-5-carboxylic acid (7 g, 39 mmol) is dissolved together with benzylalcohol (4.2 mL, 39 mmol) and 4-N,N-dimethylaminopyridine (1 g, 7.9 mmol) in 70 mL DCM and cooled in the ice bath. Then 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide (8.3 g, 43 mmol) is added and the mixture is heated to RT for 12 h. After evaporation of the solvent the residue is taken up in a little methanol and combined with 1 N hydrochloric acid (200 mL). The precipitate formed is filtered and dried. Yield: 6.9 g (65%) (A86).


The benzyl ester thus obtained (1.3 g, 4.7 mmol) is dissolved together with [methyl-(2-nitro-phenyl)-amino]-acetic acid (1.1 g, 5.2 mmol)—for synthesis see Eur. J. Org. Chem. 2003, 12, 2314-2326—and diisopropylethylamine (4.1 mL, 24 mmol) in dimethylformamide (10 mL), then combined with 2-(1H-7-azabenzotriazol-1-yl)-1,1,3,3-tetramethyl-uronium hexafluorophosphate (HATU, 2.1 g, 5.7 mmol) and stirred for 12 h at RT. After the addition of 10 mL ammonia in methanol the mixture is stirred for a further 6 h at RT, then mixed with water and extracted with DCM. The organic phase is dried and evaporated down. After purification by chromatography, the desired product benzyl 3-{2-[methyl-(2-nitrophenyl)-amino]-acetyl}-2-oxo-2,3-dihydro-1H-indole-5-carboxylate is obtained. Yield: 220 mg (10%) (A87).


A87 (190 mg, 0.41 mmol) is dissolved in methanol (30 mL), combined with palladium on activated charcoal (10%) and stirred for 12 h under hydrogen pressure (4 bar). After filtration and evaporation of the solvent, 3-[4-methyl-3,4-dihydro-1H-quinoxalin-(2Z)-ylidene]-2-oxo-2,3-dihydro-1H-indole-5-carboxylic acid is obtained.


Yield: 109 mg (82%) (A88).




embedded image


A88 (54 mg, 0.17 mmol) is converted into the corresponding butylamide according to Method W.


Yield: 13 mg (20%). [M+1]+: 377; Rt (Method B): 2.05 min; UVmax: 286 and 395 nm


The corresponding anilines can be generated from A121 and A122 using hydrogen (analogously to Method L) and these are reacted analogously to Method W to form the acetamides.














embedded image
















Ex
R1
[M + H]+






221


embedded image


423






222


embedded image


436











embedded image


The following target molecules are prepared analogously to Method F, X and W.




