WO2011131741A1 - Heterocyclic carboxylic acid amides as pdk1 inihibitors - Google Patents

Heterocyclic carboxylic acid amides as pdk1 inihibitors Download PDF

Info

Publication number
WO2011131741A1
WO2011131741A1 PCT/EP2011/056368 EP2011056368W WO2011131741A1 WO 2011131741 A1 WO2011131741 A1 WO 2011131741A1 EP 2011056368 W EP2011056368 W EP 2011056368W WO 2011131741 A1 WO2011131741 A1 WO 2011131741A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
ynyl
prop
carboxamide
optionally substituted
Prior art date
Application number
PCT/EP2011/056368
Other languages
French (fr)
Inventor
Harald Engelhardt
Christiane Kofink
Darryl Mcconnell
Original Assignee
Boehringer Ingelheim International Gmbh
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Boehringer Ingelheim International Gmbh filed Critical Boehringer Ingelheim International Gmbh
Priority to EP11716881.5A priority Critical patent/EP2560967B1/en
Priority to JP2013505483A priority patent/JP5860031B2/en
Publication of WO2011131741A1 publication Critical patent/WO2011131741A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/14Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings

Definitions

  • the present invention relates to new quinoxaline, quinoline and quinazoline compounds of general formula (1 )
  • the aim of the present invention is to indicate new compounds which can be used for the prevention and/or treatment of diseases characterised by excessive or abnormal cell proliferation.
  • the compounds according to the invention are characterised by a powerful inhibitory effect on PDK 1 and a high efficacy against tumour cells, e.g. prostate carcinoma cells, which is mediated through inhibiting PDK 1 .
  • tumour cells e.g. prostate carcinoma cells
  • the compounds have good solubility and good selectivity with regard to other signal and cell cycle kinases.
  • the importance of the PI3K-PDK1 -AKT pathway with its frequent aberrations (PTEN loss, PI3K mutation) in the major tumor indications is described in numerous scientific publications (e.g. Samuels et al. 2004; Science; 304: 554; Samuels and Velculescu 2004; Cell cycle 3; 17-19; Samuels et al. 2005; Cancer Cell; 7: 561 -573).
  • PDK1 alterations (mutations, CN gains) are rarely found isolated from other pathway alterations so that the importance of this target thrives from the fact that it acts as a central signaling node in a frequently altered pathway.
  • Active PI3K phosphorylates second messengers like Phosphatidylinositol(4.5)- bisphosphate (Ptdlns(4.5)P 2 ) to Ptdlns(3.4.5)P 3 , or Ptdlns(4)P to Ptdlns(3.4,)P 2 .
  • Ptdlns(4.5)P 2 Phosphatidylinositol(4.5)P 2
  • Ptdlns(4.5)P 2 Ptdlns(3.4.5)P 3
  • Ptdlns(4)P Ptdlns(3.4,)P 2 .
  • Such PI3K products recruit AKT and PDK1 kinases via their PH-domains (which bind the PI3 kinase products) to the plasma membrane and allow PDK1 to phosphorylate Thr308 of AKT and thereby activate the enzyme.
  • a second step important for additional AKT activation and/or AKT-substrate selection is mediated by the mTOR/rictor/SIN1 complex which phosphorylates the hydrophobic motif of AKT at Ser473.
  • Activated AKT regulates several other proteins, which are involved in cell proliferation, growth and survival.
  • PDK1 has other substrates located in the cytoplasm, e.g. p70S6K, p90RSK, PKCs and SGK, which are also implicated in regulating cell proliferation and cell survival.
  • the activation of these kinases is not dependent on phosphatidylinositols in vitro and functions properly in cells where the PDK1 -PH-domain had been destroyed by knock-in mutation (McManus et al. 2004, EMBO J. 23, 2071 -2082).
  • PDK1 mouse mutants also see Bayascas 2008; Cell cycle 7; 2978-2982).
  • PDK1 substrates (AKT, p90RSK) act at the crossroad of two important cancer signaling pathways.
  • a potent and selective inhibitor of the PDK1 kinase is predicted to inhibit tumor growth, delay relapse, or even induce objective tumor responses (i.e. complete or partial tumor shrinkage) in cancer patients.
  • compounds of general formula (1 ) wherein the groups R 1 to R 4 , Q a , Q b , Q H , L and n are defined as stated hereinafter act as inhibitors of specific signal enzymes which are involved in controlling the proliferation of cells.
  • the compounds according to the invention may be used for example for the treatment of diseases which are connected with the activity of these signal enzymes and are characterised by excessive or abnormal cell proliferation.
  • the present invention therefore relates to compounds of general formula (1 )
  • Q a is a ring system optionally substituted by one or more identical or different R a and/or R b , selected from among C 3- iocycloalkyl, C 6 -ioaryl, 5-12 membered heteroaryl and 3-14 membered heterocyclyl;
  • R 1 and R 2 are selected independently of one another from among hydrogen, halogen, C h alky I , C 2 - 4 alkenyl, C 2-4 alkynyl, Ci -4 haloalkyl, -NH 2 , -CN, -NHCi -4 alkyl, -N(Ci -4 alkyl) 2 , -OH, -OCi -4 alkyl, HO-Ci -4 alkylene, Ci -4 alkyl-0-Ci -4 alkylene, H 2 N-Ci -4 alkylene, -OCi -4 haloalkyl, (Ci -4 alkyl)NH-Ci -4 alkylene- and (Ci -4 alkyl) 2 N-Ci -4 alkylene, while alkyl, alkenyl, alkynyl and alkylene mentioned in the above groups may optionally be substituted by one or more identical or different halogens;
  • the ring system Q b is selected from among
  • each R 3 is independently selected from among halogen, Ci -4 alkyl, C 2-4 alkenyl, C 2-4 alkynyl, Ci -4 haloalkyl, -NH 2 , -CN, -NHCi -4 alkyl, -N(C 1-4 alkyl) 2 , -OH, -OCi -4 alkyl, HO-Ci -4 alkylene, HO-C 2-4 alkylene-0, Ci -4 alkyl-0-Ci -4 alkylene, Ci -4 haloalkyl-0-Ci -4 alkylene,
  • n denotes the number 0, 1 , 2 or 3 if Q b corresponds to the ring system Q b -4;
  • each n independently denotes the number 0, 1 or 2 if Q b corresponds to one of the ring systems Q b -1 , Q b -2, Q b -3 or Q b -5;
  • R 4 denotes hydrogen or Ci -4 alkyl
  • R 5 and R 6 are each independently of one another selected from among hydrogen, halogen, Ci -4 alkyl, Ci -4 haloalkyl;
  • the ring system Q H is selected from among
  • R 7 and R 9 are each independently of one another selected from among hydrogen, halogen, -NR a R a , -N(OR a )R a , -OR a , -COOR a , -COR a , -CONR a R a , -SR a , -N0 2 and -CN or denote a group optionally substituted by one or more identical or different R b and/or R c , selected from among Ci -6 alkyl , C 2-6 alkenyl, C 2-6 alkynyl, C 3- iocycloalkyl, C 6 -ioaryl, 5-12 membered heteroaryl and 3-14 membered heterocyclyl;
  • R 8 is independently selected from among hydrogen, -NR a R a , -N(OR a )R a , -OR a , -COOR a , -COR a , -CONR a R a and -SR a or denotes a group optionally substituted by one or more identical or different R b and/or R c , selected from among Ci -6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3- iocycloalkyl, C 6 -ioaryl, 5-12 membered heteroaryl and 3-14 membered heterocyclyl;
  • R 10 is independently selected from among -NR a R a , -N(OR a )R a , -OR a and -SR a or denotes a group optionally substituted by one or more identical or different R b and/or R c selected from among Ci -6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3- iocycloalkyl, C 6 -ioaryl, 5-12 membered heteroaryl and 3-14 membered heterocyclyl;
  • each R 11 and R 12 is independently selected from among R a and R b ;
  • each R g is independently selected from among hydrogen, Ci -6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3- iocycloalkyl, C 6 -ioaryl, 5-12 membered heteroaryl and 3-14 membered heterocyclyl and the above-mentioned groups R 9 may optionally be substituted by one or more identical or different substituents, selected independently of one another from among -NH 2 , -NH(Ci -6 alkyl), -N(d -6 alkyl) 2 and Ci -6 alkyl;
  • the compounds (1 ) may optionally also be present in the form of the tautomers, racemates, enantiomers, diastereomers or mixtures thereof or as the respective salts of all the above-mentioned forms.
  • Q a is a ring system optionally substituted by one or more identical or different R a and/or R b selected from among C 5 - 6 cycloalkyl, phenyl, 5-6 membered heteroaryl and 5-7 membered heterocyclyl, and
  • R a and R b are as hereinbefore defined.
  • the invention relates to compounds (1 ), wherein
  • Q a is a ring system optionally substituted by one or more identical or different R a and/or R b selected from among phenyl and 5-6 membered heteroaryl, and
  • R a and R b are as hereinbefore defined.
  • the invention relates to compounds with a structural aspect (A1 ) and (A2), wherein the ring system Q a optionally carries one or more identical or different substituents, selected from among halogen and Ci -4 alkyl.
  • Q a is selected from among phenyl, 3-fluorophenyl, 4-fluorophenyl, 3,4,5-trifluorophenyl, 3,5-difluorophenyl and 3,4-difluorophenyl.
  • Q a denotes 3,4-difluorophenyl.
  • R 1 and R 2 are selected independently of one another from among hydrogen, Ci -4 alkyl, C 2-4 alkynyl, -CN, HO-Ci -4 alkylene-, H 2 N-Ci -4 alkylene-, (Ci -4 alkyl)NH-Ci -4 alkylene-,
  • R 1 and R 2 are selected independently of one another from among hydrogen, Ci -4 alkyl, H 2 N-Ci -4 alkylene and HO-Ci -4 alkylene.
  • R 1 and R 2 are independently selected from among hydrogen, methyl, aminomethyl and hydroxymethyl.
  • the invention relates to compounds with a structural aspect (B1 ), (B2) and (B3), wherein
  • R 1 denotes hydrogen
  • the invention relates to compounds (1 ), wherein the ring system Q b is selected from among
  • each R 3 is independently selected from among halogen, Ci -4 alkyl, -OCi -4 alkyl, HO-Ci -4 alkylene, Ci -4 alkyl-0-Ci -4 alkylene, -NH(C 2 - 4 alkylene-NH 2 ), -NH[C 2 - 4 alkylene-NH(Ci -4 alkyl)] and -NH[C 2 - 4 alkylene-N(Ci -4 alkyl) 2 ];
  • n denotes the number 0, 1 , 2 or 3 if Q b corresponds to the ring system Q b -4 and each n independently denotes the number 0, 1 or 2 if Q b corresponds to one of the ring systems Q b -1 , Q b -2, Q b -3 or Q b -5,
  • the ring system Q b is selected from among
  • n has the value 0.
  • the invention relates to compounds (1 ), wherein
  • the ring system Q b is selected from among
  • Q b -2 and Q b -4 n has the value 0.
  • R 4 denotes hydrogen or methyl.
  • R 4 denotes hydrogen
  • the ring system Q H is selected from among
  • Q H -2a and 2 Q H -2d R 7 is independently selected from among hydrogen, -NR a R a , -N(OR a )R a , -OR a , -C00R 3 , -COR a , -CONR a R a and -SR a , or denotes a group optionally substituted by one or more identical or different R b and/or R c , selected from among Ci -6 alkyl, C 2 - 6 alkenyl, C 2 - 6 alkynyl, C 3- iocycloalkyl, C 6 -ioaryl, 5-12 membered heteroaryl and 3-14 membered heterocyclyl, and
  • R 11 , R 12 , R a , R b , R c and m are as hereinbefore defined.
  • the invention relates to compounds (1 ), wherein the ring system Q H is selected from among
  • R 7 is independently selected from among hydrogen, -NR a1 R a1 , -N(OR a1 )R a1 , -OR a1 , -COOR a1 , -COR a1 , -CONR a1 R a1 and -SR a1 or denotes a group optionally substituted by one or more identical or different R b1 and/or R c1 selected from among Ci -6 alkyl, C 2 - 6 alkenyl , C 2 - 6 alkynyl, C 3- iocycloalkyl, C 6 -ioaryl, 5-12 membered heteroaryl and 3-14 membered heterocyclyl;
  • each R a1 independently denotes hydrogen or a group optionally substituted by one or more identical or different R b1 and/or R c1 , selected from among Ci -6 alkyl, C 3 -i 0 cycloalkyl and 3-14 membered heterocyclyl;
  • each R b1 is independently selected from among -OR c1 , -NR c1 R c1 and halogen;
  • each R c1 independently denotes hydrogen or a group optionally substituted by one or more identical or different R d1 and/or R e1 selected from among Ci -6 alkyl, C 3 -i 0 cycloalkyl and 3-14 membered heterocyclyl;
  • each R d1 is independently selected from among -OR e1 , -CN and -C(0)OR e1 ;
  • each R e1 independently denotes hydrogen or a group optionally substituted by one or more identical or different cycloalkyl (particularly C 3 - 6 cycloalkyl) selected from among Ci -6 alkyl and C 3- i 0 cycloalkyl, and
  • R 11 , R 12 and m are as hereinbefore defined.
  • R 7 is selected from among hydrogen, -NR a1 R a1 , -OR a1 and -SR a1 , or denotes a group optionally substituted by one or more identical or different R b1 and/or R c1 , selected from among Ci -6 alky I , C 2 - 6 alkenyl, pyrrolidinyl, piperazinyl, piperidinyl, morpholinyl, homomorpholinyl, azetidinyl and homopiperazinyl;
  • each R a1 independently denotes hydrogen or a group optionally substituted by one or more identical or different R b1 and/or R c1 , selected from among Ci -6 alkyl, C 3-6 cycloalkyl, piperazinyl, piperidinyl, morpholinyl, azetidinyl and pyrrolidinyl;
  • each R b1 is independently selected from among -OR c1 , -NR c1 R c1 and halogen;
  • each R c1 independently denotes hydrogen or a group optionally substituted by one or more identical or different R d1 and/or R e1 selected from among Ci -6 alkyl, C 3 - 6 cycloalkyl, pyrrolidinyl, morpholinyl, homomorpholinyl, piperazinyl, homopiperazinyl, oxetanyl and piperidinyl;
  • each R d1 is independently selected from among -OR e1 , -CN and -C(0)OR e1 ;
  • each R e1 independently denotes hydrogen or a group optionally substituted by one or more identical or different C 3-6 cycloalkyl, selected from among Ci -6 alkyl and C 3-6 cycloalkyl, and
  • R 11 and m are as hereinbefore defined.
  • R 7 denotes -NHR a1 ;
  • R a1 is a group optionally substituted by one or more identical or different R b1 and/or R c1 , selected from among Ci -4 alkyl, cyclohexyl, cyclopentyl and piperidinyl;
  • each R b1 is independently selected from among -NR c1 R c1 and fluorine;
  • each R c1 independently denotes hydrogen or a group optionally substituted by one or more identical or different R d1 and/or R e1 , selected from among Ci -4 alkyl, cyclopropyl, morpholinyl, piperazinyl and oxetanyl;
  • each R d1 denotes -OR e1 ;
  • each R e1 independently denotes hydrogen or a Ci -4 alkyl optionally substituted by cyclopropyl
  • R 11 and m are as hereinbefore defined.
  • Q H -2a R 7 is selected from among
  • R 11 and m are as hereinbefore defined.
  • the ring system Q H is selected from among
  • R 9 is independently selected from among hydrogen, -NR a R a , -N(OR a )R a , -OR a , -C00R 3 , -COR a , -CONR a R a and -SR a , or denotes a group optionally substituted by one or more identical or different R b and/or R c , selected from among Ci -6 alkyl, C 2 - 6 alkenyl, C 2 - 6 alkynyl, C 3- iocycloalkyl, C 6 -ioaryl, 5-12 membered heteroaryl and 3-14 membered heterocyclyl, and R 11 , R 12 , R a , R b , R c and m are as hereinbefore defined.
  • the ring system Q H is selected from among
  • R 9 is independently selected from among hydrogen, -NR a2 R a2 , -N(OR a2 )R a2 , -OR a2 , -COOR 32 , -COR a2 , -CONR a2 R a2 and -SR a2 , or denotes a group optionally substituted by one or more identical or different R b2 and/or R c2 , selected from among Ci -6 alkyl, C 2 - 6 alkenyl , C 2 - 6 alkynyl, C 3- iocycloalkyl, C 6 -ioaryl, 5-12 membered heteroaryl and 3-14 membered heterocyclyl;
  • each R a2 independently denotes hydrogen or a group optionally substituted by one or more identical or different R b2 and/or R c2 selected from among Ci -6 alkyl, C 3 -i 0 cycloalkyl and 3-14 membered heterocyclyl;
  • each R b2 is independently selected from among -OR c2 , -NR c2 R c2 and halogen;
  • each R c2 independently denotes hydrogen or a group optionally substituted by one or more identical or different R d2 and/or R e2 selected from among Ci -6 alkyl, C 3 -i 0 cycloalkyl and 3-14 membered heterocyclyl;
  • each R d2 is independently selected from among -OR e2 , -CN and -C(0)OR e2 ;
  • each R e2 independently denotes hydrogen or a group optionally substituted by one or more identical or different cycloalkyl (particularly C 3 - 6 cycloalkyl) selected from among Ci -6 alkyl and C 3- i 0 cycloalkyl, and
  • R 11 , R 12 and m are as hereinbefore defined.
  • Q H -2b R 9 is selected from among hydrogen, -NR a2 R a2 , -OR a2 and -SR a2 , or denotes a group optionally substituted by one or more identical or different R b2 and/or R c2 selected from among Ci -6 alky I , C 2 - 6 alkenyl, pyrrolidinyl, piperazinyl, piperidinyl, morpholinyl, homomorpholinyl, azetidinyl and homopiperazinyl;
  • each R a2 independently denotes hydrogen or a group optionally substituted by one or more identical or different R b2 and/or R c2 , selected from among Ci -6 alkyl, C 3-6 cycloalkyl, piperazinyl, piperidinyl, morpholinyl, azetidinyl and pyrrolidinyl;
  • each R b2 is independently selected from among -OR c2 , -NR c2 R c2 and halogen;
  • each R c2 independently denotes hydrogen or a group optionally substituted by one or more identical or different R d2 and/or R e2 , selected from among Ci -6 alkyl, C 3 - 6 cycloalkyl, pyrrolidinyl, morpholinyl, homomorpholinyl, piperazinyl, homopiperazinyl, oxetanyl and piperidinyl;
  • each R d2 is independently selected from among -OR e2 , -CN and -C(0)OR e2 ;
  • each R e2 independently denotes hydrogen or a group optionally substituted by one or more identical or different C 3 - 6 cycloalkyl, selected from among Ci -6 alkyl and C 3 - 6 cycloalkyl, and
  • R 11 and m are as hereinbefore defined.
  • R 9 denotes -NHR a2 ;
  • R a2 is a group optionally substituted by one or more identical or different R b2 and/or R c2 selected from among Ci -4 alkyl, cyclohexyl, cyclopentyl and piperidinyl;
  • each R b2 is independently selected from among -NR c2 R c2 and fluorine;
  • each R c2 independently denotes hydrogen or a group optionally substituted by one or more identical or different R and/or R , selected from among Ci -4 alkyl, cyclopropyl, morpholinyl, piperazinyl and oxetanyl;
  • each R d2 denotes -OR e2 ;
  • each R e2 independently denotes hydrogen or a Ci -4 alkyl optionally substituted by cyclopropyl
  • R 11 and m are as hereinbefore defined.
  • R 9 is selected from among
  • R 11 and m are as hereinbefore defined.
  • the ring system Q H is selected from among
  • R is selected from among hydrogen, -NR a R a , -N(OR a )R a , -OR a and -SR a , or denotes a group optionally substituted by one or more identical or different R b and/or R c , selected from among Ci -6 alkyl, C 2 - 6 alkenyl, C 2 - 6 alkynyl, C 3- iocycloalkyl, C 6 -ioaryl, 5-12 membered heteroaryl and 3-14 membered heterocyclyl;
  • R 9 is independently selected from among hydrogen, halogen, -N0 2 , -CN and Ci -4 alkyl, and
  • R 11 , R 12 , R a , R b , R c and m are as hereinbefore defined.
  • the ring system Q H is selected from among
  • R is selected from among -NR R and -OR , or denotes a group optionally substituted by one or more identical or different R b3 and/or R c3 selected from among Ci -6 alkyl, C 2 - 6 alkenyl, C 2 - 6 alkynyl, 5-12 membered heteroaryl and 3-14 membered heterocyclyl;
  • R 9 is independently selected from among hydrogen, halogen, -N0 2 , -CN and Ci -4 alkyl; each R a3 independently denotes hydrogen or a group optionally substituted by one or more identical or different R b3 and/or R c3 selected from among Ci -6 alkyl, C 3 -i 0 cycloalkyl, C 6 -ioaryl and 3-14 membered heterocyclyl;
  • each R b3 is independently selected from among -OR c3 and -NR c3 R c3 ;
  • each R c3 independently denotes hydrogen or a group optionally substituted by one or more identical or different Ci -6 alkyl selected from among Ci -6 alkyl, C 3- iocycloalkyl, C 6- ioaryl and 3-14 membered heterocyclyl, and
  • R 11 , R 12 and m are as hereinbefore defined.
  • R is selected from among -NR R and -OR , or denotes a group optionally substituted by one or more identical or different R b3 and/or R c3 , selected from among Ci -6 alkyl, 5-6 membered heteroaryl and 4-6 membered heterocyclyl;
  • R 9 is independently selected from among hydrogen, halogen, -N0 2 , -CN and Ci -4 alkyl; each R independently denotes hydrogen or a group optionally substituted by one or more identical or different R b3 and/or R c3 , selected from among Ci -6 alkyl, C 3-6 cycloalkyl, phenyl, pyrrolidinyl, and piperidinyl;
  • each R b3 is independently selected from among -OR c3 and -NR c3 R c3 ;
  • each R c3 independently denotes hydrogen or a group optionally substituted by one or more identical or different Ci -6 alkyl, selected from among Ci -6 alkyl, piperazinyl, piperidinyl, tetrahydropyranyl, tetrahydrofuryl and pyrrolidinyl, and
  • R 11 and m are as hereinbefore defined.
  • R denotes -OR a3 ;
  • R 9 is independently selected from among hydrogen, halogen, -N0 2 , -CN and Ci -4 alkyl;
  • R a3 denotes a Ci -6 alkyl which is substituted by a substituent selected from among -N(Ci -4 alkyl) 2 , piperidinyl, piperazinyl or pyrrolidine, while the above-mentioned piperidinyl, piperazinyl and pyrrolidine may optionally be substituted by a Ci -4 alkyl and
  • R 11 and m are as hereinbefore defined.
  • Q H -2b R is selected from among
  • R 9 is independently selected from among hydrogen, halogen, -N0 2 , -CN and Ci -4 alkyl, and R 11 and m are as hereinbefore defined.
  • the ring system Q H is selected from among
  • R 10 is independently selected from among -NR a R a , -N(OR a )R a , -OR a and -SR a or denotes a group optionally substituted by one or more identical or different R b and/or R c selected from among Ci -6 alkyl, C 2-6 alkenyl , C 2-6 alkynyl, C 3- iocycloalkyl, C 6 -ioaryl, 5-12 membered heteroaryl and 3-14 membered heterocyclyl, and
  • R 11 , R 12 , R a , R b , R c and m are as hereinbefore defined.
  • the ring system Q H is selected from among
  • R 10 is independently selected from among -NR a4 R a4 , -N(OR a4 )R a4 , -OR a4 and -SR a4 or denotes a group optionally substituted by one or more identical or different R b4 and/or R c4 selected from among Ci -6 alkyl , C 2 - 6 alkenyl, C 2 - 6 alkynyl, C 3- iocycloalkyl, C 6 -ioaryl, 5-12 membered heteroaryl and 3-14 membered heterocyclyl;
  • each R a4 independently denotes hydrogen or a group optionally substituted by one or more identical or different R b4 and/or R c4 selected from among Ci -6 alkyl, C 3- iocycloalkyl, C 6 -ioaryl and 3-14 membered heterocyclyl;
  • each R b4 is independently selected from among -OR c4 , -NR c4 R c4 , -C(0)R c4 , -C(0)OR c4 , -C(0)NR c4 R c4 , -NHC(0)R c4 and -N(Ci -4 alkyl)C(0)R c4 ;
  • each R c4 independently of one another denotes hydrogen or a group optionally substituted by one or more identical or different R d4 and/or R e4 , selected from among Ci -6 alkyl, C 3- iocycloalkyl, C 6 -ioaryl, 5-12 membered heteroaryl and 3-14 membered heterocyclyl; each R d4 denotes -OR e1 ;
  • each R e4 independently denotes hydrogen or a group optionally substituted by one or more identical or different R f4 selected from among Ci -6 alkyl and C 6- ioaryl;
  • each R f4 denotes halogen
  • R 11 , R 12 and m are as hereinbefore defined.
  • R 10 is selected from among -NR a4 R a4 , -N(OR a4 )R a4 and -OR a4 or denotes a group optionally substituted by one or more identical or different R b4 and/or R c4 selected from among Ci -6 alkyl, phenyl, 5-6 membered heteroaryl, piperazinyl, pyrrolidinyl, morpholinyl, homomorpholinyl, homopiperazinyl and piperidinyl;
  • each R a4 independently denotes hydrogen or a group optionally substituted by one or more identical or different R b4 and/or R c4 selected from among Ci -6 alkyl, C 3-6 cycloalkyl, phenyl, azetidinyl, tetrahydropyranyl, piperidinyl and pyrrolidinyl;
  • each R b4 is independently selected from among -OR c4 , -NR c4 R c4 , -C(0)R c4 , -C(0)OR c4 , -C(0)NR c4 R c4 and -NHC(0)R c4 ;
  • each R c4 independently denotes hydrogen or a group optionally substituted by one or more identical or different R d4 and/or R e4 selected from among Ci -6 alkyl, phenyl, furyl, pyridyl, imidazolyl, thienyl, pyrazolyl, quinolinyl, oxetanyl, tetrahydrofuryl, azetidinyl, pyrrolidinyl, piperidinyl and piperazinyl;
  • each R d4 denotes -OR e4 ;
  • each R e4 independently denotes hydrogen or a group optionally substituted by one or more identical or different R f4 selected from among Ci -6 alkyl and phenyl;
  • each R f4 denotes halogen
  • R 11 and m are as hereinbefore defined.
  • the invention relates to compounds (1 ), wherein
  • R 10 is selected from among -NHMe, -NMe 2 and -NEt 2 , and
  • R 11 and m are as hereinbefore defined.
  • each R 11 and R 12 is independently selected from among hydrogen, Ci -4 alkyl and halogen, and
  • n denotes the value 0, 1 or 2.
  • each R 11 and R 12 is independently selected from among hydrogen, Ci -4 alkyl and halogen, and
  • m denotes the value 0 or 1 .
  • m has the value 0.
  • All the above-mentioned structural aspects A1 to A6, B1 to B6, C1 to C4, D1 and D2, E1 , F1 to F19 and G1 to G3 are preferred embodiments of the aspects AO, B0, CO, DO, E0, F0 and GO, respectively.
  • the structural aspects AO to A6, B0 to B6, CO to C4, DO to D2, E0 and E1 , F0 to F19 and GO to G3 relating to different molecular parts of the compounds (1 ) according to the invention may be permutated with one another as desired in combinations ABCDEFG so as to obtain preferred compounds (1 ) .
  • Each combination ABCDEFG represents and defines individual embodiments or generic amounts of compounds AOBOCODOEOFOGO according to the invention.
  • Each individual embodiment or partial quantity defined by this combination is expressly also included and is a subject of the invention.
  • the present invention further relates to all the pharmaceutically acceptable salts of compounds of general formula (1 ).
  • the present invention further relates to hydrates, solvates, polymorphs, metabolites, derivatives and prodrugs of compounds of general formula (1 ).
  • the invention relates to compounds of general formula (1 ) - or the pharmaceutically acceptable salts thereof - as medicaments.
  • the invention relates to compounds of general formula (1 ) - or the pharmaceutically acceptable salts thereof - for use in the treatment and/or prevention of cancer, infections, inflammations and autoimmune diseases.
  • the invention relates to compounds of general formula (1 ) - or the pharmaceutically acceptable salts thereof - for use in the treatment and/or prevention of cancer.
  • the invention relates to compounds of general formula (1 ) - or the pharmaceutically acceptable salts thereof - for use in the treatment and/or prevention of carci nomas of the breast, prostate or ovary, non-small-cell bronchial carcinomas (NSCLC), melanomas and chronic lymphatic leukaemias (CLL).
  • NSCLC non-small-cell bronchial carcinomas
  • CLL chronic lymphatic leukaemias
  • the invention in another aspect relates to a process for the treatment and/or prevention of cancer comprising administering a therapeutically effective amount of a compound of general formula (1 ) - or one of the pharmaceutically acceptable salts thereof - to a human being.
  • the invention in another aspect relates to a pharmaceutical preparation containing as active substance one or more compounds of general formula (1 ) - or the pharmaceutically acceptable salts thereof - optionally in combination with conventional excipients and/or carriers.
  • the invention in another aspect relates to a pharmaceutical preparation comprising a compound of general formula (1 ) - or one of the pharmaceutically acceptable salts thereof - and at least one other cytostatic or cytotoxic active substance, different from formula (1).
  • the indication of the number of members in groups that contain one or more heteroatom(s) (heteroalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, heterocyclylalkyl) relates to the total atomic number of all the ring members or chain members or the total of all the ring and chain members.
  • Alkyl denotes monovalent, saturated hydrocarbon chains, which may be present in both straight-chain (unbranched) and branched form. If an alkyl is substituted, the substitution may take place independently of one another, by mono- or polysubstitution in each case, on all the hydrogen-carrying carbon atoms.
  • C 1-5 -alkyl includes for example H 3 C-, H 3 C-CH 2 -, H 3 C-CH2-CH2-,
  • alkyl are methyl (Me; -CH 3 ), ethyl (Et; -CH 2 CH 3 ), 1 -propyl (n-propyl; n-Pr; -CH2CH2CH3) , 2-propyl (/ ' -Pr; / ' so-propyl; -CH(CH 3 ) 2 ), 1 -butyl (n-butyl; n-Bu; -CH2CH2CH2CH3), 2-methyl-1 -propyl (/ ' so-butyl; /-Bu; -CH 2 CH(CH 3 ) 2 ), 2-butyl (sec-butyl; sec-Bu; -CH(CH 3 )CH 2 CH 3 ), 2-methyl-2-propyl (ferf-butyl; f-Bu; -C(CH 3 ) 3 ), 1 -pentyl (n-pentyl; -CH2CH2CH2CH3) , 2-p
  • alkyi also applies if alkyi is a part of another group such as for example C x - y -alkylamino or C x-y -alkyloxy.
  • alkylene can also be derived from alkyi.
