Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
1,4-DICARBONYL-PIPERIDYL DERIVATIVES
Document Type and Number:
WIPO Patent Application WO/2017/076484
Kind Code:
A1
Abstract:
Compounds of the formula I in which Z, W, Q, R and Y have the meanings indicated in Claim 1, are inhibitors of Tankyrase, and can be employed, inter alia, for the treatment of diseases such as cancer, cardiovascular diseases, central nervous system injury and different forms of inflammation.

Inventors:
BUCHSTALLER HANS-PETER (DE)
DORSCH DIETER (DE)
Application Number:
PCT/EP2016/001668
Publication Date:
May 11, 2017
Filing Date:
October 07, 2016
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
MERCK PATENT GMBH (DE)
International Classes:
C07D401/14; A61K31/4545; A61K31/519; A61P35/00; C07D401/06; C07D401/12; C07D487/04
Domestic Patent References:
WO2013008217A12013-01-17
WO2013012723A12013-01-24
WO2015169421A12015-11-12
WO2015014442A12015-02-05
Other References:
MICHAEL D. SHULTZ ET AL: "Identification of NVP-TNKS656: The Use of Structure-Efficiency Relationships To Generate a Highly Potent, Selective, and Orally Active Tankyrase Inhibitor", JOURNAL OF MEDICINAL CHEMISTRY, vol. 56, no. 16, 22 August 2013 (2013-08-22), US, pages 6495 - 6511, XP055316909, ISSN: 0022-2623, DOI: 10.1021/jm400807n
ZIHAO HUA ET AL: "Development of Novel Dual Binders as Potent, Selective, and Orally Bioavailable Tankyrase Inhibitors", JOURNAL OF MEDICINAL CHEMISTRY, vol. 56, no. 24, 27 December 2013 (2013-12-27), US, pages 10003 - 10015, XP055316973, ISSN: 0022-2623, DOI: 10.1021/jm401317z
Attorney, Agent or Firm:
MERCK PATENT GMBH (DE)
Download PDF:
Claims:
Patent Claims

1. Compounds of the formula I

denotes

R2

W denotes CH2 or C(CH3)2,

Q denotes C(CH3)2, C 3), CH(CH2CH3), CH[CH(CH3)2],

CH[CH2CH(CH3)2], C_ or -O- ,

R denotes CH2( C(CH3)2> NH or N(CH3),

Y denotes Ar or Het,

R1 denotes H, F or CH3,

R2 denotes H or CH3, Ar denotes phenyl, which is unsubstituted, or mono-, di- or trisubstituted by Hal, CN, A, OR3, (CH2)mN(R3)2) COOR3 and/or CON(R3)2,

Het denotes pyrazolyl, pyridyl, pyrimidyl or pyridazinyl, each of which is unsubstituted or mono- or disubstituted by Hal, CN, A, OR3, N(R3)2 and/or (CH2)mCON(R3)2

A denotes unbranched or branched alkyl with 1 - 8 C-Atoms,

wherein one or two non-adjacent CH- and/or CH2-groups may be replaced by N- or O-atoms and wherein 1-7 H-atoms may be replaced by F, CI and/or OH,

R3 denotes H or unbranched or branched alkyl with 1 , 2, 3 or 4 C- Atoms,

Hal denotes F, CI, Br or I,

m denotes 0, 1 or 2,

n denotes 0, 1 or 2, and pharmaceutically acceptable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios.

Compounds according to Claim 1 , selected from the group

No. Name

"cr 2-{2,2-Dimethyl-4-[4-(1 -methyl-1 H-pyrazole-4-carbonyl)- piperidin-1-yl]-4-oxo-butyl}-3H-quinazolin-4-one

"C2" 2-{(S)-2-Methyl-4-[4-( 1 -methyl-1 H-pyrazole-4-carbonyl)- piperidin-1-yl]-4-oxo-butyl}-3H-quinazolin-4-one

"C3" 2-{(R)-2-Methyi-4-[4-(1 -methyl-1 H-pyrazole-4-carbonyl)- piperidin-1-yl]-4-oxo-butyl}-3H-quinazolin-4-one "C4" 2-{(S)-4-[4-(1-lsopropyl-1 H-pyrazole-4-carbonyl)-piperidin-1- yl]-2-methyl-4-oxo-butyl}-3H-quinazolin-4-one

"C5" 2-{(R)-4-[4-(1-lsopropyl-1 H-pyrazole-4-carbonyl)-piperidin-1 - yl]-2-methyl-4-oxo-butyl}-3H-quinazolin-4-one

"C6" 6-Fluoro-8-methyl-2-{(S)-2-methyl-4-[4-(1 -methyl-1 H- pyrazole-4-carbonyl)-piperidin-1-yl]-4-oxo-butyl}-3H- quinazolin-4-one

"C7" 6-Fluoro-8-methyl-2-{(R)-2-methyl-4-[4-(1 -methyl-1 H- pyrazole-4-carbonyl)-piperidin-1-yl]-4-oxo-butyl}-3H- quinazolin-4-one

"C8" 2-((S)-3-Methyl-2-{2-[4-(1-methyl-1H-pyrazole-4-carbonyl)- piperidin-1-yl]-2-oxo-ethyl}-butyl)-3H-quinazolin-4-one

"C9" 2-((R)-3-Methyl-2-{2-[4-(1 -methyl-1 H-pyrazole-4-carbonyl)- piperidin-1-yl]-2-oxo-ethyl}-butyl)-3H-quinazolin-4-one

"C10" 2-((S)-4-Methyl-2-{2-[4-(1 -methyl-1 H-pyrazole-4-carbonyl)- piperidin-1-yl]-2-oxo-ethyl}-pentyl)-3H-quinazolin-4-one

"011" 2-((R)-4-Methyl-2-{2-[4-(1 -methyl-1 H-pyrazole-4-carbonyl)- piperidin-1-yl]-2-oxo-ethyl}-pentyl)-3H-quinazolin-4-one

"C12" 2-(1 -{2-[4-(1 -Methyl-1 H-pyrazole-4-carbonyl)-piperidin-1-yl]- 2-oxo-ethyl}-cyclopropylmethyl)-3H-quinazolin-4-one

"C13" 2-{3,3-Dimethyl-4-[4-(1 -methyl-1 H-pyrazole-4-carbonyl)- piperidin-1-yl]-4-oxo-butyl}-3H-quinazolin-4-one

"C14" 2-{1 , 1 -Dimethyl-4-[4-(1 -methyl-1 H-pyrazole-4-carbonyl)- piperidin-1-yl]-4-oxo-butyl}-3H-quinazolin-4-one

"C15" 2-{4-[4-(4-Methoxy-benzoyl)-piperidin-1 -yl]-1 , 1 -dimethyl-4- oxo-butyl}-3H-quinazolin-4-one

"C16" 2-{4-[4-(6-Methoxy-pyridine-3-carbonyl)-piperidin-1-yl]-2,2- d imethy l-4-oxo-butyl}-7-methy I-3 , 7-d ihyd ro-py rrolo[2 , 3- d]pyrimidin-4-one "C17" 2-{2,2-Dimethyl-4-[4-(1 -methyl-1 H-pyrazole-4-carbonyl)- piperidin-1-yl]-4-oxo-butyl}-7-methyl-3,7-dihydro-pyrrolo[2,3- d]pyrimidin-4-one

"C18" 2-{4-[4-(1-Ethyl-1H-pyrazole-4-carbonyl)-piperidin-1-yl]-2,2- c O

dimethyl-4-oxo-butyl}-7-methyl-3,7-dihydro-pyrrolo[2,3- d]pyrimidin-4-one

"C19" 2-{4-[4-(1 -lsopropyl-1 H-pyrazole-4-carbonyl)-piperidin-1 -yl]- 2 ,2-d imethy l-4-oxo-buty l}-7-methyl-3 ,7-d ihyd ro-py rrolo[2 , 3-

10 d]pyrimidin-4-one

"C20" 6-{2,2-Dimethyl-4-[4-(1 -methyl-1 H-pyrazole-4-carbonyl)- piperidin-1 -yl]-4-oxo-butyl}-1 -methyl-1 ,5-dihydro- pyrazolo[3,4-d]pyrimidin-4-one

15 "C21" 6-{4-[4-(6-Methoxy-pyridine-3-carbonyl)-piperidin-1-yl]-2,2- dimethyl-4-oxo-butyl}-1 -methyl-1 ,5-dihydro-pyrazolo[3,4- d]pyrimidin-4-one

"C22" 6-{4-[4-(6-Methoxy-5-methyl-pyridine-3-carbonyl)-piperidin-

1 -y l]-2 ,2-d imethy l-4-oxo-buty l}-1 -methyl- 1 , 5-d ihyd ro-

20

pyrazolo[3,4-d]pyrimidin-4-one

"C23" 2-{2-[4-(4-Methoxy-benzoyl)-piperidin-1-yl]-2-oxo- ethoxymethyl}-3H-quinazo m-4-one

"C24" 2-{2-[4-(1 -Methyl-1 H-pyrazole-4-carbonyl)-piperidin-1-yl]-2-

25 oxo-ethoxymethyl}-3H-quinazolin-4-one

"C25" 2-{2-[4-(6-Methoxy-pyridine-3-carbonyl)-piperidin-1-yl]-2- oxo-ethoxymethyl}-3H-quinazolin-4-one

"C26" 6,8-Difluoro-2-{2-[4-(4-methoxy-benzoyl)-piperidin-1-yl]-2-

30 oxo-ethoxymethyl}-3H-quinazolin-4-one

"C27" 6,8-Difluoro-2-{2-[4-(1 -methyl-1 H-pyrazole-4-carbonyl)- piperidin-1-yl]-2-oxo-ethoxymethyl}-3H-quinazolin-4-one

"C28" 6,8-Difluoro-2-{2-[4-(6-methoxy-pyridine-3-carbonyl)- piperidin-1-yl]-2-oxo-ethoxymethyl}-3H-quinazolin-4-one "C29" 4-(4-Methoxy-benzoyl)-piperidine-1 -carboxylic acid [2-(4- oxo-3,4-dihydro-quinazolin-2-yl)-ethyl]-amide

"C30" 4-(4-Methoxy-benzoyl)-piperidine-1 -carboxylic acid methyl-

[2-(4-oxo-3,4-dihydro-quinazolin-2-yl)-ethyl]-amide

"C31" 4-(1 -Methyl-1 H-pyrazole-4-carbonyl)-piperidine-1 -carboxylic acid methyl-[2-(4-oxo-3,4-dihydro-quinazolin-2-yl)-ethyl]- amide

"C32" 4-{1-[3,3-Dimethyl-4-(4-oxo-3,4-dihydro-quinazolin-2-yl)- butyryl]-piperidine-4-carbonyl}-benzoic acid methyl ester

"C33" 4-{1-[3,3-Dimethyl-4-(4-oxo-3,4-dihydro-quinazolin-2-yl)- butyryl]-piperidine-4-carbonyl}-benzoic acid

"C34" 3-{1-[3,3-Dimethyl-4-(4-oxo-3,4-dihydro-quinazolin-2-yl)- butyryl]-piperidine-4-carbonyl}-benzoic acid methyl ester

"C35" 3-{1-[3,3-Dimethyl-4-(4-oxo-3,4-dihydro-quinazolin-2-yl)- butyryl]-piperidine-4-carbonyl}-benzoic acid

"C36" 3-{1-[3,3-Dimethyl-4-(8-methyl-4-oxo-3,4-dihydro-quinazolin-

2-yl)-butyryl]-piperidine-4-carbonyl}-benzoic acid methyl ester

"C37" 3-{1-[3,3-Dimethyl-4-(8-methyl-4-oxo-3,4-dihydro-quinazolin- 2-yl)-butyryl]-piperidine-4-carbonyl}-benzoic acid

"C38" 4-{1-[3,3-Dimethyl-4-(4-oxo-3,4-dihydro-quinazolin-2-yl)- butyryl]-pipertdine-4-carbonyl}-2-methyl-benzoic acid methyl ester

"C39" 4-{1-[3,3-Dimethyl-4-(4-oxo-3,4-dihydro-quinazolin-2-yl)- butyryl]-piperidine-4-carbonyl}-2-methyl-benzoic acid

"C40" 4-{1-[3,3-Dimethyl-4-(8-methyl-4-oxo-3,4-dihydro-quinazolin- 2-yl)-butyryl]-piperidine-4-carbonyl}-2-methyl-benzoic acid methyl ester

"C41" 4-{1-[3,3-Dimethyl-4-(8-methyl-4-oxo-3,4-dihydro-quinazolin-

2-yl)-butyryl]-piperidine-4-carbonyl}-2-methyl-benzoic acid "C42" 3-{1-[(R)-3-Methyl-4-(8-methyl-4-oxo-3,4-dihydro-quinazolin- 2-yl)-butyryl]-piperidine-4-carbonyl}-benzoic acid methyl ester

"C43" 3-{1-[(S)-3-Methyl-4-(8-methyl-4-oxo-3,4-dihydro-quinazolin-

2-yl)-butyryl]-piperidine-4-carbonyl}-benzoic acid methyl ester

"C44" 3-{1-[(R)-3-Methyl-4-(8-methyl-4-oxo-3,4-dihydro-quinazolin-

2-yl)-butyryl]-piperidine-4-carbonyl}-benzoic acid

"C45" 3-{1-[(S)-3-Methyl-4-(8-methyl-4-oxo-3,4-dihydro-quinazolin- 2-yl)-butyryl]-piperidine-4-carbonyl}-benzoic acid

"C46" 3-{1-[(R)-4-(6-Fluoro-8-methyl-4-oxo-3,4-dihydro-quinazolin- 2-yl)-3-methyl-butyryl]-piperidine-4-carbonyl}-benzoic acid

"C47" 3-{1-[(S)-4-(6-Fluoro-8-methyl-4-oxo-3,4-dihydro-quinazolin- 2-yl)-3-methyl-butyryl]-piperidine-4-carbonyl}-benzoic acid

"C48" 2-{4-[4-(4-Dimethylaminomethyl-benzoyl)-piperidin-1-yl]-2,2- dimethyl-4-oxo-butyl}-3H-quinazolin-4-one

"C49" 3-{2,2-Dimethyl-4-[4-(1-methyl-1 H-pyrazole-4-carbonyl)- piperidin-1-yl]-4-oxo-butyl}-2H-isoquinolin-1-one

"C50" 3-{(S)-2-Methyl-4-[4-(1 -methyl-1 H-pyrazole-4-carbonyl)- piperidin-1 -yl]-4-oxo-butyl}-2H-isoquinolin-1 -one

"C51" 3-{(R)-2-Methyl-4-[4-(1 -methyl-1 H-pyrazole-4-carbonyl)- piperidin-1 -yl]-4-oxo-butyl}-2H-isoquinolin-1 -one

"C52" 4-{ 1 -[3 , 3-Dimethy l-4-( 1 -oxo- 1 ,2-d ihyd ro-isoq u inolin-3-y I)- butyryl]-piperidine~4-carbonyl}-benzoic acid

"C53" 3-{1-[3,3-Dimethyl-4-(1-oxo-1 ,2-dihydro-isoquinolin-3-yl)- butyryl]-piperidine-4-carbonyl}-benzoic acid and pharmaceutically acceptable solvates, salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios. Process for the preparation of compounds of the formula I according to Claims 1-2 and pharmaceutically acceptable salts, solvates, tauto- mers and stereoisomers thereof, characterised in that

a compound of the formula II in which Y has the meanings indicated in Claim 1 , is reacted

with a compound of formula III

Z-W-Q-R-C(=O)-L III in which Z, W, Q and R have the meanings indicated in Claim 1 , and L denotes CI, Br, I or a free or reactively functionally modified OH group, and/or

a base or acid of the formula I is converted into one of its salts.

Medicaments comprising at least one compound of the formula I according to claim 1 and/or pharmaceutically acceptable salts, solvates, tautomers and stereoisomers thereof, including mixtures thereof in all ratios, and optionally an pharmaceutically acceptable carrier, excipient or vehicle.

Compounds for use of the formula I according to claim 1 and pharmaceutically acceptable salts, solvates, tautomers and stereoisomers thereof, including mixtures thereof in all ratios, for the treatment and/or prevention of cancer, multiple sclerosis, cardiovascular diseases, central nervous system injury and different forms of inflammation .

6. Compounds for use according to claim 6 for the treatment and/or

prevention of diseases selected from the group cancer of head, neck, eye, mouth, throat, esophagus, bronchus, larynx, pharynx, chest, bone, lung, colon, rectum, stomach, prostate, urinary bladder, uterine, cervix, breast, ovaries, testicles or other reproductive organs, skin, thyroid, blood, lymph nodes, kidney, liver, pancreas, brain, central nervous system, solid tumors and blood-borne tumors.

7. Medicaments comprising at least one compound of the formula I

according to claim 1 and/or pharmaceutically acceptable salts, solvates and stereoisomers thereof, including mixtures thereof in all ratios, and at least one further medicament active ingredient. 8. Set (kit) consisting of separate packs of

(a) an effective amount of a compound of the formula I according to claim 1 and/or pharmaceutically acceptable salts, solvates, salts and stereoisomers thereof, including mixtures thereof in all ratios, and

(b) an effective amount of a further medicament active ingredient.

Description:
1 ,4-Dicarbonyl-piperidyl derivatives

BACKGROUND OF THE INVENTION

The invention had the object of finding novel compounds having valuable properties, in particular those which can be used for the preparation of medicaments.

The present invention relates to 1 ,4-dicarbonyl-piperidyl derivatives which inhibit the activity of Tankyrases (TANKs) and poly(ADP-ribose)polymerase PARP-1. The compounds of this invention are therefore useful in treating diseases such as cancer, multiple sclerosis, cardiovascular diseases, central nervous system injury and different forms of inflammation. The present invention also provides methods for preparing these compounds,

pharmaceutical compositions comprising these compounds, and methods of treating diseases utilizing pharmaceutical compositions comprising these compounds.

The nuclear enzyme poly(ADP-ribose) polymerase-1 (PARP-1) is a member of the PARP enzyme family. This growing family of enzymes consist of PARPs such as, for example: PARP-1 , PARP-2, PARP-3 and Vault-PARP; and Tankyrases (TANKs), such as, for example: TANK-1 and TANK-2. PARP is also referred to as poly(adenosine 5'-diphospho-ribose) polymerase or PARS (poly(ADP-ribose) synthetase).

TANK-1 seems to be required for the polymerization of mitotic spindle- associated poly(ADP-ribose). The poly(ADP-ribosyl)ation activity of TANK-1 might be crucial for the accurate formation and maintenance of spindle bipolarity. Furthermore, PARP activity of TANK-1 has been shown to be required for normal telomere separation before anaphase. Interference with tankyrase PARP activity results in aberrant mitosis, which engenders a transient cell cycle arrest, probably due to spindle checkpoint activation, followed by cell death. Inhibition of tankyrases is therefore expected to have a cytotoxic effect on proliferating tumor cells (WO 2008/107478).

PARP inhibitors are described by M. Rouleau et al. in Nature Reviews,

Volume 10, 293-301 in clinical cancer studies (Table 2, page 298).

According to a review by Horvath and Szabo (Drug News Perspect 20(3), April 2007, 171-181) most recent studies demonstrated that PARP inhibitors enhance the cancer cell death primarily because they interfere with DNA repair on various levels. More recent studies have also demonstrated that PARP inhibitors inhibit angiogenesis, either by inhibiting growth factor expression, or by inhibiting growth factor-induced cellular proliferative responses. These findings might also have implications on the mode of PARP inhibitors' anticancer effects in vivo.

Also a study by Tentori et al. (Eur. J. Cancer, 2007, 43 (14) 2124-2133) shows that PARP inhibitors abrogate VEGF or placental growth factor-induced migration and prevent formation of tubule-like networks in cell-based systems, and impair angiogenesis in vivo. The study also demonstrates that growth factor-induced angiogenesis is deficient in PARP-1 knock-out mice. The results of the study provide evidence for targeting PARP for anti-angiogenesis, adding novel therapeutic implications to the use of PARP inhibitors in cancer treatment.

Defects in conserved signaling pathways are well known to play key roles in the origins and behavior of essentially all cancers (E.A.Fearon, Cancer Cell, Vol. 16, Issue 5, 2009, 366-368). The Wnt pathway is a target for anti-cancer therapy. A key feature of the Wnt pathway is the regulated proteolysis

(degradation) of β-catenin by the β-catenin destruction complex. Proteins like WTX, APC or Axin are involved in the degradation process. A proper degradation of β-catenin is important to avoid an inappropriate activation of the Wnt pathway which has been observed in many cancers. Tankyrases inhibit activity of Axin and hence inhibit the degradation of β-catenin. Consequently, tankyrase inhibitors increase degradation of β-catenin. A paper in the journal Nature not only offers important new insights into proteins regulating Wnt signaling but also further supports the approach to antagonize β-catenin levels and localization via small molecules (Huang et al., 2009;

Nature, Vol 461 , 614-620). The compound XAV939 inhibits growth of DLD-1- cancer cells. They found that XAV9393 blocked Wnt-stimulated accumulation of β-catenin by increasing the levels of the AXIN-1 and AXIN2 proteins.

Subsequent work by the authors established that XAV939 regulates AXIN levels via inhibition of tankyrases 1 and 2 (TNKS1 and TNKS2), both of which are members of the poly(ADP-ribose) polymerase (PARP) protein family (S.J. Hsiao et al., Biochimie 90, 2008, 83-92).

It has been found that the compounds according to the invention and salts thereof have very valuable pharmacological properties while being well tolerated.

The present invention specifically relates to compounds of the formula I which inhibit Tankyrase 1 and 2, to compositions which comprise these compounds, and to processes for the use thereof for the treatment of TANK-induced diseases and complaints.

The compounds of the formula I can furthermore be used for the isolation and investigation of the activity or expression of TANKs. In addition, they are particularly suitable for use in diagnostic methods for diseases in connection with unregulated or disturbed TANK activity.

The host or patient can belong to any mammalian species, for example a primate species, particularly humans; rodents, including mice, rats and hamsters; rabbits; horses, cows, dogs, cats, etc. Animal models are of interest for experimental investigations, providing a model for treatment of human disease. The susceptibility of a particular cell to treatment with the compounds according to the invention can be determined by in vitro tests. Typically, a culture of the cell is combined with a compound according to the invention at various concentrations for a period of time which is sufficient to allow active agents such as anti IgM to induce a cellular response such as expression of a surface marker, usually between about one hour and one week. In vitro testing can be carried out using cultivated cells from blood or from a biopsy sample. The amount of surface marker expressed is assessed by flow cytometry using specific antibodies recognising the marker.

The dose varies depending on the specific compound used, the specific disease, the patient status, etc. A therapeutic dose is typically sufficient considerably to reduce the undesired cell population in the target tissue while the viability of the patient is maintained. The treatment is generally continued until a considerable reduction has occurred, for example an at least about 50% reduction in the cell burden, and may be continued until essentially no more undesired cells are detected in the body.

PRIOR ART

E. Wahlberg et al., Nature Biotechnology (2012), 30(3), 283.

M. D. Shultz et al., Journal of Medicinal Chemistry 2013 , 56 (16), 6495- 6511.

In the same publication, the following benzoylpiperidine derivative is

described as Tankyrase inhibitor:

IC5o (TNKS1) = 2 nM, ICso (TNKS2) = 0.6 nM; cellular assay: ECso = 35 nM.

H. Bregman et al., Journal of Medicinal Chemistry (2013), 56(3), 1341 The following quinazolinone is described as Tankyrase inhibitor:

IC 5 o (TNKS1) = 7.4 nM, IC 5 o(TNKS2) = 4.4 nM; cellular assay: ECso = 320 nM.

The compounds of the present invention are significantly more active.

Other tankyrase inhibitors are described in WO 2013/012723, WO 2013/010092 and in WO 2013/008217.

Recently a patent (WO 2014/036022 A1) claiming further

quina below (R = F)

For this compound the following data are reported: ICso(TNKSI) = 18.8 nM, IC5o(TNKS2) = 2.59 nM; cellular assay: ECso = 247 nM in the patent application.

Oxoquinazolinyl-butanamide derivatives for the treatment of cancer are described in WO 2015/014442 A1. SUMMARY OF THE INVENTION

The invention relates to compounds of the formula I

in which

Z denotes

W denotes CH2 or C(CH3)2, Q denotes C(CH 3 )2, CH(CH 3 ), CH(CH 2 CH3), CH[CH(CH3)2],

R denotes CH2, C(CH 3 ) 2l NH or N(CH 3 ),

Y denotes Ar or Het,

R 1 denotes H, F or CH 3) R 2 denotes H or CH3,

Ar denotes phenyl, which is unsubstituted, or mono-, di- or trisubstituted by Hal, CN.A, OR 3 , (CH 2 ) m N(R 3 )2, COOR 3 and/or CON(R 3 ) 2 ,

Het denotes pyrazolyl, pyridyl, pyrimidyl or pyridazinyl, each of which is unsubstituted or mono- or disubstituted by Hal, CN, A, OR 3 , N(R 3 ) 2 and/or (CH 2 )mCON(R 3 ) 2

A denotes unbranched or branched alkyl with 1 - 8 C-Atoms, wherein one or two non-adjacent CH- and/or CH 2 -groups may be replaced by N- or O-atoms and wherein 1-7 H-atoms may be replaced by F, CI and/or OH,

R 3 denotes H or unbranched or branched alkyl with 1 , 2, 3 or 4 C-Atoms, Hal denotes F, CI, Br or I,

m denotes 0, 1 or 2,

n denotes 0, 1 or 2,

and pharmaceutically acceptable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios.