embedded image


Examples 223-360



















tret

HPLC


Ex
RX2
RX1
[min]
[M + H]+
Method




















223


embedded image




embedded image


1.76
475
B1





224


embedded image




embedded image


1.86
486
B1





225


embedded image




embedded image


1.83
462
B1





226


embedded image




embedded image


1.81
503
B1





227


embedded image




embedded image


1.75
489
B1





228


embedded image




embedded image


1.82
420
B1





229


embedded image




embedded image


1.81
475
B1





230


embedded image




embedded image


1.80
462
B1





231


embedded image




embedded image


1.89
364
B1





232


embedded image




embedded image


1.84
391
B1





233


embedded image




embedded image



479
B1





234


embedded image




embedded image


1.84
479
B1





235


embedded image




embedded image


1.95
489
B1





236


embedded image




embedded image


1.87
462
B1





237


embedded image




embedded image


1.94
463
B1





238


embedded image




embedded image


1.87
476
B1





239


embedded image




embedded image


1.79
476
B1





240


embedded image




embedded image


1.75
493
B1





241


embedded image




embedded image


1.89
392
B1





242


embedded image




embedded image


1.93
475
B1





243


embedded image




embedded image


1.74
529
B1





244


embedded image




embedded image


1.80
515
B1





245


embedded image




embedded image


1.77
516
B1





246


embedded image




embedded image


1.79
543
B1





247


embedded image




embedded image


1.85
529
B1





248


embedded image




embedded image


1.84
516
B1





249


embedded image




embedded image


1.87
553
B1





250


embedded image




embedded image


1.86
533
B1





251


embedded image




embedded image


1.67
515
B1





252


embedded image




embedded image


1.74
501
B1





253


embedded image




embedded image



502
B1





254


embedded image




embedded image


1.73
529
B1





255


embedded image




embedded image


1.79
515
B1





256


embedded image




embedded image


1.78
502
B1





257


embedded image




embedded image


1.79
489
B1





258


embedded image




embedded image


1.85
475
B1





259


embedded image




embedded image


1.83
476
B1





260


embedded image




embedded image


1.84
503
B1





261


embedded image




embedded image


1.89
476
B1





262


embedded image




embedded image


1.97
493
B1





263


embedded image




embedded image


1.91
493
B1





264


embedded image




embedded image


2.04
503
B1





265


embedded image




embedded image


2.13
557
B1





266


embedded image




embedded image


2.06
420
B1





267


embedded image




embedded image


1.53
433
B1





268


embedded image




embedded image


1.46
406
B1





269


embedded image




embedded image


1.37
433
B1





270


embedded image




embedded image


1.25
321
B1





271


embedded image




embedded image


1.45
308
B1





272


embedded image




embedded image


1.39
335
B1





273


embedded image




embedded image


1.49
406
B1





274


embedded image




embedded image


1.52
407
B1





275


embedded image




embedded image


1.45
420
B1





276


embedded image




embedded image


1.36
420
B1





277


embedded image




embedded image


1.58
516
B1





278


embedded image




embedded image


1.65
502
B1





279


embedded image




embedded image


1.85
432
B1





280


embedded image




embedded image


1.65
489
B1





281


embedded image




embedded image


1.49
477
B1





282


embedded image




embedded image


1.75
393
B1





283


embedded image




embedded image


1.64
505
B1





284


embedded image




embedded image


1.73
491
B1





285


embedded image




embedded image


1.93
421
B1





286


embedded image




embedded image


1.74
478
B1





287


embedded image




embedded image


1.67
512
B1





288


embedded image




embedded image


1.59
524
B1





289


embedded image




embedded image


1.69
499
B1





290


embedded image




embedded image


1.62
538
B1





291


embedded image




embedded image


1.59
524
B1





292


embedded image




embedded image


1.48
525
B1





293


embedded image




embedded image


1.59
510
B1





294


embedded image




embedded image


1.67
502
B1





295


embedded image




embedded image


1.62
524
B1





296


embedded image




embedded image


1.56
491
B1





297


embedded image




embedded image


1.6
488
B1





298


embedded image




embedded image


1.61
524
B1





299


embedded image




embedded image


1.73
487
B1





300


embedded image




embedded image


1.53
516
B1





301


embedded image




embedded image


1.93
601
B1





302


embedded image




embedded image


1.63
514
B1





303


embedded image




embedded image


1.54
510
B1





304


embedded image




embedded image


1.56
491
B1





305


embedded image




embedded image


1.74
513
B1





306


embedded image




embedded image


1.70
487
B1





307


embedded image




embedded image


1.60
505
B1





308


embedded image




embedded image


1.64
517
B1





309


embedded image




embedded image


1.68
519
B1





310


embedded image




embedded image


1.55
510
B1





311


embedded image




embedded image


1.77
489
B1





312


embedded image




embedded image


1.78
489
B1





313


embedded image




embedded image


1.73
533
B1





314


embedded image




embedded image


1.60
528
B1





315


embedded image




embedded image


1.64
477
B1





316


embedded image




embedded image


1.79
551
B1





317


embedded image




embedded image


1.65
501
B1





318


embedded image




embedded image


1.61
473
B1





319


embedded image




embedded image


1.69
519
B1





320


embedded image




embedded image


1.80
501
B1





321


embedded image




embedded image


1.66
519
B1





322


embedded image




embedded image


1.49
447
B1





323


embedded image




embedded image


1.90
503
B1





324


embedded image




embedded image


1.51
459
B1





325


embedded image




embedded image


1.77
510
B1





326


embedded image




embedded image


1.58
461
B1





327


embedded image




embedded image


1.6
505
B1





328


embedded image




embedded image


1.60
510
B1





329


embedded image




embedded image


1.64
510
B1





330


embedded image




embedded image


1.68
475
B1





331


embedded image




embedded image


1.63
510
B1





332


embedded image




embedded image


1.72
487
B1





333


embedded image




embedded image


1.80
489
B1





334


embedded image




embedded image


1.66
510
B1





335


embedded image




embedded image


1.88
515
B1





336


embedded image




embedded image


1.8
501
B1





337


embedded image




embedded image


1.51
457
B1





338


embedded image




embedded image


1.56
471
B1





339


embedded image




embedded image


1.63
473
B1





340


embedded image




embedded image


1.53
479
B1





341


embedded image




embedded image


1.76
509
B1





342


embedded image




embedded image


1.91
515
B1





343


embedded image




embedded image


2.00
529
B1





344


embedded image




embedded image


1.57
531
B1





345


embedded image




embedded image


1.57
531
B1





346


embedded image




embedded image


1.69
530
B1





347


embedded image




embedded image


2.11
506
B1





348


embedded image




embedded image


2.12
518
B1





349


embedded image




embedded image


1.92
478
B1





350


embedded image




embedded image


2.09
493
B1





351


embedded image




embedded image


1.94
532
B1





352


embedded image




embedded image


1.99
492
B1





353


embedded image




embedded image


1.98
519
B1





354


embedded image




embedded image


1.92
395
B1





355


embedded image




embedded image



516
B1





356


embedded image




embedded image


1.65
503
B1





357


embedded image




embedded image


1.81
502
B1





358


embedded image




embedded image



588
B1





359


embedded image




embedded image


1.93
476
B1





360


embedded image




embedded image


1.83
462
B1











embedded image


Method AD

Under an argon atmosphere, A140 (60 mg), 3-pyridylboric acid (36 mg) and 1,1′-bis(diphenylphosphino)-ferrocene-palladium(II)dichloride (8 mg) are combined with 720 μL of a mixture of THF/NMP (2:1) and with caesium carbonate (75 mg) which has been dissolved in 100 μL water. The reaction mixture is stirred for 1 h at 100° C. in a Biotage microwave. The crude product thus obtained (Example 361) is purified by RP-HPLC.


tret=1.80 min (analytical method B1)


[M+H]+=341 (analytical method B1)




embedded image


Method AE

Under an argon atmosphere, A140 (2 g), bis(pinacolate)diboran (1.63 g), potassium acetate (1.72 g) and 1,1′-bis(diphenylphosphino)-ferrocene-palladium(II)dichloride (475 mg) are combined with 15 mL anhydrous dioxane. The reaction mixture is stirred for 3 h at 80° C. and then combined with water and DCM. The organic phase is separated off, dried and filtered through silica gel. It is washed several times with ethyl acetate. The organic phases are combined and the solvent is eliminated in vacuo. This residue (A141) is reacted analogously to Method AD to form Example 362, using 2-chloropyrazine in this case.


tret=1.74 min (analytical method B1)


[M+H]+=342 (analytical method B1)


By using suitable indolinone derivatives (A139) and the corresponding heteroarylboric acids or haloheteroaromatic compounds the following compounds are prepared analogously to Method F, AD and AE.


Examples 363-430













embedded image






















tret

HPLC


Ex
R2
R3
RX1
[min]
[M + H]+
Method
















363


embedded image


H


embedded image


1.65
453
B1





364


embedded image


H


embedded image


1.69
440
B1





365


embedded image


H


embedded image


1.93
369
B1





366


embedded image


H


embedded image


1.97
460
B1





367


embedded image


H


embedded image


1.86
342
B1





368


embedded image


H


embedded image


1.79
440
B1





369


embedded image


H


embedded image


1.75
234
B1





370


embedded image


H


embedded image


1.82
427
B1





371


embedded image


H


embedded image


1.78
440
B1





372


embedded image


H


embedded image


1.75
347
B1





373


embedded image


H


embedded image


1.99
358
B1





374


embedded image


H


embedded image


1.7
426
B1





375


embedded image


H


embedded image


1.64
440
B1





376


embedded image


H


embedded image


1.82
440
B1





377


embedded image


H


embedded image


1.89
441
B1





378


embedded image


H


embedded image


1.75
453
B1





379


embedded image


H


embedded image


1.86
454
B1





380


embedded image


H


embedded image


1.75
454
B1





381


embedded image


H


embedded image


1.74
467
B1





382


embedded image


H


embedded image


1.78
479
B1





383


embedded image


H


embedded image


1.78
371
B1





384


embedded image


H


embedded image


1.91
359
B1





385


embedded image


H


embedded image


1.84
457
B1





386


embedded image


H


embedded image


1.85
444
B1





387


embedded image


H


embedded image


1.78
471
B1





388


embedded image


H


embedded image


1.81
458
B1





389


embedded image


H


embedded image


1.7
356
B1





390


embedded image


H


embedded image


1.87
371
B1





391


embedded image


H


embedded image


1.62
356
B1





392


embedded image




embedded image




embedded image


1.8
389
B1





393


embedded image




embedded image




embedded image


1.96
377
B1





394


embedded image




embedded image




embedded image


1.85
475
B1





395


embedded image




embedded image




embedded image


1.89
462
B1





396


embedded image




embedded image




embedded image


1.87
359
B1





397


embedded image


H


embedded image


1.72
445
B1





398


embedded image


H


embedded image


1.65
342
B1





399


embedded image


H


embedded image


1.9
347
B1





400


embedded image


H


embedded image


1.74
459
B1





401


embedded image




embedded image




embedded image


1.81
463
B1





402


embedded image


H


embedded image


2.06
375
B1





403


embedded image




embedded image




embedded image


1.83
487
B1





404


embedded image


H


embedded image


2.07
330
B1





405


embedded image


H


embedded image


2.05
330
B1





406


embedded image


H


embedded image


1.88
341
B1





407


embedded image


H


embedded image


1.63
342
B1





408


embedded image


H


embedded image


1.68
342
B1





409


embedded image


H


embedded image


1.67
442
B1





410


embedded image


H


embedded image


1.88
346
B1





411


embedded image


H


embedded image


1.85
347
B1





412


embedded image


H


embedded image


1.86
355
B1





413


embedded image


H


embedded image


1.91
355
B1





414


embedded image


H


embedded image


1.89
355
B1





415


embedded image


H


embedded image


1.28
357
B1





416


embedded image


H


embedded image


1.97
359
B1





417


embedded image


H


embedded image


1.93
359
B1





418


embedded image


H


embedded image


1.82
366
B1





419


embedded image


H


embedded image



366
B1





420


embedded image


H


embedded image


1.96
371
B1





421


embedded image


H


embedded image


2.07
375
B1





422


embedded image


H


embedded image


1.91
375
B1





423


embedded image


H


embedded image


1.91
375
B1





424


embedded image


H


embedded image


1.76
478
B1





425


embedded image


H


embedded image


1.77
478
B1





426


embedded image


H


embedded image


1.56
479
B1





427


embedded image


H


embedded image


1.97
385
B1





428


embedded image


H


embedded image


1.85
385
B1





429


embedded image


H


embedded image


1.99
391
B1





430


embedded image


H


embedded image


2.04
391
B1











embedded image


The following target molecules are prepared by using suitable indolinone derivatives (A142) and the corresponding diamines analogously to Method F.


