Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
INHIBITORS OF GLUTAMINYL CYCLASE
Document Type and Number:
WIPO Patent Application WO/2019/063414
Kind Code:
A1
Abstract:
The invention relates to a compound of formula (I): Formula (I) or a pharmaceutically acceptable salt, solvate or polymorph thereof, including all tautomers and stereoisomers thereof, wherein A is heteraryl selected from 1 H-benzimidazolyl and imidazo[1,2-a]pyridine, and R1, R2, R3, R4 and R5 are as defined herein, as inhibitors of glutaminyl cyclase (QC, EC 2.3.2.5) and its isoenzyme glutaminyl-peptide cyclotransferase-like protein (QPCTL). QC and QPCTL catalyze the intramolecular cyclization of N-terminal glutamine residues into pyroglutamic acid (5-oxo-prolyl, pGlu*) under liberation of ammonia and the intramolecular cyclization of N-terminal glutamate residues into pyroglutamic acid under liberation of water.

Inventors:
HEISER ULRICH (DE)
SOMMER ROBERT (DE)
Application Number:
PCT/EP2018/075494
Publication Date:
April 04, 2019
Filing Date:
September 20, 2018
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
PROBIODRUG AG (DE)
International Classes:
C07D403/04; A61K31/4184; A61P25/28; C07D471/04
Domestic Patent References:
WO2011029920A12011-03-17
Other References:
RYUJI ET AL. NIWA: "Studies on Ketene and Its Derivatives. CXXII. Reaction of Haloketenes with 1,3-Diaza-1,3-diene Compounds", CHEMICAL & PHARMACEUTICAL BULLETIN, vol. 32, no. 10, 1 January 1984 (1984-01-01), pages 4149 - 4153, XP055432389
Attorney, Agent or Firm:
MAIKOWSKI & NINNEMANN PATENTANWÄLTE PARTNERSCHAFT MBB (DE)
Download PDF:
Claims:
Claims

1 . A compound of formula (I):

or a pharmaceutically acceptable salt, solvate or polymorph thereof, including all tautomers and stereoisomers thereof, wherein:

A is heteroaryl selected from 1 H-benzoimidazolyl and imidazo[1 ,2-a]pyridine;

R1 represents hydrogen, alkyl or halogen;

R2 represents hydrogen, alkyl or halogen;

R3 represents hydrogen, alkyl or alkoxy;

R4 represents hydrogen or alkyl; and

R5 represents hydrogen, alkyl or halogen; and wherein the above alkyl or alkoxy groups are optionally substituted by one or more halogen.

2. The compound of formula (I) according to claim 1 , wherein

R1 represents hydrogen or halogen;

R2 represents hydrogen or halogen;

R3 represents hydrogen or alkoxy;

R4 represents hydrogen; and

R5 represents hydrogen or halogen; and

wherein the above alkoxy group is optionally substituted by one or more halogen

The compound of formula (I) according to claim 1 or 2, wherein A is 1 H- benzoimidazolyl.

The compound of formula (I) according to any one of claims 1 to 3, wherein R1 is hydrogen.

The compound of formula (I) according to any one of claims 1 to 3, wherein R1 is halogen, such as fluorine.

The compound of formula (I) according to any one of the preceding claims, wherein R2 is hydrogen.

The compound of formula (I) according to any one of claims 1 to 5, wherein R2 is halogen, such as fluorine.

The compound of formula (I) according to any one of the preceding claims, wherein R3 represents -0-Ci-4alkyl, optionally substituted by one or more halogen, such as fluorine.

The compound of formula (I) according to any one of the preceding claims, wherein R3 represents methoxy, difluoropropoxy or difluorobutoxy.

The compound of formula (I) according to any one of the preceding claims, wherein R3 represents 2,2-difluoropropoxy or 3,3-difluoropropoxy.

The compound of formula (I) according to any one of the preceding claims, wherein R5 is hydrogen.

The compound of formula (I) according to any one of claims 1 to 1 0, wherein R5 is halogen, such as fluorine.

The compound of formula (I) according to any one of the preceding claims or a pharmaceutically acceptable salt, solvate or polymorph thereof, including all

tautomers and stereoisomers, wherein the compound of formula (I) is a compound selected from the group consisting of

1 1 -(1 H-benzo[d]imidazol-5-yl)-4-phenylazetidin-2-one;

2 (R)-1 -(1 H-benzo[d]imidazol-6-yl)-4-phenylazetidin-2-one;

3 (S)-1 -(1 H-benzo[d]imidazol-6-yl)-4-phenylazetidin-2-one;

4 1 -(1 H-benzo[d]imidazol-5-yl)-4-(2,6-difluoro-4-methoxyphenyl)azetidin-2-one; (R)-4 4-(3,3-difluoropropoxy 2-fluorophenyl)-1 -(1 H-benzo[d]imidazol-5- yl)aze idin-2-one;

(S)-4 4-(3,3-difluoropropoxy 2-fluorophenyl)-1 -(1 H-benzo[d]imidazol-5- yl)aze idin-2-one;

(R)-4 4-(3,3-difluoropropoxy -2,3-difluorophenyl)-1 -(1 H-benzo[d]imidazol-5- yl)aze idin-2-one;

(S)-4 4-(3,3-difluoropropoxy 2,3-difluorophenyl)-1 -(1 H-benzo[d]imidazol-5- yl)aze idin-2-one;

(R)-4 4-(3,3-difluoropropoxy -2,6-difluorophenyl)-1 -(1 H-benzo[d]imidazol-5- yl)aze idin-2-one;

(S)-4 4-(3,3-difluoropropoxy 2,6-difluorophenyl)-1 -(1 H-benzo[d]imidazol-5- yl)aze idin-2-one;

(R)-4 4-(2,2-difluoropropoxy 2-fluorophenyl)-1 -(1 H-benzo[d]imidazol-5- yl)aze idin-2-one;

(S)-4 4-(2,2-difluoropropoxy 2-fluorophenyl)-1 -(1 H-benzo[d]imidazol-5- yl)aze idin-2-one;

(R)-4 4-(2,2-difluoropropoxy -2,3-difluorophenyl)-1 -(1 H-benzo[d]imidazol-5- yl)aze idin-2-one;

(S)-4 4-(2,2-difluoropropoxy 2,3-difluorophenyl)-1 -(1 H-benzo[d]imidazol-5- yl)aze idin-2-one;

(R)-4 4-(2,2-difluoropropoxy -2,6-difluorophenyl)-1 -(1 H-benzo[d]imidazol-5- yl)aze idin-2-one;

(S)-4 4-(2,2-difluoropropoxy 2,6-difluorophenyl)-1 -(1 H-benzo[d]imidazol-5- yl)aze idin-2-one;

(R)-1 H-imidazo[1 ,2-a]pyridin-7-yl)-4-phenylazetidin-2-one;

(S)-1 H-imidazo[1 ,2-a]pyridin-7-yl)-4-phenylazetidin-2-one;

4-(2,6 difluoro-4-methoxyphenyl)-1 -(H-imidazo[1 ,2-a]pyridin-7-yl)azetidin-2- one;

(R)-4- 4-(3,3-difluoropropoxy)-2-fluorophenyl)-1 -(H-imidazo[1 ,2-a]pyridin-7- yl)aze idin-2-one;

(S)-4 4-(3,3-difluoropropoxy)-2-fluorophenyl)-1 -(H-imidazo[1 ,2-a]pyridin-7- yl)aze idin-2-one;

(R)-4 4-(3,3-difluoropropoxy)-2,3-difluorophenyl)-1 -(H-imidazo[1 ,2-a]pyridin-

7-yl)azetidin-2-one; (S)-4-(4-(3,3-difluoropropoxy)-2,3-difluorophenyl)-1 -(H-imidazo[1 ,2-a]pyridin-

23

7-yl)azetidin-2-one;

(R)-4-(4-(3,3-difluoropropoxy)-2,6-difluorophenyl)-1 -(H-imidazo[1 ,2-a]pyridin-

24

7-yl)azetidin-2-one;

(S)-4-(4-(3,3-difluoropropoxy)-2,6-difluorophenyl)-1 -(H-imidazo[1 ,2-a]pyridin-

25

7-yl)azetidin-2-one;

(R)-4-(4-(2,2-difluoropropoxy)-2-fluorophenyl)-1 -(H-imidazo[1 ,2-a]pyridin-7-

26

yl)azetidin-2-one;

(S)-4-(4-(2,2-difluoropropoxy)-2-fluorophenyl)-1 -(H-imidazo[1 ,2-a]pyridin-7-

27

yl)azetidin-2-one;

(R)-4-(4-(2,2-difluoropropoxy)-2,3-difluorophenyl)-1 -(H-imidazo[1 ,2-a]pyridin-

28

7-yl)azetidin-2-one; and

(S)-4-(4-(2,2-difluoropropoxy)-2,3-difluorophenyl)-1 -(H-imidazo[1 ,2-a]pyridin-

29

7-yl)azetidin-2-one.

A pharmaceutical composition comprising a compound according to any one of claims 1 to 13 optionally in combination with one or more therapeutically acceptable diluents or carriers.

The pharmaceutical composition of claim 14, which comprises additionally at least one compound, selected from the group consisting of neuroprotectants,

antiparkinsonian drugs, amyloid protein deposition inhibitors, beta amyloid synthesis inhibitors, antidepressants, anxiolytic drugs, antipsychotic drugs and anti-multiple sclerosis drugs.

16. The pharmaceutical composition of claim 14 or 15, which comprises additionally at least one compound, selected from the group consisting of PEP-inhibitors, LiCI, inhibitors of inhibitors of DP IV or DP IV-like enzymes, acetylcholinesterase (ACE) inhibitors, PIMT enhancers, inhibitors of beta secretases, inhibitors of gamma secretases, inhibitors of neutral endopeptidase, inhibitors of Phosphodiesterase-4 (PDE-4), TNFalpha inhibitors, muscarinic M1 receptor antagonists, NMDA receptor antagonists, sigma-1 receptor inhibitors, histamine H3 antagonists,

immunomodulatory agents, immunosuppressive agents or an agent selected from the group consisting of antegren (natalizumab), Neurelan (fampridine-SR), campath

(alemtuzumab), IR 208, NBI 5788/MSP 771 (tiplimotide), paclitaxel, Anergix.MS (AG 284), SH636, Differin (CD 271 , adapalene), BAY 361677 (interleukin-4), matrix- metalloproteinase-inhibitors, interferon-tau (trophoblastin) and SAIK-MS.

7. The compound according to any one of claims 1 to 13 or the pharmaceutical

composition according to any one of claims 14 to 16 for use in the treatment of a disease selected from the group consisting of Kennedy's disease, duodenal cancer with or without Helicobacter pylori infections, colorectal cancer, Zolliger-Ellison syndrome, gastric cancer with or without Helicobacter pylori infections, pathogenic psychotic conditions, schizophrenia, infertility, neoplasia, inflammatory host responses, cancer, malign metastasis, melanoma, psoriasis, impaired humoral and cell-mediated immune responses, leukocyte adhesion and migration processes in the endothelium, impaired food intake, impaired sleep-wakefulness, impaired homeostatic regulation of energy metabolism, impaired autonomic function, impaired hormonal balance or impaired regulation of body fluids, multiple sclerosis, the Guillain-Barre syndrome, chronic inflammatory demyelinizing polyradiculoneuropathy, mild cognitive impairment, Alzheimer's disease, Familial British Dementia, Familial Danish

Dementia, neurodegeneration in Down Syndrome, Huntington's disease, rheumatoid arthritis, atherosclerosis, pancreatitis and restenosis.