  • Alkylene is bivalent, unlike alkyi, and requires two binding partners. Formally, the second valency is produced by removing a hydrogen atom in an alkyi.
  • Corresponding groups are for example -CH 3 and -CH 2 , -CH 2 CH 3 and -CH 2 CH 2 or >CHCH 3 etc.
  • C 1-4 -alkylene includes for example -(CH 2 )-, -(CH 2 -CH 2 )-, -(CH(CH 3 ))-, -(CH 2 -CH 2 -CH 2 )-, -(C(CH 3 ) 2 )-, -(CH(CH 2 CH 3 ))-, -(CH(CH 3 )-CH 2 )-, -(CH 2 -CH(CH 3 ))-, -(CH 2 -CH 2 -CH 2 )-, -(CH 2 -CH 2 -CH(CH 3 ))-, -(CH 2 -CH 2 -CH(CH 3 ))-, -(CH(CH 3 )-CH 2 -CH 2 )-,
  • alkylene examples include methylene, ethylene, propylene, 1 -methylethylene, butylene, 1 -methylpropylene, 1 .1 -dimethylethylene, 1 ,2-dimethylethylene, pentylene, 1 ,1 -dimethylpropylene, 2,2-dimethylpropylene, 1 ,2-dimethylpropylene,
  • propylene includes 1 -methylethylene and butylene includes 1 -methylpropylene, 2-methylpropylene, 1 , 1 -dimethylethylene and 1 ,2-dimethylethylene.
  • alkylene also applies if alkylene is part of another group such as for example in HO-C x - y -alkylenamino or H 2 N-Cx-y-alkylenoxy.
  • alkenyl consists of at least two carbon atoms, wherein at least two adjacent carbon atoms are joined together by a C-C double bond. If in an alkyl as hereinbefore defined having at least two carbon atoms, two hydrogen atoms on adjacent carbon atoms are formally removed and the free valencies are saturated to form a second bond, the corresponding alkenyl is formed.
  • alkenyl examples include 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-en
  • propenyl includes prop-1 -enyl and prop-2-enyl
  • butenyl includes but-1 -enyl, but-2-enyl, but-3-enyl, 1 -methyl-prop-1 -enyl, 1 -methyl-prop-2-enyl etc.
  • Alkenyl may optionally be present in the cis or trans or £ or Z orientation with regard to the double bond(s).
  • alkenyl also applies when alkenyl is part of another group such as for example in C x - y -alkenylamino or C x-y -alkenyloxy.
  • alkenylene consists of at least two carbon atoms, wherein at least two adjacent carbon atoms are joined together by a C-C double bond. If in an alkylene as hereinbefore defined having at least two carbon atoms, two hydrogen atoms at adjacent carbon atoms are formally removed and the free valencies are saturated to form a second bond, the corresponding alkenylene is formed.
  • alkenylene examples include ethenylene, propenylene, 1 -methylethenylene, butenylene, 1 -methylpropenylene, 1 ,1 -dimethylethenylene, 1 ,2-dimethylethenylene, pentenylene, 1 ,1 -dimethylpropenylene, 2,2-dimethylpropenylene, 1 ,2-dimethylpropenylene, 1 ,3-dimethylpropenylene, hexenylene etc.
  • propenylene includes 1 -methylethenylene and butenylene includes 1 -methylpropenylene, 2-methylpropenylene, 1 ,1 -dimethylethenylene and 1 ,2-dimethylethenylene.
  • Alkenylene may optionally be present in the c/ ' s or trans or £ or Z orientation with regard to the double bond(s).
  • alkenylene also applies when alkenylene is a part of another group as in for example HO-C x - y -alkenylenamino or H 2 N-Cx-y-alkenylenoxy.
  • alkynyl consists of at least two carbon atoms, wherein at least two adjacent carbon atoms are joined together by a C-C triple bond. If in an alkyl as hereinbefore defined having at least two carbon atoms, two hydrogen atoms in each case at adjacent carbon atoms are formally removed and the free valencies are saturated to form two further bonds, the corresponding alkynyl is formed.
  • alkynyl examples include ethynyl, prop-1 -ynyl, prop-2-ynyl, but-1 -ynyl, but-2-ynyl, but-3-ynyl, 1 -methyl-prop-2-ynyl, pent-1 -ynyl, pent-2-ynyl, pent-3-ynyl, pent-4-ynyl, 3-methyl-but-1 -ynyl, hex-1 -ynyl, hex-2-ynyl, hex-3-ynyl, hex-4-ynyl, hex-5-ynyl etc.
  • propynyl includes prop-1 -ynyl and prop-2-ynyl
  • butynyl includes but-1 -ynyl, but-2-ynyl, but-3-ynyl, 1 -methyl-prop-1 -ynyl, 1 -methyl-prop-2-ynyl, etc.
  • hydrocarbon chain carries both at least one double bond and also at least one triple bond, by definition it belongs to the alkynyl subgroup.
  • alkynyl also applies if alkynyl is part of another group, as in Cx-y-alkynylamino or C x-y -alkynyloxy, for example.
  • alkynylene consists of at least two carbon atoms, wherein at least two adjacent carbon atoms are joined together by a C-C triple bond. If in an alkylene as hereinbefore defined having at least two carbon atoms, two hydrogen atoms in each case at adjacent carbon atoms are formally removed and the free valencies are saturated to form two further bonds, the corresponding alkynylene is formed.
  • alkynylene examples include ethynylene, propynylene, 1 -methylethynylene, butynylene, 1 -methylpropynylene, 1 ,1 -dimethylethynylene, 1 ,2-dimethylethynylene, pentynylene, 1 ,1 -dimethylpropynylene, 2,2-dimethylpropynylene, 1 ,2-dimethylpropynylene, 1 ,3-dimethylpropynylene, hexynylene etc.
  • propynylene includes 1 -methylethynylene and butynylene includes 1 -methylpropynylene, 2-methylpropynylene, 1 ,1 -dimethylethynylene and 1 ,2- dimethylethynylene.
  • alkynylene also applies if alkynylene is part of another group, as in HO-Cx-y-alkynyleneamino or H 2 N-Cx-y-alkynyleneoxy, for example.
  • heteroatoms oxygen, nitrogen and sulphur atoms.
  • Haloalkyl (haloalkenyl, haloalkynyl) is derived from the previously defined alkyl (alkenyl, alkynyl) by replacing one or more hydrogen atoms of the hydrocarbon chain independently of one another by halogen atoms, which may be identical or different. If a haloalkyl (haloalkenyl, haloalkynyl) is to be further substituted, the substitutions may take place independently of one another, in the form of mono- or polysubstitutions in each case, on all the hydrogen-carrying carbon atoms.
  • haloalkyl haloalkenyl, haloalkynyl
  • -CCI CH 2
  • -CBr CH 2
  • -CI CH 2 , -C ⁇ C-CF 3 , -CHFCH 2 CH 3 , -CHFCH 2 CF 3 etc.
  • haloalkyl haloalkenyl, haloalkynyl
  • haloalkylene haloalkenylene, haloalkynylene
  • Haloalkylene haloalkenyl, haloalkynyl
  • the second valency is formed by removing a hydrogen atom from a haloalkyl.
  • Corresponding groups are for example -CH 2 F and -CHF-, -CHFCH 2 F and -CHFCHF- or >CFCH 2 F etc.
  • Halogen relates to fluorine, chlorine, bromine and/or iodine atoms.
  • Cycloalkyl is made up of the subgroups monocyclic hydrocarbon rings, bicyclic hydrocarbon rings and spiro-hydrocarbon rings. The systems are saturated. In bicyclic hydrocarbon rings two rings are joined together so that they have at least two carbon atoms together. In spiro-hydrocarbon rings a carbon atom (spiroatom) belongs to two rings together. If a cycloalkyl is to be substituted, the substitutions may take place independently of one another, in the form of mono- or polysubstitutions in each case, on all the hydrogen-carrying carbon atoms. Cycloalkyl itself may be linked as a substituent to the molecule via every suitable position of the ring system.
  • cycloalkyl examples include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, 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[4.1 .0]heptyl (norcaranyl), bicyclo-[3.1.1 ]heptyl (pinanyl), spiro[2.5]octyl, spiro[3.3]heptyl etc.
  • cycloalkyl also applies if cycloalkyl is part of another group as in Cx-y-cycloalkylamino or C x-y -cycloalkyloxy, for example.
  • cycloalkylene can thus be derived from the previously defined cycloalkyl.
  • Cycloalkylene unlike cycloalkyl, is bivalent and requires two binding partners. Formally, the second valency is obtained by removing a hydrogen atom from a cycloalkyl.
  • Corresponding groups are for ex cyclohexyl and (cyclohexylene).
  • cycloalkylene is part of another group as in HO-C x - y -cycloalkyleneamino or H 2 N-C x -y-cycloalkyleneoxy, for example.
  • Cycloalkenyl is also made up of the subgroups monocyclic hydrocarbon rings, bicyclic hydrocarbon rings and spiro-hydrocarbon rings. However, the systems are unsaturated, i.e. there is at least one C-C double bond but no aromatic system. If in a cycloalkyl as hereinbefore defined two hydrogen atoms at adjacent cyclic carbon atoms are formally removed and the free valencies are saturated to form a second bond, the corresponding cycloalkenyl is obtained. If a cycloalkenyl is to be substituted, the substitutions may take place independently of one another, in the form of mono- or polysubstitutions in each case, on all the hydrogen-carrying carbon atoms. Cycloalkenyl itself may be linked as a substituent to the molecule via every suitable position of the ring system.
  • cycloalkenyl examples include 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, cycl
  • cycloalkenyl also applies when cycloalkenyl is part of another group as in C x - y -cycloalkenylamino or C x-y -cycloalkenyloxy, for example.
  • cycloalkenylene can thus be derived from the previously defined cycloalkenyl.
  • Cycloalkenylene unlike cycloalkenyl, is bivalent and requires two binding partners. Formally the second valency is obtained by removing a hydrogen atom from a cycloalkenyl.
  • Corresponding groups are for example cyclopentenyl and or J or * ⁇ or * ⁇ (cyclopentenylene) etc.
  • cycloalkenylene also applies when cycloalkenylene is part of another group as in HO-C x - y -cycloalkenyleneamino or H 2 N-C x -y-cycloalkenyleneoxy, for example.
  • Aryl denotes a mono-, bi- or tricyclic group with at least one aromatic carbocycle. Preferably it denotes a monocyclic group with six carbon atoms (phenyl) or a bicyclic group with nine or ten carbon atoms (two six-membered rings or one six-membered ring with a five-membered ring), wherein the second ring may also be aromatic or, however, may also be saturated or partially saturated. If an aryl is to be substituted, the substitutions may take place independently of one another, in the form of mono- or polysubstitutions in each case, on all the hydrogen-carrying carbon atoms.
  • Aryl itself may be linked as a substituent to the molecule via every suitable position of the ring system.
  • aryl examples include phenyl, naphthyl, indanyl (2,3-dihydroindenyl), indenyl, anthracenyl, phenanthrenyl, tetrahydronaphthyl (1 ,2,3,4-tetrahydronaphthyl, tetralinyl), dihydronaphthyl (1 ,2- dihydronaphthyl), fluorenyl etc.
  • the above definition of aryl also applies when aryl is part of another group as in arylamino or aryloxy, for example.
  • arylene can also be derived from the previously defined aryl.
  • Arylene unlike aryl, is bivalent and requires two binding partners. Formally, the second valency is formed by removing a hydrogen atom from an aryl.
  • Corresponding groups are e.g.
  • arylene also applies when arylene is part of another group as in HO-aryleneamino or H 2 N-aryleneoxy for example.
  • Heteroatoms may optionally be present in all the possible oxidation stages (sulphur -> sulphoxide -SO, sulphone -S0 2 -; nitrogen -> N-oxide).
  • oxidation stages sulphur -> sulphoxide -SO, sulphone -S0 2 -; nitrogen -> N-oxide.
  • heterocyclyl there is no heteroaromatic ring, i.e. no heteratom is part of an aromatic system.
  • heterocyclyl is made up of the subgroups monocyclic heterorings, bicyclic heterorings, tricyclic heterorings and spiro-heterorings, which may be present in saturated or unsaturated form.
  • unsaturated is meant that there is at least one double bond in the ring system in question, but no heteroaromatic system is formed.
  • bicyclic heterorings two rings are linked together so that they have at least two (hetero)atoms in common.
  • spiro-heterorings a carbon atom (spiroatom) belongs to two rings together.
  • heterocyclyl If a heterocyclyl is substituted, the substitutions may take place independently of one another, in the form of mono- or polysubstitutions in each case, on all the hydrogen- carrying carbon and/or nitrogen atoms. Heterocyclyl itself may be linked as a substituent to the molecule via every suitable position of the ring system.
  • heterocyclyl examples include tetrahydrofuryl, pyrrolidinyl, pyrrolinyl, imidazolidinyl, thiazolidinyl, imidazolinyl, pyrazolidinyl, pyrazolinyl, piperidinyl, piperazinyl, oxiranyl, aziridinyl, azetidinyl, 1 ,4-dioxanyl, azepanyl, diazepanyl, morpholinyl, thiomorpholinyl, h om om orph ol i nyl , h omopi peri d i nyl , h om opi perazi nyl , h omoth i om orph ol i nyl , thiomorpholinyl-S-oxide, thiomorpholinyl-S,S-dioxide,
  • heterocyclyl also applies if heterocyclyl is part of another group as in heterocyclylamino or heterocyclyloxy for example.
  • heterocyclylene is also derived from the previously defined heterocyclyl.
  • Heterocyclylene unlike heterocyclyl, is bivalent and requires two binding partners. Formally, the second valency is obtained by removing a hydrogen atom from a heterocyclyl.
  • Corresponding groups are for example piperidinyl and or or 2,3-dihydro-1 H-pyrrolyl and H or or » or " etc.
  • heterocyclylene also applies if heterocyclylene is part of another group as in HO-heterocyclyleneamino or H 2 N-heterocyclyleneoxy for example.
  • Heteroaryl denotes monocyclic heteroaromatic rings or polycyclic rings with at least one heteroaromatic ring, which compared with the corresponding aryl or cycloalkyl (cycloalkenyl) 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, wherein the resulting group must be chemically stable.
  • the prerequisite for the presence of heteroaryl is a heteroatom and a heteroaromatic system. If a heteroaryl is to be substituted, the substitutions may take place independently of one another, in the form of mono- or polysubstitutions in each case, on all the hydrogen- carrying carbon and/or nitrogen atoms.
  • Heteroaryl itself may be linked as a substituent to the molecule via every suitable position of the ring system, both carbon and nitrogen.
  • heteroaryl examples include furyl, thienyl, pyrrolyl, oxazolyl, thiazolyl, isoxazolyl, isothiazolyl, pyrazolyl, imidazolyl, triazolyl, tetrazolyl, oxadiazolyl, thiadiazolyl, pyridyl, pyrimidyl, pyridazinyl, pyrazinyl, triazinyl, pyridyl-/V-oxide, pyrrolyl-/V-oxide, pyrimidinyl-A- oxide, pyridazinyl-/V-oxide, pyrazinyl-/V-oxide, imidazolyl-/V-oxide, isoxazolyl-/V-oxide, oxazolyl-/V-oxide, thiazolyl-/V-oxide, oxadiazolyl-/V-oxide, thiadia
  • heteroaryl also applies when heteroaryl is part of another group as in heteroarylamino or heteroaryloxy, for example.
  • heteroarylene can therefore be derived from the previously defined heteroaryl.
  • Heteroarylene unlike heteroaryl, is bivalent and requires two binding partners. Formally, the second valency is obtained by removing a hydrogen atom from a heteroaryl.
  • Corresponding groups are for ex pyrrolyl and or or or etc.
  • the above definition of heteroarylene also applies when heteroarylene is part of another group as in HO-heteroaryleneamino or H 2 N-heteroaryleneoxy, for example.
  • substituted By substituted is meant that a hydrogen atom which is bound directly to the atom under consideration, is replaced by another atom or another group of atoms (substituent). Depending on the starting conditions (number of hydrogen atoms) mono- or polysubstitution may take place on one atom. Substitution with a particular substituent is only possible if the permitted valencies of the substituent and of the atom that is to be substituted correspond to one another and the substitution leads to a stable compound (i .e. to a compound which is not converted spontaneously, e.g. by rearrangement, cyclisation or elimination).
  • substitution may be carried out by a bivalent substituent only at ring systems and requires replacement by two geminal hydrogen atoms, i.e. hydrogen atoms that are bound to the same carbon atom that is saturated prior to the substitution. Substitution by a bivalent substituent is therefore only possible at the group -CH 2 - or sulphur atoms of a ring system.
  • Stereochemistry/Solvates/Hydrates Unless stated otherwise a structural formula given in the description or in the claims or a chemical name refers to the corresponding compound itself, but also encompasses the tautomers, stereoisomers, optical and geometric isomers (e.g. enantiomers, diastereomers, EIZ isomers, etc.), racemates, mixtures of separate enantiomers in any desired combinations, mixtures of diastereomers, mixtures of the forms mentioned hereinbefore (if such forms exist) as well as salts, particularly pharmaceutically acceptable salts thereof.
  • the compounds and salts according to the invention may be present in solvated form (e.g. with pharmaceutically acceptable solvents such as e.g. water, ethanol etc.) or in unsolvated form. Generally, for the purposes of the present invention the solvated forms, e.g. hydrates, are to be regarded as of equal value to the unsolvated forms.
  • salts The term "pharmaceutically acceptable” is used herein to denote compounds, materials, compositions and/or formulations which are suitable, according to generally recognised medical opinion, for use in conjunction with human and/or animal tissue and do not have or give rise to any excessive toxicity, irritation or immune response or lead to other problems or complications, i.e. correspond overall to an acceptable risk/benefit ratio.
  • pharmaceutically acceptable salts relates to derivatives of the chemical compounds disclosed in which the parent compound is modified by the addition of acid or base.
  • pharmaceutically acceptable salts include (without being restricted thereto) salts of mineral or organic acids in relation to basic functional groups such as for example amines, alkali metal or organic salts of acid functional groups such as for example carboxylic acids, etc.
  • salts include in particular acetate, ascorbate, benzenesulphonate, benzoate, besylate, bicarbonate, bitartrate, bromide/hydrobromide, Ca-edetate/edetate, camsylate, carbonate, chloride/hydrochloride, citrate, edisylate, ethane disulphonate, estolate, esylate, fumarate, gluceptate, gluconate, glutamate, glycolate, glycollylarsnilate, hexylresorcinate, hydrabamine, hydroxymaleate, hydroxynaphthoate, iodide, isothionate, lactate, lactobionate, malate, maleate, mandelate, methanesulphonate, mesylate, methylbromide, methylnitrate, methylsulphate, mucate, napsylate, nitrate, oxalate, pamoate, pan
  • salts may be formed with cations of metals such as aluminium, calcium, lithium, magnesium, potassium, sodium, zinc, etc. (cf. also Pharmaceutical salts, Birge, S.M. et al., J. Pharm. Sci., (1977), 66, 1 -19).
  • the pharmaceutically acceptable salts of the present invention may be prepared starting from the parent compound which carries a basic or acidic functionality, by conventional chemical methods. Generally, such salts may be synthesised by reacting the free acid or base form of these compounds with a sufficient amount of the corresponding base or acid in water or an organic solvent such as for example ether, ethyl acetate, ethanol, isopropanol, acetonitrile (or mixtures thereof).
  • an organic solvent such as for example ether, ethyl acetate, ethanol, isopropanol, acetonitrile (or mixtures thereof).
  • Salts of acids other than those mentioned above, which are useful for example for purifying or isolating the compounds from the reaction mixtures (e.g. trifluoroacetates), are also to be regarded as part of the invention.
  • the letter A has the function of a ring designation in order to make it easier, for example, to indicate the attachment of the ring in question to other rings.
  • Groups or substituents are frequently selected from among a number of alternative groups/ substituents with a corresponding group designation (e.g. R a , R b etc). If such a group is used repeatedly to define a compound according to the invention in different molecular parts, it must always be borne in mind that the various uses are to be regarded as totally independent of one another.
  • a therapeutically effective amount for the purposes of this invention is meant a quantity of substance that is capable of obviating symptoms of illness or of preventing or alleviating these symptoms, or which prolong the survival of a treated patient.
  • Microwave reactions are carried out in an initiator/reactor made by Biotage or in an Explorer made by CEM in sealed containers (preferably 2, 5 or 20 ml_), preferably with stirring.
  • Thin layer chromatography is carried out on ready-made TLC plates of silica gel 60 on glass (with fluorescence indicator F-254) made by Merck.
  • HPLC high pressure chromatography
  • the com pou nd s are el uted u si n g eith er d ifferent grad ients of H 2 0/acetonitrile or H 2 0/MeOH, wherein either 0.1 % HCOOH is added to the water (acid conditions).
  • H 2 0/acetonitrile gradients are also used, and the water is made basic according to the following recipe: 5 mL of an ammonium hydrogen carbonate solution (158 g to 1 L H 2 0) and 2 mL ammonia (7M in MeOH) are made up to 1 L with H 2 0.
  • HPLC normal-phase preparative high pressure chromatography
  • the analytical HPLC (reaction monitoring) of intermediate compounds is carried out with columns made by Agilent, Waters and Phenomenex.
  • the analytical equipment is also provided with a mass detector in each case.
  • UVA is-155
  • UVA is-155
  • MS Agilent LC/MSD SL Column: Agilent Zorbax SB C8, 3.5 ⁇ , 2.1 x 50 mm, Part. No.871700-906 Eluant: A: H 2 0 + 0.2 % HCOOH; B: acetonitrile (HPLC grade) + 0.2 % HCOOH Detection: MS: Positive and negative mode
  • the compounds according to the invention are prepared by the methods of synthesis described hereinafter, in which the substituents of the general formulae have the meanings given hereinbefore. These methods are intended as an illustration of the invention, without restricting its subject matter and the scope of the compounds claimed to these examples. Where the preparation of starting compounds is not described, they are commercially obtainable or may be prepared analogously to known compounds or methods described herein. Substances described in the literature are prepared according to the published methods of synthesis.
  • the method of synthesising the nitrogen-containing heterocyclic carboxylic acids A depends on the number and position of the cyclic nitrogens in the ring Q b .
  • the grouping Q a -CR 1 R 2 - may be incorporated by nucleophilic substitution at the component A.b, which is activated by an electron-withdrawing leaving group LG, e.g. a halogen, triflate or mesylate.
  • LG e.g. a halogen, triflate or mesylate.
  • A.a is optionally deprotonated for this purpose by the addition of a base.
  • the pyrimidinonecarboxylic acid ester A.a obtained is reacted in a nucleophilic substitution with A.b and subsequently saponified to form the desired carboxylic acid A (WO 2010/0071 14, WO 2010/0071 16).
  • the derivatives used are di- and trielectrophiles, which cyclise when reacted with amines A.c (WO 2010/0071 14, WO 2010/0071 16).
  • the group LG is not a leaving group in the true sense but an electrophilic group, particularly a carbonyl, to which the hydrazine is added.
  • Malonic acid diester derivative A.d.1 (5.00 g, 25.0 mmol) is taken up in 40 mL 2-butanol, cooled to 5°C and mixed with amine A.c.1 (3.00 mL, 25.1 mmol), which has been taken up in 10 mL 2-butanol.
  • the reaction mixture is stirred for 1 h at 20°C, diluted with a further 150 mL of 2-butanol and heated to 100°C for 48 h. Then the reaction mixture is combined with 50 mL 2 N aqueous sodium hydroxide solution and 50 mL 2 N methanolic sodium hydroxide solution and stirred for 2 h at 20°C.
  • A.34 A.35 Carboxylic acid A.34 (4.00 g, 13.0 mmol) is placed in MeOH (40 mL), combined with KtBuO (30 %, 3.80 g, 32.0 mmol) and heated to 160°C for 12 min. The reaction mixture is combined with HCI (20 mL, 1 N) and DCM (30 mL), the organic phase is separated off and extracted with HCI (2 x 20 mL, 1 N).
  • the amides C required are synthesised by coupling a propargylamine B with the carboxylic acid A.
  • the amide coupling is assisted by coupling reagents such as for example DCC, DIC, TBTU, HATU, EDC or the like, or the formation of the corresponding acid chloride.
  • Carboxylic acid A.25 (1 .00 g, 3.75 mmol) is placed in DCM (10 mL), combined with TBTU (1 .73 g, 5.36 mmol), propargylamine B.1 (0.29 mL, 4.10 mmol) and NEt 3 (1 .3 mL, 9.5 mmol) and stirred for 12 h at 20°C.
  • the reaction mixture is combined with water and DCM, the organic phase is separated off and extracted 2 x with water.
  • Propargylamides C.2 - C.43 are prepared analogously to propargylamide C.1 (Table 5).
  • the ring systems Q H of compounds according to the invention can be incorporated in the target structures by various methods.
  • D* which are activated by an electron-withdrawing group EWG, particularly chlorine, bromine or iodine, and a further group which enables the substituents R 5 , R 6 , R 7 and R 8 to be inserted in their respective positions
  • EWG electron-withdrawing group
  • first of all the substituent R 5 , R 6 , R 7 or R 8 can be introduced and in this way the component D is obtained (Q H* -> Q H ).
  • D is then reacted in a SONOGASHIRA reaction with C to form the compound according to the invention (1 ) (cf. also reaction scheme D).
  • first D* may be reacted with C in the SONOGASHIRA reaction and only then is the substituent R 5 , R 6 , R 7 or R 8 introduced (cf. also reaction scheme F).
  • the position of the group EWG on the ring system Q H /Q H* denotes the linkage point of the ring system to the linker unit, i.e. in this case to the C-C triple bond.
  • Q H -2c e.g. WO 2007/104560; WO 2008/020302; WO 2006/071017
  • Anthranilic acid ED.3 (32.5 g, 21 1 mmol) is placed in glacial acetic acid (400 mL), and ICI (35.0 g, 21 1 mmol) in glacial acetic acid (100 mL) is added dropwise thereto.
  • the reaction mixture is stirred for 1 h at 20°C, then diluted again with glacial acetic acid (250 mL) and stirred for 1 h at 20°C.
  • Quinazolone Z.4 (451 mg, 1 .51 mmol) is combined with DIPEA (0.5 m L) and POCI 3 (2.5 mL, 26.7 mmol) and heated to 125°C for 1 .5 h in the MW reactor. Then the reaction mixture is evaporated to dryness and quinazoline D*.1 (456 mg, 100 %) is obtained.
  • Pyridine Z.6 (500 mg, 2.91 mmol) is taken up in triethylorthoformate (10 mL) and heated to 145°C for 18 h. Then the reaction mixture is cooled to 20°C, the precipitate is filtered off, washed with Et 2 0 (10 mL), dried and azaquinazolone Z.7 (433 mg, 82 %) is obtained. Azaquinazolone Z.7 (200 mg, 1 .10 mmol) is placed in toluene (5.0 mL), combined with ⁇ /,/V-diethylaniline (0.27 mL) and POCI 3 (0.88 mL) and heated to 1 10°C for 4 h.
  • reaction mixture is cooled to 20°C, diluted with toluene (5 mL) and H 2 0 (5 mL), washed with NaOH (3 x 10 mL, 20 %) and HCI (2 x 10 mL, 1 M), dried on Na 2 S0 4 , filtered, freed from the solvent and azaquinazoline D*.2 (165 mg, 75 %) is obtained.
  • Anthranilic acid ED.5 (1 .00 g, 4.27 mmol) is placed in 2-methoxyethanol (2.0 mL), combined with formamidine acetate (890 mg, 8.55 mmol) and heated to 125°C for 20 h.
  • the reaction mixture is added to aqueous NH 3 (200 mL, 0.01 M), the precipitate is filtered off, dried and quinazolone Z.8 (817 mg, 79 %) is obtained.
  • Quinazolone Z.8 (817 mg, 3.36 mmol) is combined with SOCI 2 (20 mL) and DMF (0.05 mL) and heated to 80°C for 2 h. The solvent is removed and quinazoline D*.3 (879 mg, 100 %) is obtained.
  • Ethoxyacrylate ED.13 (6.4 mL, 44.3 mmol) is placed in MeOH (120 mL), combined with HCI (88 mL, 2M in H 2 0) and heated to 100°C.
  • Benzaldehyde ED.12 (4.00 g, 20.1 mmol) is placed in MeOH (120 mL), slowly added dropwise to the reaction mixture and the reaction mixture is heated to 100°C for 4 h.
  • the carboxylic acid Z.9 is taken up in DMF (0.5 mL) and stirred with S0 2 CI 2 (5.0 mL) for 40 min at 20°C. The solvent is removed and the acid chloride Z.10 (495 mg, 100 %) is further reacted directly.
  • Compound D.61 is prepared analogously to quinoline D.60 (Table 10):
  • Anthranilic acid ED.18 (5.06 g, 19.2 mmol) is stirred in cone. HCI (7.0 mL) for 12 h at 20°C.
  • nitromethane (2.56 g, 42.0 mmol) is added dropwise to a mixture of ice (6.0 g, 333 mmol) and NaOH (2.53 g, 63.4 mmol), stirred for 1 h at 0°C, then heated to 20°C over 1 h , then poured onto an ice/HCI mixture (5 g/7 mL) and combined with the aniline-HCI mixture.
  • the reaction mixture is stirred for 12 h at 20°C, the precipitate is filtered off, washed with H 2 0 and dried.
  • the precipitate is taken up in acetic anhydride (35 mL), combined with KOAc (2.19 g, 22.3 mmol) and heated to 120°C for 2 h. The solvent is removed and the residue is washed with glacial acetic acid and H 2 0 and dried.
  • the electron withdrawing groups EWG may be in particular halogen (chlorine, bromine, iodine), triflate or mesylate (cf. also reaction scheme C).
  • Electron-withdrawing groups EWG that may be used include in particular halogen (chlorine, bromine, iodine), triflate or mesylate (cf. also reaction scheme C).