Examples 431-437













embedded image






















HPLC


Ex
R3
X1
tret [min]
[M + H]+
Method





431


embedded image




embedded image


1.73
420
B1





432


embedded image




embedded image


1.67
392
B1





433


embedded image




embedded image


1.79
407
B1





434


embedded image




embedded image


1.70
387
B1





435


embedded image




embedded image


1.97
430
B1





436


embedded image




embedded image


1.89
440/442
B1





437


embedded image




embedded image


1.89
454/456
B1









The following Examples may be synthesised using Method W.


Examples 438-441













embedded image





















tret

UVmax



Ex
Rc
R1
[min]
[M + H]+
[nm]
HPLC





438


embedded image




embedded image


1.97
549
218
B1





439


embedded image




embedded image


1.86
553
298
B1





440


embedded image




embedded image


1.88
571
296
B1





441


embedded image




embedded image


1.88
571
298
B1









Method AF

Similarly to Method W, 1 equiv. of the amino compound is placed in a suitable solvent (15 mL/mmol, e.g. DCM, DMF), but approx. 1.5 equiv. of an isocyanate corresponding to the desired Rc and 3 equiv. triethylamine are added. After the reaction has ended the volatile constituents are eliminated in vacuo and the product is isolated by preparative HPLC.


Examples 442-448













embedded image





















tret

UVmax



Ex.
Rc
R1
[min]
[M + H]+
[nm]
HPLC





442


embedded image




embedded image


1.94
538
234; 308
B1





443


embedded image




embedded image


1.88
574
222; 350
B1





444


embedded image




embedded image


1.82
504
218; 345
B1





445


embedded image




embedded image


1.75
528

B1





446


embedded image




embedded image


1.84
542

B1





447


embedded image




embedded image


1.84
528
250
B1





448


embedded image




embedded image


1.79
540
255
B1









Examples 449-459
Synthesis of the Examples wherein R3′═R3″═Br or R3″═NO2; R3′═NH2



embedded image


Method AG—General Procedure

A 1,5-difluoro-2-nitrobenzene (1 g) substituted according to the desired R3″ is dissolved in approx. 5 mL THF or dioxane. Depending on the desired R2, 1 equiv. of the corresponding sulphinate salt or of the corresponding amine or alcohol with DIPEA (1.1 equiv.) are added and the mixture is stirred at 50-70° C. After the first substitution has ended 1 equiv. tert.-butylethylmalonate, 3 equiv. sodium hydride and optionally NMP (2 mL) and 1 equiv. Potassium carbonate are added and the mixture is again stirred at 50-60° C. After the reaction has ended the reaction mixture is neutralised with 1 N HCl, the volatile constituents are eliminated in vacuo and the product is isolated by preparative HPLC. The sequence of the first and second substitution may optionally be reversed.


Method AH—General Procedure

The compound obtained from Method AE (1 g) is dissolved in THF (100 mL), Raney nickel (100 mg) is added and the mixture is hydrogenated under an H2 atmosphere (3-5 bar). Once the reaction has ended the reaction mixture is filtered and the filtrate is freed in vacuo from the volatile constituents. The crude product thus obtained is further used directly.


Method AI—General Procedure

The compound obtained from Method AH (1 g) is mixed with 8-15 mL of a suitable solvent (e.g. THF, toluene, dioxane). After the addition of 2-5 mL TFA it is heated to 80-100° C. After the reaction has ended the reaction mixture is combined with DCM (50 mL) and made basic with aqueous NaHCO3 solution. The organic phase is separated off and the aqueous phase is repeatedly extracted with DCM. The combined organic phases are dried, filtered and freed from the volatile constituents in vacuo. The crude product obtained is purified chromatographically by preparative HPLC.


Examples 451-455 may be synthesised according to Method W, E and F from the substances wherein R3′═NH2 obtained using Method AI:




embedded image


Examples 449-450 may be synthesised according to Method E and F from the substances wherein R3′═Br obtained using Method AG. From these, the Examples 456-459 may be prepared using Method AJ or AK:




embedded image


Method AJ (Sonogashira Reaction)

Unter an argon atmosphere the substance with R3′═Br obtained from Method F (20 mg) is combined with 2 equiv. of the alkyne substituted according to R3, 0.4 equiv. CuI, 0.1 equiv. Bis(triphenylphosphineo)palladium dichloride and 5 equiv. triethylamine with 150 μL DMF and the mixture is stirred for 5 min at RT and then at 100° C. During this time a further 8 equiv. of the alkyne are added batchwise and the mixture is stirred until the desired product has formed. Then the cooled reaction mixture is freed from the volatile constituents in vacuo and the product is isolated chromatographically by preparative HPLC.


Method AK (Suzuki Coupling)

Unter an argon atmosphere the substance with R3′═Br obtained from Method F (50 mg) is combined with 3.2 equiv. KOAc, 2.2 equiv. Bis(pinacolato)diboron or bis(neopentyl-glycolato)diboron and 0.1 equiv. 1,1′-bis(diphenylphosphino)ferrocene palladium dichloride and 1.5 ml of 1,4-dioxane. The reaction mixture is heated to 85° C. for 1-2 h, cooled to RT again and further used directly. For this, 0.5 mL methanol, 4 equiv. K2CO3, 0.1-0.2 equiv. 1,1′-bis(diphenylphosphino)ferrocene palladium dichloride and 2 equiv. of a corresponding aryl or heteroaryl halide are added, depending on the desired R3. The reaction mixture is heated to 85° C. again until the desired product has formed. Then the cooled reaction mixture is freed in vacuo from the volatile constituents and the product is isolated chromatographically by preparative HPLC.