8. A process for the preparation of a compound of formula (I) according to any one of claims 1 to 13, which comprises: preparing a compound of formula (I) from a compound of formula (II)

wherein R1 and R5 and are as defined herein for compounds of formula (I), by reacting a compound of formula (II) with brom-acetic acid in the presence of a suitable catalyst such as zinc-dust, and a protecting agent, such as trimethylsilyl chloride (TMS-CI). or preparing a compound of formula (I) from a compound of formula (III)

(III)

wherein R1 , R2, R3, R4 and R5 and are as defined herein for compounds of formula (I), by reacting a compound of formula (III) with diethylazodicarboxylate in the presence of triphenylhosphine and a suitable solvent such as tetrahydrofuran, and a deptrotection step.

Description:
INHIBITORS OF GLUTAMINYL CYCLASE

Field of the invention

The invention relates to novel azetidinone derivatives with improved pharmacokinetic properties as inhibitors of glutaminyl cyclase (QC, EC 2.3.2.5). QC catalyzes the intramolecular cyclization of N-terminal glutamine residues into pyroglutamic acid (5-oxo-prolyl, pGlu * ) under liberation of ammonia and the intramolecular cyclization of N-terminal glutamate residues into pyroglutamic acid under liberation of water. Background of the invention

Glutaminyl cyclase (QC, EC 2.3.2.5) catalyzes the intramolecular cyclization of N-terminal glutamine residues into pyroglutamic acid (pGlu * ) liberating ammonia. A QC was first isolated by Messer from the latex of the tropical plant Carica papaya in 1963 (Messer, M. 1963 Nature 4874, 1299). 24 years later, a corresponding enzymatic activity was discovered in animal pituitary (Busby, W. H. J. et al. 1987 J Biol Chem 262, 8532-8536; Fischer, W. H. and Spiess, J. 1987 Proc Natl Acad Sci U S A 84, 3628-3632). For the mammalian QC, the conversion of Gin into pGlu by QC could be shown for the precursors of TRH and GnRH (Busby, W. H. J. et al. 1987 J Biol Chem 262, 8532-8536; Fischer, W. H. and Spiess, J. 1987 Proc Natl Acad Sci U S A 84, 3628-3632). In addition, initial localization experiments of QC revealed a co- localization with its putative products of catalysis in bovine pituitary, further improving the suggested function in peptide hormone synthesis (Bockers, T. M. et al. 1995 J Neuroendocrinol 7, 445-453). In contrast, the physiological function of the plant QC is less clear. In the case of the enzyme from C. papaya, a role in the plant defense against pathogenic microorganisms was suggested (El Moussaoui, A. et al.2001 Cell Mol Life Sci 58, 556-570). Putative QCs from other plants were identified by sequence comparisons recently (Dahl, S. W. et al.2000 Protein Expr Purif 20, 27-36). The physiological function of these enzymes, however, is still ambiguous.

The QCs known from plants and animals show a strict specificity for L-Glutamine in the N- terminal position of the substrates and their kinetic behavior was found to obey the Michaelis- Menten equation (Pohl, T. et al. 1991 Proc Natl Acad Sci U S A 88, 10059-10063; Consalvo, A. P. et al. 1988 Anal Biochem 175, 131 -138; Gololobov, M. Y. et al. 1996 Biol Chem Hoppe Seyler 377, 395-398). A comparison of the primary structures of the QCs from C. papaya and that of the highly conserved QC from mammals, however, did not reveal any sequence homology (Dahl, S. W. et al. 2000 Protein Expr Purif 20, 27-36). Whereas the plant QCs appear to belong to a new enzyme family (Dahl, S. W. et al. 2000 Protein Expr Purif 20, 27-36), the mammalian QCs were found to have a pronounced sequence homology to bacterial aminopeptidases (Bateman, R. C. et al. 2001 Biochemistry 40, 1 1246-1 1250), leading to the conclusion that the QCs from plants and animals have different evolutionary origins.

Recently, it was shown that recombinant human QC as well as QC-activity from brain extracts catalyze both, the N-terminal glutaminyl as well as glutamate cyclization. Most striking is the finding, that cyclase-catalyzed Glurconversion is favored around pH 6.0 while Gln conversion to pGlu-derivatives occurs with a pH-optimum of around 8.0. Since the formation of pGlu-Αβ- related peptides can be suppressed by inhibition of recombinant human QC and QC-activity from pig pituitary extracts, the enzyme QC is a target in drug development for treatment of Alzheimer's disease.

Inhibitors of QC are described in WO 2004/098625, WO 2004/098591 , WO 2005/039548, WO 2005/075436, WO 2008/055945, WO 2008/055947, WO 2008/055950, WO2008/065141 , WO 2008/1 10523, WO 2008/128981 , WO 2008/128982, WO 2008/128983, WO 2008/128984, WO 2008/128985, WO 2008/128986, WO 2008/128987, WO 2010/026212, WO 201 1/029920, WO 201 1/107530, WO 201 1 /1 10613, WO 201 1 /131748 and WO 2012/123563 and WO 2014/140279.

EP 02 01 1 349.4 discloses polynucleotides encoding insect glutaminyl cyclase, as well as polypeptides encoded thereby and their use in methods of screening for agents that reduce glutaminyl cyclase activity. Such agents are useful as pesticides. Definitions

The terms "k" or "K " and "KD" are binding constants, which describe the binding of an inhibitor to and the subsequent release from an enzyme. Another measure is the "IC 5 o" value, which reflects the inhibitor concentration, which at a given substrate concentration results in 50 % enzyme activity.

The term "DP I V-inhibitor" or "dipeptidyl peptidase IV inhibitor" is generally known to a person skilled in the art and means enzyme inhibitors, which inhibit the catalytic activity of DP IV or DP IV-like enzymes. "DP IV-activity" is defined as the catalytic activity of dipeptidyl peptidase IV (DP IV) and DP IV- like enzymes. These enzymes are post-proline (to a lesser extent post-alanine, post-serine or post-glycine) cleaving serine proteases found in various tissues of the body of a mammal including kidney, liver, and intestine, where they remove dipeptides from the N-terminus of biologically active peptides with a high specificity when proline or alanine form the residues that are adjacent to the N-terminal amino acid in their sequence.

The term "PEP-inhibitor" or "prolyl endopeptidase inhibitor" is generally known to a person skilled in the art and means enzyme inhibitors, which inhibit the catalytic activity of prolyl endopeptidase (PEP, prolyl oligopeptidase, POP).

"PEP-activity" is defined as the catalytic activity of an endoprotease that is capable to hydrolyze post proline bonds in peptides or proteins where the proline is in amino acid position 3 or higher counted from the N-terminus of a peptide or protein substrate.

The term "QC" as used herein comprises glutaminyl cyclase (QC) and QC-like enzymes. QC and QC-like enzymes have identical or similar enzymatic activity, further defined as QC activity. In this regard, QC-like enzymes can fundamentally differ in their molecular structure from QC. Examples of QC-like enzymes are the glutaminyl-peptide cyclotransferase-like proteins (QPCTLs) from human (GenBank NM_017659), mouse (GenBank BC058181 ), Macaca fascicularis (GenBank AB168255), Macaca mulatta (GenBank XM_001 1 10995), Canis familiaris (GenBank XM_541552), Rattus norvegicus (GenBank XM_001066591 ), Mus musculus (GenBank BC058181 ) and Bos taurus (GenBank BT026254). The term "QC activity" as used herein is defined as intramolecular cyclization of N-terminal glutamine residues into pyroglutamic acid (pGlu * ) or of N-terminal L-homoglutamine or L-β- homoglutamine to a cyclic pyro-homoglutamine derivative under liberation of ammonia. See therefore schemes 1 and 2. Scheme 1 : Cyclization of glutamine by QC

Scheme 2: Cyclization of L-homoglutamine by QC

The term "EC" as used herein comprises the activity of QC and QC-like enzymes as glutamate cyclase (EC), further defined as EC activity.

The term "EC activity" as used herein is defined as intramolecular cyclization of N-terminal glutamate residues into pyroglutamic acid (pGlu * ) by QC. See therefore scheme 3. Scheme 3: N-terminal cyclization of uncharged glutamyl peptides by QC (EC)

The term "QC-inhibitor" "glutaminyl cyclase inhibitor" is generally known to a person skilled in the art and means enzyme inhibitors, which inhibit the catalytic activity of glutaminyl cyclase (QC) or its glutamyl cyclase (EC) activity. Potency of QC inhibition

In light of the correlation with QC inhibition, in preferred embodiments, the subject method and medical use utilize an agent with an IC 5 o for QC inhibition of 10 μΜ or less, more preferably of 1 μΜ or less, even more preferably of 0.1 μΜ or less or 0.01 μΜ or less, or most preferably 0.001 μΜ or less. Indeed, inhibitors with K, values in the lower micromolar, preferably the nanomolar and even more preferably the picomolar range are contemplated. Thus, while the active agents are described herein, for convenience, as "QC inhibitors", it will be understood that such nomenclature is not intending to limit the subject of the invention to a particular mechanism of action. Molecular weight of QC inhibitors

In general, the QC inhibitors of the subject method or medical use will be small molecules, e.g., with molecular weights of 500 g/mole or less, 400 g/mole or less, preferably of 350 g/mole or less, and even more preferably of 300 g/mole or less and even of 250 g/mole or less. The term "subject" as used herein, refers to an animal, preferably a mammal, most preferably a human, who has been the object of treatment, observation or experiment.

The term "therapeutically effective amount" as used herein, means that amount of active compound or pharmaceutical agent that elicits the biological or medicinal response in a tissue system, animal or human being sought by a researcher, veterinarian, medical doctor or other clinician, which includes alleviation of the symptoms of the disease or disorder being treated.

As used herein, the term "pharmaceutically acceptable" embraces both human and veterinary use: For example the term "pharmaceutically acceptable" embraces a veterinarily acceptable compound or a compound acceptable in human medicine and health care.

Throughout the description and the claims the expression "alkyl", unless specifically limited, denotes a CM 2 alkyl group, suitably a Ci- 8 alkyl group, e.g. Ci- 6 alkyl group, e.g. C1-4 alkyl group. Alkyl groups may be straight chain or branched. Suitable alkyl groups include, for example, methyl, ethyl, propyl (e.g. n-propyl and isopropyl), butyl (e.g n-butyl, iso-butyl, sec-butyl and tert-butyl), pentyl (e.g. n-pentyl), hexyl (e.g. n-hexyl), heptyl (e.g. n-heptyl) and octyl (e.g. n- octyl). The expression "alk", for example in the expressions "alkoxy", "haloalkyl" and "thioalkyl" should be interpreted in accordance with the definition of "alkyl". Exemplary alkoxy groups include methoxy, ethoxy, propoxy (e.g. n-propoxy), butoxy (e.g. n-butoxy), pentoxy (e.g. n- pentoxy), hexoxy (e.g. n-hexoxy), heptoxy (e.g. n-heptoxy) and octoxy (e.g. n-octoxy). Exemplary thioalkyl groups include methylthio-. Exemplary haloalkyl groups include fluoroalkyl e.g. CF 3 , fluoroethyl, fluropropyl, fluorobutyl, difluoroethyl, difluoropropyl and difluorobutyl.

The term "halogen" or "halo" comprises fluorine (F), chlorine (CI) and bromine (Br).

When benzimidazolyl is shown as benzimidazol-5-yl, which is represented as:

the person skilled in the art will appreciate that benzimidazol-6-yl, which is represented as:

is an equivalent structure. As employed herein, the two forms of benzimidazolyl are covered by the term "benzimidazol-5-yl".

Stereoisomers:

All possible stereoisomers of the claimed compounds are included in the present invention.

Where the compounds according to this invention have at least one chiral center, they may accordingly exist as enantiomers. Where the compounds possess two or more chiral centers, they may additionally exist as diastereomers. It is to be understood that all such isomers and mixtures thereof are encompassed within the scope of the present invention.

