AU2011226830A1 - Combinations for the treatment of diseases involving cell proliferation - Google Patents

Combinations for the treatment of diseases involving cell proliferation Download PDF

Info

Publication number
AU2011226830A1
AU2011226830A1 AU2011226830A AU2011226830A AU2011226830A1 AU 2011226830 A1 AU2011226830 A1 AU 2011226830A1 AU 2011226830 A AU2011226830 A AU 2011226830A AU 2011226830 A AU2011226830 A AU 2011226830A AU 2011226830 A1 AU2011226830 A1 AU 2011226830A1
Authority
AU
Australia
Prior art keywords
compound
synthetic
inhibitor
derivative
treatment
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
AU2011226830A
Inventor
Anke Baum
Gerd Munzert
Martin Steegmaier
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Boehringer Ingelheim International GmbH
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 AU2011226830A priority Critical patent/AU2011226830A1/en
Publication of AU2011226830A1 publication Critical patent/AU2011226830A1/en
Abandoned legal-status Critical Current

Links

Abstract

C:\NRPonb\DCC\DAR\3891776_1.DOC-22/09/2OlI The present invention relates to a pharmaceutical composition for the treatment of diseases which involve cell proliferation. The invention also relates to a method for the treatment of 5 said diseases, comprising co-administration of a compound 1 of Formula (I) wherein the groups L, RI, R2, R3, R4 and R5 have the meanings given in the claims and specification, optionally in form of its tautomers, racemates, enantiomers, diastereomers and the mixtures thereof and optionally in form of the pharmacologically acceptable acid addition salts, solvates, hydrates, polymorphs, physiologically functional derivatives or prodrugs thereof, and 10 of an effective amount of an active compound 2 and/or co-treatment with radiation therapy, in a ratio which provides an additive and synergistic effect, and to the combined use of a compound 1 of Formula (I) and of an effective amount of an active compound 2 and/or radiotherapy for the manufacture of corresponding pharmaceutical combination preparations. 4 HN HNAN N In m