Examples 449-459













embedded image























tret

UVmax



Ex
R2
R3
R1
[min]
[M + H]+
[nm]
HPLC





449


embedded image




embedded image




embedded image


1.97

262; 354
B1





450


embedded image




embedded image




embedded image


1.89

354
B1





451


embedded image




embedded image




embedded image


2.05
675
380
B1





452


embedded image




embedded image




embedded image


2.15
640
295
B1





453


embedded image




embedded image




embedded image


1.97
620

B1





454


embedded image




embedded image




embedded image


2.24
607
298
B1





455


embedded image




embedded image




embedded image


2.01
536
290
B1





456


embedded image




embedded image




embedded image


1.83
552
222; 366
B1





457


embedded image




embedded image




embedded image


2.40
564
366
B1





458


embedded image




embedded image




embedded image


2.24
584/586
366
B1





459


embedded image




embedded image




embedded image


2.11
653
374
E*





*






HPLC Method E corresponds to HPLC Method D, except that: column=Agilent Zorbax SBC8, 2.1×50 mm, 3.5μ; solvent: A=water with 0.2% formic acid added, B=acetonitrile with 0.2% formic acid added; gradient: 5% B, within 1.5 min to 95% B, then 0.5 min isocratically at 100% B and 1 min isocratically at 5% B, flow 1.2 mL/min.


Abbreviations Used














equiv.
equivalent(s)








DCM
dichloromethane



DIPEA
N,N-diisopropylethylamine



DMF
N,N-dimethylformamide



DMSO
dimethylsulphoxide



Et
ethyl



EtOAc
ethyl acetate



h
hour



HCl
hydrochloric acid



HPLC
high performance liquid chromatography



conc.
concentrated



HMDS
hexamethyldisilazane



iPr
isopropyl



Me
methyl



MeOH
methanol



min
minute



mL
millilitre



MS
mass spectrometry



N
Normal



NMP
1-methyl-2-pyrrolidone



NMR
Nuclear Magnetic Resonance spectroscopy



ppm
part per million



RP
reversed phase



RT
room temperature



TFA
trifluoroacetic acid



TBTU
O-benztriazol-1-yl-N,N,N′,N′-tetramethyluronium




tetrafluoroborate



tert
tertiary



THF
tetrahydrofuran









HPLC-Methods

Method A


HPLC: Agilent 1100 Series


MS: Agilent LC/MSD SL (LCMS1: 1100 series LC/MSD)


Column: Waters, Xterra MS C18, 2.5 μm, 2.1×30 mm, Part. No. 186000592


Solvent: A: H2O (Millipore purified purest water) with 0.1% HCOOH


B: acetonitrile (HPLC grade)


Detection: MS: Positive and negative

    • Mass range: 120-900 m/z
    • Fragmentor: 120
    • Gain EMV: 1
    • Threshold: 150
    • Stepsize: 0.25
    • UV: 254 nm
    • Bandwide: 1 (LCMS1: 2)
    • Reference: off


Spectrum: Range: 250-400 nm

    • Range step: 1.00 nm
    • Threshold: 4.00 mAU
    • Peakwidth: <0.01 min (LCMS1: >0.05 min)
    • Slit: 1 nm (LCMS1: 2 nm)


Injection: Inj. Vol.: 5 μL


Inj. Mode: Needle wash


Separation: Flow: 1.10 mL/min

    • Column temp.: 40° C.
    • Gradient: 0 min 5% solvent B
      • 0-2.5 min 5%->95% solvent B
      • 2.50-2.80 min 95% solvent B
      • 2.81-3.10 min 95%->5% solvent B


Method B

HPLC: Agilent 1100 Series


MS: 1100 Series LC/MSD (API-ES +/−3000V, Quadrupol, G1946D)


MSD Signal Settings: Scan pos 120-900, Scan neg 120-900


Column: phenomenex; Part No. 00M-4439-BO-CE; Gemini 3μ C18 110 Å; 20·2.0 mm


Eluant: A: 5 mM NH4HCO3/20 mM NH3 (pH=9.5)

    • B: acetonitrile HPLC grade


Detection: Signal: UV 254 nm (bandwide 1, reference off)

    • Spectrum: range: 250-400 nm; step: 1 nm
    • Peak width <0.01 min (0.1 s)


Injection: 10 μL standard injection


Method: B1





    • Flow: 1.0 mL/min

    • Column temperature: 40° C.






















Pump gradient:
0.0-2.5 min
 5% -> 95% solvent B




2.5-2.8 min
95% solvent B




2.8-3.1 min
95% -> 5% solvent B










Method C

HPLC: Agilent 1100 Series


MS: 1100 Series LC/MSD (Quadrupol, G1946D; API-ES)


MSD Signal Settings: Capillary Voltage +/−3000V

    • Scan pos 100-1200, Scan neg 100-1200


Column: Waters X Bridge; C18; 3.5 μm; 2.1·50 mm


Eluant: A: 5 mM NH4HCO3/20 mM NH3 (pH=9.5)

    • B: acetonitrile (HPLC grade)


Detection:


Signal: UV 254 nm (Bandwide 8, Reference off)

    • UV 230 nm (Bandwide 8, Reference off)


Spectrum: range: 190-450 nm; step: 4 nm; Threshold 1 mAU


Peakbreite: >0.03 min


Injection: 2 μL standard injection


Flow: 1.2 mL/min


Column oven.: 35° C.



















Gradient:
0.01 min
 5% B




1.25 min
95% B




2.00 min
95% B




2.01 min
 5% B




Stop time: 3.00 min










Method D

HPLC: Agilent 1100 Series


MS: LC/MSD SL (Quadrupol, G1956B; MM−ES+APCI)


MSD Signal Settings: Capillary Voltage +/−2000V

    • Corona Current [μA] +/−8
    • Charging Voltage +/−2000
    • Scan pos 105-1200, Scan neg 105-1200


column: Waters X Bridge; C18; 3.5 μm; 2.1·50 mm


eluant: A: 5 mM NH4HCO3/20 mM NH3 (pH=9.5)

    • B: acetonitrile HPLC grade


Detection:


Signal: UV 254 nm (Bandwide 4, Reference off)

    • UV 230 nm (Bandwide 4, Reference off)


Spectrum: range: 190-450 nm; step: 4 nm; Threshold 1 mAU


Peak width: >0.03 min


Injection: 2 μL standard injection


Flow: 1.2 mL/min


Column oven.: 35° C.



















Gradient:
0.01 min
 5% B




1.25 min
95% B




2.00 min
95% B




2.01 min
 5% B




Stop time: 3.00 min










The following Examples describe the biological activity of the compounds according to the invention without restricting the invention to these Examples.


As demonstrated by DNA staining followed by FACS or Cellomics Array Scan analysis, the inhibition of proliferation brought about by the majority of the compounds according to the invention is mediated above all by errors in chromosome segregation and/or cytokinesis. For this reason, massive polyploidia occurs in these cases, which may finally lead to inhibition of proliferation or even apoptosis. Moreover, many of the compounds according to the invention cause an inhibition of proliferation, by interfering with intracellular signal transduction pathways which are important to the survival of cells-primarily, but not exclusively, in cells which have become dependent on such signal pathways during their development Inhibiting these pathways in corresponding cells induces arrest in the G1 phase of the cell cycle and/or apoptosis.


On the basis of their biological properties the compounds of general formula (I) according to the invention, their isomers and the physiologically acceptable salts thereof are suitable for treating diseases characterised by excessive or abnormal cell proliferation or by the activation of survival-mediating signal pathways.


Example Aurora-B Kinase Assay

A radioactive enzyme inhibition assay was developed using E. coli-expressed recombinant Xenopus laevis Aurora B wild-type protein equipped at the N-terminal position with a GST tag (amino acids 60-361) in a complex with Xenopus laevis INCENP (amino acids 790-847), which is obtained from bacteria and purified. In equivalent manner a Xenopus laevis Aurora B mutant (G96V) in a complex with Xenopus laevis INCENP790-847 may also be used.