Preparation and isolation of stereoisomers: Where the processes for the preparation of the compounds according to the invention give rise to a mixture of stereoisomers, these isomers may be separated by conventional techniques such as preparative chromatography. The compounds may be prepared in racemic form, or individual enantiomers may be prepared either by enantiospecific synthesis or by resolution. The compounds may, for example, be resolved into their components enantiomers by standard techniques, such as the formation of diastereomeric pairs by salt formation with an optically active acid, such as (-)-di-p-toluoyl-d-tartaric acid and/or (+)-di-p-toluoyl-l-tartaric acid followed by fractional crystallization and regeneration of the free base. The compounds may also be resolved by formation of diastereomeric esters or amides, followed by chromatographic separation and removal of the chiral auxiliary. Alternatively, the compounds may be resolved using a chiral HPLC column.

Pharmaceutically acceptable salts:

In view of the close relationship between the free compounds and the compounds in the form of their salts or solvates, whenever a compound is referred to in this context, a corresponding salt, solvate or polymorph is also intended, provided such is possible or appropriate under the circumstances.

Salts and solvates of the compounds of formula (I) and physiologically functional derivatives thereof which are suitable for use in medicine are those wherein the counter-ion or associated solvent is pharmaceutically acceptable. However, salts and solvates having non- pharmaceutically acceptable counter-ions or associated solvents are within the scope of the present invention, for example, for use as intermediates in the preparation of other compounds and their pharmaceutically acceptable salts and solvates.

Suitable salts according to the invention include those formed with both organic and inorganic acids or bases. Pharmaceutically acceptable acid addition salts include those formed from hydrochloric, hydrobromic, sulfuric, nitric, citric, tartaric, phosphoric, lactic, pyruvic, acetic, trifluoroacetic, triphenylacetic, sulfamic, sulfanilic, succinic, oxalic, fumaric, maleic, malic, mandelic, glutamic, aspartic, oxaloacetic, methanesulfonic, ethanesulfonic, arylsulfonic (for example p-toluenesulfonic, benzenesulfonic, naphthalenesulfonic or naphthalenedisulfonic), salicylic, glutaric, gluconic, tricarballylic, cinnamic, substituted cinnamic (for example, phenyl, methyl, methoxy or halo substituted cinnamic, including 4-methyl and 4-methoxycinnamic acid), ascorbic, oleic, naphthoic, hydroxynaphthoic (for example 1 - or 3-hydroxy-2-naphthoic), naphthaleneacrylic (for example naphthalene-2-acrylic), benzoic, 4-methoxybenzoic, 2- or 4- hydroxybenzoic, 4-chlorobenzoic, 4-phenylbenzoic, benzeneacrylic (for example 1 ,4- benzenediacrylic), isethionic acids, perchloric, propionic, glycolic, hydroxyethanesulfonic, pamoic, cyclohexanesulfamic, salicylic, saccharinic and trifluoroacetic acid. Pharmaceutically acceptable base salts include ammonium salts, alkali metal salts such as those of sodium and potassium, alkaline earth metal salts such as those of calcium and magnesium and salts with organic bases such as dicyclohexylamine and /V-methyl-D-glucamine.

All pharmaceutically acceptable acid addition salt forms of the compounds of the present invention are intended to be embraced by the scope of this invention.

Polymorph crystal forms:

Furthermore, some of the crystalline forms of the compounds may exist as polymorphs and as such are intended to be included in the present invention. In addition, some of the compounds may form solvates with water (i.e. hydrates) or common organic solvents, and such solvates are also intended to be encompassed within the scope of this invention. The compounds, including their salts, can also be obtained in the form of their hydrates, or include other solvents used for their crystallization.

Prodrugs:

The present invention further includes within its scope prodrugs of the compounds of this invention. In general, such prodrugs will be functional derivatives of the compounds which are readily convertible in vivo into the desired therapeutically active compound. Thus, in these cases, the methods of treatment of the present invention, the term "administering" shall encompass the treatment of the various disorders described with prodrug versions of one or more of the claimed compounds, but which converts to the above specified compound in vivo after administration to the subject. Conventional procedures for the selection and preparation of suitable prodrug derivatives are described, for example, in "Design of Prodrugs", ed. H. Bundgaard, Elsevier, 1985.

Protective Groups:

During any of the processes for preparation of the compounds of the present invention, it may be necessary and/or desirable to protect sensitive or reactive groups on any of the molecules concerned. This may be achieved by means of conventional protecting groups, such as those described in Protective Groups in Organic Chemistry, ed. J.F.W. McOmie, Plenum Press, 1973; and T.W. Greene & P.G.M. Wuts, Protective Groups in Organic Synthesis, John Wiley & Sons, 1991 , fully incorporated herein by reference. The protecting groups may be removed at a convenient subsequent stage using methods known from the art.

A protecting group or protective group is introduced into a molecule by chemical modification of a functional group in order to obtain chemoselectivity in a subsequent chemical reaction. Protecting groups are e.g. alcohol protecting groups, amine protecting groups, carbonyl protecting groups, carboxylic acid protecting groups and phosphate protecting groups.

Examples for alcohol protecting groups are acetyl (Ac), benzoyl (Bz), benzyl (Bn, Bnl) β- methoxyethoxymethyl ether (MEM), mimethoxytrityl [bis-(4-methoxyphenyl)phenylmethyl, DMT], methoxymethyl ether (MOM), methoxytrityl [(4-methoxyphenyl)diphenylmethyl, MMT), p-methoxybenzyl ether (PMB), methylthiomethyl ether, pivaloyl (Piv), tetrahydropyranyl (THP), trityl (triphenylmethyl, Tr), silyl ethers (such as trimethylsilyl ether (TMS), te/t-butyldimethylsilyl ether (TBDMS), te/t-butyldimethylsilyloxymethyl ether (TOM), and triisopropylsilyl ether (TIPS)); methyl ethers and ethoxyethyl ethers (EE).

Suitable amine protecting groups are selected from carbobenzyloxy (Cbz), p-methoxybenzyl carbonyl (Moz or MeOZ), terf-butyloxycarbonyl (BOC), 9-fluorenylmethyloxycarbonyl (FMOC), acetyl (Ac), benzoyl (Bz), benzyl (Bn), p-methoxybenzyl (PMB), 3,4-dimethoxybenzyl (DMPM), p-methoxyphenyl (PMP), tosyl (Ts), and other sulfonamides (Nosyl & Nps).

Suitable carbonyl protecting groups are selected from acetals and ketals, acylals and dithianes. Suitable carboxylic acid protecting groups are selected from methyl esters, benzyl esters, tert- butyl esters, silyl esters, orthoesters, and oxazoline.

Examples for phosphate protecting groups are 2-cyanoethyl and methyl (Me)

As used herein, the term "composition" is intended to encompass a product comprising the claimed compounds in the therapeutically effective amounts, as well as any product which results, directly or indirectly, from combinations of the claimed compounds.

Carriers and Additives for galenic formulations:

Thus, for liquid oral preparations, such as for example, suspensions, elixirs and solutions, suitable carriers and additives may advantageously include water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents and the like; for solid oral preparations such as, for example, powders, capsules, gelcaps and tablets, suitable carriers and additives include starches, sugars, diluents, granulating agents, lubricants, binders, disintegrating agents and the like.

Carriers, which can be added to the mixture, include necessary and inert pharmaceutical excipients, including, but not limited to, suitable binders, suspending agents, lubricants, flavorants, sweeteners, preservatives, coatings, disintegrating agents, dyes and coloring agents.

Soluble polymers as targetable drug carriers can include polyvinylpyrrolidone, pyran copolymer, polyhydroxypropylmethacrylamidephenol, polyhydroxyethylaspartamide-phenol, or polyethyleneoxidepolyllysine substituted with palmitoyl residue. Furthermore, the compounds of the present invention may be coupled to a class of biodegradable polymers useful in achieving controlled release of a drug, for example, polyactic acid, polyepsilon caprolactone, polyhydroxy butyeric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates and cross-linked or amphipathic block copolymers of hydrogels.

Suitable binders include, without limitation, starch, gelatin, natural sugars such as glucose or betalactose, corn sweeteners, natural and synthetic gums such as acacia, tragacanth or sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and the like.

Disintegrators include, without limitation, starch, methyl cellulose, agar, bentonite, xanthan gum and the like.

Summary of the invention

Inhibitors of glutaminyl cyclase are known in the art. WO 201 1/029920 and WO 2014/140279 disclose inter alia inhibitors of glutaminyl cyclase which comprise a oxazolidinone moiety. However, for use in medicine, i.e. the prevention and therapy of diseases, there is a need for further compounds, which have improved pharmacokinetic properties in order to reduce dosing levels and thereby reducing unwanted side effects and preventing adverse events after administration to a subject. In particular, for the treatment or prevention of diseases of the central nervous system (CNS), for example neurodegenerative diseases such as Mild Cognitive Impairment, Alzheimer's disease, neurodegeneration in Downs Syndrome or Familial Alzheimer's Diseases, there is a need for new compounds, which show increased levels and increased half lifes in the CNS, e.g. in the brain and CSF.

Thus, it was the problem of the present invention to provide new compounds with improved pharmacokinetic properties, in particular for the treatment of CNS related diseases.

This problem was solved by the present invention by provision of compounds of formula (I).

According to the invention there is provided a compound of formula (I):

a (I) or a pharmaceutically acceptable salt, solvate or polymorph thereof, including all tautomers and stereoisomers thereof, wherein:

A is heteroaryl selected from 1 H-benzimidazolyl and imidazo[1 ,2-a]pyridine;

R 1 represents hydrogen, alkyl or halogen;

R 2 represents hydrogen, alkyl or halogen;

R 3 represents hydrogen, alkyl or alkoxy;

R 4 represents hydrogen or alkyl; and

R 5 represents hydrogen, alkyl or halogen; and wherein the above alkyl or alkoxy groups are optionally substituted by one or more halogen. Detailed description of the invention

Surprisingly, it was found by the inventors that compounds, which comprise a phenylazetidinone residue, result in inhibitors of glutaminyl cyclase, which have multiple advantages compared to glutaminyl cyclase inhibitors existing in the prior art. These compounds are potent inhibitors of glutaminyl cyclase (QC) as well as of the isoenzyme thereof, i.e. glutaminyl-peptide cyclotransferase-like protein (QPCTL). The inhibitor constants, such as the Ki value, of the compounds are in the low nanomolar range. The potency of the compounds could be improved by halogenation, in particular fluorination of the phenyl ring. When alkyl and alkoxy are substituted, they are typically substituted by 1 or more, such as 1 , 2 3, 4 or 5 substituents. Preferably, alkyl and alkoxy are substituted by 1 or 2 substituents, most preferably by 2 substituents. Typically the substituents are both halogen. More typically, the halogen substituents are fluorine. When R 1 , R 2 , R 3 , R 4 and R 5 represent alkyl, examples include C1 -C12 straight chain or branched alkyl groups. A suitable alkyl is a d-s alkyl, more suitably a d-e alkyl, most suitably a Ci - 4 alkyl. Aforementioned alkyl groups are unsubstituted or substituted by one or more halogen substituents, typically by 1 or 2 halogen substituents. Suitably, the halogen substituents are chlorine or fluorine. Most suitably, the halogen substituents are fluorine.