Description

Australian Patents Act 1990 - Regulation 3.2A ORIGINAL COMPLETE SPECIFICATION STANDARD PATENT Invention Title "Combinations for the treatment of diseases involving cell proliferation" The following statement is a full description of this invention, including the best method of performing it known to us:- C.\NRPortbl\DCC\DAR\3891776_ IDOC-22/09/2011 Combinations for the Treatment of Diseases involving Cell Proliferation This application is a divisional of Australian Patent Application No. 2005274384, the entire content of which is incorporated herein by reference. 5 The invention relates to new pharmaceutical compositions for the treatment of diseases involving cell proliferation, migration or apoptosis of cancer cells, or angiogenesis and the preparation thereof. The invention further relates to a method for the treatment of diseases involving cell proliferation, migration or apoptosis of cancer cells, or angiogenesis, which method comprises co-administration to a person in need of such 10 treatment and/or co-treatment of a person in need of such treatment with effective amounts of: (i) A compound 1 of Formula (I) 15 HN0 R4 RI HN a'N N2 RR 0 NH 20 Ln R~m (I) wherein the groups L, R', R 2 , R 3 , R4 and R 5 have the meanings given in the 25 claims and specification, optionally in form of its tautomers, racemates, enantiomers, diastereomers and the mixtures thereof and optionally in form of the pharmacologically acceptable acid addition salts, solvates, hydrates, polymorphs, physiologically functional derivatives or prodrugs thereof; and (ii) At least a further chemotherapeutic, immunotherapeutic or immunomodulatory, antiangiogenic, hormonal or naturally occurring, semi 30 synthetic or synthetic therapeutic agent 2; and/or WO 2006/018182 PC'I'/EP2005/008623 2 (iii) Radiotherapy or radio-immunotherapy. Background of the Invention 5 Polo-like kinases (PLKs) are serine/ threonine kinases that play important roles in regulating processes in the cell cycle. There are four PLKs disclosed in the state of the art, i.e. PLK-1,'PLK-2, PLK-3.and PLK-4.~PLKs play a role in the entry into and the exit from mitosis in mammalian cells. Especially for PLK-1 a central role with respect 10 to the regulation of mitosis was shown (Glover et al. 1998, Genes Dev. 12:3777-87; Qian et al. 2001, Mol Biol Cell. 12:1791-9). Overexpression of PLK-1 seems to be strongly associated with neoplastic cells including cancers (WO 2004/014899). Overexpression of PLK1 has been documented for various tumor types such as non small cell lung cancer, squamous cell carcinomas, breast, ovary or papillary carcinomas is as well as colorectal cancers (Wolf et al. 1997, Oncogene 14, pages 543-549; Knecht et al. 1999, Cancer Res. 59, pages 2794-2797; Wolf et al. 2000, Pathol Res Pract. 196, pages 753-759; Weichert et al. 2004, Br. J. Cancer 90, pages 815-821; Ito et al. 2004, Br. J. Cancer 90, pages 414-418; Takahashi et al. 2003, Cancer Sci. 94, pages 148 152). 20 For the treatment of diseases of oncological nature, a large number of chemotherapeutic, immunotherapeutic or immunomodulatory, antiangiogenic or hormonal agents have already been suggested, which can be used as monotherapy (treatment with one agent) or as combination therapy (simultaneous, separate or 25 sequential treatment with more than one agent) and/or which may be combined with radiotherapy or radio-immunotherapy. In this respect, chemotherapeutic agent means a naturally occurring, semi-synthetic or synthetic chemical compound which, alone or via further activation, for example with radiations in the case of radio-immunotherapy, inhibits or kills growing cells, and which can be used or is approved for use in the 30 treatment of diseases of oncological nature, which are commonly also denominated as cancers. In the literature, these agents are generally classified according to their mechanism of action. In this matter, reference can be made, for example, to the WO 2006/018182 PCT/EP2005/008623 3 classification made in "Cancer Chemotherapeutic Agents", American Chemical Society, 1995, W.O. Foye Ed. The efficacy of chemotherapeutic agents can be improved by using combination therapies with other chemotherapeutic, immunotherapeutic, irmunomodulatory, 5 antiangiogenic or hormonal compounds. Combination therapies constitute the gold standard in many settings of cancer therapy. Even if the concept of combining several therapeutic agents or therapies already has been suggested, and although various combination therapies are under investigation and in clinical trials, there is still a need for new and efficient therapeutic compositions for 10 the treatment of cancer diseases, which show advantages over standard therapies. It is the purpose of the present invention to provide a combination therapy with the PLK Inhibitors of Formula (I) for the treatment of various cancer diseases. 15 Description of the invention Thus, within the meaning of the present invention, the following classes of chemotherapeutic agents are especially of interest, although not representing a limitation: 20 * Synthetic small molecule VEGF receptor antagonists e Small molecule growth factor (GF) receptor antagonists e Inhibitors of the EGF receptor and/or VEGF receptor and/or integrin receptors or any other protein tyrosine kinase receptors, which are not classified under the 25 synthetic small-molecules e Small molecule inhibitors of the Ras/Raf/MAPK or PI3K/AKT pathways or any other scrine/threonine kinases. " Inhibitors of the Ras/Raf/MAPK or PI3K/AKT pathways or any other serine/threonine kinases, which are not classified under the synthetic small 30 molecules WO 2006/018182 PC'r/EP2005/008623 o Inhibitors directed to EGF receptor and/or VEGF receptor and/or integrin receptors or any other protein tyrosine kinase receptors, which are synthetically manufactured antibodies, antibody fragments or fusion proteins o Compounds which interact with nucleic acids and which are classified as s alkylating agents or platinum compounds o Compounds which interact with nucleic acids and which are classified as anthracyclines, as DNA intercalators or as DNA cross-linking agents o Anti-metabolites o Naturally occurring, semi-synthetic or synthetic bleomycin type antibiotics 1o (BLM-group antibiotics) o Inhibitors of DNA transcribing enzymes, especially topoisomerase I or topoisomerase II inhibitors o Chromatin modifying agents o Mitosis inhibitors, anti-mitotic agents, or cell-cycle inhibitors 15 0 Compounds interacting with or binding tubulin 0 Compounds inhibiting mitotic kinesins or other motor proteins including but not limited to Eg5, CENP-E, MCAK, Kid, MKLP-l e Proteasome inhibitors * Heat shock protein inhibitors 20 * Compounds targeting the anti-apoptotic function of Bcl-2, Bcl-xj and like molecules e Enzymes Hormones, hormone antagonists or hormone inhibitors, or inhibitors of steroid biosynthesis * Steroids 25 * Cytokines, hypoxia-selective cytotoxins, inhibitors of cytokines, lymphokines, antibodies directed against cytokines or oral and parenteral tolerance induction strategies * Supportive agents e Antiinflammatory compounds such as but not limited to COX-2 inhibitors 30 e Chemical radiation sensitizers and protectors * Photochemically activated drugs a Synthetic poly- or oligonucleotides WO 2006/018182 PCT/EP2005/008623 5 o Other chemotherapeutic or naturally occurring, semi-synthetic or synthetic therapeutic agents, such as cytotoxic antibiotics, antibodies targeting surface molecules of cancer cells, antibodies targeting growth factors or their receptors, inhibitors of metalloproteinases, inhibitors of oncogenes, inhibitors of gene 5 transcription or of RNA translation or protein expression, or complexes of rare earth elements. The beneficial effects of the invention are mainly based on the additive and synergistic effects of the combined treatment, or to an improved tolerability of the treatment by the 10 patient due, for example, to the administration of lower doses of the therapeutic agents involved. Within the meaning of the present invention, the compound 1 has the structure of the following general Formula (I): 15 N~ Ny2 HN N R4R R HR o NH Ln
R
5 "' (I) 20 wherein
R
1 , R 2 which may be identical or different, denote hydrogen or optionally substituted
CI-C
6 -alkyl, WO 2006/018182 PCT/EP2005/008623 6 or
R
1 and R 2 together denote a 2- to 5-membered alkyl bridge which may contain I to 2 heteroatoms, s R 3 denotes hydrogen or a group selected from among optionally substituted CI-C 1 2 alkyl, C 2
-C
12 -alkenyl, C 2
-C
12 -alcynyl and C 6
-C
14 -aryl, or a group selected from among optionally substituted and/or bridged C 3
-C
12 -cycloalkyl,
C
3
-C
12 -cycloalkenyl, C 7
-C
12 -polycycloalkyl, C7-C2-polycycloalkenyl, C 5
-C
12 spirocycloalkyl, C 3 -Ci 2 -heterocycloalkyl which contains 1 to 2 heteroatoms, and 10 C 3
-C
12 -heterocycloalkenyl which contains 1 to 2 heteroatoms, or R' and R 3 or R 2 and R 3 together denote a saturated or unsaturated C 3
-C
4 -alkyl bridge which may contain 1 heteroatom, is R 4 denotes a group selected from among hydrogen, -CN, hydroxy, -NR 6
R
7 and halogen, or a group selected from among optionally substituted Cx-C 6 -alkyl, C 2 -C6-alkenyl, C 2
-C
6 alkynyl, CI-C 5 -alkyloxy, C 2
-C
5 -alkenyloxy, C 2 -Cs-alkynyloxy, CI-C 6 -alkylthio, CI-C 6 alkylsulphoxo and C-C6-alkylsulphonyl, 20 L denotes a linker selected from among optionally substituted C 2 -CIO-alkyl, C 2 Clo-alkenyl, C 6
-C
14 -aryl, -C 2
-C
4 -alkyl-C 6
-C
4 -aryl, -C6-C 1 4 -aryl-CI-C 4 -alkyl, optionally bridged C 3
-C
12 -cycloalkyl and heteroaryl which contains 1 or 2 nitrogen atoms, 25 n denotes 0 or I In denotes 1 or 2
R
5 denotes a group selected from among optionally substituted morpholinyl, piperidinyl, piperazinyl, piperazinylcarbonyl, pyrrolidinyl, tropenyl, R 8 30 diketomethylpiperazinyl, sulphoxomorpholinyl, sulphonylmorpholinyl, thiomorpholinyl, -NR 8
R
9 and azacycloheptyl, R , R 7 which may be identical or different, denote hydrogen or Ci-C 4 -alkyl, WO 2006/018182 PCT/EP2005/008623 7 and
R
8 , R 9 denote unsubstituted nitrogen substituents at R 5 , which may be identical or different, denote either hydrogen or a group selected from among C-CG-alkyl, -C-C 4 alkyl-C 3
-C
1 0 -cycloalkyl, C 3 -Cio-cycloalkyl, C 6
-C
14 -aryl, -C 1
-C
4 -alkyl-C 6 -Ci 4 -aryl, 5 pyranyl, pyridinyl, pyrimidinyl, C-C 4 -alkyloxycarbonyl, C 6
-C
1 4 -arylcarbonyl, C 1
-C
4 alkylcarbonyl, C 6
-CI
4 -arylrnethyloxycarbonyl, C6-C 1 4-arylsulphonyl, C-C 4 alkylsulphonyl- and C 6
-C
1 4 -aryl-C-C4-alkylsulphonyl-, optionally in form of its tautomers, racemates, enantiomers, diastereomers and the mixtures thereof and optionally in form of the pharmacologically acceptable acid 10 addition salts, solvates, hydrates, polymorphs, physiologically functional derivatives or prodrugs thereof . Preferred compounds of Formula (1) are those wherein R1 to R 4 , R 6 and R 7 are as hereinbefore defined, and is L denotes a linker selected from among optionally substituted C 2 -Cio-alkyl, C 2 Cio-alkenyl, C 6
-C
14 -aryl, -C 2
-C
4 -alkyl-C 6
-C
1 4 -aryl, -CG-C 14 -aryl-CI-C 4 -alkyl, optionally bridged C 3
-C
12 -cycloalkyl and heteroaryl which contains 1 or 2 nitrogen atoms n denotes I m denotes 1 or 2 20 R 5 denotes a group which is bound to L via a nitrogen atom, selected from among optionally substituted morpholinyl, piperidinyl, R-piperazinyl, pyrrolidinyl, tropenyl,
R
8 -diketomethylpiperazinyl, sulphoxomorpholinyl, sulphonylmorpholinyl, thiomorpholinyl, -NR 8
R
9 and azacycloheptyl,
R
8 , R 9 denote unsubstituted nitrogen substituents at R 5 , which may be identical or 25 different, hydrogen or a group selected from among C-C 6 -alkyl, -C-C 4 -alkyl-C 3
-CIO
cycloalkyl, C 3 -Cio-cycloalkyl, C 6
-C
14 -aryl, -C-C 4 -alkyl-C 6
-C
14 -aryl, pyranyl, pyridinyl, pyrimidinyl, C-C 4 -alkyloxycarbonyl, C 6
-C
14 -arylcarbonyl, C-C 4 -alkylcarbonyl, C 6 C 14 -arylmethyloxycarbonyl, C 6
-C
14 -arylsulphonyl, C-C 4 -alkylsulphonyl and C 6
-C
4 aryl-C 1
-C
4 -alkylsulphonyl. 30 Also preferred are compounds of Formula (I), wherein R' to R 4 , R 6 and R 7 are as hereinbefore defined, WO 2006/018182 PCT/EP2005/008623 8 L denotes a linker selected from among optionally substituted C 2 -CIO-alkyl, C 2 CIO-alkenyl, C 6
-C
14 -aryl, -C 2
-C
4 -alkyl-C 6
-CI
4 -aryl, -C6-Ci 4 -aryl-CI-C 4 -alkyl, optionally bridged C 3
-C
12 -cycloalkyl and heteroaryl which contains 1 or 2 nitrogen atoms n denotes 0 or 1 5 m denotes 1 or 2 Rs denotes a group which is bound to L via a carbon atom, selected from among R piperidinyl, RBR9-piperazinyl, R 8 -pyrrolidinyl, Rs- piperazinylcarbonyl, R 8 -tropenyl, R 8 morpholinyl and R 8 -azacycloheptyl, and 10 R , R 9 denote unsubstituted nitrogen substituents at R , which may be identical or different, hydrogen or a group selected from among CI-C 6 -alkyl, -CI-C 4 -alkyl-C 3 -Cio cycloalkyl, C 3 -Clo-cycloalkyl, C 6
-C
4 -aryl, -Ci-C 4 -alkyl-C 6
-C
14 -aryl, pyranyl, pyridinyl, pyrimidinyl, C 1
-C
4 -alkyloxycarbonyl, C6-C 4 -arylcarbonyl, Ci-C 4 -alkylcarbonyl, C 6 C 14 -arylmethyloxycarbonyl, C6-C4-arylsulphonyl, Ci-C 4 -alkylsulphonyl and C 6
-C
4 is aryl-C 1
-C
4 -alkylsulphonyl, optionally in the form of the tautomers, the racemates, the enantiomers, the diastereomers and the mixtures thereof, and optionally the pharmacologically acceptable acid addition salts thereof. 20 Particularly preferred are compounds of Formula I wherein L, n, n and R 3 to R 9 are as hereinbefore defined, and R', R 2 which may be identical or different, denote a group selected from among hydrogen, Me, Et, Pr, or 25 R' and R 2 together form a C 2
-C
4 -alkyl bridge , optionally in the form of the tautomers, the racemates, the enantiomers, the diastereomers and the mixtures thereof, and optionally the pharmacologically acceptable acid addition salts thereof. 30 Especially preferred are compounds of Formula I wherein R' , R 2 , m, n and R 5 to R 8 are as hereinbefore defined, and WO 2006/018182 PCT/EP2005/008623 9
R
3 denotes a group selected from among optionally substituted C-Cio-alkyl, C 3
-C
cycloalkyl, C 3
-C
6 -heterocycloalkyl and C 6
-C
1 4 -aryl or
R
1 and R 3 or R 2 and R 3 together denote a saturated or unsaturated C 3
-C
4 -alkyl bridge which may contain 1 to 2 heteroatoms, 5 R 4 denotes a group selected from among hydrogen, OMe, OH, Me, Et, Pr, OEt, NHMe,
NH
2 , F, CL, Br, 0-propargyl, 0-butynyl, CN, SMe, NMe 2 , CONH 2 , ethynyl, propynyl, butynyl and allyl, and L denotes a linker selected from among optionally substituted phenyl, phenylmethyl, 10 cyclohexyl and branched Cl-C 6 -alkyl, optionally in the form of the tautomers, the racemates, the enantiomers, the diastereomers and the mixtures thereof, and optionally the pharmacologically acceptable acid addition salts thereof. 15 In a further embodiment, the compound j in accordance with the present invention is selected from the group consisting of the compounds of Formula (I) shown in the following Table 0 H3 H N O R~m-L--***N N~ N N N R R' R Config. Ex. R' R2 o R R4 L.-R. RI or R 27 H X R O C' a
H
3 C CH, WO 2006/018182 PCT/EP2005/008623 10 44 H X C R H ) , H SH )cH 55 H R X4 58 H X CH 3 Rvcllj ) ~cl X CHa ,F 102 H R H 3 C CH 3
K
0 H3 103 H CH 3 R O 105 H CH 3 R O CH 3 110 H CH3 R HaC 0a CH3 H 3 1 .CH 3 7 115 H x2N CF 3 R 6I WO 2006/018182 PCT/EP2005/008623 11 133 H X CH 3 R CH6 CH 134 H R 0. 234 H Xj-% CH, R ON CH H C
CH
3 x, ?H 3 240 H CH R ON SC H C NQ wherein the abbreviations X 1 , X 2 , X 3 , X 4 and X 5 used in the Table in each case denote a link to a position in the general Formula shown in the Table instead of the corresponding groups R 1 , R 2 , R 3 , R 4 and L-R'. 5 This invention thus relates to a pharmaceutical composition comprising effective amounts of: (i) A compound 1 of Formula (I) or optionally a polymorph, metabolite, 10 hydrate, preferably the mono hydrate, solvate, individual optical isomers, mixtures of the individual enantiomers or racemates thereof, or a pharmaceutically acceptable salt thereof; and (ii) At least one further chemotherapeutic or naturally occurring, semi 15 synthetic or synthetic therapeutic agent 2; WO 2006/018182 PCT/EP2005/008623 12 optionally in combination with one or more pharmaceutically acceptable excipients, and optionally adapted for a co-treatment with radiotherapy or radio-immunotherapy, in the form of a combined preparation for simultaneous, separate or sequential use in the treatment of diseases involving cell proliferation, migration or apoptosis of cancer cells, s or angiogenesis, preferably involving cell proliferation or apoptosis of cancer cells. In a preferred embodiment the instant invention is directed to a pharmaceutical composition, wherein the further chemotherapeutic or naturally occurring, semi synthetic or synthetic therapeutic agent 2 is selected from the group consisting of io compounds interacting with or binding tubulin, synthetic small molecule VEGF receptor antagonists, small molecule growth factor receptor antagonists, inhibitors of the EGF receptor and/or VEGF receptor and/or integrin receptors or any other protein tyrosine kinase receptors which are not classified under the synthetic small-molecules, inhibitors directed to EGF receptor and/or VEGF receptor and/or integrin receptors or 15 any other protein tyrosine kinase receptors, which are fusion proteins, compounds which interact with nucleic acids and which are classified as alkylating agents or platinum compounds, compounds which interact with nucleic acids and which are classified as anthracyclines, as DNA intercalators or as DNA cross-linking agents, including DNA minor-groove binding compounds, anti-metabolites, naturally 20 occurring, semi-synthetic or synthetic bleomycin type antibiotics, inhibitors of DNA transcribing enzymes, and especially the topoisomerase I or topoisomerase II inhibitors, chromatin modifying agents, mitosis inhibitors, anti-mitotic agents, cell-cycle .inhibitors, proteasome inhibitors, enzymes, hormones, hormone antagonists, hormone inhibitors, inhibitors of steroid biosynthesis, steroids, cytokines, hypoxia-selective 25 cytotoxins, inhibitors of cytokines, lymphokines, antibodies directed against cytokines, oral and parenteral tolerance induction agents, supportive agents, chemical radiation sensitizers and protectors, photo-chemically activated drugs, synthetic poly- or oligonucleotides, optionally modified or conjugated, non-steroidal anti-inflammatory drugs, cytotoxic antibiotics, antibodies targeting the surface molecules of cancer cells, 30 antibodies targeting growth factors or their receptors, inhibitors of metalloproteinases, metals, inhibitors of oncogenes, inhibitors of gene transcription or of RNA translation WO 2006/018182 PCT1EP2005/008623 13 or protein expression, complexes of rare earth elements, and photo-chemotherapeutic agents. Preferred compounds include small molecule tyrosin kinase or serine/threonine kinase 5 inhibitors, compounds interacting with nucleic acids classified as alkylating agents or anthracyclines, anti-metabolites, inhibitors of DNA transcribing enzymes such as topoisomerase I or II, tubulin binding drugs, anti-mitotic agents, antibodies targeting growth factors or their receptors and -antibodies binding-to surface molecules of cancer cells or ligands of these surface molecules in form of the hydrates and/or solvates and 10 optionally in the form of the individual optical isomers, mixtures of the individual enantiomers or racemates thereof. In another preferred embodiment the instant invention is directed to a pharmaceutical combination, wherein the further chemotherapeutic or naturally occurring, semi is synthetic or synthetic therapeutic agent 2 is selected from the group consisting of a small molecule VEGF receptor antagonist such as vatalanib (PTK-787/ZK222584), SU 5416, SU-6668, SU-11248, SU-14813, AZD-6474, AZD-2171, CP-547632, CEP-7055, AG-013736, IM-842 or GW-786034, a dual EGFR/HER2 antagonist such as gefitinib, erlotinib, CI-1033 or GW-2016, an EGFR antagonist such as iressa (ZD-1839), tarceva 20 (OSI-774), PKI-166, EKB-569, HKI-272 or herceptin, an antagonist of the mitogen activated protein kinase such as BAY-43-9006 or BAY-57-9006, a quinazoline derivative such as 4-[(3-chloro-4-fluorophenyl)amino]-6-{[4-(N,N-dimethylamino)-1 oxo-2-buten-1-yl]amino}-7-((S)-tetrahydrofuran-3-yloxy)-quinazoline or 4-[(3-chloro 4-fluoro-phenyl)amino]-6- {[4-(homomorpholin-4-yl)- 1 -oxo-2-buten- I -yl]amino 1-7 25 [(S)-(tetrahydrofuran-3-yl)oxy]-quinazoline , or a pharmaceutically acceptable salt thereof, a protein kinase receptor antagonist which is not classified under the synthetic small molecules such as atrasentan, rituximab, cetuximab, AvastinTM (bevacizumab), IMC-1C 11, erbitux (C-225), DC-101, EMD-72000, vitaxin, imatinib, a protein tyrosine kinase inhibitor which is a fusion protein such as VEGFtrap, an alkylating agent or a 30 platinum compound such as melphalan, cyclophosphamide, an oxazaphosphorine, cisplatin, carboplatin, oxaliplatin, satraplatin, tetraplatin, iproplatin, mitomycin, streptozocin, carmustine (BCNU), lomustine (CCNU), busulfan, ifosfamide, WO 2006/018182 PC'/EP2005/008623 14 streptozocin, thiotepa, chlorambucil, a nitrogen mustard such as mechlorethamine, an ethyleneimine compound, an alkylsulphonate, daunorubicin, doxorubicin (adriamycin), liposomal doxorubicin (doxil), epirubicin, idarubicin, nitoxantrone, amsacrine, dactinomycin, distamycin or a derivative thereof, netropsin, pibenzimol, mitomycin, s CC-1065, a duocarmycin, mithramycin, chromomycin, olivomycin, a phtalanilide such as propamidine or stilbamidine, an anthramycin, an aziridine, a nitrosourea or a derivative thereof, a pyrimidine or purine analogue or antagonist or an inhibitor of the nucleoside diphosphateTeductase such as -cytarabine, 5-fluorouracile (5-FU), pemetrexed, tegafur/uracil, uracil mustard, fludarabine, gemcitabine, capecitabine, 10 mercaptopurine, cladribine, thioguanine, methotrexate, pentostatin, hydroxyurea, or folic acid, a phleomycin, a bleomycin or a derivative or salt thereof, CHPP, BZPP, MTPP, BAPP, liblomycin, an acridine or a derivative thereof, a rifamycin, an actinomycin, adramycin, a camptothecin such as irinotecan (camptosar) or topotecan, an amsacrine or analogue thereof, a tricyclic carboxamide, an histonedeacetylase inhibitor is such as SAHA, MD-275, trichostatin A, CBHA, LAQ824, or valproic acid, an anti cancer drug from plants such as paclitaxel (taxol), docetaxel or taxotere, a vinca alkaloid such as navelbine, vinblastin, vincristin, vindesine or vinorelbine, a tropolone alkaloid such as colchicine or a derivative thereof, a macrolide such as maytansine, an ansamitocin or rhizoxin, an antimitotic peptide such as phomopsin or dolastatin, an 20 epipodophyllotoxin or a derivative of podophyllotoxin such as etoposide or teniposide, a steganacin, an antimitotic carbamate derivative such as combretastatin or amphetinile, procarbazine, a proteasome inhibitor such as bortezomib, an enzyme such as asparaginase, pegylated asparaginase (pegaspargase) or a thymidine-phosphorylase inhibitor, a gestagen or an estrogen such as estramustine (T-66) or megestrol, an anti 25 androgen such as flutamide, casodex, anandron or cyproterone acetate, an aromatase inhibitor such as aminogluthetimide, anastrozole, formestan or letrozole, a GNrH analogue such as leuprorelin, buserelin, goserelin or triptorelin, an anti-estrogen such as tamoxifen or its citrate salt, droloxifene, trioxifene, raloxifene or zindoxifene, a derivative of 17$-estradiol such as ICI 164,384 or ICI 182,780, aminoglutethimide, 30 formestane, fadrozole, finasteride, ketoconazole, a LH-RH antagonist such as leuprolide, a steroid such as prednisone, prednisolone, methylprednisolone, dexamethasone, budenoside, fluocortolone or triamcinolone, an interferon such as WO 2006/018182 PCT/EP2005/008623 15 interferon @, an interleukin such as IL-10 or IL-12, an anti-TNFax antibody such as etanercept, an immunomodulatory drug such as thalidomide, its R- and S-enantiomers and its derivatives, or revimid (CC-5013), a leukotrien antagonist, mitomycin C, an aziridoquinone such as BMY-42355, AZQ or EO-9, a 2-nitroimidazole such as 5 misonidazole, NLP-1 or NLA-1, a nitroacridine, a nitroquinoline, a nitropyrazoloacridine, a "dual-function" nitro aromatic such as RSU-1069 or RB-6145, CB-1954, a N-oxide of nitrogen mustard such as nitromin, a metal complex of a nitrogen mustard, an anti-CD3 or-anti-CD25 antibody, a tolerance induction agent, a biphosphonate or derivative thereof such as minodronic acid or its derivatives (YM-529, 10 Ono-5920, YH-529), zoledronic acid monohydrate, ibandronate sodium hydrate or clodronate disodium, a nitroimidazole such as metronidazole, misonidazole, benznidazole or nimorazole, a nitroaryl compound such as RSU-1069, a nitroxyl or N oxide such as SR-4233, an halogenated pyrimidine analogue such as bromodeoxyuridine, iododeoxyuridine, a thiophosphate such as WR-2721, a photo is chemically activated drug such as porfimer, photofrin, a benzoporphyrin derivative, a pheophorbide derivative, merocyanin 540 (MC-540) or tin etioporpurin, an ant-template or an anti-sense RNA or DNA such as oblimersen, a non-steroidal inflammatory drug such as acetylsalicyclic acid, mesalazin, ibuprofen, naproxen, flurbiprofen, fenoprofen, fenbufen, ketoprofen, indoprofen, pirprofen, carprofen, oxaprozin, pranoprofen, 20 miroprofen, tioxaprofen, suprofen, alminoprofen, tiaprofenic acid, fluprofen, indomethacin, sulindac, tolmetin, zomepirac, nabumetone, diclofenac, fenclofenac, alclofenac, bromfenac, ibufenac, aceclofenac, acemetacin, fentiazac, clidanac, etodolac, oxpinac, mefenamic acid, meclofenamic acid, flufenanic acid, nifluminic acid, tolfenamic acid, diflunisal, flufenisal, piroxicam, tenoxicam, lomoxicam, nimesulide, 25 meloxicam, celecoxib, rofecoxib, or a pharmaceutically acceptable salt of a non steroidal inflammatory drug, a cytotoxic antibiotic, an antibody targeting the surface molecules of cancer cells such as apolizumab or 1D09C3, an inhibitor of metalloproteinases such as TIMP-1 or TLIP-2, Zinc, an inhibitor of oncogenes such as P53 and Rb, a complex of rare earth elements such as the heterocyclic complexes of 30 lanthanides, a photo-chemotherapeutic agent such as PUVA, an inhibitor of the transcription factor complex ESX/DRIP130/Sur-2, an inhibitor of HER-2 expression, such as the heat shock protein HSP90 modulator geldanamycin and its derivative 17- WO 2006/018182 PCT/EP2005/008623 16 allylaminogeldanamycin or 17-AAG, or a therapeutic agent selected from IM-842, tetrathiornolybdate, squalamine, combrestatin A4, TNP-470, marimastat, neovastat, bicalutarnide, abarelix, oregovomab, mitumomab, TLK-286, alemtuzumab, ibritumomab, temozolomide, denileukin diftitox, aldesleukin, dacarbazine, floxuridine, 5 plicamycin, mitotane, pipobroman, plicamycin, tamoxifen and testolactone. Preferred compounds include small molecule VEGF receptor antagonist such as vatalanib (PTK-787/ZK222584), SU-5416, SU-6668, SU-11248, SU-14813, AZD 6474, EGFRHER2 antagonists such as CI-1033 or GW-2016, an EGFR antagonist such as iressa (gefitinib, ZD-1839), tarceva (erlotinib, OSI-774), PKI-166, EKB-569, HKI 10 272 or herceptin, an antagonist of the mitogen-activated protein kinase such as BAY 43-9006 or BAY-57-9006, atrasentan, rituximab, cetuximab, AvastinTm (bevacizumab), IMC-1Cl1, erbitux (C-225), DC-101, EMD-72000, vitaxin, imatinib, an alkylating agent or a platinum compound such as melphalan, cyclophosphamide, cisplatin, carboplatin, oxaliplatin, satraplatin, daunorubicin, doxorubicin (adriamycin), liposomal is doxorubicin (doxil), cpirubicin, idarubicin, a pyrimidine or purine analogue or antagonist or an inhibitor of the nucleoside diphosphate reductase such as cytarabine, 5 fluorouracile (5-FU), pemetrexed, tegafur/uracil, gemcitabine, capecitabine, mercaptopurine, methotrexate, an anti-cancer drug such as paclitaxel (taxol) or docetaxel, a vinca alkaloid such as navelbine, vinblastin, vincristin, vindesine or 20 vinorelbine, an antimitotic peptide such as dolastatin, an epipodophyllotoxin or a derivative of podophyllotoxin such as etoposide or teniposide, a non-steroidal inflammatory drug such as meloxicam, celecoxib, rofecoxib, an antibody targeting the surface molecules of cancer cells such as apolizumab or 1D09C3 or the heat shock protein HSP90 modulator geldanamycin and its derivative 17 -allylaminogeldanamycin 25 or 17-AAG. In another preferred embodiment the instant invention is directed to a pharmaceutical composition, wherein the further chemotherapeutic or naturally occurring, semi synthetic or synthetic therapeutic agent 2 is selected from the group consisting of an 30 anti-cancer drug from plants such as paclitaxel (taxol), docetaxel, a vinca alkaloid such as navelbine, vinblastin, vincristin, vindesine or vinorelbine, an alkylating agent or a platinum compound such as melphalan, cyclophosphamide, an oxazaphosphorine, WO 2006/018182 PCT/EP2005/008623 17 cisplatin, carboplatin, oxaliplatin, satraplatin. tetraplatin, iproplatin, mitomycin, streptozocin, carmustine (BCNU), lomustine (CCNU), busulfan, ifosfamide, streptozocin, thiotepa, chlorambucil, a nitrogen mustard such as mechloretharnine, an immunomodulatory drug such as thalidomide, its R- and S-enantiomers and its 5 derivatives, or revimid (CC-5013)), an ethyleneimine compound, an alkylsulphonate, daunorubicin, doxorubicin (adriamycin), liposomal doxorubicin (doxil), epirubicin, idarubicin, mitoxantrone, amsacrine, dactinomycin, distamycin or a derivative thereof, netropsin, pibenzimol, mitomycin, CC-1065, a duocarmycin, mithramycin, chromomycin, olivomycin, a phtalanilide such as propamidine or stilbamidine, an 10 anthramycin, an aziridine, a nitrosourea or a derivative thereof, a pyrimidine or purine analogue or antagonist or an inhibitor of the nucleoside diphosphate reductase such as cytarabine, 5-fluorouracile (5-FU), uracil mustard, fludarabine, gemcitabine, capecitabine, mercaptopurine, cladribine, thioguanine, methotrexate, pentostatin, hydroxyurea, or folic acid, an acridine or a derivative thereof, a rifamycin, an is actinomycin, adramycin, a camptothecin such as irinotecan (camptosar) or topotecan, an amsacrine or analogue thereof, a tricyclic carboxamide, an histonedeacetylase inhibitor such as SAHA, MD-275, trichostatin A, CBHA, LAQ824, or valproic acid, a proteasome inhibitor such as bortezomib, a small molecule VEGF receptor antagonist such as vatalanib (PTK-787/ZK222584), SU-5416, SU-6668, SU-11248, SU-14813, 20 AZD-6474, AZD-2171, CP-547632, CEP-7055, AG-013736, IM-842 or GW-786034, an antagonist of the mitogen-activated protein kinase such as BAY-43-9006 or BAY 57-9006, a dual EGFR/HER2 antagonist such as gefitinib, erlotinib, CI-1033 or GW 2016, an EGFR antagonist such as iressa (ZD-1839), tarceva (OSI-774), PKI-166, EKB 569, HKI-272 or herceptin, a quinazoline derivative such as 4-[(3-chloro-4 25 fluorophenyl)amino]-6-{ [4-(N,N-dimethylamino)-1-oxo-2-buten-1-ylJamino}-7-((S) tetrahydrofuran-3-yloxy)-quinazoline or 4-[(3-chloro-4-fluoro-phenyl)amino]-6- {[4 (homomorpholin-4-yl)-1-oxo-2-buten-1-yl]amino}-7-[(S)-(tetrahydrofuran-3-yl)oxy] quinazoline , or a pharmaceutically acceptable salt thereof, an inhibitor of the transcription factor complex ESX/DRIP130/Sur-2, an inhibitor of HER-2 expression, 30 such as the heat shock protein HSP90 modulator geldanamycin and its derivative 17 allylaminogeldanamycin or 17-AAG, a protein kinase receptor antagonist which is not classified under the synthetic small molecules such as atrasentan, rituximab, cetuximab, WO 2006/018182 PCT/EP2005/008623 18 AvastinTM (bevacizumab), IMC-1C11, erbitux (C-225), DC-101, EMD-72000, vitaxin, imatinib, and an antibody targeting the surface molecules of cancer cells such as apolizumab or 1D09C3. 