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Public Health (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)

Abstract

The present invention encompasses compounds of general formula (1) wherein the groups R1 to R4, Qa, Qb, QH, L and n are defined as in claim 1, which are suitable for the treatment of diseases characterised by excessive or abnormal cell proliferation, pharmaceutical preparations which contain such compounds and their use as medicaments.

Description

HETEROCYCLIC CARBOXYLIC ACID AMIDES AS PDK1 INIHIBITORS
The present invention relates to new quinoxaline, quinoline and quinazoline compounds of general formula (1 )
Figure imgf000002_0001
(1 )
wherein the groups R1 to R4, Qa, Qb, QH, L and n have the meanings given in the claims and specification, which are suitable for the treatment of diseases characterised by excessive or abnormal cell proliferation, pharmaceutical preparations containing such compounds and their use as medicaments. The compounds according to the invention exhibit an inhibitory effect on the serine/threonine kinase PDK 1 Background to the invention
Quinazolines as PDK1 inhibitors are described for example in WO 2010/0071 14 and WO 2010/0071 16.
The aim of the present invention is to indicate new compounds which can be used for the prevention and/or treatment of diseases characterised by excessive or abnormal cell proliferation. The compounds according to the invention are characterised by a powerful inhibitory effect on PDK 1 and a high efficacy against tumour cells, e.g. prostate carcinoma cells, which is mediated through inhibiting PDK 1 . As well as the inhibitory effect and cell potency the compounds have good solubility and good selectivity with regard to other signal and cell cycle kinases. The importance of the PI3K-PDK1 -AKT pathway with its frequent aberrations (PTEN loss, PI3K mutation) in the major tumor indications is described in numerous scientific publications (e.g. Samuels et al. 2004; Science; 304: 554; Samuels and Velculescu 2004; Cell cycle 3; 17-19; Samuels et al. 2005; Cancer Cell; 7: 561 -573).
PDK1 alterations (mutations, CN gains) are rarely found isolated from other pathway alterations so that the importance of this target thrives from the fact that it acts as a central signaling node in a frequently altered pathway.
Active PI3K phosphorylates second messengers like Phosphatidylinositol(4.5)- bisphosphate (Ptdlns(4.5)P2) to Ptdlns(3.4.5)P3, or Ptdlns(4)P to Ptdlns(3.4,)P2. Such PI3K products recruit AKT and PDK1 kinases via their PH-domains (which bind the PI3 kinase products) to the plasma membrane and allow PDK1 to phosphorylate Thr308 of AKT and thereby activate the enzyme. A second step important for additional AKT activation and/or AKT-substrate selection is mediated by the mTOR/rictor/SIN1 complex which phosphorylates the hydrophobic motif of AKT at Ser473. Activated AKT regulates several other proteins, which are involved in cell proliferation, growth and survival.
Besides AKT, PDK1 has other substrates located in the cytoplasm, e.g. p70S6K, p90RSK, PKCs and SGK, which are also implicated in regulating cell proliferation and cell survival. The activation of these kinases is not dependent on phosphatidylinositols in vitro and functions properly in cells where the PDK1 -PH-domain had been destroyed by knock-in mutation (McManus et al. 2004, EMBO J. 23, 2071 -2082). For a review on PDK1 mouse mutants also see Bayascas 2008; Cell cycle 7; 2978-2982).
In conclusion, PDK1 substrates (AKT, p90RSK) act at the crossroad of two important cancer signaling pathways.
A potent and selective inhibitor of the PDK1 kinase is predicted to inhibit tumor growth, delay relapse, or even induce objective tumor responses (i.e. complete or partial tumor shrinkage) in cancer patients.
Detailed description of the invention
It has been found that, surprisingly, compounds of general formula (1 ) wherein the groups R1 to R4, Qa, Qb, QH, L and n are defined as stated hereinafter act as inhibitors of specific signal enzymes which are involved in controlling the proliferation of cells. Thus, the compounds according to the invention may be used for example for the treatment of diseases which are connected with the activity of these signal enzymes and are characterised by excessive or abnormal cell proliferation.
The present invention therefore relates to compounds of general formula (1 )
Figure imgf000004_0001
(1 ) , wherein
(AO)
Qa is a ring system optionally substituted by one or more identical or different Ra and/or Rb, selected from among C3-iocycloalkyl, C6-ioaryl, 5-12 membered heteroaryl and 3-14 membered heterocyclyl;
(BO)
R1 and R2 are selected independently of one another from among hydrogen, halogen, Chalky I , C2-4alkenyl, C2-4alkynyl, Ci-4haloalkyl, -NH2, -CN, -NHCi-4alkyl, -N(Ci-4alkyl)2, -OH, -OCi-4alkyl, HO-Ci-4alkylene, Ci-4alkyl-0-Ci-4alkylene, H2N-Ci-4alkylene, -OCi-4haloalkyl, (Ci-4alkyl)NH-Ci-4alkylene- and (Ci-4alkyl)2N-Ci-4alkylene, while alkyl, alkenyl, alkynyl and alkylene mentioned in the above groups may optionally be substituted by one or more identical or different halogens;
(CO)
the ring system Qb is selected from among
Figure imgf000004_0002
Qb-1 Qb-2 Qb-3 Qb-4 Qb-5 each R3 is independently selected from among halogen, Ci-4alkyl, C2-4alkenyl, C2-4alkynyl, Ci-4haloalkyl, -NH2, -CN, -NHCi-4alkyl, -N(C1-4alkyl)2, -OH, -OCi-4alkyl, HO-Ci-4alkylene, HO-C2-4alkylene-0, Ci-4alkyl-0-Ci-4alkylene, Ci-4haloalkyl-0-Ci-4alkylene,
H2N-Ci-4alkylene, Ci-4alkyl-0-C2-4alkylene-0, (Ci-4alkyl)NH-Ci-4alkylene, (C1-4alkyl)2N- Ci-4alkylene, -OCi-4haloalkyl, H2N-C2-4alkylene-0-, -NH(C2-4alkylene-NH2),
-NH[C2-4alkylene-N(HCi-4alkyl)], -NH[C2-4alkylene-N(Ci-4alkyl)2],
(Ci-4alkyl)2N-C2-4alkylene-0-, (Ci-4haloalkyl)2N-C2-4alkylene-0-, (Ci-4haloalkyl)NH-C2-4alkylene-0- and (Ci-4alkyl)NH-C2-4alkylene-0-;
n denotes the number 0, 1 , 2 or 3 if Qb corresponds to the ring system Qb-4;
each n independently denotes the number 0, 1 or 2 if Qb corresponds to one of the ring systems Qb-1 , Qb-2, Qb-3 or Qb-5;
while a group R3 in the ring systems Qb-1 to Qb-5 in each case replaces a hydrogen;
(DO)
R4 denotes hydrogen or Ci-4alkyl;
(E0)
L denotes the group
Figure imgf000005_0001
and in the latter case the groups
-CR5R6- and QH may assume both a c/'s and a trans configuration with respect to the double bond;
R5 and R6 are each independently of one another selected from among hydrogen, halogen, Ci-4alkyl, Ci-4haloalkyl;
(F0)
the ring system QH is selected from among
Figure imgf000006_0001
Figure imgf000006_0002
R7 and R9 are each independently of one another selected from among hydrogen, halogen, -NRaRa, -N(ORa)Ra, -ORa, -COORa, -CORa, -CONRaRa, -SRa, -N02 and -CN or denote a group optionally substituted by one or more identical or different Rb and/or Rc, selected from among Ci-6alkyl , C2-6alkenyl, C2-6alkynyl, C3-iocycloalkyl, C6-ioaryl, 5-12 membered heteroaryl and 3-14 membered heterocyclyl;
R8 is independently selected from among hydrogen, -NRaRa, -N(ORa)Ra, -ORa, -COORa, -CORa, -CONRaRa and -SRa or denotes a group optionally substituted by one or more identical or different Rb and/or Rc, selected from among Ci-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-iocycloalkyl, C6-ioaryl, 5-12 membered heteroaryl and 3-14 membered heterocyclyl;
R10 is independently selected from among -NRaRa, -N(ORa)Ra, -ORa and -SRa or denotes a group optionally substituted by one or more identical or different Rb and/or Rc selected from among Ci-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-iocycloalkyl, C6-ioaryl, 5-12 membered heteroaryl and 3-14 membered heterocyclyl;
(GO)
each R11 and R12 is independently selected from among Ra and Rb;
m denotes the number 0, 1 or 2; each Ra independently denotes hydrogen or a group optionally substituted by one or more identical or different Rb and/or Rc, selected from among Ci-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-iocycloalkyl, C6-ioaryl, 5-12 membered heteroaryl and 3-14 membered heterocyclyl; each Rb is independently selected from among -ORc, -NRCRC, halogen, -CN, -N02, -C(0)Rc, -C(0)ORc, -C(0)NRcRc, -S(0)2Rc, -S(0)2NRcRc, -NHC(0)Rc and -N(Ci-4alkyl)C(0)Rc as well as the bivalent substituent =0, while the latter may only be a substituent in non-aromatic ring systems;
each Rc independently denotes hydrogen or a group optionally substituted by one or more identical or different Rd and/or Re, selected from among Ci-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-iocycloalkyl, C6-ioaryl, 5-12 membered heteroaryl and 3-14 membered heterocyclyl; each Rd is independently selected from among -ORe, -NReRe, halogen, -CN, -N02, -C(0)Re, -C(0)ORe, -C(0)NReRe, -S(0)2Re, -S(0)2NReRe, -NHC(0)Re and -N(Ci-4alkyl)C(0)Re, as well as the bivalent substituent =0, while the latter may only be a substituent in non-aromatic ring systems;
each Re independently denotes hydrogen or a group optionally substituted by one or more identical or different Rf and/or Rg, selected from among Ci-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-iocycloalkyl, C6-ioaryl, 5-12 membered heteroaryl and 3-14 membered heterocyclyl; each Rf is independently selected from among -OR9, -NR9R9, halogen, -CN, -N02, -C(0)R9, -C(0)OR9, -C(0)NR9R9, -S(0)2R9, -S(0)2NR9R9, -NHC(0)R9 and -N(Ci-4alkyl)C(0)R9, as well as the bivalent substituent =0, while the latter may only be a substituent in non-aromatic ring systems and
each Rg is independently selected from among hydrogen, Ci-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-iocycloalkyl, C6-ioaryl, 5-12 membered heteroaryl and 3-14 membered heterocyclyl and the above-mentioned groups R9 may optionally be substituted by one or more identical or different substituents, selected independently of one another from among -NH2, -NH(Ci-6alkyl), -N(d-6alkyl)2 and Ci-6alkyl;
while the compounds (1 ) may optionally also be present in the form of the tautomers, racemates, enantiomers, diastereomers or mixtures thereof or as the respective salts of all the above-mentioned forms.
In one aspect (A1 ) the invention relates to compounds (1 ), wherein
Qa is a ring system optionally substituted by one or more identical or different Ra and/or Rb selected from among C5-6cycloalkyl, phenyl, 5-6 membered heteroaryl and 5-7 membered heterocyclyl, and
Ra and Rb are as hereinbefore defined. In another aspect (A2) the invention relates to compounds (1 ), wherein
Qa is a ring system optionally substituted by one or more identical or different Ra and/or Rb selected from among phenyl and 5-6 membered heteroaryl, and
Ra and Rb are as hereinbefore defined. In other aspects (A3)(A4) the invention relates to compounds with a structural aspect (A1 ) and (A2), wherein the ring system Qa optionally carries one or more identical or different substituents, selected from among halogen and Ci-4alkyl.
In another aspect (A5) the invention relates to compounds (1 ) wherein
Qa is selected from among phenyl, 3-fluorophenyl, 4-fluorophenyl, 3,4,5-trifluorophenyl, 3,5-difluorophenyl and 3,4-difluorophenyl.
In another aspect (A6) the invention relates to compounds (1 ) wherein
Qa denotes 3,4-difluorophenyl.
In another aspect (B1 ) the invention relates to compounds (1 ) wherein
R1 and R2 are selected independently of one another from among hydrogen, Ci-4alkyl, C2-4alkynyl, -CN, HO-Ci-4alkylene-, H2N-Ci-4alkylene-, (Ci-4alkyl)NH-Ci-4alkylene-,
(Ci-4halolkyl)NH-Ci-4alkylene-, (Ci-4alkyl)2N-Ci-4alkylene- and
(Ci-4haloalkyl)2N-Ci-4alkylene-.
In another aspect (B2) the invention relates to compounds (1 ), wherein
R1 and R2 are selected independently of one another from among hydrogen, Ci-4alkyl, H2N-Ci-4alkylene and HO-Ci-4alkylene.
In another aspect (B3) the invention relates to compounds (1 ), wherein
R1 and R2 are independently selected from among hydrogen, methyl, aminomethyl and hydroxymethyl.
In further aspects (B4)(B5)(B6) the invention relates to compounds with a structural aspect (B1 ), (B2) and (B3), wherein
R1 denotes hydrogen.
In another aspect (C1 ) the invention relates to compounds (1 ), wherein the ring system Qb is selected from among
Figure imgf000009_0001
Qb-1 Qb-2 Qb-3 Qb-4 Qb-5 each R3 is independently selected from among halogen, Ci-4alkyl, -OCi-4alkyl, HO-Ci-4alkylene, Ci-4alkyl-0-Ci-4alkylene, -NH(C2-4alkylene-NH2), -NH[C2-4alkylene-NH(Ci-4alkyl)] and -NH[C2-4alkylene-N(Ci-4alkyl)2];
n denotes the number 0, 1 , 2 or 3 if Qb corresponds to the ring system Qb-4 and each n independently denotes the number 0, 1 or 2 if Qb corresponds to one of the ring systems Qb-1 , Qb-2, Qb-3 or Qb-5,
while a group R3 in the ring systems Qb-1 to Qb-5 in each case replaces a hydrogen.
In another as ect (C2) the invention relates to compounds (1 ), wherein
the ring system Qb is selected from among
Figure imgf000009_0002
n has the value 0. In another aspect (C3) the invention relates to compounds (1 ), wherein
the ring system Qb is selected from among
Figure imgf000009_0003
Qb-2 and Qb-4 n has the value 0.
In another aspect (C4) the invention relates to compounds (1 ), wherein
the ring system Qb to m among
Figure imgf000010_0001
In another aspect (D1 ) the invention relates to compounds (1 ), wherein
R4 denotes hydrogen or methyl.
In another aspect (D2) the invention relates to compounds (1 ), wherein
R4 denotes hydrogen.
In another aspect (E1 ) the invention relates to compounds (1 ), wherein
L denotes the group
Figure imgf000010_0002
In another aspect (F1 ) the invention relates to compounds (1 ), wherein
the ring system QH is selected from among
Figure imgf000010_0003
QH-2a and 2 QH-2d R7 is independently selected from among hydrogen, -NRaRa, -N(ORa)Ra, -ORa, -C00R3, -CORa, -CONRaRa and -SRa, or denotes a group optionally substituted by one or more identical or different Rb and/or Rc, selected from among Ci-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-iocycloalkyl, C6-ioaryl, 5-12 membered heteroaryl and 3-14 membered heterocyclyl, and
R11, R12, Ra, Rb, Rc and m are as hereinbefore defined. In another aspect (F2) the invention relates to compounds (1 ), wherein the ring system QH is selected from among
Figure imgf000011_0001
QH-2a and 2 QH-2d
R7 is independently selected from among hydrogen, -NRa1Ra1, -N(ORa1)Ra1, -ORa1, -COORa1, -CORa1, -CONRa1Ra1 and -SRa1 or denotes a group optionally substituted by one or more identical or different Rb1 and/or Rc1 selected from among Ci-6alkyl, C2-6alkenyl , C2-6alkynyl, C3-iocycloalkyl, C6-ioaryl, 5-12 membered heteroaryl and 3-14 membered heterocyclyl;
each Ra1 independently denotes hydrogen or a group optionally substituted by one or more identical or different Rb1 and/or Rc1, selected from among Ci-6alkyl, C3-i0cycloalkyl and 3-14 membered heterocyclyl;
each Rb1 is independently selected from among -ORc1, -NRc1Rc1 and halogen;
each Rc1 independently denotes hydrogen or a group optionally substituted by one or more identical or different Rd1 and/or Re1 selected from among Ci-6alkyl, C3-i0cycloalkyl and 3-14 membered heterocyclyl;
each Rd1 is independently selected from among -ORe1, -CN and -C(0)ORe1;
each Re1 independently denotes hydrogen or a group optionally substituted by one or more identical or different cycloalkyl (particularly C3-6cycloalkyl) selected from among Ci-6alkyl and C3-i0cycloalkyl, and
R11, R12 and m are as hereinbefore defined.
In another aspect (F3) the invention relates to compounds (1 ), wherein
the ring system QH denotes
Figure imgf000012_0001
QH-2a
R7 is selected from among hydrogen, -NRa1Ra1, -ORa1 and -SRa1, or denotes a group optionally substituted by one or more identical or different Rb1 and/or Rc1, selected from among Ci-6alky I , C2-6alkenyl, pyrrolidinyl, piperazinyl, piperidinyl, morpholinyl, homomorpholinyl, azetidinyl and homopiperazinyl;
each Ra1 independently denotes hydrogen or a group optionally substituted by one or more identical or different Rb1 and/or Rc1, selected from among Ci-6alkyl, C3-6cycloalkyl, piperazinyl, piperidinyl, morpholinyl, azetidinyl and pyrrolidinyl;
each Rb1 is independently selected from among -ORc1, -NRc1Rc1 and halogen;
each Rc1 independently denotes hydrogen or a group optionally substituted by one or more identical or different Rd1 and/or Re1 selected from among Ci-6alkyl, C3-6cycloalkyl, pyrrolidinyl, morpholinyl, homomorpholinyl, piperazinyl, homopiperazinyl, oxetanyl and piperidinyl;
each Rd1 is independently selected from among -ORe1, -CN and -C(0)ORe1;
each Re1 independently denotes hydrogen or a group optionally substituted by one or more identical or different C3-6cycloalkyl, selected from among Ci-6alkyl and C3-6cycloalkyl, and
R11 and m are as hereinbefore defined.
In another aspect (F4) the invention relates to compounds (1 ), wherein
the ring system QH denotes
Figure imgf000012_0002
QH-2a R7 denotes -NHRa1;
Ra1 is a group optionally substituted by one or more identical or different Rb1 and/or Rc1, selected from among Ci-4alkyl, cyclohexyl, cyclopentyl and piperidinyl;
each Rb1 is independently selected from among -NRc1Rc1 and fluorine;
each Rc1 independently denotes hydrogen or a group optionally substituted by one or more identical or different Rd1 and/or Re1, selected from among Ci-4alkyl, cyclopropyl, morpholinyl, piperazinyl and oxetanyl;
each Rd1 denotes -ORe1;
each Re1 independently denotes hydrogen or a Ci-4alkyl optionally substituted by cyclopropyl, and
R11 and m are as hereinbefore defined.
In another aspect (F5) the invention relates to compounds (1 ), wherein
the ring system QH denotes
Figure imgf000013_0001
QH-2a R7 is selected from among
Figure imgf000013_0002
Figure imgf000014_0001
R11 and m are as hereinbefore defined.
In another aspect (F6) the invention relates to compounds (1 ), wherein
the ring system QH is selected from among
Figure imgf000014_0002
QH-2b and QH-2e
R9 is independently selected from among hydrogen, -NRaRa, -N(ORa)Ra, -ORa, -C00R3, -CORa, -CONRaRa and -SRa, or denotes a group optionally substituted by one or more identical or different Rb and/or Rc, selected from among Ci-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-iocycloalkyl, C6-ioaryl, 5-12 membered heteroaryl and 3-14 membered heterocyclyl, and R11, R12, Ra, Rb, Rc and m are as hereinbefore defined.
In another aspect (F7) the invention relates to compounds (1 ), wherein
the ring system QH is selected from among
Figure imgf000015_0001
QH-2b and QH-2e
R9 is independently selected from among hydrogen, -NRa2Ra2, -N(ORa2)Ra2, -ORa2, -COOR32, -CORa2, -CONRa2Ra2 and -SRa2, or denotes a group optionally substituted by one or more identical or different Rb2 and/or Rc2, selected from among Ci-6alkyl, C2-6alkenyl , C2-6alkynyl, C3-iocycloalkyl, C6-ioaryl, 5-12 membered heteroaryl and 3-14 membered heterocyclyl;
each Ra2 independently denotes hydrogen or a group optionally substituted by one or more identical or different Rb2 and/or Rc2 selected from among Ci-6alkyl, C3-i0cycloalkyl and 3-14 membered heterocyclyl;
each Rb2 is independently selected from among -ORc2, -NRc2Rc2 and halogen;
each Rc2 independently denotes hydrogen or a group optionally substituted by one or more identical or different Rd2 and/or Re2 selected from among Ci-6alkyl, C3-i0cycloalkyl and 3-14 membered heterocyclyl;
each Rd2 is independently selected from among -ORe2, -CN and -C(0)ORe2;
each Re2 independently denotes hydrogen or a group optionally substituted by one or more identical or different cycloalkyl (particularly C3-6cycloalkyl) selected from among Ci-6alkyl and C3-i0cycloalkyl, and
R11, R12 and m are as hereinbefore defined.
In another aspect (F8) the invention relates to compounds (1 ), wherein
the ring system QH denotes
Figure imgf000015_0002
QH-2b R9 is selected from among hydrogen, -NRa2Ra2, -ORa2 and -SRa2, or denotes a group optionally substituted by one or more identical or different Rb2 and/or Rc2 selected from among Ci-6alky I , C2-6alkenyl, pyrrolidinyl, piperazinyl, piperidinyl, morpholinyl, homomorpholinyl, azetidinyl and homopiperazinyl;
each Ra2 independently denotes hydrogen or a group optionally substituted by one or more identical or different Rb2 and/or Rc2, selected from among Ci-6alkyl, C3-6cycloalkyl, piperazinyl, piperidinyl, morpholinyl, azetidinyl and pyrrolidinyl;
each Rb2 is independently selected from among -ORc2, -NRc2Rc2 and halogen;
each Rc2 independently denotes hydrogen or a group optionally substituted by one or more identical or different Rd2 and/or Re2, selected from among Ci-6alkyl, C3-6cycloalkyl, pyrrolidinyl, morpholinyl, homomorpholinyl, piperazinyl, homopiperazinyl, oxetanyl and piperidinyl;
each Rd2 is independently selected from among -ORe2, -CN and -C(0)ORe2;
each Re2 independently denotes hydrogen or a group optionally substituted by one or more identical or different C3-6cycloalkyl, selected from among Ci-6alkyl and C3-6cycloalkyl, and
R11 and m are as hereinbefore defined.
In another aspect (F9) the invention relates to compounds (1 ), wherein
the ring system QH denotes
Figure imgf000016_0001
QH-2b
R9 denotes -NHRa2;
Ra2 is a group optionally substituted by one or more identical or different Rb2 and/or Rc2 selected from among Ci-4alkyl, cyclohexyl, cyclopentyl and piperidinyl;
each Rb2 is independently selected from among -NRc2Rc2 and fluorine;
each Rc2 independently denotes hydrogen or a group optionally substituted by one or more identical or different R and/or R , selected from among Ci-4alkyl, cyclopropyl, morpholinyl, piperazinyl and oxetanyl;
each Rd2 denotes -ORe2;
each Re2 independently denotes hydrogen or a Ci-4alkyl optionally substituted by cyclopropyl, and
R11 and m are as hereinbefore defined.
In another aspect (F10) the invention relates to compounds (1), wherein
the ring system QH denotes
Figure imgf000017_0001
R9 is selected from among
Figure imgf000017_0002
Figure imgf000018_0001
R11 and m are as hereinbefore defined.
In another aspect (F11 ) the invention relates to compounds (1 ), wherein
the ring system QH is selected from among
Figure imgf000018_0002
QH-2b and QH-2e
R is selected from among hydrogen, -NRaRa, -N(ORa)Ra, -ORa and -SRa, or denotes a group optionally substituted by one or more identical or different Rb and/or Rc, selected from among Ci-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-iocycloalkyl, C6-ioaryl, 5-12 membered heteroaryl and 3-14 membered heterocyclyl;
R9 is independently selected from among hydrogen, halogen, -N02, -CN and Ci-4alkyl, and
R11, R12, Ra, Rb, Rc and m are as hereinbefore defined.
In another aspect (F12) the invention relates to compounds (1 ), wherein
the ring system QH is selected from among
Figure imgf000019_0001
QH-2b and 2 QH-2e
R is selected from among -NR R and -OR , or denotes a group optionally substituted by one or more identical or different Rb3 and/or Rc3 selected from among Ci-6alkyl, C2-6alkenyl, C2-6alkynyl, 5-12 membered heteroaryl and 3-14 membered heterocyclyl; R9 is independently selected from among hydrogen, halogen, -N02, -CN and Ci-4alkyl; each Ra3 independently denotes hydrogen or a group optionally substituted by one or more identical or different Rb3 and/or Rc3 selected from among Ci-6alkyl, C3-i0cycloalkyl, C6-ioaryl and 3-14 membered heterocyclyl;
each Rb3 is independently selected from among -ORc3 and -NRc3Rc3;
each Rc3 independently denotes hydrogen or a group optionally substituted by one or more identical or different Ci-6alkyl selected from among Ci-6alkyl, C3-iocycloalkyl, C6-ioaryl and 3-14 membered heterocyclyl, and
R11, R12 and m are as hereinbefore defined.
In another aspect (F13) the invention relates to compounds (1 ), wherein
the ring system QH denotes
Figure imgf000019_0002
QH-2b
R is selected from among -NR R and -OR , or denotes a group optionally substituted by one or more identical or different Rb3 and/or Rc3, selected from among Ci-6alkyl, 5-6 membered heteroaryl and 4-6 membered heterocyclyl;
R9 is independently selected from among hydrogen, halogen, -N02, -CN and Ci-4alkyl; each R independently denotes hydrogen or a group optionally substituted by one or more identical or different Rb3 and/or Rc3, selected from among Ci-6alkyl, C3-6cycloalkyl, phenyl, pyrrolidinyl, and piperidinyl;
each Rb3 is independently selected from among -ORc3 and -NRc3Rc3;
each Rc3 independently denotes hydrogen or a group optionally substituted by one or more identical or different Ci-6alkyl, selected from among Ci-6alkyl, piperazinyl, piperidinyl, tetrahydropyranyl, tetrahydrofuryl and pyrrolidinyl, and
R11 and m are as hereinbefore defined.
In another aspect (F14) the invention relates to compounds (1 ), wherein
the ring system QH denotes
Figure imgf000020_0001
QH-2b
R denotes -ORa3;
R9 is independently selected from among hydrogen, halogen, -N02, -CN and Ci-4alkyl;
Ra3 denotes a Ci-6alkyl which is substituted by a substituent selected from among -N(Ci-4alkyl)2, piperidinyl, piperazinyl or pyrrolidine, while the above-mentioned piperidinyl, piperazinyl and pyrrolidine may optionally be substituted by a Ci-4alkyl and
R11 and m are as hereinbefore defined.
In another aspect (F15) the invention relates to compounds (1 ), wherein
the ring system QH denotes
Figure imgf000020_0002
QH-2b R is selected from among
Figure imgf000021_0001
R9 is independently selected from among hydrogen, halogen, -N02, -CN and Ci-4alkyl, and R11 and m are as hereinbefore defined.
In another aspect (F16) the invention relates to compounds (1 ), wherein
the ring system QH is selected from among
Figure imgf000021_0002
QH-2c and QH-2f
R10 is independently selected from among -NRaRa, -N(ORa)Ra, -ORa and -SRa or denotes a group optionally substituted by one or more identical or different Rb and/or Rc selected from among Ci-6alkyl, C2-6alkenyl , C2-6alkynyl, C3-iocycloalkyl, C6-ioaryl, 5-12 membered heteroaryl and 3-14 membered heterocyclyl, and
R11, R12, Ra, Rb, Rc and m are as hereinbefore defined.
In another aspect (F17) the invention relates to compounds (1 ), wherein
the ring system QH is selected from among
Figure imgf000022_0001
Qh-2C and QH-2f
R10 is independently selected from among -NRa4Ra4, -N(ORa4)Ra4, -ORa4 and -SRa4 or denotes a group optionally substituted by one or more identical or different Rb4 and/or Rc4 selected from among Ci-6alkyl , C2-6alkenyl, C2-6alkynyl, C3-iocycloalkyl, C6-ioaryl, 5-12 membered heteroaryl and 3-14 membered heterocyclyl;
each Ra4 independently denotes hydrogen or a group optionally substituted by one or more identical or different Rb4 and/or Rc4 selected from among Ci-6alkyl, C3-iocycloalkyl, C6-ioaryl and 3-14 membered heterocyclyl;
each Rb4 is independently selected from among -ORc4, -NRc4Rc4, -C(0)Rc4, -C(0)ORc4, -C(0)NRc4Rc4, -NHC(0)Rc4 and -N(Ci-4alkyl)C(0)Rc4;
each Rc4 independently of one another denotes hydrogen or a group optionally substituted by one or more identical or different Rd4 and/or Re4, selected from among Ci-6alkyl, C3-iocycloalkyl, C6-ioaryl, 5-12 membered heteroaryl and 3-14 membered heterocyclyl; each Rd4 denotes -ORe1;
each Re4 independently denotes hydrogen or a group optionally substituted by one or more identical or different Rf4 selected from among Ci-6alkyl and C6-ioaryl;
each Rf4 denotes halogen and
R11, R12 and m are as hereinbefore defined.
In another aspect (F18) the invention relates to compounds (1 ), wherein
the ring system QH denotes
Figure imgf000023_0001
QH-2c
R10 is selected from among -NRa4Ra4, -N(ORa4)Ra4 and -ORa4 or denotes a group optionally substituted by one or more identical or different Rb4 and/or Rc4 selected from among Ci-6alkyl, phenyl, 5-6 membered heteroaryl, piperazinyl, pyrrolidinyl, morpholinyl, homomorpholinyl, homopiperazinyl and piperidinyl;
each Ra4 independently denotes hydrogen or a group optionally substituted by one or more identical or different Rb4 and/or Rc4 selected from among Ci-6alkyl, C3-6cycloalkyl, phenyl, azetidinyl, tetrahydropyranyl, piperidinyl and pyrrolidinyl;
each Rb4 is independently selected from among -ORc4, -NRc4Rc4, -C(0)Rc4, -C(0)ORc4, -C(0)NRc4Rc4 and -NHC(0)Rc4;
each Rc4 independently denotes hydrogen or a group optionally substituted by one or more identical or different Rd4 and/or Re4 selected from among Ci-6alkyl, phenyl, furyl, pyridyl, imidazolyl, thienyl, pyrazolyl, quinolinyl, oxetanyl, tetrahydrofuryl, azetidinyl, pyrrolidinyl, piperidinyl and piperazinyl;
each Rd4 denotes -ORe4;
each Re4 independently denotes hydrogen or a group optionally substituted by one or more identical or different Rf4 selected from among Ci-6alkyl and phenyl;
each Rf4 denotes halogen and
R11 and m are as hereinbefore defined. In another aspect (F19) the invention relates to compounds (1 ), wherein
the ring system QH denotes
Figure imgf000024_0001
QH-2c
R10 is selected from among -NHMe, -NMe2 and -NEt2, and
R11 and m are as hereinbefore defined.
In another aspect (G1 ) the invention relates to compounds (1 ), wherein
each R11 and R12 is independently selected from among hydrogen, Ci-4alkyl and halogen, and
m denotes the value 0, 1 or 2.
In another aspect (G2) the invention relates to compounds (1 ), wherein
each R11 and R12 is independently selected from among hydrogen, Ci-4alkyl and halogen, and
m denotes the value 0 or 1 .
In another aspect (G3) the invention relates to compounds (1 ), wherein
m has the value 0.
All the above-mentioned structural aspects A1 to A6, B1 to B6, C1 to C4, D1 and D2, E1 , F1 to F19 and G1 to G3 are preferred embodiments of the aspects AO, B0, CO, DO, E0, F0 and GO, respectively. The structural aspects AO to A6, B0 to B6, CO to C4, DO to D2, E0 and E1 , F0 to F19 and GO to G3 relating to different molecular parts of the compounds (1 ) according to the invention may be permutated with one another as desired in combinations ABCDEFG so as to obtain preferred compounds (1 ) . Each combination ABCDEFG represents and defines individual embodiments or generic amounts of compounds AOBOCODOEOFOGO according to the invention. Each individual embodiment or partial quantity defined by this combination is expressly also included and is a subject of the invention.
The present invention further relates to all the pharmaceutically acceptable salts of compounds of general formula (1 ).
The present invention further relates to hydrates, solvates, polymorphs, metabolites, derivatives and prodrugs of compounds of general formula (1 ).
In another aspect the invention relates to compounds of general formula (1 ) - or the pharmaceutically acceptable salts thereof - as medicaments.
In another aspect the invention relates to compounds of general formula (1 ) - or the pharmaceutically acceptable salts thereof - for use in the treatment and/or prevention of cancer, infections, inflammations and autoimmune diseases.
In another aspect the invention relates to compounds of general formula (1 ) - or the pharmaceutically acceptable salts thereof - for use in the treatment and/or prevention of cancer.
In another aspect the invention relates to compounds of general formula (1 ) - or the pharmaceutically acceptable salts thereof - for use in the treatment and/or prevention of carci nomas of the breast, prostate or ovary, non-small-cell bronchial carcinomas (NSCLC), melanomas and chronic lymphatic leukaemias (CLL).
In another aspect the invention relates to a process for the treatment and/or prevention of cancer comprising administering a therapeutically effective amount of a compound of general formula (1 ) - or one of the pharmaceutically acceptable salts thereof - to a human being. In another aspect the invention relates to a pharmaceutical preparation containing as active substance one or more compounds of general formula (1 ) - or the pharmaceutically acceptable salts thereof - optionally in combination with conventional excipients and/or carriers.
In another aspect the invention relates to a pharmaceutical preparation comprising a compound of general formula (1 ) - or one of the pharmaceutically acceptable salts thereof - and at least one other cytostatic or cytotoxic active substance, different from formula (1). Definitions
Terms that are not specifically defined here have the meanings that are apparent to the skilled man in the light of the overall disclosure and the context as a whole.
As used herein, the following definitions apply, unless stated otherwise:
The use of the prefix Cx-y, wherein x and y each represent a natural number (x < y), indicates that the chains or ring structure or combination of chains and ring structure as a whole, specified and mentioned in direct association, may consist of a maximum of y and a minimum of x carbon atoms.
The indication of the number of members in groups that contain one or more heteroatom(s) (heteroalkyl, heteroaryl, heteroarylalkyl, heterocyclyl, heterocyclylalkyl) relates to the total atomic number of all the ring members or chain members or the total of all the ring and chain members.
Alkyl denotes monovalent, saturated hydrocarbon chains, which may be present in both straight-chain (unbranched) and branched form. If an alkyl is substituted, the substitution may take place independently of one another, by mono- or polysubstitution in each case, on all the hydrogen-carrying carbon atoms.
The term "C1-5-alkyl" includes for example H3C-, H3C-CH2-, H3C-CH2-CH2-,
H3C-CH(CH3)-, H3C-CH2-CH2-CH2-, H3C-CH2"CH(CH3)-, H3C-CH(CH3)-CH2-, H3C-C(CH3)2_, H3C-CH2-CH2-CH2-CH2-, H3C-CH2"CH2-CH(CH3)-, H3C-CH2-CH(CH3)-CH2-, H3C-CH(CH3)-CH2-CH2-, H3C-CH2-C(CH3)2-, H3C-C(CH3)2-CH2-, H3C-CH(CH3)-CH(CH3)- and H3C-CH2-CH(CH2CH3)-.
Further examples of alkyl are methyl (Me; -CH3), ethyl (Et; -CH2CH3), 1 -propyl (n-propyl; n-Pr; -CH2CH2CH3) , 2-propyl (/'-Pr; /'so-propyl; -CH(CH3)2), 1 -butyl (n-butyl; n-Bu; -CH2CH2CH2CH3), 2-methyl-1 -propyl (/'so-butyl; /-Bu; -CH2CH(CH3)2), 2-butyl (sec-butyl; sec-Bu; -CH(CH3)CH2CH3), 2-methyl-2-propyl (ferf-butyl; f-Bu; -C(CH3)3), 1 -pentyl (n-pentyl; -CH2CH2CH2CH2CH3) , 2-pentyl (-CH(CH3)CH2CH2CH3), 3-pentyl (-CH(CH2CH3)2), 3-methyl-1 -butyl (/so-pentyl; -CH2CH2CH(CH3)2), 2-methyl-2-butyl (-C(CH3)2CH2CH3), 3-methyl-2-butyl (-CH(CH3)CH(CH3)2), 2,2-dimethyl-1 -propyl (neo-pentyl; -CH2C(CH3)3), 2-methyl-1 -butyl (-CH2CH(CH3)CH2CH3), 1 -hexyl (n-hexyl; -CH2CH2CH2CH2CH2CH3), 2-hexyl (-CH(CH3)CH2CH2CH2CH3), 3-hexyl (-CH(CH2CH3)(CH2CH2CH3)), 2-methyl-2-pentyl (-C(CH3)2CH2CH2CH3), 3-methyl-2-pentyl (-CH(CH3)CH(CH3)CH2CH3) , 4-methyl-2-pentyl (-CH(CH3)CH2CH(CH3)2), 3-methyl-3-pentyl (-C(CH3)(CH2CH3)2), 2-methyl-3-pentyl (-CH(CH2CH3)CH(CH3)2), 2,3-dimethyl-2-butyl (-C(CH3)2CH(CH3)2), 3 , 3-dimethyl-2-butyl (-CH(CH3)C(CH3)3), 2,3-dimethyl-l -butyl (-CH2CH(CH3)CH(CH3)CH3), 2,2-dimethyl-l -butyl
(-CH2C(CH3)2CH2CH3), 3,3-dimethyl-l -butyl (-CH2CH2C(CH3)3), 2-methyl-1 -pentyl (-CH2CH(CH3)CH2CH2CH3) , 3-methyl-1 -pentyl (-CH2CH2CH(CH3)CH2CH3), 1 -heptyl (n-heptyl), 2-methyl-1 -hexyl, 3-methyl-1 -hexyl, 2,2-dimethyl-1 -pentyl,
2,3-dimethyl-l -pentyl, 2,4-dimethyl-1 -pentyl, 3,3-dimethyl-1 -pentyl, 2,2, 3-trimethyl-1 -butyl, 3-ethyl-1 -pentyl, 1 -octyl (n-octyl), 1 -nonyl (n-nonyl); 1 -decyl (n-decyl) etc.
By the terms propyl, butyl, pentyl, hexyl, heptyl, octyl, nonyl, decyl etc. without any further definition are meant saturated hydrocarbon groups with the corresponding number of carbon atoms, wherein all isomeric forms are included.
The above definition for alkyi also applies if alkyi is a part of another group such as for example Cx-y-alkylamino or Cx-y-alkyloxy.
The term alkylene can also be derived from alkyi. Alkylene is bivalent, unlike alkyi, and requires two binding partners. Formally, the second valency is produced by removing a hydrogen atom in an alkyi. Corresponding groups are for example -CH3 and -CH2, -CH2CH3 and -CH2CH2 or >CHCH3 etc.
Th e term "C1-4-alkylene" includes for example -(CH2)-, -(CH2-CH2)-, -(CH(CH3))-, -(CH2-CH2-CH2)-, -(C(CH3)2)-, -(CH(CH2CH3))-, -(CH(CH3)-CH2)-, -(CH2-CH(CH3))-, -(CH2-CH2-CH2-CH2)-, -(CH2-CH2-CH(CH3))-, -(CH(CH3)-CH2-CH2)-,
-(CH2-CH(CH3)-CH2)-, -(CH2-C(CH3)2)-, -( C ( C H3)2-CH2)-, -(CH(CH3)-CH(CH3))-, -(CH2-CH(CH2CH3))-, -(CH(CH2CH3)-CH2)-, -(CH(CH2CH2CH3))-, -(CHCH(CH3) 2)- and -C(CH3)(CH2CH3)-.
Other examples of alkylene are methylene, ethylene, propylene, 1 -methylethylene, butylene, 1 -methylpropylene, 1 .1 -dimethylethylene, 1 ,2-dimethylethylene, pentylene, 1 ,1 -dimethylpropylene, 2,2-dimethylpropylene, 1 ,2-dimethylpropylene,
1 ,3-dimethylpropylene, hexylene etc.
By the generic terms propylene, butylene, pentylene, hexylene etc. without any further definition are meant all the conceivable isomeric forms with the corresponding number of carbon atoms, i.e. propylene includes 1 -methylethylene and butylene includes 1 -methylpropylene, 2-methylpropylene, 1 , 1 -dimethylethylene and 1 ,2-dimethylethylene. The above definition for alkylene also applies if alkylene is part of another group such as for example in HO-Cx-y-alkylenamino or H2N-Cx-y-alkylenoxy.
Unlike alkyl, alkenyl consists of at least two carbon atoms, wherein at least two adjacent carbon atoms are joined together by a C-C double bond. If in an alkyl as hereinbefore defined having at least two carbon atoms, two hydrogen atoms on adjacent carbon atoms are formally removed and the free valencies are saturated to form a second bond, the corresponding alkenyl is formed.
Examples of alkenyl are 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.
By the generic terms propenyl, butenyl, pentenyl, hexenyl, butadienyl, pentadienyl, hexa- dienyl, heptadienyl, octadienyl, nonadienyl, decadienyl etc. without any further definition are meant all the conceivable isomeric forms with the corresponding number of carbon atoms, i.e. propenyl includes prop-1 -enyl and prop-2-enyl, butenyl includes but-1 -enyl, but-2-enyl, but-3-enyl, 1 -methyl-prop-1 -enyl, 1 -methyl-prop-2-enyl etc.
Alkenyl may optionally be present in the cis or trans or £ or Z orientation with regard to the double bond(s).
The above definition for alkenyl also applies when alkenyl is part of another group such as for example in Cx-y-alkenylamino or Cx-y-alkenyloxy. Unlike alkylene, alkenylene consists of at least two carbon atoms, wherein at least two adjacent carbon atoms are joined together by a C-C double bond. If in an alkylene as hereinbefore defined having at least two carbon atoms, two hydrogen atoms at adjacent carbon atoms are formally removed and the free valencies are saturated to form a second bond, the corresponding alkenylene is formed.
Examples of alkenylene are ethenylene, propenylene, 1 -methylethenylene, butenylene, 1 -methylpropenylene, 1 ,1 -dimethylethenylene, 1 ,2-dimethylethenylene, pentenylene, 1 ,1 -dimethylpropenylene, 2,2-dimethylpropenylene, 1 ,2-dimethylpropenylene, 1 ,3-dimethylpropenylene, hexenylene etc.
By the generic terms propenylene, butenylene, pentenylene, hexenylene etc. without any further definition are meant all the conceivable isomeric forms with the corresponding number of carbon atoms, i.e. propenylene includes 1 -methylethenylene and butenylene includes 1 -methylpropenylene, 2-methylpropenylene, 1 ,1 -dimethylethenylene and 1 ,2-dimethylethenylene.
Alkenylene may optionally be present in the c/'s or trans or £ or Z orientation with regard to the double bond(s).
The above definition for alkenylene also applies when alkenylene is a part of another group as in for example HO-Cx-y-alkenylenamino or H2N-Cx-y-alkenylenoxy.
Unlike alkyl, alkynyl consists of at least two carbon atoms, wherein at least two adjacent carbon atoms are joined together by a C-C triple bond. If in an alkyl as hereinbefore defined having at least two carbon atoms, two hydrogen atoms in each case at adjacent carbon atoms are formally removed and the free valencies are saturated to form two further bonds, the corresponding alkynyl is formed.
Examples of alkynyl are ethynyl, prop-1 -ynyl, prop-2-ynyl, but-1 -ynyl, but-2-ynyl, but-3-ynyl, 1 -methyl-prop-2-ynyl, pent-1 -ynyl, pent-2-ynyl, pent-3-ynyl, pent-4-ynyl, 3-methyl-but-1 -ynyl, hex-1 -ynyl, hex-2-ynyl, hex-3-ynyl, hex-4-ynyl, hex-5-ynyl etc.
By the generic terms propynyl, butynyl, pentynyl, hexynyl, heptynyl, octynyl, nonynyl, decynyl etc. without any further definition are meant all the conceivable isomeric forms with the corresponding number of carbon atoms, i.e. propynyl includes prop-1 -ynyl and prop-2-ynyl, butynyl includes but-1 -ynyl, but-2-ynyl, but-3-ynyl, 1 -methyl-prop-1 -ynyl, 1 -methyl-prop-2-ynyl, etc.
If a hydrocarbon chain carries both at least one double bond and also at least one triple bond, by definition it belongs to the alkynyl subgroup.
The above definition for alkynyl also applies if alkynyl is part of another group, as in Cx-y-alkynylamino or Cx-y-alkynyloxy, for example.
Unlike alkylene, alkynylene consists of at least two carbon atoms, wherein at least two adjacent carbon atoms are joined together by a C-C triple bond. If in an alkylene as hereinbefore defined having at least two carbon atoms, two hydrogen atoms in each case at adjacent carbon atoms are formally removed and the free valencies are saturated to form two further bonds, the corresponding alkynylene is formed.
Examples of alkynylene are ethynylene, propynylene, 1 -methylethynylene, butynylene, 1 -methylpropynylene, 1 ,1 -dimethylethynylene, 1 ,2-dimethylethynylene, pentynylene, 1 ,1 -dimethylpropynylene, 2,2-dimethylpropynylene, 1 ,2-dimethylpropynylene, 1 ,3-dimethylpropynylene, hexynylene etc.
By the generic terms propynylene, butynylene, pentynylene, hexynylene etc. without any further definition are meant all the conceivable isomeric forms with the corresponding number of carbon atoms, i.e. propynylene includes 1 -methylethynylene and butynylene includes 1 -methylpropynylene, 2-methylpropynylene, 1 ,1 -dimethylethynylene and 1 ,2- dimethylethynylene.
The above definition for alkynylene also applies if alkynylene is part of another group, as in HO-Cx-y-alkynyleneamino or H2N-Cx-y-alkynyleneoxy, for example.
By heteroatoms are meant oxygen, nitrogen and sulphur atoms.
Haloalkyl (haloalkenyl, haloalkynyl) is derived from the previously defined alkyl (alkenyl, alkynyl) by replacing one or more hydrogen atoms of the hydrocarbon chain independently of one another by halogen atoms, which may be identical or different. If a haloalkyl (haloalkenyl, haloalkynyl) is to be further substituted, the substitutions may take place independently of one another, in the form of mono- or polysubstitutions in each case, on all the hydrogen-carrying carbon atoms.
Examples of haloalkyl (haloalkenyl, haloalkynyl) are -CF3, -CHF2, -CH2F, -CF2CF3, -CHFCFs, -CH2CF3, -CF2CH3, -CHFCH3, -CF2CF2CF3, -CF2CH2CH3, -CF=CF2, -CCI=CH2, -CBr=CH2, -CI=CH2, -C≡C-CF3, -CHFCH2CH3, -CHFCH2CF3 etc.
From the previously defined haloalkyl (haloalkenyl, haloalkynyl) are also derived the terms haloalkylene (haloalkenylene, haloalkynylene). Haloalkylene (haloalkenyl, haloalkynyl), unlike haloalkyl, is bivalent and requires two binding partners. Formally, the second valency is formed by removing a hydrogen atom from a haloalkyl.
Corresponding groups are for example -CH2F and -CHF-, -CHFCH2F and -CHFCHF- or >CFCH2F etc.
The above definitions also apply if the corresponding halogen groups are part of another group.
Halogen relates to fluorine, chlorine, bromine and/or iodine atoms. Cycloalkyl is made up of the subgroups monocyclic hydrocarbon rings, bicyclic hydrocarbon rings and spiro-hydrocarbon rings. The systems are saturated. In bicyclic hydrocarbon rings two rings are joined together so that they have at least two carbon atoms together. In spiro-hydrocarbon rings a carbon atom (spiroatom) belongs to two rings together. If a cycloalkyl is to be substituted, the substitutions may take place independently of one another, in the form of mono- or polysubstitutions in each case, on all the hydrogen-carrying carbon atoms. Cycloalkyl itself may be linked as a substituent to the molecule via every suitable position of the ring system.
Examples of cycloalkyl are cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, 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[4.1 .0]heptyl (norcaranyl), bicyclo-[3.1.1 ]heptyl (pinanyl), spiro[2.5]octyl, spiro[3.3]heptyl etc.
The above definition for cycloalkyl also applies if cycloalkyl is part of another group as in Cx-y-cycloalkylamino or Cx-y-cycloalkyloxy, for example.
If the free valency of a cycloalkyl is saturated, then an alicyclic group is obtained.
The term cycloalkylene can thus be derived from the previously defined cycloalkyl. Cycloalkylene, unlike cycloalkyl, is bivalent and requires two binding partners. Formally, the second valency is obtained by removing a hydrogen atom from a cycloalkyl. Corresponding groups are for ex cyclohexyl and
Figure imgf000031_0001
(cyclohexylene).
The above definition for cycloalkylene also applies if cycloalkylene is part of another group as in HO-Cx-y-cycloalkyleneamino or H2N-Cx-y-cycloalkyleneoxy, for example.
Cycloalkenyl is also made up of the subgroups monocyclic hydrocarbon rings, bicyclic hydrocarbon rings and spiro-hydrocarbon rings. However, the systems are unsaturated, i.e. there is at least one C-C double bond but no aromatic system. If in a cycloalkyl as hereinbefore defined two hydrogen atoms at adjacent cyclic carbon atoms are formally removed and the free valencies are saturated to form a second bond, the corresponding cycloalkenyl is obtained. If a cycloalkenyl is to be substituted, the substitutions may take place independently of one another, in the form of mono- or polysubstitutions in each case, on all the hydrogen-carrying carbon atoms. Cycloalkenyl itself may be linked as a substituent to the molecule via every suitable position of the ring system.
Examples of cycloalkenyl are 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, bicyclo[2.2.1 ]hepta-2,5-dienyl
(norborna-2,5-dienyl), bicyclo[2.2.1 ]hept-2-enyl (norbornenyl), spiro[4.5]dec-2-ene etc.
The above definition for cycloalkenyl also applies when cycloalkenyl is part of another group as in Cx-y-cycloalkenylamino or Cx-y-cycloalkenyloxy, for example.
If the free valency of a cycloalkenyl is saturated, then an unsaturated alicyclic group is obtained.
The term cycloalkenylene can thus be derived from the previously defined cycloalkenyl. Cycloalkenylene, unlike cycloalkenyl, is bivalent and requires two binding partners. Formally the second valency is obtained by removing a hydrogen atom from a cycloalkenyl. Corresponding groups are for example cyclopentenyl and or J or *^ or *^ (cyclopentenylene) etc.
The above definition for cycloalkenylene also applies when cycloalkenylene is part of another group as in HO-Cx-y-cycloalkenyleneamino or H2N-Cx-y-cycloalkenyleneoxy, for example.
Aryl denotes a mono-, bi- or tricyclic group with at least one aromatic carbocycle. Preferably it denotes a monocyclic group with six carbon atoms (phenyl) or a bicyclic group with nine or ten carbon atoms (two six-membered rings or one six-membered ring with a five-membered ring), wherein the second ring may also be aromatic or, however, may also be saturated or partially saturated. If an aryl is to be substituted, the substitutions may take place independently of one another, in the form of mono- or polysubstitutions in each case, on all the hydrogen-carrying carbon atoms. Aryl itself may be linked as a substituent to the molecule via every suitable position of the ring system.
Examples of aryl are phenyl, naphthyl, indanyl (2,3-dihydroindenyl), indenyl, anthracenyl, phenanthrenyl, tetrahydronaphthyl (1 ,2,3,4-tetrahydronaphthyl, tetralinyl), dihydronaphthyl (1 ,2- dihydronaphthyl), fluorenyl etc. The above definition of aryl also applies when aryl is part of another group as in arylamino or aryloxy, for example.
If the free valency of an aryl is saturated, then an aromatic group is obtained.
The term arylene can also be derived from the previously defined aryl. Arylene, unlike aryl, is bivalent and requires two binding partners. Formally, the second valency is formed by removing a hydrogen atom from an aryl. Corresponding groups are e.g.
Figure imgf000033_0001
The above definition for arylene also applies when arylene is part of another group as in HO-aryleneamino or H2N-aryleneoxy for example.
Heterocyclyl denotes ring systems, which are derived from the previously defined cycloalkyl, cycloalkenyl and aryl by replacing one or more of the groups -CH2- independently of one another in the hydrocarbon rings by the groups -0-, -S- or -NH- or by replacing one or more of the groups =CH- by the group =N-, wherein a total of not more than five heteroatoms may be present, at least one carbon atom may be present between two oxygen atoms and between two sulphur atoms or between one oxygen and one sulphur atom and the ring as a whole must have chemical stability. Heteroatoms may optionally be present in all the possible oxidation stages (sulphur -> sulphoxide -SO, sulphone -S02-; nitrogen -> N-oxide). In a heterocyclyl there is no heteroaromatic ring, i.e. no heteratom is part of an aromatic system.
A direct result of the derivation from cycloalkyl, cycloalkenyl and aryl is that heterocyclyl is made up of the subgroups monocyclic heterorings, bicyclic heterorings, tricyclic heterorings and spiro-heterorings, which may be present in saturated or unsaturated form. By unsaturated is meant that there is at least one double bond in the ring system in question, but no heteroaromatic system is formed. In bicyclic heterorings two rings are linked together so that they have at least two (hetero)atoms in common. In spiro-heterorings a carbon atom (spiroatom) belongs to two rings together. If a heterocyclyl is substituted, the substitutions may take place independently of one another, in the form of mono- or polysubstitutions in each case, on all the hydrogen- carrying carbon and/or nitrogen atoms. Heterocyclyl itself may be linked as a substituent to the molecule via every suitable position of the ring system.
Examples of heterocyclyl are tetrahydrofuryl, pyrrolidinyl, pyrrolinyl, imidazolidinyl, thiazolidinyl, imidazolinyl, pyrazolidinyl, pyrazolinyl, piperidinyl, piperazinyl, oxiranyl, aziridinyl, azetidinyl, 1 ,4-dioxanyl, azepanyl, diazepanyl, morpholinyl, thiomorpholinyl, h om om orph ol i nyl , h omopi peri d i nyl , h om opi perazi nyl , h omoth i om orph ol i nyl , thiomorpholinyl-S-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-S-oxide, tetrahydrothienyl-S,S-dioxide, homothiomorpholinyl-S-oxide,
2.3- dihydroazet, 2H-pyrrolyl, 4/-/-pyranyl, 1 ,4-dihydropyridinyl, 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,
1 .4- dioxa-spiro[4.5]decyl, 1 -oxa-3.8-diaza-spiro[4.5]decyl, 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.
Further examples are the structures illustrated below, which may be attached via each hydrogen-carrying atom (exchanged for hydrogen):
Figure imgf000034_0001
Figure imgf000035_0001