When R 3 represents alkoxy, examples include -O-C1-12 straight chain or branched alkoxy groups. A suitable alkoxy is a -O-d-s alkyl, more suitably a -O-d-e alkyl, most suitably a -O- C1 -4 alkyl. Eaxmples of alkoxy include methoxy, ethoxy, propoxy and and butoxy groups. Aforementioned alkoxy groups are substituted by one or more halogen substituents, typically by 1 or 2 halogen substituents. Most suitably, the halogen substituents are fluorine. Examples of substituted alkoxy groups include dichloromethoxy, difluoromethoxy, dichloropropoxy, e.g. 2,2-dichloropropoxy and 3,3-dichloropropoxy, difluoropropo, e.g. 2,2-difluoropropoxy and 3,3- difluropropoxy, dichlorobutoxy and difluorobutoxy. Particularly advantageous compounds according to the present invention have been obtained, when both are fluorinated: (i) the phenyl ring, wherein at least one of R 1 , R 2 , and R 5 is fluorine, and (ii) the substituent at position R 3 .

In one particular embodiment of the invention, there is provided a compound of formula (I):

or a pharmaceutically acceptable salt, solvate or polymorph thereof, including all tautomers and stereoisomers thereof, wherein:

R 1 represents hydrogen or halogen; R 2 represents hydrogen or halogen; R 3 represents hydrogen or alkoxy; R 4 represents hydrogen; and

R 5 represents hydrogen or halogen; and wherein the above alkoxy group is optionally substituted by one or more halogen.

In a preferred embodiment, A is 1 H-benzoimidazolyl, especially 1 H-benzimidazol-5-yl or 1 H-benzimidazol-6-yl, and the compound of formula (I) is a compound of formula (la):

wherein R 1 , R 2 , R 3 , R 4 and R 5 are as defiend herein.

In a preferred embodiment, A is imidazo[1 ,2-a]pyridine and the compound of formula (I) is a compound of formula (lb):

wherein R 1 , R 2 , R 3 , R 4 and R 5 are as defiend herein.

When R 1 represents halogen, halogen is preferably chlorine or fluorine.

In a preferred embodiment, R 1 is hydrogen.

In another preferred embodiment, R 1 is halogen, most preferably fluorine.

When R 2 represents halogen, halogen is preferably chlorine or fluorine.

In a preferred embodiment, R 2 is hydrogen.

In another preferred embodiment, R 2 is halogen, most preferably fluorine.

More suitably, R 3 represents -0-Ci-4alkyl, substituted by one or more halogen, such as fluorine.

Preferably, R 3 represents methoxy, difluoropropoxy or difluorobutoxy. More preferably, R 3 represents methoxy or difluoropropoxy.

In a most preferred embodiment, R 3 is hydrogen.

In another most preferred embodiment, R 3 is methoxy.

In another most preferred embodiment, R 3 represents 2,2-difluoropropoxy or 3,3- difluoropropoxy.

R 4 is preferably hydrogen.

When R 5 represents halogen, halogen is preferably chlorine or fluorine.

In a preferred embodiment, R 5 is hydrogen.

In another preferred embodiment, R 5 is halogen, most preferably fluorine.

In one embodiment, R 1 , R 2 , R 3 , R 4 and R 5 are hydrogen.

Especially preferred according to the present invention are compounds of formula (I), wherein R 3 is alkoxy as described herein.

Further preferred according to the present invention are compounds, in which the phenyl ri in the compound of formula (I) is substituted by at least one halogen, i.e. at least one of R 1 R 2 , R 4 and R 5 is halogen. In one embodiment, R 1 and R 5 are halogen and R 2 and R 4 are hydrogen.

In a further embodiment, R 1 is halogen and R 2 , R 4 and R 5 are hydrogen.

In a further embodiment, R 1 and R 2 are halogen and R 4 and R 5 are hydrogen.

Preferred according to the present invention are compounds of formula (I), wherein at least one of R 1 , R 2 , R 4 and R 5 is fluorine.

More preferably, one of R 1 , R 2 , R 4 and R 5 is fluorine.

Further more preferably, two of R 1 , R 2 , R 4 and R 5 are fluorine. Most preferably, R 1 is fluorine and R 2 , R 4 and R 5 are hydrogen; or

R 1 and R 5 are fluorine and R 2 and R 4 are hydrogen; or

R 1 and R 2 are fluorine and R 4 and R 5 are hydrogen.

Preferred compounds of formula (I) are characterized by the following embodiments:

R 1 is fluorine;

R 2 is hydrogen;

R 3 is methoxy, 2,2-difluoropropoxy or 3,3-difluoropropoxy;

R 4 is hydrogen; and

R 5 is fluorine; or

R 1 is fluorine,

R 2 is hydrogen,

R 3 is 2,2-difluoropropoxy or 3,3-difluoropropoxy;

R 4 is hydrogen; and

R 5 is hydrogen; or

R 1 is fluorine,

R 2 is fluorine,

R 3 is 2,2-difluoropropoxy or 3,3-difluoropropoxy;

R 4 is hydrogen; and

R 5 is hydrogen; or

R 1 is fluorine,

R 2 is hydrogen,

R 3 is 2,2-difluoropropoxy or 3,3-difluoropropoxy;

R 4 is hydrogen; and R 5 is fluorine; Processes

According to a further aspect of the invention there is provided a process for preparing a compound of formula (I) which comprises:

(a) preparing a compound of formula (I) from a compound of formula (II):

wherein

R 1 and R 5 and are as defined herein for compounds of formula (I).

The process typically involves reacting a compound of formula (II) with brom-acetic acid in the presence of a suitable catalyst such as zinc-dust, and a protecting agent, such as trimethylsilyl chloride (TMS-CI). A non-limiting example of the methodology of process (a) is described in Method 1 herein.

(b) preparing a compound of formula (I) from a compound of formula (III):

(III)

wherein

R 1 , R 2 , R 3 , R 4 and R 5 and are as defined herein for compounds of formula (I). Process (b) typically involves reacting a compound of formula (III) with diethylazodicarboxylate in the presence of triphenylhosphine and a suitable solvent such as tetrahydrofuran, and a deptrotection step. A non-limiting example of the methodology of process (b) is described in Method 2 herein. Compounds of formula (I) and intermediate compounds may also be prepared using techniques analogous to those known to a skilled person, or described herein.

Novel intermediates are claimed as an aspect of the present invention. Therapeutic uses

Physiological substrates of QC (EC) in mammals are, e.g. amyloid beta-peptides (3-40), (3- 42), (1 1 -40 and (1 1 -42), ABri, ADan, Gastrin, Neurotensin, FPP, CCL 2, CCL 7, CCL 8, CCL 16, CCL 18, Fractalkine, Orexin A, [Gln 3 ]-glucagon(3-29), [Gln 5 ]-substance P(5-1 1 ) and the peptide QYNAD. For further details see table 1 . The compounds and/or combinations according to the present invention and pharmaceutical compositions comprising at least one inhibitor of QC (EC) are useful for the treatment of conditions that can be treated by modulation of QC activity.

Table 1 : Amino acid sequences of physiological active peptides with an N-terminal glutamine residue, which are prone to be cyclized to final pGlu

Peptide Amino acid sequence Function

Abeta(1 -42) Asp-Ala-Glu-Phe-Arg-His-Asp-Ser- Plays a role in

Gly-Tyr-Glu-Val-His-His-Gln-Lys- neurodegeneration, e.g. in

Leu-Val-Phe-Phe-Ala-Glu-Asp-Val- Alzheimer's Disease, Familial

Gly-Ser-Asn-Lys-Gly-Ala-lle-lle-Gly- British Dementia, Familial

Leu-Met-Val-Gly-Gly-Val-Val-lle-Ala Danish Dementia, Down

Syndrome Peptide Amino acid sequence Function

Abeta(1 -40) Asp-Ala-Glu-Phe-Arg-His-Asp-Ser- Plays a role in

Gly-Tyr-Glu-Val-His-His-Gln-Lys- neurodegeneration, e.g. in

Leu-Val-Phe-Phe-Ala-Glu-Asp-Val- Alzheimer's Disease, Familial

Gly-Ser-Asn-Lys-Gly-Ala-lle-lle-Gly- British Dementia, Familial

Leu-Met-Val-Gly-Gly-Val-Val Danish Dementia, Down

Syndrome

Abeta(3-42) Glu-Phe-Arg-His-Asp-Ser-Gly-Tyr- Plays a role in

Glu-Val-His-His-Gln-Lys-Leu-Val- neurodegeneration, e.g. in

Phe-Phe-Ala-Glu-Asp-Val-Gly-Ser- Alzheimer's Disease, Familial

Asn-Lys-Gly-Ala-lle-lle-Gly-Leu-Met- British Dementia, Familial

Val-Gly-Gly-Val-Val-lle-Ala Danish Dementia, Down

Syndrome

Abeta(3-40) Glu-Phe-Arg-His-Asp-Ser-Gly-Tyr- Plays a role in

Glu-Val-His-His-Gln-Lys-Leu-Val- neurodegeneration, e.g. in

Phe-Phe-Ala-Glu-Asp-Val-Gly-Ser- Alzheimer's Disease, Familial

Asn-Lys-Gly-Ala-lle-lle-Gly-Leu-Met- British Dementia, Familial

Val-Gly-Gly-Val-Val Danish Dementia, Down

Syndrome

Abeta( 1 1 -42) Glu-Val-His-His-Gln-Lys-Leu-Val- Plays a role in

Phe-Phe-Ala-Glu-Asp-Val-Gly-Ser- neurodegeneration, e.g. in Asn-Lys-Gly-Ala-lle-lle-Gly-Leu-Met- Alzheimer's Disease, Familial Val-Gly-Gly-Val-Val-lle-Ala British Dementia, Familial

Danish Dementia, Down Syndrome

Abeta( 1 1 -40) Glu-Val-His-His-Gln-Lys-Leu-Val- Plays a role in

Phe-Phe-Ala-Glu-Asp-Val-Gly-Ser- neurodegeneration, e.g. in Asn-Lys-Gly-Ala-lle-lle-Gly-Leu-Met- Alzheimer's Disease, Familial Val-Gly-Gly-Val-Val British Dementia, Familial

Danish Dementia, Down Syndrome

ABri EASNCFA IRHFENKFAV ETLIC Pyroglutamated form plays a

SRTVKKNIIEEN role in Familial British Dementia Peptide Amino acid sequence Function

ADan EASNCFA IRHFENKFAV ETLIC Pyroglutamated form plays a

FNLFLNSQEKHY role in Familial Danish

Dementia

Gastrin 17 QGPWL EEEEEAYGWM DF (amide) Gastrin stimulates the stomach mucosa to produce and secrete Swiss-Prot: P01350 hydrochloric acid and the

pancreas to secrete its digestive enzymes. It also stimulates smooth muscle contraction and increases blood circulation and water secretion in the stomach and intestine.

Neurotensin QLYENKPRRP YIL Neurotensin plays an endocrine or paracrine role in the

Swiss-Prot: P30990 regulation of fat metabolism. It causes contraction of smooth muscle.

FPP QEP amide A tripeptide related to thyrotrophin releasing hormone (TRH), is found in seminal plasma. Recent evidence obtained in vitro and in vivo showed that FPP plays an important role in regulating sperm fertility.

TRH QHP amide TRH functions as a regulator of the biosynthesis of TSH in the

Swiss-Prot: P20396 anterior pituitary gland and as a neurotransmitter/

neuromodulator in the central and peripheral nervous systems. Peptide Amino acid sequence Function

GnRH QHWSYGL RP(G) amide Stimulates the secretion of gonadotropins; it stimulates the

Swiss-Prot: P01 148 secretion of both luteinizing and follicle-stimulating hormones.

CCL16 (small QPKVPEW VNTPSTCCLK Shows chemotactic activity for inducible cytokine YYEKVLPRRL VVGYRKALNC lymphocytes and monocytes A16) HLPAIIFVTK RNREVCTNPN but not neutrophils. Also shows

DDWVQEYIKD PNLPLLPTRN potent myelosuppressive

Swiss-Prot: 015467 LSTVKIITAK NGQPQLLNSQ activity, suppresses

proliferation of myeloid progenitor cells. Recombinant SCYA16 shows chemotactic activity for monocytes and THP-1 monocytes, but not for resting lymphocytes and neutrophils. Induces a calcium flux in THP-1 cells that were desensitized by prior expression to RANTES.