5 Preferred compounds include small molecule receptor antagonists such aus vatalanib, SU 11248 or AZD-6474, EGFR or HER2 antagonists such as gefitinib, erlotinib, CI 1033 or Herceptin, antibodies such as bevacizumab, cetuximab, rituximab, DNA alkylating drugs such as cisplatin, oxaliplatin or carboplatin, anthracyclines such as doxorubicin or epirubicin, an antimetabolite such as 5-FU, pemetrexed, gemcitabine or 10 capecitabine, a camptothecin such as irinotecan or topotecan, an anti-cancer drug such as paclitaxel or docetaxel, an epipodophyllotoxin such as etoposide or teniposide, a proteasome inhibitor such as bortezomib or antiinflammatory drugs such as celecoxib or rofecoxib., optionally in form of the pharmaceutically acceptable salts, in form of the hydrates and/or solvates and optionally in the form of the individual optical isomers, is mixtures of the individual enantiomers or racemates thereof. In another preferred embodiment the instant invention is directed to a pharmaceutical composition as defined hereinbefore, wherein the further chemotherapeutic or naturally occurring, semi-synthetic or synthetic therapeutic agent 2 is the quinazoline derivative 20 4-[(3-chloro-4-fluorophenyl)amino]-6-{[4-(N,N-dimethylamino)-1-oxo-2-buten-1 yl]amino}-7-((S)-tetrahydrofuran-3-yloxy)-quinazoline or a pharmaceutically acceptable salt thereof In another preferred embodiment the instant invention is directed to a pharmaceutical 25 composition as defined hereinbefore, wherein the further chemotherapeutic or naturally occurring, semi-synthetic or synthetic therapeutic agent 2 is the di-maleic acid salt of the compound 4-[(3-chloro-4-fluorophenyl)amino]-6-{[4-(N,N-dimethylamino)-1-oxo 2-buten-1-yl]amino }-7-((S)-tetrahydrofuran-3-yloxy)-quinazoline, or 4-[(3-chloro-4 fluoro-phenyl)amino]-6-{[4-(homomorpholin-4-yl)-1-oxo-2-buten-1-yl]amino}-7-[(S) 30 (tetrahydrofuran-3-yl)oxy]-quinazoline, or the tautomers, stereoisomers or a pharmaceutically acceptable salt thereof.
WO 2006/018182 PCT/EP2005/008623 19 In another preferred embodiment the instant invention is directed to a pharmaceutical composition as defined hereinbefore, wherein the further chemotherapeutic or naturally occurring, semi-synthetic or synthetic therapeutic agent 2 is the 4-[(3-chloro-4-fluoro phenyl)amino]-6-{ [4-(homomorpholin-4-yl)- 1-oxo-2-buten- 1 -yl]amino)-7-[(S) s (tetrahydrofuran-3-yl)oxy)-quinazoline , or a pharmaceutically acceptable salt thereof. In another preferred embodiment the instant invention is directed to a pharmaceutical composition as defined hereinbefore, wherein the further chemotherapeutic or naturally occurring, semi-synthetic or synthetic therapeutic agent 2 is the 3-Z-[1-(4-(N-((4 10 methyl-piperazin-1-yl)-methylcarbonyl)-N-methyl-amino)-anilino)-1-phenyl methylene]-6-methoxycarbonyl-2-indolinone, or a polymorph, metabolite or pharmaceutically acceptable salt thereof. In another preferred embodiment the instant invention is directed to a pharmaceutical 15 composition as defined hereinbefore, wherein the further chemotherapeutic or naturally occurring, semi-synthetic or synthetic therapeutic agent 2 is the monoethanesulfonate salt of 3-Z-[1-(4-(N-((4-methyl-piperazin-1-yl)-methylcarbonyl)-N-methyl-amino) anilino)-1-phenyl-methylenel-6-methoxycarbonyl-2-indolinone. 20 In another preferred embodiment the instant invention is directed to a pharmaceutical composition as defined hereinbefore, wherein the further chemotherapeutic or naturally occurring, semi-synthetic or synthetic therapeutic agent 2 is the 3-Z-[1-(4 dimethylaminomethylanilino)- 1-(4-(2-carboxyethyl)phenyl)methylene]-6-fluoro-2 indolinone, or a polymorph, metabolite or pharmaceutically acceptable salt thereof. 25 In another preferred embodiment the instant invention is directed to a pharmaceutical composition, wherein the further chemotherapeutic or naturally occurring, semi synthetic or synthetic therapeutic agent 2 is irinotecan, topotecan, oxaliplatin, docetaxel, paclitaxel, gemcitabine, pemetrexed, cisplatin, carboplatin, bevacizumab, cetuximab, 30 gefitinib or erlotinib, particularly preferred irinotecan, docetaxel, gemcitabine, topotecan or paclitaxel.
WO 2006/018182 PCT/EP2005/008623 20 In another preferred embodiment the instant invention is directed to a pharmaceutical composition as defined hereinbefore, wherein the further naturally occurring, semi synthetic or synthetic therapeutic agent 2 is a compound which reduces the transport of hyaluronan mediated by one or more ABC transporters, or drug transport inhibitor, such s as a P-glycoprotein (P-gp) inhibitor molecule or inhibitor peptide, an MRP1 inhibitor, an antibody directed against and capable of blocking the ABC transporter, an antisense oligomer, iRNA, siRNA or aptamer directed against one or more ABC transporters. Examples of P-glycoprotein (P-gp) inhibitor molecules in accordance with the-present invention are zosuquidar (LY 335973), its salts (especially the trichloride salt) and its 10 polymorphs, cyclosporin A (also known as cyclosporine), verapamil or its R-isomer, tamoxifen, quinidine, d-alpha tocopheryl polyethylene glycol 1000 succinate, VX-710, PSC833, phenothiazine, GF120918 (II), SDZ PSC 833, TMBY, MS-073, S-9788, SDZ 280-446, XR(9051) and functional derivatives, analogues and isomers of these. 15 Furthermore, where the compounds 2 carries an acidic moiety, suitable pharmaceutically acceptable salts thereof may include alkali metal salts (e.g. sodium or potassium salts), alkaline earth metal salts (e. g. calcium or magnesium salts) and salts formed with suitable organic ligands (e.g. quaternary ammonium salts). 20 The compounds 2 may have chiral centers and may occur as racemates, racernic mixtures and as individual diastereomers, or enantiomers with all isomeric forms being included in the present invention. Hence, where a compound is chiral, the separate enantiomers, substantially free of the others, are included within the scope of the invention. Further included are all mixtures of the two enantiomers. Also included 25 within the scope of the invention are polymorphs and hydrates of the compounds of the instant invention. The present invention includes within its scope prodrugs of a compound .. of Formula (I) and of the further active ingredient 2. In general, such prodrugs will be functional 30 derivatives of the compounds or active ingredients of this invention which are readily convertible in vivo into the required compound.
WO 2006/018182 PCT/EP2005/008623 21 In a further embodiment the invention relates to a composition as defined hereinbefore, which inhibits the proliferation of various human tumour cell lines including but not limited to Saos-2, H4, MDA-MB-435S, MVDA-MB453, MCF7, HeLa S3, HCT116, Colo 205, HT29, FaDu, HL-60, K-562, THP-1, HepG2, A549, NCI-H460, GRANTA 5 519, Raji, Ramos, BRO, SKOV-3, BxPC-3, Mia CaPa-2, DU145, PC-3, NCI-N87, MES-SA, SK-UT-1B and A431. Another embodiment of the invention relates to the use of a pharmaceutical composition as defined hereinbefore for the preparation of a medicament for the treatment of 10 oncological diseases, such as malignant human neoplasias. In a preferred embodiment the instant invention relates to the use of a pharmaceutical composition as defined hereinbefore, wherein the oncological disease is selected from the group consisting of solid tumours. 15 In a further preferred embodiment the invention relates to the use of a pharmaceutical composition as defined hereinbefore, wherein the oncological disease is selected from the group consisting of urogenital cancers (such as prostate cancer, renal cell cancers, bladder cancers), gynecological cancers (such as ovarian cancers, cervical cancers, 20 endormetrial cancers), lung cancer, gastrointestinal cancers (such as colorectal cancers, pancreatic cancer, gastric cancer, esophageal cancers, hepatocellular cancers, cholangiocellular cancers), head and neck cancer, malignant mesothelioma, breast cancer, malignant melanoma or bone and soft tissue sarcomas. 25 In a further preferred embodiment the invention relates to the use of a pharmaceutical composition as defined hereinbefore wherein the oncological disease is selected from the group consisting of refractory or relapsed multiple myeloma, acute or chronic myelogenous leukaemia, myelodysplastic syndrome,acute lymphoblastic leukaemia, Hodgkin's or non-Hodgkin's lymphoma. 30 In a further preferred embodiment, the disease is hormone sensitive or hormone refractory prostate cancer, ovarian carcinoma, or small cell lung cancer.
WO 2006/018182 PCT/EP2005/008623 22 In a further preferred embodiment the invention relates to the use of a composition as defined hereinbefore, wherein the oncological disease is characterized by inappropriate cellular proliferation, migration, apoptosis or angiogenesis, preferably by inappropriate 5 cellular proliferation. Inappropriate cell proliferation means cellular proliferation resulting from inappropriate cell growth, from excessive cell division, from cell division at an accelerated rate and/or from inappropriate cell survival. In a further preferred embodiment the invention relates to the use according to the io invention, wherein the disease is cancer selected from the group consisting of carcinomas, sarcomas, melanomas, myelomas, hematological neoplasias, lymphomas and childhood cancers. Examples of carcinomas within the scope of the invention include but are not limited to is adenocarcinoma (AC), squamous cell carcinoma (SCC) and mixed or undifferentiated carcinomas. Carcinomas within the scope of the invention include but are not limited to the following histologies: " Head and neck tumours: SCC, AC, transitional cell cancers, mucoepidermoid cancers, undifferentiated carcinomas; 20 0 Central nervous system tumours: Astrocytoma, glioblastoma, meningeoma, neurinoma, schwannoma, ependymoma, hypophysoma, oligodendroglioma, medulloblastoma; " Bronchial and mediastinal tumours: o Bronchial tumours: 25 - Small cell lung cancers (SCLC): oat-cell lung cancer, intermediate cell cancer, combined oat-cell lung cancer; - Non-small cell lung cancers (NSCLC): SCC, spindle cell carcinoma, AC, bronchioalveolar carcinoma, large cell NSCLC, clear cell NSCLC; o Mesothelioma; 30 o Thymoma; o Thyroid carcinomas: papillary, follicular, anaplastic, medullary; e Tumours of the gastrointestinal tract: WO 2006/018182 PC'I'/EP2005/008623 23 o Esophageal cancers: SCC, AC, anaplastic, carcinoid, sarcoma; o Gastric cancers: AC, adenosquamous, anaplastic; o Colorectal cancers: AC, including hereditary forms of AC, carcinoid, sarcoma; 5 o Anal cancers: SCC, transitional epithelial cancer, AC, basal cell carcinoma; o Pancreatic cancers: AC, including ductal and acinary cancers, papillary, adenosquamous, undifferentiated, tumours of the endocrine pancreas; o -Hepatocellular carcinoma, cholangiocarcinoma, angiosarcoma, hepatoblastoma; 10 o Biliary carcinomas: AC, SCC, small cell, undifferentiated; o Gastrointestinal stroma tumours (GIST); 0 Gynaecological cancers: o Breast cancers: AC, including invasive ductal, lobular and medullary cancers, tubular, mucinous cancers, Paget-carcinoma, inflammatory i5 carcinoma, ductal and lobular carcinoma in situ; o Ovarian cancers: Epithelial tumours, stroma tumours, germ cell tumours, undifferentiated tumours; o Cervical cancers: SCC, AC, mixed and undifferentiated tumours; o Endometrial cancers: AC, SCC, mixed, undifferentiated tumours; 20 o Vulvar cancers: SCC, AC; o Vaginal cancers: SCC, AC; e Urinary tract and testicular cancers: o Testicular cancers: seminoma; o Non-seminomatous germ cell tumours: teratoma, embryonal cell carcinoma, 25 choriocarcinoma, yolk sac tumour, mixed, Sertoli and Leydig-cell tumours; o Extragonadal germ cell tumours; o Prostate cancers: AC, small cell, SCC; o Renal cell cancers: AC, including clear cell, papillary and chromophobous carcinomas, hereditary forms (e.g. von-Hippel-Lindau syndrome), 30 nephroblastoma; o Urinary bladder cancers: transitional cell (urothelial) cancers, SCC, AC; o Urethral cancers: SCC, transitional cell cancers, AC; WO 2006/018182 PCT/EP2005/008623 24 o Penile cancers: SCC; o Tumours of endocrine tissue: o Thyroid cancers: papillary, follicular, anaplastic, medullary carcinomas, including MEN syndrome; 5 o Tumours of the endocrine pancreas; o Carcinoids; o Adrenal tumours, e.g. Pheochromocytoma. Examples of sarcomas within the scope of the invention include but are not limited to 10 Ewing-sarcoma, osteosarcoma or osteogenic sarcoma, chondrosarcoma, synovial sarcoma, leiomyosarcoma, rhabdomyosarcoma, mesothelial sarcoma or mesothelioma, fibrosarcoma, angiosarcoma or hemangioendothelioma, liposarcoma, glioma or astrocytoma, myxosarcoma, malignant fibrous histiocytoma, mesenchymous or mixed mesodermal tumour, neuroblastoma and clear cell sarcoma. 15 Examples of skin tumors within the scope of the invention include but are not limited to basal cell carcinoma, Merkel cell carcinoma, sebaceous carcinoma, fibroxanthoma, malignant fibrous histiocytoma, and skin sarcoma. 20 Examples of melanomas within the scope of the invention include but are not limited to superficial spreading melanoma, nodular and lentigo-maligna melanoma. Examples of myelomas within the scope of the invention include but are not limited to immunocytona, plasmocytoma and multiple myeloma. 25 Examples of childhood cancers within the scope of the invention include but are not limited to Wilms' tumor, neuroblastoma, retinoblastoma, rhabdomyosarcoma, Ewing's sarcoma and peripheral primitive neuroectodermal tumors, germ cell tumors and childhood lymphoma and leukemias. 30 In another preferred embodiment the invention relates to the use of a composition as defined hereinbefore, wherein the hematologic cancer is leukemia.
WO 2006/018182 PCT/EP2005/008623 25 Further examples of hematologic neoplasias within the scope of the invention include but are not limited to acute or chronic leukemias of myeloid, erythroid or lymphatic origin, myelodysplastic syndromes (MDS) and myeloproliferative syndromes (MPS, 5 such as chronic myelogeneous leukemia, osteomyelofibrosis, polycythemia vera or essential thrombocythemia). Examples of lymphomas within the scope of the invention include but are not limited to: 10 o Hodgkin-lymphoma; o Non-Hodgkin-lymphomas: T- and B-cell lymphomas o B-cell lymphomas: Low and intermediate grade: Chronic lymphocytic leukemia (CLL), prolymphocytic leukemia (PLL), small lymphocytic lymphoma, hairy is cell leukemia, plasmacytoid lymphoma, mantle cell lymphoma, follicular lymphoma, marginal zone lymphoma including MALT-lymphoma; = High grade: diffuse large B-cell lymphoma (DLBCL including immunoblastic and centroblastic variants), lymphoblastic, Burkitt's lymphoma; 20 o T-cell lymphomas: - Low grade: T-CLL, T-PLL, Mycosis fungoides, Sezary-syndrome; " High grade: Anaplastic large cell, T-immunoblastic and lymphoblastic. In another preferred embodiment the invention relates to the use according to the 25 invention, wherein the disease is cancer selected from the group consisting of mixed tumours, undifferentiated tumours and metastases thereof. Examples of mixed tumours within the scope of the invention include but are not limited to adenosquamous carcinomas, mixed mesodermal tumours, carcinosarcomas 30 and teratocarcinomas.
WO 2006/018182 PCT/EP2005/008623 26 Examples of undifferentiated, other tumours or metastases thereof within the scope of the invention include but are not limited to undifferentiated tumours, carcinomas of unknown primary (CUP), metastases of unknown primary (MUP) and pheochromocytoma, carcinoids. s In a further embodiment the invention relates to the use of a composition as defined hereinbefore, for the preparation of a medicament for the treatment of autoimmune disorders selected from the group consisting of amyloidosis, systemic lupus erythematosus, rheumatoid arthritis, Crohn's disease, multiple sclerosis, systemic 1o sclerosis (scleroderma), mixed connective tissue disease, Sjugren's syndrome, ankylosing spondylitis, autoimmune vasculitis, Behcet's syndrome, psoriasis, autoimmune arthritis, sarcoidosis and diabetes mellitus. In a further embodiment the invention relates to the use of a pharmaceutical is composition as defined hereinbefore for the preparation of a medicament for the treatment of further non-oncological diseases, such as diabetic retinopathy and rheumatoid arthritis. In a further embodiment the invention relates to the use of a composition as defined 20 hereinbefore wherein the composition according to the invention is administered orally, enterically, transdermally, intravenously, peritoneally or by injection, preferably intravenously. In a further embodiment the invention relates to a pharmaceutical combination 25 preparation kit for the treatment of diseases involving cell proliferation, migration or apoptosis of myeloma cells, or angiogenesis, comprising a therapeutically effective amount of a compound I of Formula (I) in accordance with the present invention, or a polymorph, hydrate, metabolite or pharmaceutically acceptable salt thereof, and at least a further chemotherapeutic or naturally occurring, semi-synthetic or synthetic 30 therapeutic agent 2, and optionally adapted for a co-treatment with radiotherapy or radio-immunotherapy, characterised in that the compound 1 of Formula (I) is comprised within a first compartment and the further chemotherapeutic or naturally WO 2006/018182 PCT/EP2005/008623 27 occurring, semi-synthetic or synthetic therapeutic agent 2 is comprised within a second compartment, such that the administration to a patient in need thereof can be simultaneous, separate or sequential. 5 In a preferred embodiment the invention relates to a pharmaceutical combination preparation kit, wherein the formulation of the compound i of Formula (I) in accordance with the present invention is for oral administration or injection. In a further embodiment the invention relates to the use of a pharmaceutical 10 combination or a pharmaceutical combination preparation kit, for the manufacture of a medicament, optionally adapted for a co-treatment with radiotherapy or radio immunotherapy, to treat diseases involving cell proliferation, migration or apoptosis of cancer cells, or angiogenesis, in a human or non-human.mammalian body. is In a further embodiment the invention relates to the use of an effective amount of a compound 1 of Formula (I) or a polymorph, hydrate, metabolite or pharmaceutically acceptable salt thereof, in combination with at least a further chemotherapeutic or naturally occurring, semi-synthetic or synthetic therapeutic agent 2, for the manufacture of a pharmaceutical combination preparation, optionally adapted for a co-treatment with 20 radiotherapy or radio-immunotherapy, for simultaneous, separate or sequential use in the treatment of diseases involving cell proliferation, migration or apoptosis of cancer cells, or angiogenesis, in a human or non-human mammalian body. In a further embodiment the invention relates to a method for the treatment of diseases 2s involving cell proliferation, migration or apoptosis of cancer cells, or angiogenesis, which method comprises simultaneous, separate or sequential co-administration of effective amounts of: (i) a compound 1 of Formula (I) or a polymorph, metabolite, hydrate, solvate, an individual optical isomer, mixtures of the individual 30 enantiomers or racemates thereof, or a pharmaceutically acceptable salt thereof; and WO 2006/018182 PC'I/EP2005/008623 28 (ii) at least a further chemotherapeutic or naturally occurring, semi synthetic or synthetic therapeutic agent 2; in the form of a combined preparation optionally adapted for a co-treatment with s radiotherapy or radio-immunotherapy, to a person in need of such treatment. In a further embodiment the invention relates to the uses described above, characterised in that a compound 1 of Formula (I), or its polymorph, metabolite, hydrate, solvate, an individual optical isomer, mixtures of the individual enantiomers or racemates thereof, 1o or a pharmaceutically acceptable salt thereof, is administered intermittent or in a daily dosage such that the plasma level of the active substance lies between 10 and 5000 nM for at least 12 hours of the dosing interval. The term "therapeutically effective amount" shall mean that amount of a drug or is pharmaceutical agent that will elicit the biological or medical response of a tissue, system, animal or human that is being sought by a researcher or clinician. As used herein, the term "composition" is intended to encompass a product comprising the specified ingredients in the specified amounts, as well as any product which results, 20 directly or indirectly, from a combination of the specified ingredients in the specified amounts. As already mentioned before, within the meaning of the present invention, the components 1 and 2 of the composition for a combination therapy may be administered 25 separately (which implies that they are formulated separately) or together (which implies that they are formulated together). Hence, the administration of one element of the combination of the present invention may be prior to, concurrent to, or subsequent to the administration of the other element of the combination. 30 In accordance with the present invention, the elements of the combination of 1 and 2 may be administered by oral (including buccal or sublingual), enterical, parenteral (e.g., intramuscular, intraperitoneal, intravenous, transdermal or subcutaneous injection, or WO 2006/018182 PCT/EP2005/008623 29 implant), nasal, vaginal, rectal, or topical (e.g. ocular eyedrops) routes of administratic.. and may be formulated, alone or together, in suitable dosage unit formulations containing conventional non-toxic pharmaceutically acceptable carriers, adjuvants and vehicles appropriate for each route of administration. 5 In a preferred embodiment the element I of the combination in accordance with the invention is administered orally, enterically, transdermally, intravenously, peritoneally or by injection, preferably intravenously. 10 The pharmaceutical compositions for the administration of the components I and 2 of this invention may conveniently be presented in dosage unit form and may be prepared by any of the methods well known in the art of pharmacy. All methods include the step of bringing the active ingredient into association with the carrier which is constituted of one or more accessory ingredients. In general, the 15 pharmaceutical compositions are prepared by uniformly and intimately bringing the active ingredients into association with a liquid carrier or a finely divided solid carrier or both, and then, if necessary, shaping the product into the desired dosage form. In the pharmaceutical compositions the active compounds are included in an amount sufficient to produce the desired pharmacologic effect. 20 The pharmaceutical compositions containing the active ingredients 1 and 2, separately or together, that are suitable for oral administration may be in the form of discrete units such as hard or soft capsules, tablets, troches or lozenges, each containing a predetermined amount of the active ingredients, or in the form of a dispersible powder 25 or granules, or in the form of a solution or a suspension in an aqueous liquid or non aqueous liquid, or in the form of syrups or elixirs, or in the form of an oil-in-water emulsion or a water-in-oil emulsion. Dosage forms intended for oral use may be prepared according to any method known to 30 the art for the manufacture of pharmaceutical formulations and such compositions. The excipients used may be, for example: (a) inert diluents such as mannitol, sorbitol, WO 2006/018182 PCT/EP2005/008623 30 calcium carbonate, pregelatinized starch, lactose, calcium phosphate or sodium phosphate; (b) granulating and disintegrating agents, such as povidone, copovidone, hydroxypropylmethylcellulose, corn starch, alginic acid, crospovidone, sodiumstarchglycolate, croscarmellose, or polacrilin potassium ; (c) binding agents such 5 as microcrystalline cellulose or acacia ; and (d) lubricating agents such as magnesium stearate, stearic acid, fumaric acid or talc. In some cases, formulations for oral use may be in the form of hard gelatin or HPMC (hydroxypropylmethylcellulose) capsules wherein the active ingredients I or 2, 10 separately or together, is mixed with an inert solid diluent, for example pregelatinized starch, calcium carbonate, calcium phosphate or kaolin, or dispensed via a pellet formulation. They may also be in the form of soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin, medium chain triglycerides or olive oil. 15 The tablets, capsules or pellets may be uncoated or they may be coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a delayed action or sustained action over a longer period. For example, a time delay material such as celluloseacetate phtalate or hydroxypropylcellulose acetate 20 succinate or sustained release material such as ethylcellulose or ammoniomethacrylate copolymer (type B) may be employed. Liquid dosage forms for oral administration in accordance with the present invention include pharmaceutically acceptable emulsions, solutions, suspensions, syrups, and 25 elixirs containing inert diluents commonly used in the art, such as water. Besides such inert diluents, compositions can also include adjuvants, such as wetting agents, emulsifying and suspending agents, and sweetening, flavoring, perfuming and preserving agents. 