Figure imgf000036_0001
Figure imgf000037_0001
The above definition of heterocyclyl also applies if heterocyclyl is part of another group as in heterocyclylamino or heterocyclyloxy for example.
If the free valency of a heterocyclyl is saturated, then a heterocyclic group is obtained.
The term heterocyclylene is also derived from the previously defined heterocyclyl. Heterocyclylene, unlike heterocyclyl, is bivalent and requires two binding partners. Formally, the second valency is obtained by removing a hydrogen atom from a heterocyclyl. Corresponding groups are for example piperidinyl and or or
Figure imgf000037_0002
2,3-dihydro-1 H-pyrrolyl and H or or » or " etc.
The above definition of heterocyclylene also applies if heterocyclylene is part of another group as in HO-heterocyclyleneamino or H2N-heterocyclyleneoxy for example.
Heteroaryl denotes monocyclic heteroaromatic rings or polycyclic rings with at least one heteroaromatic ring, which compared with the corresponding aryl or cycloalkyl (cycloalkenyl) 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, wherein the resulting group must be chemically stable. The prerequisite for the presence of heteroaryl is a heteroatom and a heteroaromatic system. If a heteroaryl is to be substituted, the substitutions may take place independently of one another, in the form of mono- or polysubstitutions in each case, on all the hydrogen- carrying carbon and/or nitrogen atoms. Heteroaryl itself may be linked as a substituent to the molecule via every suitable position of the ring system, both carbon and nitrogen.
Examples of heteroaryl are furyl, thienyl, pyrrolyl, oxazolyl, thiazolyl, isoxazolyl, isothiazolyl, pyrazolyl, imidazolyl, triazolyl, tetrazolyl, oxadiazolyl, thiadiazolyl, pyridyl, pyrimidyl, pyridazinyl, pyrazinyl, triazinyl, pyridyl-/V-oxide, pyrrolyl-/V-oxide, pyrimidinyl-A- oxide, pyridazinyl-/V-oxide, pyrazinyl-/V-oxide, imidazolyl-/V-oxide, isoxazolyl-/V-oxide, oxazolyl-/V-oxide, thiazolyl-/V-oxide, oxadiazolyl-/V-oxide, thiadiazolyl-/V-oxide, triazolyl-/V- oxide, tetrazolyl-/V-oxide, indolyl, isoindolyl, benzofuryl, benzothienyl, benzoxazolyl, benzothiazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazolyl, indazolyl, isoquinolinyl, quinolinyl, quinoxalinyl, cinnolinyl, phthalazinyl, quinazolinyl, benzotriazinyl, indolizinyl, oxazolopyridyl, imidazopyridyl, naphthyridinyl, benzoxazolyl, pyridopyridyl, purinyl, pteridinyl, benzothiazolyl, imidazopyridyl, imidazothiazolyl, quinolinyl-/V-oxide, indolyl-/V- oxide, isoquinolyl-/V-oxide, quinazolinyl-/V-oxide, quinoxalinyl-/V-oxide, phthalazinyl-/V- oxide, indolizinyl-/V-oxide, indazolyl-/V-oxide, benzothiazolyl-/V-oxide, benzimidazolyl-/V- oxide etc.
Further examples are the structures illustrated below, which may be attached via each hydrogen-carrying atom (exchanged for hydrogen):
Figure imgf000038_0001
Figure imgf000039_0001
The above definition of heteroaryl also applies when heteroaryl is part of another group as in heteroarylamino or heteroaryloxy, for example.
If the free valency of a heteroaryl is saturated, a heteroaromatic group is obtained.
The term heteroarylene can therefore be derived from the previously defined heteroaryl. Heteroarylene, unlike heteroaryl, is bivalent and requires two binding partners. Formally, the second valency is obtained by removing a hydrogen atom from a heteroaryl. Corresponding groups are for ex pyrrolyl and
Figure imgf000039_0002
or or or etc. The above definition of heteroarylene also applies when heteroarylene is part of another group as in HO-heteroaryleneamino or H2N-heteroaryleneoxy, for example.
By substituted is meant that a hydrogen atom which is bound directly to the atom under consideration, is replaced by another atom or another group of atoms (substituent). Depending on the starting conditions (number of hydrogen atoms) mono- or polysubstitution may take place on one atom. Substitution with a particular substituent is only possible if the permitted valencies of the substituent and of the atom that is to be substituted correspond to one another and the substitution leads to a stable compound (i .e. to a compound which is not converted spontaneously, e.g. by rearrangement, cyclisation or elimination).
Bivalent substituents such as =S, =NR, =NOR, =NNRR, =NN(R)C(0)NRR, =N2 or the like, may only be substituents at carbon atoms, wherein the bivalent substituent =0 may also be a substituent at sulphur. Generally, substitution may be carried out by a bivalent substituent only at ring systems and requires replacement by two geminal hydrogen atoms, i.e. hydrogen atoms that are bound to the same carbon atom that is saturated prior to the substitution. Substitution by a bivalent substituent is therefore only possible at the group -CH2- or sulphur atoms of a ring system.
Stereochemistry/Solvates/Hydrates: Unless stated otherwise a structural formula given in the description or in the claims or a chemical name refers to the corresponding compound itself, but also encompasses the tautomers, stereoisomers, optical and geometric isomers (e.g. enantiomers, diastereomers, EIZ isomers, etc.), racemates, mixtures of separate enantiomers in any desired combinations, mixtures of diastereomers, mixtures of the forms mentioned hereinbefore (if such forms exist) as well as salts, particularly pharmaceutically acceptable salts thereof. The compounds and salts according to the invention may be present in solvated form (e.g. with pharmaceutically acceptable solvents such as e.g. water, ethanol etc.) or in unsolvated form. Generally, for the purposes of the present invention the solvated forms, e.g. hydrates, are to be regarded as of equal value to the unsolvated forms.
Salts: The term "pharmaceutically acceptable" is used herein to denote compounds, materials, compositions and/or formulations which are suitable, according to generally recognised medical opinion, for use in conjunction with human and/or animal tissue and do not have or give rise to any excessive toxicity, irritation or immune response or lead to other problems or complications, i.e. correspond overall to an acceptable risk/benefit ratio.
The term "pharmaceutically acceptable salts" relates to derivatives of the chemical compounds disclosed in which the parent compound is modified by the addition of acid or base. Examples of pharmaceutically acceptable salts include (without being restricted thereto) salts of mineral or organic acids in relation to basic functional groups such as for example amines, alkali metal or organic salts of acid functional groups such as for example carboxylic acids, etc. These salts include in particular acetate, ascorbate, benzenesulphonate, benzoate, besylate, bicarbonate, bitartrate, bromide/hydrobromide, Ca-edetate/edetate, camsylate, carbonate, chloride/hydrochloride, citrate, edisylate, ethane disulphonate, estolate, esylate, fumarate, gluceptate, gluconate, glutamate, glycolate, glycollylarsnilate, hexylresorcinate, hydrabamine, hydroxymaleate, hydroxynaphthoate, iodide, isothionate, lactate, lactobionate, malate, maleate, mandelate, methanesulphonate, mesylate, methylbromide, methylnitrate, methylsulphate, mucate, napsylate, nitrate, oxalate, pamoate, pantothenate, phenyl acetate, phosphate/diphosphate, polygalacturonate, propionate, salicylate, stearate, subacetate, succinate, su lpham ide, su lphate, tan nate, tartrate, teoclate, tol uenesu lphonate, triethiodide, ammonium, benzathine, chloroprocaine, choline, diethanolamine, ethylenediamine, meglumin and procaine. Other pharmaceutically acceptable salts may be formed with cations of metals such as aluminium, calcium, lithium, magnesium, potassium, sodium, zinc, etc. (cf. also Pharmaceutical salts, Birge, S.M. et al., J. Pharm. Sci., (1977), 66, 1 -19).
The pharmaceutically acceptable salts of the present invention may be prepared starting from the parent compound which carries a basic or acidic functionality, by conventional chemical methods. Generally, such salts may be synthesised by reacting the free acid or base form of these compounds with a sufficient amount of the corresponding base or acid in water or an organic solvent such as for example ether, ethyl acetate, ethanol, isopropanol, acetonitrile (or mixtures thereof).
Salts of acids other than those mentioned above, which are useful for example for purifying or isolating the compounds from the reaction mixtures (e.g. trifluoroacetates), are also to be regarded as part of the invention.
In a representation such as for example
Figure imgf000042_0001
the letter A has the function of a ring designation in order to make it easier, for example, to indicate the attachment of the ring in question to other rings.
For bivalent groups in which it is crucial to determine which adjacent groups they bind and with which valency, the corresponding binding partners are indicated in brackets, where necessary for clarification purposes, as in the following representations:
Figure imgf000042_0002
or (R2)-C(0)NH- or (R2)-NHC(0)-;
Groups or substituents are frequently selected from among a number of alternative groups/ substituents with a corresponding group designation (e.g. Ra, Rb etc). If such a group is used repeatedly to define a compound according to the invention in different molecular parts, it must always be borne in mind that the various uses are to be regarded as totally independent of one another.
By a therapeutically effective amount for the purposes of this invention is meant a quantity of substance that is capable of obviating symptoms of illness or of preventing or alleviating these symptoms, or which prolong the survival of a treated patient.
List of abbreviations
Figure imgf000042_0003
DCM dichloromethane
DEA diethylamine
DIC diisopropylcarbodiimide
A/-ethyl-/V,/V-diisopropylamine
DIPEA
(HIJNIG base)
DMF /V,/V-dimethylformamide
DMSO dimethylsulphoxide
N-(3-dimethylaminopropyl)-N4-
EDC
ethylcarbodiimide hydrochloride
ESI electron spray ionization
Et ethyl
EtOH ethanol
h hour
0-(7-azabenzotriazol-1 -yl)-A/,A/,A/',/V'-
HATU
tetramethyl-uronium hexafluorophosphate
HPLC high performance liquid chromatography
Hijnig base /V-ethyl-/V,/V-diisopropylamine
/' iso
cat. catalyst, catalytic
cone. concentrated
LC liquid chromatography
sin. solution
Me methyl
MeOH methanol
min minutes
MPLC medium pressure liquid chromatography
MS mass spectrometry
MW microwave
NMP N-methylpyrrolidone
PBS phosphate-buffered saline solution
1 ,1 '-bis(diphenylphosphino)ferrocene-
Pd-DPPF
palladium(ll)-dichloride dichloromethane
PDK1 phosphoinositide-dependent kinase 1
Ph phenyl PI3K phosphatidylinositol-3-kinase
PKT protein kinase B
Pr propyl
Rf (Rf) retention factor
RP reversed phase
RT ambient temperature
s second
0-(benzotriazol-1 -yl)-/V,/V,/V',/V'-
TBTU
tetramethyl-uronium tetrafluoroborate
TEA triethylamine
tert. tertiary
Tf triflate
TFA trifluoroacetic acid
THF tetrahydrofuran
TMS trimethylsilyl
Tos tosyl
tRet. retention time (HPLC)
TRIS tris(hydroxymethyl)-aminomethane
UV ultraviolet
Features and advantage of the present invention will become apparent from the following detailed Examples, which illustrate the fundamentals of the invention by way of example, without restricting its scope: Preparation of the compounds according to the invention
General
Unless stated otherwise, all the reactions are carried out in commercially obtainable apparatus using methods that are commonly used in chemical laboratories. Starting materials that are sensitive to air and/or moisture are stored under protective gas and corresponding reactions and manipulations therewith are carried out under protective gas (nitrogen or argon).
The compounds are named according to the Beilstein rules using the Autonom software (Beilstein). If a compound is to be represented both by a structural formula and by its nomenclature, in the event of a conflict the structural formula is decisive.
Microwave reactions are carried out in an initiator/reactor made by Biotage or in an Explorer made by CEM in sealed containers (preferably 2, 5 or 20 ml_), preferably with stirring.
Chromatography
Thin layer chromatography is carried out on ready-made TLC plates of silica gel 60 on glass (with fluorescence indicator F-254) made by Merck.
The preparative high pressure chromatography (HPLC) of the example compounds according to the invention is carried out with columns made by Waters (names: Sunfire C1 8, 5 m, 30 x 100 mm Part. No. 186002572; X-Bridge C18, 5 [Ji m , 30 x 100 mm Part. No. 186002982).
The com pou nd s are el uted u si n g eith er d ifferent grad ients of H20/acetonitrile or H20/MeOH, wherein either 0.1 % HCOOH is added to the water (acid conditions). For chromatography under basic conditions H20/acetonitrile gradients are also used, and the water is made basic according to the following recipe: 5 mL of an ammonium hydrogen carbonate solution (158 g to 1 L H20) and 2 mL ammonia (7M in MeOH) are made up to 1 L with H20.
The normal-phase preparative high pressure chromatography (HPLC) of the example compounds according to the invention is carried out with colu mns made by Macherey & Nagel (name: Nucleosil, 50-7, 40 x 250 mm) and VDSoptilab (name: Kromasil 100 NH2, 10 μ Μ , 50 x 250 mm). The compounds are eluted using different gradients of DCIW MeOH, with 0.1 % NH3 added to the MeOH.
The analytical HPLC (reaction monitoring) of intermediate compounds is carried out with columns made by Agilent, Waters and Phenomenex. The analytical equipment is also provided with a mass detector in each case.
HPLC mass spectroscopy/UV spectrometry
The retention times/MS-ESI+ for characterising the example compounds according to the invention are produced using an HPLC-MS apparatus (high performance liquid chromatography with mass detector) made by Agilent. Compounds that elute at the injection peak are given the retention time tRet. = 0.00. HPLC-methods
Preparative
prep. HPLC1
HPLC: 333 and 334 Pumps
Column: Waters X-Bridge C18, 5 [Jim , 30 x 100 mm, Part. No. 186002982
Eluant: A: 10 mM NH4HC03 in H20; B: acetonitrile (HPLC grade)
Detection: UVA is-155
Flow: 50 mL/min
Gradient: 0.00 min: 5 % B
3.00 - 15.00 min: variable (see individual methods) 15.00 - 17.00 min: 100 % B
prep. HPLC2
HPLC: 333 and 334 Pumps
Column: Waters Sunfire C18, 5 [Jim , 30 x 100 mm, Part. No. 186002572
Eluant: A: H20 + 0.2 % HCOOH; B: acetonitrile (HPLC grade) + 0.2 % HCOOH
Detection: UVA is-155
Flow: 50 mL/min
Gradient: 0.00 min: 5 % B
3.00 - 15.00 min: variable (see individual methods) 15.00 - 17.00 min: 100 % B
analytical
LCMSBAS1
HPLC: Agilent 1 100 Series
MS: Agilent LC/MSD SL
Column: Phenomenex Mercury Gemini C 1 8, 3 μηη , 2 x 20 mm , Part. No. 00M- 4439-BO-CE
Eluant: A: 5 mM NH4HCO3/20 mM NH3 in H20; B: acetonitrile (H PLC grade) Detection: MS: Positive and negative mode
Mass range: 120 - 900 m/z
Flow: 1 .00 mL/min
Column temp. : 40°C
Gradient: 0.00 min: 5 % B 0.00-2.50 min: 5 %→ 95 % B
2.50-2.80 min: 95 % B
2.81 - 3.10 min: 95 %→ 5 % B
HPLC: Agilent 1100 Series
MS: Agilent LC/MSD SL
Column: Waters X-Bridge C18, 2.1 x 50mm, 3.5μηι
Eluant: A: 5 mM NH4HCO3/20 mM NH3 in H20; B: acetonitrile (HPLC grade) Detection: MS: Positive and negative mode
Mass range 105- 1200 m/z
Flow: 1.20 mL/min
Column temp.:35°C
Gradient: 0.01 min: 5% B
0.01 - 1.25 min: 5 % -» 95 % B
1.25-2.00 min: 95 % B
2.00-2.01 min: 95 % -» 5 % B
FE CB4/FE CBM2
HPLC: Agilent 1100 Series
MS: Agilent LC/MSD SL
Column: Agilent Zorbax Extend C18, 3.5 μηι, 2.1 x 50 mm, Part. No.
735700-902
Eluant: A: 5 mM NH4HCO3/20 mM NH3 in H20; B: acetonitrile (HPLC grade)
Detection: MS: Positive and negative mode
Mass range: 105 - 1200 m/z
Flow: 1.20 mL/min
Column temp.: 35°C
Gradient: 0.01 min: 5% B
0.01 - 1.25 min: 5 %→ 95 % B
1.25-2.00 min: 95 % B
2.00-2.01 min: 95 %→ 5 % B
Agilent 1100 Series
MS: Agilent LC/MSD SL Column: Agilent Zorbax SB C8, 3.5 μηι, 2.1 x 50 mm, Part. No.871700-906 Eluant: A: H20 + 0.2 % HCOOH; B: acetonitrile (HPLC grade) + 0.2 % HCOOH Detection: MS: Positive and negative mode
Mass range: 105- 1200 m/z
Flow: 1.20 mL/min
Column temp. : 35°C
Gradient: 0.01 min: 5% B
0.01 - 1.25 min: 5 %→95 % B
1.25-2.00 min: 95 % B
2.00-2.01 min: 95 %→ 5 % B
_BAS
HPLC: Agilent 1100 Series
MS: 1100 Series LC/MSD SL (MM-ES+APCI, +3000V, Quadrupole, G1956B)
Column: Waters X-Bridge C18, 3.5 [Jim, 135 A, 2.1 x 30 mm, Part.No.: 186003020 Eluant: A: 5 mM NH4HCO3/20 mM NH3 (pH = 9.5); B: acetonitrile (HPLC grade) Detection: MS: Positive mode
Mass range: 120-750 m/z
Flow: 1.00 mL/min
Column temp. : 35°C
Gradient: 0.0 - 1.0 min 15% -» 95% B
1.0- 1.6 min 95 % B
1.6- 1.7 min 95% -» 15% B
1.7- 2.3 min 15% B
_FEC
HPLC: Agilent 1100 Series
MS: 1200 Series LC/MSD (API-ES + 3000V, Quadrupole, G6140A)
Column: Agilent Zorbax SB C8, 3.5 μηι, 80 A, 2.1 x 50 mm, Part. No.:
871700-906
Eluant: A: water + 0.1 % HCOOH; B: acetonitrile (HPLC grade) + 0.1 % HCOOH Detection: MS: Positive mode
Mass range: 120-750 m/z
Flow: 1.10 mL/min
Column temp. : 45°C Gradient: 0.0 - 1.75 min 15%→95% B
1.75- 1.9 min 95 % B
1.9 - 1.92 min 95%→15% B
1.92-2.1 min 15% B
1 BAS MeOH POS
HPLC: Agilent 1100 Series
MS: 1100 Series LC/MSD SL (MM-ES+APCI, + 3000V, Quadrupole,
G1956B)
Column: Waters X-Bridge C18, 3.5 [Jim, 135 A, 2.1 x 30 mm, Part. No.:
186003020
Eluant: A: 5 mM NH4HCO3/20 mM NH3 (pH = 9.5); B: MeOH (HPLC grade)
Detection: MS: Positive
Mass range: 120 - 750 m/z
Flow: 1.00 mL/min
Column temp.: 40°C
Gradient: 0.0 - 1.0 min 20 %→ 95 % B
1.0-2.0 min 95 % B
2.0-2.1 min 95 %→20 % B
2.1 -2.3 min 20 % B
BFEC
HPLC: Agilent 1100/1200 Series
MS: Agilent 1100 LC/MSD SL
Column: Waters X-Bridge C18, 5 μηι, 2.1 x 50 mm
Eluant: A: H20 + 0.1 % NH4OH/NH4HC03; B: acetonitrile
Detection: MS: Positive and negative mode
Mass range: 100 - 1200 m/z
Flow: 1.00 mL/min
Column temp.: 35°C
Gradient: 0.01 min: 5% B
0.01 - 1.50 min: 5% 100% B
1.50-2.10 min: 100% B
2.10-2.20 min: 100 % ^ 5% B AFEC
HPLC: Agilent 1100/1200 Series
MS: Agilent 1100 LC/MSD SL
Column: Waters Sunfire C18, 5 μηι, 2.1 x 50 mm
Eluant: A: H20 + 0.1 % HCOOH; B: acetonitrile
Detection: MS: Positive and negative mode
Mass range: 100 - 1200 m/z
Flow: 1.00 mL/min
Column temp.: 35°C
Gradient: 0.01 min: 5 % B
0.01 - 1.50 min: 5%→100%B
1.50-2.10 min: 100 % B
2.10-2.20 min: 100%→5%B
FECS2
HPLC: Agilent 1100/1200 Series
MS: Agilent 1100 LC/MSD SL
Column: Sunfire C18, δμηη, 2.1 x 50 mm
Eluant: A: H20 + 0.2 % HCOOH; B: MeOH
Detection: ESI
Mass range: 100 - 1200 m/z
Flow: 1.20 mL/min
Column temp.: 35°C
Gradient: 0.01 min: 5 % B
0.01 - 1.50 min: 5%→100%B
1.50-2.00 min: 100 % B
FECBM4
HPLC: Agilent 1100/1200 Series
MS: Agilent 1100 LC/MSD SL
Column: Waters Sunfire, δμηη, 2.1 x 50 mm
Eluant: A: H20 + 5 mM NH4HC03, 19 mM NH3; B: MeOH
Detection: MS: Positive and negative mode
Mass range: 100 - 1200 m/z
Flow: 1.20 mL/min Column temp.: 35°C
Gradient: 0.01 min: 5% B
0.01 - 1.25 min: 5 % -» 95 % B 1.25- 1.50 min: 95% -» 100 % B 1.50-2.25 min 100% B
FECB6
HPLC: Agilent 1100/1200 Series
MS: Agilent 1100 LC/MSD SL
Column: Waters X-Bridge C18, 5 μηι, 2.1 x 50
Eluant: A: H20 + 5 mM NH4HC03, 19 mM NH3
Detection: ESI
Mass range: 100 - 1200 m/z
Flow: 1.20 mL/min
Column temp.: 35°C
Gradient: 0.01 min: 5% B
0.01 - 1.24 min: 5 %→100 1.24-2.10 min: 100% B
FSUN
HPLC: Agilent 1100/1200 Series
MS: Agilent 1100 LC/MSD SL
Column: Waters Sunfire, 3.5 μηι, 2.1 x 50 mm
Eluant: A: H20 + 0.2 % HCOOH; B: CH3CN
Detection: ESI
Mass range: 100 - 1200 m/z
Flow: 1.20 mL/min
Column temp.: 35°C
Gradient: 0.01 min: 5% B
0.01 - 1.50 min: 5 %→95 1.50-2.00 min: 100% B FSUN2
HPLC: Agilent 1100/1200 Series
MS: Agilent 1100 LC/MSD SL Column: Waters Sunfire, 5.0 μηη, 2.1 x 50 mm
Eluant: A: H20 + 0.2 % HCOOH; B: CH3CN
Detection: ESI
Mass range: 100 - 1200 m/z
Flow: 1 .20 ml_/min
Column temp.: 35°C
Gradient: 0.01 min: 5 % B
0.01 - 1 .50 min: 5 %→ 95 % B
1 .50 - 2.00 min: 100 % B
The compounds according to the invention are prepared by the methods of synthesis described hereinafter, in which the substituents of the general formulae have the meanings given hereinbefore. These methods are intended as an illustration of the invention, without restricting its subject matter and the scope of the compounds claimed to these examples. Where the preparation of starting compounds is not described, they are commercially obtainable or may be prepared analogously to known compounds or methods described herein. Substances described in the literature are prepared according to the published methods of synthesis.
a) Synthesis of free cyclic carboxylic acids A
Reaction scheme A
Figure imgf000053_0001
A.d
The method of synthesising the nitrogen-containing heterocyclic carboxylic acids A depends on the number and position of the cyclic nitrogens in the ring Qb.
Starting from the esters A.a the grouping Qa-CR1R2- may be incorporated by nucleophilic substitution at the component A.b, which is activated by an electron-withdrawing leaving group LG, e.g. a halogen, triflate or mesylate. A.a is optionally deprotonated for this purpose by the addition of a base.
A pyridazinone ring system (Qb = pyridazinone) is synthesised starting from pyruvic acid derivatives which are cyclised with cyanoacetohydrazides. Subsequent saponification of the nitrile yields the corresponding carboxylic acid (Helv. Chim. Acta, 1954, 37, 1467), which after esterification yields the desired pyridazinonecarboxylic acid ester A.a. This is reacted in a nucleophilic substitution with A.b and subsequently saponified to form the desired carboxylic acid A.
A pyrimidinone ring system (Qb = pyrimidinone) is synthesised by cyclisation of malonic acid diester derivatives with amidines. The pyrimidinonecarboxylic acid ester A.a obtained is reacted in a nucleophilic substitution with A.b and subsequently saponified to form the desired carboxylic acid A (WO 2010/0071 14, WO 2010/0071 16).
A pyridinone ring system (Qb = pyridinone) is synthesised starting from malonic acid - diester derivatives A.d. The derivatives used are di- and trielectrophiles, which cyclise when reacted with amines A.c (WO 2010/0071 14, WO 2010/0071 16).
A pyrazinone ring system (Qb = pyrazinone) is synthesised starting from glyoxal derivatives and aminomalonamide amidines. First of all the corresponding amino- pyrazinecarboxamides are obtained, which are saponified to form the carboxylic acid. After hydrolysis of the amine by diazotisation and esterification of the carboxylic acid the desired pyrazinone ring system A.a is obtained (J. Am. Chem. Soc, 1959, 81, 2472-4). This is reacted in a nucleophilic substitution with A.b and then saponified to form the desired carboxylic acid A.
A pyrazolone ring system (Qb = pyrazolone) is obtained starting from malonic acid diester derivatives A.d, which cyclise with hydrazines A.e. The group LG is not a leaving group in the true sense but an electrophilic group, particularly a carbonyl, to which the hydrazine is added.
Using the methods of synthesis described above, starting from the cyclic carboxylic acid esters A.a or their precursors A.d after reaction with A.b or A.c, first of all carboxylic acid esters A* are obtained. These are each saponified to form the free acid A. In the grouping -COOR" , it is possible to have groups R" which make this saponification easy and gentle. Such groups include in particular methyl, ethyl, ie f-butyl and benzyl esters, wherein others are known to the skilled man from their general specialist knowledge.
The required educts A.b, A.c and A.e, as well as A.a and A.d, are commercially obtainable, already described in the literature or may be prepared analogously to published methods. ethod for synthesising A.1
Figure imgf000055_0001
A.1
Malonic acid diester derivative A.d.1 (5.00 g, 25.0 mmol) is taken up in 40 mL 2-butanol, cooled to 5°C and mixed with amine A.c.1 (3.00 mL, 25.1 mmol), which has been taken up in 10 mL 2-butanol. The reaction mixture is stirred for 1 h at 20°C, diluted with a further 150 mL of 2-butanol and heated to 100°C for 48 h. Then the reaction mixture is combined with 50 mL 2 N aqueous sodium hydroxide solution and 50 mL 2 N methanolic sodium hydroxide solution and stirred for 2 h at 20°C. The reaction mixture is acidified with HCI (50 mL, 1 N) and extracted with DCM (3 x 50 mL), dried on MgS04, filtered, the solvent is eliminated in vacuo and A.1 (6.08 g, 90 %; MS(M+H)+ = 266; t 0.00 min; method LCMSBAS1 ) is obtained.
Compounds A.2 - A.24-3 are prepared analogously to A.1 (Table 1 ):
MS (M±H)r;
# Structure tRet. HPLC [min] HPLC method
,= 0.00 LCMSBAS1
,= 0.00 LCMSBAS1
Figure imgf000055_0002
Figure imgf000056_0001
Figure imgf000057_0001
.10 M+H= 260; tRet.= 0.20 1_BAS_MEOH_POS
A.11 M+H= 278; tRet = 0.00 LCMSBAS1
A.12 M+H= 296; tRet = 0.00 LCMSBAS1
Figure imgf000057_0002
Figure imgf000058_0001
A.16 M+H= 274; tRet = 0.24 1_BAS_MEOH_POS
A.17 M+H= 266; tRet = 0.00 LCMSBAS1
Figure imgf000058_0002
A.18 M+H= 265; tRet = 1.66 FECS
Figure imgf000058_0003
CI
Figure imgf000059_0001
Figure imgf000060_0001
Method of svnthesising A.25
Figure imgf000060_0002
Sodium hydride (60 %; 1.40 g, 36.0 mmol) is placed in DMF (70 mL), mixed with carboxylic acid ester A.a.1 (5.00 g, 32.0 mmol) and stirred for 45 min at 20°C. Then benzylbromide A.b.1 (4.10 mL, 32.0 mmol) is metered in and the mixture is stirred for a further 3 h at 20°C. The reaction mixture is combined with HCI (50 mL, 1 N) and DCM (50 mL), the organic phase is separated off and extracted again with HCI (2 x 30 mL). Then the organic phase is dried, the solvent is eliminated in vacuo and carboxylic acid ester A*.25 (10.0 g, 89 %; HPLC-MS: MS(M+H)+ = 281 ; tRel = 1 .57 min; method FEC3) is obtained.
Carboxylic acid ester A*.25 (1 .9 g, 5.6 mmol) is taken up in MeOH (7.3 mL) and mixed with NaOH (6.2 mL, 1 M). After 16 h at 20°C the mixture is diluted with water and extracted with DCM. The organic phase is discarded, the aqueous phase is acidified and extracted with DCM. The organic phase is dried on Na2S04, filtered, the solvent is eliminated in vacuo and the free carboxylic acid A.25 (694 mg, 47 %; HPLC-MS: MS(M+H)+ = 267; tRet = 0.29 min; method FECB4) is obtained.
Method of synthesising A.26
Figure imgf000061_0001
Sodium hydride (60 %; 105 mg, 2.60 mmol) is placed in DMF (1 .50 mL), mixed with carboxylic acid ester A.a.2 (400 mg, 2.40 mmol) and stirred for 45 min at 20°C. Then benzyl bromide A.b.1 (0.31 mL, 2.4 mmol) is metered in and the mixture is stirred for a further 48 h at 20°C. The reaction mixture is combined with HCI (50 mL, 1 N) and DCM (50 mL), the organic phase is separated off and extracted again with HCI (2 x 30 mL). Then the organic phase is dried, the solvent is eliminated in vacuo and carboxylic acid ester A*.26 (487 mg, 69 %; HPLC-MS: MS(M+H)+ = 295; tRel = 2.09 min; method AFEC) is obtained.
Carboxylic acid ester A*.26 (487 mg, 1 .71 mmol) is taken up in MeOH (4.0 mL) and mixed with NaOH (2.0 mL, 1 N). After 2 h at 20°C the mixture is diluted with water and extracted with DCM. The organic phase is discarded, the aqueous phase is acidified and extracted with DCM. The organic phase is dried on Na2S04, the solvent is eliminated in vacuo and the free carboxylic acid A.26 (HPLC-MS: MS(M-H)" = 279 ; t 0.00 min; method LCMSBAS1 ) is obtained.
Compounds A.27 - A.33 are prepared analogously to compound A.26 and if necessary subjected to chiral chromatography before the saponification (Table 2):
Figure imgf000062_0001
Figure imgf000063_0001
* Gilson HPLC apparatus, chiral column (Daicel Chiralpack IC, 250 x 20 mm).
Eluant: 35 % n-heptane, 65 % DCM/EtOH/diethylamine (2000:100:2.6)
Method of synthesising A.34
Figure imgf000063_0002
ED.1 Z.1 A.a.3
Figure imgf000063_0003
A*.34, 98 %
3. NaO
Figure imgf000063_0004
A.34 Carboxylic acid ester ED.1 (2.00 g, 10.4 mmol) is placed in dioxane (9.0 mL), combined with HCI (10.4 mL, 1 M) and stirred for 20 h at 90°C. Then the reaction mixture is cooled to 0°C, the precipitate is filtered off, washed with water and pyridazinonic acid Z.1 (955 mg, 53 %; HPLC-MS: MS(M-H)" = 173; tRet = 0.00 min; method LCMSBAS1 ) is obtained. Pyridazinonic acid Z.1 (955 mg, 5.4 mmol) is placed in MeOH (6.0 mL), combined with H CI (6.0 m L, 4M in d ioxane) and stirred for 20 h at 50°C. The reaction mixture is combined with H20 and DCM, the organic phase is separated off and extracted with H20 (2 x 1 0 m L). Then the organic phase is dried on MgS04, filtered off, the solvent is eliminated in vacuo and carboxylic acid ester A.a.3 (890 mg, 86 %; HPLC-MS: MS(M+H)+ = 189; tRet = 1.47 min; method AFEC) is obtained.
Sodium hydride (60 %; 93 mg, 2.3 mmol) is placed in DMF (1 .5 mL), combined with carboxylic acid ester A.a.3 (397 mg, 2.10 mmol) and stirred for 45 min at 20°C. Then benzyl bromide A.b.1 (0.27 mL, 2.1 mmol) is metered in and the mixture is stirred for a further 2 h at 20°C. The reaction mixture is combined with HCI (5 mL, 1 N) and DCM (5 mL), the organic phase is separated off and extracted again with HCI (2 x 5 mL). Then the organic phase is dried on MgS04, filtered off, the solvent is eliminated in vacuo and carboxylic acid ester A*.34 (922 mg, 98 %; H PLC-MS: MS(M+H)+ = 31 5; tRet = 2.2 min; method AFEC) is obtained.
Carboxylic acid ester A*.34 (470 mg, 1 .00 mmol) is taken up in MeOH (3.0 mL) and combined with 1 N NaOH (1 .3 mL). After 5 h at 20°C the mixture is diluted with water and extracted with DCM. The organic phase is discarded, the aqueous phase is acidified with HCI (5 mL, 1 M) and extracted with DCM (3 x 5 mL). The organic phase is dried, the solvent is eliminated in vacuo and the free carboxylic acid A. 34 (149 mg, 48 %; HPLC- MS: MS(M-H)" = 299; tRet = 0.76 min; method LCMSBAS1 ) is obtained. Method of synthesising A.35
Figure imgf000064_0001
A.34 A.35 Carboxylic acid A.34 (4.00 g, 13.0 mmol) is placed in MeOH (40 mL), combined with KtBuO (30 %, 3.80 g, 32.0 mmol) and heated to 160°C for 12 min. The reaction mixture is combined with HCI (20 mL, 1 N) and DCM (30 mL), the organic phase is separated off and extracted with HCI (2 x 20 mL, 1 N). Then the organic phase is dried on Na2S04, filtered off, the solvent is eliminated in vacuo and carboxylic acid A.34 (3.62 g, 94 %; HPLC-MS: MS(M-H)" = 295; tRet = 1.00 min; method 1_FEC) is obtained.