The invention also relates to the optically active forms (stereoisomers), the enantiomers, the racemates, the diastereomers and the hydrates and solvates of these compounds.

Moreover, the invention relates to pharmaceutically acceptable derivatives of compounds of formula I.

The term solvates of the compounds is taken to mean adductions of inert solvent molecules onto the compounds which form owing to their mutual attractive force. Solvates are, for example, mono- or dihydrates or

alkoxides.

It is understood, that the invention also relates to the solvates of the salts. The term pharmaceutically acceptable derivatives is taken to mean, for example, the salts of the compounds according to the invention and also so-called prodrug compounds.

As used herein and unless otherwise indicated, the term "prodrug" means a derivative of a compound of formula I that can hydrolyze, oxidize, or otherwise react under biological conditions (in vitro or in vivo) to provide an active compound, particularly a compound of formula I. Examples of prodrugs include, but are not limited to, derivatives and metabolites of a compound of0

formula I that include biohydrolyzable moieties such as biohydrolyzable amides, biohydrolyzable esters, biohydrolyzable carbamates, biohydrolyzable carbonates, biohydrolyzable ureides, and biohydrolyzable phosphate analogues. In certain embodiments, prodrugs of compounds with carboxyl5 functional groups are the lower alkyl esters of the carboxylic acid. The

carboxylate esters are conveniently formed by esterifying any of the carboxylic acid moieties present on the molecule. Prodrugs can typically be prepared using well- known methods.

0 The expression "effective amount" denotes the amount of a medicament or of a pharmaceutical active ingredient which causes in a tissue, system, animal or human a biological or medical response which is sought or desired, for example, by a researcher or physician.

^ In addition, the expression "therapeutically effective amount" denotes an

amount which, compared with a corresponding subject who has not received this amount, has the following consequence:

improved treatment, healing, prevention or elimination of a disease, syndrome, condition, complaint, disorder or side-effects or also the reduction° in the advance of a disease, complaint or disorder.

The expression "therapeutically effective amount" also encompasses the amounts which are effective for increasing normal physiological function. 5 The invention also relates to the use of mixtures of the compounds of the formula I, for example mixtures of two diastereomers, for example in the ratio 1 :1 , 1 :2, 1 :3, 1 :4, 1 :5, 1:10, 1 :100 or 1 :1000.

These are particularly preferably mixtures of stereoisomeric compounds.

"Tautomers" refers to isomeric forms of a compound that are in equilibrium with each other. The concentrations of the isomeric forms will depend on the environment the compound is found in and may be different depending upon, for example, whether the compound is a solid or is in an organic or aqueous solution.

The invention relates to the compounds of the formula I and salts thereof and to a process for the preparation of compounds of the formula I and pharmaceutically acceptable salts, solvates, tautomers and stereoisomers thereof, characterised in that

a compound of the formula II

in which Y has the meanings indicated in Claim 1 , is reacted

with a compound of formula III

Z-W-Q-R-C(=0)-L III in which Z, W, Q and R have the meanings indicated in Claim 1 , and L denotes CI, Br, I or a free or reactively functionally modified OH group, and/or a base or acid of the formula I is converted into one of its salts.

Above and below, the radicals Z, W, Q, R and Y have the meanings indicated for the formula I, unless explicitely stated otherwise.

A denotes alkyl, this is unbranched (linear) or branched, and has 1 , 2, 3, 4, 5, 6, 7 or 8 C atoms. A preferably denotes methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl or tert-butyl, furthermore also pentyl, 1-, 2- or 3-methylbutyl, 1 ,1- , 1 ,2- or 2,2-dimethylpropyl, 1-ethylpropyl, hexyl, 1- , 2- , 3- or 4-methylpentyl, 1 ,1- , 1 ,2- , 1 ,3- , 2,2- , 2,3- or 3,3-dimethylbutyl, 1- or 2-ethylbutyl, 1-ethyl-1-methylpropyl, 1-ethyl-2-methylpropyl, 1 ,1,2- or 1 ,2,2-trimethylpropyl, furthermore preferably, for example, trifluoromethyl. A very particularly preferably denotes alkyl having 2, 3, 4, 5 or 6 C atoms, preferably ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, pentyl, hexyl, trifluoromethyl, pentafluoroethyl or 1 ,1 ,1-trifluoroethyl.

Moreover, A denotes preferably CH2OCH3, CH2CH2OH or CH2CH2OCH3.

Throughout the invention, all radicals which occur more than once may be identical or different, i.e. are independent of one another.

The compounds of the formula I may have one or more chiral centres and can therefore occur in various stereoisomeric forms. The formula I encompasses all these forms.

Accordingly, the invention relates, in particular, to the compounds of the formula I in which at least one of the said radicals has one of the preferred meanings indicated above. Some preferred groups of compounds may be expressed by the following sub-formulae la to le, which conform to the formula I and in which the radicals not designated in greater detail have the meaning indicated for the formula I, but in which in la R 1 denotes H, F or CH3; W

- 11 - denotes H or CH3;

denotes phenyl, which is unsubstituted, or mono-, di- or trisubstituted by Hal, CN, A, OR 3 , N(R 3 ) 2 , COOR 3 and/or CON(R 3 ) 2 ;

denotes pyrazolyl, pyridyl, pyrimidyl or pyridazinyl, each of which is unsubstituted or mono- or disubstituted by Hal, CN, A, OR 3 and/or N(R 3 ) 2 ;

denotes CH2 or C(CH3)2,

denotes C(CH 3 )2, CH(CH 3 ), CH(CH 2 CH3), CH[CH(CH 3 ) 2 ],

CH[CH 2 CH(CH 3 ) 2 ], C or -O- ,

denotes CH 2 , C(CH 3 ) 2 , NH or N(CH 3 ), denotes Ar or Het,

denotes H, F or CH 3 ,

denotes H or CH 3 ,

denotes phenyl, which is unsubstituted, or mono-, di- or trisubstituted by Hal, CN.A, OR 3 , N(R 3 ) 2 , COOR 3 and/or CON(R 3 ) 2 ,

denotes pyrazolyl, pyridyl, pyrimidyl or pyridazinyl, each of which is unsubstituted or mono- or disubstituted by Hal, CN, A, OR 3 , N(R 3 ) 2 and/or (CH 2 ) m CON(R 3 ) 2

denotes unbranched or branched alkyl with 1 - 8 C-Atoms, wherein one or two non-adjacent CH- and/or CH 2 -groups may be replaced by N- or O-atoms and wherein 1-7 H- atoms may be replaced by F, CI and/or OH,

denotes H or unbranched or branched alkyl with 1 , 2, 3 or 4 C-Atoms,

denotes F, CI, Br or I, m denotes 0, 1 or 2,

n denotes 0, 1 or 2,

and pharmaceutically acceptable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios.

The compounds of the formula I and also the starting materials for their preparation are, in addition, prepared by methods known per se, as described in the literature (for example in the standard works, such as

Houben-Weyl, Methoden der organischen Chemie [Methods of Organic Chemistry], Georg-Thieme-Verlag, Stuttgart), to be precise under reaction conditions which are known and suitable for the said reactions. Use can also be made here of variants known per se which are not mentioned here in greater detail.

The starting compounds of the formula II and III are generally known. If they are novel, however, they can be prepared by methods known per se.

Compounds of the formula I can preferably be obtained by reacting a compound of the formula II with a compound of the formula III.

In the compounds of the formula III, L preferably denotes CI, Br, I or a free or reactively modified OH group, such as, for example, an activated ester, an imidazolide or alkylsulfonyloxy having 1-6 C atoms (preferably methyl- sulfonyloxy or trifluoromethylsulfonyloxy) or arylsulfonyloxy having 6-10 C atoms (preferably phenyl- or p-tolylsulfonyloxy).

The reaction is generally carried out in the presence of an acid-binding agent, preferably an organic base, such as DIPEA, triethylamine, dimethyl- aniline, pyridine or quinoline.

The addition of an alkali or alkaline earth metal hydroxide, carbonate or bicarbonate or another salt of a weak acid of the alkali or alkaline earth met- als, preferably of potassium, sodium, calcium or caesium, may also be favourable.

Depending on the conditions used, the reaction time is between a few minutes and 14 days, the reaction temperature is between about -30° and 140°, normally between -10° and 90°, in particular between about 0° and about 70°.

Examples of suitable inert solvents are hydrocarbons, such as hexane, petroleum ether, benzene, toluene or xylene; chlorinated hydrocarbons, such as trichloroethylene, 1 ,2-dichloroethane, carbon tetrachloride, chloroform or dichloromethane; alcohols, such as methanol, ethanol, isopropa- nol, n-propanol, n-butanol or tert-butanol; ethers, such as diethyl ether, diisopropyl ether, tetrahydrofuran (THF) or dioxane; glycol ethers, such as ethylene glycol monomethyl or monoethyl ether, ethylene glycol dimethyl ether (diglyme); ketones, such as acetone or butanone; amides, such as acetamide, dimethylacetamide or dimethylformamide (DMF); nitriles, such as acetonitrile; sulfoxides, such as dimethyl sulfoxide (DMSO); carbon disulfide; carboxylic acids, such as formic acid or acetic acid; nitro compounds, such as nitromethane or nitrobenzene; esters, such as ethyl acetate, or mixtures of the said solvents.

Particular preference is given to acetonitrile, 1 ,2-dichloroethane, dichloromethane and/or DMF.

Pharmaceutical salts and other forms

The said compounds according to the invention can be used in their final non-salt form. On the other hand, the present invention also encompasses the use of these compounds in the form of their pharmaceutically acceptable salts, which can be derived from various organic and inorganic acids and bases by procedures known in the art. Pharmaceutically acceptable salt forms of the compounds of the formula I are for the most part prepared by conventional methods. If the compound of the formula I contains a car- boxyl group, one of its suitable salts can be formed by reacting the compound with a suitable base to give the corresponding base-addition salt. Such bases are, for example, alkali metal hydroxides, including potassium hydroxide, sodium hydroxide and lithium hydroxide; alkaline earth metal hydroxides, such as barium hydroxide and calcium hydroxide; alkali metal alkoxides, for example potassium ethoxide and sodium propoxide; and various organic bases, such as piperidine, diethanolamine and N-methyl- glutamine. The aluminium salts of the compounds of the formula I are likewise included. In the case of certain compounds of the formula I, acid- addition salts can be formed by treating these compounds with pharmaceutically acceptable organic and inorganic acids, for example hydrogen halides, such as hydrogen chloride, hydrogen bromide or hydrogen iodide, other mineral acids and corresponding salts thereof, such as sulfate, nitrate or phosphate and the like, and alkyl- and monoarylsulfonates, such as ethanesulfonate, toluenesulfonate and benzenesulfonate, and other organic acids and corresponding salts thereof, such as acetate, trifluoro- acetate, tartrate, maleate, succinate, citrate, benzoate, salicylate, ascor- bate and the like. Accordingly, pharmaceutically acceptable acid-addition salts of the compounds of the formula I include the following: acetate, adi- pate, alginate, arginate, aspartate, benzoate, benzenesulfonate (besylate), bisulfate, bisulfite, bromide, butyrate, camphorate, camphorsulfonate, caprylate, chloride, chlorobenzoate, citrate, cyclopentanepropionate, diglu- conate, dihydrogenphosphate, dinitrobenzoate, dodecylsulfate, ethanesulfonate, fumarate, formate, galacterate (from mucic acid), galacturonate, glucoheptanoate, gluconate, glutamate, glycerophosphate, hemisuccinate, hemisulfate, heptanoate, hexanoate, hippurate, hydrochloride, hydro- bromide, hydroiodide, 2-hydroxyethanesulfonate, iodide, isethionate, iso- butyrate, lactate, lactobionate, malate, maleate, malonate, mandelate, metaphosphate, methanesulfonate, methylbenzoate, monohydrogenphos- phate, 2-naphthalenesulfonate, nicotinate, nitrate, oxalate, oleate, palmo- ate, pectinate, persulfate, phenylacetate, 3-phenylpropionate, phosphate, phosphonate, phthalate, but this does not represent a restriction. Furthermore, the base salts of the compounds according to the invention include aluminium, ammonium, calcium, copper, iron(lll), iron(ll), lithium, magnesium, manganese(lll), manganese(ll), potassium, sodium and zinc salts, but this is not intended to represent a restriction. Of the above-men- tioned salts, preference is given to ammonium; the alkali metal salts sodium and potassium, and the alkaline earth metal salts calcium and magnesium. Salts of the compounds of the formula I which are derived from pharmaceutically acceptable organic non-toxic bases include salts of primary, secondary and tertiary amines, substituted amines, also including naturally occurring substituted amines, cyclic amines, and basic ion exchanger resins, for example arginine, betaine, caffeine, chloroprocaine, choline, N.N'-dibenzylethylenediamine (benzathine), dicyclohexylamine, diethanolamine, diethylamine, 2-diethylaminoethanol, 2-dimethylamino- ethanol, ethanolamine, ethylenediamine, N-ethylmorpholine, N-ethyl- piperidine, glucamine, glucosamine, histidine, hydrabamine, isopropyl- amine, lidocaine, lysine, meglumine, N-methyl-D-glucamine, morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethanolamine, triethylamine, trimethylamine, tripropylamine and tris- (hydroxymethyl)methylamine (tromethamine), but this is not intended to represent a restriction. Compounds of the present invention which contain basic nitrogen-containing groups can be quaternised using agents such as (Ci-C4)alkyl halides, for example methyl, ethyl, isopropyl and tert-butyl chloride, bromide and iodide; di(Ci-C4)alkyl sulfates, for example dimethyl, diethyl and diamyl sulfate; (Cio-Ci8)alkyl halides, for example decyl, dodecyl, lauryl, myristyl and stearyl chloride, bromide and iodide; and aryl(Ci-C4)alkyl halides, for example benzyl chloride and phenethyl bromide. Both water- and oil-soluble compounds according to the invention can be prepared using such salts. The above-mentioned pharmaceutical salts which are preferred include acetate, trifluoroacetate, besylate, citrate, fumarate, gluconate, hemisucci- nate, hippurate, hydrochloride, hydrobromide, isethionate, mandelate, meglumine, nitrate, oleate, phosphonate, pivalate, sodium phosphate, stea- rate, sulfate, subsalicylate, tartrate, thiomalate, tosylate and trometh- amine, but this is not intended to represent a restriction.

Particular preference is given to hydrochloride, dihydrochloride, hydro- bromide, maleate, mesylate, phosphate, sulfate and succinate.

The acid-addition salts of basic compounds of the formula I are prepared by bringing the free base form into contact with a sufficient amount of the desired acid, causing the formation of the salt in a conventional manner. The free base can be regenerated by bringing the salt form into contact with a base and isolating the free base in a conventional manner. The free base forms differ in a certain respect from the corresponding salt forms thereof with respect to certain physical properties, such as solubility in polar solvents; for the purposes of the invention, however, the salts otherwise correspond to the respective free base forms thereof.

As mentioned, the pharmaceutically acceptable base-addition salts of the compounds of the formula I are formed with metals or amines, such as alkali metals and alkaline earth metals or organic amines. Preferred metals are sodium, potassium, magnesium and calcium. Preferred organic amines are Ν,Ν'-dibenzylethylenediamine, chloroprocaine, choline, di- ethanolamine, ethylenediamine, N-methyl-D-glucamine and procaine.

The base-addition salts of acidic compounds according to the invention are prepared by bringing the free acid form into contact with a sufficient amount of the desired base, causing the formation of the salt in a conventional manner. The free acid can be regenerated by bringing the salt form into contact with an acid and isolating the free acid in a conventional man- ner. The free acid forms differ in a certain respect from the corresponding salt forms thereof with respect to certain physical properties, such as solubility in polar solvents; for the purposes of the invention, however, the salts otherwise correspond to the respective free acid forms thereof.

If a compound according to the invention contains more than one group which is capable of forming pharmaceutically acceptable salts of this type, the invention also encompasses multiple salts. Typical multiple salt forms include, for example, bitartrate, diacetate, difumarate, dimeglumine, diphosphate, disodium and trihydrochloride, but this is not intended to represent a restriction.

With regard to that stated above, it can be seen that the expression "pharmaceutically acceptable salt" in the present connection is taken to mean an active ingredient which comprises a compound of the formula I in the form of one of its salts, in particular if this salt form imparts improved pharmacokinetic properties on the active ingredient compared with the free form of the active ingredient or any other salt form of the active ingredient used earlier. The pharmaceutically acceptable salt form of the active ingredient can also provide this active ingredient for the first time with a desired pharmacokinetic property which it did not have earlier and can even have a positive influence on the pharmacodynamics of this active ingredient with respect to its therapeutic efficacy in the body.

Isotopes

There is furthermore intended that a compound of the formula I includes isotope-labelled forms thereof. An isotope-labelled form of a compound of the formula I is identical to this compound apart from the fact that one or more atoms of the compound have been replaced by an atom or atoms having an atomic mass or mass number which differs from the atomic mass or mass number of the atom which usually occurs naturally.

Examples of isotopes which are readily commercially available and which can be incorporated into a compound of the formula I by well-known methods include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, fluorine and chlorine, for example 2 H, 3 H, 13 C, 1 C, 5 N, 8 0, 17 0, 31 P, 32 P, 35 S, 18 F and 36 CI, respectively. A compound of the formula I, a prodrug, thereof or a pharmaceutically acceptable salt of either which contains one or more of the above-mentioned isotopes and/or other iso-topes of other atoms is intended to be part of the present invention. An isotope-labelled compound of the formula I can be used in a number of beneficial ways. For example, an isotope-labelled compound of the formula I into which, for example, a radioisotope, such as 3 H or 14 C, has been incorporated is suitable for medicament and/or substrate tissue distribution assays. These radioisotopes, i.e. tritium ( 3 H) and carbon-14 ( 14 C), are particularly preferred owing to simple preparation and excellent detectabihty. Incorporation of heavier isotopes, for example deuterium ( 2 H), into a compound of the formula I has therapeutic advantages owing to the higher metabolic stability of this isotope-labelled compound. Higher metabolic stability translates directly into an increased in vivo half-life or lower dosages, which under most circumstances would represent a preferred embodiment of the present invention. An isotope-labelled compound of the formula I can usually be prepared by carrying out the procedures disclosed in the synthesis schemes and the related

description, in the example part and in the preparation part in the present text, replacing a non-isotope-labelled reactant by a readily available isotope-labelled reactant.

Deuterium ( 2 H) can also be incorporated into a compound of the formula I for the purpose in order to manipulate the oxidative metabolism of the compound by way of the primary kinetic isotope effect. The primary kinetic isotope effect is a change of the rate for a chemical reaction that results from exchange of isotopic nuclei, which in turn is caused by the change in ground state energies necessary for covalent bond formation after this isotopic exchange. Exchange of a heavier isotope usually results in a lowering of the ground state energy for a chemical bond and thus cause a reduction in the rate in rate-limiting bond breakage. If the bond breakage occurs in or in the vicinity of a saddle-point region along the coordinate of a multi-product reaction, the product distribution ratios can be altered substantially. For explanation: if deuterium is bonded to a carbon atom at a non-exchangeable position, rate differences of k /kD = 2-7 are typical. If this rate difference is successfully applied to a compound of the formula I that is susceptible to oxidation, the profile of this compound in vivo can be drastically modified and result in improved pharmacokinetic properties.

When discovering and developing therapeutic agents, the person skilled in the art attempts to optimise pharmacokinetic parameters while retaining desirable in vitro properties. It is reasonable to assume that many compounds with poor pharmacokinetic profiles are susceptible to oxidative metabolism. In vitro liver microsomal assays currently available provide valuable information on the course of oxidative metabolism of this type, which in turn permits the rational design of deuterated compounds of the formula I with improved stability through resistance to such oxidative meta-bolism. Significant improvements in the pharmacokinetic profiles of compounds of the formula I are thereby obtained, and can be expressed quantitatively in terms of increases in the in vivo half-life (t1/2),

concen-tra-tion at maximum therapeutic effect (Cmax), area under the dose response curve (AUC), and F; and in terms of reduced clearance, dose and materi-als costs.

The following is intended to illustrate the above: a compound of the formula I which has multiple potential sites of attack for oxidative

metabolism, for example benzylic hydrogen atoms and hydrogen atoms bonded to a nitrogen atom, is prepared as a series of analogues in which various combinations of hydrogen atoms are replaced by deuterium atoms, so that some, most or all of these hydrogen atoms have been replaced by deuterium atoms. Half-life determinations enable favourable and accurate determination of the extent of the extent to which the improve-ment in resistance to oxidative metabolism has improved. In this way, it is deter-mined that the half-life of the parent compound can be extended by up to 100% as the result of deuterium-hydrogen exchange of this type.

Deuterium-hydrogen exchange in a compound of the formula I can also be used to achieve a favourable modification of the metabolite spectrum of the starting compound in order to diminish or eliminate undesired toxic metabolites. For example, if a toxic metabolite arises through oxidative carbon-hydrogen (C-H) bond cleavage, it can reasonably be assumed that the deuterated analogue will greatly diminish or eliminate production of the unwanted metabolite, even if the particular oxidation is not a rate- determining step. Further information on the state of the art with respect to deuterium-hydrogen exchange may be found, for example in Hanzlik et al., J. Org. Chem. 55, 3992-3997, 1990, Reider et al., J. Org. Chem. 52, 3326- 3334, 1987, Foster, Adv. Drug Res. 14, 1-40, 1985, Gillette et al,

Biochemistry 33(10) 2927-2937, 1994, and Jarman et al. Carcinogenesis 16(4), 683-688, 1993.

The invention furthermore relates to medicaments comprising at least one compound of the formula I and/or pharmaceutically acceptable derivatives, solvates and stereoisomers thereof, including mixtures thereof in all ratios, and optionally excipients and/or adjuvants.

Pharmaceutical formulations can be administered in the form of dosage units which comprise a predetermined amount of active ingredient per dosage unit. Such a unit can comprise, for example, 0.5 mg to 1 g, preferably 1 mg to 700 mg, particularly preferably 5 mg to 100 mg, of a compound according to the invention, depending on the condition treated, the method of administration and the age, weight and condition of the patient, or pharmaceutical formulations can be administered in the form of dosage units which comprise a predetermined amount of active ingredient per dosage unit. Preferred dosage unit formulations are those which comprise a daily dose or part-dose, as indicated above, or a corresponding fraction thereof of an active ingredient. Furthermore, pharmaceutical formulations of this type can be prepared using a process which is generally known in the pharmaceutical art.

Pharmaceutical formulations can be adapted for administration via any desired suitable method, for example by oral (including buccal or sublingual), rectal, nasal, topical (including buccal, sublingual or transdermal), vaginal or parenteral (including subcutaneous, intramuscular, intravenous or intradermal) methods. Such formulations can be prepared using all processes known in the pharmaceutical art by, for example, combining the active ingredient with the excipient(s) or adjuvant(s).

Pharmaceutical formulations adapted for oral administration can be administered as separate units, such as, for example, capsules or tablets; powders or granules; solutions or suspensions in aqueous or non-aqueous liquids; edible foams or foam foods; or oil-in-water liquid emulsions or water-in-oil liquid emulsions.

Thus, for example, in the case of oral administration in the form of a tablet or capsule, the active-ingredient component can be combined with an oral, non-toxic and pharmaceutically acceptable inert excipient, such as, for example, ethanol, glycerol, water and the like. Powders are prepared by comminuting the compound to a suitable fine size and mixing it with a pharmaceutical excipient comminuted in a similar manner, such as, for example, an edible carbohydrate, such as, for example, starch or mannitol. A flavour, preservative, dispersant and dye may likewise be present.

Capsules are produced by preparing a powder mixture as described above and filling shaped gelatine shells therewith. Glidants and lubricants, such as, for example, highly disperse silicic acid, talc, magnesium stearate, calcium stearate or polyethylene glycol in solid form, can be added to the powder mixture before the filling operation. A disintegrant or solubiliser, such as, for example, agar-agar, calcium carbonate or sodium carbonate, may likewise be added in order to improve the availability of the medicament after the capsule has been taken.