CCL8 (small QPDSVSI PITCCFNVIN Chemotactic factor that attracts inducible cytokine RKIPIQRLES YTRITNIQCP monocytes, lymphocytes, A8) KEAVIFKTKR GKEVCADPKE basophils and eosinophils. May

RWVRDSMKHL DQIFQNLKP play a role in neoplasia and

Swiss-Prot: P80075 inflammatory host responses.

This protein can bind heparin. Peptide Amino acid sequence Function

CCL2 (MCP-1 , small QPDAINA PVTCCYNFTN Chemotactic factor that attracts inducible cytokine A2) RKISVQRLAS YRRITSSKCP monocytes and basophils but

KEAVIFKTIV AKEICADPKQ not neutrophils or eosinophils.

Swiss-Prot: P13500 KWVQDSMDHL DKQTQTPKT Augments monocyte anti-tumor activity. Has been implicated in the pathogenesis of diseases characterized by monocytic infiltrates, like psoriasis, rheumatoid arthritis or atherosclerosis. May be involved in the recruitment of monocytes into the arterial wall during the disease process of atherosclerosis. Binds to CCR2 and CCR4.

CCL18 (small QVGTNKELC CLVYTSWQIP Chemotactic factor that attracts inducible cytokine QKFIVDYSET SPQCPKPGVI lymphocytes but not monocytes A18) LLTKRGRQIC ADPNKKWVQK or granulocytes. May be

YISDLKLNA involved in B cell migration into

Swiss-Prot: P55774 B cell follicles in lymph nodes.

Attracts naive T lymphocytes toward dendritic cells and activated macrophages in lymph nodes, has chemotactic activity for naive T cells, CD4+ and CD8+ T cells and thus may play a role in both humoral and cell-mediated immunity responses. Peptide Amino acid sequence Function

Fractal kine QHHGVT KCNITCSKMT The soluble form is chemotactic (neurotactin) SKIPVALLIH YQQNQASCGK for T cells and monocytes, but

RAIILETRQH RLFCADPKEQ not for neutrophils. The

Swiss-Prot: P78423 WVKDAMQHLD RQAAALTRNG membrane-bound form

GTFEKQIGEV KPRTTPAAGG promotes adhesion of those MDESVVLEPE ATGESSSLEP leukocytes to endothelial cells. TPSSQEAQRA LGTSPELPTG May play a role in regulating VTGSSGTRLP PTPKAQDGGP leukocyte adhesion and VGTELFRVPP VSTAATWQSS migration processes at the APHQPGPSLW AEAKTSEAPS endothelium binds to CX3CR1 . TQDPSTQAST ASSPAPEENA PSEGQRVWGQ GQSPRPENSL EREEMGPVPA HTDAFQDWGP GSMAHVSVVP VSSEGTPSRE PVASGSWTPK AEEPIHATMD PQRLGVLITP VPDAQAATRR QAVGLLAFLG LLFCLGVAMF TYQSLQGCPR KMAGEMAEGL RYIPRSCGSN SYVLVPV

CCL7 (small inducible QPVGINT STTCCYRFIN Chemotactic factor that attracts cytokine A7) KKIPKQRLES YRRTTSSHCP monocytes and eosinophils, but

REAVIFKTKL DKEICADPTQ not neutrophils. Augments

Swiss-Prot: P80098 KWVQDFMKHL DKKTQTPKL monocyte anti-tumor activity.

Also induces the release of gelatinase B. This protein can bind heparin. Binds to CCR1 , CCR2 and CCR3. Peptide Amino acid sequence Function

Orexin A (Hypocretin- QPLPDCCRQK TCSCRLYELL Neuropeptide that plays a 1 ) HGAGNHAAGI LTL significant role in the regulation of food intake and sleep-

Swiss-Prot 043612 wakefulness, possibly by

coordinating the complex behavioral and physiologic responses of these

complementary homeostatic functions. It plays also a broader role in the homeostatic regulation of energy metabolism, autonomic function, hormonal balance and the regulation of body fluids. Orexin-A binds to both 0X1 R and 0X2R with a high affinity.

Substance P RPK PQQFFGLM Belongs to the tachykinins.

Tachykinins are active peptides which excite neurons, evoke behavioral responses, are potent vasodilators and secretagogues, and contract (directly or indirectly) many smooth muscles.

QYNAD Gln-Tyr-Asn-Ala-Asp Acts on voltage-gated sodium channels.

Glutamate is found in positions 3, 1 1 and 22 of the amyloid β-peptide. Among them the mutation from glutamic acid (E) to glutamine (Q) in position 22 (corresponding to amyloid precursor protein APP 693, Swissprot P05067) has been described as the so called Dutch type cerebroarterial amyloidosis mutation.

The β-amyloid peptides with a pyroglutamic acid residue in position 3, 1 1 and/or 22 have been described to be more cytotoxic and hydrophobic than the amyloid β-peptides 1 -40(42/43) (Saido T.C. 2000 Medical Hypotheses 54(3): 427-429). The multiple N-terminal variations, e.g. Abeta(3-40), Abeta(3-42), Abeta(1 1 -40) and Abeta (1 1 - 42) can be generated by the β-secretase enzyme β-site amyloid precursor protein-cleaving enzyme (BACE) at different sites (Huse J.T. et al. 2002 J. Biol. Chem. 277 (18): 16278-16284), and/or by aminopeptidase or dipeptidylaminopeptidase processing from the full lenght peptides Abeta(1 -40) and Abeta(1 -42). In all cases, cyclization of the then N-terminal occuring glutamic acid residue is catalyzed by QC.

Transepithelial transducing cells, particularly the gastrin (G) cell, co-ordinate gastric acid secretion with the arrival of food in the stomach. Recent work showed that multiple active products are generated from the gastrin precursor, and that there are multiple control points in gastrin biosynthesis. Biosynthetic precursors and intermediates (progastrin and Gly-gastrins) are putative growth factors; their products, the amidated gastrins, regulate epithelial cell proliferation, the differentiation of acid-producing parietal cells and histamine-secreting enterochromaffin-like (ECL) cells, and the expression of genes associated with histamine synthesis and storage in ECL cells, as well as acutely stimulating acid secretion. Gastrin also stimulates the production of members of the epidermal growth factor (EGF) family, which in turn inhibit parietal cell function but stimulate the growth of surface epithelial cells. Plasma gastrin concentrations are elevated in subjects with Helicobacter pylori, who are known to have increased risk of duodenal ulcer disease and gastric cancer (Dockray, G.J. 1999 J Physiol 15 315-324).

The peptide hormone gastrin, released from antral G cells, is known to stimulate the synthesis and release of histamine from ECL cells in the oxyntic mucosa via CCK-2 receptors. The mobilized histamine induces acid secretion by binding to the H(2) receptors located on parietal cells. Recent studies suggest that gastrin, in both its fully amidated and less processed forms (progastrin and glycine-extended gastrin), is also a growth factor for the gastrointestinal tract. It has been established that the major trophic effect of amidated gastrin is for the oxyntic mucosa of stomach, where it causes increased proliferation of gastric stem cells and ECL cells, resulting in increased parietal and ECL cell mass. On the other hand, the major trophic target of the less processed gastrin (e.g. glycine-extended gastrin) appears to be the colonic mucosa (Koh, T.J. and Chen, D. 2000 Regul Pept 9337-44).

Neurotensin (NT) is a neuropeptide implicated in the pathophysiology of schizophrenia that specifically modulates neurotransmitter systems previously demonstrated to be misregulated in this disorder. Clinical studies in which cerebrospinal fluid (CSF) NT concentrations have been measured revealed a subset of schizophrenic patients with decreased CSF NT concentrations that are restored by effective antipsychotic drug treatment. Considerable evidence also exists concordant with the involvement of NT systems in the mechanism of action of antipsychotic drugs. The behavioral and biochemical effects of centrally administered NT remarkably resemble those of systemically administered antipsychotic drugs, and antipsychotic drugs increase NT neurotransmission. This concatenation of findings led to the hypothesis that NT functions as an endogenous antipsychotic. Moreover, typical and atypical antipsychotic drugs differentially alter NT neurotransmission in nigrostriatal and mesolimbic dopamine terminal regions, and these effects are predictive of side effect liability and efficacy, respectively (Binder, E. B. et al. 2001 Biol Psychiatry 50 856-872).

Fertilization promoting peptide (FPP), a tripeptide related to thyrotrophin releasing hormone (TRH), is found in seminal plasma. Recent evidence obtained in vitro and in vivo showed that FPP plays an important role in regulating sperm fertility. Specifically, FPP initially stimulates nonfertilizing (uncapacitated) spermatozoa to "switch on" and become fertile more quickly, but then arrests capacitation so that spermatozoa do not undergo spontaneous acrosome loss and therefore do not lose fertilizing potential. These responses are mimicked, and indeed augmented, by adenosine, known to regulate the adenylyl cyclase (AC)/cAMP signal transduction pathway. Both FPP and adenosine have been shown to stimulate cAMP production in uncapacitated cells but inhibit it in capacitated cells, with FPP receptors somehow interacting with adenosine receptors and G proteins to achieve regulation of AC. These events affect the tyrosine phosphorylation state of various proteins, some being important in the initial "switching on", others possibly being involved in the acrosome reaction itself. Calcitonin and angiotensin II, also found in seminal plasma, have similar effects in vitro on uncapacitated spermatozoa and can augment responses to FPP. These molecules have similar effects in vivo, affecting fertility by stimulating and then maintaining fertilizing potential. Either reductions in the availability of FPP, adenosine, calcitonin, and angiotensin II or defects in their receptors contribute to male infertility (Fraser, L.R. and Adeoya-Osiguwa, S. A. 2001 Vitam Horm 63, 1 -28).