Expression and Purification


The coding sequence for Aurora-B60-361 from Xenopus laevis is cloned into a modified version of pGEX-6T (Amersham Biotech) via BamHI and SalI cutting sites. The vector contains two cloning cassettes which are separated by a ribosomal binding site, allowing bi-cistronic expression. In this configuration Xenopus laevis Aurora B is expressed by the first cassette, and the Xenopus laevis INCENP790-847 is expressed by the second cassette. The resulting vector is pAUB-IN847.


First of all the E. coli strain BL21 (DE3) is co-transformed with pUBS520 helper plasmid and pAUB-IN847, after which protein expression is induced using 0.3 mM IPTG at an OD600 of 0.45-0.7. The expression is then continued for approx. 12-16 h at 23-25° C. with agitation.


The bacteria are then removed by centrifuging and the pellet is lysed in lysis buffer (50 mM Tris/C1 pH 7.6, 300 mM NaCl, 1 mM DTT, 1 mM EDTA, 5% glycerol, Roche Complete Protease Inhibitor tablets) using ultrasound, using 20-30 mL lysis buffer per liter of E. coli culture. The lysed material is freed from debris by centrifugation (12000 rpm, 45-60 min, JA20 rotor). The supernatant is incubated with 300 μL of equilibrated GST Sepharose Fast Flow (Amersham Biosciences) per liter of E. coli culture for 4-5 h at 4° C. Then the column material is washed with 30 volumes of lysis buffer and then equilibrated with 30 volumes of cleavage buffer (50 mM Tris/C1 pH 7.6, 150 mM NaCl, 1 mM DTT, 1 mM EDTA). To cleave the GST tag from Aurora B, 10 units of Precision Protease (Amersham Biosciences) are used per milligram of substrate and the mixture is incubated for 16 h at 4° C. The supernatant which contains the cleavage product is loaded onto a 6 mL Resource Q column (Amersham Biosciences) equilibrated with ion exchange buffer (50 mM Tris/C1 pH 7.6, 150 mM NaCl, 1 mM DTT, 1 mM EDTA). The Aurora B/INCENP complex is caught as it flows through, then concentrated and loaded onto a Superdex 200 size exclusion chromatography (SEC) column equilibrated with SEC buffer (10 mM Tris/C1 pH 7.6, 150 mM NaCl, 1 mM DTT, 1 mM EDTA). Fractions which contain the AuroraB/INCENP complex are collected and concentrated using Vivaspin concentrators (molecular weight exclusion 3000-5000 Da) to a final concentration of 12 mg/mL. Aliquots (e.g. 240 ng/μL) for kinase assays are transferred from this stock solution into freezing buffer (50 mM Tris/Cl pH 8.0, 150 mM NaCl, 0.1 mM EDTA, 0.03% Brij-35, 10% glycerol, 1 mM DTT) and stored at −80° C.


Aurora B Kinase Assay

Test substances are placed in a polypropylene dish (96 wells, Greiner #655 201), in order to cover a concentration frame of 10 μM-0.0001 μM. The final concentration of DMSO in the assay is 5%. 30 μL of protein mix (50 mM tris/C1 pH 7.5, 25 mM MgCl2, 25 mM NaCl, 167 μM ATP, 10 ng Xenopus laevis Aurora B/INCENP complex in freezing buffer) are pipetted into the 10 μL of test substance provided in 25% DMSO and this is incubated for 15 min at RT. Then 10 μL of peptide mix (100 mM tris/C1 pH 7.5, 50 mM MgCl2, 50 mM NaCl, 5 μM NaCl, 5 μM DTT, 1 μCi gamma-P33-ATP [Amersham], 50 μM substrate peptide [biotin-EPLERRLSLVPDS or multimers thereof, or biotin-EPLERRL SLVPKM or multimers thereof, or biotin-LRRWSLGLRRWSLGLRRWSLGLRRWSLG]) are added. The reaction is incubated for 75 min (ambient temperature) and stopped by the addition of 180 μL of 6.4% trichloroacetic acid and incubated for 20 min on ice. A multiscreen filtration plate (Millipore, MAIP NOB10) is equilibrated first of all with 100 μL 70% ethanol and then with 180 μL trichloroacetic acid and the liquids are eliminated using a suitable suction apparatus. Then the stopped kinase reaction is applied. After 5 washing steps with 180 μL 1% trichloroacetic acid in each case the lower half of the dish is dried (10-20 min at 55° C.) and 25 μL scintillation cocktail (Microscint, Packard #6013611) is added. Incorporated gamma-phosphate is quantified using a Wallac 1450 Microbeta Liquid Scintillation Counter. Samples without test substance or without substrate peptide are used as controls. IC50 values are obtained using Graph Pad Prism software.


The effect of the compounds according to the invention on kinases that inhibit signal transduction pathways, e.g. on Serine-Threonine Kinase PDK1, is determined in in vitro kinase assays with recombinantly produced protein. In this assay, the compounds exhibit good to very good efficacy, i.e. an IC50 value of for example less than 1 μmmol/L, generally less than 0.1 μmol/L.


Example of PDK1 Kinase Assay

Recombinant human PDK1 enzyme (aa 52-556) linked at its N-terminal end to His6 is isolated from baculovirus-infected insect cells. Purified enzyme may be obtained for example from the University of Dundee, Scotland. The following components are combined in a well of a 96-well round-based dish (Messrs. Greiner bio-one, No. 650101):

    • 7.5 μL of compound to be tested in varying concentrations (e.g. Starting at 10 μM, and diluted 1:5) in 3.33% DMSO (final concentration 1% DMSO)/assay buffer (50 mM Tris pH 7.5, 0.05% β-mercaptoethanol, 10 mM Mg-acetate)
    • 7.5 μL PDK1 (10 ng/well) and PDKtide (KTFCGTPEYLAPEVRREPRILSEEEQEMF RDFDYIADWC, synthesised by Pepceuticals Limited, Nottingham, United Kingdom; 25 μM final concentration) PDK1 and PDKtide are together diluted accordingly in assay buffer; PDKtide is present in this mixture as an 83.3 μM solution.
    • 10 μL ATP solution (25 μM ATP with 0.5 μCi/well gamma-P33-ATP)


The reaction is started by adding the ATP solution and the mixture is incubated for 30 min at ambient temperature; at the start of the reaction the dishes are shaken gently. The reaction is stopped by the addition of 5 μL/well 0.5 M phosphoric acid (H3PO4) and incubated for about 20 min at ambient temperature. The precipitate is transferred by harvesting onto filter plates (96-well microtitre filter plate: UniFilter GF/C; Messrs Perkin Elmer; No. 6005174), then washed 6 times with 50 mM H3PO4 and dried at 60° C. Then the plate is stuck down with sealing tape, 25 μL/well of scintillation solution (Microscint 0; Messrs. Perkin Elmer; No. 6013611) are added and the amount of P33 precipitated is measured using the Wallac Betacounter. The measured data are evaluated using Graphpad Prism software.


The antiproliferative activity of the compounds according to the invention is determined on cultivated human tumour cells, for example on NCI-H460 or PC-3 cells. The compounds exhibit good to very good activity, i.e. For example an EC50 value in the PC-3 proliferation test of less than 5 μmol/L, generally less than 1 μmol/L.