In addition, if desired or necessary, suitable binders, lubricants and disin- tegrants as well as dyes can likewise be incorporated into the mixture. Suitable binders include starch, gelatine, natural sugars, such as, for example, glucose or beta-lactose, sweeteners made from maize, natural and synthetic rubber, such as, for example, acacia, tragacanth or sodium alginate, carboxymethylcellulose, polyethylene glycol, waxes, and the like. The lubricants used in these dosage forms include sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and the like. The disintegrants include, without being restricted thereto, starch, methylcellulose, agar, bentonite, xanthan gum and the like. The tablets are formulated by, for example, preparing a powder mixture, granulating or dry-pressing the mixture, adding a lubricant and a disintegrant and pressing the entire mixture to give tablets. A powder mixture is prepared by mixing the compound comminuted in a suitable manner with a diluent or a base, as described above, and optionally with a binder, such as, for example, carboxymethylcellulose, an alginate, gelatine or polyviny^ pyrrolidone, a dissolution retardant, such as, for example, paraffin, an absorption accelerator, such as, for example, a quaternary salt, and/or an absorbant, such as, for example, bentonite, kaolin or dicalcium phosphate. The powder mixture can be granulated by wetting it with a binder, such as, for example, syrup, starch paste, acadia mucilage or solutions of cellulose or polymer materials and pressing it through a sieve. As an alternative to granulation, the powder mixture can be run through a tabletting machine, giving lumps of non-uniform shape, which are broken up to form granules. The granules can be lubricated by addition of stearic acid, a stearate salt, talc or mineral oil in order to prevent sticking to the tablet casting moulds. The lubricated mixture is then pressed to give tablets. The compounds according to the invention can also be combined with a free-flowing inert excipient and then pressed directly to give tablets without carrying out the granulation or dry-pressing steps. A transparent or opaque protective layer consisting of a shellac sealing layer, a layer of sugar or polymer material and a gloss layer of wax may be present. Dyes can be added to these coatings in order to be able to differentiate between different dosage units.

Oral liquids, such as, for example, solution, syrups and elixirs, can be prepared in the form of dosage units so that a given quantity comprises a pre- specified amount of the compound. Syrups can be prepared by dissolving the compound in an aqueous solution with a suitable flavour, while elixirs are prepared using a non-toxic alcoholic vehicle. Suspensions can be formulated by dispersion of the compound in a non-toxic vehicle. Solubilisers and emulsifiers, such as, for example, ethoxylated isostearyl alcohols and polyoxyethylene sorbitol ethers, preservatives, flavour additives, such as, for example, peppermint oil or natural sweeteners or saccharin, or other artificial sweeteners and the like, can likewise be added.

The dosage unit formulations for oral administration can, if desired, be encapsulated in microcapsules. The formulation can also be prepared in such a way that the release is extended or retarded, such as, for example, by coating or embedding of particulate material in polymers, wax and the like.

The compounds of the formula I and pharmaceutically salts, tautomers and stereoisomers thereof can also be administered in the form of liposome delivery systems, such as, for example, small unilamellar vesicles, large unilamellar vesicles and multilamellar vesicles. Liposomes can be formed from various phospholipids, such as, for example, cholesterol, stearylamine or phosphatidylcholines. The compounds of the formula I and the salts, tautomers and

stereoisomers thereof can also be delivered using monoclonal antibodies as individual carriers to which the compound molecules are coupled. The compounds can also be coupled to soluble polymers as targeted

medicament carriers. Such polymers may encompass polyvinylpyrrolidone, pyran copolymer, polyhydroxypropylmethacrylamidophenol, polyhydroxy- ethylaspartamidophenol or polyethylene oxide polylysine, substituted by palmitoyl radicals. The compounds may furthermore be coupled to a class of biodegradable polymers which are suitable for achieving controlled release of a medicament, for example polylactic acid, poly-epsilon-capro- lactone, polyhydroxybutyric acid, polyorthoesters, polyacetals, polydihy- droxypyrans, polycyanoacrylates and crosslinked or amphipathic block copolymers of hydrogels.

Pharmaceutical formulations adapted for transdermal administration can be administered as independent plasters for extended, close contact with the epidermis of the recipient. Thus, for example, the active ingredient can be delivered from the plaster by iontophoresis.

Pharmaceutical compounds adapted for topical administration can be formulated as ointments, creams, suspensions, lotions, powders, solutions, pastes, gels, sprays, aerosols or oils.

For the treatment of the eye or other external tissue, for example mouth and skin, the formulations are preferably applied as topical ointment or cream. In the case of formulation to give an ointment, the active ingredient can be employed either with a paraffinic or a water-miscible cream base. Alternatively, the active ingredient can be formulated to give a cream with an oil-in-water cream base or a water-in-oil base. Pharmaceutical formulations adapted for topical application to the eye include eye drops, in which the active ingredient is dissolved or suspended in a suitable carrier, in particular an aqueous solvent.

Pharmaceutical formulations adapted for topical application in the mouth encompass lozenges, pastilles and mouthwashes.

Pharmaceutical formulations adapted for rectal administration can be administered in the form of suppositories or enemas.

Pharmaceutical formulations adapted for nasal administration in which the carrier substance is a solid comprise a coarse powder having a particle size, for example, in the range 20-500 microns, which is administered in the manner in which snuff is taken, i.e. by rapid inhalation via the nasal passages from a container containing the powder held close to the nose. Suitable formulations for administration as nasal spray or nose drops with a liquid as carrier substance encompass active-ingredient solutions in water or oil.

Pharmaceutical formulations adapted for administration by inhalation encompass finely particulate dusts or mists, which can be generated by various types of pressurised dispensers with aerosols, nebulisers or insufflators.

Pharmaceutical formulations adapted for vaginal administration can be administered as pessaries, tampons, creams, gels, pastes, foams or spray formulations.

Pharmaceutical formulations adapted for parenteral administration include aqueous and non-aqueous sterile injection solutions comprising antioxidants, buffers, bacteriostatics and solutes, by means of which the formulation is rendered isotonic with the blood of the recipient to be treated; and aqueous and non-aqueous sterile suspensions, which may comprise suspension media and thickeners. The formulations can be administered in single-dose or multidose containers, for example sealed ampoules and vials, and stored in freeze-dried (lyophilised) state, so that only the addition of the sterile carrier liquid, for example water for injection purposes, immediately before use is necessary. Injection solutions and suspensions prepared in accordance with the recipe can be prepared from sterile powders, granules and tablets.

It goes without saying that, in addition to the above particularly mentioned constituents, the formulations may also comprise other agents usual in the art with respect to the particular type of formulation; thus, for example, formulations which are suitable for oral administration may comprise flavours.

A therapeutically effective amount of a compound of the formula I depends on a number of factors, including, for example, the age and weight of the animal, the precise condition that requires treatment, and its severity, the nature of the formulation and the method of administration, and is ultimately determined by the treating doctor or vet. However, an effective amount of a compound according to the invention is generally in the range from 0.1 to 100 mg/kg of body weight of the recipient (mammal) per day and particularly typically in the range from 1 to 10 mg/kg of body weight per day. Thus, the actual amount per day for an adult mammal weighing 70 kg is usually between 70 and 700 mg, where this amount can be administered as a single dose per day or usually in a series of part-doses (such as, for example, two, three, four, five or six) per day, so that the total daily dose is the same. An effective amount of a salt or solvate or of a physiologically functional derivative thereof can be determined as the fraction of the effective amount of the compound according to the invention perse. It can be assumed that similar doses are suitable for the treatment of other conditions mentioned above. A combined treatment of this type can be achieved with the aid of simultaneous, consecutive or separate dispensing of the individual components of the treatment. Combination products of this type employ the compounds according to the invention.

The invention furthermore relates to medicaments comprising at least one compound of the formula I and/or pharmaceutically acceptable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios, and at least one further medicament active ingredient.

The invention also relates to a set (kit) consisting of separate packs of

(a) an effective amount of a compound of the formula I and/or pharmaceutically acceptable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios,

and

(b) an effective amount of a further medicament active ingredient.

The set comprises suitable containers, such as boxes, individual bottles, bags or ampoules. The set may, for example, comprise separate ampoules, each containing an effective amount of a compound of the formula I and/or pharmaceutically acceptable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios,

and an effective amount of a further medicament active ingredient in dissolved or lyophilised form.

"Treating" as used herein, means an alleviation, in whole or in part, of symptoms associated with a disorder or disease, or slowing, or halting of further progression or worsening of those symptoms, or prevention or prophylaxis of the disease or disorder in a subject at risk for developing the disease or disorder. The term "effective amount" in connection with a compound of formula (I) can mean an amount capable of alleviating, in whole or in part, symptoms associated with a disorder or disease, or slowing or halting further progression or worsening of those symptoms, or preventing or providing prophylaxis for the disease or disorder in a subject having or at risk for developing a disease disclosed herein, such as inflammatory conditions, immunological conditions, cancer or metabolic conditions.

In one embodiment an effective amount of a compound of formula (I) is an amount that inhibits a tankyrase in a cell, such as, for example, in vitro or in vivo. In some embodiments, the effective amount of the compound of formula (I) inhibits tankyrase in a cell by 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or 99%, compared to the activity of tankyrase in an untreated cell. The effective amount of the compound of formula (I), for example in a pharmaceutical composition, may be at a level that will exercise the desired effect; for example, about 0.005 mg/kg of a subject's body weight to about 10 mg/kg of a subject's body weight in unit dosage for both oral and parenteral administration.

USE

The present compounds are suitable as pharmaceutical active ingredients for mammals, especially for humans, in the treatment of cancer, multiple sclerosis, cardiovascular diseases, central nervous system injury and different forms of inflammation. The present invention encompasses the use of the compounds of the formula I and/or pharmaceutically acceptable salts, tautomers and

stereoisomers thereof for the preparation of a medicament for the treatment or prevention of cancer, multiple sclerosis, cardiovascular diseases, central nervous system injury and different forms of

inflammation. Examples of inflammatory diseases include rheumatoid arthritis, psoriasis, contact dermatitis, delayed hypersensitivity reaction and the like.

Also encompassed is the use of the compounds of the formula I and/or pharmaceutically acceptable salts, tautomers and stereoisomers thereof for the preparation of a medicament for the treatment or prevention of a tankyrase-induced disease or a tankyrase-induced condition in a mammal, in which to this method a therapeutically effective amount of a compound according to the invention is administered to a sick mammal in need of such treatment. The therapeutic amount varies according to the specific disease and can be determined by the person skilled in the art without undue effort.

The expression "tankyrase-induced diseases or conditions" refers to pathological conditions that depend on the activity of one or more tankyrases. Diseases associated with tankyrase activity include cancer, multiple sclerosis, cardiovascular diseases, central nervous system injury and different forms of inflammation .

The present invention specifically relates to compounds of the formula I and pharmaceutically acceptable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios,

for the use for the treatment of diseases in which the inhibition, regulation and/or modulation inhibition of tankyrase plays a role.

The present invention specifically relates to compounds of the formula I and pharmaceutically acceptable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios, for the use for the inhibition of tankyrase. The present invention specifically relates to compounds of the formula I and pharmaceutically acceptable salts, tautomers and stereoisomers thereof, including mixtures thereof in all ratios, for the use for the treatment of cancer, multiple sclerosis, cardiovascular diseases, central nervous system injury and different forms of inflammation.

The present invention specifically relates to methods for treating or preventing cancer, multiple sclerosis, cardiovascular diseases, central nervous system injury and different forms of inflammation , comprising administering to a subject in need thereof an effective amount of a compound of formula I or a pharmaceutically acceptable salt, tautomer, stereoisomer or solvate thereof.

Representative cancers that compounds of formula I are useful for treating or preventing include, but are not limited to, cancer of the head, neck, eye, mouth, throat, esophagus, bronchus, larynx, pharynx, chest, bone, lung, colon, rectum, stomach, prostate, urinary bladder, uterine, cervix, breast, ovaries, testicles or other reproductive organs, skin, thyroid, blood, lymph nodes, kidney, liver, pancreas, brain, central nervous system, solid tumors and blood-borne tumors.

Representative cardiovascular diseases that compounds of formula I are useful for treating or preventing include, but are not limited to, restenosis, atherosclerosis and its consequences such as stroke, myocardial infarction, ischemic damage to the heart, lung, gut, kidney, liver, pancreas, spleen or brain.

The present invention relates to a method of treating a proliferative,

autoimmune, anti inflammatory or infectious disease disorder that

comprises administering to a subject in need thereof a therapeutically effective amount of a compound of formula I .

Preferably, the present invention relates to a method wherein the disease is a cancer. W 201

- 31 -

Particularly preferable, the present invention relates to a method wherein the disease is a cancer, wherein administration is simultaneous, sequential or in alternation with administration of at least one other active drug agent. The disclosed compounds of the formula I can be administered in combination with other known therapeutic agents, including anticancer agents.

As used here, the term "anticancer agent" relates to any agent which is administered to a patient with cancer for the purposes of treating the cancer.

The anti-cancer treatment defined above may be applied as a monotherapy or may involve, in addition to the herein disclosed compounds of formula I, conventional surgery or radiotherapy or medicinal therapy. Such medicinal therapy, e.g. a chemotherapy or a targeted therapy, may include one or more, but preferably one, of the following anti-tumor agents:

Alkylating agents

such as altretamine, bendamustine, busulfan, carmustine, chlorambucil, chlormethine, cyclophosphamide, dacarbazine, ifosfamide, improsulfan, tosilate, lomustine, melphalan, mitobronitol, mitolactol, nimustine, ranimustine, temozolomide, thiotepa, treosulfan, mechloretamine, carboquone;

apaziquone, fotemustine, glufosfamide, palifosfamide, pipobroman,

trofosfamide, uramustine, TH-302 4 , VAL-083 4 ;

Platinum Compounds

such as carboplatin, cisplatin, eptaplatin, miriplatine hydrate, oxaliplatin, lobaplatin, nedaplatin, picoplatin, satraplatin;

lobaplatin, nedaplatin, picoplatin, satraplatin;

DNA altering agents

such as amrubicin, bisantrene, decitabine, mitoxantrone, procarbazine, trabectedin, clofarabine;

amsacrine, brostallicin, pixantrone, laromustine 1 3 ;

Topoisomerase Inhibitors such as etoposide, irinotecan, razoxane, sobuzoxane, teniposide, topotecan; amonafide, belotecan, elliptinium acetate, voreloxin;

Microtubule modifiers

such as cabazitaxel, docetaxel, eribulin, ixabepilone, paclitaxel, vinblastine, vincristine, vinorelbine, vindesine, vinflunine;

fosbretabulin, tesetaxel;

Antimetabolites

such as asparaginase 3 , azacitidine, calcium levofolinate, capecitabine,0 cladribine, cytarabine, enocitabine, floxuridine, fludarabine, fluorouracil,

gemcitabine, mercaptopurine, methotrexate, nelarabine, pemetrexed, pralatrexate, azathioprine, thioguanine, carmofur;

doxifluridine, elacytarabine, raltitrexed, sapacitabine, tegafur 2 3 , trimetrexate; Anticancer antibiotics

such as bleomycin, dactinomycin, doxorubicin, epirubicin, idarubicin, levamisole, miltefosine, mitomycin C, romidepsin, streptozocin, valrubicin, zinostatin, zorubicin, daunurobicin, plicamycin;

aclarubicin, peplomycin, pirarubicin;

Hormones/Antagonists

such as abarelix, abiraterone, bicalutamide, buserelin, calusterone, chlorotrianisene, degarelix, dexamethasone, estradiol, fluocortolone fluoxymesterone, flutamide, fulvestrant, goserelin, histrelin, leuprorelin,5 megestrol, mitotane, nafarelin, nandrolone, nilutamide, octreotide,

prednisolone, raloxifene, tamoxifen, thyrotropin alfa, toremifene, trilostane, triptorelin, diethylstilbestrol;

acolbifene, danazol, deslorelin, epitiostanol, orteronel, enzalutamide 1 3 ;

Q Aromatase inhibitors

such as aminoglutethimide, anastrozole, exemestane, fadrozole, letrozole, testolactone;

formestane;

Small molecule kinase inhibitors

5

such as crizotinib, dasatinib, eriotinib, imatinib, lapatinib, nilotinib, pazopanib, regorafenib, ruxolitinib, sorafenib, sunitinib, vandetanib, vemurafenib, bosutinib, gefitinib, axitinib;

afatinib, alisertib, dabrafenib, dacomitinib, dinaciclib, dovitinib, enzastaurin, nintedanib, lenvatinib, linifanib, linsitinib, masitinib, midostaurin, motesanib, neratinib, orantinib, perifosine, ponatinib, radotinib, rigosertib, tipifarnib,

5

tivantinib, tivozanib, trametinib, pimasertib, brivanib alaninate, cediranib, apatinib 4 , cabozantinib S-malate 1 3 , ibrutinib 1 - 3 , icotinib 4 , buparlisib 2 , cipatinib 4 , cobimetinib 1 ' 3 , idelalisib 1 fedratinib 1 , XL-647 4 ;

Photosensitizers

10 such as methoxsalen 3 ;

porfimer sodium, talaporfin, temoporfin;

Antibodies

such as alemtuzumab, besilesomab, brentuximab vedotin, cetuximab,

«15 denosumab, ipilimumab, ofatumumab, panitumumab, rituximab, tositumomab, trastuzumab, bevacizumab, pertuzumab 2 - 3 ;

catumaxomab, elotuzumab, epratuzumab, farletuzumab, mogamulizumab, necitumumab, nimotuzumab, obinutuzumab, ocaratuzumab, oregovomab, ramucirumab, rilotumumab, siltuximab, tocilizumab, zalutumumab,

20

zanolimumab, matuzumab, dalotuzumab 1 2 * 3 , onartuzumab 1,3 , racotumomab , tabalumab 1 3 , EMD-525797 4 , nivolumab 1 3 ;

Cytokines

such as aldesleukin, interferon alfa 2 , interferon alfa2a 3 , interferon alfa2b 2 3 ; 25 celmoleukin, tasonermin, teceleukin, oprelvekin 1 3 , recombinant interferon

beta-1a 4 ;

Drug Conjugates

such as denileukin diftitox, ibritumomab tiuxetan, iobenguane 1123,

2Q prednimustine, trastuzumab emtansine, estramustine, gemtuzumab,

ozogamicin, aflibercept;

cintredekin besudotox, edotreotide, inotuzumab ozogamicin, naptumomab estafenatox, oportuzumab monatox, technetium (99mTc) arcitumomab 1 3 , vintafolide 1 3 ;

^ Vaccines such as sipuleucel 3 ; vitespen 3 , emepepimut-S 3 , oncoVAX 4 , rindopepimut 3 , troVax 4 , MGN-1601 4 , MGN-1703 4 ;

Miscellaneous

alitretinoin, bexarotene, bortezomib, everolimus, ibandronic acid, imiquimod, lenalidomide, lentinan, metirosine, mifamurtide, pamidronic acid,

pegaspargase, pentostatin, sipuleucel 3 , sizofiran, tamibarotene, temsirolimus, thalidomide, tretinoin, vismodegib, zoledronic acid, vorinostat;

celecoxib, cilengitide, entinostat, etanidazole, ganetespib, idronoxil, iniparib, ixazomib, lonidamine, nimorazole, panobinostat, peretinoin, plitidepsin, pomalidomide, procodazol, ridaforolimus, tasquinimod, telotristat, thymalfasin, tirapazamine, tosedostat, trabedersen, ubenimex, valspodar, gendicine 4 , picibanil 4 , reolysin 4 , retaspimycin hydrochloride 1 3 , trebananib 2 3 , virulizin 4 , carfilzomib 1 3 , endostatin 4 , immucothel 4 , belinostat 3 , MGN-1703 4 ;

1 Prop. INN (Proposed International Nonproprietary Name)

2 Rec. INN (Recommended International Nonproprietary Names)

3 USAN (United States Adopted Name)

4 no INN.

The following abbreviations refer respectively to the definitions below:

aq (aqueous), h (hour), g (gram), L (liter), mg (milligram), MHz (Megahertz), min. (minute), mm (millimeter), mmol (millimole), mM (millimolar), m.p.

(melting point), eq (equivalent), mL (milliliter), L (microliter), ACN (acetonitrile), AcOH (acetic acid), CDC (deuterated chloroform), CD3OD (deuterated methanol), CH3CN (acetonitrile), c-hex (cyclohexane), DCC (dicyclohexyl carbodiimide), DCM (dichloromethane), DIC (diisopropyl carbodiimide), DIEA (diisopropylethyl-amine), DMF (dimethylformamide), DMSO

(dimethylsulfoxide), DMSO-de (deuterated dimethylsulfoxide), EDC (1-(3- dimethyl-amino-propyl)-3-ethylcarbodiimide), ESI (Electro-spray ionization), EtOAc (ethyl acetate), EfcO (diethyl ether), EtOH (ethanol), HATU

(dimethylamino-([1 ,2,3]triazolo[4,5-b]pyridin-3-yloxy)-methylene]-dimethyl- ammonium hexafluorophosphate), HPLC (High Performance Liquid Chromatography), i-PrOH (2-propanol), K2CO3 (potassium carbonate), LC (Liquid Chromatography), MeOH (methanol), MgSO4 (magnesium sulfate), MS (mass spectrometry), MTBE (Methyl tert-butyl ether), NaHCO3 (sodium bicarbonate), NaBH4 (sodium borohydride), NMM (N-methyl morpholine),

NMR (Nuclear Magnetic Resonance), PyBOP (benzotriazole-1-yl-oxy-tris- pyrrolidino-phosphonium hexafluorophosphate), RT (room temperature), Rt (retention time), SPE (solid phase extraction), TBTU (2-(1-H-benzotriazole-1- yl)-1 ,1 ,3,3-tetramethyluromium tetrafluoro borate), TEA (triethylamine), TFA (trifluoroacetic acid), THF (tetrahydrofuran), TLC (Thin Layer

Chromatography), UV (Ultraviolet).

Description of the in vitro assays

Abbreviations:

GST = Glutathione-S-transferase

FRET= Fluorescence resonance energy transfer

HTRF® = (homogenous time resolved fluorescence)

HEPES = 4-(2-hydroxyethyl)-1-piperazine ethanesulfonic acid buffer DTT = Dithiothreitol BSA = bovine serum albumin CHAPS = detergent;

CHAPS = 3-[(3-cholamidopropyl)dimethylammonio]-1 -propanesulfonate

Streptavidin-XLent® is a high grade streptavidin-XL665 conjugate for which the coupling conditions have been optimized to yield a conjugate with enhanced performances for some assays, particularly those requiring high sensitivity.

Measurement of cellular inhibition of tankyrase

Since Tankyrases have been described to modulate cellular level of Axin2 (Huang et al., 2009; Nature) the increase of Axin2 level is used as read-out for determination of cellular inhibition of Tankyrases in a Luminex based assay.

Cells of the colon carcinoma cell line DLD1 are plated in 96 well plates with 1.5x10 4 cells per well. Next day, cells are treated with a serial dilution of test compound in seven steps as triplicates with a final DMSO

concentration of 0.3%. After 24 hours, cells are lysed in lysis buffer (20mM Tris/HCI pH 8.0, 150 mM NaCI, 1% NP40, 10% Glycerol) and lysates are cleared by centrifugation through a 96 well filter plate (0.65pm). Axin2 protein is isolated from cell lysates by incubation with a monoclonal anti- Axin2 antibody (R&D Systems #MAB6078) that is bound to fluorescent carboxybeads. Then, bound Axin2 is specifically detected with a polyclonal anti-Axin2 antibody (Cell Signaling #2151) and an appropriate PE- fluorescent secondary antibody. The amount of isolated Axin2 protein is determined in a Luminex 200 machine (Luminex Corporation) according to the manufacturer's instruction by counting 100 events per well. Inhibition of Tankyrase by test compounds results in higher levels of Axin2 which directly correlates with an increase of detectable fluorescence. As controls cells are treated with solvent alone (neutral control) and with a Tankyrase reference inhibitor IWR-2 (3E-06 M) which refers as control for maximum increase of Axin2. For analysis, the obtained data are normalized against the untreated solvent control and fitted for determination of the EC50

values using the Assay Explorer software (Accelrys).

Description of the PARP1 assay

Biochemical activity testing of PARP-1 : Autoparsylation assay

The autoparsylation assay is run in two steps: the enzymatic reaction in which His-tagged Parp-1 transfers biotinylated ADP-ribose/ADP-ribose to itself from biotinylated NAD/NAD as co-substrate and the detection reaction where a time resolved FRET between cryptate labelled anti-His antibody bound to the His tag of the enzyme and Xlent® labelled-streptavidin bound the biotin- parsylation residue is analysed. The autoparsylation activity is detectable directly via the increase in HTRF signal.