CCL2 (MCP-1 ), CCL7, CCL8, CCL16, CCL18 and fractalkine play an important role in pathophysiological conditions, such as suppression of proliferation of myeloid progenitor cells, neoplasia, inflammatory host responses, cancer, psoriasis, rheumatoid arthritis, atherosclerosis, vasculitis, humoral and cell-mediated immunity responses, leukocyte adhesion and migration processes at the endothelium, inflammatory bowel disease, restenosis, pulmonary fibrosis, pulmonary hypertention, liver fibrosis, liver cirrhosis, nephrosclerosis, ventricular remodeling, heart failure, arteriopathy after organ transplantations and failure of vein grafts. A number of studies have underlined in particular the crucial role of MCP-1 for the development of atherosclerosis (Gu, L, et al., (1998) Mol.Cell 2, 275-281 ; Gosling, J., et al., (1999) J Clin. Invest 103, 773-778); rheumatoid arthritis (Gong, J. H., et al., (1997) J Exp. Med 186, 131 - 137; Ogata, H., et al., (1997) J Pathol. 182, 106-1 14); pancreatitis (Bhatia, M., et al., (2005) Am.J Physiol Gastrointest. Liver Physiol 288, G1259-G1265); Alzheimer's disease (Yamamoto, M., et al., (2005) Am.J Pathol. 166, 1475-1485); lung fibrosis (Inoshima, I., et al., (2004) Am.J Physiol Lung Cell Mol. Physiol 286, L1038-L1044); renal fibrosis (Wada, T., et al., (2004) J Am.Soc.Nephrol. 15, 940-948), and graft rejection (Saiura, A., et al., (2004) Arterioscler. Thromb. Vase. Biol. 24, 1886-1890). Furthermore, MCP-1 might also play a role in gestosis (Katabuchi, H., et al., (2003) Med Electron Microsc. 36, 253-262), as a paracrine factor in tumor development (Ohta, M., et al., (2003) Int.J Oncol. 22, 773-778; Li, S., et al., (2005) J Exp.Med 202, 617-624), neuropathic pain (White, F. A., et al., (2005) Proc. Natl. Acad.Sci.U.S.A) and AIDS (Park, I. W., Wang, J. F., and Groopman, J. E. (2001 ) Blood 97, 352-358; Coll, B., et al., (2006) Cytokine 34, 51 -55). MCP-1 levels are increased in CSF of AD patients and patients showing mild cognitive impairment (MCI) (Galimberti, D., et al., (2006) Arch. Neurol. 63, 538-543). Furthermore, MCP- 1 shows an increased level in serum of patients with MCI and early AD (Clerici, F., et al., (2006) Neurobiol.Aging 27, 1763-1768). Several cytotoxic T lymphocyte peptide-based vaccines against hepatitis B, human immunodeficiency virus and melanoma were recently studied in clinical trials. One interesting melanoma vaccine candidate alone or in combination with other tumor antigens, is the decapeptide ELA. This peptide is a Melan-A/MART-1 antigen immunodominant peptide analog, with an N-terminal glutamic acid. It has been reported that the amino group and gamma-carboxylic group of glutamic acids, as well as the amino group and gamma- carboxamide group of glutamines, condense easily to form pyroglutamic derivatives. To overcome this stability problem, several peptides of pharmaceutical interest have been developed with a pyroglutamic acid instead of N-terminal glutamine or glutamic acid, without loss of pharmacological properties. Unfortunately compared with ELA, the pyroglutamic acid derivative (PyrELA) and also the N-terminal acetyl-capped derivative (AcELA) failed to elicit cytotoxic T lymphocyte (CTL) activity. Despite the apparent minor modifications introduced in PyrELA and AcELA, these two derivatives probably have lower affinity than ELA for the specific class I major histocompatibility complex. Consequently, in order to conserve full activity of ELA, the formation of PyrELA must be avoided (Beck A. et al. 2001 , J Pept Res 57(6):528-38.).

Orexin A is a neuropeptide that plays a significant role in the regulation of food intake and sleep-wakefulness, possibly by coordinating the complex behavioral and physiologic responses of these complementary homeostatic functions. It plays also a role in the homeostatic regulation of energy metabolism, autonomic function, hormonal balance and the regulation of body fluids.

Recently, increased levels of the pentapeptide QYNAD were identified in the cerebrospinal fluid (CSF) of patients suffering from multiple sclerosis or Guillain-Barre syndrome compared to healthy individuals (Brinkmeier H. et al. 2000, Nature Medicine 6, 808-81 1 ). There is a big controversy in the literature about the mechanism of action of the pentapeptide Gln-Tyr-Asn- Ala-Asp (QYNAD), especially its efficacy to interact with and block sodium channels resulting in the promotion of axonal dysfunction, which are involved in inflammatory autoimmune diseases of the central nervous system. But recently, it could be demonstrated that not QYNAD, but its cyclized, pyroglutamated form, pEYNAD, is the active form, which blocks sodium channels resulting in the promotion of axonal dysfunction. Sodium channels are expressed at high density in myelinated axons and play an obligatory role in conducting action potentials along axons within the mammalian brain and spinal cord. Therefore, it is speculated that they are involved in several aspects of the pathophysiology of inflammatory autoimmune diseases, especially multiple sclerosis, the Guillain-Barre syndrome and chronic inflammatory demyelinizing polyradiculoneuropathy.

Furthermore, QYNAD is a substrate of the enzyme glutaminyl cyclase (QC, EC 2.3.2.5), which is also present in the brain of mammals, especially in human brain. Glutaminyl cyclase catalyzes effectively the formation of pEYNAD from its precursor QYNAD. Accordingly, the present invention provides the use of the compounds of formula (I) for the preparation of a medicament for the prevention or alleviation or treatment of a disease selected from the group consisting of mild cognitive impairment, Alzheimer's disease, Familial British Dementia, Familial Danish Dementia, neurodegeneration in Down Syndrome, Huntington's disease, Kennedy's disease, ulcer disease, duodenal cancer with or w/o Helicobacter pylori infections, colorectal cancer, Zolliger-Ellison syndrome, gastric cancer with or without Helicobacter pylori infections, pathogenic psychotic conditions, schizophrenia, infertility, neoplasia, inflammatory host responses, cancer, malign metastasis, melanoma, psoriasis, rheumatoid arthritis, atherosclerosis, pancreatitis, restenosis, impaired humoral and cell- mediated immune responses, leukocyte adhesion and migration processes in the endothelium, impaired food intake, impaired sleep-wakefulness, impaired homeostatic regulation of energy metabolism, impaired autonomic function, impaired hormonal balance or impaired regulation of body fluids, multiple sclerosis, the Guillain-Barre syndrome and chronic inflammatory demyelinizing polyradiculoneuropathy. Furthermore, by administration of a compound according to the present invention to a mammal it can be possible to stimulate the proliferation of myeloid progenitor cells.

In addition, the administration of a QC inhibitor according to the present invention can lead to suppression of male fertility.

In a preferred embodiment, the present invention provides the use of inhibitors of QC (EC) activity in combination with other agents, especially for the treatment of neuronal diseases, artherosclerosis and multiple sclerosis. The present invention also provides a method of treatment of the aforementioned diseases comprising the administration of a therapeutically active amount of at least one compound of formula (I) to a mammal, preferably a human.

Most preferably, said method and corresponding uses are for the treatment of a disease selected from the group consisting of mild cognitive impairment, Alzheimer's disease, Familial British Dementia, Familial Danish Dementia, neurodegeneration in Down Syndrome, Parkinson's disease and Chorea Huntington, comprising the administration of a therapeutically active amount of at least one compound of formula (I) to a mammal, preferably a human. Even preferably, the present invention provides a method of treatment and corresponding uses for the treatment of rheumatoid arthritis, atherosclerosis, pancreatitis and restenosis.

Pharmaceutical combinations

In a preferred embodiment, the present invention provides a composition, preferably a pharmaceutical composition, comprising at least one QC inhibitor optionally in combination with at least one other agent selected from the group consisting of nootropic agents, neuroprotectants, antiparkinsonian drugs, amyloid protein deposition inhibitors, beta amyloid synthesis inhibitors, antidepressants, anxiolytic drugs, antipsychotic drugs and anti-multiple sclerosis drugs.

Most preferably, said QC inhibitor is a compound of formula (I) of the present invention.

More specifically, the aforementioned other agent is selected from the group consisting of beta- amyloid antibodies, vaccines, cysteine protease inhibitors, PEP-inhibitors, LiCI, acetylcholinesterase (AChE) inhibitors, PIMT enhancers, inhibitors of beta secretases, inhibitors of gamma secretases, inhibitors of aminopeptidases, preferably inhibitors of dipeptidyl peptidases, most preferably DP IV inhibitors; inhibitors of neutral endopeptidase, inhibitors of Phosphodiesterase-4 (PDE-4), TNFalpha inhibitors, muscarinic M1 receptor antagonists, NMDA receptor antagonists, sigma-1 receptor inhibitors, histamine H3 antagonists, immunomodulatory agents, immunosuppressive agents, MCP-1 antagonists or an agent selected from the group consisting of antegren (natalizumab), Neurelan (fampridine- SR), campath (alemtuzumab), IR 208, NBI 5788/MSP 771 (tiplimotide), paclitaxel, Anergix.MS (AG 284), SH636, Differin (CD 271 , adapalene), BAY 361677 (interleukin-4), matrix- metalloproteinase-inhibitors (e.g. BB 76163), interferon-tau (trophoblastin) and SAIK-MS.

Furthermore, the other agent may be, for example, an anti-anxiety drug or antidepressant selected from the group consisting of

(a) Benzodiazepines, e.g. alprazolam, chlordiazepoxide, clobazam, clonazepam, clorazepate, diazepam, fludiazepam, loflazepate, lorazepam, methaqualone, oxazepam, prazepam, tranxene,

(b) Selective serotonin re-uptake inhibitors (SSRI's), e.g. citalopram, fluoxetine,

fluvoxamine, escitalopram, sertraline, paroxetine,

(c) Tricyclic antidepressants, e.g. amitryptiline, clomipramine, desipramine, doxepin, imipramine

(d) Monoamine oxidase (MAO) inhibitors,

(e) Azapirones, e.g. buspirone, tandopsirone,

(f) Serotonin-norepinephrine reuptake inhibitors (SNRI's), e.g. venlafaxine, duloxetine,

(g) Mirtazapine,

(h) Norepinephrine reuptake inhibitors (NRI's), e.g. reboxetine,

(i) Bupropione, (j) Nefazodone,

(k) beta-blockers,

(I) NPY-receptor ligands: NPY agonists or antagonists.

In a further embodiment, the other agent may be, for example, an anti-multiple sclerosis drug selected from the group consisting of

a) dihydroorotate dehydrogenase inhibitors, e.g. SC-12267, teriflunomide, MNA-715, HMR-1279 (syn. to HMR-1715, MNA-279),

b) autoimmune suppressant, e.g. laquinimod,

c) paclitaxel,

d) antibodies, e.g. AGT-1 , anti-granulocyte-macrophage colony-stimulating factor (GM- CSF) monoclonal antibody, Nogo receptor modulators, ABT-874, alemtuzumab (CAMPATH), anti-OX40 antibody, CNTO-1275, DN-1921 , natalizumab (syn. to AN- 100226, Antegren, VLA-4 Mab), daclizumab (syn. to Zenepax, Ro-34-7375, SMART anti-Tac), J-695, priliximab (syn. to Centara, CEN-000029, cM-T412), MR A, Dantes, anti-IL-12-antibody,

e) peptide nucleic acid (PNA) preparations, e.g. reticulose,

f) interferon alpha, e.g. Alfaferone, human alpha interferon (syn. to Omniferon, Alpha Leukoferon),

g) interferon beta, e.g. Frone, interferon beta-1 a like Avonex, Betron (Rebif), interferon beta analogs, interferon beta-transferrin fusion protein, recombinant interferon beta-1 b like Betaseron,

h) interferon tau,

i) peptides, e.g. AT-008, AnergiX.MS, Immunokine (alpha-lmmunokine-NNS03), cyclic peptides like ZD-7349,

j) therapeutic enzymes, e.g. soluble CD8 (sCD8),

k) multiple sclerosis-specific autoantigen-encoding plasmid and cytokine-encoding plasmid, e.g. BHT-3009;

I) inhibitor of TNF-alpha, e.g. BLX-1002, thalidomide, SH-636,

m) TNF antagonists, e.g. solimastat, lenercept (syn. to RO-45-2081 , Tenefuse), onercept

(STNFR1 ), CC-1069,

n) TNF alpha, e.g. etanercept (syn. to Enbrel, TNR-001 )

o) CD28 antagonists, e.g. abatacept,

p) Lck tyrosine kinase inhibitors,

q) cathepsin K inhibitors, r) analogs of the neuron-targeting membrane transporter protein taurine and the plant- derived calpain inhibitor leupeptin, e.g. Neurodur,

s) chemokine receptor-1 (CCR1 ) antagonist, e.g. BX-471 ,

t) CCR2 antagonists,

u) AMPA receptor antagonists, e.g. ER-167288-01 and ER-099487, E-2007, talampanel, v) potassium channel blockers, e.g. fampridine,

w) tosyl-proline-phenylalanine small-molecule antagonists of the VLA-4/VCAM interaction, e.g. TBC-3342,

x) cell adhesion molecule inhibitors, e.g. TBC-772,

y) antisense oligonucleotides, e.g. EN-101 ,

z) antagonists of free immunoglobulin light chain (IgLC) binding to mast cell receptors, e.g. F-991 ,

aa) apoptosis inducing antigens, e.g. Apogen MS,

bb) alpha-2 adrenoceptor agonist, e.g. tizanidine (syn. to Zanaflex, Ternelin, Sirdalvo, Sirdalud, Mionidine),

cc) copolymer of L-tyrosine, L-lysine, L-glutamic acid and L-alanine, e.g. glatiramer acetate