Compounds A.36 and A.37 are repared analogously to compound A.35 (Table 3):
Figure imgf000065_0001
ethod of svnthesising A.38
Figure imgf000066_0001
A.38
Carboxylic acid ED.2 (300 mg, 2.20 mmol) is placed in cone. H2S04 (1 .3 mL), cooled to 0°C, a mixture of NaN02 (149 mg, 2.20 mmol) in cone. H2S04 (1.6 mL) is added dropwise and the mixture is stirred for 1 h. Then the reaction mixture is added dropwise to an ice- water mixture with vigorous stirring, the precipitate formed is filtered off and pyrazinonic acid Z.2 (200 mg, 66 %; MS(M-H)" = 139; t 0.00 min; method LCMSBAS1 ) is obtained.
Pyrazinonic acid Z.2 (200 mg, 1.4 mmol) is placed in MeOH (10 mL), combined with HCI (0.1 mL, 4 M in dioxane) and stirred for 12 h at 20°C. Then the solvent is removed and carboxylic acid ester A.a.4 (212 mg, 96 %; HPLC-MS: MS(M-H)" = 153; t 0.00 min; method LCMSBAS1 ) is obtained.
Sodium hydride (60 %; 61 mg, 1 .5 mmol) is placed in DMF (1 .2 mL), combined with carboxylic acid ester A.a.4 (212 mg, 1 .4 mmol) and stirred for 45 min at 20°C. Then benzyl bromide A.b.1 (0.18 mL, 1 .4 mmol) is metered in and the mixture is stirred for a further 24 h at 20°C. The reaction mixture is combined with HCI (5 mL, 1 M) and DCM (5 mL), the organic phase is separated off and extracted with HCI (2 x 5 mL, 1 N). Then the organic phase is dried on Na2S04, filtered off, the solvent is eliminated in vacuo and carboxylic acid ester A*.38 (166 mg, 43 %; HPLC-MS: MS(M+H)+ = 281 ; tRet = 1 .63 min; method FECS) is obtained. Carboxylic acid ester A*.38 (1 66 mg, 0.60 mmol) is taken up in MeOH (4.0 mL) and combined with NaOH (0.71 mL, 1 M). After 5 h at 20°C the mixture is diluted with water and extracted with DCM. The organic phase is discarded, the aqueous phase is acidified with HCI (5 mL, 1 M) and extracted with DCM. The organic phase is dried on Na2S04, filtered off, the solvent is eliminated in vacuo and the free carboxylic acid A. 38 (150 mg, 95 %; HPLC-MS: MS(M+H)+ = 267; tRet = 1.59 min; method FECS) is obtained.
Method of synthesising A.39 and A.40
Figure imgf000067_0001
A.39
A.40
Acetyldiethyl malonate A.d.2 (100 mg 0.49 mmol) is placed in cone, acetic acid (0.5 mL) and combined with benzylhydrazine A.e.1 (97 mg, 0.49 mmol). The mixture is stirred for 3 h at 95°C. Then the solvent is removed and carboxylic acid ester A*.39 (45 mg, 35 %; HPLC-MS: MS(M+H)+ = 261 ; tRel = 0.39 min; method LCMSBAS1 ) is obtained.
Carboxylic acid ester A*.39 (22 mg, 0.09 mmol) is placed in THF (0.3 mL) and combined with Cs2C03 (30 mg, 0.09 mmol). After 15 min at 20°C Mel (5 μί, 0.09 mmol) is added and the mixture is stirred for a further 16 h at 20°C. After elimination of the solvent carboxylic acid ester A*.40 (15 mg, 65 %; H PLC-MS: MS(M+H)+ = 275; tRet = 1 .24 min; method LCMSBAS1 ) is obtained.
Carboxylic acid ester A*.40 (35 mg, 0.13 mmol) is placed in THF (0.6 mL), combined with NaOH (0.5 mL, 1 N) and stirred for 4 h at 50°C. The reaction mixture is combined with HCI (3 mL, 1 N) and DCM, the organic phase is separated off, extracted with HCI (2 x 5 mL, 1 N), dried, the solvent is eliminated in vacuo and carboxylic acid A.40 (31 mg, 99 %; HPLC-MS: MS(M+H)+ = 247; tRel = 1.59 min; method FSUN) is obtained.
Compounds A.41 - A.46 are prepared analogously to compound A.40 and if necessary subjected to chiral chromatography before the saponification (Table 4):
HPLC method
1
1
1
Figure imgf000068_0001
MS (M+Hf ;
od 1
Figure imgf000069_0001
*Gilson HPLC apparatus, chiral column (Daicel Chiralpack IC, 250 x 20 mm). Eluant: 35 % n-heptane, 65 % DCM/EtOH/diethylamine (2000:100:2.6) b) Synthesis of propargylamides C
Reaction scheme B
Figure imgf000069_0002
The amides C required are synthesised by coupling a propargylamine B with the carboxylic acid A. The amide coupling is assisted by coupling reagents such as for example DCC, DIC, TBTU, HATU, EDC or the like, or the formation of the corresponding acid chloride.
The synthesis components which are to be used in the above reaction schemes are optionally provided with common protective groups when used. Therefore, additional intermediate steps may be needed to eliminate these protective groups.
With regard to the feasibility of the reaction methods illustrated and described in the above reaction schemes reference is made to WO 2008/005457. In the cited specification, pyridinonecarboxylic acids A are amidated in a variety of ways. Method of synthesising C.1
Figure imgf000070_0001
C.1
Carboxylic acid A.25 (1 .00 g, 3.75 mmol) is placed in DCM (10 mL), combined with TBTU (1 .73 g, 5.36 mmol), propargylamine B.1 (0.29 mL, 4.10 mmol) and NEt3 (1 .3 mL, 9.5 mmol) and stirred for 12 h at 20°C. The reaction mixture is combined with water and DCM, the organic phase is separated off and extracted 2 x with water. Then the organic phase is dried on Na2S04, the solvent is eliminated in vacuo, the crude product is purified by chromatography (90: 10 to 60:40 in 12 min H20/CH3CN) and carboxylic acid amide C.1 (940 mg, 83 %; H PLC-MS: MS(M+H)+ = 304; = 1 -45 min; method LCMSBAS1 ) is obtained.
Propargylamides C.2 - C.43 are prepared analogously to propargylamide C.1 (Table 5).
MS (M+HT;
# Structure tRet. HPLC [min] HPLC method
M+H= 304; tRet = 1 .45 LCMSBAS1
M+H= 267; tRet = 1.43 LCMSBAS1
Figure imgf000070_0002
MS (M+Hf ;
# Structure tRet. HPLC [min] HPLC method
M+H= 285; tRet = 1.42 LCMSBAS1
M+H= 285; tRet = 1.45 LCMSBAS 1
M+H= 303; tRet = 1.53 LCMSBAS1
M+H= 317; tRet = 1.66 LCMSBAS 1
M+H= 317; tRet = 1.66 LCMSBAS 1
Figure imgf000071_0001
Figure imgf000072_0001
Figure imgf000073_0001
Figure imgf000074_0001
MS (M+Hf ;
# Structure tRet. HPLC [min] HPLC method
M+H= 307; tRet = 1.62 LCMSBAS1
M+H= 334; tRet = 1.62 LCMSBAS1
M+H= 348; tRet = 1.93 LCMSBAS1
M+H= 372; tRet = 1.80 LCMSBAS1
M+H=334; tRet = 1.88 LCMSBAS1
M+H= 390; tRet = 1.86 1_FEC
Figure imgf000075_0001
Figure imgf000076_0001
MS (M+Hf ;
# Structure tRet. HPLC [min] HPLC method
C.40 0.98 1 FEC
1.23 LCMSBAS1
Figure imgf000077_0001
c) Synthesis of quinazoline, quinoxaline and quinoline compounds D (EWG-QH) Reaction scheme C
^ J©— *~ EWG J©—
EWG
cf. reaction scheme E cf. reaction scheme D
Figure imgf000078_0001
The ring systems QH of compounds according to the invention, i.e. embodiments QH-2a to QH-2f, can be incorporated in the target structures by various methods. Starting from bisfunctional components D*, which are activated by an electron-withdrawing group EWG, particularly chlorine, bromine or iodine, and a further group which enables the substituents R5, R6, R7 and R8 to be inserted in their respective positions, first of all the substituent R5, R6, R7 or R8 can be introduced and in this way the component D is obtained (QH* -> QH). D is then reacted in a SONOGASHIRA reaction with C to form the compound according to the invention (1 ) (cf. also reaction scheme D). In an alternative sequence first D* may be reacted with C in the SONOGASHIRA reaction and only then is the substituent R5, R6, R7 or R8 introduced (cf. also reaction scheme F). The position of the group EWG on the ring system QH/QH* denotes the linkage point of the ring system to the linker unit, i.e. in this case to the C-C triple bond.
As another alternative it is possible to carry out the SONOGASHIRA reaction between N-protected propargylamines B and D* or D and couple the intermediates R4-NH-L-QH or R4-NH-L-QH* obtained with the acids A (cf. this method of inserting alkenyl linkers, reaction schemes G and H). The final substituents R5, R6, R7 or R8 (QH -> QH*) are introduced for example by nucleophilic substitution, SUZUKI coupling, amidation (optionally activation with DCC, DIC, TBTU, HATU, EDC or the like), reductive alkylation or, however, during the ring formation of the corresponding system QH. Other possibilities are BUCHWALD-HARTWIG, NEGISHI , HECK or SONOGASHIRA reactions. The activating groups that are suitable for these reactions are well known in the art.
The synthesis of bisfunctional components D*which are suitable for incorporating the ring systems QH in compounds (1 ) according to the invention, is described in the literature. They may be prepared using published methods or can be directly obtained commercially: QH-2b: e.g. Tetrahedron Lett. 2002, 6209; WO 2007/133637;
QH-2c: e.g. WO 2007/104560; WO 2008/020302; WO 2006/071017
QH-2d: e.g. WO 2008/138878;
QH-2e: e.g. US 4.996.213 (S.3) and J. Org. Chem. 1971 , 1331 ; Polish J. Chem. 1983,
587;
QH-2f : e.g. US 2009/036430; WO 2009/003669; WO 2006/135993;
Method of synthesising the quinazolines D*.1 and D.1
Figure imgf000080_0001
Anthranilic acid ED.3 (32.5 g, 21 1 mmol) is placed in glacial acetic acid (400 mL), and ICI (35.0 g, 21 1 mmol) in glacial acetic acid (100 mL) is added dropwise thereto. The reaction mixture is stirred for 1 h at 20°C, then diluted again with glacial acetic acid (250 mL) and stirred for 1 h at 20°C. The reaction mixture is filtered, the filter cake is washed with glacial acetic acid and acetonitrile and anthranilic acid Z.3 (45.3 g, 70 %; HPLC-MS: MS(M-H)" = 276; tRet = 0.00 min; method LCMSBAS1 ) is obtained.
Anthranilic acid Z.3 (17.3 g, 50.0 mmol) and formamidine acetate (25.0 g, 238 mmol) are placed in EtOH (500 mL) and heated to 95°C for 6 h. Then the reaction mixture is cooled to 50°C, the precipitate is filtered off, the filter cake is washed with EtOH and quinazolone Z.4 (1 1 .8 g, 69 %; HPLC-MS: MS(M-H)" = 285; tRel = 1 .13 min; method LCMSBAS1 ) is obtained.
Quinazolone Z.4 (451 mg, 1 .51 mmol) is combined with DIPEA (0.5 m L) and POCI3 (2.5 mL, 26.7 mmol) and heated to 125°C for 1 .5 h in the MW reactor. Then the reaction mixture is evaporated to dryness and quinazoline D*.1 (456 mg, 100 %) is obtained.
Quinazoline D*.1 (530 mg, 1.70 mmol) is combined with methylamine (15 mL, 2M in THF) in the ultrasound bath and stirred for 1 h at 20°C. The solvent is removed, the crude product is purified by chromatography (EtOAc) and quinazoline D.1 (320 mg, 62 %; HPLC-MS: MS(M+H)+ = 300; tRet = 1 .43 min; method LCMSBAS1 ) is obtained. Method of svnthesising the azaquinazolines D*.2 and D.2
Figure imgf000081_0001
D.2 D*.2 Z.7
Pyridine ED.4 (10.0 g, 54.5 mmol) is taken up in H2S04 (150 mL, 90 %) and heated to 70°C for 3.5 h. Then the reaction mixture is cooled to 20°C, added dropwise to ice water, the precipitate is filtered off, washed with H20, dried and pyridine Z.5 (10.5 g, 95 %) is obtained.
Pyridine Z.5 (2.00 g, 1 0.0 mmol) and iron filings (2.77 g, 49.6 mol) are taken up in EtOH/H20 (100 mL, 1 :1 ), combined with NH4CI (265 mg, 4.91 mmol) and heated to 60°C for 1 h. The solvent is removed, the residue is taken up in DCM/MeOH (50 mL, 9:1 ), filtered through Celite, the solvent is eliminated again and pyridine Z.6 (1 .57 g, 92 %) is obtained.
Pyridine Z.6 (500 mg, 2.91 mmol) is taken up in triethylorthoformate (10 mL) and heated to 145°C for 18 h. Then the reaction mixture is cooled to 20°C, the precipitate is filtered off, washed with Et20 (10 mL), dried and azaquinazolone Z.7 (433 mg, 82 %) is obtained. Azaquinazolone Z.7 (200 mg, 1 .10 mmol) is placed in toluene (5.0 mL), combined with Λ/,/V-diethylaniline (0.27 mL) and POCI3 (0.88 mL) and heated to 1 10°C for 4 h. The reaction mixture is cooled to 20°C, diluted with toluene (5 mL) and H20 (5 mL), washed with NaOH (3 x 10 mL, 20 %) and HCI (2 x 10 mL, 1 M), dried on Na2S04, filtered, freed from the solvent and azaquinazoline D*.2 (165 mg, 75 %) is obtained.
Azaquinazoline D*.2 (1 .15 g, 5.75 mmol) is placed in DCM (5.0 mL), cooled to 0°C, combined with methylamine (3.5 mL, 7.19 mmol, 2 M in THF) and DIPEA (3.0 mL), the reaction mixture is heated to 20°C and stirred for 1 h at 20°C. The solvent is removed, the residue is taken u p in H20, the precipitate is filtered off, washed with H20 and azaquinazoline D.2 (640 mg, 57 %; HPLC-MS: MS(M+H)+ = 195; tRet. = 1 .85 min; method 1_BAS).
Method of synthesising the quinazolines D*.3 and D.3
Figure imgf000082_0001
D.3
Anthranilic acid ED.5 (1 .00 g, 4.27 mmol) is placed in 2-methoxyethanol (2.0 mL), combined with formamidine acetate (890 mg, 8.55 mmol) and heated to 125°C for 20 h. The reaction mixture is added to aqueous NH3 (200 mL, 0.01 M), the precipitate is filtered off, dried and quinazolone Z.8 (817 mg, 79 %) is obtained.
Quinazolone Z.8 (817 mg, 3.36 mmol) is combined with SOCI2 (20 mL) and DMF (0.05 mL) and heated to 80°C for 2 h. The solvent is removed and quinazoline D*.3 (879 mg, 100 %) is obtained.
Quinazoline D*.3 (879 mg, 3.36 mmol) is combined with methylamine (30 mL, 2 M in THF) and stirred for 1 h at 20°C. Th e solvent is eliminated, the residue is purified by chromatography (35:65 to 80:20 in 12 min MeOH/H20) and quinoline D.3 (555 mg, 65 %; HPLC-MS: MS(M+H)+ = 256/258; tRet = 1.86 min; method FECBM4) is obtained. Method of svnthesising quinazoline D.4
Figure imgf000083_0001
Aminoalcohol ED.6 (98 μΙ_, 0.76 mmol) is placed in DMF, combined with K2C03 (105 mg, 0.76 mmol) and stirred for 5 min at 20°C. Then quinazoline D*.4 (200 mg, 0.69 mmol) is added and the mixture is stirred for 30 min at 150°C. The solvent is removed and D.4 (24 mg, 10 %; HPLC-MS: MS(M+H)+ = 358; tRet = 1.78 min; method LCMSBAS1 ) is obtained.
Compounds D.5 - D.14 are repared analogously to quinazolines D.1 to D.4 (Table 6):
Figure imgf000083_0002
Figure imgf000084_0001
Method of svnthesising quinoxaline D.15
Figure imgf000085_0001
Quinoxaline D*.5 (100 mg, 0.41 mmol) is placed in isopropanol (0.5 mL), mixed with aminopiperidinethanol ED.7 (296 mg, 2.05 mmol) and DIPEA (0.7 mL) and stirred for 24 h at 100°C. The solvent is removed, the residue is purified by chromatography (95:5 to 50:50 in 6 min H20/CH3CN) and quinoxaline D.15 (46 mg, 32 %) is obtained.
Com ounds D.16-D.50 are prepared analogously to quinoxaline D.15 (Table 7):
Figure imgf000085_0002
tRet. HPLC [min]
2.08 LCMSBAS1 1.84 LCMSBAS1 2.03 LCMSBAS1 2.16 LCMSBAS1 2.18 LCMSBAS1 1.70 LCMSBAS1 1.58 LCMSBAS1 1.60 LCMSBAS1 1.63 LCMSBAS1 1.70 LCMSBAS1 1.93 LCMSBAS1
Figure imgf000086_0001
□ 32 ί ί ϋ M+H=306/308; tRet = 2.01 LCMSBAS1
N N Br tRet. HPLC [min]
D.33 M+H=307/309; tRet = 1.31 LCMSBAS1
D.34 M+H=335/337; tRet = 1.75 LCMSBAS1 M+H=347/349; tRet.= 1.77 LCMSBAS1
D.36 M+H=321/323; tRet = 1.76 LCMSBAS1
D.37 M+H=321/323; tRet = 1.76 LCMSBAS1
M+H=266/268; tRet = 1.81 LCMSBAS1
M+H=322/324; tRet = 1.56 LCMSBAS1
M+H=365/367; tRet = 1.35 LCMSBAS1 M+H=351/353; tRet = 1.31 LCMSBAS1
D 42 M+H=322/324; tRet = 1.34 LCMSBAS1
D.43 M+H=322/324; tRet = 1.70 LCMSBAS1
D 44 M+H=391/393; tRet.=1.67 LCMSBAS1
Figure imgf000087_0001
Structure . ^,Κΐ '■ , HPLC method
tRet. HPLC [min]
D 45 M+H=444/446; tRet.= 1.75 LCMSBAS1
M+H=349/351 ; tRet.= 1.52 LCMSBAS1
M+H=307/309; tRet = 1.63 LCMSBAS1
D.48 M+H=353/355; tRet = 1.40 LCMSBAS1 M+H=353/355; tRet = 1.40 LCMSBAS1
M+H=363/365; tRet = 1.31 LCMSBAS1
Figure imgf000088_0001
Method of svnthesising quinoxaline D.51
NBoc
ED.8
Figure imgf000088_0002
D*.5 D.51
Methylene piperidine ED.8 (243 mg, 1 .23 mmol) is combined with 9-BBN (2.46 mL, 0.5 M in THF) and heated to 65°C for 1 h. Then the mixture is cooled to 20°C, quinoxaline D*.5 (300 mg, 1 .23 mmol), Pd(dppf) (50 mg, 10 mol %) and K2C03 (195 mg, 1 .41 mmol) are placed in DMF/H20 (1 .2 mL, 5: 1 ) and slowly the freshly prepared boronate is added dropwise. The reaction mixture is heated to 60°C for 2 h, cooled, combined with NaHC03 and extracted with DCM (3 x 10 mL). The solvent is removed, the residue is purified by chromatography (45:55 to 90: 1 0 in 6 min CH3CN/H20) and quinoxaline D.51 (250 mg, 50 %) is obtained.
Method of synthesisin quinoxaline D.52
Figure imgf000089_0001
THF/NMP
D*.5 D.52
Boric acid ester ED.9 (0.07 mL, 0.35 mmol), quinoxaline D*.5 (50 mg, 0.21 mmol), Pd(dppf) (16 mg, 10 mol %) and Cs2C03 (0.12 mL, 0.67 mmol, 70 % in H20) are placed in THF/NMP (10.6 mL, 2:1 ) and the reaction mixture is heated to 50°C for 1 h . Then the reaction mixture is cooled to 20°C, combined with NaHC03 and extracted with DCM (3 x 10 mL). The solvent is removed, the residue is purified by chromatography (90:10 to 80:20 in 20 min hexane/EtOAc) and quinoxaline D.52 (31 mg, 61 %) is obtained.
Method of synthesisin quinoxaline D.53
Figure imgf000089_0002
D*.5 D.53
Boric acid ester ED.10 (0.07 mL, 0.35 mmol), quinoxaline D*.5 (50 mg, 0.21 mmol), Pd(dppf) (16 mg, 10 mol %) and Cs2C03 (0.12 mL, 0.67 mmol, 70 % in H20) are placed in THF/NMP (10.6 mL, 2:1 ) and the reaction mixture is heated to 50°C for 1 h. The reaction mixture is cooled to 20°C, combined with NaHC03 and extracted with DCM (3 x 10 mL). The solvent is removed, the residue is purified by chromatography (80:20 to 70:30 in 20 min hexane/EtOAc) and quinoxaline D.53 (35 mg, 65 %) is obtained. ethod of svnthesising quinoline D.54
Figure imgf000090_0001
D*.6 0 54
Quinoline D*.6 (50 mg, 0.17 mmol) is placed in DCM (1 .0 mL), combined with DIPEA (0.06 mL, 0.34 mmol) and piperidinone ED.11 (0.1 mL, 0.85 mmol) and stirred for 1 h at 20°C. Then CH3COOH (0.48 mL) and NaBH(OAc)3 (268 mg, 1 .27 mmol) are added and the mixture is stirred for 3 h at 20°C. The solvent is removed, the residue is purified by chromatography (20:80 to 80:20 in 12 min CH3CN/H20) and quinoline D.54 (50 mg, 92 %; HPLC-MS: MS(M+H)+ = 320/322; tRel = 1.37 min; method FSUN2) is obtained.
Compounds D.55 and D.56 are prepared analo ously to quinoline D.54 (Table 8):
Figure imgf000090_0002
Method of synthesising quinoline D.57
Figure imgf000091_0001
Ethoxyacrylate ED.13 (6.4 mL, 44.3 mmol) is placed in MeOH (120 mL), combined with HCI (88 mL, 2M in H20) and heated to 100°C. Benzaldehyde ED.12 (4.00 g, 20.1 mmol) is placed in MeOH (120 mL), slowly added dropwise to the reaction mixture and the reaction mixture is heated to 100°C for 4 h. Then it is made basic with NaHC03, the solvent is eliminated, the residue is purified by chromatography (90:10 to 40:60 in 15 min hexane/EtOAc) and quinoline D.57 (4.08 g, 51 %; HPLC-MS: MS(M+H)+ = 266/268; tRet = 1 .50 min; method LCMSBAS1 ) is obtained.
Compounds D.58 and D.59 are repared analogously to quinoline D.57 (Table 9):
Figure imgf000091_0002
ethod of svnthesising quinoline D.60
Figure imgf000092_0001
D.60
Quinoline D.58 (500 mg, 1 .58 mmol) is placed in dioxane (5.0 mL), combined with LiOH (3.2 mL, 3.2 mmol, 1 M in H20) and stirred for 20 min at 20°C. Then the mixture is diluted with H20, adjusted to pH = 1 with HCI and the carboxylic acid Z.9 is filtered off.
The carboxylic acid Z.9 is taken up in DMF (0.5 mL) and stirred with S02CI2 (5.0 mL) for 40 min at 20°C. The solvent is removed and the acid chloride Z.10 (495 mg, 100 %) is further reacted directly.
Dimethylamine (1 .9 mL, 3.7 mmol, 2 M in THF) is placed in THF (10 mL), cooled to -10°C and combined with acid chloride Z.10 (215 mg, 0.76 mmol). The reaction mixture is heated to 20°C over 1 h, then diluted with H20 (10 mL), extracted with DCM (3 x 20 mL), dried on MgS04, filtered, the solvent is eliminated and quinoline D.60 (185 mg, 84 %; HPLC-MS: MS(M+H)+ = 293/295; tRel = 1.48 min; method LCMSBAS1 ) is obtained.
Compound D.61 is prepared analogously to quinoline D.60 (Table 10):
MS (Μ+Η ;
# Structure HPLC method
tRet. HPLC [min]
D.60 LCMSBAS1
o D.61 M+H=279/281 ; tRet = 1 .45 LCMSBAS1
Figure imgf000093_0001
Method of synthesising quinoline D*.7
Figure imgf000093_0002
D.57 D*.7
Quinolinester D.57 (3.80 g, 9.46 mmol) is placed in THF (50 mL), cooled to -20°C and combined with DIBAL-H (57.0 mL, 56.9 mmol , 1 M in DCM). The reaction mixture is heated to 20°C over 2 h, then it is diluted with H20, the solvent is eliminated, the residue is purified by chromatography (90:10 to 40:60 in 15 min hexane/EtOAc) and quinoline D*.7 (716 mg, 29 %) is obtained.
Method of synthesising quinoline D*.8
Figure imgf000093_0003
D*.7 D*.8
Alcohol D*.7 (716 mg, 2.70 mmol) is placed in DCM (9 mL), combined with Mn02 (705 mg, 8.12 mmol) and stirred for 2 d at 20°C. The reaction mixture is filtered through Celite, the solvent is eliminated and product D*.8 (577 mg, 63 %) is obtained.
Method of synthesising quinoline D.62
N\___ N— \
Figure imgf000093_0004
D*.8 D.62
Aldehyde D*.8 (82 mg, 0.35 mmol) is placed in DCM (1 .5 mL), combined with amine ED.14 (0.18 mL, 1 .39 mmol) and CH3COOH (0.09 mL) and stirred for 10 min at 20°C. Then NaBH(OAc)3 (1 10 mg, 0.52 mmol) is added, the mixture is stirred for 12 h at 20°C, the solvent is eliminated, the residue is purified by chromatography (20:80 to 80:20 in 15 min CH3CN/H20) and quinoline D.62 (68 mg, 59 %) is obtained.
Compounds D.63 - D.67 are repared analogously to quinoline D.62 (Table 1 1 ):
Figure imgf000094_0001
Method of synthesising quinoline D.68
Figure imgf000094_0002
NaH (43 mg, 1.07 mmol, 60 %) is placed in NMP (2.0 mL), combined with aminoalcohol ED.15 (0.17 mL, 0.91 mmol) and quinoline D*.9 (200 mg, 0.83 mmol) and stirred for 12 h at 20°C. Then the reaction mixture is diluted with H20 (5 mL), extracted with DCM (3 x 5 mL), dried on MgS04, filtered, the solvent is eliminated and quinoline D.68 (255 mg, 90 %; HPLC-MS: MS(M+H)+ = 309/31 1 ; tRel = 1.86 min; method LCMSBAS1 ) is obtained.
Compounds D.69 - D.91 -9 are prepared analogously to quinoline D.68 (Table 12):
Figure imgf000095_0001
MS (M+Hf ;
tructure HPLC method
tRet. HPLC [min] S1
Figure imgf000096_0001
MS (Μ+Η ;
tructure HPLC method
tRet, HPLC [min] tRet.= 2.26 BFEC ; tRet = 1.50 BFEC ; tRet.= 0.15 AFEC ; tRet = 1.63 AFEC ; tRet = 175 AFEC ; tRet = 2.24 AFEC Ret =1.34 LCMSBAS1 Ret =1.34 LCMSBAS1
Figure imgf000097_0001
Figure imgf000098_0001
Method of svnthesising quinoline D.91 -11
Figure imgf000099_0001
D.91-9 D.91-11
Quinoline D.91 -9 (100 mg, 0.23 mmol) is taken up in DMF (3ml_), combined with K2C03 (94 mg, 1 mmol) and stirred for 5 min at RT. Then ED.15-1 (45 mg, 0.24 mmol) is added and the mixture is stirred for 12 h at RT. Then K2C03 (94 mg, 1 mmol) and chloride ED.15-1 (45 mg, 0.24 mmol) are added once again and the mixture is stirred for a further 12 h at RT, then 48 h at 60°C. The reaction mixture is diluted with H20 (5 mL), extracted with EtOAc (3 x 5 mL), dried on MgS04, filtered, the solvent is removed, the crude product is purified by chromatography (10:90 to 80:20 in 10 min CH3CN/H20) and quinoline D.91 - 11 (HPLC-MS: MS(M+H)+ = 482; tRel = 1.36 min; method LCMSBAS1 ) is obtained.
Compounds D.91 -12 and D.91 -13 are prepared analogously to quinoline D.91 -11 (Table 12a):
MS (Μ+Η ;
# Structure HPLC method
tRet. HPLC [min]
Figure imgf000099_0002
M+H=482; tRet = 1.36 LCMSBAS1
Figure imgf000099_0003
tructure t tRe M t.S HPLrC+ [m'in i] HPLC method
D.91-12 M+H=363; tRet.= 2.16 AFEC
D.91-13 M+H=383; tRet = 1.92 LCMSBAS1
Figure imgf000100_0001
Method of synthesising quinoline D*.1 Q
Figure imgf000100_0002
D*.10
Aniline ED.16 (5.00 g, 22.4 mmol) and cyanoacrylate ED.17 (3.86 g, 22.4 mmol) are placed in toluene (12 mL) and heated to 150°C for 18 h. Then the reaction mixture is cooled to 20°C, the precipitate is filtered off, washed with diethyl ether (12 mL), dried, and intermediate Z.11 (6.46 g, 84 %) is obtained.
Some of the intermediate Z.11 (1 .54 g, 4.50 mmol) is taken up in Ph20 (10 mL) and heated to 250°C for 18 h. Then the reaction mixture is cooled to 20°C, the precipitate is filtered off, washed with diethyl ether (12 mL), dried and quinolinone Z.12 (930 mg, 70 %) is obtained.
Quinolinone Z.12 (930 mg, 3.14 mmol) is taken up in POCI3 (3.0 mL) and the mixture is heated to 160°C for 5 h. The solvent is removed, the residue is combined with NaHC03 (10 mL) and ice water, the precipitate is filtered off, washed with H20 (1 0 m L) and quinoline DM 0 (908 mg, 92 %) is obtained.
Method of synthesising quinoline D*.11
Figure imgf000101_0001
Anthranilic acid ED.18 (5.06 g, 19.2 mmol) is stirred in cone. HCI (7.0 mL) for 12 h at 20°C. In a second round flask nitromethane (2.56 g, 42.0 mmol) is added dropwise to a mixture of ice (6.0 g, 333 mmol) and NaOH (2.53 g, 63.4 mmol), stirred for 1 h at 0°C, then heated to 20°C over 1 h , then poured onto an ice/HCI mixture (5 g/7 mL) and combined with the aniline-HCI mixture. The reaction mixture is stirred for 12 h at 20°C, the precipitate is filtered off, washed with H20 and dried.
The precipitate is taken up in acetic anhydride (35 mL), combined with KOAc (2.19 g, 22.3 mmol) and heated to 120°C for 2 h. The solvent is removed and the residue is washed with glacial acetic acid and H20 and dried.
The residue is taken up in POCI3 (20 mL, 209 mmol) and heated to 120°C for 40 min. The reaction mixture is evaporated down to one third, poured onto ice water, washed with H20, dried and quinoline D*.11 (4.0 g, 95 %) is obtained. Method of synthesising quinoline D.92
Figure imgf000102_0001
D*.11 D.92
Quinoline D*.11 (1 .00 g, 2.99 mmol) is placed in DMSO (0.5 mL), combined with ethylamine (202 mg, 4.49 mmol) and NEt3 (1 .81 g, 15.0 mmol) and stirred for 2 h at 20°C and 4 h at 50°C. The precipitate is filtered off, washed with CH3CN (5 mL), dried and quinoline D.92 (450 mg, 43 %; HPLC-MS: MS(M+H)+ = 344; tRet = 1 .58 min; method LCMSBAS1 ) is obtained.
Compounds D.93 and D.94 are repared analogously to quinoline D.92 (Table 13):
Figure imgf000102_0002
d) Synthesis of compounds according to the invention (1 ) Reaction scheme D
Figure imgf000103_0001
C and D are linked together by SONOGASHIRA reaction. The electron withdrawing groups EWG may be in particular halogen (chlorine, bromine, iodine), triflate or mesylate (cf. also reaction scheme C).
Method of synthesising 1-1
Figure imgf000103_0002
D.4 1-1
Iodide D.4 (24 mg, 0.07 mmol), propargylamide C.1 (24 mg, 0.08 mmol), Pd(PPh3)4 (1 .5 mg, 10 mol %) and Cul (0.2 mg, 10 mol %) are placed in DMSO (0.3 ml_), combined with DIPEA (0.07 mL) and stirred for 1 h at 40°C. The reaction mixture is mixed with water and DCM, the organic phase is separated off and extracted another 2 x with water. Then the organic phase is dried, the solvent is eliminated in vacuo, the crude product is purified by chromatography (20:80 to 80:20 in 15 min CH3CN/H20) and quinazoline 1-1 (6 mg, 17 %; HPLC-MS: MS(M+H)+ = 533; tRet = 1.85 min; method LCMSBAS1 ) is obtained.
Compounds I-2 - I-8 may be prepared analogously to compound 1-1 (Table 14): 103
Figure imgf000104_0001
Figure imgf000105_0001
Method of svnthesising quinoxaline 1-9
Figure imgf000106_0001
1-9
Quinoxaline D.15 (46 mg, 0.13 mmol), propargylamide C.1 (50 mg, 0.16 mmol), Pd(dppf) (6 mg, 10 mol %) and Cul (6 mg, 20 mol %) are placed in DMSO (0.5 mL), combined with DIPEA (0.28 mL) and stirred for 3 h at 90°C. The solvent is removed, the residue is purified by chromatography (10:90 to 60:40 in 6 min CH3CN/H20) and product I-9 (38 mg, 42 %; HPLC-MS: MS(M+H)+ = 574; tRel = 1.69 min; method LCMSBAS1 ) is obtained.
Compounds 1-10 - I-65 are prepared analogously to quinoxaline I-9 (Table 15):
MS (M±H)*; HPLC
tRet. HPLC [min] method
M+H=574; tRet = 1.69 LCMSBAS1 106
Figure imgf000107_0001
107
Figure imgf000108_0001
108
Figure imgf000109_0001
Figure imgf000110_0001
110
Figure imgf000111_0001
111
Figure imgf000112_0001
112
Figure imgf000113_0001
113
Figure imgf000114_0001
114
Figure imgf000115_0001
115
Figure imgf000116_0001
Figure imgf000117_0001
method
08 LCMSBAS1
LCMSBAS1
77 LCMSBAS1
88 LCMSBAS1
Figure imgf000117_0002
Figure imgf000118_0001
118
Figure imgf000119_0001
MS (M±H)r; HPLC
Structure
tRet. HPLC [min] method
M-H=597; tRet = 1 .85 LCMSBAS1
M+H=607; tRet = 170 LCMSBAS1
M+H=614; tRet = 1.95 LCMSBAS1
M+H=678; tRet = 1.90 LCMSBAS1
Figure imgf000120_0001
120
Figure imgf000121_0001
Method of svnthesising quinoxaline 1-66
Figure imgf000122_0001
1-66
Quinoxaline D.51 (250 mg, 0.62 mmol), propargylamide C.1 (205 mg, 0.68 mmol), Pd(dppf) (30 mg, 10 mol %) and Cul (26 mg, 20 mol %) are placed in DMSO (2.0 mL), combined with DIPEA (1 .2 mL) and stirred for 3 h at 50°C. The solvent is removed, the residue is purified by chromatography (30:70 to 95:5 in 6 min CH3CN/H20) and the protected quinoxaline Boc-l-66 (372 mg, 88 %) is obtained.
Boc-l-66 (372 mg, 0.59 mmol) is placed in DCM (10.0 mL), combined with TFA (1 .0 mL) and stirred for 1 h at 20°C. It is made basic with NaHC03 (10 mL), extracted with DCM (3 x 20 m L), the solvent is removed and product I-66 (260 mg, 83 %; HPLC-MS: MS(M+H)+ = 529; tRet = 2.00 min; method LCMSBAS1 ) is obtained. ethod of svnthesising quinoxaline 1-67
Figure imgf000123_0001
1-66 1-67
Quinoxaline 1-66 (35 mg, 0.07 mmol) is placed in DMA (0.8 mL), combined with formaldehyde (5.9 μΙ_, 37 %, 0.07 mmol) and acetic acid (4 μΙ_, 0.07 mmol) and stirred for 1 h at 20°C. Then NaBH(OAc)3 (16 mg, 0.07 mmol) is added and the mixture is stirred for another 12 h at 20°C. It is diluted with MeOH (10 mL), then the solvent is eliminated, it is purified by chromatography (20:80 to 80:20 in 6 min CH3CN/H20) and quinoxaline I-67 (16 mg, 45 %; HPLC-MS: MS(M+H)+ = 543; tRet = 1 .91 min; method LCMSBAS1 ) is obtained.
Compounds 1-68 and 1-69 are prepared analogously to quinoxaline 1-67 (Table 16):
MS (M+H) ; HPLC
Structure
tRet. HPLC [min] method
I -66 M+H=529; tRet = 2.00 LCMSBAS1 
Figure imgf000124_0001
Method of svnthesising quinoline 1-70
Figure imgf000125_0001
1-70
Quinoline D.54 (50 mg, 0.18 mmol), propargylamide C.1 (55 mg, 0.16 mmol), Pd(dppf) (7 mg, 10 mol %) and Cul (7 mg, 20 mol %) are placed in DMSO (0.40 mL), combined with DIPEA (0.31 mL) and stirred for 3 h at 90°C. The solvent is removed, the residue is purified by chromatography (20:80 to 80:20 in 6 min CH3CN/H20) and quinoline I-70 (28 mg, 28 %; HPLC-MS: MS(M+H)+ = 543; tRet = 1 .80 min; method LCMSBAS1 ) is obtained.
Com ounds 1-71 - I-78 are prepared analogously to quinoline I-70 (Table 17):
Figure imgf000125_0002
Figure imgf000126_0001
method
91 LCMSBAS1
97 LCMSBAS1
86 LCMSBAS1
Figure imgf000126_0002