30 Aqueous suspensions in accordance with the present invention normally contain the active materials 1 and 2, separately or together, in admixture with excipients suitable for the manufacture of aqueous suspensions. Such excipients may be (a) suspending agents WO 2006/018182 PCT/EP2005/008623 31 such as hydroxy ethylcellulose, sodium carboxymethylcellulose, methylcellulose, hydroxypropylmethylcellulose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia; (b) dispersing or wetting agents which may be (b. 1) a naturally occurring phosphatide such as lecithin, (b.2) a condensation product of an alkylene 5 oxide with a fatty acid, for example, polyoxyethylene stearate, (b.3) a condensation product of ethylene oxide with a long chain aliphatic alcohol, for example heptadecaethyleneoxycetanol, (b.4) a condensation product of ethylene oxide with a partial ester derived from a fatty acid and a hexitol such as polyoxyethylene sorbitol monooleate, or (b.5) a condensation product of ethylene oxide with a partial ester 10 derived from a fatty acid and a hexitol anhydride, for example polyoxyethylene sorbitan monooleate. The aqueous suspensions may also contain: one or more preservatives, for example, ethyl or n-propyl p-hydroxybenzoate; one or more coloring agents; one or more is flavoring agents; and one or more sweetening agents, such as sucrose or saccharin. Oily suspensions in accordance with the present invention may be formulated by suspending the active ingredients 1 and 2, separately or together, in a vegetable oil, for 20 example arachis (peanut) oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin. The oily suspensions may contain a thickening agent, for example beeswax, hard paraffin or cetyl alcohol. Sweetening agents and flavoring agents may be added to provide a palatable oral preparation. These compositions may be prepared by the addition of an antioxidant such as ascorbic acid. 25 Dispersible powders and granules are suitable formulations for the preparation of an aqueous suspension in accordance with the present invention. In these formulations the active ingredients 1 and 2 are present, separately or together, in admixture with a dispersing or wetting agent, a suspending agent and one or more preservatives. Suitable 30 examples of dispersing or wetting agents, suspending agents and preservatives are those already mentioned hereinbefore. Additional excipients such as, for example, sweetening, flavouring and colouring agents may also be present. Suitable examples of WO 2006/018182 PCTIEP2005/008623 32 excipients are those already mentioned hereinbefore. The pharmaceutical compositions of the invention may also be in the form of oil-in water emulsions. The oily phase may be a vegetable oil such as olive oil or arachis s (peanut) oil, or a mineral oil such as liquid paraffin or a mixture thereof. Suitable emulsifying agents may be (a) naturally-occurring gums such as gum acacia and gum tragacanth, (b) naturally-occurring phosphatides such as soybean and lecithin, (c) esters or partial esters derived from fatty acids and hexitol anhydrides, for example 1o sorbitan monooleate, (d) condensation products of said partial esters with ethylene oxide, for example polyoxyethylene sorbitan monooleate. The emulsions may also contain sweetening and flavouring agents. Syrups and elixirs in accordance with the present invention may be formulated with 15 sweetening agents, for example glycerol, propylene glycol, sorbitol or sucrose. Such formulations may also contain a preservative and flavoring and coloring agents. The pharmaceutical compositions containing 1 and 2, separately or together, may be in the form of a sterile injectable aqueous or oleagenous suspension or solution. The 20 suspension may be formulated according to known methods using those suitable dispersing or wetting agents and suspending agents which have been mentioned hereinbefore. A suitable sterile injectable preparation may also be a sterile injectable solution or suspension in a non toxic parenterally-acceptable diluent or solvent, for example a solution in 1,3-butane-diol. Examples of suitable acceptable vehicles and 25 solvents that may be employed are water, Ringer's solution and an isotonic sodium chloride solution. In addition, sterile, fixed oils may conventionally be employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed, including synthetic mono-or diglycerides. In addition, fatty acids such as oleic acid find use in the preparation of injectables in accordance with the present invention. 30 Preparations for parenteral administration according to the present invention containing 1 and 2, separately or together, include sterile aqueous or non-aqueous solutions, WO 2006/018182 PCT/EP2005/008623 33 suspension, or emulsions. Examples of suitables non-aqueous solvents or vehicles for the preparations in accordance with the present invention are propylene glycol, polyethylene glycol, 5 vegetable oils, such as olive oil and corn oil, gelatin, and injectable organic esters such as ethyl oleate. Such dosage forms may also contain adjuvants such as preserving, wetting, emulsifying, and dispersing agents. They may be sterilized by, for example, by filtration through a bacteria-retaining filter, by incorporating sterilizing agents into the compositions, by irradiating the compositions, or by heating the compositions. They io may also be manufactured in the form of sterile solid compositions which can be reconstituted in sterile water, or some other sterile injectable medium immediately before use. The elements 1 and 2 of the combination of this invention may also be administered in 15 the form of suppositories for rectal administration. Such compositions can be prepared by mixing the active ingredient with a suitable non-irritating excipient which is solid at ordinary temperatures but liquid at the rectal temperature and will therefore melt in the rectum to release the active ingredient. Such materials are cocoa butter, hard fat, and polyethylene glycols. 20 Compositions for buccal, nasal or sublingual administration in accordance with the present invention may be prepared with standard excipients well known in the art. For topical administration, the elements 1 and 2 of the combination -of this invention 25 may be formulated, separately or together, in liquid or semi-liquid preparations. Examples of suitable preparations are: liniments, lotions, applications; oil-in-water or water-in-oil emulsions such as creams, ointments, jellies or pastes, including tooth pastes; solutions or suspensions such as drops. 30 The dosage of the active ingredients in the compositions in accordance with the present invention may be varied, although the amount of the active ingredients I and 2 shall be such that a suitable dosage form is obtained. Hence, the selected dosage and the selected WO 2006/018182 PCT/EP2005/008623 34 dosage form shall depend on the desired therapeutic effect, the route of administration and the duration of the treatment. Suitable dosage ranges for the combination are from the maximal tolerated dose for the single agent to lower doses, e.g. to one tenth of the maximal tolerated dose. 5 In the following, the present invention is illustrated via examples of pharmaceutical compositions comprising a compound 1 of chemical structure (I) in combination with one of the aforementioned combination partners 2, and by in vivo combination studies showing the potency of the combination to inhibit the proliferationn and/or to induce the 10 apoptosis of tumour cells. In these examples, the compound 1 of chemical structure (I) is 4-[[(7R)-8-cyclopentyl-7-ethyl-5,6,7,8-tetrahydro-5-methyl-6-oxo-2 pteridinyl]amino]-3-methoxy-N-(1-methyl-4-piperidinyl)-benzamide, which is a compound of Formula (I) according to the invention (Exemplified compound Nr. 46 in Table 1). 15 Combination of exemplified compound Nr. 46 of Table I and irinotecan (HCT 116 colon cancer model combination study) Objective of the study 20 Exemplified compound Nr. 46 of Table 1 is a potent and selective inhibitor of the serine/threonine kinase PLK-1. irinotecan (sold under the Trade name Campto*) is a standard chemotherapeutic agent for treatment of colorectal carcinomas. Previous studies have shown that exemplified compound Nr. 46 of Table 1 and irinotecan are active on HCT 116 derived tumors in nude mice. The 25 goal of the present study was to assess the anti-cancer efficacy of suboptimal doses of exemplified compound Nr. 46 of Table 1, irinotecan and the combination of exemplified compound Nr. 46 of Table 1 and irinotecan, in the human colon carcinoma model HCT 116 grown as xenograft in nude mice. Suboptimal doses of both compounds were used to facilitate the detection of 30 additive, synergistic or antagonistic effects. Design of the study WO 2006/018182 PCT/EP2005/008623 35 Model: Human colon carcinoma model HCT 116 grown as subcutaneous xenografts in nude mice. 5 Treatment groups (10 animals per group): Controls Vehicle, i.v., once weekly for 6 weeks ((q7d)x6) Exemplified compound Nr. 46 of Table 1 30 mg/kg, i.v., once weekly for 10 weeks ((q7d)x10) Irinotecan 12.5 mg/kg, i.p., once weekly for 10 weeks ((q7d)xlO) 10 Combination 30 mg/kg exemplified compound Nr. 46 of Table 1, i.v., once weekly for 10 weeks ((q7d)xlO) and 12.5 mg/kg irinotecan, i.p., once weekly (-1h after exemplified compound Nr. 46 of Table 1) for 10 weeks ((q7d)xlO) Is Tumor volumes and animal weights were recorded 3 times per week. Evaluation of therapy results was based on the absolute volumes of individual tumors. Material and Methods: Mice were female BomTac:NMRI-nu/nu. Exemplified compound Nr. 46 of 20 Table 1 was dissolved in hydrochloric acid (0.1 N) diluted with 0.9% NaCl and injected intravenously into the tail vein. irinotecan infusion concentrate was diluted with 0.9% NaCl and injected intraperitoneally. The administration volume was 10 ml per kg body weight for both compounds. HCT 116 tumors were established from cultured HCT 116 cells. Tumor volumes were determined 25 three times a week using a caliper. The weight of mice was determined as an indicator of tolerability on the same days. Plasma samples were taken on the last treatment day. Main results (see Figures 1.1-1.3) 30 Brief description of the Figures WO 2006/018182 PCT/EP2005/008623 36 Figure 1.1 HCT 116 tumor responses to treatment with 30 mg/kg exemplified compound Nr. 46 of Table 1, 12.5 mg/kg irinotecan or both. HCT 116 tumor-bearing mice were treated intravenously with 30 mg/kg 5 exemplified compound Nr. 46 of Table 1 once weekly ((q7d)x1O), with 12.5 mg/kg irinotecan once weekly ((q7d)xlO), with both in parallel ((q7d)xlO) or once weekly with the vehicle only, and median tumor volumes were plotted over time. Day I was the first day, day 64 the last day of treatment and day 121 the final day of the study. The triangles 1o indicate the treatment days. Figure 1.2 Days until HCT 116 tumors reach 1000 mm 3 in volume. HCT 116 tumor-bearing mice were treated with 30 mg/kg exemplified is compound Nr. 46 of Table 1. i.v. once weekly ((q7d)x10), with 12.5 mg/kg irinotecan i.p. once weekly ((q7d)x10), or a combination of both compounds ((q7d)x10) at respective doses. Vehicle treated mice (once weekly) were used as controls. Individual days until HCT 116 tumors reach 1000 mm 3 in volume were plotted. Each symbol represents one 20 individual tumor. The horizontal lines represent the mean days. Figure 1.3 Change of body weight in response to treatment with 30 mg/kg exemplified compound Nr. 46 of Table 1, 12.5 mg/kg irinotecan or both. 2S HCT 116 tumor-bearing mice were treated intravenously with 30 mg/kg exemplified compound Nr. 46 of Table 1 once weekly ((q7d)xlO), with 12.5 mg/kg irinotecan once weekly ((q7d)xlO), with both in parallel ((q7d)xlO) or once weekly with the vehicle only and average changes of body weight were plotted over time. Day 1 was the first day, day 64 the 30 last day of treatment and day 121 the final day of the study. The triangles indicate the treatment days. The triangles indicate the treatment days.
WO 2006/018182 PCT/EP2005/008623 37 Results on day 39 (end of the controls): 30 mg/kg exemplified compound Nr. 46 of Table 1 i.v. significantly delays HCT 116 tumor growth (T/C = 20 %, p<0.001) 5 12.5 mg/kg irinotecan i.p. significantly delays tumor growth (T/C = 25 %, p<0.001) Combined administration of 30 mg/kg exemplified compound Nr. 46 of 10 Table 1 and of 12.5 mg/kg irinotecan significantly delays tumor growth (T/C = 8 %, p<0.001). 30 mg/kg exemplified compound Nr. 46 of Table 1, 12.5 mg/kg irinotecan and their combination are well tolerated. Control mice gained 15 10.3 % body weight. Mice treated with 30 mg/kg exemplified compound Nr. 46 of Table 1 showed 8.6 % body weight increase, mice treated with 12.5 mg/kg irinotecan gained 5.9 % body weight in average, and mice treated with the combination gained 5.5. % body weight. 20 Results on day 121 (end of the study): Weekly treatment (until day 64) with exemplified compound Nr. 46 of Table 1, irinotecan or a combination thereof delays the average time to reach 1000 mm3 in tumor volume for 33.7 days, 35.1 days or 56.0 days, 25 respectively. Conclusions A treatment with suboptimal doses of exemplified compound Nr. 46 of Table 1 30 or irinotecan significantly delays tumor growth and is well tolerated.
WO 2006/018182 PCT/EP2005/008623 38 Treatment with the combination of suboptimal doses of exemplified compound Nr. 46 of Table 1 and irinotecan shows a significant growth delay and a higher efficacy than either of the compounds alone, without a decrease in tolerability. 5 The comparison of the growth delay (time until 1000 mm 3 in tumor size) shows an additive/synergistic effect. Combination of exemplified compound Nr. 46 of Table 1 and docetaxel 10 (NCI-H460 lung model) Objective of the study Exemplified compound Nr. 46 of Table 1 is a potent and selective inhibitor of 15 the PLK1 serine/threonine kinase, Docetaxel (sold under the Trade Name Taxotere") is a standard chemotherapeutic agent for treatment of lung cancer. Previous studies have shown that exemplified compound Nr. 46 of Table I is active on nude mice xenografts derived from the human lung cancer cell line NCI-H460. The goal of the present study was to assess the anti-cancer effects of 20 suboptimal doses of exemplified compound Nr. 46 of Table 1 and docetaxel on NCI-H460 tumor growth when administered alone or in combination. Suboptimal doses of both compounds were used to facilitate the detection of additive, synergistic or antagonistic effects. 25 Design of the study Model: Human non-small cell lung carcinoma model NCI-H460 grown as subcutaneous xenografts in nude mice. 30 Treatment groups (intravenous administration, 10 animals per group): Controls Vehicle, once weekly for 4 weeks ((q7d)x4) Exemplified compound Nr. 46 of Table 1 WO 2006/018182 PCT/EP2005/008623 39 50 mg/kg, once weekly for 4 weeks ((q7d)x4) Docetaxel 15 mg/kg, once weekly for 4 weeks ((q7d)x4) Combination 50 mg/kg exemplified compound Nr. 46 of Table 1, once weekly for 4 weeks ((q7d)x4) and 15 mg/kg Docetaxel, 5 once weekly (3 days after exemplified compound Nr. 46 of Table 1) for 4 weeks ((q7d)x4) Tumor volumes and animal weights were recorded 3 times per week.Evaluation of therapy results was based on the absolute volumes of individual tumors. 10 Material and methods: Mice were female BomTac:NvIRI-nu/nu. Exemplified compound Nr. 46 of Table 1 was dissolved in hydrochloric acid (0.1 N) diluted with 0.9% NaCl and injected intravenously into the tail vein. Docetaxel infusion concentrate was is diluted with 0.9% NaCI and injected intravenously. The administration volume was 10 ml per kg body weight. NCI-H460 tumors were established from cultured NCI-H460 cells. Tumor volumes were determined three times a week using a caliper. The weight of mice was determined as an indicator of tolerability on the same days. Plasma samples were taken on the last treatment 20 day. Main results (see Figures 2.1-2.3) 25 Brief description of the Figures Figure 2.1 NCI-H460 tumor responses to treatment with 50 mg/kg exemplified compound Nr. 46 of Table 1, 15 mg/kg Docetaxel or both. 30 NCI-H460 tumor-bearing mice were treated intravenously with 50 mg/kg exemplified compound Nr. 46 of Table 1 once weekly ((q7d)x4), with 15 mg/kg Docetaxel once weekly ((q7d)x4), with both in parallel or once WO 2006/018182 PCT/EP2005/008623 40 weekly with the vehicle only, and median tumor volumes were plotted over time. Day 1 was the first day, day 25 the last day of treatment and day 43 the last day of the calculation of the median tumor volume. The triangles indicate the treatment days. 5 Figure 2.2 Days until NCI-H460 tumors reach 1000 mm 3 in volume. NCI-H460 tumor-bearing mice were treated with 50 mg/kg exemplified compound Nr. 46 of Table 1 i.v. once weekly ((q7d)x4), with 15 mg/kg 10 Docetaxel i.v. once weekly ((q7d)x4), or a combination of both compounds at the same doses. Vehicle treated mice (once weekly) were used as controls. Individual days until NCI-H460 tumors reach 1000 mm 3 in volume were plotted. Each symbol represents one individual tumor. The horizontal lines represent the median days. 15 Figure 2.3 Change of body weight in response to treatment with 50 mg/kg exemplified compound Nr. 46 of Table 1, 15 mg/kg Docetaxel or both. NCI-H460 tumor-bearing mice were treated intravenously with 50 mglkg 20 exemplified compound Nr. 46 of Table 1 once weekly ((q7d)x4), with 15 mg/kg Docetaxel once weekly ((q7d)x4), with both in parallel or once weekly with the vehicle only and average changes of body weight were plotted over time. Day 1 was the first day, day 25 the last day of treatment and day 43 the last day of the calculation of the median body 25 weights. The triangles indicate the treatment days. Results on day 17 (end of the controls) 50 mg/kg exemplified compound Nr. 46 of Table 1 i.v. once weekly does 30 not significantly delay NCI-H460 tumor growth (T/C = 65 %, p>0.05).
WO 2006/018182 1CT/EP2005/008623 41 15 mg/kg Docetaxel i.v. once weekly significantly delays tumor growth (T/C = 42 %, p<0.05). Combined administration of 50 mg/kg exemplified compound Nr. 46 of 5 Table I and of 15 mg/kg Docetaxel significantly delays tumor growth (T/C = 26 %, p<0.0O1). 50 mg/kg exemplified compound Nr. 46 of Table 1 is well tolerated. 10 15 mg/kg Docetaxel administered alone or in combination is not well tolerated. Mice treated with 15 mg/kg Docetaxel on average lost 4.8 % body weight until day 17. The combination of exemplified compound Nr. 46 of Table 1 and Docetaxel induced a body weight loss of 8.3. %. is Results until day 71 (end of the study) Weekly treatment (until day 25) with exemplified compound Nr. 46 of Table 1, Docetaxel or a combination thereof delays the average time to reach 1000 mm 3 in tumor volume for 4.0 days, 10.5 days or 28.0 days 20 compared to the controls, respectively. Mice treated with 15 mg/kg Docetaxel further lost body weight (up to 9.4 % on day 24) and one mouse had to be euthanized due to severe loss of body weight. Mice treated simultaneously with exemplified compound 25 Nr. 46 of Table 1 and Docetaxel further lost body weight (up to 10.4 % on day 24) and two mice had to be euthanized due to severe loss of body weight. Conclusions 30 A treatment with suboptimal doses of exemplified compound Nr. 46 of Table 1 does not significantly delay tumor growth (T/C =65 %, p>0.05).
WO 2006/018182 PCT/EP2005/008623 42 Docetaxel significantly delays tumor growth (T/C = 42 %, p<0.05). A combination of exemplified compound Nr. 46 of Table 1 and Docetaxel 5 shows a significant growth delay compared to the controls (T/C = 26 %, p<0.001). The difference to the single treatment with exemplified compound Nr. 46 of Table I is also significant (p<0.01), indicating that the two agents might act at least additively. 10 Combination of exemplified compound Nr. 46 of Table 1 and Gemcitabine (BxPC-3 pancreas model) Objective of the study is Exemplified compound Nr. 46 of Table 1 is a potent and selective inhibitor of the serine/threonine kinase PLK1. Gemcitabine (sold under the Trade Name Gemzar*) is a standard chemotherapeutic agent for treatment of pancreatic adenocarcinomas .The goal of the present study was to assess the anti-cancer efficacy of suboptimal doses of exemplified compound Nr. 46 of Table 1, 20 gemcitabine and their combination in the human pancreas adenocarcinoma model BxPC-3 grown as xenograft in nude mice. Suboptimal doses of both compounds were used to facilitate the detection of additive, synergistic or antagonistic effects. 25 Design of the study Model: Human adenocarcinoma model BxPC-3 grown as subcutaneous xenografts in nude mice. 30 Treatment groups (10 animals per group): Controls Vehicle, i.v., once weekly for 4 weeks ((q7d)x4) Exemplified compound Nr. 46 of Table 1 WO 2006/018182 PCI/EP2005/008623 43 50 mg/kg, i.v., once weekly for 6 weeks ((q7d)x6) Gemcitabine 100 mg/kg, i.p., once weekly for 6 weeks ((q7d)x6) Combination 50 mg/kg exemplified compound Nr. 46 of Table 1, i.v., once weekly for 6 weeks ((q7d)x6) and 100 mg/kg 5 gemcitabine, i.p., once weekly (-1h after exemplified compound Nr. 46 of Table 1) for 6 weeks ((q7d)x6) Tumor volumes and animal weights were recorded 3 times per week. Evaluation of therapy results was based on the absolute volumes of individual tumors. 10 Material and Methods: Mice were female BomTac:NMRI-nu/nu. Exemplified compound Nr. 46 of Table 1 was dissolved in hydrochloric acid (0.1 N) diluted with 0.9% NaCl and injected intravenously into the tail vein. Gemcitabin infusion concentrate was i5 diluted with 0.9% NaCl and injected intraperitoneally. The administration volume was 10 ml per kg body weight for both compounds. BxPC-3 tumours were established from cultured BxPC-3. Tumour volumes were determined three times a week using a calliper. The weight of mice was determined as an indicator of tolerability on the same days. Plasma samples were taken on the last 20 treatment day. Main results (see Figures 3.1-3.2) Brief description of the Figures 25 Figure 3.1 BxPC-3 tumor responses to treatment with 50 mg/kg exemplified compound Nr. 46 of Table 1, 100 mg/kg gemcitabine or both. BxBC-3 tumor-bearing mice were treated intravenously with 50 mg/kg 30 exemplified compound Nr. 46 of Table 1 once weekly ((q7d)x6), with 100 mg/kg gemcitabine once weekly ((q7d)x6), with both in parallel or once weekly with the vehicle only, and median tumour volumes were WO 2006/018182 PCT/EP2005/008623 44 plotted over time. Day 1 was the first day, day 36 the last day of treatment and day 26 the last day of the calculation of the median tumour volume, 5 Figure 3.2 Change of body weight in response to treatment with 50 mg/kg exemplified compound Nr. 46 of Table 1, 100 mg/kg gemcitabine or both. BxPC-3 tumour-bearing mice were treated intravenously with 50 mg/kg 10 exemplified compound Nr. 46 of Table 1 once weekly ((q7d)x6), with 100 mg/kg gemcitabine once weekly ((q7d)x6), with both in parallel or once weekly with the vehicle only and average changes of body weight were plotted over time. Day 1 was the first day, day 36 the last day of treatment and day 43 the last day of the calculation of the median body 15 weights. The triangles indicate the treatment days. Results on day 26 (end of the controls): 50 mg/kg exemplified compound Nr. 46 of Table 1 i.v. significantly 20 delays HCT 116 tumour growth (T/C = 28 %). 100 mg/kg gemcitabine i.p. only marginally delays tumour growth (T/C = 65%) Higher doses of gemcitabine were not tolerated. 25 A combined administration of 50 mg/kg exemplified compound Nr. 46 of Table 1 and of 100 mg/kg gemcitabine delays tumour growth to the same extend as exemplified compound Nr. 46 of Table 1 alone (T/C = 24%). 30 50 mg/kg exemplified compound Nr. 46 of Table 1, 100 mg/kg gemcitabine and their combination were well tolerated. Control mice gained 6.2 % body weight. Mice treated with 50 mg/kg exemplified WO 2006/018182 PC'T/EP2005/008623 45 compound Nr. 46 of Table I showed 8.2 % body weight increase, mice treated with 100 mg/kg gemcitabine gained 8.8 % in average and mice treated with the combination gained 8.5. % body weight. 5 Results on day 43 (end of the study): Weekly treatment (until day 36) with exemplified compound Nr. 46 of Table 1 or a combination of exemplified compound Nr. 46 of Table I and gemcitabine delays the average time to reach 1000 mm in tumour 10 volume to the same extend. There is no significant difference between the two treatment groups. Conclusions 15 Treatment with suboptimal doses of exemplified compound Nr. 46 of Table 1 significantly delays tumour growth and is tolerated well. In contrast, the maximal tolerated dose of gemcitabine does not result in significant anti-tumour activity. 20 Treatment with the combination of suboptimal doses of exemplified compound Nr. 46 of Table I and gemcitabine shows a comparable growth delay as exemplified compound Nr. 46 of Table 1 alone, indicating that the two agents do not act antagonistically in this model. 25 A process for the manufacture of exemplified compound Nr. 46 of Table 1, i.e. the compound 4-[[(7R)-8-cyclopentyl-7-ethyl-5,6,7,8-tetrahydro-5-methyl-6-oxo-2 pteridinyl] amino]-3-methoxy-N-(1-methyl-4-piperidinyl)-benzamide, is described in WO 03/20722 as well as in WO 04/76454, which are incorporated herein by reference. 30 However, for the sake of completeness, a process for the manufacture of the compound 4 -[[(7R)-8-cyclopentyl-7-ethyl-5,6,7,8-tetrahydro-5-methyl-6-oxo-2-pteridinyl] amino] 3-methoxy-N-(1-methyl-4-piperidinyl)-benzamide is described as well hereinafter. This WO 2006/018182 PC'T/EP2005/008623 46 method is to be understood as an illustration of the invention without restricting it to the subject matter thereof. Synthesis of 4-[[(7R)-8-cyclopentyl-7-ethyl-5,6.7,8-tetrahydro-5-methyl-6-oxo-2 s pteridinvllaminol-3-methoxy-N-(1-methyl-4-piperidinyl)-benzamide For the synthesis, first of all an intermediate compound Z3 is prepared as described below. HN N N 0 OH 10 54.0 g (0.52 mol) D-2-aminobutyric acid are suspended in 540 mL methanol and slowly combined with 132 g (1.1 mol) thionyl chloride while cooling with ice. The mixture is refluxed for 1.5 h and then evaporated down. The oil remaining is combined with 540 mL tert-butylmethylether and the colourless crystals formed are suction filtered. is Yield: 78.8 g of a compound Z3a (colourless crystals) 74.2 g of the compound Z3a and 43.5 mL (0.49 mol) cyclopentanone are dissolved in 800 mL dichloromethane. After the addition of 40.0 g (0.49 mol) sodium acetate and 150.0 g (0.71 mol) sodium triacetoxyborohydride at 0*C the mixture is stirred for 12 h 20 at ambient temperature and then 500 mL of 20% sodium hydrogen carbonate solution arc added. The aqueous phase is extracted with dichloromethane. The combined organic phases are washed with water, dried over MgSO 4 and evaporated down. Yield: 85.8 g of a compound Z3b (light yellow oil) WO 2006/018182 PCT/EP2005/008623 47 40.0 g of the compound Z3b and 30.0 g (0.22 mol) potassium carbonate are suspended in 600 mL acetone and combined with 45.0 g (0.23 mol) 2,4-dichloro-5-nitropyrimidin in 200 mL acetone while cooling with ice. After 12 h a further 5.0 g 2,4-dichloro-5 nitropyrimidin are added and stirred for 3 h. The reaction mixture is evaporated down, 5 taken up in 800 mL ethyl acetate and 600 mL water and the aqueous phase is extracted with ethyl acetate. The combined organic phases are washed with water, dried over MgSO 4 and evaporated down. Yield: 75.0 g of a compound Z3c (brown oil) 10 100 g of the compound Z3c are dissolved in 650 mL glacial acetic acid and at 70"C 20 g of iron powder are added batchwise. The mixture is stirred for 1 h at 70*C, then for 1.5 h at 100*C and then filtered hot through diatomite (kieselguhr). The reaction mixture is evaporated down, taken up in methanol/dichloromethane, applied to silica gel and purified with ethyl acetate by Soxhlet extraction. The solvent is removed and the 15 residue stirred with methanol. Yield: 30.0 g of a compound Z3d (light brown crystals) 25.0 g of the compound Z3d and 6.5 mL (0.1 mol) methyl iodide are placed in 250 mL dimethylacetamide and at -10*C 3.8 g (0.95 mol) sodium hydride as a 60% dispersion 20 in mineral oil is added. It is stirred for 20 min at 0 0 C, then for 30 min at ambient temperature and finally ice is added. The reaction mixture is evaporated down and combined with 300 mL water. The precipitate formed is suction filtered and washed with petroleum ether. Yield: 23.0 g of a compound Z3e (colourless solid) 25 6.0 g of the compound Z3e and 5.1 g (31 mmol) 4-anino-3-methoxybenzoic acid are suspended in 90 mL ethanol and 350 mL water, combined with 3.5 mL concentrated hydrochloric acid and refluxed for 48 h. The reaction mixture is evaporated down, the residue stirred with methanol/diethyl ether and the precipitate formed is suction filtered. 