The autoparsylation assay is performed as 384-well HTRF® (Cisbio, Codolet, France) assay format in Greiner low volume nb 384-well microtiter plates. 35 nM His-tagged Parp-1 (human, recombinant, Enzo Life Sciences GmbH, Lorrach, Germany) and a mixture of 125 nM bio-NAD (Biolog, Life science Inst., Bremen, Germany) and 800 nM NAD as co-substrate are incubated in a total volume of 6 μΙ (100 rtiM Tris/HCI, 4 mM Mg-chloride, 0.01 % IGEPAL® CA630, 1mM DTT , 0.5 % DMSO, pH 8, 13 ng/μΙ activated DNA (BPS

Bioscience, San Diego, US)) in the absence or presence of the test compound (10 dilution concentrations) for 150 min at 23°C. The reaction is stopped by the addition of 4 pi of the Stop/detection solution (70 nM SA- Xlent® (Cisbio, Codolet, France), 2.5 nM Anti-His-K® (Eu-labelled anti-His, Cisbio, Codolet, France) in 50 mM HEPES, 400 mM KF, 0.1 % BSA, 20 mM EDTA, pH 7.0). After 1h incubation at room temperature the HTRF iss measured with an Envision multimode reader (Perkin Elmer LAS Germany GmbH) at excitation wavelength 340 nm (laser mode) and emission

wavelengths 615 nm and 665 nm. The ratio of the emission signals is determined. The full value used is the inhibitor-free reaction. The

pharmacological zero value used is Olaparib (LCIabs, Woburn, US) in a final concentration of 1 μΜ. The inhibitory values (IC50) are determined using either the program Symyx Assay Explorer® or Condosseo® from GeneData.

Description of the TNKS1 and TNKS2 ELISA assay

Biochemical activity testing of TNKS 1 and 2: activity ELISA (Autoparsylation assay)

For analysis of autoparsylation activity of TNKS 1 and 2 an activity ELISA iss performed: In the first step GST tagged TNKS is captured on a Glutathione coated plate. Then the activity assay with biotinylated NAD is performed in the absence/presence of the compounds. During the enzymatic reaction GST tagged TNKS transfers biotinylated ADP-ribose to itself from biotinylated NAD as co-substrate. For the detection streptavidin-HRP conjugate is added that binds to the biotinylated TNKS and is thereby captured to the plates. The amount of biotinylated resp. autoparsylated TNKS is detected with a

luminescence substrate for HRP. The level of the luminescence signal correlats directly with the amount of autoparsylated TNKS and therefore with activity of TNKS.

The acitivity ELISA is performed in 384 well Glutathione coated microtiter plates (Express capture Glutathione coated plate, Biocat, Heidelberg,

Germany). The plates are pre-equilibrated with PBS. Then the plates are incubated with 50 μΙ 20 ng/well GST-tagged Tnks-1 (1023-1327 aa, prepared in-house), respectively GST-tagged Tnks-2 (873-1166 aa, prepared in-house) in assay buffer (50 mM HEPES, 4 mM Mg-chloride, 0.05 % Pluronic F-68, 2 mM DTT, pH 7.7) overnight at 4°C. The plates are washed 3 times with PBS- Tween-20. The wells are blocked by incubation at room temperature for 20 minutes with 50 pi blocking buffer (PBS, 0.05 % Tween-20, 0.5 % BSA).

Afterwards the plates are washed 3 times with PBS-Tween-20. The enzymatic reaction is performed in 50 μΙ reaction solution (50 mM HEPES, 4 mM Mg- chloride, 0.05 % Pluronic F-68, 1.4 mM DTT, 0.5 % DMSO, pH 7.7) with 10 μΜ bio-NAD (Biolog, Life science Inst., Bremen, Germany) as co-substrate in the absence or presence of the test compound (10 dilution concentrations) for 1 hour at 30°C. The reaction is stopped by 3 times washing with PBS-Tween- 20. For the detection 50 μΙ of 20ng^l Streptavidin, HRP conjugate (MoBiTec, Gottingen, Germany) in PBS/0.05%Tween-20/0.01%BSA are added and the plates are incubated for 30 minutes at room temperature. After three times washing with PBS-Tween-20 50 μΙ of SuperSignal ELISA Femto Maximum sensitivity substrate solution (ThermoFisherScientific (Pierce), Bonn,

Germany) are added. Following a 1 minute incubation at room temperature luminescence signals are measured with an Envision multimode reader (Perkin Elmer LAS Germany GmbH) at 700 nm. The full value used is the inhibitor-free reaction. The pharmacological zero value used is XAV-939

(Tocris) in a final concentration of 5 μΜ. The inhibitory values (IC50) are determined using either the program Symyx Assay Explorer® or Condosseo® from GeneData.

Above and below, all temperatures are indicated in°C. In the following ex- amples, "conventional work-up" means: water is added if necessary, the pH is adjusted, if necessary, to values between 2 and 10, depending on the constitution of the end product, the mixture is extracted with ethyl acetate or dichloromethane, the phases are separated, the organic phase is dried over sodium sulfate and evaporated, and the residue is purified by chromatography on silica gel and/or by crystallisation. Rf values on silica gel; eluent: ethyl acetate/methanol 9:1. Test Method Microsomal Stability (Intrinsic Clearance)

A microsomal stability assay is used to measure in vitro clearance (Clint). The assay involves measuring the rate of disappearance of a compound due to its intrinsic attitude to be metabolized ("intrinsic" meaning that the disappearance is not affected by other properties like permeability, binding etc. that play a role when quantifying in vivo clearance). The microsomal stability (intrinsic clearance, Clint) and thus metabolic stability is generally given as μΙ/min/mg protein. It can be visualized as the volume of solution that 1 mg of microsomes is able to clear of the compound in one minute.

Instrumentation

A Tecan Genesis workstation (RSP 150/8) was used for to perform the microsomal incubations. Analysis was carried out using a Waters

ACQUITY UPLC system coupled to an ABSciex API3000 mass

spectrometer. Data analysis was performed using Assay Explorer (Symyx).

UPLC conditions

Column: Acquity UPLC BEH C18, 2.1 x 50mm, 1.7 pm (Waters)

Mobile phases: A = 0.1 % formic acid in water; B = acetonitrile Gradient Time % A % B initial 90 10

0.47 5 95

0.65 5 95

0.66 90 10

Flow rate: 0.750 mL/min; Detection: ESI, MRM; Injection: 10 pL; Column temperature: 50°C

Chemicals

• Potassium phosphate buffer: 0.05 M potassium phosphate buffer pH 7.4 containing 1 mM MgC

• NADPH (nicotinamide adenine dinucleotide phosphate): 22.5 mg

NADPH-Na4 in 1.8 ml potassium phosphate buffer

• Acetonitrile: 50 Vol% acetonitrile (1 volume acetonitrile, 1 volume water)

• DMSO: 20 Vol% DMSO in water

• Stock solution of 20 mg/ml human or mouse liver microsomes

(protein)/ml in phosphate buffer

Stock solution of 10 mM compound in 100% DMSO

1 H NMR was recorded on Bruker DPX-300, DRX-400, AVII-400 or on a 500 MHz spectrometer, using residual signal of deuterated solvent as internal reference. Chemical shifts (δ) are reported in ppm relative to the residual solvent signal (δ = 2.49 ppm for 1 H NMR in DMSO-de). 1 H NMR data are reported as follows: chemical shift (multiplicity, coupling constants, and number of hydrogens). Multiplicity is abbreviated as follows: s (singlet), d (doublet), t (triplet), q (quartet), m (multiplet), br (broad). HPLC/MS conditions (A):

Gradient: A:B = 96:4 to 0:100 in 3.4 min; Flow rate: 2.40 ml/min

A: Water + formic acid (0.05 %); B: Acetonitrile + formic acid (0.04 %)

Column: Chromolith SpeedROD RP-18e, 50 x 4.6 mm 2

Wavelength: 220 nm HPLC/MS conditions (B):

Gradient: 0 min: 5% B, 8 min: 100% B, 8.1 min: 100% B, 8.5 min: 5% B, 10 min 5% B

flow rate: 2.0 mLJmin

A: Water + TFA (0.1 %Vol.); B: Acetonitrile + TFA (0.1 %Vol.)

Column: XBridge C8, 3.5 pm, 4.6 x 50 mm

Wavelength: 220 nm

HPLC/MS conditions (C):

Gradient: 0 min: 10% B, 2.5 min: 95% B, 4.5 min: 95% B, 4.6 min: 10% B, 6 min 10% B

flow rate: 1.5 mL/min

A: Water + TFA (0.1%Vol.); B: Acetonitrile + TFA (0.1%Vol.)

Column: Atlantis dC18, 4.6 x 50 mm, 5 pm

Wavelength: 220 nm

Pharmacological data

Table 1 Inhibition of tankyrases

of representative compounds of the formula I

Compound ECso [M] Compound No. EC 50 [M]

No. TNKS TNKS

cellular assay cellular assay

"C1" 4.50E-09 "C32" 1.10E-07 7/076484

- 42 -

"C2" 4.70 E-09 "C33" 1.50E-07

"C3" 3.00 E-08 "C34" 9.90E-09

"C4" 4.70 E-09 "C35" 5.50E-06

"C5" 3.90 E-08 "C36" 3.40E-09

"C6" 1.70 E-10 "C37" 4.80E-07

"C7" 1.10 E-09 "C38" 9.90E-10

"C8" 9.20 E-09 "C39" 1.30E-07

"C9" 1.40 E-07 "C40" 2.90E-09

"C10" 9.00 E-08 "C41" 1.10E-08

"C11" 5.00 E-09 "C42" 2.50E-08

"C12" 2.30 E-08 "C43" 4.70E-10

"C13" 2.30 E-06 "C44" 5.30E-06

"C14" 2.70 E-07 "C45" 8.90E-08

"C15" 6.20 E-08 "C46" 1.00E-05

"C16" 4.10 E-09 "C47" 3.00E-09

"C17" 6.90 E-09 "C48" 1.30E-07

"C18" 2.80 E-09 "C49" 1.30E-09

"C19" 9.30 E-09 "C50" 6.80E-10

"C20" 1.90 E-08 "C51" 1.20E-08

"C21" 1.60 E-08 "C52" 8.00E-08

"C22" 7.90 E-09 "C53" 8.70E-07

"C23" 2.60 E-08

"C24" 1.00 E-07

"C25" 6.40 E-08

"C26" 2.30 E-08

"C27" 2.40 E-07

"C28" 1.00 E-07

"C29" 9.60 E-09

"C30" 1.60 E-09

"C31" 2.30 E-08 The compounds shown in Table 1 are particularly preferred compounds according to the invention.

Table 2 Inhibition of tankyrases

of representative compounds of the formula I

Compound IC 5 o [M] IC 5 o [M] ICso [M]

No. PARP TNKS1 TNKS2

ELISA ELISA

"C1" 1.20 E-05 5.70 E-10 3.70 E-10

"C2" 1.70 E-06 3.20 E-10 1.90 E-10

"C3" 1.60 E-05 1.70 E-09 1.90 E-09

"C4" 1.90 E-06 8.40 E-10 4.50 E-10

"C5" 3.20 E-06 1.20 E-08 4.40 E-09

"C6" 3.00 E-08 3.10 E-10 2.00 E-10

"C7" 4.30 E-08 6.60 E-10 5.90 E-10

"C8" 2.10 E-05 5.00 E-10 5.30 E-10

"C9" >3,00E-5 1.40 E-08 7.90 E-09

"C10" 2.60 E-05 1.50 E-08 7.90 E-09

"C11" 1.40 E-05 5.20 E-10 4.00 E-10

"C12" 7.10 E-06 2.20 E-09 1.00 E-09

"C13" 1.10 E-05 6.10 E-07 4.20 E-07

"C14" 1.30 E-05 6.30 E-08 4.40 E-08

"C15" 4.20 E-06 9.20 E-09 5.00 E-09

"C16" 9.00 E-06 6.10 E-10 4.10 E-10

"C17" >3,00E-5 4.70 E-10 3.30 E-10

"C18" >3,00E-5 5.80 E-10 3.6 E-10

"C19" 7.80 E-06 1.30 E-09 5.80 E-10

"C20" >3,00E-5 9.20 E-10 7.30 E-10

"C21" >3,00E-5 3.60 E-09 2.10 E-09

"C22" >3,00E-5 2.20 E-09 1.8 E-09

"C23" 9.00 E-07 6.30 E-09 2.10 E-09 7/076484

- 44 -

"C24" 3.80 E-06 2.70 E-08 1.20 E-08

"C25" 1.60 E-06 3.60 E-08 1.20 E-08

"C26" 9.50 E-07 7.70 E-09 3.00 E-09

"C27" 3.30 E-06 2.90 E-08 1.30 E-08

"C28" 4.40 E-07 3.40 E-08 1.20 E-08

"C29" 8.10 E-07 1.80 E-09 8.40 E-10

"C30" 2.00 E-06 6.20 E-10 5.70 E-10

"C31" 5.30 E-06 3.00 E-09 2.20 E-09

"C32" 6.80E-10 5.60E-10

"C33" 1.70E-05 5.10E-10 4.50E-10

"C34" 4.80E-10 5.00E-10

"C35" 2.90E-05 4.20E-09 3.30E-09

"C36" 7.20E-06 7.90E-10 2.10E-09

"C37" 1.50E-06 9.50E-10 2.00E-09

"C38" 5.80E-10 3.20E-10

"C39" 4.30E-10 6.10E-10

"C40" 3.20E-06 1.50E-09 4.70E-09

"C41" 3.50E-06 5.90E-10 8.80E-10

"C42" 1.10E-06 4.50E-10 1.00E-09

"C43" 7.90E-07 2.30E-10 6.30E-10

"C44" 3.10E-07 7.90E-09 8.80E-09

"C45" 3.90E-07 4.50E-10 5.50E-10

"C46" 8.20E-09 2.80E-09

"C47" 1.70E-07 3.50E-10 2.20E-10

"C48" 1.40E-08 5.30E-09

"C49" 3.40E-06 3.00E-10 2.10E-10

"C50" 2.70E-06 4.40E-10 2.40E-10

"C51" 1.00E-06 1.00E-09 7.20E-10

"C52" 1.00E-06 3.30E-10 2.50E-10

"C53" 9.20E-06 1.10E-09 6.90E-10 Explanation: 3.00 E-06 means 3.00 x 10 6

The compounds shown in Table 2 are particularly preferred compounds according to the invention.

Synthesis of intermediates

A1 : 3,3-Dimethyl-4-(4-oxo-3,4-dihydro-quinazolin-2-yl)-butyric acid

2-Aminobenzamide (4.79 g; 35.173 mmol) and 3,3-dimethylglutaric anhydride (5.00 g; 35.173 mmol) were suspended in toluene (80.0 mL) and the mixture was stirred at reflux for 5 h. The mixture was evaporated, the residue dissolved in 2 N sodium hydroxide solution (50.0 mL) and stirred at 90 °C for 3 h. The mixture was neutralized with 2N HCI solution (50 mL). The precipitated solid was filtered by suction, washed with distilled water and dried in vacuum. Yield: 7.63 g colorless solid; LC/MS (A), Rt: 1.53 min; (M+H) 261.1

A2: 3-Methyl-4-(4-oxo-3,4-dihydro-quinazolin-2-yl)-butyric acid

Acid A2 was prepared as described for acid A1. Yield: 609 mg (85%) colorless solid; LC/MS (A), Rt: 1.37 min- (M+H) 247.1

A3: 4-(6-Fluoro-8-methyl-4-oxo-3,4-dihydro-quinazolin-2-yl)-3-me thyl-butyric acid

Acid A3 was prepared as described for acid A1. Yield: 633 mg (55%) colorless solid; LC/MS (A), Rt: 1.81 min; (M+H) 279.1

A4: 4-Methyl-3-(4-oxo-3,4-dihydro-quinazolin-2-ylmethyl)-pentano ic acid

A4.1 : 4-lsopropyl-dihydro-pyran-2,6-dione

3-lsopropyl-pentanedioic acid (12.36 g; 67.408 mmol) was dissolved in acetic anhydride (38.23 ml; 404.446 mmol) and stirred at 130 °C overnight. The reaction mixture was gradually cooled to room temperature, the excess acetic anhydride was evaporated under vacuum and the residue was purified by distillation. Yield: 4.30 g (41%) light yellow liquid; bp: 105-108 °C (0.5 mbar) A4.2: 4-Methyl-3-(4-oxo-3,4-dihydro-quinazolin-2-ylmethyl)-pentano ic acid Preparation as described for acid A.1 using A4.1. Yield: 851 mg (89%) beige solid; LC/MS (A), Rt: 1.61 min; (M+H) 275.1

A5: 5-Methyl-3-(4-oxo-3,4-dihydro-quinazolin-2-ylmethyl)-hexanoi c acid

A5. : 4-lsobutyl-dihydro-pyran-2,6-dione

Preparation and purification as described for A4.1. Yield: 8.10 g (61%) light yellow liquid; bp: 120 °C (0.8 mbar) A5.2: 5-Methyl-3-(4-oxo-3,4-dihydro-quinazolin-2-ylmethyl)-hexanoi c acid Preparation as described for acid A1 using A5.1. Yield: 1.85 g (89%) beige solid; LC/MS (A), Rt: 1.61 min; (M+H) 275.1

A6: [1 -(4-Oxo-3,4-dihydro-quinazolin-2-ylmethyl)-cyclopropyl]-acet ic acid

A6.1 : 6-Oxa-spiro[2.5]octane-5,7-dione

(l-Carboxymethyl-cyclopropyl)-acetic acid (496.0 mg; 3.073 mmol) was dissolved in acetic anhydride (12.0 mL; 126.949 mmol) and stirred at 130 °C over night. The reaction mixture was gradually cooled to room temperature and the excess acetic anhydride was evaporated under vacuum. The light beige residue was co-evaporated two times with toluene, finally dried under vacuum (400 mg (93%) beige solid) and used in the next step without further purification.

A6.2: [1-(4-Oxo-3,4-dihydro-quinazolin-2-ylmethyl)-cyclopropyl]-ac etic acid Preparation as described for acid A1 using A6.1. Yield: 303 mg (82%) beige solid; LC/MS (A), Rt: 1.46 min; (M+H) 259.1

A7: 2,2-Dimethyl-4-(4-oxo-3,4-dihydro-quinazolin-2-yl)-butyric acid

Preparation as described for acid A1 using 3,3-dimethyltetrahydropyran-2,6- dione. Yield: 1.23 g (61%) colorless solid; LC/MS (A), Rt: 1.55 min; (M+H) 261.1 A8: 3 , 3-D imethyl-4-(7-methy 1-4-0X0-4 , 7-d ihyd ro-3 H-py rrolo[2 , 3-d]pyrimid in-2- yl)-butyric acid

A8.1 : 2-Amino-1-methyl-1H-pyrrole-3-carbonitrile

To a stirred solution of malononitrile (14.85 g; 224.79 mmol) in

dichloromethane (300.0 mL) (2,2-dimethoxy-ethyl)-methyl-amine (24.6 mL; 224.79 mmol) was added and the mixture was stirred for 10 min at ambient temperature. Toluene-4-sulfonic acid (77.42 g; 449.58 mmol) was added and reaction mixture was stirred for 48 h at ambient temperature. The reaction mixture was concentrated under vacuum and the residue was rendered basic with saturated sodium bicarbonate solution and extracted with ethyl acetate. The combined organic layers were washed with saturated brine solution, dried over anhydrous sodium sulfate, filtered and concentrated under vacuum. The residue was purified by column chromatography (eluent: ethyl acetate in petroleum ether (35%-40%)). Yield: 5.00 g (18%) pale brown solid; 1 H NMR (400 MHz, DMSO-de) δ 6.17 (s, 1H), 5.96 (s, 1H), 5.74 (s, 2H), 3.34 (s, 3H). A8.2: 4-(3-Cyano-1-methyl-1 H-pyrrol-2-ylcarbamoyl)-3,3-dimethyl-butyric acid To a stirred solution of 2-Amino-1-methyl-1H-pyrrole-3-carbonitrile (265.0 mg; 2.187 mmol) in dry toluene (3.00 mL) 4,4-dimethyl-dihydro-pyran-2,6-dione (466.4 mg; 3.281 mmol) was added and the mixture was heated to 135 °C for 15 h. The reaction mixture was concentrated under reduced pressure and the crude residue (174.0 mg brown gum) was used in the next step without further purification.

A8.3: 3,3-Dimethyl-4-(7-methyl-4-oxo-4,7-dihydro-3H-pyrrolo[2,3-d] pyrimidin- 2-yl)-butyric acid

To a stirred solution of A8.2 in 10% aqueous potassium hydroxide (3.0 mL) hydrogen peroxide (30% solution; 6.0 mL) was added at 0 °C. The mixture was allowed to warm to room temperature and stirred for 30 min. The mixture was then heated to 75 °C for 12 h. The reaction mixture was cooled to 0 °C, acidified (pH 4) using acetic acid and extracted with dichloromethane. The combined organic layers were dried with sodium sulfate, filtered by suction and concentrated under reduced pressure. Yield: 172 mg (99 %) colorless oil; LC/MS (B), Rt: 1.91 min; (M+H) 264.2

A9: 3,3-Dimethyl-4-(1-methyl-4-oxo-4,5-dihydro-1 H-pyrazolo[3,4-d]pyrimidin-6- yl)-butyric acid

5-Amino-1-methyl-1 H-pyrazole-4-carboxylic acid amide (1.49 g; 10.618 mmol and 4,4-dimethyl-dihydro-pyran-2,6-dione (2.26 g; 15.898 mmol) was heated to 170 °C for 6 h. The reaction mixture was cooled to room temperature, 10% aqueous sodium hydroxide solution (56.00 ml_) was added and the mixture was heated to 100 °C for 2h. The reaction mixture was cooled to 0 °C, acidified with acetic acid and extracted with dichloromethane. The combined organic layers were dried with sodium sulfate, filtered by suction and

concentrated. The residue was triturated with petrol ether and ethyl acetate (1/1), the resulting solid was filtered and dried. Yield: 0.80 g (28%) colorless solid; LC/MS (B), Rt: 1.84 min; (M+H) 265.0. 1 H NMR (400 MHz, DMSO-de) δ 13.00 (brs, 1 H), 12.12 (brs, 1 H), 7.92 (s, 1 H), 3.86 (s, 3H), 2.68 (s, 2H), 2.22 (s, 2H), 1.04 (s, 6H).

A10: (6,8-Difluoro-4-oxo-3,4-dihydro-quinazolin-2-ylmethoxy)-acet ic acid

2-Amino-3,5-difluoro-benzamide (1.72 g; 10.00 mmol) and [1,4]-dioxane-2,6- dione (1.16 g; 10.00 mmol) were suspended in toluene (21.0 ml_) and the mixture was stirred at reflux for 3.5 h. The mixture was evaporated to dryness, sodium hydroxide solution (2 N; 17.0 ml; 459.03 mmol) was added and the mixture was stirred at 80 °C for 4 h. The reaction mixture was cooled to room temperature, acidified with acetic acid and concentrated. The aqueous residue was purified by flash chromatography (Companion RF; 510 g Si50 silica gel column). Yield: 1.78 g (66%) pale brown solid; LC/MS, Rt: 1.44; (M+H) 271.0; 1 H NMR (400 MHz, Methanol-d 4 ) δ 7.71 (ddd, J = 8.3, 2.9, 1.6 Hz, 1 H), 7.52 (ddd, J = 10.2, 8.7, 2.8 Hz, 1H), 4.64 (s, 2H), 4.21 (s, 2H).