Figure imgf000127_0001
Figure imgf000128_0001
Method of svnthesising quinoline 1-79
Figure imgf000128_0002
Quinoline D.59 (60 mg, 0.17 mmol), propargylamide C.1 (58 mg, 0.19 mmol), Pd(PPh)3 (20 mg, 10 mol %) and Cul (3 mg, 10 mol %) are placed in DMSO (1 .0 mL), combined with DIPEA (0.1 1 mL) and stirred for 30 min at 100°C. The solvent is removed, the residue is purified by chromatography (20:80 to 80:20 in 6 min CH3CN/H20) and quinoline I-79 (49 mg, 54 %; HPLC-MS: MS(M+H)+ = 521 ; tRet = 2.21 min; method LCMSBAS1 ) is obtained.
Compounds I-80 - I-87 are prepared analogously to quinoline I-79 (Table 18): 
Figure imgf000129_0001

Figure imgf000130_0001
Figure imgf000131_0001
Method of svnthesising quinoline I-88
Figure imgf000131_0002
I-88
Quinoline D.68 (61 mg, 0.20 mmol), propargylamide C.1 (83 mg, 0.21 mmol), Pd(PPh)3 (16 mg, 10 mol %) and Cul (3 mg, 10 mol %) are placed in DMSO (1 .0 mL), combined with DEA (0.1 mL) and stirred for 20 min at 80°C. The solvent is removed, the residue is purified by chromatography (99:1 to 80:20 in 6 min DCM/MeOH) and quinoline I-88 (18 mg, 17 %; HPLC-MS: MS(M+H)+ = 532; tRet = 1 .84 min; method LCMSBAS1 ) is obtained.
Compounds I-89 - 1-125.23 are prepared analogously to quinoline I-88 (Table 19): 131
Figure imgf000132_0001
ı32
Figure imgf000133_0001
ı33
Figure imgf000134_0001