(syn. to Copaxone, COP-1 , copolymer-1 ),

dd) topoisomerase II modulators, e.g. mitoxantrone hydrochloride,

ee) adenosine deaminase inhibitor, e.g. cladribine (syn. to Leustatin, Mylinax, RWJ- 26251 ),

ff) interleukin-10, e.g. ilodecakin (syn. to Tenovil, Sch-52000, CSIF),

gg) interleukin-12 antagonists, e.g. lisofylline (syn. to CT-1501 R, LSF, lysofylline), hh) Ethanaminum, e.g. SRI-62-834 (syn. to CRC-8605, NSC-614383),

ii) immunomodulators, e.g. SAIK-MS, PNU-156804, alpha-fetoprotein peptide (AFP), IPDS,

jj) retinoid receptor agonists, e.g. adapalene (syn. to Differin, CD-271 ),

kk) TGF-beta, e.g. GDF-1 (growth and differentiation factor 1 ),

ii) TGF-beta-2, e.g. BetaKine,

mm) MMP inhibitors, e.g. glycomed,

nn) phosphodiesterase 4 (PDE4) inhibitors, e.g. RPR-122818,

oo) purine nucleoside phosphorylase inhibitors, e.g. 9-(3-pyridylmethyl)-9-deazaguanine, peldesine (syn. to BCX-34, TO-200),

mm) alpha-4/beta-1 integrin antagonists, e.g. ISIS-104278,

qq) antisense alpha4 integrin (CD49d), e.g. ISIS-17044, ISIS-27104,

rr) cytokine-inducing agents, e.g. nucleosides, ICN-17261 , ss) cytokine inhibitors,

tt) heat shock protein vaccines, e.g. HSPPC-96,

uu) neuregulin growth factors, e.g. GGF-2 (syn. to neuregulin, glial growth factor 2), vv) cathepsin S - inhibitors,

ww) bropirimine analogs, e.g. PNU-56169, PNU-63693,

xx) Monocyte chemoattractant protein-1 inhibitors, e.g. benzimidazoles like MCP-1 inhibitors, LKS-1456, PD-064036, PD-064126, PD-084486, PD-172084, PD-172386.

Further, the present invention provides pharmaceutical compositions e.g. for parenteral, enteral or oral administration, comprising at least one QC inhibitor, optionally in combination with at least one of the other aforementioned agents.

These combinations provide a particularly beneficial effect. Such combinations are therefore shown to be effective and useful for the treatment of the aforementioned diseases. Accordingly, the invention provides a method for the treatment of these conditions.

The method comprises either co-administration of at least one QC inhibitor and at least one of the other agents or the sequential administration thereof. Co-administration includes administration of a formulation, which comprises at least one QC inhibitor and at least one of the other agents or the essentially simultaneous administration of separate formulations of each agent.

Beta-amyloid antibodies and compositions containing the same are described, e.g. in WO/2009/065054, WO/2009/056490, WO/2009/053696, WO/2009/033743, WO/2007/1 13172, WO/2007/022416, WO 2006/137354, WO 2006/1 18959, WO 2006/1031 16, WO 2006/095041 , WO 2006/081 171 , WO 2006/066233, WO 2006/066171 , WO 2006/066089, WO 2006/066049, WO 2006/055178, WO 2006/046644, WO 2006/039470, WO 2006/036291 , WO 2006/026408, WO 2006/016644, WO 2006/014638, WO 2006/014478, WO 2006/008661 , WO 2005/123775, WO 2005/120571 , WO 2005/105998, WO 2005/081872, WO 2005/080435, WO 2005/02851 1 , WO 2005/025616, WO 2005/025516, WO 2005/023858, WO 2005/018424, WO 2005/01 1599, WO 2005/000193, WO 2004/108895, WO 2004/098631 , WO 2004/080419, WO 2004/071408, WO 2004/069182, WO 2004/067561 , WO 2004/044204, WO 2004/032868, WO 2004/031400, WO 2004/029630, WO 2004/029629, WO 2004/024770, WO 2004/024090, WO 2003/104437, WO 2003/089460, WO 2003/086310, WO 2003/077858, WO 2003/074081 , WO 2003/070760, WO 2003/063760, WO 2003/055514, WO 2003/051374, WO 2003/048204, WO 2003/045128, WO 2003/040183, WO 2003/039467, WO 2003/016466, WO 2003/015691 , WO 2003/014162, WO 2003/012141 , WO 2002/088307, WO 2002/088306, WO 2002/074240, WO 2002/046237, WO 2002/046222, WO 2002/041842, WO 2001/062801 , WO 2001/012598, WO 2000/077178, WO 2000/072880, WO 2000/063250, WO 1999/060024, WO 1999/027944, WO 1998/044955, WO 1996/025435, WO 1994/017197, WO 1990/014840, WO 1990/012871 , WO 1990/012870, WO 1989/006242.

The beta-amyloid antibodies may be selected from, for example, polyclonal, monoclonal, chimenic or humanized antibodies. Furthermore, said antibodies may be useful to develop active and passive immune therapies, i.e. vaccines and monoclonal antibodies.

Suitable examples of beta-amyloid antibodies are ACU-5A5, huC091 (Acumen/Merck); PF- 4360365, RI-1014, RI-1219, RI-409, RN-1219 (Rinat Neuroscience Corp (Pfizer Inc)); the nanobody therapeutics of Ablynx/Boehringer Ingelheim; beta-amyloid-specific humanized monoclonal antibodies of Intellect Neurosciences/IBL; m266, m266.2 (Eli Lilly & Co.); AAB-02 (Elan); bapineuzumab (Elan); BAN-2401 (Bioarctic Neuroscience AB); ABP-102 (Abiogen Pharma SpA); BA-27, BC-05 (Takeda); R-1450 (Roche); ESBA-212 (ESBATech AG); AZD- 3102 (AstraZeneca) and beta-amyloid antibodies of Mindset BioPharmaceuticals Inc. Especially preferred are antibodies, which recognize the N-terminus of the Αβ peptide. A suitable antibody, which recognizes the Αβ-Ν-Terminus is, for example Acl-24 (AC Immune SA).

Monoclonal antibodies against beta-amyloid peptide are disclosed in WO 2007/068412, WO/2008/156621 and WO/2010/012004. Respective chimeric and humanized antibodies are disclosed in WO 2008/01 1348 and WO/2008/060364. Vaccine composition for treating an amyloid-associated disease is disclosed in WO/2002/096937, WO/2005/014041 , WO 2007/06841 1 , WO/2007/097251 , WO/2009/029272, WO/2009/054537, WO/2009/090650 WO/2009/095857, WO/2010/016912, WO/2010/01 1947, WO/2010/01 1999, WO/2010/044464.

Suitable vaccines for treating an amyloid-associated disease are, e.g. Affitopes AD-01 and AD-02 (GlaxoSmithKline), ACC-01 and ACC-02 (Elan/Wyeth), CAD-1 06 (Novartis / Cytos Biotechnology), Suitable cysteine protease inhibitors are inhibitors of cathepsin B. Inhibitors of cathepsin B and compositions containing such inhibitors are described, e.g. in WO/2008/077109, WO/2007/038772, WO 2006/060473, WO 2006/042103, WO 2006/039807, WO 2006/021413, WO 2006/021409, WO 2005/097103, WO 2005/007199, WO2004/084830, WO 2004/078908, WO 2004/026851 , WO 2002/094881 , WO 2002/027418, WO 2002/021509, WO 1998/046559, WO 1996/021655.

Examples of suitable PIMT enhancers are 10-aminoaliphatyl-dibenz[b, f] oxepines described in WO 98/15647 and WO 03/057204, respectively. Further useful according to the present invention are modulators of PIMT activity described in WO 2004/039773.

Inhibitors of beta secretase and compositions containing such inhibitors are described, e.g. in WO/2010/094242, WO/2010/058333, WO/2010/021680, WO/2009/108550, WO/2009/042694, WO/2008/054698, WO/2007/051333, WO/2007/021793, WO/2007/019080, WO/2007/019078, WO/2007/01 1810, WO03/059346, WO2006/099352, WO2006/078576, WO2006/060109, WO2006/057983, WO2006/057945, WO2006/055434, WO2006/044497, WO2006/034296, WO2006/034277, WO2006/029850, WO2006/026204, WO2006/014944, WO2006/014762, WO2006/002004, US 7,109,217, WO2005/1 13484, WO2005/103043, WO2005/103020, WO2005/065195, WO2005/051914, WO2005/044830, WO2005/032471 , WO2005/018545, WO2005/004803, WO2005/004802, WO2004/062625, WO2004/043916, WO2004/013098, WO03/099202, WO03/043987, WO03/039454, US 6,562,783, WO02/098849 and WO02/096897.

Suitable examples of beta secretase inhibitors for the purpose of the present invention are WY-25105 (Wyeth); Posiphen, (+)-phenserine (TorreyPines / NIH); LSN-2434074, LY- 2070275, LY-2070273, LY-2070102 (Eli Lilly & Co.); PNU-159775A, PNU-178025A, PNU- 17820A, PNU-33312, PNU-38773, PNU-90530 (Elan / Pfizer); KMI-370, KMI-358, kmi-008 (Kyoto University); OM-99-2, OM-003 (Athenagen Inc.); AZ-12304146 (AstraZeneca / Astex); GW-840736X (GlaxoSmithKline pic), DNP-004089 (De Novo Pharmaceuticals Ltd.) and CT- 21 166 (CoMentis Inc.).

Inhibitors of gamma secretase and compositions containing such inhibitors are described, e.g. in WO/2010/090954, WO/2009/01 1851 , WO/2009/008980, WO/2008/147800, WO/2007/084595, WO2005/008250, WO2006/004880, US 7, 122,675, US 7,030,239, US 6,992,081 , US 6,982,264, WO2005/097768, WO2005/028440, WO2004/101562, US 6,756,51 1 , US 6,683,091 , WO03/066592, WO03/014075, WO03/013527, WO02/36555, WO01/53255, US 7,109,217, US 7,101 ,895, US 7,049,296, US 7,034,182, US 6,984,626, WO2005/040126, WO2005/030731 , WO2005/014553, US 6,890,956, EP 1334085, EP 1263774, WO2004/101538, WO2004/00958, WO2004/08991 1 , WO2004/073630, WO2004/069826, WO2004/039370, WO2004/031 139, WO2004/031 137, US 6,713,276, US 6,686,449, WO03/091278, US 6,649,196, US 6,448,229, WO01 /77144 and WO01 /66564.