A11 : 3,3-Dimethyl-4-(8-methyl-4-oxo-3,4-dihydro-quinazolin-2-yl)- buty acid

Acid A11 was prepared as described for acid A1. Yield: 1.29 g (77%) pale- yellow solid; LC/MS (A), Rt: 1.82 min; (M+H) 275.1

A 12 : 3-Methy l-4-(8-methy I-4-OXO-3 ,4-d ihyd ro-q uinazoli n-2-y l)-buty ric acid

Acid A12 was prepared as described for acid A1. Yield: 1.51 g (82%) pale- brown solid; LC/MS (A), Rt: 1.72 min; (M+H) 261.1

A13: 4-(6-Fluoro-8-methyl-4-oxo-3,4-dihydro-quinazolin-2-yl)-3-me thyl- butyric acid

Acid A13 was prepared as described for acid A1. Yield: 0.63 g (55%) colorless solid; LC/MS (A), Rt: 1.81 min; (M+H) 279.1

A14: 3,3-Dimethyl-4-(1-oxo-1 ,2-dihydro-isoquinolin-3-yl)-butyric acid

A14.1 : Toluene-4-sulfonic acid 2,2-dimethyl-pent-4-ynyl ester

To a solution of 2,2-dimethyl-pent-4-yn-1-ol (3.12 g; 27.852 mmol) in dry pyridine (40.0 mL) 4-toluenesulfonylchloride (6.37 g; 33.423 mmol) was added at room temperature and the mixture was stirred overnight. The solvent was removed and the residue was once coevaporated with toluene. The residue was diluted with water and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate, filtered and concentrated in vacuo. The residue was purified by RP-flash chromatography (CombiFlashRF 200). Yield: 5.00 g (67%) beige oil; LC/MS (A), Rt: 2.50 min; (M+H) 267.1

A14.2: Toluene-4-sulfonic acid 2,2-dimethyl-3-(1-oxo-1H-isochromen-3-yl)- propyl ester

To 2-iodobenzoic acid (1.52 g; 6.125 mmol) and potassium carbonate (1.69 g; 12.249 mmol) DMF (16.8 mL) was added under argon atmosphere and the mixture was stirred for 15 min at room temperature. A14.1 (1.63 g; 6.125 mmol) and copper(l) iodide (583.0 mg; 3.062 mmol) were added and the reaction mixture was stirred overnight under argon atmosphere at 65 °C. The reaction mixture was allowed to cool to ambient temperature, filtered over celite and washed with ethyl acetate. The filtrate was concentrated in vacuo and the residue purified by RP-flash

chromatography (CombiFlashRF 200). Yield: 1.71 g (72%) orange oil; LC/MS (A), Rt: 2.59 min; (M+H) 387.1

14.3: 3,3-Dimethyl-4-(1-oxo-1 H-isochromen-3-yl)-butyronitrile

To a solution of A14.2 (1.70 g; 4.397 mmol) in dry DMSO (32 mL) potassium cyanide (429.5 mg; 6.596 mmol) was added and the mixture was stirred at 90 °C overnight. The mixture was cooled to room

temperature, diluted with water (40 mL) and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate, filtered and concentrated in vacuo. The residue was purified by flash chromatography (CombiFlashRF 200). Yield: 229 mg (22%) orange oil; LC/MS (A), Rt: 2.15 min; (M+H) 242.2

14.4: 3,3-Dimethyl-4-(1-oxo-1 ,2-dihydro-isoquinolin-3-yl)-butyronitrile Compound A14.3 (229.0 mg; 0.951 mmol) was dissolved in a solution of ammonia in methanol (7 M; 14.0 mL) and heated in a microwave for 1.5 h at 130 °C. The reaction mixture was evaporated to dryness. DMSO and subsequently acetonitrile and water was added, precipitating the solid was filtered off by suction, washed with water dried under high vacuum. Yield: 133 mg (58%) colorless solid; LC/MS (A), Rt: 1.87 min; (M+H) 241.2

A14.5: 3,3-Dimethyl-4-(1 -oxo-1 ,2-dihydro-isoquinolin-3-yl)-butyric acid To A14.4 (105.0 mg; 0.435 mmol) sodium hydroxide solution (6 N; 2.18 ml_; 13.061 mmol) was added and the suspension was stirred at 100 °C overnight 100 °C. The solution was poured onto water, acidified with HCI solution (0.1 N) and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate, filtered and concentrated in vacuo. Yield: 112 mg (99%) colorless solid; LC/MS (A), Rt: 1.85 min; (M+H) 260.1

A15: 3-Methyl-4-(1-oxo-1,2-dihydro-isoquinolin-3-yl)-butyric acid

A15.1 : Toluene-4-sulfonic acid 1-methyl-but-3-ynyl ester

4-Pentyn-2-ol (4.12 g; 48.932 mmol) was dissolved in pyridine (25.0 mL). 4-Toluenesulfonylchloride ( 0.26 g; 53.826 mmol) was added and the solution was stirred at room temperature overnight. The solvent was removed and the residue was once coevaporated with toluene. The residue was diluted with water and aqueous citric acid solution (10%) and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate, filtered and concentrated in vacuo. The residue was purified by flash chromatography (CombiFlashRF 200). Yield: 10.52 g (90%) colorless liquid; LC/MS (A), Rt: 2.30 min; (M+H) 239.1 A15.2: 2-(1-Methyl-but-3-ynyl)-malonic acid diethyl ester

Diethyl malonate (1.49 ml; 9.748 mmol) dissolved in DMF (11.3 mL) was added dropwise to a suspension of sodium hydride (60% in paraffin oil; 501.8 mg; 12.547 mmol) in DMF (22.5 mL). The reaction mixture was stirred 1 h under nitrogen atmosphere. Compound A15.1 (2.30 g; 9.652 mmol) dissolved in DMF (4.5 mL) was added and the mixture was stirred at 100 °C overnight. The reaction was cooled to room temperature, diluted with water and extracted with ethyl acetate. The combined organic layers were washed with brine, dried over sodium sulfate, filtered and

concentrated in vacuo. The residue was purified by flash chromatography (CombiFlashRF 200). Yield: 1.06 g (49%) colorless oil

A15.3: 2-[1-Methyl-2-( 1-OXO-1 H-isochromen-3-yl)-ethyl]-malonic acid diethyl ester

2-lodobenzoic acid (706.0 mg; 2.845 mmol) was dissolved in DMF (11.4 mL) and potassium carbonate (786.4 mg; 5.690 mmol) was added. The mixture was stirred for 15 min at room temperature under argon

atmosphere. Copper iodide (270.8 mg; 1.422 mmol) was added and the reaction mixture was stirred at 65 °C overnight. The reaction mixture was allowed to cool to ambient temperature, filtered over celite, washed with DMF and the filtrate was concentrated in vacuo. The residue was diluted with ethyl acetate and washed with water. A solid precipitated between the phases, which was removed by filtration. The organic phase was washed with brine, dried over sodium sulfate, filtered and evaporated to dryness. The residue was combined with the isolated precipitate and purified by flash chromatography (CombiFlashRF 200). Yield: 391 mg (40%) colorless oil; LC/MS (A), Rt: 2.45-2.49 min; (M+H) 347.1

A15.4: 3-Methyl-4-(1-oxo-1 ,2-dihydro-isoquinolin-3-yl)-butyric acid ethyl ester

Compound A15.3 (347.0 mg; 0.529 mmol) was dissolved in a solution of ammonia in methanol (7M; 8 mL) and stirred for 1 h in a Mikrowave (CEM) at 120 °C. The mixture was evaporated to dryness. The residue was purified by flash chromatography (CombiFlashRF 200). The isolated product (137 mg) was a mixture of the methyl and ethyl ester of the target compound, which was used in the next step without separation.

A15.5: 3-Methyl-4-(1-oxo-1 ,2-dihydro-isoquinolin-3-yl)-butyric acid

Saponification of compound A15.4 with sodium hydroxide solution (2 N) in 1,4-dioxane 80 °C for 2 h followed by usual work-up yielded the title compound. Yield: 119 mg (100%) colorless solid; LC/MS (A), Rt: 1.69 min; (M+H) 246.1

B1 : (1-Methyl-1 H-pyrazol-4-yl)-piperidin-4-yl-methanone hydrochloride 3

B1.1 : 4-lodo-1-methyl-1 H-pyrazole (1.12 g; 5.385 mmol) and 4-(Methoxy- methyl-carbamoyl)-piperidine-1-carboxylic acid tert-butyl ester (1.47 g; 5.385 mmol) were dissolved in dry THF (15 mL) under argon. While stirring the clear light yellow solution was cooled down to -60 °C and butyllithium (15% solution in n-hexane) (3.72 mL; 5.923 mmol) was added dropwise at this temperature over a period of 10 min. The reaction mixture was stirred for 30 min between - 60 and -45 °C, then slowly warmed to room temperature and stirred over night. The reaction mixture was cooled to 0 °C, quenched with 10% citric acid solution, diluted with ethyl acetate (70 mL) and washed with water and brine, dried with Na2S04, filtered and evaporated to dryness. The oily residue was purified by flash chromatography (Companion RF; 120 g Si50 silica gel column). Yield: 999 mg (63%) light green oil;

1 H NMR (500 MHz, DMSO-de) δ 8.42 (s, 1H), 7.94 (d, J = 0.7 Hz, 1 H), 3.97 (d, J = 12.6 Hz, 2H), 3.87 (s, 3H), 3.15 (tt, J = 11.4, 3.6 Hz, 1H), 2.93-2.75 (m, 2H), 1.76-1.67 (m, 2H), 1.33-1.46 (m, 11 H). LC/MS (A), Rt: 1.93 min; (M+H; BOC-cleaved mass) 238.1

B1.2: (1-Methyl-1H-pyrazol-4-yl)-piperidin-4-yl-methanone hydrochloride Boc-cleavage from B1.1 afforded the title compound. Colorless solid; LC/MS (A), Rt: 0.34/0.47 min; (M+H) 194.2

B2: (1 -Ethyl- 1 H-pyrazol-4-yl)-piperidin-4-yl-methanone hydrochloride

Preparation as described for B1 ; off-white solid; LC/MS (B), Rt: 1.26 min;

(M+H) 208.2;

1 H NMR (400 MHz, DMSO-de) δ 9.27 (brs, 1H), 8.75 (brs, 1H), 8.49 (s, H), 7.99 (s, 1 H), 4.20-4.11 (m, 2H), 3.31-3.22 (m, 3H), 2.99-2.88 (m, 2H), 1.93- 1.68 (m, 4H), 1.42-1.31 (m, 3H).

B3: 1-lsopropyl-1 H-pyrazol-4-yl)-piperidin-4-yl-methanone hydrochloride

Preparation as described for B1 ; off-white solid; LC/MS (C), Rt: 1.54 min; (M+H) 222.2

B4: (6-Methoxy-pyridin-3-yl)-piperidin-4-yl-methanone hydrochloride

B4.1 : 4-(6-Methoxy-pyridine-3-carbonyl)-piperidine-1-carboxylic acid tert-butyl ester

To a solution of 5-Bromo-2-methoxy-pyridine (6.60 g; 34.40 mmol) in THF (132 mL) under nitrogen atmosphere, n-butyl lithium (1.6 M in hexanes) (25.80 mL; 41.28 mmol) was added drop wise at -78 °C and stirred for 1 h at the same temperature. A solution of 4-(Methoxy-methyl-carbamoyl)-piperidine-1- carboxylic acid tert-butyl ester (10.52 g; 37.84 mmol) in THF (25 mL) was added dropwise at -78 °C and stirred for 4 h at -78 °C. The reaction mixture was then slowly allowed to attain room temperature and stirred for 12 h. The reaction mixture was quenched by saturated NhUCI solution (250 mL) and extracted with ethyl acetate (2 x 300 mL). The combined organic layers were washed with water (200 mL) and brine (200 mL), dried over anhydrous sodium sulfate and concentrated. The crude material was purified by column

chromatography using silica gel (60-120) and petrol ether/ethyl acetate as gradient elution to afford 4-(6-Methoxy-pyridine-3-carbonyl)-piperidine-1- carboxylic acid tert-butyl ester (5.00 g; 44.5 %) as a pale yellow oil.

1H NMR (400 MHz, CDC ) δ 8.80 (d, J = 2.3 Hz, 1H), 8.14 (dd, J = 2.4, 8.7 Hz, 1H), 6.82 (d, J = 8.8 Hz, 1H), 4.20-4.17 (m, 2H), 4.02 (s, 3H), 3.35-3.27

(m, 1H), 2.92-2.86 (m, 2H), 1.85-1.82 (m, 2H), 1.76-1.66 (m, 2H), 1 ,47 (s, 9H). LC/MS (B), Rt: 4.64 min; (M+H; BOC-cleaved mass) 265.0

B4.2: (6-Methoxy-pyridin-3-yl)-piperidin-4-yl-methanone hydrochloride

Boc-cleavage from B4.1 afforded the title compound. Colorless solid; LC/MS (B), Rt: 1.84 min; (M+H) 221.0;

H NMR (400 MHz, DMSO-de) δ 9.21 (s, 1 H), 8.91 (d, J = 1.08 Hz, 2H), 8.23- 8.20 (m, 1 H), 6.95 (d, J = 8.76 Hz, 1H), 6.55 (bs, 3H), 6.09 (bs, 2H), 3.94 (s, 3H), 3.78-3.67 (m, 1H), 3.29-3.26 (m, 2H), 3.04-2.95 (m, 2H), 1.93-1.90 (m, 2H), 1.82-1.71 (m, 2H).

B5: (6-Methoxy-5-methyl-pyridin-3-yl)-piperidin-4-yl-methanone hydrochloride

Preparation as described for B4; pale-brown solid; LC/MS (C), Rt: 1.89 min; (M+H) 235.0

B6: Methyl 4-(piperidine-4-carbonyl)benzoate hydrochloride

B6.1 : tert-Butyl 4-(p-tolylsulfonylhydrazono)piperidine-1 -carboxylate

To 4-Methylbenzenesulfonohydrazide (9.30 g; 49.937 mmol) and 4-Oxo-

10 piperidine-1-carboxylic acid tert-butyl ester (9.95 g; 49.937 mmol) dry

methanol (45.0 mL) was added under argon atmosphere and the mixture was stirred at room temperature for 45 min. The reaction mixture was evaporated to dryness. The solid residue was triturated with MTB-ether

^ and dried under vacuum at 40 °C overnight. Yield: 17.05 g (93%) colorless solid; LC/MS (A), Rt: 2.24 min; (M+H) 368.2

B6.2: 4-(4-Bromo-benzoyl)-piperidine-1-carboxylic acid tert-butyl ester Compound B6.1 (1.00 g; 2.721 mmol), 4-bromobenzaldehyde (0.604 g; 3.266 mmol) and cesium carbonate (0.436 ml; 5.443 mmol) were

20

suspended in dry ,4-dioxane (10.0 mL) under argon atmosphere. The mixture was heated to 110 °C and stirred overnight at this temperature. The reaction mixture was cooled to room temperature, quenched with water (30 mL) and extracted with MTB-ether. The combined organic layers 25 were washed with 5% citric acid solution, saturated NaHC03 solution and brine, dried with sodium sulfate, filtered by suction and evaporated to dryness. The solid residue was triturated with petrolether/MTB-ether (1 :1), filtered by suction, washed with petrolether/MTB-ether (3:1) and dried. From the filtrate further product was isolated by flash chromatography (Companion RF; 40 g Si50 silica gel column). Yield: 657 mg (66%) colorless solid; LC/MS (A), Rt: 2.72 min; (M+H-t-Bu) 312.0/314.0

B6.3: 4-(4-Methoxycarbonyl-benzoyl)-piperidine-1-carboxylic acid tert-butyl ester

^ In an autoclave, a solution of compound B6.2 (0.60 g; 1.629 mmol) and

230.8 mg (2.281 mmol) triethylamine in methanol (9 mL) and THF (20 mL) was flushed with nitrogen. (1 ,1'-Bis(diphenylphosphino)- ferrocen)dichloropalladium(ll), dichioromethane (13.0 mg; 0.016 mmol) was added. Then the autoclave was filled with carbon monoxide and the mixture stirred at 70 °C and a pressure of (max.) 3.7 bar for 17.5 h. The autoclave was brought to atmospheric pressure. The reaction mixture was evaporated to dryness. The crude residue was purified by flash

chromatography (Companion RF; 80 g Si50 silica gel column). Yield: 0.512 g (90%) light yellow solid; LC/MS (A), Rt: 2.50 min; (M+H-t-Bu) 274.2 B6.4: 4-(Piperidine-4-carbonyl)-benzoic acid methyl ester hydrochloride Compound B6.3 (0.512 g; 1.474 mmol) was dissolved in dry 1 ,4-dioxane (2.6 mL) and a solution of HCI in dioxane (4N; 1.105 mL; 4.421 mmol) was added. The colorless solution was stirred at room temperature for 1 h. A white suspension was formed. Further HCI solution (4N; 1.105 mL; 4.421 mmol) was added and the suspension was stirred overnight at room temperature. The reaction mixture was evaporated to dryness. The solid residue was suspended in MTB-ether, filtered by suction, washed with little MTB-ether and dried under vacuum at 50 °C for 2 h. Yield: 0.403 g (96%) colorless solid; LC/MS (A), Rt: 1.22 min; (M+H) 248.2

B7: Methyl 3-(piperidine-4-carbonyl)benzoate hydrochloride

Preparation as described for B6; pale-brown solid; LC/MS (C), Rt: 1.24 min; (M+H) 248.2

B8: (4-Dimethylaminomethyl-phenyl)-piperidin-4-yl-methanone

hydrochloride

B8.1 : 4-(4-Dimethylaminomethyl-benzoyl)-piperidine-1-carboxylic acid tert- butyl ester

Preparation from compound B6.1 (500.0 mg; 1.361 mmol) and 4- dimethylaminomethylbenzaldehyde (266.5 mg; 1.633 mmol) as described for compound B6.2. Yield: 269 mg (57%) yellow oil; LC/MS (A), Rt: 1.58 min; (M+H) 347.3

B8.2: (4-Dimethylaminomethyl-phenyl)-piperidin-4-yl-methanone

hydrochloride

Deprotection of B8.1 was performed as described for compound B6.4.

Yield: 220.5 mg (100%) orange solid; LC/MS (A), Rt: 0.38 min; (M+H) 247.3

Example 1 : 2-{2,2-Dimethyl-4-[4-(1-methyl-1 H-pyrazole-4-carbonyl)-piperidin- 1-yl]-4-oxo-butyl}-3H-quinazolin-4-one ("C1")

A1 (350.0 mg; 1.303 mmol), B1 (299.3 mg; 1.303 mmol) and

[dimethylamino(triazolo[4,5-b]pyridin-3-yloxy)methylene]- dimethyl-ammonium hexafluorophosphate (743.1 mg; 1.954 mmol) were placed in a vial and suspended in DMF (8.0 mL). Ethyl-diisopropyl-amine (886.3 μΙ; 5.212 mmol) was added and the mixture was stirred for 30 min at room temperature. The mixture was diluted with water (50 ml_) and extracted with ethyl acetate. The combined organic layers were washed with brine, dried with sodium sulfate, filtered by suction and evaporated to dryness. The red-brown, oily residue was purified by chromatography (Companion RF; 80 g SI50 silica gel column). The red-brown oil was freeze-dried. Yield: 338 mg (60%) pale brown solid; LC/MS (A), Rt: 1.73 min; (M+H) 436.2; 1 H NMR (400 MHz, DMSO-de) δ 12.49 (s, 1 H), 8.44 (s, 1H), 8.09 (dd, J = 7.9, 1.5 Hz, 1H), 7.97 (s, 1H), 7.80-7.74 (m, 1H), 7.61 (d, J = 8.1 Hz, 1H), 7.49-7.44 (m, 1H), 4.58-4.50 (m, 1H), 4.17-4.07 (m, 1H), 3.88 (s, 3H), 3.32-3.13 (m, 2H), 2.82-2.64 (m, 3H), 2.50-2.46 (m, 2H), 1.87-1.74 (m, 2H), 1.58-1.33 (m, 2H), 1.11-1.04 (m, 6H).

Example 2: 2-{(S)-2-Methyl-4-[4-(1 -methyl- 1 H-pyrazole-4-carbonyl)-piperidin- 1-yl]-4-oxo-butyl}-3H-quinazolin-4-one ("C2")

Example 3: 2-{(R)-2-Methyl-4-[4-(1-methyM H-pyrazole-4-carbonyl)-piperidin- 1-yl]-4-oxo-butyl}-3H-quinazolin-4-one ("C3")

A2 (277.4 mg; 1.126 mmol) and B1 (258.8 mg; 1.126 mmol) were coupled described for example 1. Yield: 237 mg (50%) orange oil

The preparative separation of the enantiomers was performed by SFC (column: ChiralCel OJ-H; eluent: CO2:methanol (containing 0.5%

diethylamine) - 88:12). The combined fractions were evaporated to dryness and freeze-dried.

Example 2: 76 mg beige solid; LC/MS (A), Rt: 1.55 min; (M+H) 422.2; 1 H NMR 5 (500 MHz, DMSO-de) δ 12.01 (s, 1H), 8.43 (s, 1H), 8.10-8.05 (m, 1H), 7.95 (s,

1 H), 7.80-7.72 (m, 1H), 7.59 (d, J = 8.1 Hz, 1 H), 7.45 (t, J = 7.5 Hz, 1H), 4.43- 4.30 (m, 1 H), 4.03-3.91 (m, 1 H), 3.88 (s, 3H), 3.22 (tt, J = 11.3, 3.6 Hz, 1H), 3.17-3.07 (m, 1H), 2.68-2.57 (m, 2H), 2.57-2.42 (m, 3H, overlapped with 10 DMSO-de), 2.28-2.17 (m, 1H), 1.83-1.69 (m, 2H), 1.57-1.43 (m, 1H), 1.39-1.21

(m, 1H), 0.95 (d, J = 5.9 Hz, 3H).

Example 3: 98 mg beige solid; LC/MS (A), Rt: 1.55 min; (M+H) 422.2; 1 H NMR (500 MHz, DMSO-de) δ 11.81 (s, 1 H), 8.43 (s, 1H), 8.11-8.04 (m, 1H), 7.95 (s,

-15 1 H), 7.80-7.73 (m, 1H), 7.59 (d, J = 8.1 Hz, 1 H), 7.45 (t, J = 7.5 Hz, 1H), 4.43- 4.28 (m, 1 H), 4.06-3.92 (m, 1 H), 3.88 (s, 3H), 3.22 (tt, J = 11.3, 3.6 Hz, 2H), 3.16-3.06 (m, 1 H), 2.70-2.57 (m, 2H), 2.57-2.41 (m, 3H, overlapped with DMSO-de), 2.30-2.16 (m, 1 H), 1.82-1.66 (m, 2H), 1.58-1.40 (m, 1H), 1.40-1.19 o (m, 1 H), 0.95 (d, J = 5.9 Hz, 3H).

Example 4: 2-{(S)-4-[4-(1-lsopropyl-1 H-pyrazole-4-carbonyl)-piperidin-1-yl]-2- methyl-4-oxo-butyl}-3H-quinazolin-4-one ("C4")

Example 5: 2-{(R)-4-[4-(1-lsopropyl-1 H-pyrazole-4-carbonyl)-piperidin methyl-4-oxo-butyl}-3H-quinazolin-4-one ("C5") 5

A2 (204.1 mg; 0.829 mmol) and B3 (213.6 mg; 0.829 mmol) were coupled as described for example 1. Yield: 200 mg (54%) colorless oil

The preparative separation of the enantiomers was performed by SFC

(column: ChiralPak AD-H; eluent: C02:ethanol (containing 0.5% diethylamine) - 60:40). The combined fractions were evaporated to dryness and freeze- dried.

Example 4: 92 mg beige solid; LC/MS (A), Rt: 1.73 min; (M+H) 450.3; H NMR (500 MHz, DMSO-de) δ 12.12 (brs, 1 H), 8.49 (s, 1H), 8.10-8.04 (m, 1 H), 7.97 (s, 1H), 7.76 (t, J = 7.5 Hz, 1H), 7.59 (d, J = 8.1 Hz, 1 H), 7.45 (t, J = 7.5 Hz, 1 H), 4.54 (hept, J = 6.7 Hz, 1 H), 4.42-4.32 (m, 1 H), 4.05-3.94 (m, 1H), 3.29- 3.20 (m, 1 H), 3.18-3.08 (m, 1 H), 2.70-2.58 (m, 2H), 2.58-2.43 (m, 3H), 2.29- 2.19 (m, 1H), 1.82-1.69 (m, 2H), 1.58-1.46 (m, 1H), 1.44 (d, J = 6.7 Hz, 6H), 1.38-1.23 (m, 1H), 0.95 (d, J = 5.9 Hz, 3H).

Example 5: 97 mg beige solid; LC/MS (A), Rt: 1.74 min; (M+H) 450.2; 1 H NMR (500 MHz, DMSO-de) δ 12.06 (brs, 1 H), 8.49 (s, 1H), 8.11-8.04 (m, 1 H), 7.97 (s, 1 H), 7.76 (t, J = 7.6 Hz, 1 H), 7.59 (d, J = 8.1 Hz, 1 H), 7.45 (t, J = 7.4 Hz, 1H), 4.54 (hept, J = 6.7 Hz, 1H), 4.42-4.32 (m, 1H), 4.04-3.95 (m, 1H), 3.28- 3.21 (m, 1 H), 3.18-3.07 (m, 1H), 2.73-2.58 (m, 2H), 2.58-2.41 (m, 3H), 2.29- 2.18 (m, 1 H), 1.82-1.69 (m, 2H), 1.57-1.46 (m, 1 H), 1.44 (d, J = 6.7 Hz, 6H), 1.38-1.22 (m, H), 0.95 (d, J = 5.9 Hz, 3H).

Example 6: 6-Fluoro-8-methyl-2-{(S)-2-methyl-4-[4-(1-methyl-1 H-pyrazole-4- carbonyl)-piperidin-1-yl]-4-oxo-butyl}-3H-quinazolin-4-one ("C6")

A3 (172.7 mg; 0.621 mmol) and B1 (142.6 mg; 0.621 mmol) were coupled as described for example 1. Yield: 187 mg (66%) yellow oil

The preparative separation of the enantiomers was performed by HPLC (column: ChiralPak AD-H; eluent: n-heptane:ethanol - 30:70). The combined fractions were evaporated to dryness and freeze-dried.