Measurement of the Inhibition of Proliferation on Cultivated Human Tumour Cells

To measure proliferation on cultivated human tumour cells, cells of lung tumour cell line NCI-H460 (obtained from American Type Culture Collection (ATCC)) are cultivated in RPMI 1640 medium (Gibco) and 10% foetal calf serum (Gibco) and harvested in the log growth phase. Then the NCI-H460 cells are placed in 96-well flat-bottomed plates (Falcon) at a density of 1000 cells per well in RPMI 1640 medium and incubated overnight in an incubator (at 37° C. and 5% CO2). The active substances are added to the cells in various concentrations (dissolved in DMSO; DMSO final concentration: 0.1%). After 72 h incubation 20 μL AlamarBlue reagent (AccuMed International) is added to each well, and the cells are incubated for a further 5-7 h. After incubation the colour change of the AlamarBlue reagent is determined in a Wallac Microbeta fluorescence spectrophotometer. EC50 values are calculated using Standard Levenburg Marquard algorithms (GraphPadPrizm).


To measure proliferation on the prostate carcinoma tumour cell line PC-3 (obtained from American Type Culture Collection (ATCC)) the cells are cultivated in Ham's F12K (Gibco) and 10% foetal calf serum (Gibco) and harvested in the log growth phase. Then the PC-3 cells are placed in 96-well plates (Costar) at a density of 2000 cells per well and incubated overnight in an incubator (at 37° C. and 5% CO2), while on each plate 16 wells are used as controls (8 wells with cells to which only DMSO solution has been added (should yield 30-50% maximum value of reduced AlamarBlue), 4 wells containing only medium (medium control, after the addition of oxidised AlamarBlue reagent the back-ground signal is obtained) and 4 wells where again only medium is added (after the addition of reduced AlamarBlue reagent it acts as a maximum value)). The active substances are added to the cells in various concentrations (dissolved in DMSO; DMSO final concentration: 0.2%) (in each case as a double or triple measurement). After 5 days incubation 20 μl AlamarBlue reagent (Serotec) are added to each well, and the cells are incubated for a further 5-7 h. As a control, 20 μL reduced AlamarBlue reagent is added to each of 4 wells (AlamarBlue reagent which is autoclaved for 30 min). After incubation the colour change of the AlamarBlue reagent in the individual wells is determined in a SpectraMax Photometer (Molecular Devices) (extinction 530 nm, emission 590 nm, 5 sec measuring time). The amount of AlamarBlue reagent reacted represents the metabolic activity of the cells. The relative cell activity is calculated in relation to the control (PC-3 cells without inhibitor) and the active substance concentration which inhibits the cell activity by 50% (EC50) is derived. The values are calculated from the average of two or three individual measurements.


Cell cycle analyses are carried out for example using FACS analyses (Fluorescence Activated Cell Sorter) or by Cellomics Array Scan (CellCycle Analysis).


FACS Analysis

Propidium iodide (PI) binds stoichiometrically to double-stranded DNA, and is thus suitable for determining the proportion of cells in the G1, S, and G2/M phase of the cell cycle on the basis of the cellular DNA content. Cells in the G0 and G1 phase have a diploid DNA content (2N), whereas cells in the G2 or mitosis phase have a 4N DNA content.


For PI staining, for example, 1.75×106 NCI-H460 cells are seeded onto a 75 cm2 cell culture flask, and after 24 h either 0.1% DMSO is added as control or the substance is added in various concentrations (in 0.1% DMSO). The cells are incubated for 42 h with the substance or with DMSO. Then the cells are detached with trypsin and centrifuged. The cell pellet is washed with buffered saline solution (PBS) and the cells are then fixed with 80% ethanol at −20° C. for at least 2 h. After another washing step with PBS the cells are permeabilised with Triton X-100 (Sigma; 0.25% in PBS) on ice for 5 min, and then incubated with a solution of PI (Sigma; 10 μg/ml) and RNAse (Serva; 1 mg/mL) in the ratio 9:1 for at least 20 min in the dark.


The DNA measurement is carried out in a Becton Dickinson FACS Analyzer, with an argon laser (500 mW, emission 488 nm); data are obtained and evaluated using the DNA Cell Quest Programme (BD).


Cellomics Array Scan

NCI-H460 cells are seeded into 96-well flat-bottomed dishes (Falcon) in RPMI 1640 medium (Gibco) with 10% foetal calf serum (Gibco) in a density of 2000 cells per well and incubated overnight in an incubator (at 37° C. and 5% CO2). Analogously thereto, PC-3 cells are cultivated in Ham's F12K (Gibco) and 10% foetal calves' serum (Gibco) and harvested in the log growth phase. Then the PC-3 cells are placed in 96-well dishes (FALCON black/clear bottom (#353948)) at a density of 3000 cells per well and incubated overnight in an incubator (at 37° C. and 5% CO2). The active substances are added to the cells in various concentrations (dissolved in DMSO; DMSO final concentration: 0.1%). After 42 h incubation the medium is suction filtered, the cells are fixed for 10 min with 4% formaldehyde solution and Triton X-100 (1:200 in PBS) at ambient temperature and simultaneously permeabilised, and then washed twice with a 0.3% BSA solution (Calbiochem). Then the DNA is stained by the addition of 50 μL/well of 4′,6-diamidino-2-phenylindole (DAPI; Molecular Probes) in a final concentration of 300 nM for 1 h at ambient temperature, in the dark. Alternatively, 50 μL/well of Hoechst 33342 (Invitrogen) in PBS may be used for the DNA staining (1 h at RT, final concentration: 5 μg/mL). The preparations are then carefully washed twice with PBS, the dishes are stuck down with black adhesive film and analysed in the Cellomics ArrayScan using the CellCycle BioApplication programme and visualised and evaluated using Spotfire.


The substances of the present invention are Aurora B and/or PDK1 kinase inhibitors. On the basis of their biological properties the compounds of general formula (I) according to the invention, their isomers and the physiologically acceptable salts thereof are suitable for treating diseases characterised by excessive or abnormal cell proliferation or by aberrant activation of the phosphatidylinositol-3-kinase (PI3K)-PDK1-AKT signal pathway.


Such diseases include for example: viral infections (e.g. HIV and Kaposi's sarcoma); inflammatory and autoimmune diseases (e.g. colitis, arthritis, Alzheimer's disease, glomerulonephritis and wound healing); bacterial, fungal and/or parasitic infections; leukaemias, lymphomas and solid tumours (e.g. carcinomas and sarcomas), skin diseases (e.g. psoriasis); diseases based on hyperplasia which are characterised by an increase in the number of cells (e.g. fibroblasts, hepatocytes, bones and bone marrow cells, cartilage or smooth muscle cells or epithelial cells (e.g. endometrial hyperplasia)); bone diseases and cardiovascular diseases (e.g. restenosis and hypertrophy).