30 Yield: 6.3 g of a compound Z3 (light beige crystals) WO 2006/018182 PC'I'/EP2005/008623 48 4-[[(7R)-8-cyclopentyl-7-ethyl-5,6,7,8--tetrahydro-5-methyl-6-oxo-2-pteridinyllamino] 3-methoxy-N-(1-methyl-4-piperidinyl)-benzamide is obtained as described below. 0.15 g of the compound Z3, 0.12 g TBTU, 0.12 mL DIPEA are dissolved in 5 mL 5 dichloromethane and stirred for 30 minutes at 25*C. Then 50 mg 1-methyl-4 aminopiperidin are added and the mixture is stirred for a further 2.5 hours at 25"C. The solution is then extracted with water and then evaporated down. The residue is dissolved in warm ethyl acetate and crystallised from ether and petroleum ether. Yield: 0.025 g of white crystals. M.p.: 203'C as the base. 10 All compounds of Formula (I) according to the invention may be prepared by the synthesis methods A described hereinafter, while the substituents of general Formula (Al) to (A9) have the meanings given hereinbefore. This method is to be understood as an illustration of the invention without restricting it to the subject matter thereof. Is Method A STEP 1A 20 A compound of Formula (Al) is reacted with a compound of Formula (A2) to obtain a compound of Formula (A3) (Diagram 1A). This reaction may be carried out according to WO 00/43369 or WO 00/43372. Compound (Al) is commercially obtainable, for example, from City Chemical LLC, 139 Allings Crossing Road, West Haven, CT, 06516, USA. Compound (A2) may be prepared by procedures known from the 25 literature: (a) F. Effenberger, U. Burkhart, J. Willfahrt Liebigs Ann. Chen. 1986, 314 333; (b) T. Fukuyama, C.-K. Jow, M. Cheung, Tetrahedron Lett. 1995, 36, 6373-6374; (c) R. K. Olsen, I. Org. Chen. 1970, 35, 1912-1915; (d) F.E. Dutton, B.H. Byung Tetrahedron Lett. 1998, 30, 5313-5316; (e) J. M. Ranajuhi, M. M. Joullie Synth. Commun. 1996, 26, 1379-1384.). 30 Diagram 1A WO 2006/018182 PCT/EP2005/008623 49 0 0 R N " + Hy)ffR 3 CI N C + R 0 C1 N NR R--7 O,1 () (1)2 R (A3) In Step 1A, I equivalent of the compound (Al) and I to 1.5 equivalents, preferably 1.1 equivalents of a base, preferably potassium carbonate, potassium hydrogen carbonate, s sodium carbonate or sodium hydrogen carbonate, calcium carbonate, most preferably potassium carbonate, are stirred in a diluent optionally mixed with water, for example acetone, tetrahydrofuran, diethylether, cyclohexane, petroleum ether or dioxane, preferably cyclohexane or diethylether. 10 At a temperature of 0 to 15 *C, preferably 5 to 10 'C, 1 equivalent of an amino acid of Formula (A2), dissolved in an organic solvent, for example acetone, tetrahydrofurane, diethylether, cyclohexane or dioxane, is added dropwise. The reaction mixture is heated to a temperature of 18'C to 30 *C, preferably about 22'C, with stirring and then stirred for a further 10 to 24 hours, preferably about 12 hours. Then the diluent is distilled off, is the residue is combined with water and the mixture is extracted two to three times with an organic solvent, such as diethylether or ethyl acetate, preferably ethyl acetate. The combined organic extracts are dried and the solvent is distilled off. The residue (compound A3) may be used in Step 2 without any prior purification. 20 STEP2A The compound obtained in Step 1A (A3) is reduced at the nitro group and cyclised to form the compound of Formula (A4) (Diagram 2A). 25 Diagram2A WO 2006/018182 PCT/EP12005/008623 50 0 II. H N- N ;O R 3 R Cl N Reduction C j-" R2 13 R 0, R- R R o (A4) (A3) In Step 2A, 1 equivalent of the nitro compound (A3) is dissolved in an acid, preferably glacial acetic acid, formic acid or hydrochloric acid, preferably glacial acetic acid, and 5 heated to 50 to 70*C, preferably about 60*C. Then a reducing agent, for example zinc, tin or iron, preferably iron filings, is added to complete the exothermic reaction and the mixture is stirred for 0.2 to 2 hours, preferably 0.5 hours, at 100 to 125 'C, preferably at about 117 *C. After cooling to ambient temperature the iron salt is filtered off and the solvent is distilled off. The residue is taken up in a solvent or mixture of solvents, for 10 example ethyl acetate or dichloromethane/ methanol 9/1 and semisaturated NaCl solution, and filtered through kieselgur, for example. The organic phase is dried and evaporated down. The residue (compound (A4)) may be purified by chromatography or by crystallisation or used as the crude product in Step 3A of the synthesis. is STEP3A The compound obtained in Step 2A (A4) may be reacted by electrophilic substitution as shown in Diagram 3A to obtain the compound of Formula (A5). 20 Diagram 3A H N :0 N N 0 CI N CN
R
2 R R (A4) (A5) WO 2006/018182 PCT/E P2005/008623 51 In Step 3A 1 equivalent of the amide of Formula (A4) is dissolved in an organic solvent, for example dimethylformamide or dimethylacetamide, preferably dimethylacetamide, and cooled to about -5 to 5 *C, preferably 0 0 C. 5 Then 0.9 to 1.3 equivalents of sodium hydride and 0.9 to 1.3 equivalents of a methylating reagent, e.g. methyl iodide, are added. The reaction mixture is stirred for 0.1 - 3 hours, preferably about 1 hour, at about 0 to 10 "C, preferably at about 5 'C, and may optionally be left to stand for a further 12 hours at this temperature. The reaction mixture is poured onto ice water and the precipitate is isolated. The residue (compound 1o (A5)) may be purified by chromatography, preferably over silica gel, or by crystallisation, or used as the crude product in step 4A of the synthesis. Step 4A is The amination of the compound (A5) obtained in Step 3A to yield the compound of Formula (A9) (Diagram 4A) may be carried out using the methods known from the literature, for variants 4.1 A from e.g. (a) M.P.V. Boarland, J.F.W. McOmie J. Chem. Soc. 1951, 1218-1221 or (b) F. H. S. Curd, F. C. Rose J. Chem. Soc. 1946, 343-348, for variants 4.2 A from e.g. (a) Banks J. Am. Chem. Soc. 1944, 66, 1131, (b) Ghosh and 20 Dolly J. Indian Chem. Soc. 1981, 58, 512-513 or (c) N. P. Reddy and M. Tanaka Tetrahedron Lett. 1997, 38, 4807-4810. Diaeram 4A WO 2006/018182 PCT/EIP2005/008623 52
NH
2
R
4 (A 6) 0 OH NR R R (A5) 4 43 R R (A7) 0 OR HN R R4 Ra (AS) 0 OR For example, in variant 4.1 A, 1 equivalent of the compound (AS) and 1 to 3 equivalents, preferably about 2 equivalents of the compound (A6) are heated without a solvent or in an organic solvent such as for example sulpholane, dimethylformamide, s dimethylacetamide, toluene, N-methylpyrrolidone, dimethylsulphoxide or dioxane, preferably sulpholane, for 0.1 to 4 hours, preferably 1 hour, at 100 to 220 *C, preferably at about 160 'C. After cooling, the product (A9) is crystallised by the addition of organic solvents or mixtures of solvents, e.g. diethylether/methanol, ethyl acetate, methylene chloride, or diethylether, preferably diethylether/methanol 9/1, or purified by 10 chromatography.
WO 2006/018182 PCTI/EP2005/008623 53 For example, in variant 4.2 A , 1 equivalent of the compound (A5) and 1 to 3 equivalents of the compound (A6) are stirred with acid, for example 1-10 equivalents of 10-38% hydrochloric acid and/or an alcohol, for example ethanol, propanol, butanol, s preferably ethanol, at reflux temperature for 1 to 48 hours, preferably about 5 hours. The product precipitated (A9) is filtered off and optionally washed with water, dried and crystallised from a suitable organic solvent. For example, in variant 4.3 A ,1 equivalent of the compound (A5) and 1 to 3 equivalents 1o of the compound (A7) are dissolved in a solvent, for example toluene or dioxane and combined with a phosphine ligand, for example 2,2'-bis-(diphenylphosphino)-1,1' binaphthyl and a palladium catalyst, for example tris(dibenzylidene-acetone) dipalladium(0) and a base, for example caesium carbonate, and refluxed for 1-24 h, preferably 17 h. The reaction mixture is purified for example over silica gel and the is product (A8) is isolated from the solution or obtained by suitable crystallisation. The product (A8) is dissolved in a suitable solvent, for example dioxane and mixed with acid, for example semiconcentrated hydrochloric acid, for example in the ratio of solvent to acid of 3: 1. Then the mixture is refluxed for 1 - 48 h, for example 12 h, and the precipitate formed is isolated. If desired the product (A9) is purified by 20 crystallisation. Step 5A Diagram 5A 25 WO 2006/018182 IC'I'/EP2005/008623 54 HI M' R2 + NH--L-R H R2 R3 (A10) R 3 (All) 0 OH 0 N H For example, 1 equivalent of the compound (A9) is dissolved with 1 equivalent of an activating reagent, e.g. O-benzotriazolyl-N,N,N',N'-tetramethyluronium tetrafluoroborate (TBTU) and a base, for example 1.5 equivalents of s diisopropylethylamine (DIPEA) in an organic diluent, for example dichloromethane, tetrahydrofuran, dimethylformamide, N-methylpyrrolidone, dimethylacetamide, preferably dichloromethane or dimethylformamide. After the addition of I equivalent of the amine (Al0) the reaction mixture is stirred for 0.1 to 24 hours, preferably about 2 hours at 20'C to 100*C. The product of Formula (All) is obtained for example by 10 crystallisation or chromatographic purification. The compounds of general Formula (I) may be synthesised analogously to the following examples of synthesis. The numbering of the Examples corresponds to the numbering used in Table 1. These Examples are, however, intended only as examples of is procedures to illustrate the invention further, without restricting the invention to their subject matter. The preparation of some intermediate compounds used to synthesise the compounds is also described hereinafter. 20 Preparation of the acids WO 2006/018182 PC'I'/EP2005/008623 55 To synthesise the compounds of Examples 94 and 95 of Table 1, first an intermediate compound ZI HN N N 0 OH zi is prepared as described hereinafter. 5 50.0 g (0.48 mol) of D-alanine methyl ester x HCl and 49.1 g (0.50 mol) cyclohexanone are placed in 300 rnL dichloromethane and then combined with 41.0 g (0.50 mol) sodium acetate and 159.0 g (0.75 mol) sodium triacetoxyborohydride. The mixture is stirred overnight and then 300 mL of 10% sodium hydrogen carbonate solution are 10 added. The aqueous phase is extracted with dichloromethane. The combined organic phases are washed with 10% sodium hydrogen carbonate solution, dried over Na 2
SO
4 and evaporated down. Yield: 72.5 g of a compound Z1a (clear liquid) is 72.5 g of the compound ZIa are placed in 500 mL water and 76.6 g (0.39 mol) of 2,4 dichloro-5-nitropyrimidine in 500 mL diethyl ether are added. At a temperature of -5*C 100 mL 10% potassium hydrogen carbonate solution are added dropwise. The mixture is stirred for 3 h at -5 0 C and for a further 12 h at ambient temperature. The organic phase is separated off and dried over Na 2
SO
4 . On evaporation, the product crystallizes 20 out. Yield: 48.0 g of a compound ZIb (yellow crystals) 48.0 g of the compound Zib are dissolved in 350 mL glacial acetic acid and heated to 60"C. 47.5 g of iron powder are added, while the temperature rises to 105*C. The 25 reaction mixture is stirred for three hours at 80"C, then filtered hot through cellulose WO 2006/018182 PC'I'/EP2005/008623 56 and evaporated down. The residue is stirred in water and ethyl acetate, suction filtered and the light-grey precipitate is washed with ethyl acetate. The filtrate is washed with dilute ammonia and water, the organic phase is dried over Na 2
SO
4 , filtered through activated charcoal and evaporated down. Some more light-grey solids are obtained. 5 Yield: 29.5 g of a compound ZIc (light-grey crystals) 32.1 g of the compound Zlc are placed in 300 mL dimethylacetamide and combined with 13 mL (0.2 mol) methyl iodide. At -5*C 6.4 g (0.16 mol) sodium hydride as a 60% dispersion in mineral oil is added batchwise. After 2 h the reaction mixture is poured 1o onto 800 mL ice water. The precipitate formed is suction filtered and washed with petroleum ether. Yield: 33.0 g of a compound Zld (beige crystals) 4.0 g of the compound Zid and 2.3 g (15 mmol) 4-amino-3-methylbenzoic acid are is suspended in 50 mL ethanol and 120 mL water, combined with 2 mL concentrated hydrochloric acid and refluxed for 48 h. The precipitate formed on cooling is suction filtered and washed with water, ethanol and diethyl ether. Yield: 2.9 g of a compound ZI (colourless crystals) 20 To synthesise the compounds Example 188 and Example 203 of Table 1, first of all an intermediate compound Z2 N 0 HN N CI 0 OH Z2 is prepared as described below. 25 WO 2006/018182 PC'/EP2005/008623 57 A solution of 128.2 g (0.83 mol) D-alanine ethyl ester x HCI and 71.5 g (0.85 mol) cyclopentanone in 1500 mL dichloromethane is combined with 70.1 (0.85 mol) sodium acetate and 265.6 g (1.25 mol) sodium triacetoxyborohydride. The reaction mixture is stirred for 12 h and then poured into 1.5 L of a 10% sodium hydrogen carbonate 5 solution. The aqueous phase is extracted with dichloromethane. The combined organic phases are dried over Na 2
SO
4 and evaporated down. Yield: 143.4 g of a compound Z2a (colourless oil) 66.0 g of the compound Z2a are placed in 500 mL water and combined with 85.0 g 10 (0.44 mol) 2,4-dichloro-5-nitropyrimidine in 500 mL diethyl ether. At -5"C 100 mL 10% potassium hydrogen carbonate solution are added dropwise and the reaction mixture is stirred for 48 h at ambient temperature. The aqueous phase is extracted with diethyl ether, the combined organic phases are dried over Na 2
SO
4 and evaporated down. The dark red solid is stirred with petroleum ether and suction filtered. is Yield: 88.0 g of a compound Z2b (yellow crystals) 88.0 g of the compound Z2b are dissolved in 1000 mL glacial acetic acid and at 60*C combined batchwise with 85 g iron powder, while the temperature rises to 110*C. It is stirred for 1 h at 60 0 C, then suction filtered hot through cellulose and evaporated down. 20 The brown solid is stirred with 700 mL water and suction filtered. Yield: 53.3 g of a compound Z2c (light brown crystals) 53.3 g of the compound Z2c are dissolved in 300 mL dimethylacetamide and combined with 13 mL (0.21 mol) methyl iodide. At -5*C 5.0 g (0.21 mol) sodium hydride as a 25 60% dispersion in mineral oil are added batchwise. After 12 h the reaction mixture is poured onto 1000 mL ice water and the precipitate formed is suction filtered. Yield: 40.0 g of a compound Z2d (colourless crystals) 4.0 g of the compound Z2d and 2.8 g (16 mmol) 4-amino-3-chlorbenzoic acid are 30 suspended in 25 mL ethanol and 60 mL water, combined with 3 mL concentrated hydrochloric acid and refluxed for 43 h. The precipitate formed on cooling is suction filtered and washed with water, ethanol and diethyl ether.
WO 2006/018182 PCI'/EP2005/008623 58 Yield: 0.9 g of a compound Z2 (colourless crystals) To synthesise the compounds of Examples 19, 21, 22, 23, 45, 46, 55, 58, 116, 128, 131, 133, 134, 136, 138, 177, 217, 231, 239, 46, 184, 166 and 187 of Table 1, first of all an 5 intermediate compound Z3 N N '. HN N N 0 0 OH Z3 is prepared as described below. 54.0 g (0.52 mol) D-2-aninobutyric acid are suspended in 540 mL methanol and slowly 1o combined with 132 g (1.1 mol) thionyl chloride while cooling with ice. The mixture is refluxed for 1.5 h and then evaporated down. The oil remaining is combined with 540 mL tert-butylmethylether and the colourless crystals formed are suction filtered. Yield: 78.8 g of a compound Z3a (colourless crystals) is 74.2 g of the compound Z3a and 43.5 mL (0.49 mol) cyclopentanone are dissolved in 800 mL dichloromethane. After the addition of 40.0 g (0.49 mol) sodium acetate and 150.0 g (0.71 mol) sodium triacetoxyborohydride at 0*C the mixture is stirred for 12 h at ambient temperature and then 500 mL of 20% sodium hydrogen carbonate solution are added. The aqueous phase is extracted with dichloromethane. The combined organic 20 phases are washed with water, dried over MgSO 4 and evaporated down. Yield: 85.8 g of a compound Z3b (light yellow oil) 40.0 g of the compound Z3b and 30.0 g (0.22 mol) potassium carbonate are suspended in 600 mL acetone and combined with 45.0 g (0.23 mol) 2,4-dichloro-5-nitropyrimidin WO 2006/018182 PCT/EP2005/(108623 59 in 200 mL acetone while cooling with ice. After 12 h a further 5.0 g 2,4-dichloro-5 nitropyrimidin are added and stirred for 3 h. The reaction mixture is evaporated down, taken up in 800 mL ethyl acetate and 600 mL water and the aqueous phase is extracted with ethyl acetate. The combined organic phases are washed with water, dried over s MgSO 4 and evaporated down. Yield: 75.0 g of a compound Z3c (brown oil) 100 g of the compound Z3c are dissolved in 650 mL glacial acetic acid and at 70*C 20 g of iron powder are added batchwise. The mixture is stirred for 1 h at 70*C, then for 1.5 1o h at 100*C and then filtered hot through kieselgur. The reaction mixture is evaporated down, taken up in methanol/dichloromethane, applied to silica gel and purified with ethyl acetate by Soxhlet extraction. The solvent is removed and the residue stirred with methanol. Yield: 30.0 g of a compound Z3d (light brown crystals) 15 25.0 g of the compound Z3d and 6.5 mL (0.1 mol) methyl iodide are placed in 250 mL dimethylacetamide and at -10'C 3.8 g (0.95 mol) sodium hydride as a 60% dispersion in mineral oil is added. It is stirred for 20 min at 0 0 C, then for 30 min at ambient temperature and finally ice is added. The reaction mixture is evaporated down and 20 combined with 300 mL water. The precipitate formed is suction filtered and washed with petroleum ether. Yield: 23.0 g of a compound Z3e (colourless solid) 6.0 g of the compound Z3e and 5.1 g (31 mmol) 4-amino-3-methoxybenzoic acid are 25 suspended in 90 mL ethanol and 350 mL water, combined with 3.5 mL concentrated hydrochloric acid and refluxed for 48 h. The reaction mixture is evaporated down, the residue stirred with methanol/diethyl ether and the precipitate formed is suction filtered. Yield: 6.3 g of a compound Z3 (light beige crystals) 30 To synthesise the compound of Examples 81, 82, 93 and 137 of Table 1, first of all an intermediate compound Z4 WO 2006/018182 PCI/EP2005/008623 60 5 N 0 HN N N 0 0 OH Z4 is prepared as described below. 10 25.0 g (0.19 mol) of ethyl 1-aminocyclopropane-1-carboxylate x HCl and 16.8 g (0.20 mol) of cyclopentanone are dissolved in 300 mL of dichloromethane and combined with 16.4 g (0.20 mol) of sodium acetate and 61.7 g (0.29 mol) of sodium triacetoxyborohydride. It is stirred overnight and the reaction mixture is then poured is onto 400 mL of 10% sodium hydrogen carbonate solution. The aqueous phase is extracted with dichloromethane. The combined organic phases are dried over Na 2 S0 4 and evaporated down. Yield: 34.5 g of a compound Z4a (colourless oil) 20 42.5 g (0.22 mol) of 2,4-dichloro-5-nitropyrimidine in 350 mL of diethyl ether are added to a mixture of 34.5 g of the compound Z4a in 350 mIL water. At -5 0 C the mixture is combined with 80 mL 10% potassium hydrogen carbonate solution and stirred overnight at ambient temperature. The aqueous phase is extracted with diethyl ether. The combined organic phases are dried over Na 2
SO
4 and evaporated down. 25 Yield: 53.8 g of a compound Z4b (brown oil) WO 2006/018182 PCT/EP2005/008623 61 20.1 g of the compound Z4b are dissolved in 200 mL glacial acetic acid and combined batchwise at 60'C with 19.1 g iron powder, during which time the temperature rose to 100"C. The mixture is stirred for 3 h at 60*C, then suction filtered through cellulose and 5 evaporated down. The residue is stirred in water and ethyl acetate and the yellow precipitate is suction filtered. The filtrate is washed with dilute ammonia and water, the organic phase dried over Na 2
SO
4 and evaporated down. After the addition of diethyl ether additional product crystallised out. Yield: 4.0 g of a compound Z4c (yellow crystals) 10 7.8 g of the compound Z4c and 2.6 mL (0.04 mol) methyl iodide are dissolved in 100 mL dimethylacetamide and at -5*C 1.5 g (0.04 mol) sodium hydride are added batchwise as a 60% dispersion in mineral oil. After 2 h the reaction mixture is poured onto ice water and the precipitate formed is suction filtered. 15 Yield: 7.5 g of a compound Z4d (light brown crystals) 3.0 g of the compound Z4d and 1.9 g (11 mmol) 4-amino-3-methoxybenzoic acid are suspended in 40 mL ethanol and 80 mL water, combined with 2 mL concentrated hydrochloric acid and refluxed for 20 h. A further 0.5 g of 4-amino-3-methoxybenzoic 20 acid are added and refluxed for48 h. The precipitate formed on cooling is suction filtered and washed with water, ethanol and diethyl ether. Yield: 2.1 g of a compound Z4 (colourless crystals) m.p.: 222-223*C To synthesise the compounds of Examples 162, 43, 53, 161, 202, 211, 215 and 212 of 25 Table 1, first of all an intermediate compound Z5 N O HN N 0 OH WO 2006/018182 PCI/EP2005/008623 62 is prepared as described below. 5 A mixture of 73.4 mL (0.5 mol) ethyl 2-bromoisobutyrate, 87.1 mL (0.75 mol) of 3-methyl-1-butylamine, 82.5 g (0.6 mol) sodium iodide and 76.0 g (0.6 mol) of potassium carbonate in 1000 mL ethyl acetate is refluxed for3 days. Any salts present are filtered off and the filtrate evaporated down. 10 Yield: 97.0 g of a compound Z5a (red oil) Z5 49.0 g (0.25 mol) of 2,4-dichloro-5-nitropyrimidine and 38.3 g (0.28 mol) of potassium carbonate are suspended in 500 mL acetone and at 0*C combined with 93.0 g of the compound Z5a in 375 mL acetone. The reaction mixture is stirred overnight at ambient is temperature, filtered and evaporated down. The residue dissolved in ethyl acetate is washed with water and the organic phase dried over MgSO 4 and evaporated down. Yield: 102.7 g of a compound Z5b (brown oil) 22.7 g of the compound Z5b are dissolved in 350 mL glacial acetic acid and at 60*C 20 combined batchwise with 17.4 g iron powder. After the addition ends the mixture is refluxed for 0.5 h, filtered hot and evaporated down. The residue is taken up in 200 mL dichloromethane/methanol (9:1) and washed with sodium chloride solution. The organic phase is suction filtered through kieselguhr, dried over MgSO 4 , evaporated down and purified by column chromatography (eluant: ethyl acetate/cyclohexane 1:1). 25 Yield: 1.9 g of a compound Z5c (colourless crystals) 1.9 g of the compound Z5c are dissolved in 32 mL dimethylacetamide and while cooling with ice combined with 0.3 g (7 mmol) sodium hydride as a 60% dispersion in mineral oil. After 10 min 0.5 mL (7 mmol) methyl iodide are added and stirred for 3 h 30 at ambient temperature. The reaction mixture is evaporated down and combined with water. The precipitate formed is suction filtered and washed with petroleum ether. Yield: 1.6 g of a compound Z5d (colourless crystals) WO 2006/018182 PCI/EP2005/008623 63 14.0 g of the compound Z5d and 10.0 g (0.06 mol) 4 -amino-3-methoxybenzoic acid are suspended in 200 mL dioxane and 80 mL water, combined with 10 mL concentrated hydrochloric acid and refluxed for 40 h. The precipitate formed on cooling is suction s filtered and washed with water, dioxane and diethyl ether. Yield: 13.9 g of a compound Z5 (colourless crystals) To synthesise the compounds of Examples 88, 194, 229 and 89 of Table 1, first of all an intermediate compound Z6 N 0 N N HN N N 1-0 0 OH 10 Z6 is prepared as described below. 15 6.0 g (0.06 mol) L-2-aminobutyric acid is placed in 80 mL 0.5 M sulphuric acid and at 0*C combined with 5.5 g (0.08 mol) sodium nitrite in 15 mL water. The reaction mixture is stirred for 22 h at 0*C, combined with ammonium sulphate and filtered. The filtrate is extracted with diethyl ether and the combined organic dried over MgSO 4 and evaporated down. 20 Yield: 6.0 g of a compound Z6a (yellow oil) 200 mL methanol are combined successively with 65.0 mL (0.89 mol) thionyl chloride and 76.0 g of the compound Z6a in 50 mL methanol while cooling with ice. The WO 2006/018182 PCI/EP2005/008623 64 resulting mixture is stirred for 1 h at 0"C and 2 h at ambient temperature and then the methanol and remaining thionyl chloride are eliminated in vacuo at 0 0 C. Yield: 40.0 g of a compound Z6b (yellow oil) 5 30.0 mL (0.17 mol) of trifluoromethanesulphonic acid anhydride are placed in 150 mL dichloromethane and while cooling with ice a solution of 20.0 g of the compound Z6b and 14.0 mL (0.17 mol) pyridine in 50 mL dichloromethane is added within one hour. The mixture is stirred for 2 h at ambient temperature, any salts formed are suction filtered and then washed with 100 mL water. The organic phase is dried over MgSO 4 10 and evaporated down. Yield: 42.0 g of a compound Z6c (light yellow oil) 42.0 g of the compound Z6c in 200 mL dichloromethane is added dropwise within one hour to a solution of 15.5 mL (0.17 mol) of aniline and 24.0 mL (0.17 mol) of is triethylamine in 400 mL dichloromethane while cooling with ice. The mixture is stirred for 1 h at ambient temperature and a further 2 h at 35*C. The reaction mixture is washed with water, dried over MgSO 4 and evaporated down. The residue remaining is purified by distillation (95-100*C, 1*10~3 mbar). Yield: 14.0 of a compound Z6d (colourless oil) 20 14.0 g of the compound Z6d and 16.0 g (0.1 mol) potassium carbonate are suspended in 100 mL acetone and at 10"C combined with 16.0 g (0.08 mol) of 2,4-dichloro-5 nitropyrimidine. The mixture is stirred for 4 h at 40'C, any salts formed are suction filtered and the filtrate evaporated down. The residue is taken up in 300 mL ethyl 25 acetate and washed with water. The organic phase is dried over MgSO 4 and evaporated down. Yield: 31.0 g of a compound Z6e (brown oil) 31.0 g of the compound Z6e are dissolved in 200 mL glacial acetic acid and at 60 0 C 30 combined batchwise with 10 g iron powder, during which time the temperature rose to 85*C. The-mixture is stirred for a further hour at 60*C, filtered through kieselguhr and evaporated down. The residue is stirred with methanol.
WO 2096/018182 PCI/EP2005/008623 65 Yield: 4.5 g of a compound Z6f (brown crystals) At -20'C 0.6 g (16 mmol) of sodium hydride as a 60% dispersion in mineral oil are added batchwise to a mixture of 4.5 g of the compound Z6f and 1.0 mL (16 mmol) 5 methyl iodide in 100 mL dimethylacetamide. After 1 h the reaction mixture is combined with 50 mL water and evaporated down. The residue is stirred with 200 mL water, the precipitate is suction filtered and washed with petroleum ether. Yield: 4.5 g of a compound Z6g (colourless crystals) 10 A suspension of 1.5 g of the compound Z6g and 1.4 g (8 mmol) of methyl 4-amino-3 methoxybenzoate in 30 mL toluene is combined with 0.4 g (0.6 mmol) of 2,2'-bis (diphenylphosphino)-1,1'-binaphthyl, 0.23 g (0.3 mmol) of tris(dibenzylideneacetone) dipalladium(0) and 7.0 g (21 mmol) of caesium carbonate and refluxed for17 h. The reaction mixture is applied to silica gel and purified by column chromatography (eluant: 15 dichloromethane/methanol 9:1). Yield: 1.7 g of a compound Z6h (yellow crystals) 1.7 g of the compound Z6h are dissolved in 50 mL dioxane, combined with 15 mL of semiconcentrated hydrochloric acid and refluxed forl2 h. After cooling the precipitate 20 formed is suction filtered. Yield: 1.1 g of a compound Z6 (colourless solid) To synthesise the compounds of Examples 26, 20,32, 56, 101, 112 and 209 of Table 1, first of all an intermediate compound Z7 2N N H N N 0 O OH Z7 WO 2006/018182 PCTI/EP2005/008623 66 5 is prepared as described below. 50.0 g (0.36 mol) D-alanine methyl ester x HCl is suspended in 500 mL of dichloromethane and 35 mL of acetone and combined with 30.0 g (0.37 inol) of sodium acetate and 80.0 g (0.38 rnol) of sodium triacetoxyborohydride. The mixture is stirred to for 12 h and then poured onto 400 mL of 10% sodium hydrogen carbonate solution. The organic phase is dried over Na 2
SO
4 and evaporated down. Yield: 51.0 g of a compound Z7a (yellow oil) A suspension of 51.0 g of the compound Z7a in 450 mL water is combined with 80.0 g 15 (0.41 mol) of 2,4-dichloro-5-nitropyridine in 450 mL of diethyl ether. At -5'C 100 niL of 10% potassium hydrogen carbonate solution are added dropwise. The reaction mixture is stirred for 3 h, the organic phase dried over Na 2
SO