Example 6: 84 mg colorless solid; LC/MS (A), Rt: 1.90 min; (M+H) 454.2; 1 H NMR (500 MHz, DMSO-de) δ 12.26 (s, 1 H), 8.42 (s, 1H), 7.95 (s, 1H), 7.59- 7.52 (m, 2H), 4.41-4.30 (m, 1 H), 3.98-3.89 (m, 1 H), 3.87 (s, 3H), 3.21 (tt, J = 11.3, 3.4 Hz, 1 H), 3.11 (t, J = 12.6 Hz, 1 H), 2.69-2.44 (m, 8H), 2.33-2.21 (m, 1 H), 1.79-1.69 (m, 2H), 1.55-1.42 (m, 1 H), 1.38-1.24 (m, 1 H), 0.97 (d, J = 5.8 Hz, 3H).

Example 7: 90 mg colorless solid; LC/MS (A), Rt: 1.90 min; (M+H) 454.2; 1 H NMR (500 MHz, DMSO-de) δ 12.26 (s, 1H), 8.42 (s, 1H), 7.94 (s, 1H), 7.59- 7.51 (m, 2H), 4.41-4.30 (m, 1H), 3.99-3.89 (m, 1 H), 3.87 (s, 3H), 3.21 (tt, =

11.3, 3.5 Hz, 1 H), 3.11 (t, J = 12.6 Hz, 1 H), 2.69-2.44 (m, 8H), 2.33-2.20 (m, 1 H), 1.81-1.68 (m, 2H), 1.55-1.41 (m, 1 H), 1.38-1.21 (m, 1 H), 0.97 (d, J

Hz, 3H).

Example 8: 2-((S)-3-Methyl-2-{2-[4-(1-methyl-1 H-pyrazole-4-carbonyl)- piperidin-1-yl]-2-oxo-ethyl}-butyl)-3H-quinazolin-4-one ("C8")

Example 9: 2-((R)-3-Methyl-2-{2-[4-(1-methyl-1 H-pyrazoIe-4-carbonyl)- piperidin-1-yl]-2-oxo-ethyl}-butyl)-3H-quinazolin-4-one ("C9")

A4 (202.0 mg; 0.737 mmol) and B1 (169.2 mg; 0.737 mmol) were coupled as described for example 1. Yield: 258 mg (78%) yellow oil

The preparative separation of the enantiomers was performed by HPLC (column: ChiralPak AD-H; eluent: n-heptane:2-propanol - 50:50). The combined fractions were evaporated to dryness and freeze-dried.

Example 8: 94 mg beige solid; LC/MS (A), Rt: 1.76 min; (M+H) 450.2; H NMR (400 MHz, DMSO-de, 90 °C) δ 11.84 (brs, 1H), 8.30 (s, 1 H), 8.06 (dd, J = 7.9, 1.3 Hz, 1 H), 7.87 (s, 1 H), 7.75-7.68 (m, 1 H), 7.55 (d, J = 8.1 Hz, 1 H), 7.41 (t, J = 7.5 Hz, 1 H), 4.04 (brs, 2H), 3.86 (s, 3H), 3.22-3.08 (m, 1H), 3.06-2.90 (m, 2H, overlapped with HDO), 2.72-2.60 (m, 1H), 2.58-2.45 (m, 2H), 2.45-2.26 (m, 2H), 1.88-1.62 (m, 3H), 1.58-1.23 (m, 2H), 0.98-0.84 (m, 6H). Example 9: 86 mg beige solid; LC/MS (A), Rt: 1.76 min; (M+H) 450.2; 1 H NMR (400 MHz, DMSO-de, 90 °C) δ 11.84 (brs, 1H), 8.30 (s, 1H), 8.06 (dd, J = 7.9, 1.2 Hz, 1H), 7.87 (s, 1 H), 7.75-7.67 (m, 1 H), 7.55 (d, J = 8.1 Hz, 1 H), 7.44- 7.36 (m, 1H), 4.04 (brs, 2H), 3.86 (s, 3H), 3.14 (tt, J = 10.9, 3.9 Hz, 1H), 3.07- 2.92 (m, 2H, overlapped with HDO), 2.71-2.61 (m, 1H), 2.54-2.45 (m, 2H), 2.44-2.29 (m, 2H), 1.89-1.64 (m, 3H), 1.61-1.21 (m, 2H), 0.97-0.85 (m, 6H).

Example 10: 2-((S)-4-Methyl-2-{2-[4-(1-methyl-1 H-pyrazole-4-carbonyl)- piperidin-1-yl]-2-oxo-ethyl}-pentyl)-3H-quinazolin-4-one ("C10")

Example 1 : 2-((R)-4-Methyl-2-{2-[4-(1-methyl-1 H-pyrazole-4-carbonyl)- piperidin-1-yl]-2-oxo-ethyl}-pentyl)-3H-quinazolin-4-one ("C11")

A5 (208.2 mg; 0.722 mmol) and B1 (165.9 mg; 0.722 mmol) were coupled as described for example 1. Yield: 269 mg (80%) yellow oil

The preparative separation of the enantiomers was performed by SFC (column: ChiralPak AS-H; eluent: C02:2-propanol (containing 0.5% diethylamine) - 80:20). The combined fractions were evaporated to dryness and freeze-dried.

Example 10: 80 mg colorless solid; LC/MS (A), Rt: 1.90 min; (M+H) 464.3; 1 H NMR (500 MHz, DMSO-de) δ 12.17 (s, 1H), 8.42 (s, 1 H), 8.07 (d, J = 7.8 Hz, 1H), 7.95 (s, 1 H), 7.81-7.70 (m, 1 H), 7.63-7.52 (m, 1H), 7.51-7.39 (m, 1H), 4.35-4.23 (m, 1 H), 4.04-3.94 (m, 1H), 3.88 (s, 3H), 3.19 (tt, J = 11.4, 3.6 Hz, 1H), 3.15-3.05 (m, 1 H), 2.71-2.39 (m, 5H), 2.34-2.19 (m, 1 H), 1.82-1.60 (m, 3H), 1.60-1.45 (m, 1H), 1.35-1.14 (m, 3H), 0.91-0.79 (m, 6H).

Example 11 : 85.5 mg colorless solid; LC/MS (A), Rt: 1.90 min; (M+H) 464.3; H NMR (500 MHz, DMSO-de) δ 12.13 (brs, 1 H), 8.42 (s, 1H), 8.07 (d, J = 7.8 Hz, 1 H), 7.95 (s, 1H), 7.79-7.71 (m, 1H), 7.60-7.54 (m, 1 H), 7.487.40 (m, 1H), 4.35-4.24 (m, 1H), 4.03-3.94 (m, 1H), 3.88 (s, 3H), 3.19 (tt, J = 11.4, 3.6 Hz, 1 H), 3.15-3.04 (m, 1H), 2.71-2.39 (m, 5H), 2.33-2.19 (m, 1H), 1.82-1.60 (m, 3H), 1.60-1.44 (m, 1 H), 1.37-1.14 (m, 3H), 0.91- 0.79 (m, 6H).

Example 12: 2-(1-{2-[4-(1-Methyl-1 H-pyrazole-4-carbonyl)-piperidin-1-yl]-2- oxo-ethyl}-cyclopropylmethyl)-3H-quinazolin-4-one ("C12")

A6 (180.0 mg; 0.697 mmol) and B1 (160.0 mg; 0.697 mmol) were coupled as described for example 1. Yield: 225 mg (75%) orange solid; LC/MS (A), Rt: 1.64 min; (M+H) 434.3; H NMR (400 MHz, DMSO-de) δ 12.14 (s, 1 H), 8.43 (s, 1 H), 8.08 (dd, J = 7.9, 1.2 Hz, 1 H), 7.96 (s, 1 H), 7.76 (ddd, J = 8.5, 7.2, 1.6 Hz, 1 H), 7.60 (d, J = 7.8 Hz, 1 H), 7.52-7.38 (m, 1 H), 4.41 (d, J = 12.9 Hz, 1 H), 3.99-3.80 (m, 4H), 3.28-3.15 (m, 1H), 3.08 (t, J = 12.2 Hz, 1 H), 2.74-2.57 (m, 3H), 2.57-2.43 (m, 2H), 1.74 (t, J = 14.8 Hz, 2H), 1.60-1.44 (m, 1H), 1.43-1.28 (m, 1 H), 0.61 (d, J = 5.5 Hz, 2H), 0.45 (d, J = 5.4 Hz, 2H).

Example 13: 2-{3,3-Dimethyl-4-[4-(1-methyl-1H-pyrazole-4-carbonyl)-piper idin- 1-yl]-4-oxo-butyl}-3H-quinazolin-4-one ("C13")

A7 (71.4 mg; 0.275 mmol) and B1 (69.4 mg; 0.302 mmol) were coupled as described for example 1. Yield: 36.5 mg (31%) colorless solid; LC/MS (A), Rt: 1.68 min; (M+H) 436.2; 1 H NMR (400 MHz, DMSO-de) δ 12.15 (s, 1 H), 8.43 (s, 1 H), 8.07 (dd, J = 7.9, 1.3 Hz, 1 H), 7.95 (s, 1 H), 7.76 (td, J = 7.8, 7.2, 1.6 Hz, 1 H), 7.59 (d, J = 7.8 Hz, 1 H), 7.50-7.31 (m, 1H), 4.45-4.20 (m, 2H), 3.87 (s, 3H), 3.29-3.17 (m, 1H), 3.02-2.85 (m, 2H), 2.63-2.54 (m, 2H), 2.11-2.01 (m, 2H), 1.87-1.72 (m, 2H), 1.57-1.41 (m, 2H), 1.24 (s, 6H).

Example 14: 2-{1 ,1-Dimethyl-4-[4-(1 -methyl- 1 H-pyrazole-4-carbonyl)-piperidin- 1-yl]-4-oxo-butyl}-3H-quinazolin-4-one ("C14")

14.1: 2,2-Dimethyl-5-[4-(1-methyl-1 H-pyrazole-4-carbonyl)-piperidin-1-yl]-5- oxo-pentanoic acid

B1 (294.0 mg; 1.280 mmol) and potassium carbonate (353.8 mg; 2.560 mmol) were suspended in toluene (5.0 ml_) and stirred for 15 min at room

temperature. 3,3-dimethyltetrahydropyran-2,6-dione (181.9 mg; 1.280 mmol) was added, the mixture was heated to 110 °C and stirred for 1.5 h. The heating was removed, water (2.0 mL) was added and the mixture was stirred for 30 min at 100 °C. The mixture was cooled to room temperature and the aqueous layers was acidified with 2 N HCI solution. The mixture was diluted with water (10 mL) and extracted with ethyl acetate. The combined organic layers were washed with brine, dried with sodium sulfate, filtered by suction and evaporated to dryness. Yield: 342 mg (80%) light yellow solid; LC/MS (A), Rt: 1.48 min; (M+H) 336.2

14.2: 2,2-Dimethyl-5-[4-(1-methyl-1H-pyrazole-4-carbonyl)-piperidi n-1-yl]-5- oxo-pentanoyl chloride

Compound 14.1 (342.0 mg; 1.020 mmol) was suspended in acetonitrile (2.0 mL). Thionyl chloride (147.9 μΙ; 2.039 mmol) was added dropwise under argon and the mixture was stirred for 1 h at room temperature. The light brown solution was diluted with toluene (5.0 mL) and then evaporated to dryness. The oily residue was used in the next step without further purification.

14.3: 2-{2,2-Dimethyl-5-[4-(1-methyl-1 H-pyrazole-4-carbonyl)-piperidin-1-yl]-5- oxo-pentanoylamino}-benzamide

2-Aminobenzamide (123.1 mg; 0.904 mmol) was suspended in

dichloromethane (4.0 mL) and cooled to 0 °C. Triethylamine (0.5 mL; 3.618 mmol) was added under argon followed by the dropwise addition of compound 14.2 (360.8 mg; 0.904 mmol), dissolved in dichloromethane (4.0 mL). The clear, light yellow solution was stirred at 0 °C for 30 min, warmed to room temperature and stirred for another 30 min. The mixture was heated to 50 °C and stirred overnight. The reaction mixture was cooled to room temperature, diluted with dichloromethane (20 mL), washed once with 5% citric acid solution and once with saturated NaHC03 solution, dried with sodium sulfate, filtered by suction and evaporated to dryness. The oily residue was purified by flash chromatography (Companion RF; 24 g Si50 silica gel column). Yield: 63.5 mg (15%) light green foam; LC/MS (A), Rt: 1.64 min; (M+H) 454.3

14.4: 2-{1 ,1-Dimethyl-4-[4-(1-methyl-1 H-pyrazole-4-carbonyl)-piperidin-1-yl]-4- oxo-butyl}-3H-quinazolin-4-one

A mixture of compound 14.3 (60.6 mg; 0.134 mmol) and sodium hydroxide solution (2 N, 0.33 mL; 0.668 mmol) was heated to 100 °C and stirred for 45 min. The reaction mixture was diluted with water (5 mL) and extracted with ethyl acetate. The combined organic layers were washed with brine, dried with sodium sulfate, filtered by suction and evaporated to dryness. The oily residue was purified by flash chromatography (Companion RF; 12 g Si50 silica gel column) and prep. HPLC (Agilent1260 HPLC; column: Waters SunFire C18 5pm 30x150mm). The combined fractions were evaporated to an aqueous residue, rendered basic with saturated NaHC03 solution and extracted with ethyl acetate. The combined organic layers were washed with brine, dried with sodium sulfate, filtered by suction and evaporated to dryness and finally freeze-dried. Yield: 9.5 mg (16%) colorless solid; LC/MS (A), Rt: 1.73 min; (M+H) 436.3

H NMR (400 MHz, DMSO-de) δ 11.83 (s, 1 H), 8.41 (s, 1H), 8.09 (dd, J = 7.9, 1.3 Hz, 1H), 7.94 (s, 1H), 7.81-7.73 (m, 1H), 7.60 (d, J = 7.8 Hz, 1H), 7.51- 7.43 (m, 1H), 4.35 (d, J = 12.7 Hz, 1H), 3.94-3.78 (m, 4H), 3.19 (tt, J = 11.4, 3.6 Hz, 1 H), 3.14-3.00 (m, 1H), 2.69-2.56 (m, 1H), 2.33-2.10 (m, 2H), 1.99 (t, J = 8.2 Hz, 2H), 1.81-1.67 (m, 2H), 1.57-1.42 (m, 1 H), 1.38-1.21 (m, 7H).

Example 15: 2-{4-[4-(4-Methoxy-benzoyl)-piperidin-1-yl]-1 ,1-dimethyl-4- butyl}-3H-quinazolin-4-one ("C15")

Preparation as described for Example 14 (steps 14.1-14.4). Yield: 18 mg (39%) colorless solid; LC/MS (A), Rt: 2.15 min; (M+H) 462.2

1H NMR (400 MHz, DMSO-de) δ 11.83 (s, 1 H), 8.08 (dd, J = 7.9, 1.3 Hz, 1 H), 8.00-7.93 (m, 2H), 7.80-7.73 (m, 1 H), 7.60 (d, J = 7.9 Hz, 1 H), 7.50-7.43 (m, 1H), 7.08-7.01 (m, 2H), 4.35 (d, J = 13.6 Hz, 1 H), 3.93-3.78 (m, 4H), 3.61 (tt, J = 11.2, 3.5 Hz, 1 H), 3.14 (t, J = 11.2 Hz, 1 H), 2.69 (t, J = 11.2 Hz, 1 H), 2.35- 2.11 (m, 2H), 2.00 (t, J = 8.2 Hz, 2H), 1.74 (d, J = 12.2 Hz, 2H), 1.58-1.42 (m, 1H), 1.42-1.25 (m, 7H).

Example 16: 2-{4-[4-(6-Methoxy-pyridine-3-carbonyl)-piperidin-1 -yl]-2,2- d imethy l-4-oxo-buty l}-7-methy I-3 , 7-d ihyd ro-py rrolo[2 , 3-d]py rimid in-4-one ("C16")

To a stirred solution A8 (86.0 mg; 0.327 mmol) in dichloromethane (1.0 ml_) and DMF (0.2 ml_) B4 (100.6 mg; 0.392 mmol), triethylamine (0.12 ml_; 0.817 mmol) and T3P (50% in ethyl acetate; 598.0 mg; 0.817 mmol) were added and the mixture was stirred at room temperature for 12 h. The reaction mixture was concentrated under reduced pressure, diluted with dichloromethane and washed with 10% sodium bicarbonate solution, water, and brine. The organic layer was dried over sodium sulfate, filtered by suction and concentrated. The residue was purified by prep. HPLC. Yield: (84.0 mg (55%) colorless solid; LC/MS (B), Rt: 3.82 min; (M+H) 466.0

1 H NMR (400 MHz, DMSO-de) δ 12.01 (s, 1 H), 8.93 (s, 1H), 8.24 (dd, J = 2.40, 8.40 Hz, 1 H), 7.04 (d, J = 3.20 Hz, 1 H), 6.96 (d, J = 8.80 Hz, 1 H), 6.41 (s, 1 H), 4.60-4.49 (m, 1 H), 4.17-4.07 (m, 1H), 3.96 (s, 3H), 3.76-3.65 (m, 4H), 3.28- 3.19 (m, 1 H), 2.87-2.78 (m, 1H), 2.68 (s, 3H), 2.48-2.42 (m, 2H), 1.90-1.80 (m, 2H), 1.61-1.32 (m, 2H), 1.05 (s, 6H).

Example 17: 2-{2,2-Dimethyl-4-[4-(1-methyl-1 H-pyrazole-4-carbonyl)-piperidin- 1-yl]-4-oxo-butyl}-7-methyl-3,7-dihydro-pyrrolo[2,3-d]pyrimi din-4-one ("C17")

Preparation according to the procedure described for example 16 using A8 and B1. Yield: 85 mg (59%) colorless solid; LC/MS (B), Rt: 3.08 min; (M+H) 439.0; 1 H NMR (400 MHz, DMSO-de) δ 12.02 (s, 1H), 8.46 (s, 1H), 7.98 (s, 1 H), 7.04 (d, J = 3.60 Hz, 1H), 6.41 (d, J = 3.20 Hz, 1H), 4.59-4.50 (m, 1 H), 4.17-4.06 (m, 1H), 3.89 (s, 3H), 3.68 (s, 3H), 3.30-3.22 (m, 1 H), 3.21-3.12 (m, 1H), 2.80-2.62 (m, 3H), 2.48-2.41 (m, 2H), 1.88-1.75 (m, 2H),1.59-1.32 (m, 2H), 1.05 (s,6 H).

Example 18: 2-{4-[4-(1-Ethyl-1 H-pyrazole-4-carbonyl)-piperidin-1-yl]-2,2- dimethy -oxo-butylH-methyl-S -dihydro-pyrrolop.S-dlpyrimidin^-one ("C18")

Preparation according to the procedure described for example 16 using A8 and B2. Yield: 25 mg (40%) off-white solid; LC/MS (B), Rt: 3.31 min; (M+H) 453.2; H NMR (400 MHz, DMSO-de) δ 12.02 (s, 1 H), 8.50 (s, 1 H), 7.99 (s, 1H), 7.04 (d, J = 3.2 Hz, 1H), 6.40 (d, J = 3.2 Hz, 1 H), 4.59-4.51 (m, 1 H), 4.23- 4.08 (m, 3H) 3.67 (s, 3H), 3.32-3.24 (m, 1 H), 3.22-3.11 (m, 1 H), 2.80-2.62 (m, 3H), 2.48-2.41 (d, J = 3.6 Hz, 2H), 1.88-1.73 (m, 2H), 1.59-1.31 (m, 5H), 1.18 (8, 6H).

Example 19: 2-{4-[4-(1-lsopropyl-1 H-pyrazole-4-carbonyl)-piperidin-1-yl]-2,2- dimethyl-4-oxo-butyl}-7-methyl-3,7-dihydro-pyrrolo[2,3-d]pyr imidin-4-one

("C19")

Preparation according to the procedure described for example 16 using A8 and B3. Yield: 26 mg (40%) off-white solid; LC/MS (B), Rt: 3.57 min; (M+H) 467.2; 1 H NMR (400 MHz, DMSO-de) δ 12.02 (s, 1 H), 8.52 (s, 1Η), 7.99 (s, 1H), 7.04 (d, J = 3.2 Hz, 1H), 6.40 (d, J = 3.2 Hz, 1H), 4.62-4.51 (m, 2H), 4.18- 4.07 (m, 1H), 3.67 (s, 3H), 3.31-3.26 (m, 1H), 3.23-3.11 (m, 1 H), 2.80-2.63 (m, 3H), 2.47-2.38 (m, 2H), 1.89-1.75 (m, 2H), 1.58-1.33 (m, 8H), 1.05 (s, 6H).

Example 20: 6-{2,2-Dimethyl-4-[4-(1-methyl-1 H-pyrazole-4-carbonyl)-piperidin- 1 -yl]-4-oxo-butyl}-1 -methyl- 1 ,5-dihydro-pyrazolo[3,4-d]pyrimidin-4-one ("C20")

Preparation according to the procedure described for example 16 using A9 and B1. Yield: 70 mg (43%) colorless solid; LC/MS (B), Rt: 2.92 min; (M+H) 440.0; H NMR (400 MHz, DMSO-de) δ 12.35 (s, H), 8.46 (s, 1 H), 8.02-7.97 (m, 2H), 4.58-4.49 (m, 1H), 4.17-4.08 (m, 1 H), 3.88 (s, 3H), 3.87 (s, 3H), 3.30- 3.23 (m, 1 H), 3.21-3.11 (m, 1 H), 2.81-2.64 (m, 3H), 2.48-2.40 (m, 2H), 1.88- 1.74 (m, 2H), 1.58-1.30 (m, 2H), 1.07 (s, 6H).

Example 21 : 6-{4-[4-(6-Methoxy-pyridine-3-carbonyl)-piperidin-1-yl]-2,2- dimethyM-oxo-butylJ-l-methyl-I .S-dihydro-pyrazolotS^-dlpyrimidin^-one

("C21")

Preparation according to the procedure described for example 16 using A9 and B4. Yield: 60 mg (35%) colorless solid; LC/MS (B), Rt: 3.64 min; (M+H) 467.3; 1 H NMR (400 MHz, DMSO-de) δ 8.93 (d, J = 2.4 Hz, 1 H), 8.24 (dd, J = 8.4, 2.8 Hz, 1 H), 8.00 (s, 1 H), 6.96 (d, J = 8.8 Hz, 1 H), 4.58-4.50 (m, 1 H), 4.17-4.08 (m, 1 H), 3.95 (s, 3H), 3.87 (s, 3H), 3.74- 3.66 (m, 1 H), 3.28-3.20 (m, 1 H), 2.87-2.78 (m, 1 H), 2.72 (s, 2H), 2.48-2.38 (m, 2H), 1.89 -1.78 (m, 2H), 1.59-1.32 (m, 2H), 1.1 (s, 6H).

Example 22: 6-{4-[4-(6-Methoxy-5-methyl-pyridine-3-carbonyl)-piperidin-1 -yl]-

2,2-dimethyl-4-oxo-butyl}-1-methyl-1 > 5-dihydro-pyrazolo[3 I 4-d]pyrimidin-4-one

("C22")

Preparation according to the procedure described for example 16 using A9 and B5. Yield: 30 mg (17%) colorless solid; LC/MS (B), Rt: 4.13 min; (M+H) 481.2; H NMR (400 MHz, DMSO-de) δ 12.34 (s, 1 H), 8.77 (d, J = 2.4 Hz, 1 H), 8.08 (d, J = 1.2 Hz, 1H), 7.99 (s, 1H), 4.58-4.49 (m, 1H), 4.18-4.07 (m, 1H), 3.98 (s, 3H), 3.87 (s, 3H), 3.74-3.63 (m, 1 H), 3.28-3.17 (m, 1H), 2.89-2.71 (m, 1H), 2.72 (s, 2H), 2.48-2.38 (m, 2H), 2.21 (s, 3H), 1.90-1.77 (m, 2H), 1.62- 1.31 (m, 2H), 1.07 (s, 6H).

Example 23: 2-{2-[4-(4-Methoxy-benzoyl)-piperidin-1 -yl]-2-oxo-ethoxymethyl}- 3H-quinazolin-4-one ("C23")

[(4-oxo-3,4-dihydroquinazolin-2-yl)methoxy]acetic acid (117.1 mg; 0.50 mmol) and (4-Methoxy-phenyl)-piperidin-4-yl-methanone hydrochloride (140.7 mg; 0.55 mmol) were coupled in THF/DMF - 3/1 as described for example 1. Yield: 42 mg (19%) yellow solid; LC/MS (A), Rt: 1.95 min; (M+H) 436.2

H NMR (400 MHz, DMSO-de) δ 12.53 (s, 1 H), 8.12 (dd, J = 8.0, 1.5 Hz, 1 H), 8.03-7.96 (m, 2H), 7.83-7.78 (m, 1H), 7.66-7.62 (m, 1 H), 7.54-7.49 (m, 1H), 7.09-7.03 (m, 2H), 4.52 (s, 2H), 4.46 (d, J = 8.1 Hz, 2H), 4.44-4.36 (m, 1H), 3.85 (s, 3H), 3.81-3.73 (m, 1H), 3.68 (tt, J = 11.1 , 3.6 Hz, 1H), 3.23-3.13 (m, 1 H), 2.91-2.80 (m, 1 H), 1.86-1.73 (m, 2H), 1.66-1.53 (m, 1H), 1.50-1.37 (m, 1H).