For example, the following cancers may be treated with compounds according to the invention, without being restricted thereto: brain tumours such as for example acoustic neurinoma, astrocytomas such as pilocytic astrocytomas, fibrillary astrocytoma, protoplasmic astrocytoma, gemistocytary astrocytoma, anaplastic astrocytoma and glioblastoma, brain lymphomas, brain metastases, hypophyseal tumour such as prolactinoma, HGH (human growth hormone) producing tumour and ACTH producing tumour (adrenocorticotropic hormone), craniopharyngiomas, medulloblastomas, meningeomas and oligodendrogliomas; nerve tumours (neoplasms) such as for example tumours of the vegetative nervous system such as neuroblastoma sympathicum, ganglioneuroma, paraganglioma (pheochromocytoma, chromaffinoma) and glomus-caroticum tumour, tumours on the peripheral nervous system such as amputation neuroma, neurofibroma, neurinoma (neurilemmoma, Schwannoma) and malignant Schwannoma, as well as tumours of the central nervous system such as brain and bone marrow tumours; intestinal cancer such as for example carcinoma of the rectum, colon, anus, small intestine and duodenum; eyelid tumours such as basalioma or basal cell carcinoma; pancreatic cancer or carcinoma of the pancreas; bladder cancer or carcinoma of the bladder; lung cancer (bronchial carcinoma) such as for example small-cell bronchial carcinomas (oat cell carcinomas) and non-small cell bronchial carcinomas such as plate epithelial carcinomas, adenocarcinomas and large-cell bronchial carcinomas; breast cancer such as for example mammary carcinoma such as infiltrating ductal carcinoma, colloid carcinoma, lobular invasive carcinoma, tubular carcinoma, adenocystic carcinoma and papillary carcinoma; non-Hodgkin's lymphomas (NHL) such as for example Burkitt's lymphoma, low-malignancy non-Hodgkin's lymphomas (NHL) and mucosis fungoides; uterine cancer or endometrial carcinoma or corpus carcinoma; CUP syndrome (Cancer of Unknown Primary); ovarian cancer or ovarian carcinoma such as mucinous, endometrial or serous cancer; gall bladder cancer; bile duct cancer such as for example Klatskin tumour; testicular cancer such as for example seminomas and non-seminomas; lymphoma (lymphosarcoma) such as for example malignant lymphoma, Hodgkin's disease, non-Hodgkin's lymphomas (NHL) such as chronic lymphatic leukaemia, leukaemic reticuloendotheliosis, immunocytoma, plasmocytoma (multiple myeloma), immunoblastoma, Burkitt's lymphoma, T-zone mycosis fungoides, large-cell anaplastic lymphoblastoma and lymphoblastoma; laryngeal cancer such as for example tumours of the vocal cords, supraglottal, glottal and subglottal laryngeal tumours; bone cancer such as for example osteochondroma, chondroma, chondroblastoma, chondromyxoid fibroma, osteoma, osteoid osteoma, osteoblastoma, eosinophilic granuloma, giant cell tumour, chondrosarcoma, osteosarcoma, Ewing's sarcoma, reticulo-sarcoma, plasmocytoma, giant cell tumour, fibrous dysplasia, juvenile bone cysts and aneurysmatic bone cysts; head and neck tumours such as for example tumours of the lips, tongue, floor of the mouth, oral cavity, gums, palate, salivary glands, throat, nasal cavity, paranasal sinuses, larynx and middle ear; liver cancer such as for example liver cell carcinoma or hepatocellular carcinoma (HCC); leukaemias, such as for example acute leukaemias such as acute lymphatic/lymphoblastic leukaemia (ALL), acute myeloid leukaemia (AML); chronic leukaemias such as chronic lymphatic leukaemia (CLL), chronic myeloid leukaemia (CML); stomach cancer or gastric carcinoma such as for example papillary, tubular and mucinous adenocarcinoma, signet ring cell carcinoma, adenosquamous carcinoma, small-cell carcinoma and undifferentiated carcinoma; melanomas such as for example superficially spreading, nodular, lentigo-maligna and acral-lentiginous melanoma; renal cancer such as for example kidney cell carcinoma or hypernephroma or Grawitz's tumour; oesophageal cancer or carcinoma of the oesophagus; penile cancer; prostate cancer; throat cancer or carcinomas of the pharynx such as for example nasopharynx carcinomas, oropharynx carcinomas and hypopharynx carcinomas; retinoblastoma; vaginal cancer or vaginal carcinoma; plate epithelial carcinomas, adenocarcinomas, in situ carcinomas, malignant melanomas and sarcomas; thyroid carcinomas such as for example papillary, follicular and medullary thyroid carcinoma, as well as anaplastic carcinomas; spinalioma, epidormoid carcinoma and plate epithelial carcinoma of the skin; thymomas, cancer of the urethra and cancer of the vulva.


Apart from pathologically/morphologically manifested types of tumour, these compounds according to the invention may also be used to treat “biomarker-defined” cell anomalies or those characterised by other molecular-biological and/or genetic methods, primarily, but not exclusively, those which are characterised by changes stable over time (stable over at least 3 cell divisions), in the amount or state of activation of genes or their gene products such as the phosphatidylinositol-3-kinase family, PDK1, AKT/PK family, RAS family, tyrosine-kinase receptors, cytokine receptors, chemokine receptors.


The new compounds may be used for the prevention, short-term or long-term treatment of the above-mentioned diseases, optionally also in combination with radiotherapy or other “state-of-the-art” compounds, such as e.g. cytostatic or cytotoxic substances, cell proliferation inhibitors, anti-angiogenic substances, steroids or antibodies.


The compounds of general formula (1) may be used on their own or in combination with other active substances according to the invention, optionally also in combination with other pharmacologically active substances.


Chemotherapeutic agents which may be administered in combination with the compounds according to the invention, include, without being restricted thereto, hormones, hormone analogues and antihormones (e.g. tamoxifen, toremifene, raloxifene, fulvestrant, megestrol acetate, flutamide, nilutamide, bicalutamide, aminoglutethimide, cyproterone acetate, finasteride, buserelin acetate, fludrocortinsone, fluoxymesterone, medroxyprogesterone, octreotide), aromatase inhibitors (e.g. anastrozole, letrozole, liarozole, vorozole, exemestane, atamestane), LHRH agonists and antagonists (e.g. goserelin acetate, luprolide), inhibitors of growth factors (growth factors such as for example “platelet derived growth factor” and “hepatocyte growth factor”, inhibitors are for example “growth factor” antibodies, “growth factor receptor” antibodies and tyrosinekinase inhibitors, such as for example gefitinib, imatinib, lapatinib and trastuzumab); antimetabolites (e.g. Antifolates such as methotrexate, raltitrexed, pyrimidine analogues such as 5-fluorouracil, capecitabin and gemcitabin, purine and adenosine analogues such as mercaptopurine, thioguanine, cladribine and pentostatin, cytarabine, fludarabine); antitumour antibiotics (e.g. anthracyclins such as doxorubicin, daunorubicin, epirubicin and idarubicin, mitomycin-C, bleomycin, dactinomycin, plicamycin, streptozocin); platinum derivatives (e.g. cisplatin, oxaliplatin, carboplatin); alkylation agents (e.g. estramustin, meclorethamine, melphalan, chlorambucil, busulphan, dacarbazin, cyclophosphamide, ifosfamide, temozolomide, nitrosoureas such as for example carmustin and lomustin, thiotepa); antimitotic agents (e.g. Vinca alkaloids such as for example vinblastine, vindesin, vinorelbin and vincristine; and taxanes such as paclitaxel, docetaxel); topoisomerase inhibitors (e.g. epipodophyllotoxins such as for example etoposide and etopophos, teniposide, amsacrin, topotecan, irinotecan, mitoxantron) and various chemotherapeutic agents such as amifostin, anagrelid, clodronat, filgrastin, interferon alpha, leucovorin, rituximab, procarbazine, levamisole, mesna, mitotane, pamidronate and porfimer.