Figure imgf000135_0001

Figure imgf000136_0001

Figure imgf000137_0001

Figure imgf000138_0001

Figure imgf000139_0001
Figure imgf000140_0001
method
LCMSBAS1
LCMSBAS1
LCMSBAS1
LCMSBAS1
LCMSBAS1
Figure imgf000140_0002
Figure imgf000141_0001
method
LCMSBAS1
LCMSBAS1
LCMSBAS1
LCMSBAS1
LCMSBAS1
Figure imgf000141_0002
Figure imgf000142_0001
method
04 LCMSBAS1
10 LCMSBAS1
83 LCMSBAS1
84 LCMSBAS1
60 LCMSBAS1
Figure imgf000142_0002
Figure imgf000143_0001
method
LCMSBAS1
LCMSBAS1
LCMSBAS1
Figure imgf000143_0002
ı43
Figure imgf000144_0001
Figure imgf000145_0001
Quinoline D.92 (350 mg, 1.02 mmol), propargylamide C.1 (371 mg, 1 .22 mmol), Pd(PPh)3 (60 mg, 10 mol %) and Cul (2 mg, 5 mol %) are placed in DMSO (1 .0 mL), combined with DIPEA (0.54 mL) and stirred for 30 min at 50°C. The solvent is removed, the residue is taken up in H20 (5 mL), extracted with DCM (3 x 5 mL), dried on MgS04, filtered, the solvent is removed, the residue is purified by chromatography (95:5 to 40:60 in 6 min H20/MeOH) and product 1-126 (306 mg, 60 %; HPLC-MS: MS(M+H)+ = 519; tRet = 1 .88 min; method LCMSBAS1 ) is obtained. Com ounds 1-127 and 1-128 are prepared analogously to quinoline 1-126 (Table 20):
Figure imgf000145_0002
Figure imgf000146_0001
e) Synthesis of quinazoline, quinoxaline and quinoline compounds E
Reaction scheme E
Figure imgf000146_0002
C and D* are linked by SONOGASHIRA reaction. Electron-withdrawing groups EWG that may be used include in particular halogen (chlorine, bromine, iodine), triflate or mesylate (cf. also reaction scheme C).
Method of synthesising quinazoline E.1
Figure imgf000146_0003
E.1
Iodide D*.4 (1 .60 g, 5.00 mmol), propargylamide C.1 (1 .71 g, 5.50 mmol), Pd(PPh3)4 (1 17 mg, 10 mol %) and Cul (20 mg, 10 mol %) are placed in DMSO (20 mL), combined with DIPEA (5 mL) and stirred for 3 h at 40°C. The reaction mixture is combined with water and DCM, the organic phase is separated off and extracted another 2 x with water. Then the organic phase is dried, the solvent is eliminated in vacuo, the crude product is purified by chromatography and quinazoline E.1 (2.12 g, 46 %; HPLC-MS: MS(M-H)" = 464; tRet.= 1.93 min; method LCMSBAS1 ) is obtained.
Method of synthesising quinoline E.2
Figure imgf000147_0001
Quinoline D*.12 (50 mg, 0.18 mmol), propargylamide C.1 (55 mg, 0.16 mmol), Pd(dppf) (7 mg, 10 mol %) and Cul (7 mg, 20 mol %) are placed in DMSO (0.40 mL), combined with DIPEA (0.31 mL) and stirred for 3 h at 90°C. The solvent is removed, the residue is purified by chromatography (20:80 to 80 :20 in 6 min CH3CN/H20) and quinoline E.2 (28 mg, 28 %) is obtained.
Method of synthesising quinoline E.3
Figure imgf000147_0002
Quinoline D*.9 (1 .00 g, 4.12 mmol), propargylamide C.1 (1 .50 g, 4.95 mmol), Pd(PPh)3 (238 mg, 10 mol %) and Cul (32 mg, 5 mol %) are placed in DMSO (10.0 mL), combined with DIPEA (7.7 mL) and stirred for 2 h at 50°C. The solvent is removed, the residue is purified by chromatography (95:5 to 40:60 in 6 min H20/MeOH) and quinoline E.3 (1.51 g, 79 % HPLC-MS: MS(M+H)+ = 465; tRel = 2.14 min; method FECB6) is obtained.
Method of synthesising quinoline E.4
Figure imgf000148_0001
Quinoline D*.10 (1 00 mg, 0.32 mmol), propargylamide C.1 (1 15 mg, 0.38 mmol), Pd(PPh)3 (18 mg, 10 mol %) and Cul (3 mg, 5 mol %) are placed in DMSO (1 .0 mL), combined with DIPEA (0.29 mL) and stirred for 3 h at 20°C. The solvent is removed, the residue is taken up in H20 (5 mL), extracted with DCM (3 x 5 mL), dried on MgS04, filtered, the solvent is removed and quinoline E.4 (61 mg, 39 %) is obtained. f) Synthesis of other compounds (1 ) according to the invention
Reaction scheme F
Figure imgf000148_0002
(1)
Cf. remarks relating to reaction scheme C. Method of svnthesising 11-1
Figure imgf000149_0001
Quinazoline E.1 (300 mg, 0.23 mmol) is placed in DMSO (2 mL), combined with NEt3 (1 16 mg, 0.96 mmol) and then added dropwise to methylamine (0.65 mL, 1 .3 mL). The reaction mixture is stirred for 2 h at 20°C, freed from the solvent, the crude product is purified by chromatography (95:5 to 40:60 in 6 min H20/MeOH) and quinazoline 11-1 (1 15 mg, 78 %; HPLC-MS: MS(M+H)+ = 461 ; tRet = 1 .55 min; method LCMSBAS1 ) is obtained.
Quinazolines II-2 - II-63 are prepared analogously to quinazoline 11-1 (Table 21 ):
MS (M±H) ; HPLC
Structure
tRet. HPLC [min] method
11-1 M+H=461 ; tRet = 1.55 LCMSBAS1
11 -2 M+H=516; tRet = 1.58 LCMSBAS1
11 -3 M+H=565; tRet = 1.98 LCMSBAS1
Figure imgf000149_0002

Figure imgf000150_0001
150
Figure imgf000151_0001
Figure imgf000152_0001
LCMSBAS1
LCMSBAS1
LCMSBAS1 -17 LCMSBAS1 -18 LCMSBAS1
Figure imgf000152_0002
ı52
Figure imgf000153_0001
ı53
Figure imgf000154_0001
ı54
Figure imgf000155_0001
ı55
Figure imgf000156_0001


Figure imgf000158_0001

Figure imgf000159_0001

Figure imgf000160_0001
160
Figure imgf000161_0001
Method of svnthesising 11-64
Figure imgf000162_0001
(128 mg, 0.92 mmol) and Pd(dppf) (15 mg, 10 mol %) are placed in THF/NMP (3 mL, 2:1 ) and the reaction mixture is stirred for 3 h at 100°C and for 12 h at 20°C. Then HCI (1 mL, 4M in dioxane) is added, the mixture is heated to 50°C for 30 min, freed from the solvent, the crude product is purified by chromatography (95:5 to 40:60 in 6 min H20/MeOH) and quinazoline II-64 (30 mg, 33 %; HPLC-MS: MS(M+H)+ = 498; t 1 .55 min; method LCMSBAS1 ) is obtained. Quinazolines 11-65 - 11-67 are prepared analogously to quinazoline 11-64 (Table 22):
Figure imgf000162_0002
Figure imgf000163_0001
Method of s nthesising quinoline II-68
Figure imgf000163_0002
Quinoline E.2 (60 mg, 0.13 mmol) is placed in DMF, combined with HATU (58 mg, 0.15 mmol) and DIPEA (0.09 mL) and stirred for 10 min at 20°C. Then amine ED.20 (0.04 mL, 0.32 mmol) is added and the mixture is stirred for 12 h at 20°C. The solvent is removed, the residue is purified by chromatography (20:80 to 80:20 in 15 min CH3CN/H20) and quinoline II-68 (28 mg, 28 %; HPLC-MS: MS(M+H)+ = 559; tRet = 1 .68 min; method LCMSBAS1 ) is obtained. Compounds 11-69 - 11-87 are prepared analogously to quinoline 11-68 (Table 23): ı63
Figure imgf000164_0001
ı64
Figure imgf000165_0001
ı65
Figure imgf000166_0001
ı66
Figure imgf000167_0001
Figure imgf000168_0001
Figure imgf000168_0002
E.3
11-88
Quinoline E.3 (100 mg, 0.22 mmol), boric acid ester ED.21 (189 mg, 0.65 mmol), Pd(dppf) (17 mg, 10 mol %) and K2C03 (149 mg, 1 .07 mmol) are placed in TH F/NMP (3.0 mL, 2: 1 ) and stirred for 1 h at 100°C. Then the reaction mixture is diluted with H20 (5 mL), extracted with DCM (3 x 5 mL), dried on MgS04, filtered and the solvent is eliminated. The residue is taken up in DCM (1 .0 mL), combined with TFA (2.0 mL) and stirred for 12 h at 20°C. The reaction mixture is made basic with NaHC03 (5 mL), extracted with DCM (3 x 5 mL), dried on MgS04, filtered, the solvent is eliminated and the residue is purified by chromatography (95:5 to 40:60 in 6 min H20/MeOH) and quinoline 11-88 (1 7 mg, 1 6 %; HPLC-MS: MS(M+H)+ = 496; tRei = 1 .78 min; method LCMSBAS1 ) is obtained. Compounds II-89 - II-93 are prepared analogously to quinoline II-88 (Table 24):
Figure imgf000169_0001
Figure imgf000170_0001
Method of svnthesising quinoline II-94
Figure imgf000170_0002
E.4 II-94
Quinoline E.4 (30 mg, 0.06 mmol) is combined with dimethylamine (0.31 mL, 0.61 mmol, 2 M in TH F), DI PEA (1 6 μ Ι_) is added and the mixture is stirred for 1 h at 20°C. The solvent is eliminated, the residue is purified by chromatography (95:5 to 40:60 in 6 min H20/MeOH) and quinoline II-94 (17 mg, 16 %; HPLC-MS: MS(M+H)+ = 498; tRel = 1 .88 min; method LCMSBAS1 ) is obtained.
Compounds II-95 - 11-106 are prepared analogously to quinoline II-94 (Table 25):
170
Figure imgf000171_0001
171
Figure imgf000172_0001
Figure imgf000173_0001
94 LCMSBAS1
78 LCMSBAS1
11-106 M+H=588; tRet.= 2.13 LCMSBAS1
Figure imgf000173_0002
g) Method of synthesising quinazolines F (with alkenyl linkers)
Reaction scheme G
Figure imgf000173_0003
EWG = electron withdrawing group, e.g. halogen, triflate,
mesylate
PG = protecting group In order to incorporate alkenyl linkers L the components D are first of all reacted with protected allylamines B in a HECK reaction to obtain the amine intermediates F. After the reaction of B with D the product obtained still contains the protective group PG, which is cleaved. The protective group PG used may be any of the amino protecting groups known in organic synthesis.
Method of synthesising quinazoline F.1
Figure imgf000174_0001
Quinazoline DM 3 (3.17 g, 13.0 mmol) is placed in DCM/MeCN (6 mL, 2:1 ), combined with DIPEA (0.33 mL) and methylamine (0.81 mL, 2 M in THF) and stirred for 1.5 h at 20°C. Then the solvent is removed, the residue is taken up in H20 (15 mL), filtered off, washed with H20 (3 x 15 mL) and Et20 (3 mL), dried in vacuo and quinazoline D.95 (2.93 g, 95 %; HPLC-MS: MS(M+H)+ = 238; tRel = 1 .13 min; method LCMSBAS1 ) is obtained.
Quinazoline D.95 (1 .1 9 g, 5.00 mmol), Pd(OAc)2 (1 12 mg, 10 mol %) and tri(o- tolyl)phosphine (31 1 mg, 20 mol %) are placed in CH3CN/NMP (12 mL, 5:1 ), combined with allylamine B.2 (1 .59 g, 6.00 mmol) and then with DIPEA (1 .8 mL) and heated to 1 10°C for 1 .5 h. The reaction mixture is filtered, then H20 (50 mL) is added, the mixture is extracted with DCM (3 x 30 mL), dried on Na2S04, filtered, freed from the solvent, the residue is purified by chromatography (97:3 to 82:18 in 6 min DCM/MeOH) and quinazoline Z.13 (415 mg, 90 %; HPLC-MS: MS(M+H)+ = 415; tRet = 1.92 min; method LCMSBAS1 ) is obtained.
Quinazoline Z.13 (2.06 g, 5.00 mmol) is taken up in HCI (10 mL, 4M in dioxane) and stirred for 24 h at 20°C. The precipitate is filtered off, freed from the solvent and quinazoline F.1 (1 .31 g, 70 %; HPLC-MS: MS(M+H)+ = 215; tRet = 1 .08 min; method LCMSBAS1 ) is obtained. h) Synthesis of quinazolines (1 ) according to the invention with alkenyl linkers Reaction scheme H
Figure imgf000175_0001
A (1 )
Amide couplings which are supported in the first reaction step by coupling reagents such as for example DCC, DIC, TBTU, HATU, EDC or the like are used for the amide linking of the acids A and amines F.
Method of synthesising quinazoline III— 1
Figure imgf000175_0002
Carboxylic acid A.46 (22 mg, 0.07 mmol) is placed in DMSO (0.2 ml_), combined with DIPEA (56 μΙ_) and HATU (40 mg, 0.1 0 mmol) and stirred for 1 0 m in at 20°C. Then quinazoline F.1 (30 mg, 0.08 mmol) is added and the mixture is stirred for 3 h at 20°C. The solvent is eliminated, the residue is purified by chromatography (97:3 to 82: 18 in 6 min H20/MeOH) and quinazoline 111-1 (21 mg, 60 %; HPLC-MS: MS(M+H)+ = 509; tRel = 1 .44 min; method LCMSBAS1 ) is obtained. Quinazoline 111-1 (Table 26):
Figure imgf000176_0001
The following Examples describe the biological activity of the compounds according to the invention, without restricting the invention to these Examples.
Compounds of general formula (1 ) are characterised by their many possible applications in the therapeutic field. Particular mention should be made of those applications in which the inhibition of specific signal enzymes, particularly the inhibiting effect on the proliferation of cultivated human tumour cells but also on the proliferation of other cells such as endothelial cells, for example, are involved. The activity of the compounds according to the invention on the kinase PDK1 which inhibits the signal transduction pathway is determined in an in vitro kinase assay with recombinantly prepared protein:
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 (Greiner bio-one, No. 650101 ):
15 μΙ_ of compound to be tested in varying concentrations (e.g. starting at 10 μΜ, and diluted in steps of 1 :5) in APT buffer (50mM Tris/CI pH 7.5; 0.05% β-mercaptoethanol; 10mM Mg-acetate; 0.0166 % Tween 20; 3.33 % DMSO)
15 μΙ_ His6-PDK1 (aa 52-556) 3.33 ng/well) and PDKtide
(KTFCGTPEYLAPEVRRE PRILSEEEQEMFRDFDYIADWC), synthesised by Pepceuticals Limited, Nottingham, United Kingdom; 25 μΜ final concentration); His6-PDK1 and PDKtide are together diluted accordingly in assay buffer (50 mM tris pH 7.5, 0.05 % β-mercaptoethanol, 10 mM Mg-acetate); PDKtide is present in this mixture as an 83.3 μΜ solution. These 30 μΙ are routinely incubated for 30 min at RT.
• 20 μΙ_ ATP solution (25 μΜ ATP with 1.0 μΟί/ννβΙΙ gamma-P33-ATP). The final concentration of Tween 20 is 0.005%.
The reaction is started by adding the ATP solution and the mixture is incubated for 90 min at RT. At the start of the reaction the dishes are shaken gently. The reaction is stopped by the addition of 50 μ-Jwell of 500 mM phosphoric acid (H3P04) and incubated for about 20 min at RT. The precipitate is transferred by harvesting onto filter plates (96-well microtitre filter plate: UniFilter GF/C; Perkin Elmer; No. 6005174), then washed 6 times with 50 mM H3P04 and dried at 60°C. Then the plate is stuck down with sealing tape, 25 [\Uwe\\ of scintillation solution (Microscint 0; Perkin Elmer; No. 601361 1 ) are added and the amount of P33 precipitated is measured using the Wallac Betacounter. The measured data are evaluated using Graphpad Prism software.
Table 27 shows the IC50 values of the example compounds of types I and II determined using the above assay.
Table 27
Figure imgf000177_0001
I -2 37 1-15 68 I -28 21 1-41 3
I -3 8 1-16 33 I -29 41 I -42 3
I -4 311 1-17 95 I -30 2 I -43 5
I -5 229 1-18 17 1-31 5 I -44 11
I -6 68 1-19 4 I -32 8 I -45 8
I -7 94 I -20 4 I -33 40 I -46 7
I -8 103 1-21 149 I -34 3 I -47 4
I -9 3 I -22 6 I -35 21 I -48 3
1-10 11 I -23 8 I -36 87 I -49 9
1-11 88 I -24 117 I -37 5 I -50 1
1-12 35 I -25 42 I -38 56 1-51 2
1-13 34 I -26 588 I -39 21 I -52 4
Figure imgf000178_0001
■54 8 I -89 23 1-124 2 11 -3 91
55 4 I -90 1 10 1-125 2 II-9 443
56 3 1-91 38 1-125.1 3 11-10 229
■57 9 I -92 128 1-125.2 5 11-11 135
58 8 I -93 12 1-125.3 5 11-12 161
59 5 I -94 55 1-125.4 12 11-13 184
-60 7 I -95 33 1-125.5 16 11-14 158
61 2 I -96 74 1-125.6 26 11-15 445
■62 2 I -97 38 1-125.7 32 11-16 218
63 4 I -98 25 1-125.8 46 11-17 488
■64 5 I -99 36 1-125.9 2 11-18 41
65 7 1-100 14 1-125.10 3 11-19 500
-66 654 1-101 5 1-125.11 4 11 -20 13
■67 940 1-102 13 1-125.12 6 11-21 515
-68 233 1-103 33 1-125.13 6 11 -22 371
-69 64 1-104 71 1-125.14 60 11 -23 142
70 1 1-105 47 1-125.15 1 1 11 -24 77
■71 4 1-106 1 19 1-125.16 1 1 11 -25 126
■72 2 1-107 212 1-125.17 2 11 -26 199
73 4 1-108 1 16 1-125.18 2 11 -27 52
■74 8 1-109 166 1-125.19 3 11 -28 1 10
75 1 1-110 137 1-125.20 17 11 -29 156
76 2 1-111 29 1-125.21 2 11 -30 21
■77 4 1-112 71 1-125.22 3 11-31 1 14
78 9 1-113 15 1-125.23 4 11 -32 124
79 80 1-114 23 1-126 86 11 -33 79
-80 132 1-115 780 1-127 98 11 -34 59
81 66 1-116 1000 1-128 1 16 11 -35 59
-82 1 19 1-117 1000 11-1 12 11 -36 468
83 62 1-118 853 11 -2 171 11 -37 44
-84 21 1-119 554 11 -3 134 11 -38 97
85 185 1-120 656 II-4 26 11 -39 137
-86 86 1-121 156 II-5 16 11 -40 74
■87 23 1-122 3 11 -6 98 11-41 61
Figure imgf000179_0001
-43 8 11 -60 8 11 -77 53 11 -94 27
-44 40 11-61 57 11 -78 402 11 -95 544
-45 63 11 -62 16 11 -79 201 11 -96 607
-46 6 11 -63 14 11 -80 64 11 -97 1055
-47 1 17 I -64 57 11-81 20 11 -98 184
-48 15 11 -65 58 11 -82 531 11 -99 683
-49 50 11 -66 98 11 -83 286 11-100 276
-50 182 11 -67 96 11 -84 405 11-101 148
-51 770 11 -68 20 11 -85 1000 11-102 871
-52 6 11 -69 21 11 -86 355 11-103 77
-53 486 11 -70 185 11 -87 24 11-104 147
-54 8 11-71 1000 11 -88 19 11-105 46
-55 60 11 -72 45 11 -89 35 11-106 414
-56 6 11 -73 31 11 -90 86
-57 357 11 -74 12 11-91 14
-58 10 11 -75 45 11 -92 56
The antiproliferative activity of the compounds according to the invention is determined in the proliferation test on cultivated human tumour cells and/or in a cell cycle analysis, for example on PC-3 tumour cells: Inhibition of proliferation on cultivated human tumour cells (PC-3)
To measure proliferation on 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 % C02), wherein 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 background 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 d incubation 20 μΙ AlamarBlue reagent (Serotec) are added to each well, and the cells are incubated for a further 5-7 h. As a control, 20 μΙ 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 s 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.
Many of the compounds according to the invention cause inhibition of proliferation by interfering with intracellular signal transduction pathways which are important for cell survival, predominantly, but not exclusively, in cells which have become dependent on these signal pathways during their development.
Compounds (1 ) according to the invention generally demonstrate good activity in cell assays of this kind, i.e. for example an EC50 value in the PC-3 proliferation test of less than 10 μη"ΐοΙ/Ι_, often less than 5 μmol L.
Biomarker inhibition:
The substances of the present invention bring about cellular inhibition of PDK1 -substrates. Examples of the latter are Phospho-Thr308/AKT, Phospho-Ser221 ,227/RSK, or phosphorylation sites on p70S6 kinase (Thr229). In order to determine the inhibitory effect, the cells are treated with substance for 2 h, for example, lysed and analysed by Western Blot and/or BioPlex analysis for phosphoproteins of this kind. Commercially obtainable phospho-specific antibodies to the above-mentioned phosphorylation sites are used.
In PC-3 or other signal pathway-mutated cell lines as a rule EC50 values of less than 5 μη"ΐοΙ/Ι_, often less than 0.5 μη"ΐοΙ/Ι_, are achieved with the present compounds on these phosphorylation sites compared with the carrier control and after standardisation to the corresponding whole protein. On the basis of their biological properties the compounds of general formula (1 ) according to the invention, their tautomers, racemates, enantiomers, diastereomers, mixtures thereof and the salts of all the above-mentioned forms 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). They are also suitable for protecting proliferating cells (e.g. hair, intestinal, blood and progenitor cells) from DNA damage caused by radiation, UV treatment and/or cytostatic treatment.
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 pi locytic astrocytomas , fi bril lary 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 tumou r (adrenocorticotropic hormone), craniopharyngiomas, medu lloblastomas, 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 carcinoma, colorectal carcinoma, anal carcinoma, carcinoma of the large bowel, tumours of the 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 (NSCLC) 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, 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 exam ple 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.
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, fludrocortisone, 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 tyrosine kinase inhibitors, such as for example cetuximab, 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.
Other possible combination partners are 2-chlorodesoxyadenosine, 2-fluorodesoxycytidine, 2-methoxyoestradiol, 2C4, 3-alethine, 131 -I-TM-601 , 3 CPA, 7-ethyl-10-hydroxycamptothecin, 16-aza-epothilone B, A 105972, A 204197, aldesleukin, alitretinoin, altretamine, alvocidib, amonafide, anthrapyrazole, AG-2037, AP-5280, apaziquone, apomine, aranose, arglabin, arzoxifene, atamestane, atrasentan, auristatin PE, AVLB, AZ10992, ABX-EGF, ARRY-300, ARRY-142886/AZD-6244, ARRY-704/AZD-8330, AS-703026 , azacyti d i n e , azae poth i l on e B , azon afi d e , BAY-43-9006, BBR-3464, BBR-3576, bevacizumab, biricodar dicitrate, BCX-1777, bleocin, BLP-25, BMS-184476, BMS-247550, BMS-188797, BMS-275291 , BNP-1350, BNP-7787, BI BW 2992, B I B F 1 120 , bleomycin ic acid , bleomycin A, bleomycin B, bryostatin-1 , bortezomib, brostallicin, busulphan, CA-4 prodrug, CA-4, CapCell, calcitriol, canertinib, canfosfamide, capecitabine, carboxyphthalatoplatin, CCI-779, CEP-701 , CEP-751 , CBT-1 cefixime, ceflatonin, ceftriaxone, celecoxib, celmoleukin, cemadotin, CH4987655/RO-4987655, chlorotrianisene, cilengitide, ciclosporin, CDA-II, CDC-394, CKD-602, clofarabin, colchicin, combretastatin A4, CHS-828, CLL-Thera, CMT-3 cryptophycin 52, CTP-37, CP-461 , CV-247, cyanomorpholinodoxorubicin, cytarabine, D 24851 , decitabine, deoxorubicin, deoxyrubicin, deoxycoformycin, depsipeptide, desoxyepothilone B, dexamethasone, dexrazoxanet, diethylstilbestrol, diflomotecan, didox, DMDC, dolastatin 10, doranidazole, E7010, E-6201 , edatrexat, edotreotide, efaproxiral, eflornithine, EKB-569, EKB-509, elsamitrucin, epothilone B, epratuzumab, ER-86526, eriotinib, ET-18-OCH3, ethynylcytidine, ethynyloestradiol, exatecan, exatecan mesylate, exemestane, exisulind, fenretinide, floxuridine, folic acid, FOLFOX, FOLFIRI, formestane, galarubicin, gallium maltolate, gefinitib, gemtuzumab, gimatecan, glufosfamide, GCS-IOO, G17DT immunogen, GMK, GPX-100, GSK-5126766, GSK-1 120212, GW2016, granisetron, hexamethylmelamine, histamine, homoharringtonine, hyaluronic acid, hydroxyurea, hydroxyprogesterone caproate, ibandronate, ibritumomab, idatrexate, idenestrol, IDN-5109, IMC-1 C1 1 , immunol, indisulam, interferon alpha-2a, interferon alpha-2b, interleukin-2, ionafarnib, iproplatin, irofulven, isohomohalichondrin-B, isoflavone, isotretinoin, ixabepilone, JRX-2, JSF-154, J-107088, conjugated oestrogens, kahalid F, ketoconazole, KW-2170, lobaplatin, leflunomide, lenograstim, leuprolide, leuporelin, lexidronam, LGD-1550, linezolid, lutetium texaphyrin, lometrexol, losoxantrone, LU 223651 , lurtotecan, mafosfamide, marimastat, mechloroethamine, methyltestosteron, methylprednisolone, MEN-10755, MDX-H210, MDX-447, MGV, midostaurin, minodronic acid, mitomycin, mivobulin, MK-2206, MLN518, motexafin gadolinium, MS-209, MS-275, MX6, neridronate, neovastat, nimesulide, nitroglycerin, nolatrexed, norelin, N-acetylcysteine, 06-benzylguanine, omeprazole, oncophage, ormiplatin, ortataxel, oxantrazole, oestrogen, patupilone, pegfilgrastim, PCK-3145, pegfilgrastim, PBI-1402, PEG-paclitaxel, PEP-005, P-04, PKC412, P54, PI-88, pelitinib, pemetrexed, pentrix, perifosine, perillylalcohol, PG-TXL, PG2, PLX-4032/RO-5185426, PT-100, picoplatin, pivaloyloxymethylbutyrate, pixantrone, phenoxodiol O, PKI166, plevitrexed, plicamycin, polyprenic acid, porfiromycin, prednisone, prednisolone, quinamed, quinupristin, RAF-265, ramosetron, ranpirnase, RDEA-1 19/BAY 869766, rebeccamycin analogues, revimid, RG-7167, rhizoxin, rhu-MAb, risedronate, rituximab, rofecoxib, Ro-31 -7453, RO-5126766, RPR 109881A, rubidazone, rubitecan, R-flurbiprofen, S-9788, sabarubicin, SAHA, sargramostim, satraplatin, SB 408075, SU5416, SU6668, SDX-101 , semustin, seocalcitol, SM-1 1355, SN-38, SN-4071 , SR-27897, SR-31747, SRL-172, sorafenib, spiroplatin, squalamine, suberanilohydroxamic acid, sutent, T 900607, T 138067, TAS-103, tacedinaline, talaporfin, tariquitar, taxotere, taxoprexin, tazarotene, tegafur, temozolamide, tesmilifene, testosterone, testosterone propionate, tesmilifene, tetraplatin, tetrodotoxin, tezacitabine, thalidomide, theralux, therarubicin, thymectacin, tiazofurin, tipifarnib, tirapazamine, tocladesine, tomudex, toremofin, trabectedin, TransMID-107, transretinic acid, traszutumab, tretinoin, triacetyluridine, triapine, trimetrexate, TLK-286TXD 258, urocidin, valrubicin, vatalanib, vincristine, vinflunine, virulizin, WX-UK1 , vectibix, xeloda, XELOX, XL-281 , XL-518/R-7420, YM-51 1 , YM-598, ZD-4190, ZD-6474, ZD-4054, ZD-0473, ZD-6126, ZD-9331 , ZDI839, zoledronat and zosuquidar. 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 th e usu al m ethods , preferably by ora l 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 m ixed together. The mixtu re 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 )
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