Example 24: 2-{2-[4-(1 -Methyl- 1 H-pyrazole-4-carbonyl)-piperidin-1 -yl]-2-oxo- ethoxymethyl}-3H-quinazolin-4-one ("C24")

[(4-oxo-3,4-dihydroquinazolin-2-yl)methoxy]acetic acid (50.0 mg; 0.213 mmol) and B1 (53.9 mg; 0.235 mmol) were coupled as described for example 1. Yield: 51 mg (58%) colorless solid; LC/MS (A), Rt: 1.53 min; (M+H) 410.1 1 H NMR (400 MHz, DMSO-de) δ 12.56 (s, 1H), 8.44 (s, 1H), 8.13 (dd, J = 7.9, 1.2 Hz, 1H), 7.97 (s, 1H), 7.81 (ddd, J = 8.5, 7.3, 1.5 Hz, 1H), 7.65 (d, J = 7.7 Hz, 1 H), 7.52 (ddd, J = 8.1 , 7.2, 1.0 Hz, 1 H), 4.53 (s, 2H), 4.47 (d, J = 8.6 Hz, 2H), 4.44-4.35 (m, 1H), 3.88 (s, 3H), 3.82-3.71 (m, 1H), 3.26 (tt, J = 11.5, 3.7 Hz, 1H), 3.18-3.04 (m, 1H), 2.86-2.74 (m, 1H), 1.87-1.71 (m, 2H), 1.69-1.50 (m, 1H), 1.50-1.33 (m, 1H).

Example 25: 2-{2-[4-(6-Methoxy-pyridine-3-carbonyl)-piperidin-1 -yl]-2-oxo- ethoxymethyl}-3H-quinazolin-4-one ("C25")

[(4-oxo-3,4-dihydroquinazolin-2-yl)methoxy]acetic acid (50.0 mg; 0.213 mmol) and B4 (51.7 mg; 0.235 mmol) were coupled as described for example 1. Yield: 45 mg (49%) colorless solid; LC/MS (A), Rt: 1.83 min; (M+H) 437.2 H NMR (400 MHz, DMSO-de) δ 12.55 (s, 1 H), 8.92 (d, J = 2.2 Hz, 1 H), 8.24 (dd, J = 8.7, 2.5 Hz, 1H), 8.13 (dd, J = 7.9, 1.2 Hz, 1 H), 7.81 (td, J = 7.8, 7.2, 1.5 Hz, 1H), 7.65 (d, J = 7.8 Hz, 1 H), 7.58-7.48 (m, 1 H), 6.96 (d, J = 8.7 Hz, 1 H), 4.53 (s, 2H), 4.47 (d, = 8.6 Hz, 2H), 4.41 (d, J = 12.6 Hz, 1H), 3.96 (s, 3H), 3.78 (d, J = 13.4 Hz, 1 H), 3.74-3.63 (m, 1H), 3.18 (t, J = 11.8 Hz, 1H), 2.86 (t, J = 11.5 Hz, 1H), 1.91-1.75 (m, 2H), 1.60 (qd, J = 13.1 , 3.8 Hz, 1H), 1.44 (qd, J = 12.4, 3.5 Hz, 1H).

Example 26: 6,8-Difluoro-2-{2-[4-(4-methoxy-benzoyl)-piperidin-1-yl]-2-o xo- ethoxymethyl}-3H-quinazolin-4-one ("C26")

A10 (297.2 mg; 1.10 mmol) and (4-Methoxy-phenyl)-piperidin-4-yl-methanone hydrochloride (255.7 mg; 1.00 mmol) were coupled as described for example 1. Yield: 33 mg (7%) colorless solid; LC/MS (A), Rt: 2.13 min; (M+H) 472.2 1 H NMR (400 MHz, DMSO-de) δ 12.89 (s, 1H), 8.07-7.91 (m, 2H), 7.80 (ddd, J = 10.5, 9.1 , 2.9 Hz, 1 H), 7.66 (ddd, J = 8.4, 2.8, 1.4 Hz, 1 H), 7.16-6.96 (m, 2H), 4.55 (s, 2H), 4.47 (d, J = 6.2 Hz, 2H), 4.43-4.34 (m, 1 H), 3.85 (s, 3H), 3.82-3.74 (m, 1H), 3.68 (tt, J = 11.0, 3.7 Hz, 1H), 3.23-3.12 (m, 1 H), 2.92-2.80 (m, 1H), 1.88-1.70 (m, 2H), 1.67-1.49 (m, 1 H), 1.49-1.30 (m, 1H).

Example 27: 6,8-Difluoro-2-{2-[4-(1 -methyl-1 H-pyrazole-4-carbonyl)-piperidin- 1-yl]-2-oxo-ethoxymethyl}-3H-quinazolin-4-one ("C27")

A10 (50.0 mg; 0.185 mmol) and B1 (46.8 mg; 0.204 mmol) were coupled as described for example 1. Yield: 39.5 mg (48%) colorless solid; LC/MS (A), Rt:

1.69 min; (M+H) 446.1

1 H NMR (400 MHz, DMSO-de) δ 12.91 (s, 1 H), 8.44 (s, 1H), 7.97 (s, 1H), 7.82 (ddd, J = 10.5, 9.1 , 2.9 Hz, 1 H), 7.67 (ddd, J = 8.4, 2.7, 1.3 Hz, 1H), 4.55 (s, 2H), 4.48 (d, J = 6.3 Hz, 2H), 4.42-4.35 (m, 1 H), 3.89 (s, 3H), 3.83-3.74 (m,

1H), 3.37-3.20 (m, overlapped with water signal, 1H), 3.19-3.05 (m, 1H), 2.89-

2.70 (m, 1H), 1.88-1.70 (m, 2H), 1.69-1.51 (m, 1H), 1.51-1.31 (m, 1H).

Example 28: 6,8-Difluoro-2-{2-[4-(6-methoxy-pyridine-3-carbonyl)-piperid in-1 - yl]-2-oxo-ethoxymethyl}-3H-quinazolin-4-one ("C28")

A10 (50.0 mg; 0.185 mmol) and B4 (45.7 mg; 0.204 mmol) were coupled as described for example 1. Yield: 30.5 mg (35%) colorless solid; LC/MS (A), Rt: 2.00 min; (M+H) 473.2

1 H NMR (400 MHz, DMSO-de) δ 12.91 (brs, 1 H), 8.91 (d, J = 2.2 Hz, 1H), 8.23 (dd, J = 8.7, 2.5 Hz, 1H), 7.81 (ddd, J = 10.5, 9.1 , 2.9 Hz, 1 H), 7.67 (ddd, J = 8.4, 2.7, 1.3 Hz, 1H), 6.95 (d, J = 8.7 Hz, 1H), 4.55 (s, 2H), 4.53-4.42 (m, 2H), 4.40 (d, J = 13.0 Hz, 1H), 3.95 (s, 3H), 3.84-3.73 (m, 1 H), 3.73-3.62 (m, 1 H), 3.18 (t, J = 12.4 Hz, 1 H), 2.85 (t, J = 11.8 Hz, 1 H), 1.89-1.77 (m, 2H), 1.66- 1.52 (m, 1H), 1.52-1.36 (m, 1 H).

Example 29: 4-(4-Methoxy-benzoyl)-piperidine-1-carboxylic acid [2-(4-oxo-3,4- dihydro-quinazolin-2-yl)-ethyl]-amide ("C29")

29.1 : 3-{[4-(4-Methoxy-benzoyl)-piperidine-1 -carbonyl]-amino}-propionic acid ethyl ester

(4-Methoxy-phenyl)-piperidin-4-yl-methanone hydrochloride (178.7 mg; 0.699 mmol) was suspended in dichloromethane (8.0 ml_). N-Ethyldiisopropylamine (0.145 ml; 0.838 mmol) was added under argon and the mixture was stirred at room temperature for 5 min. Ethyl 3-isocyanatopropionate (93.7 μΙ; 0.699 mmol) was added dropwise to the suspension over a period of 5 min. After complete addition a clear solution was formed, which was stirred for additional 15 min. The reaction mixture was diluted with dichloromethane (30 mL), washed once 5% citric acid solution and once with water, dried with sodium sulfate, filtered by suction and evaporated to dryness. The colorless oil (273 mg; LC/MS (A), Rt: 1.93 min; (M+H) 363.2) was used in the next step without further purification.

29.2: 3-{[4-(4-Methoxy-benzoyl)-piperidine-1-carbonyl]-amino}-prop ionic acid Compound 29.1 (273.0 mg) was dissolved in 1 ,4-dioxane (1.5 mL) and sodium hydroxide solution (2 N; 0.70 mL) was added to the solution and the mixture was stirred at room temperature for 30 min. The reaction mixture was treated with HCI solution (2 N; 0.70 mL), diluted with water and extracted with ethyl acetate. The combined organic layers were washed with brine, dried with sodium sulfate, filtered by suction and evaporated to dryness. Yield: 107 mg

(44%) colorless solid; LC/MS (A), Rt: 1.66 min; (M+H) 335.2

29.3: 4-(4-Methoxy-benzoyl)-piperidine-1-carboxylic acid [2-(2-carbamoyl- phenylcarbamoyl)-ethyl]-amide

2-Aminobenzamide (41.3 mg; 0.304 mmol) and compound 29.2 (101.5 mg; 0.304 mmol) were coupled as described for example 1. The reaction mixture was diluted with water and extracted ethyl acetate. The combined organic layers were washed each with 5% citric acid solution, saturated NaHC03 solution and brine, dried with sodium sulfate, filtered by suction and

evaporated to dryness. The residue (206 mg brown oil; LC/MS (A), Rt: 1.80; (M+H) 453.2) was used in the next step without further purification.

29.4: 4-(4-Methoxy-benzoyl)-piperidine-1-carboxylic acid [2-(4-oxo-3,4- dihydro-quinazolin-2-yl)-ethyl]-amide

Compound 29.3 (206.0 mg) was stirred in sodium hydroxide solution (2 N; 1.0 ml_) at 100 °C for 30 min. The reaction mixture was cooled to room

temperature, neutralized with 2N HCI solution and extracted with ethyl acetate. The combined organic layers were washed with brine, dried with sodium sulfate, filtered by suction and evaporated to dryness. The residue was triturated with acetonitrile, filtered by suction, washed with a small amount of acetonitrile and dried under high vacuum at 80 °C for 2h. Yield: 77 mg (45%) colorless solid; LC/MS (A), Rt: 1.82; (M+H) 435.2

1 H NMR (400 MHz, DMSO-de) δ 12.14 (s, 1 H), 8.07 (dd, J = 7.9, 1.2 Hz, 1 H), 7.97 (d, J = 8.9 Hz, 2H), 7.85-7.67 (m, 1H), 7.59 (d, J = 8.0 Hz, 1 H), 7.53-7.29 (m, 1H), 7.05 (d, J = 8.9 Hz, 2H), 6.61 (t, J = 5.5 Hz, 1H), 3.94 (d, J = 13.3 Hz, 2H), 3.84 (s, 3H), 3.63-3.41 (m, 3H), 2.83 (t, J = 11.6 Hz, 2H), 2.75 (t, J = 6.8 Hz, 2H), 1.67 (d, J = 11.0 Hz, 2H), 1.40 (qd, J = 12.8, 3.8 Hz, 2H).

Example 30: 4-(4-Methoxy-benzoyl)-piperidine-1-carboxylic acid methyl-[2-(4- oxo-3,4-dihydro-quinazolin-2-yl)-ethyl]-amide ("C30")

30.1 : [2-(2-Carbamoyl-phenylcarbamoyl)-ethyl]-methyl-carbamic acid tert-butyl ester

2-Aminobenzamide (250.00 mg; 1.836 mmol), 3-[(tert-Butoxycarbonyl)- (methyl)amino]propanoic acid (410.5 mg; 2.020 mmol) and

[dimethylamino(triazolo[4,5-b]pyridin-3-yloxy)methylene]- dimethyl-ammonium hexafluorophosphate (HATU) (1.047 g; 2.754 mmol) were suspended in DMF (3.0 ml_). N-Ethyldiisopropylamine (0.94 ml_; 5.509 mmol) was added and the mixture was stirred for 2 h at room temperature. A clear brown solution was formed after a few minutes, which was stirred for further 30 min at room temperature.

The reaction mixture was diluted with water and extracted with ethyl acetate. The combined organic layers were washed each with 5% citric acid solution, saturated NaHC03 solution and brine, dried with sodium sulfate, filtered by suction and evaporated to dryness. The residue (672 mg brown solid; LC/MS (A), Rt: 1.81 min; (M(-BOC)+H) 222.1) was used in the next step without further purification.

30.2: Methyl-[2-(4-oxo-3,4-dihydro-quinazolin-2-yl)-ethyl]-carbami c acid tert- butyl ester

Compound 30.1 (672.0 mg) was stirred in sodium hydroxide solution (2 N; 4.9 ml_) at 100 °C for 30 min. The reaction mixture cooled to room temperature, neutralized with 2N HCI solution and extracted with ethyl acetate. The combined organic layers were washed with brine, dried with sodium sulfate, filtered by suction and evaporated to dryness. The solid residue was triturated with acetonitrile, filtered by suction, washed with a small amount of acetonitrile and dried. Yield: 443 mg (75%) colorless solid; LC/MS (A), Rt: 1.73 min;

(M+H) 304.2

30.3: 2-[2-(methylamino)ethyl]-3H-quinazolin-4-one, dihydrochloride

Compound 30.2 (443.0 mg; 1.460 mmol) was suspended in 1 ,4-dioxane (5.0 mL). A solution of HCI in 1 ,4-dioxane (4 M, 7.302 ml; 29.207 mmol) and water (1.0 mL) was added and the suspension was stirred at room temperature for 1 h. The reaction mixture was evaporated to dryness and the residue was lyophilized. Yield: 402 mg (100%) colorless solid; LC/MS (A), Rt: 0.85; (M+H) 204.2

30.4: 4-(4-Methoxy-benzoyl)-piperidine-1-carboxylic acid methyl-[2-(4-oxo-3,4- dihydro-quinazolin-2-yl)-ethyl]-amide

(4-Methoxy-phenyl)-piperidin-4-yl-methanone; hydrochloride (200.0 mg; 0.782 mmol) was suspended in dry THF (5.0 mL). N-Ethyldiisopropylamine (0.54 mL; 3.128 mmol) was added and the mixture was cooled to 0 °C. A solution of bis(trichloromethyl) carbonate (243.7 mg; 0.821 mmol), dissolved in THF (1.0 mL), was added dropwise over a period of 3 min and the mixture was stirred for 30 min at 0-5 °C. Compound 30.3 (216.0 mg; 0.782 mmol) was added followed by the slow addition of sodium hydroxide solution (2 N; 1.17 mL; 2.346 mmol). The clear, light yellow solution was stirred at room temperature for 3 h. Further sodium hydroxide solution (2 N; 0.60 mL; 1.200 mmol) was added and the mixture was stirred at room temperature for 2.5 h. The reaction mixture was diluted with water (30 mL) and ethyl acetate (30 mL). Between the two layers a light yellow precipitate was formed. It was filtered off by suction, washed with water and a small amount of acetonitrile, and purified by flash chromatography (Companion RF; 24 g Si50 silica gel column). Yield: 81.5 mg (23%) light yellow solid; LC/MS (A), Rt: 1.91 min; (M+H) 449.2

1 H NMR (400 MHz, DMSO-de) δ 12.22 (s, 1H), 8.06 (dd, J = 7.9, 1.2 Hz, 1 H), 8.00-7.89 (m, 2H), 7.75 (ddd, J = 8.5, 7.2, 1.5 Hz, 1 H), 7.58 (d, J = 8.0 Hz, 1H), 7.50-7.37 (m, 1 H), 7.11-6.95 (m, 2H), 3.84 (s, 3H), 3.56 (t, J = 7.0 Hz, 2H), 3.53-3.41 (m, 3H), 2.85 (t, J = 7.0 Hz, 2H), 2.82 (s, 3H), 2.81-2.73 (m, 2H), 1.62 (d, J = 10.8 Hz, 2H), 1.45 (qd, J = 12.6, 3.6 Hz, 2H). Example 31 : 4-(1-Methyl-1 H-pyrazole-4-carbonyl)-piperidine-1-carboxylic acid methyl-[2-(4-oxo-3,4-dihydro-quinazolin-2-yl)-ethyl]-amide ("C31 ")

Preparation as described for example 30 using compound 30.3 (180.3 mg; 0.653 mmol) and B1 (150.0 mg; 0.653 mmol). Yield: 40 mg (14%) light green solid; LC/MS (A), Rt: 1.52 min; (M+H) 423.2

1 H NMR (400 MHz, DMSO-de) δ 12.20 (s, 1 H), 8.39 (s, 1 H), 8.06 (dd, J = 7.9, 1.3 Hz, 1H), 7.92 (s, 1H), 7.81-7.71 (m, 1H), 7.58 (d, J = 8.0 Hz, 1H), 7.44 (t, J = 7.1 Hz, 1H), 3.87 (s, 3H), 3.56 (t, J = 7.0 Hz, 2H), 3.46 (d, J = 13.2 Hz, 2H), 3.07 (tt, J = 11.7, 3.6 Hz, 1 H), 2.90-2.78 (m, 5H), 2.71 (t, J = 11.5 Hz, 2H), 1.62 (d, J = 10.6 Hz, 2H), 1.55-1.38 (m, 2H).

Following examples are prepared analogously



The following derivatives were prepared analogously:

Example 32: 4-{1-[3,3-Dimethyl-4-(4-oxo-3,4-dihydro-quinazolin-2-yl)-but yryl]- piperidine-4-carbonyl}-benzoic acid methyl ester ("C32")

Yield: 110 mg (100%) colorless solid; LC/MS (A), Rt: 2.19 min; (M+H) 490.2;

1 H NMR (500 MHz, DMSO-de) δ 12.46 (s, 1H), 8.16-8.07 (m, 5H), 7.78 (td, J = 7.8, 7.3, 1.5 Hz, 1 H), 7.62 (d, J - 8.0 Hz, 1 H), 7.50-7.45 (m, 1 H), 4.54 (d, J = 13.0 Hz, 1 H), 4.14 (d, J = 13.5 Hz, 1 H), 3.91 (s, 3H), 3.78 (tt, J = 11.1 , 3.4 Hz, 1 H), 3.30-3.23 (m, 1 H), 2.85 (t, J = 11.6 Hz, 1 H), 2.72 (d, J = 13.0 Hz, 1 H), 2.67 (d, J = 13.0 Hz, 1H), 2.50 (s, 2H), 1.87 (t, J = 14.0 Hz, 2H), 1.54 (qd, J = 12.6, 3.6 Hz, 1H), 1.43 (qd, J = 12.7, 3.8 Hz, 1H), 1.10 (s, 3H), 1.09 (s, 3H).

Example 33: 4-{1 -[3,3-Dimethyl-4-(4-oxo-3,4-dihydro-quinazolin-2-yl)-butyryl ]- piperidine-4-carbonyl}-benzoic acid ("C33")

Yield: 65.5 mg (75%) colorless solid; LC/MS (A), Rt: 1.92 min; (M+H) 476.2 1 H NMR (500 MHz, DMSO-de) δ 13.33 (s, br, 1 H), 12.47 (s, 1H), 8.14-8.05 (m, 5H), 7.81-7.74 (m, 1 H), 7.62 (d, J = 8.0 Hz, 1 H), 7.50-7.44 (m, 1 H), 4.54 (d, J = 12.9 Hz, 1H), 4.13 (d, J = 13.5 Hz, 1H), 3.78 (tt, J = 1 .1, 3.4 Hz, 1H), 3.29- 3.22 (m, 1 H), 2.85 (t, J = 11.6 Hz, 1 H), 2.72 (d, J = 13.0 Hz, 1 H), 2.67 (d, J = 13.0 Hz, 1 H), 2.50 (s, 2H), 1.94-1.81 (m, 2H), 1.54 (qd, J = 12.9, 12.5, 3.4 Hz, 1 H), 1.43 (qd, J = 12.8, 3.8 Hz, 1H), 1.10 (s, 3H), 1.09 (s, 3H).

Example 34: 3-{1 -[3,3-Dimethyl-4-(4-oxo-3,4-dihydro-quinazolin-2-yl)-butyryl ]- piperidine-4-carbonyl}-benzoic acid methyl ester ("C34")

Yield: 82 mg (75%) colorless solid; LC/MS (A), Rt: 2.17 min; (M+H) 490.2 1 H NMR (500 MHz, DMSO-de) δ 12.48 (s, 1H), 8.49 (t, J = 1.6 Hz, 1H), 8.31 (dt, J = 7.8, 1.3 Hz, 1 H), 8.22 (dt, J = 7.8, 1.3 Hz, 1 H), 8.10 (dd, J = 7.9, 1.3 Hz, 1H), 7.78 (ddd, J = 8.5, 7.2, 1.6 Hz, 1H), 7.73 (t, J = 7.8 Hz, 1 H), 7.62 (d, J = 8.0 Hz, 1H), 7.47 (ddd, J = 8.1 , 7.2, 1.0 Hz, 1 H), 4.54 (d, J = 13.1 Hz, 1 H), 4.13 (d, J = 13.5 Hz, 1 H), 3.92 (s, 3H), 3.81 (tt, J = 11.1 , 3.6 Hz, 1H), 3.34- 3.25 (m, 1 H), 2.87 (t, J = 11.6 Hz, 1 H), 2.72 (d, J = 13.0 Hz, 1 H), 2.67 (d, J = 13.0 Hz, 1 H), 2.50 (s, 2H), 1.92-1.82 (m, 2H), 1.55 (qd, J = 12.7, 3.6 Hz, 1H), 1.44 (qd, J = 12.8, 3.8 Hz, 1H), 1.10 (s, 3H), 1.09 (s, 3H).

Example 35: 3-{1 -[3,3-Dimethyl-4-(4-oxo-3,4-dihydro-quinazolin-2-yl)-butyryl ]- piperidine-4-carbonyl}-benzoic acid ("C35")

Yield: 62 mg (92%) colorless solid; LC/MS (A), Rt: 1.92 min; (M+H) 476.2 1 H NMR (500 MHz, DMSO-de) δ 13.30 (s, br, 1 H), 12.48 (s, 1H), 8.49 (t, J = 1.6 Hz, 1 H), 8.27 (dt, J = 7.8, 1.3 Hz, 1 H), 8.20 (dt, J = 7.7, 1.3 Hz, 1 H), 8.10 (dd, J = 7.9, 1.3 Hz, 1 H), 7.78 (ddd, J = 8.5, 7.2, 1.6 Hz, 1 H), 7.70 (t, J = 7.8 Hz, 1H), 7.62 (d, J= 7.9 Hz, 1H), 7.47 (ddd, J = 8.0, 7.2, 1.0 Hz, 1H), 4.54 (d, J= 13.0 Hz, 1H), 4.13 (d, J= 13.5 Hz, 1H), 3.81 (tt, J= 11.1, 3.6 Hz, 1H), 3.35-3.25 (m, H), 2.87 (t, J= 11.6 Hz, 1H), 2.72 (d, J= 13.0 Hz, 1H), 2.67 (d, J= 13.0 Hz, 1H), 2.50 (s, 2H), 1.93-1.81 (m, 2H), 1.56 (qd, J= 12.6, 3.5 Hz, 1H), 1.44 (qd, J= 12.8, 3.8 Hz, 1H), 1.10 (s, 3H), 1.09 (s, 3H).

Example 36: 3-{1-[3,3-Dimethyl-4-(8-methyl-4-oxo-3,4-dihydro-quinazolin- 2- yl)-butyryl]-piperidine-4-carbonyl}-benzoic acid methyl ester ("C36")

Yield: 99 mg (91%) colorless solid; LC/MS (A), Rt: 2.43 min; (M+H) 504.3 1 H NMR (500 MHz, DMSO-de) δ 12.34 (s, 1H), 8.48 (t, J= 1.6 Hz, 1H), 8.30 (dt, J= 7.8, 1.3 Hz, 1H), 8.22 (dt, J= 7.8, 1.3 Hz, 1H), 7.95-7.92 (m, 1H), 7.73 (t, J = 7.8 Hz, 1 H), 7.64 (d, J = 7.0 Hz, 1 H), 7.34 (t, J = 7.6 Hz, 1 H), 4.52 (d, J = 13.1 Hz, 1H), 4.10 (d, J= 13.2 Hz, 1H), 3.91 (s, 3H), 3.79 (tt, J = 11.1, 3.4 Hz, 1H), 3.26 (t, J= 10.0 Hz, 1H), 2.84 (t, J= 11.5 Hz, 1H), 2.73 (s, 2H), 2.61- 2.44 (m, 5H, overlap with DMSO), 1.85 (t, J = 12.6 Hz, 2H), 1.61-1.47 (m, 1H), 1.47-1.35 (m, 1H), 1.11 (s, 6H).