Suitable preparations include for example tablets, capsules, suppositories, solutions,—particularly solutions for injection (s.c., i.v., i.m.) and infusion—elixirs, emulsions or dispersible powders. The content of the pharmaceutically active compound(s) should be in the range from 0.1 to 90 wt.-%, preferably 0.5 to 50 wt.-% of the composition as a whole, i.e. In amounts which are sufficient to achieve the dosage range specified below. The doses specified may, if necessary, be given several times a day.


Suitable tablets may be obtained, for example, by mixing the active substance(s) with known excipients, for example inert diluents such as calcium carbonate, calcium phosphate or lactose, disintegrants such as corn starch or alginic acid, binders such as starch or gelatine, lubricants such as magnesium stearate or talc and/or agents for delaying release, such as carboxymethyl cellulose, cellulose acetate phthalate, or polyvinyl acetate. The tablets may also comprise several layers.


Coated tablets may be prepared accordingly by coating cores produced analogously to the tablets with substances normally used for tablet coatings, for example collidone or shellac, gum arabic, talc, titanium dioxide or sugar. To achieve delayed release or prevent incompatibilities the core may also consist of a number of layers. Similarly the tablet coating may consist of a number of layers to achieve delayed release, possibly using the excipients mentioned above for the tablets.


Syrups or elixirs containing the active substances or combinations thereof according to the invention may additionally contain a sweetener such as saccharine, cyclamate, glycerol or sugar and a flavour enhancer, e.g. a flavouring such as vanillin or orange extract. They may also contain suspension adjuvants or thickeners such as sodium carboxymethyl cellulose, wetting agents such as, for example, condensation products of fatty alcohols with ethylene oxide, or preservatives such as p-hydroxybenzoates.


Solutions for injection and infusion are prepared in the usual way, e.g. With the addition of isotonic agents, preservatives such as p-hydroxybenzoates, or stabilisers such as alkali metal salts of ethylenediamine tetraacetic acid, optionally using emulsifiers and/or dispersants, whilst if water is used as the diluent, for example, organic solvents may optionally be used as solvating agents or dissolving aids, and transferred into injection vials or ampoules or infusion bottles.


Capsules containing one or more active substances or combinations of active substances may for example be prepared by mixing the active substances with inert carriers such as lactose or sorbitol and packing them into gelatine capsules.


Suitable suppositories may be made for example by mixing with carriers provided for this purpose, such as neutral fats or polyethyleneglycol or the derivatives thereof.


Excipients which may be used include, for example, water, pharmaceutically acceptable organic solvents such as paraffins (e.g. petroleum fractions), vegetable oils (e.g. groundnut or sesame oil), mono- or polyfunctional alcohols (e.g. ethanol or glycerol), carriers such as e.g. natural mineral powders (e.g. kaolins, clays, talc, chalk), synthetic mineral powders (e.g. highly dispersed silicic acid and silicates), sugars (e.g. cane sugar, lactose and glucose) emulsifiers (e.g. lignin, spent sulphite liquors, methylcellulose, starch and polyvinylpyrrolidone) and lubricants (e.g. magnesium stearate, talc, stearic acid and sodium lauryl sulphate).


The preparations are administered by the usual methods, preferably by oral or transdermal route, most preferably by oral route. For oral administration the tablets may, of course contain, apart from the abovementioned carriers, additives such as sodium citrate, calcium carbonate and dicalcium phosphate together with various additives such as starch, preferably potato starch, gelatine and the like. Moreover, lubricants such as magnesium stearate, sodium lauryl sulphate and talc may be used at the same time for the tabletting process. In the case of aqueous suspensions the active substances may be combined with various flavour enhancers or colourings in addition to the excipients mentioned above.


For parenteral use, solutions of the active substances with suitable liquid carriers may be used.


The dosage for intravenous use is from 1-1000 mg per hour, preferably between 5 and 500 mg per hour.


However, it may sometimes be necessary to depart from the amounts specified, depending on the body weight, the route of administration, the individual response to the drug, the nature of its formulation and the time or interval over which the drug is administered. Thus, in some cases it may be sufficient to use less than the minimum dose given above, whereas in other cases the upper limit may have to be exceeded. When administering large amounts it may be advisable to divide them up into a number of smaller doses spread over the day.


The formulation examples which follow illustrate the present invention without restricting its scope:


Examples of Pharmaceutical Formulations
















A)
Tablets
per tablet



















active substance according to formula (1)
100 mg



lactose
140 mg



corn starch
240 mg



polyvinylpyrrolidone
 15 mg



magnesium stearate
 5 mg




500 mg










The finely ground active substance, lactose and some of the corn starch are mixed together. The mixture is screened, then moistened with a solution of polyvinylpyrrolidone in water, kneaded, wet-granulated and dried. The granules, the remaining corn starch and the magnesium stearate are screened and mixed together. The mixture is compressed to produce tablets of suitable shape and size.

















B)
Tablets
per tablet



















active substance according to formula (1)
80 mg



lactose
55 mg



corn starch
190 mg 



microcrystalline cellulose
35 mg



polyvinylpyrrolidone
15 mg



sodium-carboxymethyl starch
23 mg



magnesium stearate
 2 mg




400 mg 










The finely ground active substance, some of the corn starch, lactose, microcrystalline cellulose and polyvinylpyrrolidone are mixed together, the mixture is screened and worked with the remaining corn starch and water to form a granulate which is dried and screened. The sodiumcarboxymethyl starch and the magnesium stearate are added and mixed in and the mixture is compressed to form tablets of a suitable size.
















C)
Ampoule solution




















active substance according to formula (1)
50
mg



sodium chloride
50
mg



water for inj.
5
ml










The active substance is dissolved in water at its own pH or optionally at pH 5.5 to 6.5 and sodium chloride is added to make it isotonic. The solution obtained is filtered free from pyrogens and the filtrate is transferred under aseptic conditions into ampoules which are then sterilised and sealed by fusion. The ampoules contain 5 mg, 25 mg and 50 mg of active substance.

Claims
  • 1. A compound of formula (I),
  • 2. The compound according to claim 1, wherein R7 denotes hydrogen or C1-6alkyl.
  • 3. The compound according to claim 1, wherein R4 denotes hydrogen.
  • 4. A pharmaceutical preparation, comprising as active substance one or more compounds of formula (1) according to claim 1 with one or more conventional excipients or carriers.
Priority Claims (1)
Number Date Country Kind
07110049 Jun 2007 EP regional
PCT Information
Filing Document Filing Date Country Kind 371c Date
PCT/EP2008/057150 6/9/2008 WO 00 3/18/2010
Publishing Document Publishing Date Country Kind
WO2008/152014 12/18/2008 WO A
Foreign Referenced Citations (2)
Number Date Country
02074742 Sep 2002 WO
03084951 Oct 2003 WO
Related Publications (1)
Number Date Country
20100222331 A1 Sep 2010 US