Patent Claims
1. Compounds of general formula (1 )
Figure imgf000190_0001
(1 ) , wherein
Qa is a ring system optionally substituted by one or more identical or different Ra and/or Rb, selected from among C3-iocycloalkyl, C6-ioaryl, 5-12 membered heteroaryl and 3-14 membered heterocyclyl;
R1 and R2 are selected independently of one another from among hydrogen, halogen, Ci-4alkyl, C2-4alkenyl, C2-4alkynyl, Ci-4haloalkyl, -NH2, -CN, -NHCi-4alkyl, -N(Ci-4alkyl)2, -OH, -OCi-4a l ky l , H 0-Ci-4alkylene-, Ci-4alkyl-0-Ci-4alkylene-, H2N-Ci-4alkylene-, -OCi-4haloalkyl, (Ci-4alkyl)NH-Ci-4alkylene- and (Ci-4alkyl)2N-Ci-4alkylene, while alkyl, alkenyl, alkynyl and alkylene mentioned in the above groups may optionally be substituted by one or more identical or different halogens;
the ring system Qb is selected from among
Figure imgf000190_0002
each R3 is independently selected from among halogen, Ci-4alkyl, C2-4alkenyl, C2-4alkynyl, Ci-4haloalkyl, -NH2, -CN, -NHCi-4alkyl, -N(C1-4alkyl)2, -OH, -OCi-4alkyl, HO-Ci-4alkylene-, HO-C2-4alkylene-0-, Ci-4alkyl-0-Ci-4alkylene-, Ci-4haloalkyl-0-Ci-4alkylene-,
H2N-Ci-4alkylene-, Ci-4alkyl-0-C2-4alkylene-0-, (Ci-4alkyl)NH-Ci-4alkylene-,
(Ci-4alkyl)2N-Ci-4alkylene-, -OCi-4haloalkyl, H2N-C2-4alkylene-0-, -NH(C2-4alkylene-NH2), -NH[C2-4alkylene-N(HCi-4alkyl)], -NH[C2-4alkylene-N(Ci-4alkyl)2],
(Ci-4alkyl)2N-C2-4alkylene-0-, (Ci-4haloalkyl)2N-C2-4alkylene-0-, (Ci-4haloalkyl)NH-C2-4alkylene-0- and (Ci-4alkyl)NH-C2-4alkylene-0-;
n denotes the number 0, 1 , 2 or 3 if Qb corresponds to the ring system Qb-4;
each n independently denotes the number 0, 1 or 2 if Qb corresponds to one of the ring systems Qb-1 , Qb-2, Qb-3 or Qb-5;
while a group R3 in the ring systems Qb-1 to Qb-5 in each case replaces a hydrogen;
R4 denotes hydrogen or Ci-4alkyl;
L denotes the group
Figure imgf000191_0001
and in the latter case the groups -CR5R6- and QH may assume both a c/'s and a trans configuration with respect to the double bond;
R5 and R6 are each independently of one another selected from among hydrogen , halogen, Ci-4alkyl, Ci-4haloalkyl;
the ring system QH is selected from among
Figure imgf000191_0002
QH-2a QH-2b QH-2c
Figure imgf000191_0003
QH-2d QH-2e QH-2f
R7 and R9 are each independently of one another selected from among hydrogen, halogen, -NRaRa, -N(ORa)Ra, -ORa, -COORa, -CORa, -CONRaRa, -SRa, -N02 and -CN or denote a group optionally substituted by one or more identical or different Rb and/or Rc, selected from among Ci-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-iocycloalkyl, C6-ioaryl , 5-12 membered heteroaryl and 3-14 membered heterocyclyl;
R8 is independently selected from among hydrogen, -NRaRa, -N(ORa)Ra, -ORa, -COORa, -CORa, -CONRaRa and -SRa or denotes a group optionally substituted by one or more identical or different Rb and/or Rc, selected from among Ci-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-iocycloalkyl, C6-ioaryl, 5-12 membered heteroaryl and 3-14 membered heterocyclyl;
R10 is independently selected from among -NRaRa, -N(ORa)Ra, -ORa and -SRa or a group optionally substituted by one or more identical or different Rb and/or Rc, selected from a mon g Ci-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-iocycloalkyl, C6-ioaryl, 5-12 membered heteroaryl and 3-14 membered heterocyclyl;
each R11 and R12 is independently selected from among Ra and Rb;
m denotes the number 0, 1 or 2; each Ra independently denotes hydrogen or a group optionally substituted by one or more identical or different Rb and/or Rc, selected from among Ci-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-iocycloalkyl, C6-ioaryl, 5-12 membered heteroaryl and 3-14 membered heterocyclyl; each Rb is independently selected from among -ORc, -NRCRC, halogen, -CN, -N02, -C(0)Rc, -C(0)ORc, -C(0)NRcRc, -S(0)2Rc, -S(0)2NRcRc, -NHC(0)Rc and -N(Ci-4alkyl)C(0)Rc as well as the bivalent substituent =0, while the latter may only be a substituent in non-aromatic ring systems;
each Rc independently denotes hydrogen or a group optionally substituted by one or more identical or different Rd and/or Re, selected from among Ci-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-iocycloalkyl, C6-ioaryl, 5-12 membered heteroaryl and 3-14 membered heterocyclyl; each Rd is independently selected from among -ORe, -NReRe, halogen, -CN, -N02, -C(0)Re, -C(0)ORe, -C(0)NReRe, -S(0)2Re, -S(0)2NReRe, -NHC(0)Re and -N(Ci-4alkyl)C(0)Re, as well as the bivalent substituent =0, while the latter may only be a substituent in non-aromatic ring systems;
each Re independently denotes hydrogen or a group optionally substituted by one or more identical or different Rf and/or Rg, selected from among Ci-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-iocycloalkyl, C6-ioaryl, 5-12 membered heteroaryl and 3-14 membered heterocyclyl; each Rf is independently selected from among -OR9, -NR9R9, halogen, -CN, -N02, -C(0)R9, -C(0)OR9, -C(0)NR9R9, -S(0)2R9, -S(0)2NR9R9, -NHC(0)R9 and -N(Ci-4alkyl)C(0)R9, as well as the bivalent substituent =0, while the latter may only be a substituent in non-aromatic ring systems and
each Rg is independently selected from among hyd rogen , Ci-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-iocycloalkyl, C6-ioaryl , 5-12 membered heteroaryl and 3-14 membered heterocyclyl;
while the compounds (1 ) may optionally also be present in the form of the tautomers, racemates, enantiomers, diastereomers or mixtures thereof or as the respective salts of all the above-mentioned forms.
2. Compounds according to claim 1 , wherein
Qa is a ring system optionally substituted by one or more identical or different Ra and/or Rb selected from among C5-6cycloalkyl, phenyl, 5-6 membered heteroaryl and 5-7 membered heterocyclyl, and
Ra and Rb are defined as in claim 1.
3. Compounds according to one of claims 1 or 2, wherein
the ring system QH is selected from among
Figure imgf000193_0001
QH-2a and QH-2d
R7 is independently selected from among hydrogen, -NRaRa, -N(ORa)Ra, -ORa, -COORa, -CORa, -CONRaRa and -SRa, or denotes a group optionally substituted by one or more identical or different Rb and/or Rc selected from among Ci-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-iocycloalkyl, C6-ioaryl, 5-12 membered heteroaryl and 3-14 membered heterocyclyl, and
R11, R12, Ra, Rb, Rc and m are defined as in claim 1 .
4. Compounds according to claim 3, wherein
the ring system QH is selected from among
Figure imgf000194_0001
QH-2a and QH-2d
R7 is independently selected from among hydrogen, -NRa1Ra1, -N(ORa1)Ra1, -ORa1, -COORa1, -CORa1, -CONRa1Ra1 and -SRa1, or denotes a group optionally substituted by one or more identical or different Rb1 and/or Rc1 selected from among Ci-6alkyl, C2-6alkenyl , C2-6alkynyl, C3-iocycloalkyl, C6-ioaryl, 5-12 membered heteroaryl and 3-14 membered heterocyclyl;
each Ra1 independently denotes hydrogen or a group optionally substituted by one or more identical or different Rb1 and/or Rc1 selected from among Ci-6alkyl, C3-i0cycloalkyl and 3-14 membered heterocyclyl;
each Rb1 is independently selected from among -ORc1, -NRc1Rc1 and halogen;
each Rc1 independently denotes hydrogen or a group optionally substituted by one or more identical or different Rd1 and/or Re1 selected from among Ci-6alkyl, C3-i0cycloalkyl and 3-14 membered heterocyclyl;
each Rd1 is independently selected from among -ORe1, -CN and -C(0)ORe1;
each Re1 independently denotes hydrogen or a group optionally substituted by one or more identical or different C3-6cycloalkyl selected from among Ci-6alkyl and C3-i0cycloalkyl, and
R11, R12 and m are defined as in claim 1.
5. Compounds according to one of claims 1 or 2, wherein
the ring system QH is selected from among
Figure imgf000195_0001
-2b and QH-2e
R9 is independently selected from among hydrogen, -NRaRa, -N(ORa)Ra, -ORa, -C00R3, -CORa, -CONRaRa and -SRa, or a group optionally substituted by one or more identical or different Rb and/or Rc selected from among Ci-6alky l , C2-6alkenyl, C2-6alkynyl, C3-iocycloalkyl, C6-ioaryl, 5-12 membered heteroaryl and 3-14 membered heterocyclyl, and
R11, R12, Ra, Rb, Rc and m are defined as in claim 1 .
6. Compounds according to claim 5, wherein
the ring system QH is selected from among
Figure imgf000195_0002
QH-2b and QH-2e R9 is independently selected from among hydrogen, -NRa2Ra2, -N(ORa2)Ra2, -ORa2, -COORa2, -CORa2, -CONRa2Ra2 and -SRa2, or denotes a group optionally substituted by one or more identical or different Rb2 and/or Rc2 selected from among Ci-6alkyl, C2-6alkenyl , C2-6alkynyl, C3-iocycloalkyl, C6-ioaryl, 5-12 membered heteroaryl and 3-14 membered heterocyclyl;
each Ra2 independently denotes hydrogen or a group optionally substituted by one or more identical or different Rb2 and/or Rc2 selected from among Ci-6alkyl, C3-i0cycloalkyl and 3-14 membered heterocyclyl;
each Rb2 is independently selected from among -ORc2, -NRc2Rc2 and halogen;
each Rc2 independently denotes hydrogen or a group optionally substituted by one or more identical or different Rd2 and/or Re2 selected from among Ci-6alkyl, C3-i0cycloalkyl and 3-14 membered heterocyclyl;
each Rd2 is independently selected from among -ORe2, -CN and -C(0)ORe2;
each Re2 independently denotes hydrogen or a group optionally substituted by one or more identical or different C3-6cycloalkyl selected from among Ci-6alkyl and C3-iocycloalkyl, and
R11, R12 and m are defined as in claim 1.
7. Compounds according to claim 6, wherein
the ring system QH denotes
Figure imgf000196_0001
QH-2b R9 is selected from among hydrogen, -NRa2Ra2, -ORa2 and -SRa2, or denotes a group optionally substituted by one or more identical or different Rb2 and/or Rc2 selected from among Ci-6alky I , C2-6alkenyl, pyrrolidinyl, piperazinyl, piperidinyl, morpholinyl, homomorpholinyl, azetidinyl and homopiperazinyl;
each Ra2 independently denotes hydrogen or a group optionally substituted by one or more identical or different Rb2 and/or Rc2 selected from among Ci-6alkyl, C3-6cycloalkyl, piperazinyl, piperidinyl, morpholinyl, azetidinyl and pyrrolidinyl;
each Rb2 is independently selected from among -ORc2, -NRc2Rc2 and halogen;
each Rc2 independently denotes hydrogen or a group optionally substituted by one or more identical or different Rd2 and/or Re2 selected from among Ci-6alkyl, C3-6cycloalkyl, pyrrolidinyl, morpholinyl, homomorpholinyl, piperazinyl, homopiperazinyl, oxetanyl and piperidinyl;
each Rd2 is independently selected from among -ORe2, -CN and -C(0)ORe2;
each Re2 independently denotes hydrogen or a group optionally substituted by one or more identical or different C3-6cycloalkyl selected from among Ci-6alkyl and C3-6cycloalkyl, and R11 and m are defined as in claim 1.
8. Compounds according to one of claims 1 or 2, wherein
the ring system QH is selected from among
Figure imgf000197_0001
QH-2b and R is selected from among hydrogen, -NRaRa, -N(ORa)Ra, -ORa and -SRa, or denotes a group optionally substituted by one or more identical or different Rb and/or Rc selected from among Ci-6alkyl, C2-6alkenyl, C2-6alkynyl, C3-iocycloalkyl, C6-ioaryl, 5-12 membered heteroaryl and 3-14 membered heterocyclyl;
R9 is independently selected from among hydrogen, halogen, -N02, -CN and Ci-4alkyl, and
R11, R12, Ra, RD, Rc and m are defined as in claim 1 .
9. Compounds according to claim 8, wherein
the ring system QH is selected from among
Figure imgf000197_0002
R is selected from among -NRa R and -OR , or denotes a group optionally substituted by one or more identical or different Rb3 and/or Rc3 selected from among Ci-6alkyl, C2-6alkenyl, C2-6alkynyl, 5-12 membered heteroaryl and 3-14 membered heterocyclyl;
R9 is independently selected from among hydrogen, halogen, -N02, -CN and Ci-4alkyl; each Ra3 independently denotes hydrogen or a group optionally substituted by one or more identical or different Rb3 and/or Rc3 selected from among Ci-6alkyl, C3-i0cycloalkyl, C6-ioaryl and 3-14 membered heterocyclyl;
each Rb3 is independently selected from among -ORc3 and -NRc3Rc3;
each Rc3 independently denotes hydrogen or a group optionally substituted by one or more identical or different Ci-6alkyl selected from among Ci-6alkyl, C3-iocycloalkyl, C6-ioaryl and 3-14 membered heterocyclyl, and
R11, R12 and m are defined as in claim 1.
10. Compounds according to one of claims 1 or 2, wherein
the ring system QH is selected from among
Figure imgf000198_0001
QH-2c and QH-2f R10 is independently selected from among -NRaRa, -N(ORa)Ra, -ORa and -SRa or denotes a group optionally substituted by one or more identical or different Rb and/or Rc selected from among Ci-6alkyl, C2-6alkenyl , C2-6alkynyl, C3-iocycloalkyl, C6-ioaryl, 5-12 membered heteroaryl and 3-14 membered heterocyclyl, and
R11, R12, Ra, Rb, Rc and m are defined as in claim 1 .
11. Compounds according to claim 10, wherein
the ring system QH is selected from among
Figure imgf000198_0002
QH-2c and QH-2f
R10 is independently selected from among -NRa4Ra4, -N(ORa4)Ra4, -ORa4 and -SRa4 or denotes a group optionally substituted by one or more identical or different Rb4 and/or Rc4 selected from among Ci-6alkyl , C2-6alkenyl, C2-6alkynyl, C3-iocycloalkyl, C6-ioaryl, 5-12 membered heteroaryl and 3-14 membered heterocyclyl;
each Ra4 independently denotes hydrogen or a group optionally substituted by one or more identical or different Rb4 and/or Rc4 selected from among Ci-6alkyl, C3-iocycloalkyl, C6-ioaryl and 3-14 membered heterocyclyl;
each Rb4 is independently selected from among -ORc4, -NRc4Rc4, -C(0)Rc4, -C(0)ORc4, -C(0)NRc4Rc4, -NHC(0)Rc4 and -N(Ci-4alkyl)C(0)Rc4;
each Rc4 independently denotes hydrogen or a group optionally substituted by one or more identical or different Rd4 and/or Re4 selected from among Ci-6alkyl, C3-iocycloalkyl, C6-ioaryl, 5-12 membered heteroaryl and 3-14 membered heterocyclyl;
each Rd4 denotes -ORe1;
each Re4 independently denotes hydrogen or a group optionally substituted by one or more identical or different Rf4 selected from among Ci-6alkyl and C6-ioaryl;
each Rf4 denotes halogen and
R11, R12 and m are defined as in claim 1.
12. Compounds according to claim 1 :
I-88 1 -[(3,4-difluorophenyl)methyl]-N-[3-[4-[3-(dimethylamino)propoxy]quinolin-6- yl]prop-2-ynyl]-6-oxopyrimidine-5-carboxamide;
1-101 1 -[(3,4-difluorophenyl)methyl]-6-oxo-N-[3-[4-[(3S)-pyrrolidin-3-yl]oxyquinolin-
6-yl]prop-2-ynyl]pyrimidine-5-carboxamide;
1-125.9 N-[3-[4-[[(3R)-1 -methylpyrrolidin-3-yl]methoxy]quinolin-6-yl]prop-2-ynyl]-2- oxo-1 -[(3,4,5-trifluorophenyl)methyl]pyridine-3-carboxamide;
1-125.1 1 N-[3-[4-[[(3S)-1 -methylpyrrolidin-3-yl]methoxy]quinolin-6-yl]prop-2-ynyl]-2- oxo-1 -[(1 R)-1 -(3,4,5-trifluorophenyl)ethyl]pyridine-3-carboxamide; 1-125.15 1 -[(1 R)-1 -(3,4-difluorophenyl)-2-hydroxyethyl]-N-[3-(4-ethoxyquinolin-6- yl)prop-2-ynyl]-2-oxopyridine-3-carboxamide;
1-125.17 N-[3-[4-[[(3R)-1 -methylpiperidin-3-yl]methoxy]quinolin-6-yl]prop-2-ynyl]-2- oxo-1 -[(3,4,5-trifluorophenyl)methyl]pyridine-3-carboxamide;
1-125.20 N-[3-[4-[[(3S)-1 -(2-morpholin-4-ylethyl)piperidin-3-yl]methoxy]quinolin-6- yl]prop-2-ynyl]-2-oxo-1 -[(1 R)-1 -(3,4,5-trifluorophenyl)ethyl]pyridine-3- carboxamide;
1-125.22 1 -[(3,4-difluorophenyl)methyl]-2-oxo-N-[3-[4-[[(3R)-1 -propan-2-ylpiperidin-3- yl]methoxy]quinolin-6-yl]prop-2-ynyl]pyridine-3-carboxamide;
1-125.21 1 -[(1 R)-2-hydroxy-1 -(3,4,5-trifluorophenyl)ethyl]-N-[3-[4-[[(3R)-1 - methylpiperidin-3-yl]methoxy]quinolin-6-yl]prop-2-ynyl]-2-oxopyridine-3- carboxamide;
1-125.19 N-[3-[4-[[(3S)-1 -(2-morpholin-4-ylethyl)piperidin-3-yl]methoxy]quinolin-6- yl]prop-2-ynyl]-2-oxo-1 -[(3,4,5-trifluorophenyl)methyl]pyridine-3- carboxamide;
I- 125.16 1 -[(1 S)-1 -(3,4-difluorophenyl)-2-hydroxyethyl]-N-[3-[4-(oxan-4- ylmethoxy)quinolin-6-yl]prop-2-ynyl]-2-oxopyridine-3-carboxamide;
II- 59 2-[(3,4-difluorophenyl)methyl]-6-methoxy-N-[3-[4-(methylamino)quinazolin-6- yl]prop-2-ynyl]-3-oxopyridazine-4-carboxamide;
I-93 1 -[(3,4-difluorophenyl)methyl]-N-[3-[4-[3-(dimethylamino)propoxy]quinolin-6- yl]prop-2-ynyl]-2-oxopyridine-3-carboxamide;
1-122 1 -[(3,4-difluorophenyl)methyl]-6-oxo-N-[3-[4-(pyrrolidin-3-ylmethoxy)quinolin-
6-yl]prop-2-ynyl]pyrimidine-5-carboxamide;
1-125.4 1 -[(1 S)-1 -(3,4-difluorophenyl)-2-hydroxyethyl]-2-oxo-N-[3-[4-(oxolan-3- ylmethoxy)quinolin-6-yl]prop-2-ynyl]pyridine-3-carboxamide;
I-30 1 -[(1 R)-1 -(3,4-difluorophenyl)ethyl]-N-[3-[3-[(1 -methylpiperidin-4- yl)amino]quinoxalin-6-yl]prop-2-ynyl]-2-oxopyridine-3-carboxamide; I-40 1 -[(3,4-difluorophenyl)methyl]-N-[3-[3-[(4-morpholin-4- ylcyclohexyl)amino]quinoxalin-6-yl]prop-2-ynyl]-6-oxopyrimidine-5- carboxamide;
I-42 N-[3-[3-[[4-[4-(cyclopropylmethyl)piperazin-1 -yl]cyclohexyl]amino]quinoxalin- 6-yl]prop-2-ynyl]-1 -[(3,4-difluorophenyl)methyl]-6-oxopyrimidine-5- carboxamide;
I-46 1 -[(3,4-difluorophenyl)methyl]-N-[3-[3-[(1 -ethylpiperidin-4- yl)amino]quinoxalin-6-yl]prop-2-ynyl]-2-oxopyridine-3-carboxamide;
1-51 1 -[(1 S)-1 -(3,4-difluorophenyl)ethyl]-N-[3-[3-[(1 -ethylpiperidin-4- yl)amino]quinoxalin-6-yl]prop-2-ynyl]-2-oxopyridine-3-carboxamide;
I-52 1 -[(1 S)-1 -(3,4-difluorophenyl)ethyl]-N-[3-[3-[[1 -(2-methoxyethyl)piperidin-4- yl]amino]quinoxalin-6-yl]prop-2-ynyl]-2-oxopyridine-3-carboxamide; 1-72 1 -[(3,4-difluorophenyl)methyl]-N-[3-[3-[(1 -ethylpiperidin-4-yl)amino]quinolin-
6-yl]prop-2-ynyl]-2-oxopyridine-3-carboxamide;
I-74 1 -[(3,4-difluorophenyl)methyl]-N-[3-[3-[[1 -(2-methoxyethyl)piperidin-4- yl]amino]quinolin-6-yl]prop-2-ynyl]-2-oxopyridine-3-carboxamide; I-75 1 -[(1 S)-1 -(3,4-difluorophenyl)ethyl]-N-[3-[3-[(1 -ethylpiperidin-4- yl)amino]quinolin-6-yl]prop-2-ynyl]-2-oxopyridine-3-carboxamide; I-78 1 -[(1 S)-1 -(3,4-difluorophenyl)ethyl]-N-[3-[3-[[1 -(2-methoxyethyl)piperidin-4- yl]amino]quinolin-6-yl]prop-2-ynyl]-2-oxopyridine-3-carboxamide; I-47 1 -[(3,4-difluorophenyl)methyl]-N-[3-[3-[[(3S,4R)-1 -ethyl-3-fluoropiperidin-4- yl]amino]quinoxalin-6-yl]prop-2-ynyl]-6-oxopyrimidine-5-carboxamide;
I-48 1 -[(1 R)-1 -(3,4-difluorophenyl)-2-hydroxyethyl]-N-[3-[3-[[1 -(2-methoxyethyl)- piperidin-4-yl]amino]quinoxalin-6-yl]prop-2-ynyl]-2-oxopyridine-3- carboxamide;
I-49 1 -[(3,4-difluorophenyl)methyl]-N-[3-[3-[[1 -(2-methoxyethyl)piperidin-4- yl]amino]quinoxalin-6-yl]prop-2-ynyl]-2-oxopyridine-3-carboxamide;
I-53 2-[(3,4-difluorophenyl)methyl]-N-[3-[3-[[1 -(2-methoxyethyl)piperidin-4- yl]amino]quinoxalin-6-yl]prop-2-ynyl]-3-oxopyridazine-4-carboxamide;
I-57 1 -[(3,4-difluorophenyl)methyl]-N-[3-[3-[[1 -(oxetan-3-yl)piperidin-4- yl]amino]quinoxalin-6-yl]prop-2-ynyl]-2-oxopyridine-3-carboxamide;
1-61 N-[3-[3-[[4-[4-(cyclopropylmethyl)piperazin-1 -yl]cyclohexyl]amino]quinoxalin-
6-yl]prop-2-ynyl]-1 -[(1 S)-1 -(3-fluorophenyl)-2-hydroxyethyl]-2-oxopyridine-3- carboxamide;
I-73 1 -[(3,4-difluorophenyl)methyl]-N-[3-[3-[[1 -(2-methoxyethyl)piperidin-4- yl]amino]quinolin-6-yl]prop-2-ynyl]-6-oxopyrimidine-5-carboxamide;
I-77 1 -[(1 R)-1 -(3,4-difluorophenyl)-2-hydroxyethyl]-N-[3-[3-[[1 -(2-methoxyethyl)- piperidin-4-yl]amino]quinolin-6-yl]prop-2-ynyl]-2-oxopyridine-3-carboxamide;
I- 87 1 -[(3,4-difluorophenyl)methyl]-6-oxo-N-[3-(3-piperidin-4-yloxyquinolin-6- yl)prop-2-ynyl]pyrimidine-5-carboxamide;
II- 88 1 -[(3,4-difluorophenyl)methyl]-6-oxo-N-[3-[4-(1 H-pyrrol-3-yl)quinolin-6- yl]prop-2-ynyl]pyrimidine-5-carboxamide.
13. Compounds of general formula (1 ) according to one of claims 1 to 12 - or the pharmaceutically acceptable salts thereof - as medicaments.
14. Compounds of general formula (1 ) according to one of claims 1 to 12 - or the pharmaceutically acceptable salts thereof - for use in the treatment and/or prevention of cancer, infections, inflammations and autoimmune diseases.
15. Pharmaceutical preparation containing as active substance one or more compounds of general formula (1 ) according to one of claims 1 to 12 - or the pharmaceutically acceptable salts thereof - optionally in combination with conventional excipients and/or carriers.
16. Pharmaceutical preparation comprising a compound of general formula (1 ) according to one of claims 1 to 12 - or one of the pharmaceutically acceptable salts thereof - and at least one other cytostatic or cytotoxic active substance, different from formula (1 ).
PCT/EP2011/056368 2010-04-21 2011-04-20 Heterocyclic carboxylic acid amides as pdk1 inihibitors WO2011131741A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
EP11716881.5A EP2560967B1 (en) 2010-04-21 2011-04-20 Heterocyclic carboxylic acid amides as PDK1 inihibitors
JP2013505483A JP5860031B2 (en) 2010-04-21 2011-04-20 Heterocyclic carboxylic amides as PDK1 inhibitors

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP10160552 2010-04-21
EP10160552.5 2010-04-21

Publications (1)

Publication Number Publication Date
WO2011131741A1 true WO2011131741A1 (en) 2011-10-27

Family

ID=42306748

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2011/056368 WO2011131741A1 (en) 2010-04-21 2011-04-20 Heterocyclic carboxylic acid amides as pdk1 inihibitors

Country Status (6)

Country Link
US (1) US8575203B2 (en)
EP (1) EP2560967B1 (en)
JP (1) JP5860031B2 (en)
TW (1) TW201204362A (en)
UY (1) UY33349A (en)
WO (1) WO2011131741A1 (en)

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013075084A1 (en) * 2011-11-18 2013-05-23 Constellation Pharmaceuticals Modulators of methyl modifying enzymes, compositions and uses thereof
WO2013075083A1 (en) * 2011-11-18 2013-05-23 Constellation Pharmaceuticals Modulators of methyl modifying enzymes, compositions and uses thereof
WO2013135671A1 (en) 2012-03-13 2013-09-19 Basf Se Fungicidal pyrimidine compounds
WO2014055955A1 (en) 2012-10-05 2014-04-10 Rigel Pharmaceuticals, Inc. Gdf-8 inhibitors
WO2015036058A1 (en) 2013-09-16 2015-03-19 Basf Se Fungicidal pyrimidine compounds
WO2015036059A1 (en) 2013-09-16 2015-03-19 Basf Se Fungicidal pyrimidine compounds
US9085583B2 (en) 2012-02-10 2015-07-21 Constellation—Pharmaceuticals, Inc. Modulators of methyl modifying enzymes, compositions and uses thereof
WO2016133838A1 (en) 2015-02-20 2016-08-25 Rigel Pharmaceuticals, Inc. Gdf-8 inhibitors
US9745305B2 (en) 2013-03-15 2017-08-29 Constellation Pharmaceuticals, Inc. Modulators of methyl modifying enzymes, compositions and uses thereof
US9969716B2 (en) 2013-08-15 2018-05-15 Constellation Pharmaceuticals, Inc. Indole derivatives as modulators of methyl modifying enzymes, compositions and uses thereof
US10457640B2 (en) 2016-10-19 2019-10-29 Constellation Pharmaceuticals, Inc. Synthesis of inhibitors of EZH2
US10577350B2 (en) 2015-08-28 2020-03-03 Constellation Pharmaceuticals, Inc. Crystalline forms of (R)-N-((4-methoxy-6-methyl-2-oxo-1,2-dihydropyridin-3-yl)methyl)-2-methyl-1-(1-(1-(2,2,2-trifluoroethyl)piperidin-4-yl)ethyl)-1H-indole-3-carboxamide

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2571877B1 (en) * 2010-05-17 2018-08-15 Boehringer Ingelheim International GmbH 1h-imidazo[4,5-c]quinolines
CN102775358B (en) * 2012-08-22 2015-05-27 山东齐都药业有限公司 Preparation method of 6-fluoro-3-hydroxy-2-pyrazinamide
MX2018004109A (en) 2015-10-05 2018-09-27 Univ Columbia Activators of autophagic flux and phospholipase d and clearance of protein aggregates including tau and treatment of proteinopathies.

Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4996213A (en) 1988-06-09 1991-02-26 Sanofi Derivatives of 4-amino 3-carboxy naphthyridines and their pharmaceutical compositions
WO2006071017A1 (en) 2004-12-29 2006-07-06 Hanmi Pharm. Co., Ltd. Quinazoline derivatives for inhibiting cancer cell growth and method for the preparation thereof
WO2006135993A1 (en) 2005-06-24 2006-12-28 Gilead Sciences, Inc. Pyrido(3,2-d)pyrimidines and pharmaceutical compositions useful for treating hepatitis c.
WO2007104560A1 (en) 2006-03-15 2007-09-20 Grünenthal GmbH Substituted 4-amino-quinazoline derivatives as regulators of metabotropic glutamate receptors and their use for producing drugs
WO2007133637A2 (en) 2006-05-10 2007-11-22 Renovis, Inc. Amide derivatives as ion-channel ligands and pharmaceutical compositions and methods of using the same
WO2008005457A2 (en) 2006-06-30 2008-01-10 Sunesis Pharmaceuticals Pyridinonyl pdk1 inhibitors
WO2008020302A2 (en) 2006-08-17 2008-02-21 Pfizer Products Inc. Heteroaromatic quinoline-based compounds as phosphodiesterase (pde) inhibitors
WO2008138878A2 (en) 2007-05-10 2008-11-20 Æterna Zentaris Gmbh Novel pyridopyrazine derivatives, process of manufacturing and uses thereof
WO2009003669A2 (en) 2007-06-29 2009-01-08 4 Aza Ip Nv Pyrido(3,2-d)pyrimidines and pharmaceutical compositions useful for medical treatment
WO2010007114A2 (en) 2008-07-16 2010-01-21 Boehringer Ingelheim International Gmbh New chemical compounds
WO2010007116A2 (en) 2008-07-16 2010-01-21 Boehringer Ingelheim International Gmbh New chemical compounds

Patent Citations (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4996213A (en) 1988-06-09 1991-02-26 Sanofi Derivatives of 4-amino 3-carboxy naphthyridines and their pharmaceutical compositions
WO2006071017A1 (en) 2004-12-29 2006-07-06 Hanmi Pharm. Co., Ltd. Quinazoline derivatives for inhibiting cancer cell growth and method for the preparation thereof
US20090036430A1 (en) 2004-12-30 2009-02-05 Steven Cesar Alfons De Jonghe Pyrido(3,2-d)pyrimidines and pharmaceutical compositions useful for medical treatment
WO2006135993A1 (en) 2005-06-24 2006-12-28 Gilead Sciences, Inc. Pyrido(3,2-d)pyrimidines and pharmaceutical compositions useful for treating hepatitis c.
WO2007104560A1 (en) 2006-03-15 2007-09-20 Grünenthal GmbH Substituted 4-amino-quinazoline derivatives as regulators of metabotropic glutamate receptors and their use for producing drugs
WO2007133637A2 (en) 2006-05-10 2007-11-22 Renovis, Inc. Amide derivatives as ion-channel ligands and pharmaceutical compositions and methods of using the same
WO2008005457A2 (en) 2006-06-30 2008-01-10 Sunesis Pharmaceuticals Pyridinonyl pdk1 inhibitors
WO2008020302A2 (en) 2006-08-17 2008-02-21 Pfizer Products Inc. Heteroaromatic quinoline-based compounds as phosphodiesterase (pde) inhibitors
WO2008138878A2 (en) 2007-05-10 2008-11-20 Æterna Zentaris Gmbh Novel pyridopyrazine derivatives, process of manufacturing and uses thereof
WO2009003669A2 (en) 2007-06-29 2009-01-08 4 Aza Ip Nv Pyrido(3,2-d)pyrimidines and pharmaceutical compositions useful for medical treatment
WO2010007114A2 (en) 2008-07-16 2010-01-21 Boehringer Ingelheim International Gmbh New chemical compounds
WO2010007116A2 (en) 2008-07-16 2010-01-21 Boehringer Ingelheim International Gmbh New chemical compounds

Cited By (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2013075084A1 (en) * 2011-11-18 2013-05-23 Constellation Pharmaceuticals Modulators of methyl modifying enzymes, compositions and uses thereof
WO2013075083A1 (en) * 2011-11-18 2013-05-23 Constellation Pharmaceuticals Modulators of methyl modifying enzymes, compositions and uses thereof
US9409865B2 (en) 2011-11-18 2016-08-09 Constellation_Pharmaceuticals, Inc. Modulators of methyl modifying enzymes, compositions and uses thereof
US9206128B2 (en) 2011-11-18 2015-12-08 Constellation Pharmaceuticals, Inc. Modulators of methyl modifying enzymes, compositions and uses thereof
US20140288123A1 (en) * 2011-11-18 2014-09-25 Constellation Pharmaceuticals, Inc. Modulators of methyl modifying enzymes, compositions and uses thereof
US9051269B2 (en) 2011-11-18 2015-06-09 Constellation Pharmaceuticals, Inc. Modulators of methyl modifying enzymes, compositions and uses thereof
US10016405B2 (en) 2012-02-10 2018-07-10 Constellation Pharmaceuticals, Inc. Modulators of methyl modifying enzymes, compositions and uses thereof
USRE47428E1 (en) 2012-02-10 2019-06-11 Constellation Pharmaceuticals, Inc. Modulators of methyl modifying enzymes, compositions and uses thereof
US9085583B2 (en) 2012-02-10 2015-07-21 Constellation—Pharmaceuticals, Inc. Modulators of methyl modifying enzymes, compositions and uses thereof
US9371331B2 (en) 2012-02-10 2016-06-21 Constellation Pharmaceuticals, Inc. Modulators of methyl modifying enzymes, compositions and uses thereof
US9980952B2 (en) 2012-02-10 2018-05-29 Constellation Pharmaceuticals, Inc. Modulators of methyl modifying enzymes, compositions and uses thereof
US9469646B2 (en) 2012-02-10 2016-10-18 Constellation Pharmaceuticals, Inc. Modulators of methyl modifying enzymes, compositions and uses thereof
WO2013135671A1 (en) 2012-03-13 2013-09-19 Basf Se Fungicidal pyrimidine compounds
WO2014055955A1 (en) 2012-10-05 2014-04-10 Rigel Pharmaceuticals, Inc. Gdf-8 inhibitors
US9745305B2 (en) 2013-03-15 2017-08-29 Constellation Pharmaceuticals, Inc. Modulators of methyl modifying enzymes, compositions and uses thereof
US9969716B2 (en) 2013-08-15 2018-05-15 Constellation Pharmaceuticals, Inc. Indole derivatives as modulators of methyl modifying enzymes, compositions and uses thereof
WO2015036059A1 (en) 2013-09-16 2015-03-19 Basf Se Fungicidal pyrimidine compounds
WO2015036058A1 (en) 2013-09-16 2015-03-19 Basf Se Fungicidal pyrimidine compounds
WO2016133838A1 (en) 2015-02-20 2016-08-25 Rigel Pharmaceuticals, Inc. Gdf-8 inhibitors
US10577350B2 (en) 2015-08-28 2020-03-03 Constellation Pharmaceuticals, Inc. Crystalline forms of (R)-N-((4-methoxy-6-methyl-2-oxo-1,2-dihydropyridin-3-yl)methyl)-2-methyl-1-(1-(1-(2,2,2-trifluoroethyl)piperidin-4-yl)ethyl)-1H-indole-3-carboxamide
US10457640B2 (en) 2016-10-19 2019-10-29 Constellation Pharmaceuticals, Inc. Synthesis of inhibitors of EZH2

Also Published As

Publication number Publication date
TW201204362A (en) 2012-02-01
JP5860031B2 (en) 2016-02-16
UY33349A (en) 2011-12-01
EP2560967B1 (en) 2017-12-20
US20120094976A1 (en) 2012-04-19
JP2013525333A (en) 2013-06-20
US8575203B2 (en) 2013-11-05
EP2560967A1 (en) 2013-02-27

Similar Documents

Publication Publication Date Title
EP2552905B1 (en) B-Raf kinase inhibitors
US9296748B2 (en) Pyridinones
DK2925761T3 (en) Triazole pyrazines as BRD4 inhibitors for use in the treatment of cancer
US8889684B2 (en) Azaindolylphenyl sulfonamides as serine/threonine kinase inhibitors
US9428513B2 (en) Triazolopyrazine
WO2011131741A1 (en) Heterocyclic carboxylic acid amides as pdk1 inihibitors
US8865703B2 (en) Pyridyltriazoles
JP5809288B2 (en) Oxindole pyrimidines as IGF1R receptor inhibitors
JP6128133B2 (en) 5,8-Dihydro-6H-pyrazolo [3,4-H] quinazoline as an IGF-1R / IR inhibitor
EP2571877B1 (en) 1h-imidazo[4,5-c]quinolines
US9428515B2 (en) Benzimidazole derivatives
US8710055B2 (en) Triazolylphenyl sulfonamides as serine/threonine kinase inhibitors
OA19258A (en) Triazolopyrazine

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 11716881

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2013505483

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2011716881

Country of ref document: EP