4 and evaporated down. Yield: 74 g of a compound Z7b (yellow oil) 20 18.6 g of the compound Z7b are dissolved in 200 mL glacial acetic acid and at 60*C combined batchwise with 20.0 g iron powder. The mixture is stirred for 2 h at 60*C and then suction filtered through cellulose. The residue is dissolved in ethyl acetate and washed with water and concentrated ammonia. The organic phase is dried over Na 2
SO
4 and evaporated down. The residue is crystallised from diethyl ether. 25 Yield: 9.8 g of a compound Z7c (colourless crystals) 17.0 g of the compound Z7c and 7 mL (0.1 mol) methyl iodide are dissolved in 200 mL dimethylacetamide and at -5*C combined with 4.0 g (0.1 mol) of sodium hydride as a 60% dispersion in mineral oil. The reaction mixture is stirred for 30 min and then 30 poured onto 300 mL ice water. The precipitate formed is suction filtered and stirred with petroleum ether. Yield: 14.8 g of a compound Z7d (beige crystals) WO 2006/018182 PC'T/EI'200'5/008623 67 0.9 g of the compound Z7d and 1.5 g (9 mmol) 4-arnino-3-methoxybenzoic acid are heated to 210*C for 30 min. After cooling the residue is stirred with ethyl acetate and the precipitate obtained is suction filtered. 5 Yield: 1.2 g of a compound Z7 (grey crystals) The following acids can, for example, be prepared analogously to the methods of synthesis hereinbefore described. N ~-N"' HN N HN N 0 OH 0 OH 10 Z8 Z9 N 0 HN N N HN N N 0 OH 0 OH Z10 Z11 is Synthesis of the amino components L-R5 The following amines, WO 2006/018182 P1CT/EP2005/008623 68 1,1-dimethyl-2-dimethylamino-1-yl-ethylamine and 1,1-dinethyl-2-piperidin-1-yl ethylamine, H12N -H2N
-N
may be obtained as follows. The compounds may be prepared according to the following references: (a) S. Schuetz et al. Arzneimittel-Forschung 1971, 21, 739-763, (b) V. M. Belikov et al.Tetrahedron 1970, 26, 1199-1216 and (c) E.B. Butler and McMillan J. Amer. Chem. Soc. 1950, 72, 2978. 10 Other amines can be prepared as follows, in a modified manner compared with the literature described above. 1.1-dimethyl-2-morpholin-1-yl-ethylamine 15
H
2 N 8.7 mL morpholine and 9.3 mL 2-nitropropane are prepared while cooling the reaction with ice, 7.5 mL formaldehyde (37%) and 4 mL of a 0.5 mol/L NaOH solution are slowly added dropwise (<10*C). Then the mixture is stirred for lh at 25*C and 1h at 20 50*C. The solution is treated with water and ether and the aqueous phase is extracted 3x with ether. The combined organic phase is dried over NaSO4 and combined with HCI in dioxane (4mol/l), the precipitate formed is suction filtered. Yield: 21.7 of white powder 5 g of the white powder are dissolved in 80 mL methanol and with the addition of 2 g RaNi treated with hydrogen at 35*C and 50 psi for 40 minutes. This yields 3.6 g of 1,1 25 dimethyl-2-morpholin- 1 -yl-ethylamine.
WO 2006/018182 PC'r/EI'2005/008623 69 The following amines can be prepared analogously. 1,1- dimethyl-N-methvlpiperazin-1-yl-ethylamine
H
2 N
N-
5 1.1-dimethyl-2-pyrrolidin-1-yl-ethvlamine
H
2 N N 1.3 -DIMORPH OLIN-2-AMINO-PROPANE
NH
2 N N 10 5 g of 1,3 Dimorpholine-2-nitropropane obtained from Messrs. Aldrich is dissolved in 80 mL methanol and treated with hydrogen for 5.5 h at 30*C and 50 psi with the addition of 2 g RaNi. This yields 4.2 g of 1,3 dimorpholin-2-amino-propane. 4-Aminobenzylmorpholine
H
2
N
WO 2006/018182 PCI/EP2005/008623 70 The preparation of this amine is described in the following reference: S. Mitsuru et al. J. Med. Chem. 2000, 43, 2049-2063. 5 4-amino- 1-tetrahydro-4H-pyran-4-yl-piperidine
H
2 N N O 20 g (100 mmol) of 4-tert-butyloxycarbony-aminopiperidine are dissolved in 250 mL
CH
2
CI
2 and stirred for 12 h at RT with 10 g (100 mmol) tetrahydro-41--pyran-4-one and 42 g (200 mmol) NaBH(OAc) 3 . Then water and potassium carbonate are added, the 10 organic phase is separated off, dried and the solvent is eliminated in vacuo. The residue is dissolved in 200 mL CH 2
CI
2 and stirred for 12 h at RT with 100 mL trifluoroacetic acid. The solvent is eliminated in vacuo, the residue taken up with CHC 3 and evaporated down again, then taken up in acetone and the hydrochloride is precipitated with ethereal HCL. Yield: 14.3 g (56%). 15 Cis- and trans-4-morpholino-cyclohexylamine
H
2 N .-.
O
H
2 N 20 3.9 g (30 mmol) of 4-dibenzylcyclohexanone are dissolved in 100 mL of CH 2
CI
2 and stirred for 12 h at RT with 3.9 g (45 mmol) of morpholine and 9.5 g (45 mmol) NaBH(OAc) 3 . Then water and potassium carbonate are added, the organic phase is separated off, dried and the solvent is eliminated in vacuo. The residue is purified through a silica gel column (about 20 mL silica gel; about 500 mL of ethyl acetate 90/ 25 methanol 10 + 1% concentrated ammonia). The appropriate fractions are evaporated down in vacuo. Yield: 6.6 g (60%) of cis-isomer and 2 g (18%) of trans-isomer.
WO 2006/018182 PCT/EP'2005/008623 71 Alternatively, the trans-dibenzyl-4--norpholino-cyclohexylamine may be prepared by the following method: 33 g (112 mmol) of 4-dibenzylcyclohexanone are dissolved in 300 mL MeOH, 5 combined with 17.4 g (250 mmol) of hydroxylamine hydrochloride and stirred for 4 h at 60*C. The solvent is evaporated down in vacuo, combined with 500 mL water and 50 g potassium carbonate and extracted twice with 300 mL of dichloromethane. The organic phase is dried, evaporated down in vacuo, the residue is crystallised from petroleum ether, dissolved in 1.5 L of EtOH and heated to 70'C. 166 g of sodium are added 10 batchwise and the mixture is refluxed until the sodium dissolves. The solvent is eliminated in vacuo, the residue combined with 100 mL water and extracted twice with 400 mL of ether. The organic phase is washed with water, dried, evaporated down in vacuo and the trans isomer is isolated using a column (about 1.5 L silica gel; about 2 L of ethyl acetate 80/ methanol 20 + 2 % concentrated ammonia). Yield: 12.6 g (41.2%). 15 6.8 g (23 mmol) of trans-1-amino-4-dibenzylaminocyclohexane is dissolved in 90 mL of DMF and stirred for 8 h at 100*C with 5 mL (42 mmol) of 2,2'-dichloroethyl ether and 5 g of potassium carbonate. After cooling 30 mL of water is added, the precipitated crystals are suction filtered and purified through a short column (about 20 mL silica gel, 20 about 100 mL ethyl acetate). The residue is crystallised from methanol and concentrated HCI as the dihydrochloride. Yield: 7.3 g (72.4%). Trans-4-morpholino-cyclohexylamine 25 7.2 g (16.4 mmol) of trans-dibenzyl-4-morpholino-cyclohexylamine are dissolved in 100 mL of MeOH and hydrogenated on 1.4 g of Pd/C (10%) at 30-50'C. The solvent is eliminated in vacuo and the residue is crystallised from ethanol and concentrated HCl. Yield: 3.9 g (93%). 30 The cis isomer may be prepared analogously. Cis- and trans-4-piperidino-cyclohexylamine WO 2006/018182 PCI'/EI'2005/008623 72
H
2 N
H
2 N Trans-dibenzyl-4-piperidino-cyclohexylamine 5 2.0 g (6.8 mm6l) of trans-1amino-4-dibenzylaminocyclohexane (see Example 2) is dissolved in 50 mL DMF and stirred for 48 h at RT with 1.6 g (7 mmol) of 1,5-dibromopentane and 2 g of potassium carbonate. The mixture is cooled, combined with water, extracted twice with 100 mL of dichloromethane, dried and the solvent is eliminated in vacuo. The residue is purified over a column ( about 100 mL silica gel, 10 about 500 mL ethyl acetate 80/meth-nol 20 +1% concentrated ammonia). The desired fractions are evaporated down in vacuo and crystallised from petroleum ether. Yield: 1.2 g (49%). Trans-4-piperidino-cyclohexylamine 15 1.7 g (4.8 mmol) of trans-dibenzyl-4-piperidino-cyclohexylamine are dissolved in 35 mL MeOH and hydrogenated on 350 mg of Pd/C (10%) at 20 0 C. The solvent is eliminated in vacuo and the residue crystallised from ethanol and concentrated HCL. Yield: 1.1 g (78%). 20 The cis isomer may be prepared analogously. Cis- and trans-4-(4-phenyl-piperazin-1-yl)-cyclohexylamine 2 -H 25 4.1 g (25.3 mmol) of 4-dibenzylcyclohexanone is dissolved in 50 mL of dichloromethane and stirred for 12 h at RT with 7.4 g (25.3 mmol) of WO 2006/018182 PCT/E P12005/008623 73 N-phenylpyperazine and 7.4 g (35 mnol) of NaBH(OAc) 3 . Then water and potassium carbonate are added, the organic phase is separated off, dried and the solvent is eliminated in vacuo. The residue is purified over a silica gel column (ethyl acetate 80/ methanol 20 + 0.5% concentrated ammonia). Yield: 1.7 g (15.8%) of cis-isomer and s 0.27 (2.5%) of trans-isomer. Trans-4-(4-phenyl-piperazin-1-yl)-cyclohexylamine 270 mg (0,61 mmol) of trans-dibenzyl-[4-(4-phenyl-piperazin-1-yl)-cyclohexyl]-amine 10 are dissolved in 5 mL MeOH and hydrogenated on 40 mg of Pd/C (10%) at 20-30 *C. The solvent is eliminated in vacuo and the residue crystallised from ethanol and concentrated HCL. Yield: 110 mg (69%). The cis isomer may be prepared analogously. 15 Cis- and trans-4-(4-cyclopropylmethyl-piperazin-1-yl)-cyclohexylamine HN N H 2 N N N 9.8 g (33.4 mmol) of 4-dibenzylcyclohexanone is dissolved in 100 mL dichloromethane 20 and stirred for 12 h at RT with 5.6 g (40 mmol) of N-cyclopropylmethylpiperazine and 8.5 g (40 mmol) of NaBH(OAc) 3 . Then water and potassium carbonate are added, the organic phase is separated off, dried and the solvent is eliminated in vacuo. The residue is purified over a silica gel column (about 50 mL silica gel, about 3 L ethyl acetate 95/ methanol 5 + 0.25% concentrated ammonia. The appropriate fractions are evaporated 25 down in vacuo. The faster eluting cis compound crystallised from ethyl acetate. The trans-compound is crystallised from ethanol + concentrated HCL. Yield: 8.5 g (61%) cis-isomer and 2.2 (13%) trans-isomer. cis-4-(4-cyclopropylmethyl-piperazin-1-yl)-cyclohexylamine WO 2006/018182 PC'I'/EP2005/008623 7-4 8.5 g (20 mmol) of cis-dibenzyl-[4-( 4 -cyclopropylmethyl-piperazin-1-yl)-cyclohexyl] amine are dissolved in 170 mL MeOH and hydrogenated on 1.7 g Pd/C (10%) at 30-50 'C. The solvent is eliminated in vacuo and the residue is crystallised from ethanol and s concentrated HCl. Yield: 4.4 g (91%). The trans-isomer may be prepared analogously. Synthesis of the Examples 10 Example 152 0.15g of the compound Z1O, 0.14 g TBTU, 0.13 mL DIPEA are dissolved in dichloromethane and stirred for 20 minutes at 25'C. Then 90 yAL 1-(3-aminopropyl)-4 Is methylpiperazine is added and stirred for a further 2 hours at 25*C. The solution is then diluted with dichloromethane and extracted with water. The product is precipitated by the addition of petroleum ether, ether and ethyl acetate to the organic phase. Yield: 0.16 g of beige solid. 20 Example 164 0.10g of the compound ZI0, 0.1 g TBTU, 0.08 mL DIPEA are dissolved in 4 mL dichloromethane and stirred for 20 minutes at 25*C. Then 44 pL dimethylaminopropylamine are added and stirred for a further 2 hours at 25"C. The 25 solution is then diluted with dichloromethane and extracted with water. The product is precipitated by the addition of petroleum ether, ether and acetone to the organic phase. Yield: 0.08 g yellow solid. Example 242 30 0.15g of the compound Z1O, 0.14 g TBTU, 0.13 mL DIPEA are dissolved in 5 mL dichloromethane and stirred for 20 minutes at 25"C. Then 75 pL 1-(2-aninoethyl) WO 2006/018182 PCI/EP2005/008623 75 piperidine are added and stirred for a further 2 hours at 25'C. The solution is then diluted with dichloromethane and extracted with water. The product is precipitated by the addition of petroleum ether, ether and ethyl acetate to the organic phase. Yield: 0.14 g yellow solid. 5 Example 188 0.1 g of the compound Z2, 0.09 g TBTU, 0.05 mL DIPEA are dissolved in 15 mL dichloromethane and stirred for 20 minutes at 25*C. Then 33 mg 1-methyl-4 10 aminopiperidin are added and the mixture is stirred for a further 3 hours at 25*C. The solution is extracted with 20 mL water, then evaporated down in vacuo. The product is crystallised using ether. Yield: 0.047 g of white crystals. Example 203 15 0.1 g of the compound Z2, 0.09 g TBTU, 0.5 mL DIPEA are dissolved in 15 mL dichloromethane and stirred for 30 minutes at 25'C. Then 50 mg 4-amino-1 benzylpiperidin are added and the mixture is stirred for a further 3 hours at 25*C. The solution is extracted with 20 mL water, then evaporated down in vacuo. Then the 20 residue is chromatographed over silica gel and the isolated product is crystallised with ether. Yield: 0.015 g of white crystals. Example 94 25 0.17 g of the compound ZI, 0.19 g TBTU, 0.11 mL DIPEA are dissolved in 50 mL dichloromethane and stirred for 30 minutes at 25*C. Then 63 mg of 1-methyl-4 aminopiperidine are added and the mixture is stirred for a further 17 hours at 25*C. 50 mL of water and 1 g of potassium carbonate are added to the solution and the organic phase is separated off using a phase separation cartridge, then evaporated down in 30 vacuo. Then the product is purified by silica gel chromatography and the purified product is crystallised with ether. Yield: 0.1 g of white crystals.
WO 2006/018182 PCT/EP2005/008623 76 Example 95 0.17 g of the compound Z1, 0.19 g TBTU, 0.11 mL DIPEA are dissolved in 50 mL dichloromethane and stirred for 30 minutes at 25'C. Then 77 mg of exo-3-$-ainino s tropane are added and the mixture is stirred for a further 17 hours at 25*C. 50 mL of water and 1 g of potassium carbonate are added to the solution and the organic phase is separated off using a phase separation cartridge, then evaporated down in vacuo. Then the product is purified by silica gel chromatography and the purified product is crystallised with ether. Yield: 0.03 g of white crystals. 10 Example 46 0.15 g of the compound Z3, 0.12 g TBTU, 0.12 mL DIPEA are dissolved in 5 mL dichloromethane and stirred for 30 minutes at 25"C. Then 50 mg 1-methyl-4 15 aminopiperidin are added and the mixture is stirred for a further 2.5 hours at 25*C. The solution is then extracted with water and then evaporated down. The residue is dissolved in warm ethyl acetate and crystallised from ether and petroleum ether. Yield: 0.025 g of white crystals. 20 Example 80 0.2 g of the compound Z8, 0.2 g of TBTU, 0.1 mL of DIPEA are dissolved in 10 mL dichloromethane and stirred for 30 minutes at 25*C. Then 100 mg of 1-methyl-4 aminopiperidine are added and the mixture is stirred for a further 17 hours at 254C. The 25 solution is then extracted with a dilute potassium carbonate solution and evaporated down. The residue is crystallised using ether. Yield: 0.12 g of white crystals. Example 190 30 0.2 g of compound Z8, 0.2 g of TBTU, 0.3 miL of DIPEA are dissolved in 5 mL dichloromethane and stirred for lh at 25*C. Then 0.13 g of 4-amino-1-benzylpiperidine are added and the mixture is stirred for a further hour at 25'C. The solution is then WO 2006/018182 PCT/EP2005/008623 77 diluted with 10 mL methylene chloride and extracted with 20 mL water. Then the product is purified over silica gel and crystallised from ethyl acetate and ether. Yield: 0.23 g of the compound Z8. 0.23 g of the benzylamine Z8 are dissolved in 10 mL methanol, combined with 50 mg 5 of Pd/C and hydrogenated under 3 bar for 3h at 25'C. By adding petroleum ether and ethyl acetate white crystals are produced. These are chromatographed over silica gel and crystallised from ethyl acetate and ether. Yield: 0.075 g of white crystals. Example 196 10 0.lg of compound Z10, 0.09 g of TBTU, 0.3 mL of DIPEA are dissolved in 4 mL of dichloromethane and stirred for 20 minutes at 25"C. Then 67 mg xx amine is added and stirred for a further 2 hours at 25"C. The solution is then diluted with dichloromethane and extracted with water. It is then chromatographed over silica gel and the residue is is dissolved in acetone, combined with ethereal HC] and the precipitate formed is isolated. Yield: 0.09 g light yellow solid. Example 166 20 0.1 g of the compound Z1O, 0.11 g of TBTU, 0.14 mL of DIPEA are dissolved in 2 mL dimethylformamide and stirred for 3h at 50*C. Then 55 mg of 4-morpholinomethylphenylamine is added. The reaction mixture is then cooled to ambient temperature within 17 h. Then the dimethylformamide is eliminated in vacuo, the residue is taken up in dichloromethane and extracted with water. It is then 25 chromatographed over silica gel and the product crystallised from ethyl acetate and ether. Yield: 0.06 g yellowish crystals. Example 81 30 0.2 g of the compound Z4, 0.2 g of TBTU, 0.1 mL of DIPEA are dissolved in 10 mL dichloromethane and stirred for 30 minutes at 25"C. Then 0.1 g of 1-methyl-4 aminopiperidine are added and the mixture is stirred for a further 17 hours at 25"C. The WO 2006/018182 PCT/EP2005/008623 78 solution is then extracted with aqueous potassium carbonate solution and then evaporated down. The product is crystallised using ether. Yield: 0.16 g of white crystals. s Example 162 0.1 g of the compound Z5, 0.07 g of TBTU, 0.15 mL of DIPEA are dissolved in 5 mL dichloromethane and stirred for 20 minutes at 25*C. Then 0.04 g 1-methyl-4 aminopiperidine are added and the mixture is stirred for a further 2 hours at 25*C. The 10 solution is then diluted with 15 mL dichloromethane and extracted with 20 mL water. The residue is dissolved in MeOH and acetone, combined with 1 mL ethereal HC and evaporated down. A crystalline product is produced using ether, ethyl acetate and a little MeOH. Yield: 0.1 g of white crystals. 15 Example 88 0.1 g of the compound Z6, 0.12 g of TBTU, 0.12 mL of DIPEA are in 10 mL dichloromethane dissolved and stirred for 30 minutes at 25"C. Then 0.04 g of 1-methyl 4-aminopiperidine are added and the mixture is stirred for a further 2 hours at 25*C. The 20 solution is then diluted with 10 mL dichloromethane and extracted with 10 mL water. A crystalline product is produced using ether, ethyl acetate and petroleum ether. Yield: 0.6 g of white crystals. Example 89 25 0.1 g of the compound Z6, 0.08 g of TBTU, 0.08 mL of DIPEA are dissolved in 10 mL dichloromethane and stirred for 30 minutes at 25*C. Then 37 puL g N,N dimethylneopentanediamine are added and the mixture is stirred for a further 2 hours at 25*C. The solution is then diluted with 10 mL dichloromethane and extracted with 10 30 mL water. The product is then chromatographed over silica gel and crystallised from ethyl acetate, ether and petroleum ether. Yield: 0.005 g of white crystals.
WO 2006/018182 PCT/EP2005/008623 79 Example 26 0.15 g of the compound Z7, 0.16 g of TBTU, 1 mL of DIPEA are dissolved in 5 mL dichloromethane and stirred for 30 minutes at 25"C. Then 0.1 g s 4-morpholinocyclohexylamine are added and the mixture is stirred for a further 17 hours at 25*C. The residue is then combined with 10 mL of 10% potassium carbonate solution, the precipitate is isolated and washed with water. It is then dissolved in dichloromethane and evaporated down again. The product is crystallised from ethyl acetate. Yield: 0.1 g of white crystals. 10 Example 9 150 mg of the compound Z9 and 93 mg of amine are dissolved in 5 mL dichloromethane and stirred with 160 mg of TBTU and 1 mL of DIPEA for 12 h at RT. 15 The solvent is eliminated in vacuo, the residue is combined with 10 mL of 10% potassium carbonate solution. The precipitate is suction filtered, washed with water, taken up in dichloromethane, dried and the solvent eliminated in vacuo. The residue is crystallised from ethyl acetate. Yield: 82.0 mg. 20 Example 16 150 mg of the compound Z8 and 73 mg of trans-4-piperidino-cyclohexylamine are dissolved in 5 mL dichloromethane and stirred with 160 mg (0.50 mmol) of TBTU and 1 mL of DIPEA for 12 h at RT. The solvent is eliminated in vacuo, the residue is 25 combined with 10 mL of 10% potassium carbonate solution. The precipitate is suction filtered, washed with water, taken up in dichloromethane, dried and the solvent eliminated in vacuo. The residue is crystallised from ethyl acetate. Yield: 87.0 mg. 30 100 mg of the compound Z9 and 42 mg of 3-amino-1-ethyl-pyrolidine are dissolved in 10 mL dichloromethane and stirred with 90 mg of TBTU and 0.5 mL of DIPEA for 12 h at RT. The solvent is eliminated in vacuo, the residue is combined with 10 ml- of 10% WO 2006/0 18182 PCT/EP2005/008623 potassium carbonate solution. The precipitate is suction filtered, washed with water, taken up in dichloromethane, dried and the solvent is eliminated in vacuo. The residue is crystallised from ethyl acetate/petroleum ether. Yield: 24.0 mg. 5 Example 120 100 mg of the compound Z11 and 73 mg of 4-amino-ltetrahydro-4H-pyran-4-yl piperidine are dissolved in 10 mL dichloromethane and stirred with 90 mg of TBTU and 0.5 mL of DIPEA for I h at RT. The solvent is eliminated in vacuo, the residue is to combined with 10 mL of 10% potassium carbonate solution. The precipitate is suction filtered, washed with water, taken up in dichloromethane, dried and the solvent is eliminated in vacuo. The residue is crystallised from ethyl acetate/petroleum ether. Yield: 89 mg. 15 Example 212 150 mg of the compound Z5 and 150 mg of trans-4-(4-cyclopropylmcthyl-piperazin-l yl)-cyclohexylamine (as the hydrochloride) are dissolved in 5 mL of dichloromethane and stirred with 160 mg of TBTU and 2 mL of DIPEA for 2 h at RT. The solvent is 20 eliminated in vacuo, the residue is combined with 10 mL of 10% potassium carbonate solution. The precipitate is suction filtered, washed with water, taken up in dichloromethane, dried and the solvent eliminated in vacuo. The residue is purified over a column (20 mL silica gel, 300 mL ethyl acetate 90/ methanol 10 + 2% concentrated ammonia). The appropriate fractions are evaporated down in vacuo and crystallised 25 from ethyl acetate. Yield: 140 mg. Example 232 390 mg of the compound ZI 1 and 240 mg of trans-4-(4-tbutyloxycarbonyl-piperazin-1 30 yl)-cyclohexylamine are dissolved in 2.5 mL of NMP and stirred with 482 mg of TBTU and 1 mL triethylamine for 2 h at RT. Then 100 mL of water and 200 mg of potassium carbonate are added, the precipitate is suction filtered, washed with water and purified WO 2006/018182 PCT/FIP2005/008623 81 through a silica gel column. The appropriate fractions are evaporated down in vacuo, dissolved in 2 mL dichloromethane, combined with 2 mL of trifluoroacetic acid and stirred for 2 h at RT, combined with another 100m] of water and 200 mg potassium carbonate and the precipitate is suction filtered and washed with water. Then the s precipitate is purified through a silica gel column. The appropriate fractions are evaporated down in vacuo and the residue is crystallised from ethanol and concentrated hydrochloric acid. Yield: 95 mg. Example 213 10 60 mg of the compound of Example 232 is dissolved in 10 mL ethyl acetate and stirred with 1 mL of acetic anhydride and 1 mL of triethylamine for 30 min. at RT. The solvent is eliminated in vacuo, the residue combined with water and ammonia, the crystals precipitated are suction filtered and washed with water and a little cold acetone. is Yield: 40 mg. Example 218 1.2 g of the compound Z9 and 0.5g of 1,4-dioxaspiro[4.5]dec-8-ylamine 20 were dissolved in 20 mL dichloromethane and stirred with 1.28 g of TBTU and 4 mL of triethylamine for 12 h at RT. Then 50 mL of water and 0.5 g of potassium carbonate are added, the organic phase is separated off, dried and evaporated down in vacuo. The residue is crystallised from ethyl acetate, combined with 25 mL of 1 N hydrochloric acid and 20 mL of methanol and stirred for 30 min at 50*C. The methanol is eliminated 25 in vacuo, the precipitate is suction filtered, washed with water and dried. The residue is taken up in 20 mL dichloromethane, stirred with 0.5 g of thiomorpholine and 0.5 g of NaBH(OAc) 3 for 12 h at RT .Then water and potassium carbonate are added, the organic phase is separated off, dried and the solvent is eliminated in vacuo. The residue is purified over a silica gel column. The appropriate fractions are 30 evaporated down in vacuo and the hydrochloride is precipitated with ethereal HCL. Yield: 86 mg of trans-isomer; amorphous powder.
C iNRPostblDCCDAR\389I776-DOC-22/09/2011 - 82 Example 187 200 mg of the compound Z3 in 5 mL dichloromethane is combined with 0.1 mL of 5 diisopropylethylamine and 180 mg of TBTU and stirred for 30 min. Then 191 mg of 4 (4-methyl-piperazin- I -yl)-phenylamine are added and the mixture is stirred overnight. The reaction mixture is combined with water and the aqueous phase extracted with dichloromethane. The combined organic phases are dried over Na 2
SO
4 and evaporated down. The residue is purified by column chromatography (eluant: 10 dichloromethane/methanol 100:7). Yield: 128 mg (light yellow crystals). The compounds of Formula (I) listed in Table 1, inter alia, are obtained analogously to the procedure described hereinbefore. The abbreviations Xi, X 2 , X 3 , X 4 and X 5 used in Table 1 in each case denote a link to a position in the general Formula shown 15 under Table I instead of the corresponding groups R', R 2 , R 3 , R 4 and L-R'. Throughout this specification and the claims which follow, unless the context requires otherwise, the word "comprise", and variations such as "comprises" and "comprising", will be understood to imply the inclusion of a stated integer or step or group of integers or steps but 20 not the exclusion of any other integer or step or group of integers or steps. The reference in this specification to any prior publication (or information derived from it), or to any matter which is known, is not, and should not be taken as an acknowledgment or admission or any form of suggestion that that prior publication (or information derived from it) 25 or known matter forms part of the common general knowledge in the field of endeavour to which this specification relates.
WO 20061(018182 PCT/EI'2005/008623 E a I: CCC Z-4J a:x
OE
WO 2006/018182 PCIAEP200S5/008623 84 3rr! ~cc: WO 2006/018182 PC17EP20O5/0O8623 IND -, ~ ir 000 WO 2006/018182 PCT/EIP2005/008623 86 00 >Z- 0 /0 >a--a WO 2006/018182 PCT/EP12005/008623 87 C- 0 0.-0 0-0 0 L m 3:mT oo__ C7%__ I I I I WO 2006/018182 PCT/EP2005/008623 88 U-0 ,0- ,0- XI0 I" / / >a--a>4i- >C0 cc:" IdI I1 C1 N IqC1 WO 20061018182 PCT7I1I2005/008623 89 66 0e- 0 000 WO 2006/018182 PCTJEP2005/008623 90 3r C.)~L C. C) >4-10 >f-( x-C 3r I II cn qr0 WO 2006/018182 PCTIEP2005/008623 91 :Il 00
[-(-I--
WO 2006/018182 PCTIEP2005/008623 92 X"I cc a: a: cc 0 > > WO 2006/018182 PCTIEP2005/008623 93 cc cc cc cc cc A i§ tr x- /e oo I Ii Cin 0 C WO 2006/018182 PCT[EP2005/008623 94 23: Cf'0 '0 t10 '0 0 WO 2006/018182 PCT/EP20051008623 95 U-0 Kcc M- _ __c 00 0%0'-n' WO 2006/018182 PCTIEP200IOO8623 96 00_ / '0 cc a: cc cc Il~ CO WO 2006/018182 PCTIEP2005/008623 97 0 0) 0 0r cc c0rC WO 2006/018182 PCTIEP2005/008623 98
/W
WO 2006/018182 PCT/EP2005/008623 99 0 0 /0 I =l 00 CA 0 WO 2006/018182 PCT/EP2005/008623 100 _ _ _ -oo ~ I1 00 00 Co0o 00 WO 2006/018182 PCT/EP2005/008623 101 C.X, Nr- XA > I 000 00 00 C WO 2006/018182 PCT1EP2005/008623 102 > *0 0 C.)- 01- C.)o Ir.)r a:ira cc cc / __>___ 5e k4 I WO 2006/018182 PCTIEP2005/008623 103 >e-0 00 - WO 2006/018182 PCTIEP2005/008623 104 t iti/ a WO 2006/018182 PCTIEP2005/008623 105 0 10 0 0 C1 4 WO 2006/018182 PCTIEP2005/008623 106 3r cc cc WO 2006/018182 PCT/EP2005/008623 107 r-o --oo >-o 00 0 C1 4 -*4 WO 2006/018182 PCTIEP2005/008623 108 ___ __ _ -o- xo-o x-- D >C-, --- 11 / CCQ a: x- X X x- Al. Ae
N
WO 2006/018182 PCT/EP2005/008623 109 0 0 OO CO -QO KOJ C V)) WO 2006/018182 PCT/EP2005/008623 110 00- <>o-o x- K)< <)O / cc = = = I> cnr M Cf) en-4 WO 2006/018182 PCT/EP2005/008623 0 0 0 >r-g 00 ON C)-4 4 cn O I WO 2006/018182 PCTIEP200S/008623 112 0 CC( a: a: c cc a: xv ___ __ _ _ _ _ _ _ _ _ _ __ __ WO 2006/018182 PCTIEP2005/008623 113 0 00 ON WO 2006/018182 PCT[EP200S!008623 114 jx I 3f a: _c a: I I I 3: tn ' IdtInInI WO 2006/018182 PC1'1EP2005/008623 115 ifK-t 0 0 000
V.)
WO 2006/018182 PCTIEP2005/008623 116 cr c cccc, cn r- WO 2006/018182 PCT/EP2005/008623 117 -P~K ~2 K 9-C( 3: 3: 2: : 3: 00 C0 WO 2006/018182 PCTIEP200S/008623 118 e-I a: cc WO 2006/018182 PCTIEP2005/008623 119 -0P 10 0 0-0 00 0 t- 00 0 00 -4-4 -4 -4 1-4 WO 2006/018182 PCT/EP2005/008623 120 %nI I t o 00 00 00 00E WO 2006/0 18182 PCT/EP2005/008623 121 0/ ccir I I 00 00- WO 2006/018182 PCT/EP2005/008623 122 &1 -I ~~-O >lD __ cc x0cc _ _ _ _ _ I II WO 2006/018182 PCU/EP2005/008623 123 0 0 x cc 000 WO 2006/018182 PCT[P2005i008623 124 X, O-r C5 U-0/ I fI a:I 0 0 WO 2006/018182 PCT/EP2005/008623 125 00 0N -C4 c'4 C,4 WO 2006/018182 PCTIEP2005/008623 126 0z >- XI0 > >c--cJ >L-\ ccc xc N N C9C= WO 2006/018182 PCT[EP200S/008623 127 >eGO OQ oO & I :3: -- = I -14 e4q WO 2006/018182 PCTIEP2005/008623 128 X 0/0 >e6' O-C' I I I 3r4 WO 2006/018182 PCTIEP200S/008623 129 __OfV - 3rO 3r 3 >o
>
WO 2006/018182 PCT/EP2005/008623 130 >c cc cco Cf-) ~-KD ____ _ __ ___ ____ WO 2006/018182 PCTIEP2005/008623 131 cn cn Iq N WO 2006/018182 PCTIEP2005/008623 132 Lcc: C14