Example 37: 3-{1-[3,3-Dimethyl-4-(8-methyl-4-oxo-3,4-dihydro-quinazolin- 2- yl)-butyryl]-piperidine-4-carbonyl}-benzoic acid ("C37")

Yield: 65 mg (80%) colorless solid; LC/MS (A), Rt: 2.18 min; (M+H) 490.2 H NMR (500 MHz, DMSO-de) δ 13.30 (s, br, 1H), 12.34 (s, 1 H), 8.48 (s, 1H), 8.26 (d, J = 7.8 Hz, 1H), 8.22-8.16 (m, 1H), 7.96-7.91 (m, 1H), 7.70 (t, J = 7.7 Hz, 1 H), 7.64 (d, J = 7.1 Hz, 1H), 7.34 (t, J = 7.6 Hz, 1H), 4.52 (d, J = 13.0 Hz, 1 H), 4.10 (d, J = 13.5 Hz, 1 H), 3.78 (tt, J = 11.1, 3.5 Hz, 1H), 3.29-3.21 (m, 1H, overlap with water), 2.84 (t, J = 11.5 Hz, 1H), 2.73 (s, 2H), 2.57-2.47 (m, 5H, overlap with DMSO), 1.90-1.80 (m, 2H), 1.53 (q, J = 11.3 Hz, 1H), 1.47- 1.31 (m, 1 H), 1.11 (s, 6H).

Example 38: 4-{1 -[3,3-Dimethyl-4-(4-oxo-3,4-dihydro-quinazolin-2-yl)-butyryl ]- piperidine-4-carbonyl}-2-methyl-benzoic acid methyl ester ("C38")

Yield: 51 mg (49%) pale-brown solid; LC/MS (A), Rt: 2.30 min; (M+H) 504.3 1 H NMR (500 MHz, DMSO-de) δ 12.48 (s, 1 H), 8.10 (dd, J = 7.9, 1.3 Hz, 1 H), 7.96-7.88 (m, 3H), 7.81-7.76 (m, H), 7.62 (d, J = 7.9 Hz, 1H), 7.50-7.45 (m, 1 H), 4.54 (d, J = 13.1 Hz, 1 H), 4.13 (d, J = 13.5 Hz, 1H), 3.88 (s, 3H), 3.77 (tt, J = 11.1 , 3.5 Hz, 1 H), 3.30-3.18 (m, 1H), 2.85 (t, J = 11.6 Hz, 1 H), 2.78-2.64 (m, 2H), 2.59 (s, 3H), 2.53-2.48 (m, 2H, overlap with DMSO), 1.90-1.80 (m, 2H), 1.59-1.47 (m, 1H), 1.42 (qd, J = 12.8, 3.8 Hz, 1 H), 1.11-1.04 (m, 6H). Example 39: 4-{1 -[3,3-Dimethyl-4-(4-oxo-3,4-dihydro-quinazolin-2-yl)-butyryl ]- piperidine-4-carbonyl}-2-methyl-benzoic acid ("C39")

Yield: 24 mg (63%) pale-brown solid; LC/MS (A), Rt: 2.01 min; (M+H) 490.2 1 H NMR (500 MHz, DMSO-de) δ 13.17 (s, br, 1H), 12.47 (s, 1 H), 8.09 (dd, J = 7.9, 1.3 Hz, 1H), 7.94-7.85 (m, 3H), 7.77 (td, = 7.8, 7.2, 1.5 Hz, 1H), 7.61 (d, J = 8.0 Hz, 1 H), 7.49-7.43 (m, 1H), 4.53 (d, J = 13.0 Hz, 1 H), 4.12 (d, J = 13.4 Hz, 1H), 3.76 (tt, J = 11.1 , 3.5 Hz, 1H), 3.35-3.21 (m, 1H), 2.84 (t, J = 11.6 Hz, 1 H), 2.71 (d, J = 13.0 Hz, 1 H), 2.66 (d, J = 13.1 Hz, 1H), 2.58 (s, 3H), 2.49 (s, 2H), 1.91-1.79 (m, 2H), 1.52 (qd, = 12.7, 3.5 Hz, 1H), 1.41 (qd, J = 12.8, 3.8 Hz, 1H), 1.09 (s, 3H), 1.08 (s, 3H).

Example 40: 4-{1-[3,3-Dimethyl-4-(8-methyl-4-oxo-3,4-dihydro-quinazolin- 2- yl)-butyryl]-piperidine-4-carbonyl}-2-methyl-benzoic acid methyl ester ("C40")

Yield: 101 mg (98%) colorless solid; LC/MS (A), Rt: 2.55 min; (M+H) 518.2 1 H NMR (500 MHz, DMSO-de) δ 12.34 (s, 1H), 7.96-7.88 (m, 4H), 7.64 (d, J = 7.1 Hz, 1 H), 7.34 (t, J = 7.6 Hz, 1H), 4.52 (d, J = 13.0 Hz, 1 H), 4.10 (d, J = 13.5 Hz, 1H), 3.88 (s, 3H), 3.75 (tt, J = 11.0, 3.3 Hz, 1 H), 3.28-3.16 (m, 1 H), 2.82 (t, J = 11.7 Hz, 1 H), 2.73 (s, 2H), 2.59 (s, 3H), 2.55-2.49 (m, 5H, overlap with DMSO), 1.90-1.80 (m, 2H), 1.59-1.45 (m, 1H), 1.45-1.31 (m, 1H), 1.11 (s, 6H).

Example 41 : 4-{1-[3,3-Dimethyl-4-(8-methyl-4-oxo-3,4-dihydro-quinazolin- 2- yl)-butyryl]-piperidine-4-carbonyl}-2-methyl-benzoic acid ("C41 ")

Yield: 70 mg (84%) colorless solid; LC/MS (A), Rt: 2.25 min; (M+H) 504.3 1 H NMR (500 MHz, DMSO-de) δ 13.17 (s, br, 1 H), 12.34 (s, 1H), 7.95-7.84 (m, 4H), 7.63 (d, J = 7.2 Hz, 1 H), 7.33 (t, J = 7.6 Hz, 1 H), 4.51 (d, J = 12.9 Hz, 1 H), 4.09 (d, J = 13.4 Hz, 1 H), 3.73 (tt, J = 11.1 , 3.4 Hz, 1H), 3.30-3.17 (m, 1 H), 2.81 (t, J = 11.6 Hz, 1H), 2.72 (s, 2H), 2.58 (s, 3H), 2.57-2.49 (m, 5H), 1.89-1.77 (m, 2H), 1.50 (qd, J = 12.9, 3.6 Hz, 1 H), 1.38 (qd, J = 12.7, 3.8 Hz, 1H), 1.10 (s, 6H).

Example 42: 3-{1-[(R)-3-Methyl-4-(8-methyl-4-oxo-3,4-dihydro-quinazolin- 2-yl)- butyryl]-piperidine-4-carbonyl}-benzoic acid methyl ester ("C42")

Example 43: 3-{1-[(S)-3-Methyl-4-(8-methyl-4-oxo-3,4-dihydro-quinazolin- 2-yl)- butyryl]-piperidine-4-carbonyl}-benzoic acid methyl ester ("C43")

A12 (83.0 mg; 0.317 mmol) and B7 (100.0 mg; 0.343 mmol) were coupled as described for example 1 ; Yield: 151 mg (97%) brown oil.

The preparative separation of the enantiomers was performed by SFC

(column: ChiralPak AD-H; eluent: C02:methanol (containing 0.5%

diethylamine) - 60:40). The combined fractions were evaporated to dryness and freeze-dried.

Example 42: 15 mg pale-yellow solid; LC/MS (A), Rt: 2.24 min; (M+H) 490.2; 1 H NMR (500 MHz, DMSO-ds) δ 12.12 (s, 1 H), 8.45 (t, J = 1.3 Hz, 1H), 8.27 (dt, J = 7.8, 1.3 Hz, 1 H), 8.20 (dt, J = 7.7, 1.2 Hz, 1 H), 7.91 (d, J = 7.9 Hz, 1 H), 7.71 (t, J = 7.8 Hz, 1 H), 7.62 (d, J = 7.2 Hz, 1H), 7.32 (t, J = 7.6 Hz, 1H), 4.35 (d, J = 10.0 Hz, 1 H), 4.00-3.83 (m, 4H), 3.81-3.66 (m, 1H), 3.21 (t, J = 12.9

Hz, 1H), 2.73 (q, J = 10.8 Hz, 1H), 2.67-2.43 (m, 7H), 2.34-2.20 (m, 1 H), 1.85- 1.73 (m, 2H), 1.58-1.43 (m, 1 H), 1.40-1.20 (m, 1H), 0.98 (d, J = 6.0 Hz, 3H). Example 43: 15 mg pale-yellow solid; LC/MS (A), Rt: 2.24 min; (M+H) 490.2; H NMR (500 MHz, DMSO-de) δ 12.12 (s, 1H), 8.45 (t, J = 1.5 Hz, 1H), 8.28 (dt, J = 7.8, 1.3 Hz, 1 H), 8.20 (dt, J = 7.7, 1.2 Hz, 1 H), 7.91 (d, J = 7.8 Hz, 1 H), 7.71 (t, J = 7.8 Hz, 1 H), 7.62 (d, J = 7.2 Hz, 1H), 7.32 (t, J = 7.6 Hz, 1H), 4.35 (d, J = 9.9 Hz, 1H), 4.00-3.83 (m, 4H), 3.80-3.69 (m, 1 H), 3.26-3.13 (m, 1 H), 2.80-2.68 (m, 1H), 2.68-2.41 (m, 7H), 2.36-2.21 (m, 1 H), 1.87-1.71 (m, 2H), 1.58-1.43 (m, 1 H), 1.39-1.20 (m, 1 H), 0.98 (d, J = 6.0 Hz, 3H).

Example 44: 3-{1-[(R)-3-Methyl-4-(8-methyl-4-oxo-3,4-dihydro-quinazolin- 2-yl)- butyryl]-piperidine-4-carbonyl}-benzoic acid ("C44")

Yield: 42 mg (36%) pale-brown solid; LC/MS (A), Rt: 2.00 min; (M+H) 476.2 1 H NMR (500 MHz, DMSO-de) δ 13.29 (s, br, 1H), 12.13 (s, 1 H), 8.46 (s, 1H), 8.24 (d, J = 7.8 Hz, 1 H), 8.19 (d, J = 7.8 Hz, 1 H), 7.92 (d, J = 7.8 Hz, 1 H), 7.69 (t, J = 7.7 Hz, 1H), 7.63 (d, J = 7.2 Hz, 1 H), 7.33 (t, J = 7.6 Hz, 1H), 4.41-4.30 (m, 1 H), 3.95 (t, J = 12.9 Hz, 1 H), 3.75 (t, J = 10.6 Hz, 1H), 3.26-3.13 (m, 1H, overlap with water), 2.74 (q, J = 10.8 Hz, 1H), 2.69-2.61 (m, 1H), 2.61-2.47 (m, 6H, overlap with DMSO), 2.36-2.23 (m, 1H), 1.81 (t, J = 11.4 Hz, 2H), 1.59-1.44 (m, 1H), 1.42-1.27 (m, 1H), 0.99 (d, J = 6.0 Hz, 3H).

Example 45: 3-{1 -[(S)-3-Methyl-4-(8-methyl-4-oxo-3,4-dihydro-quinazolin-2-y I)- butyryl]-piperidine-4-carbonyl}-benzoic acid ("C45")

Yield: 41 mg (34%) pale-brown solid; LC/MS (A), Rt: 2.00 min; (M+H) 476.2 1 H NMR (500 MHz, DMSO-de) δ 13.32 (s, br, 1H), 12.13 (s, 1H), 8.46 (s, 1H), 8.22-8.14 (m, 2H), 7.92 (d, J = 7.8 Hz, 1 H), 7.69-7.59 (m, 2H), 7.33 (t, J = 7.6 Hz, 1 H), 4.37 (d, J = 10.0 Hz, 1 H), 3.95 (t, J = 12.8 Hz, 1H), 3.74 (t, J = 10.4 Hz, 1H), 3.50-3.04 (m, 1H, overlap with HDO), 2.74 (q, J = 11.1 Hz, 1H), 2.70- 2.60 (m, 1 H), 2.60-2.44 (m, 6H, overlap with DMSO), 2.35-2.20 (m, 1 H), 1.80 (t, J = 11.5 Hz, 2H), 1.59-1.44 (m, 1 H), 1.41-1.23 (m, 1 H), 0.99 (d, J = 5.9 Hz, 3H). Example 46: 3-{1-[(R)-4-(6-Fluoro-8-methyl-4-oxo-3,4-dihydro-quinazolin- 2-yl)- 3-methyl-butyryl]-piperidine-4-carbonyl}-benzoic acid ("C46")

Yield: 24 mg (42%) colorless solid; LC/MS (A), Rt: 2.08 min; (M+H) 494.2

1 H NMR (500 MHz, DMSO-de) δ 14.01-12.50 (m, 1 H), 12.26 (s, 1H), 8.47-8.44 (m, 1H), 8.22-8.16 (m, 2H), 7.66 (t, J = 7.7 Hz, 1 H), 7.59-7.52 (m, 2H), 4.39- 4.30 (m, 1 H), 3.99-3.89 (m, 1H), 3.78-3.69 (m, 1 H), 3.26-3.16 (m, 1H), 2.78- 2.68 (m, 1 H), 2.66-2.60 (m, 1H), 2.59-2.51 (m, 6H), 2.34-2.21 (m, 1H), 1.85- 1.74 (m, 2H), 1.56-1.44 (m, 1H), 1.38-1.25 (m, 1 H), 0.97 (d, J = 5.8 Hz, 3H).

Example 47: 3-{1 -[(S)-4-(6-Fluoro-8-methyl-4-oxo-3,4-dihydro-quinazolin-2-yl )- 3-methyl-butyryl]-piperidine-4-carbonyl}-benzoic acid ("C47")

Yield: 37 mg (67%) colorless solid; LC/MS (A), Rt: 2.09 min; (M+H) 494.1 H NMR (400 MHz, DMSO-de) δ 13.26 (s, 1 H), 12.26 (s, 1H), 8.47-8.43 (m, 1 H), 8.26-8.21 (m, 1H), 8.20-8.16 (m, 1H), 7.68 (t, J = 7.7 Hz, 1H), 7.59-7.52 (m, 2H), 4.34 (d, J = 12.6 Hz, 1H), 3.94 (t, J = 12.5 Hz, 1 H), 3.74 (t, J = 11.3 Hz, 1H), 3.21 (t, J = 12.9 Hz, 1 H), 2.73 (q, J = 11.9, 11.5 Hz, 1 H), 2.67-2.55 (m, 3H), 2.52 (s, 3H), 2.50-2.43 (m, 1 H), 2.34-2.20 (m, 1 H), 1.85-1.73 (m, 2H), 1.58-1.43 (m, 1 H), 1.39-1.22 (m, 1 H), 0.97 (d, J = 5.5 Hz, 3H).

Example 48: 2-{4-[4-(4-Dimethylaminomethyl-benzoyl)-piperidin-1 -yl]-2,2- dimethyl-4-oxo-butyl}-3H-quinazolin-4-one ("C48")

A1 (125.5 mg; 0.317 mmol) and B8 (150.0 mg; 0.530 mmol) were coupled as described for example 1. Yield: 38 mg (16%) colorless solid; LC/MS (A), Rt: 1.55 min; (M+H) 489.3

H NMR (500 MHz, DMSO-de) δ 12.48 (s, 1H), 8.09 (dd, J = 8.0, 1.5 Hz, 1H), 7.99-7.95 (m, 2H), 7.79-7.75 (m, 1 H), 7.62-7.59 (m, 1 H), 7.48-7.44 (m, 3H), 4.57-4.51 (m, 1H), 4.16-4.09 (m, 1 H), 3.76-3.69 (m, 1H), 3.52-3.44 (m, 2H), 3.29-3.22 (m, 1 H), 2.87-2.80 (m, 1 H), 2.73-2.64 (m, 2H), 2.49-2.47 (m, 2H), 2.17 (s, 6H), 1.89-1.79 (m, 2H), 1.58-1.49 (m, 1 H), 1.47-1.36 (m, 1H), 1.09 (s, 3H), 1.08 (s, 3H).

Example 49: 3-{2,2-Dimethyl-4-[4-(1-methyl-1 H-pyrazole-4-carbonyl)- piperidin-1-yl]-4-oxo-butyl}-2H-isoquinolin-1-one ("C49")

A14 (22.0 mg; 0.085 mmol) and B1 (23.4 mg; 0.102 mmol) were coupled as described for example 1. Yield: 22 mg (60%) colorless solid; LC/MS (A), Rt: 2.08 min; (M+H) 435.3

1 H NMR (500 MHz, DMSO-de) δ 11.64 (s, 1H), 8.45 (s, 1H), 8.15-8.12 (m, 1H), 7.98-7.97 (m, 1H), 7.68-7.62 (m, 1H), 7.59-7.55 (m, 1 H), 7.44-7.39 (m, 1 H), 6.35-6.32 (m, 1 H), 4.61-4.55 (m, 1H), 4.16-4.09 (m, 1H), 3.88 (s, 3H), 3.32- 3.24 (m, 1H), 3.23-3.16 (m, 1H), 2.79 (td, J = 12.7, 2.8 Hz, 1 H), 2.61-2.52 (m, 2H), 2.34 (s, 2H), 1.88-1.77 (m, 2H), 1.56-1.47 (m, 1H), 1.47-1.38 (m, 1 H), 1.05-1.00 (m, 6H).

Example 50: 3-{(S)-2-Methyl-4-[4-(1-methyl-1 H-pyrazole-4-carbonyl)-piperidin- 1-yl]-4-oxo-butyl}-2H-isoquinolin-1-one ("C50")

Example 51 : 3-{(R)-2-Methyl-4-[4-(1-methyl-1 H-pyrazole-4-carbonyl)-piperidin- 1 -yl]-4-oxo-butyl}-2H-isoquinolin-1 -one ("C51 ")

A15 (118.2 mg; 0.482 mmol) and B1 (110.7 mg; 0.482 mmol) were coupled as described for example 1. Yield: 140 mg 68%) pale-yellow foam

The preparative separation of the enantiomers was performed by SFC

(column: ChiralPak AD-H; eluent: C02:2-propanol (containing 0.5%

diethylamine) - 60:40). The combined fractions were evaporated to dryness and freeze-dried.

Example 50: 57 mg colorless solid; LC/MS (A), Rt: 1.83 min; (M+H) 421.3; 1 H NMR (500 MHz, DMSO-de) δ 11.24 (s, 1H), 8.44 (s, 1 H), 8.13 (d, J = 7.9 Hz, 1 H), 7.96 (s, 1 H), 7.65 (t, J = 7.5 Hz, 1 H), 7.57 (d, J = 7.9 Hz, 1 H), 7.45-7.38 (m, 1H), 6.34 (s, 1 H), 4.47-4.35 (m, 1 H), 3.99-3.83 (m, 4H), 3.23 (t, J = 10.6 Hz, 1H), 3.12 (t, J = 12.9 Hz, 1H), 2.67 (t, J = 12.5 Hz, 1H), 2.56-2.50 (m, 1H), 2.43-2.16 (m, 4H), 1.76 (d, J = 12.7 Hz, 2H), 1.49 (qd, J = 12.7, 3.8 Hz, 1 H), 1.42-1.24 (m, 1 H), 0.95-0.89 (m, 3H). Example 51 : 52 mg colorless solid; LC/MS (A), Rt: 1.83 min; (M+H) 421.2; H NMR (500 MHz, DMSO-de) δ 11.24 (s, 1H), 8.44 (s, 1 H), 8.13 (d, J = 8.0 Hz, 1H), 7.96 (s, 1H), 7.65 (t, J = 7.5 Hz, 1H), 7.57 (d, J = 7.9 Hz, 1 H), 7.41 (t, J = 7.5 Hz, 1H), 6.34 (s, 1H), 4.46-4.37 (m, 1 H), 3.98-3.84 (m, 4H), 3.23 (t, J = 10.6 Hz, 1H), 3.12 (t, J = 12.9 Hz, 1H), 2.67 (t, J = 12.5 Hz, 1H), 2.56-2.48 (m, 1 H), 2.43-2.17 (m, 4H), 1.76 (d, J = 12.7 Hz, 2H), 1.49 (qd, J = 12.7, 3.7 Hz, 1H), 1.40-1.25 (m, 1 H), 0.95-0.89 (m, 3H).

Example 52: 4-{1-[3,3-Dimethyl-4-(1-oxo-1 ,2-dihydro-isoquinolin-3-yl)-butyryl]- piperidine-4-carbonyl}-benzoic acid ("C52")

Yield: 53 mg (84%) colorless solid; LC/MS (A), Rt: 2.22 min; (M+H) 475.2

1 H NMR (500 MHz, DMSO-de) δ 13.31 (s, 1 H), 11.62 (s, 1H), 8.15-8.06 (m, 5H), 7.67-7.63 (m, 1H), 7.59-7.55 (m, 1H), 7.43-7.39 (m, 1 H), 6.35-6.31 (m, 1 H), 4.60-4.54 (m, 1 H), 4.17-4.10 (m, 1 H), 3.82-3.74 (m, 1 H), 3.31-3.22 (m, 1 H), 2.91-2.83 (m, 1 H), 2.62-2.53 (m, 2H), 2.38-2.31 (m, 2H), 1.94-1.82 (m, 2H), 1.58-1.48 (m, 1 H), 1.48-1.38 (m, 1 H), 1.03 (s, 3H), 1.03 (s, 3H).

Example 53: 3-{1-[3,3-Dimethyl-4-(1-oxo-1 ,2-dihydro-isoquinolin-3-yl)-butyryl]- piperidine-4-carbonyl}-benzoic acid ("C53")

Yield: 56 mg (56%) colorless solid; LC/MS (A), Rt: 2.21 min; (M+H) 475.2 1 H NMR (500 MHz, DMSO-de) δ 13.27 (s, 1 H), 11.63 (s, 1 H), 8.50-8.47 (m, 1 H), 8.26 (d, J = 7.7 Hz, 1 H), 8.21-8.18 (m, 1 H), 8.15-8.11 (m, 1 H), 7.69 (t, J 7.7 Hz, 1 H), 7.67-7.62 (m, 1 H), 7.57 (d, J = 7.9 Hz, 1 H), 7.44-7.39 (m, 1 H),

6.34- 6.31 (m, 1 H), 4.60-4.54 (m, 1 H), 4.16-4.10 (m, 1 H), 3.84-3.77 (m, 1 H),

3.35- 3.27 (m, 1 H), 2.93-2.86 (m, 1 H), 2.61-2.53 (m, 2H), 2.38-2.32 (m, 2H), 1.93-1.82 (m, 2H), 1.60-1.49 (m, 1 H), 1.49-1.40 (m, 1 H), 1.06-1.00 (m, 6H).

The following examples relate to medicaments:

Example A: Injection vials

A solution of 100 g of an active ingredient of the formula I and 5 g of disodium hydrogenphosphate in 3 I of bidistilled water is adjusted to pH 6.5 using 2 N hydrochloric acid, sterile filtered, transferred into injection vials, lyophilised under sterile conditions and sealed under sterile conditions. Each injection vial contains 5 mg of active ingredient.

Example B: Suppositories

A mixture of 20 g of an active ingredient of the formula I with 100 g of soya lecithin and 1400 g of cocoa butter is melted, poured into moulds and allowed to cool. Each suppository contains 20 mg of active ingredient.

Example C: Solution

A solution is prepared from 1 g of an active ingredient of the formula I, 9.38 g of NaH 2 P0 4 · 2 H 2 O, 28.48 g of Na 2 HP0 4 · 12 H2O and 0.1 g of benzalkonium chloride in 940 ml of bidistilled water. The pH is adjusted to 6.8, and the solution is made up to 1 I and sterilised by irradiation. This solution can be used in the form of eye drops.

Example D: Ointment

500 mg of an active ingredient of the formula I are mixed with 99.5 g of Vaseline under aseptic conditions. Example E: Tablets

A mixture of 1 kg of active ingredient of the formula I, 4 kg of lactose, 1.2 kg of potato starch, 0.2 kg of talc and 0.1 kg of magnesium stearate is pressed in a conventional manner to give tablets in such a way that each tablet contains 10 mg of active ingredient.

Example F: Dragees

Tablets are pressed analogously to Example E and subsequently coated in a conventional manner with a coating of sucrose, potato starch, talc, traga- canth and dye.

Example G: Capsules

2 kg of active ingredient of the formula I are introduced into hard gelatine capsules in a conventional manner in such a way that each capsule contains 20 mg of the active ingredient.

Example H: Ampoules

A solution of 1 kg of active ingredient of the formula I in 60 I of bidistilled water is sterile filtered, transferred into ampoules, lyophilised under sterile conditions and sealed under sterile conditions. Each ampoule contains 10 mg of active ingredient.