Claims (4)

1. A pharmaceutical composition comprising effective amounts of: (i) A compound 1 of Formula (I) HN K N R R4 HN3 O NH n R M Rm a wherein R', R 2 which may be identical or different, denote hydrogen or optionally substituted C1-C6-allcyl, or R' and R 2 together denote a 2- to 5-membered alkyl bridge which may contain 1 to 2 heteroatoms, R 3 denotes hydrogen or a group selected from among optionally substituted CI-C 1 2 -alkyl, C 2 -C 1 2 -alkenyl, C2-C 1 2 -alkynyl and C 6 -C 14 -aryl, or a group selected from among optionally substituted and/or bridged C 3 -C 1 2 cycloalkyl, C 3 -C 1 2 -cycloalkenyl, C7-C2-polycycloalkyl, C 7 -C 1 2 - WO 2006/018182 PCT/EP2005/008623 134 polycycloalkenyl, C 5 -C 1 2 -spirocycloalkyl, C3-C2-heterocycloalkyl which contains I to 2 heteroatoms, and C 3 -C12-heterocycloalkenyl which contains 1 to 2 heteroatoms, or 5 R' and R3 or R 2 and R 3 together denote a saturated or unsaturated C 3 -C 4 -alkyl bridge which may contain 1 heteroatom, R 4 denotes a group selected from among hydrogen, -CN, hydroxy, -NR 6 R 7 and halogen, or 10 a group selected from among optionally substituted CI-C 6 -alkyl, C 2 -C 6 -alkenyl, C 2 -C 6 -alkynyl, C 1 -Cs-alkyloxy, C 2 -C 5 -alkenyloxy, C 2 -Cs-alkynyloxy, CI-C 6 alkylthio, CI-C 6 -alkylsulphoxo and C 1 -C 6 -alkylsulphonyl, L denotes a linker selected from among optionally substituted C2-Clo-alkyl, 15 C 2 -CIO-alkenyl, C 6 -C 1
4-aryl, -C 2 -C 4 -alkyl-C6-C4-aryl, -C 6 -C 14 -aryl-CI-C 4 -alkyl, optionally bridged C 3 -C 12 -cycloalkyl and heteroaryl which contains 1 or 2 nitrogen atoms, n denotes 0 or 1 20 m denotes 1 or 2 R 5 denotes a group selected from among optionally substituted morpholinyl, piperidinyl, piperazinyl, piperazinylcarbonyl, pyrrolidinyl, tropenyl, R 8 diketomethylpiperazinyl, sulphoxomorpholinyl, sulphonylmorpholinyl, 25 thiomorpholinyl, -NR 8 R 9 and azacycloheptyl, R 6, R7 which may be identical or different, denote hydrogen or C 1 -C 4 -alkyl, and R, R 9 denote unsubstituted nitrogen substituents at R 5 , which may be identical or different, denote either hydrogen or a group selected from among CI-C 6 -alkyl, 30 -Ci-C 4 -alkyl-C 3 -Cio-cycloalkyl, C 3 -Cio-cycloalkyl, C 6 -C4-aryl, -CI-C 4 -alkyl-C6-C 4 -aryl, pyranyl, pyridinyl, pyrimidinyl, CI-C 4 alkyloxycarbonyl, C 6 -C4-arylcarbonyl, C-C 4 -alkylcarbonyl, C6-C4- WO 2006/018182 PCT/EP2005/008623 135 arylmethyloxycarbonyl, C 6 -C 1 4 -arylsulphonyl, Ci-C 4 -alkylsulphonyl- and C 6 C 14 raryl-C-C 4 -alkylsulphonyl-, optionally in form of its tautomers, racemates, enantiomers, diastereomers and the mixtures thereof and optionally in form of the pharmacologically acceptable 5 acid addition salts, solvates, hydrates, polymorphs, physiologically functional derivatives or prodrugs thereof; and 10 (ii) at least one further chemotherapeutic or naturally occurring, semi synthetic or synthetic therapeutic agent 2; optionally in combination with one or more pharmaceutically acceptable excipients, and optionally adapted for a co-treatment with radiotherapy or radio-immunotherapy, in is the form of a combined preparation for simultaneous, separate or sequential use in the treatment of diseases involving cell proliferation, migration or apoptosis of cancer cells, or angiogenesis. 2. The pharmaceutical composition according to claim 1, wherein the compound 1 20 is selected from the group consisting of the compounds of Formula (I) shown in the following Table O HH H- N O NN N NN R R 4 Config. Example R' R 2 2 RR4 L-RR WO 2006/018182 PCT/EP2005/008623 136 27 H R CH 2 HaC CH 3 0 44 H R H 55 H R ) CH3 0 58 H R C 0.0 HY- OH jH 3 102 H R H3C CH3 103 H CH3 R 105 H 0% R OC 110 H R H HC CH, a 3 WO 2006/018182 PCT/EP2005/008623 137 115 H CH, R 133 H X CH R NCH, O o 134 H X CH 3 R O,'CH, 234 H X CHs R CH 3 HC CH, 240 H CH R O % KC c, wherein the abbreviations X 1 , X 2 , X 3 , X4 and Xs used in the Table in each case denote a link to a position in the general Formula shown in the Table instead of the corresponding groups R1, R 2 , R 3 , R 4 and L-RI. 5 3. A pharmaceutical composition according to claim 1 or 2, wherein the further chemotherapeutic or naturally occurring, semi-synthetic or synthetic therapeutic agent 2 is selected from the group consisting of synthetic small molecule VEGF receptor antagonists, small molecule growth factor receptor antagonists, inhibitors of the EGF 10 receptor and/or VEGF receptor and/or integrin receptors or any other protein tyrosine kinase receptors which are not classified under the synthetic small-molecules, inhibitors directed to EGF receptor and/or VEGF receptor and/or integrin receptors or any other WO 2006/018182 PCT/EP2005/008623 138 protein tyrosine kinase receptors, which are fusion proteins, compounds which interact with nucleic acids and which are classified as alkylating agents or platinum compounds, compounds which interact with nucleic acids and which are classified as anthracyclines, as DNA intercalators or as DNA cross-linking agents, including DNA minor-groove 5 binding compounds, anti-metabolites, naturally occurring, semi-synthetic or synthetic bleomycin type antibiotics, inhibitors of DNA transcribing enzymes, and especially the topoisomerase I or topoisomerase II inhibitors, chromatin modifying agents, nitosis inhibitors, anti-mitotic agents, cell-cycle inhibitors, proteasome inhibitors, enzymes, hormones, hormone antagonists, hormone inhibitors, inhibitors of steroid biosynthesis, io steroids, cytokines, hypoxia-selective cytotoxins, inhibitors of cytokines, lymphokines, antibodies directed against cytokines, oral and parenteral tolerance induction agents, supportive agents, chemical radiation sensitizers and protectors, photo-chemically activated drugs, synthetic poly- or oligonucleotides, optionally modified or conjugated, non-steroidal anti-inflammatory drugs, cytotoxic antibiotics, antibodies targeting growth 15 factors or their receptors, antibodies targeting the surface molecules of cancer cells, inhibitors of metalloproteinases, metals, inhibitors of oncogenes, inhibitors of gene transcription or of RNA translation or protein expression, complexes of rare earth elements, compounds which reduces the transport of hyaluronan mediated by one or more ABC transporters, and photo-chemotherapeutic agents. 20 4. A pharmaceutical combination according to claim 1 or 2, wherein the further chemotherapeutic or naturally occurring, semi-synthetic or synthetic therapeutic agent 2 is selected from the group consisting of a small molecule VEGF receptor antagonist such as vatalanib (PTK-787/ZK222584), SU-5416, SU-6668, SU-11248, SU-14813, 25 AZD-6474, AZD-2171, CP-547632, CEP-7055, AG-013736, IM-842 or GW-786034, a dual EGFR/HER2 antagonist such as gefitinib, erlotinib, CI-1033 or GW-2016, an EGFR antagonist such as iressa (ZD-1839), tarceva (OSI-774), PKI-166, EKB-569, HKI-272 or herceptin, an antagonist of the mitogen-activated protein kinase such as BAY-43-9006 or BAY-57-9006, a quinazoline derivative such as 4-[(3-chloro-4 30 fluorophenyl)amino]-6- { [4-(NN-dimethylamino)-1-oxo-2-buten-1-yl] amino} -7-((S) tetrahydrofuran-3-yloxy)-quinazoline or 4-[(3-chloro-4-fluoro-phenyl)amino]-6-{ [4 (homomorpholin-4-yl)-1-oxo-2-buten-1-yl]amino}-7-[(S)-(tetrahydrofuran-3-yl)oxy]- WO 2006/018182 PCT/EP2005/008623 139 quinazoline , or a pharmaceutically acceptable salt thereof, a protein kinase receptor antagonist which is not classified under the synthetic small molecules such as atrasentan, rituximab, cetuximab, Avastinrm (bevacizumab), IMC-1C1 1, erbitux (C 225), DC-101, EMD-72000, vitaxin, imatinib, a protein tyrosine kinase inhibitor which s is a fusion protein such as VEGFtrap, an alkylating agent or a platinum compound such as melphalan, cyclophosphamide, an oxazaphosphorine, cisplatin, carboplatin, oxaliplatin, satraplatin, tetraplatin, iproplatin, mitomycin, streptozocin, carmustine (BCNU), lomustine (CCNU), busulfan, ifosfamide, streptozocin, thiotepa, chlorambucil, a nitrogen mustard such as mechlorethamine, an ethyleneimine 10 compound, an alkylsulphonate, daunorubicin, doxorubicin (adriamycin), liposomal doxorubicin (doxil), epirubicin, idarubicin, mitoxantrone, amsacrine, dactinomycin, distamycin or a derivative thereof, netropsin, pibenzimol, mitomycin, CC-1065, a duocarmycin, mithramycin, chromomycin, olivomycin, a phtalanilide such as propamidine or stilbamidine, an anthramycin, an aziridine, a nitrosourea or a derivative 15 thereof, a pyrimidine or purine analogue or antagonist or an inhibitor of the nucleoside diphosphate reductase such as cytarabine, 5-fluorouracile (5-FU), pemetrexed, tegafur/uracil, uracil mustard, fludarabine, gemcitabine, capecitabine, mercaptopurine, cladribine, thioguanine, methotrexate, pentostatin, hydroxyurea, or folic acid, a phleomycin, a bleomycin or a derivative or salt thereof, CHPP, BZPP, MIPP, BAPP, 20 liblomycin, an acridine or a derivative thereof, a rifamycin, an actinomycin, adramycin, a camptothecin such as irinotecan or topotecan, an amsacrine or analogue thereof, a tricyclic carboxamide, an histonedeacetylase inhibitor such as SAHA, MD-275, trichostatin A, CBHA, LAQ824, or valproic acid, an anti-cancer drug from plants such as paclitaxel (taxol), docetaxel or taxotere, a vinca alkaloid such as navelbine, 25 vinblastin, vincristin, vindesine or vinorelbine, a tropolone alkaloid such as colchicine or a derivative thereof, a macrolide such as maytansine, an ansamitocin or rhizoxin, an antimitotic peptide such as phomopsin or dolastatin, an epipodophyllotoxin or a derivative of podophyllotoxin such as etoposide or teniposide, a steganacin, an antimitotic carbamate derivative such as combretastatin or amphetinile, procarbazine, a 30 proteasome inhibitor such as bortezomib, an enzyme such as asparaginase, pegylated asparaginase (pegaspargase) or a thymidine-phosphorylase inhibitor, a gestagen or an estrogen such as estramustine (T-66) or megestrol, an anti-androgen such as flutamide, WO 2006/018182 PCT/EP2005/008623 140 casodex, anandron or cyproterone acetate, an aromatase inhibitor such as aminogluthetimide, anastrozole, formestan or letrozole, a GNrH analogue such as leuprorelin, buserelin, goserelin or triptorelin, an anti-estrogen such as tamoxifen or its citrate salt, droloxifene, trioxifene, raloxifene or zindoxifene, a derivative of 170 s estradiol such as ICI 164,384 or ICI 182,780, aminoglutethimide, formestane, fadrozole, finasteride, ketoconazole, a LH-RH antagonist such as leuprolide, a steroid such as prednisone, prednisolone, methylprednisolone, dexamethasone, budenoside, fluocortolone or triamcinolone, an interferon such as interferon p, an interleukin such as IL-10 or IL-12, an anti-TNFa antibody such as etanercept, an immunomodulatory drug 1o such as thalidomide, its R- and S-enantiomers and its derivatives, or revimid (CC-5013), a leukotrien antagonist, mitomycin C, an aziridoquinone such as BMY-42355, AZQ or EO-9, a 2-nitroimidazole such as misonidazole, NLP-1 or NLA-1, a nitroacridine, a nitroquinoline, a nitropyrazoloacridine, a "dual-function" nitro aromatic such as RSU 1069 or RB-6145, CB-1954, a N-oxide of nitrogen mustard such as nitromin, a metal Is complex of a nitrogen mustard, an anti-CD3 or anti-CD25 antibody, a tolerance induction agent, a biphosphonate or derivative thereof such as minodronic acid or its derivatives (YM-529, Ono-5920, YH-529), zoledronic acid monohydrate, ibandronate sodium hydrate or clodronate disodium, a nitroimidazole such as metronidazole, misonidazole, benznidazole or nimorazole, a nitroaryl compound such as RSU-1069, a 20 nitroxyl or N-oxide such as SR-4233, an halogenated pyrimidine analogue such as bromodeoxyuridine, iododeoxyuridine, a thiophosphate such as WR-2721, a photo chemically activated drug such as porfimer, photofrin, a benzoporphyrin derivative, a pheophorbide derivative, merocyanin 540 (MC-540) or tin etioporpurin, an ant-template or an anti-sense RNA or DNA such as oblimersen, a non-steroidal inflammatory drug 25 such as acetylsalicyclic acid, mesalazin, ibuprofen, naproxen, flurbiprofen, fenoprofen, fenbufen, ketoprofen, indoprofen, pirprofen, carprofen, oxaprozin, pranoprofen, miroprofen, tioxaprofen, suprofen, alminoprofen, tiaprofenic acid, fluprofen, indomethacin, sulindac, tolmetin, zomepirac, nabumetone, diclofenac, fenclofenac, alclofenac, bromfenac, ibufenac, aceclofenac, acemetacin, fentiazac, cidanac, etodolac, 30 oxpinac, mefenamic acid, meclofenamic acid, flufenamic acid, nifluminic acid, tolfenamic acid, diflunisal, flufenisal, piroxicam, tenoxicam, lornoxicam, nimesulide, meloxicam, celecoxib, rofecoxib, or a pharmaceutically acceptable salt of a non- WO 2006/018182 PCT/EP2005/008623 141 steroidal inflammatory drug, a cytotoxic antibiotic, an antibody targeting the surface molecules of cancer cells such as apolizumab or 1D09C3, an inhibitor of metalloproteinases such as TIMP-1 or TIMP-2, Zinc, an inhibitor of oncogenes such as P53 and Rb, a complex of rare earth elements such as the heterocyclic complexes of s lanthanides, a photo-chemotherapeutic agent such as PUVA, an inhibitor of the transcription factor complex ESX/DRIP130/Sur-2, an inhibitor of HER-2 expression, such as the heat shock protein HSP90 modulator geldanamycin and its derivative 17 allylaminogeldanamycin or 17-AAG, a compound which reduces the transport of hyaluronan mediated by one or more ABC transporters selected from a P-glycoprotein 1o (P-gp) inhibitor molecule or inhibitor peptide, an MRP1 inhibitor, an antibody directed against and capable of blocking the ABC transporter, an antisense oligomer, iRNA, siRNA or aptamer directed against one or more ABC transporters, or a therapeutic agent selected from IM-842, tetrathiomolybdate, squalamine, combrestatin A4, TNP-470, marimastat, neovastat, bicalutamide, abarelix, oregovomab, mitumomab, TLK-286, 15 alemtuzumab, ibritumomab, temozolomide, denileukin diftitox, aldesleukin, dacarbazine, floxuridine, plicamycin, mitotane, pipobroman, plicamycin, tamloxifen and testolactone.
5. A pharmaceutical composition according to claim 1 or 2, wherein the further 20 chemotherapeutic or naturally occurring, semi-synthetic or synthetic therapeutic agent 2 is selected from the group consisting of an anti-cancer drug from plants such as paclitaxel (taxol), docetaxel or taxotere, a vinca alkaloid such as navelbine, vinblastin, vincristin, vindesine or vinorelbine, a vinca alkaloid such as navelbine, vinblastin, vincristin, vindesine or vinorelbine, an alkylating agent or a platinum compound such as 25 melphalan, cyclophosphamide, an oxazaphosphorine, cisplatin, carboplatin, oxaliplatin, satraplatin, tetraplatin, iproplatin, mitomycin, streptozocin, carmustine (BCNU), lomustine (CCNU), busulfan, ifosfamide, streptozocin, thiotepa, chlorambucil, a nitrogen mustard such as mechlorethamine, an immunomodulatory drug such as thalidomide, its R- and S-enantiomers and its derivatives, or revimiid (CC-5013)), an 30 ethyleneimine compound, an alkylsulphonate, daunorubicin, doxorubicin (adriamycin), liposomal doxorubicin (doxil), epirubicin, idarubicin, mitoxantrone, amsacrine, dactinomycin, distamycin or a derivative thereof, netropsin, pibenzimol, mitomycin, WO 2006/018182 PCTIEP2005/008623 142 CC-1065, a duocarmycin, mithramycin, chromomycin, olivomycin, a phtalanilide such as propamidine or stilbamidine, an anthramycin, an aziridine, a nitrosourea or a derivative thereof, a pyrimidine or purine analogue or antagonist or an inhibitor of the nucleoside diphosphate reductase such as cytarabine, 5-fluorouracile (5-FU), s pemetrexed, tegafur/uracil, uracil mustard, fludarabine, gemcitabine, capecitabine, mercaptopurine, cladribine, thioguanine, methotrexate, pentostatin, hydroxyurea, or folic acid, an acridine or a derivative thereof, a rifamycin, an actinomycin, adramycin, a camptothecin such as irinotecan or topotecan, an amsacrine or analogue thereof, a tricyclic carboxamide, an histonedeacetylase inhibitor such as SAHA, MD-275, 10 trichostatin A, CBHA, LAQ824, or valproic acid, a proteasome inhibitor such as bortezomib, a small molecule VEGF receptor antagonist such as vatalanib (FTK 787/ZK222584), SU-5416, SU-6668, SU-11248, SU-14813, AZD-6474, AZD-2171, CP-547632, CEP-7055, AG-013736, IM-842 or GW-786034, an antagonist of the mitogen-activated protein kinase such as BAY-43-9006 or BAY-57-9006, a dual is EGFR/HER2 antagonist such as gefitinib, erlotinib, CI-1033 or GW-2016, an EGFR antagonist such as iressa (ZD-1839), tarceva (OSI-774), PKI-166, EKB-569, HKI-272 or herceptin, a quinazoline derivative such as 4-[(3-chloro-4-fluorophenyl)amino]-6 {[4-(NN-dimethylamino)-1-oxo-2-buten-1-yl]amino }-7-((S)-tetrahydrofuran-3-yloxy) quinazoline or 4-[(3-chloro-4-fluoro-phenyl)aniino]-6-{[4-(homomorpholin-4-yl)-1 20 oxo-2-buten-1-yl]amino}-7-[(S)-(tetrahydrofuran-3-yl)oxy]-quinazoline , or a pharmaceutically acceptable salt thereof, an inhibitor of the transcription factor complex ESX/DRIP130/Stir-2, an inhibitor of HER-2 expression, such as the heat shock protein HSP90 modulator geldanamycin and its derivative 17-allylaminogeldanamycin or 17 AAG, a protein kinase receptor antagonist which is not classified under the synthetic 25 small molecules such as atrasentan, rituximab, cetuximab, AvastinTm (bevacizumab), IMC-1C1 1, erbitux (C-225), DC-101, EMD-72000, vitaxin, imatinib, a P-glycoprotein (P-gp) inhibitor molecule such as zosuquidar (LY 335973), its salts (especially the trichloride salt) and its polymorphs, cyclosporin A, verapamil or its R-isomer, tamoxifen, quinidine, d-alpha tocopheryl polyethylene glycol 1000 succinate, VX-710, 30 PSC833, phenothiazine, GF120918 (II), SDZ PSC 833, TMBY, MS-073, S-9788, SDZ
280-446, XR(9051) and functional derivatives, analogues and isomers of these, or an antibody targeting the surface molecules of cancer cells such as apolizumab or 1D09C3. WO 2006/018182 PCT/EP2005/008623 143 6. The pharmaceutical composition according to claim 1, wherein the further chemotherapeutic or naturally occurring, semi-synthetic or synthetic therapeutic agent 2 is the quinazoline derivative 4-[(3-chloro-4-fluorophenyl)amino]-6-{[4-(N,N 5 dimethylamino)-1-oxo-2-buten-1-yl]amino }-7-((S)-tetrahydrofuran-3-yloxy) quinazoline or a pharmaceutically acceptable salt thereof. 7. The pharmaceutical combination according to claim 1, wherein the further chemotherapeutic or naturally occurring, semi-synthetic or synthetic therapeutic agent 2 1o is the di-maleic acid salt of the compound 4-[(3-chloro-4-fluorophenyl)amino]-6-{[4 (N,N-dimethylamino)-1-oxo-2-buten-1-yl]amino}-7-((S)-tetrahydrofuran-3-yloxy) quinazoline, or 4-[(3-chloro-4-fluoro-phenyl)amino]-6-{[4-(homomorpholin-4-yl)-1 oxo-2-buten-1-yl]amino }-7-[(S)-(tetrahydrofuran-3-yl)oxy]-quinazoline, or the tautomers, stereoisomers or a pharmaceutically acceptable salt thereof. 15 8. The pharmaceutical combination according to claim 1, wherein the further chemotherapeutic or naturally occurring, semi-synthetic or synthetic therapeutic agent 2 is 4-[(3-chloro-4-fluoro-phenyl)amino]-6-{[4-(homomorpholin-4-yl)-1-oxo-2-buten-l yllamino}-7-[(S)-(tetrahydrofuran-3-yl)oxy]-quinazoline , or the tautomers, 20 stereoisomers or a pharmaceutically acceptable salt thereof. 9. The pharmaceutical combination according to claim 1, wherein the further chemotherapeutic or naturally occurring, semi-synthetic or synthetic therapeutic agent 2 is 3-Z-[l-(4-(N-((4-methyl-piperazin-1-yl)-methylcarbonyl)-N-methyl-anino)-anilino) 25 1-phenyl-methylene]-6-methoxycarbonyl-2-indolinone, or a polymorph, metabolite or pharmaceutically acceptable salt thereof. 10. The pharmaceutical combination according to claim 1, wherein the further chemotherapeutic or naturally occurring, semi-synthetic or synthetic therapeutic agent 2 30 is the monoethanesulfonate salt of 3-Z-[1-(4-(N-((4-methyl-piperazin-1-yl) methylcarbonyl)-N-methyl-amino)-anilino)-l-phenyl-methylene]-6-methoxycarbonyl 2-indolinone. WO 2006/018182 PCT/EP2005/008623 144 11. The pharmaceutical combination according to claim 1, wherein the further chemotherapeutic or naturally occurring, semi-synthetic or synthetic therapeutic agent 2 is 3-Z-[1-(4-dimethylaminomethylanilino)-1-(4-(2-carboxyethyl)phenyl)methylene]-6 s fluoro-2-indolinone, or a polymorph, metabolite or pharmaceutically acceptable salt thereof. 12. The pharmaceutical composition according to claim 1, wherein the further chemotherapeutic or naturally occurring, semi-synthetic or synthetic therapeutic agent 2 1o is selected from the group consisting of irinotecan, topotecan, oxaliplatin, docetaxel, paclitaxel, gemcitabine, pemetrexed, cisplatin, carboplatin, bevacizumab, cetuximab, gefitinib or erlotinib. 13. Use of a pharmaceutical composition according to any one of claims 1 to 12 for is the preparation of a medicament for the treatment of an oncological disease. 14. Use according to claim 13, wherein the oncological disease is selected from the group consisting of solid tumours. 20 15. Use according to claim 13 wherein the oncological disease is selected from the group consisting of urogenital cancers, lung cancers, gastrointestinal cancers, head and neck cancer, malignant mesotheliomas, breast cancer, malignant melanoma, childhood cancers, and bone or soft tissue sarcomas. 25 16. Use according to claim 13 wherein the oncological disease is selected from the group consisting of refractory or relapsed multiple myeloma, acute or chronic myelogenous leukaenia, myelodysplastic syndrome, myeloproliferativesyndromes, acute lymphoblastic leukaemia, Hodgkin's and non-Hodgkin's lymphoma. 30 17. Use of a pharmaceutical composition according to any one of claims 1 to 12 for the preparation of a medicament for the treatment of a non-oncological disease. WO 2006/018182 PCTIEP2005/008623 145 18. Use according to claim 17, wherein the non-oncological disease is selected from the group consisting of autoimmune disorders, diabetic retinopathy and rheumatoid arthritis. s 19. A pharmaceutical combination preparation kit for the treatment of diseases involving cell proliferation, migration or apoptosis of cancer cells, or angiogenesis, comprising a therapeutically effective amount of: (i) a compound 1 of Formula (I) as defined in claims 1 or 2; and 10 (ii) at least a further chemotherapeutic or naturally occurring, semi synthetic or synthetic therapeutic agent 2 as defined in any one of claims 3 to 12; 15 and optionally adapted for a co-treatment with radiotherapy or radio-immunotherapy; characterised in that the compound 1 of Formula (1) is comprised within a first compartment and the further chemotherapeutic or naturally occurring, semi-synthetic or synthetic therapeutic agent 2 is comprised within a second compartment, such that the 20 administration to a patient in need thereof can be simultaneous, separate or sequential. 20. The pharmaceutical combination preparation kit in accordance with claim 19, wherein the formulation of the compound 1 of Formula (I) is for oral administration or injection. 25 21. Use of a pharmaceutical composition according to any one of claims 1 to 12 or a pharmaceutical combination preparation kit according to claims 19 or 20, for the manufacture of a medicament, optionally adapted for a co-treatment with radiotherapy or radio-immunotherapy, to treat diseases involving cell proliferation, migration or 3o apoptosis of cancer cells, or angiogenesis, in a human or non-human mammalian body. WO 2006/018182 PCT/IEP2005/008623 146 22. Use of an effective amount of a compound 1 of Formula (I) as defined in claims 1 or 2 in combination with at least a further chemotherapeutic or naturally occurring, semi-synthetic or synthetic therapeutic agent 2 as defined in any one of claims 3 to 12, for the manufacture of a pharmaceutical combination preparation, optionally adapted for 5 a co-treatment with radiotherapy or radio-immunotherapy, for simultaneous, separate or sequential use in the treatment of diseases involving cell proliferation, migration or apoptosis of cancer cells, or angiogenesis, in a human or non-human mammalian body. 23. A method for the treatment of diseases involving cell proliferation, migration or 10 apoptosis of cancer cells, or angiogenesis, which method comprises simultaneous, separate or sequential co-administration of effective amounts of: (iii) a compound 1 of Formula (I) as defined in claims 1 or 2; and (iv) at least a further chemotherapeutic or naturally occurring, semi Is synthetic or synthetic therapeutic agent 2 as defined in any one of claims 3 to 12; in the form of a combined preparation optionally adapted for a co-treatment with radiotherapy or radio-immunotherapy, to a person in need of such treatment. 20 24. Use according to any one of claims 13 to 18, 21 or 22, characterised in that the compound 1 of Formula (I), or its polymorph, hydrate, metabolite or pharmaceutically acceptable salt, is administered intermittent or in a daily dosage such that the plasma level of the active substance lies between 10 and 5000 nM for at least 12 hours of the 25 dosing interval.
AU2011226830A 2004-08-14 2011-09-23 Combinations for the treatment of diseases involving cell proliferation Abandoned AU2011226830A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2011226830A AU2011226830A1 (en) 2004-08-14 2011-09-23 Combinations for the treatment of diseases involving cell proliferation

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
EP04019361.7 2004-08-14
EP04019448.2 2004-08-17
AU2011226830A AU2011226830A1 (en) 2004-08-14 2011-09-23 Combinations for the treatment of diseases involving cell proliferation

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU2005274384A Division AU2005274384B2 (en) 2004-08-14 2005-08-09 Combinations for the treatment of diseases involving cell proliferation

Publications (1)

Publication Number Publication Date
AU2011226830A1 true AU2011226830A1 (en) 2011-10-13

Family

ID=45442061

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2011226830A Abandoned AU2011226830A1 (en) 2004-08-14 2011-09-23 Combinations for the treatment of diseases involving cell proliferation

Country Status (1)

Country Link
AU (1) AU2011226830A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112218865A (en) * 2018-04-24 2021-01-12 沃泰克斯药物股份有限公司 Pteridinone compounds and uses thereof

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112218865A (en) * 2018-04-24 2021-01-12 沃泰克斯药物股份有限公司 Pteridinone compounds and uses thereof
CN112218865B (en) * 2018-04-24 2024-03-12 沃泰克斯药物股份有限公司 Pteridinone compounds and uses thereof

Similar Documents

Publication Publication Date Title
AU2005274384B2 (en) Combinations for the treatment of diseases involving cell proliferation
US20180086744A1 (en) Combination treatment of cancer comprising egfr/her2 inhibitors
JP6736559B2 (en) Triazolopyrimidine compounds and uses thereof
RU2492864C2 (en) Method of treating cancer carrying egfr mutations
JP2008509948A5 (en)
JP2010514676A5 (en)
AU2011226830A1 (en) Combinations for the treatment of diseases involving cell proliferation
ES2368245T3 (en) COMBINATIONS FOR THE TREATMENT OF DISEASES THAT INVOLVE A CELL PROLIFERATION.
WO2019049891A1 (en) METHOD FOR TREATING CANCER BY COMBINATION OF Trk INHIBITOR AND KINASE INHIBITOR

Legal Events

Date Code Title Description
MK4 Application lapsed section 142(2)(d) - no continuation fee paid for the application