Suitable gamma secretase inhibitors for the purpose of the present invention are GSI-953, WAY-GSI-A, WAY-GSI-B (Wyeth); MK-0752, MRK-560, L-852505, L-685-458, L-852631 , L- 852646 (Merck & Co. Inc.); LY-450139, LY-41 1575, AN-37124 (Eli Lilly & Co.); BMS-299897, BMS-433796 (Bristol-Myers Squibb Co.); E-2012 (Eisai Co. Ltd.); EHT-0206, EHT-206 (ExonHit Therapeutics SA); NGX-555 (TorreyPines Therapeutics Inc.) and Semagacestat (Eli Lilly). DP IV-inhibitors and compositions containing such inhibitors are described, e.g. in US6,01 1 ,155; US6,107,317; US6,1 10,949; US6,124,305; US6,172,081 ; W099/61431 , W099/67278, W099/67279, DE19834591 , WO97/40832, WO95/15309, W098/19998, WO00/07617, WO99/38501 , W099/46272, WO99/38501 , WO01/68603, WO01/40180, WO01/81337, WO01/81304, WO01/55105, WO02/02560, WO01/34594, WO02/38541 , WO02/083128, WO03/072556, WO03/002593, WO03/000250, WO03/000180, WO03/000181 , EP1258476, WO03/002553, WO03/002531 , WO03/002530, WO03/004496, WO03/004498, WO03/024942, WO03/024965, WO03/033524, WO03/035057, WO03/035067, WO03/037327, WO03/040174, WO03/045977, WO03/055881 , WO03/057144, WO03/057666, WO03/068748, WO03/068757, WO03/082817, WO03/101449, WO03/101958, WO03/104229, WO03/74500, WO2004/007446, WO2004/007468, WO2004/018467, WO2004/018468, WO2004/018469, WO2004/026822, WO2004/032836, WO2004/033455, WO2004/037169, WO2004/041795, WO2004/043940, WO2004/048352, WO2004/050022, WO2004/052850, WO2004/058266, WO2004/064778, WO2004/069162, WO2004/071454, WO2004/076433, WO2004/076434, WO2004/087053, WO2004/089362, WO2004/099185, WO2004/103276, WO2004/103993, WO2004/108730, WO2004/1 10436, WO2004/1 1 1041 , WO2004/1 12701 , WO2005/000846, WO2005/000848, WO2005/01 1581 , WO2005/01691 1 , WO2005/023762, WO2005/025554, WO2005/026148, WO2005/030751 , WO2005/033106, WO2005/037828, WO2005/040095, WO2005/044195, WO2005/047297, WO2005/051950, WO2005/056003, WO2005/056013, WO2005/058849, WO2005/075426, WO2005/082348, WO2005/085246, WO2005/087235, WO2005/095339, WO2005/095343, WO2005/095381 , WO2005/108382, WO2005/1 13510, WO2005/1 16014, WO2005/1 16029, WO2005/1 18555, WO2005/120494, WO2005/121089, WO2005/121 131 , WO2005/123685, WO2006/995613; WO2006/009886; WO2006/013104; WO2006/017292; WO2006/019965; WO2006/020017; WO2006/023750; WO2006/039325; WO2006/041976; WO2006/047248; WO2006/058064; WO2006/058628; WO2006/066747; WO2006/066770 and WO2006/068978.

Suitable DP IV-inhibitors for the purpose of the present invention are for example Sitagliptin, des-fluoro-sitagliptin (Merck & Co. Inc.); vildagliptin, DPP-728, SDZ-272-070 (Novartis) ; ABT- 279, ABT-341 (Abbott Laboratories); denagliptin, TA-6666 (GlaxoSmithKline pic); SYR-322 (Takeda San Diego Inc.); talabostat (Point Therapeutics Inc.); Ro-0730699, R-1499, R-1438 (Roche Holding AG); FE-99901 1 (Ferring Pharmaceuticals); TS-021 (Taisho Pharmaceutical Co. Ltd.); GRC-8200 (Glenmark Pharmaceuticals Ltd.); ALS-2-0426 (Alantos Pharmaceuticals Holding Inc.); ARI-2243 (Arisaph Pharmaceuticals Inc.); SSR-162369 (Sanofi-Synthelabo); MP-513 (Mitsubishi Pharma Corp.); DP-893, CP-867534-01 (Pfizer Inc.); TSL-225, TMC-2A (Tanabe Seiyaku Co. Ltd.); PHX-1 149 (Phenomenix Corp.); saxagliptin (Bristol-Myers Squibb Co.); PSN-9301 ((OSI) Prosidion), S-40755 (Servier); KRP-104 (ActivX Biosciences Inc.); sulphostin (Zaidan Hojin); KR-62436 (Korea Research Institute of Chemical Technology); P32/98 (Probiodrug AG); Bl-A, Bl-B (Boehringer Ingelheim Corp.); SK-0403 (Sanwa Kagaku Kenkyusho Co. Ltd.); and NNC-72-2138 (Novo Nordisk A/S).

Other preferred DP IV-inhibitors are

(i) dipeptide-like compounds, disclosed in WO 99/61431 , e.g. N-valyl prolyl, O-benzoyl hydroxylamine, alanyl pyrrolidine, isoleucyl thiazolidine like L-allo-isoleucyl thiazolidine, L- threo-isoleucyl pyrrolidine and salts thereof, especially the fumaric salts, and L-allo-isoleucyl pyrrolidine and salts thereof;

(ii) peptide structures, disclosed in WO 03/002593, e.g. tripeptides;

(iii) peptidylketones, disclosed in WO 03/033524;

(vi) substituted aminoketones, disclosed in WO 03/040174;

(v) topically active DP IV-inhibitors, disclosed in WO 01/14318;

(vi) prodrugs of DP IV-inhibitors, disclosed in WO 99/67278 and WO 99/67279; and

(v) glutaminyl based DP IV-inhibitors, disclosed in WO 03/072556 and WO 2004/099134.

Suitable beta amyloid synthesis inhibitors for the purpose of the present invention are for example Bisnorcymserine (Axonyx Inc.); (R)-flurbiprofen (MCP-7869; Flurizan) (Myriad Genetics); nitroflurbiprofen (NicOx); BGC-20-0406 (Sankyo Co. Ltd.) and BGC-20-0466 (BTG pic), RQ-00000009 (RaQualia Pharma Inc).

Suitable amyloid protein deposition inhibitors for the purpose of the present invention are for example SP-233 (Samaritan Pharmaceuticals); AZD-103 (Ellipsis Neurotherapeutics Inc.); AAB-001 (Bapineuzumab), AAB-002, ACC-001 (Elan Corp pic); Colostrinin (ReGen Therapeutics pic); Tramiprosate (Neurochem); AdPEDI-(amyloid-beta1 -6)1 1 ) (Vaxin Inc.); MPI-127585, MPI-423948 (Mayo Foundation); SP-08 (Georgetown University); ACU-5A5 (Acumen / Merck); Transthyretin (State University of New York); PTI-777, DP-74, DP 68, Exebryl (ProteoTech Inc.); m266 (Eli Lilly & Co.); EGb-761 (Dr. Willmar Schwabe GmbH); SPI- 014 (Satori Pharmaceuticals Inc.); ALS-633, ALS-499 (Advanced Life Sciences Inc.); AGT- 160 (ArmaGen Technologies Inc.); TAK-070 (Takeda Pharmaceutical Co. Ltd.); CHF-5022, CHF-5074, CHF-5096 and CHF-5105 (Chiesi Farmaceutici SpA.), SEN-1 1 76 and SEN-1 329 (Senexis Ltd.), AGT-160 (ArmaGen Technologies), Davunetide (Allon Therapeutics), ELND-005 (Elan Corp / Transition Therapeutics) and nilvadipine (Archer Pharmaceuticals).

Suitable PDE-4 inhibitors for the purpose of the present invention are for example Doxofylline (Instituto Biologico Chemioterapica ABC SpA.); idudilast eye drops, tipelukast, ibudilast (Kyorin Pharmaceutical Co. Ltd.); theophylline (Elan Corp.); cilomilast (GlaxoSmithKline pic); Atopik (Barrier Therapeutics Inc.); tofimilast, CI-1044, PD-189659, CP-220629, PDE 4d inhibitor BHN (Pfizer Inc.); arofylline, LAS-37779 (Almirall Prodesfarma SA.); roflumilast, hydroxypumafentrine (Altana AG), tetomilast (Otska Pharmaceutical Co. Ltd.); tipelukast, ibudilast (Kyorin Pharmaceutical), CC-10004 (Celgene Corp.); HT-0712, IPL-4088 (Inflazyme Pharmaceuticals Ltd.); MEM-1414, MEM-1917 (Memory Pharmaceuticals Corp.); oglemilast, GRC-4039 (Glenmark Pharmaceuticals Ltd.); AWD-12-281 , ELB-353, ELB-526 (Elbion AG); EHT-0202 (ExonHit Therapeutics SA.); ND-1251 (Neuro3d SA.); 4AZA-PDE4 (4 AZA Bioscience NV.); AVE-81 12 (Sanofi-Aventis); CR-3465 (Rottapharm SpA.); GP-0203, NCS- 613 (Centre National de la Recherche Scientifique); KF-19514 (Kyowa Hakko Kogyo Co. Ltd.); ONO-6126 (Ono Pharmaceutical Co. Ltd.); OS-0217 (Dainippon Pharmaceutical Co. Ltd.); IBFB-13001 1 , IBFB-150007, IBFB-130020, IBFB-140301 (IBFB Pharma GmbH); IC-485 (ICOS Corp.); RBx-14016 and RBx-1 1082 (Ranbaxy Laboratories Ltd.). A preferred PDE-4- inhibitor is Rolipram. MAO inhibitors and compositions containing such inhibitors are described, e.g. in WO2006/091988, WO2005/007614, WO2004/089351 , WO01/26656, WO01 /12176, WO99/57120, W099/571 19, W099/13878, WO98/40102, WO98/01 157, WO96/20946, WO94/07890 and W092/21333.

Suitable MAO-inhibitors for the purpose of the present invention are for example Linezolid (Pharmacia Corp.); RWJ-416457 (RW Johnson Pharmaceutical Research Institute); budipine (Altana AG); GPX-325 (BioResearch Ireland); isocarboxazid; phenelzine; tranylcypromine; indantadol (Chiesi Farmaceutici SpA.); moclobemide (Roche Holding AG); SL-25.1 131 (Sanofi-Synthelabo); CX-1370 (Burroughs Wellcome Co.); CX-157 (Krenitsky Pharmaceuticals Inc.); desoxypeganine (HF Arzneimittelforschung GmbH & Co. KG); bifemelane (Mitsubishi-Tokyo Pharmaceuticals Inc.); RS-1636 (Sankyo Co. Ltd.); esuprone (BASF AG); rasagiline (Teva Pharmaceutical Industries Ltd.); ladostigil (Hebrew University of Jerusalem); safinamide (Pfizer), NW-1048 (Newron Pharmaceuticals SpA.), EVT-302 (Evotec), .

Suitable histamine H3 antagonists for the purpose of the present invention are, e.g. ABT-239, ABT-834 (Abbott Laboratories); 3874-H1 (Aventis Pharma); UCL-2173 (Berlin Free University), UCL-1470 (BioProjet, Societe Civile de Recherche); DWP-302 (Daewoong Pharmaceutical Co Ltd); GSK-189254A, GSK-207040A (GlaxoSmithKline Inc.); cipralisant, GT-2203 (Gliatech Inc.); Ciproxifan (INSERM), 7S,2S-2-(2-Aminoethyl)-1 -(1 H-imidazol-4- yl)cyclopropane (Hokkaido University); JNJ-17216498, JNJ-5207852 (Johnson & Johnson); NNC-0038-0000-1049 (Novo Nordisk A/S); and Sch-79687 (Schering-Plough). PEP inhibitors and compositions containing such inhibitors are described, e.g. in JP 01042465, JP 03031298, JP 04208299, WO 00/71 144, US 5,847,155; JP 09040693, JP 10077300, JP 05331072, JP 05015314, WO 95/15310, WO 93/00361 , EP 0556482, JP 06234693, JP 01068396, EP 0709373, US 5,965,556, US 5,756,763, US 6,121 ,31 1 , JP 63264454, JP 64000069, JP 63162672, EP 0268190, EP 0277588, EP 0275482, US 4,977,180, US 5,091 ,406, US 4,983,624, US 5,1 12,847, US 5,100,904, US 5,254,550, US 5,262,431 , US 5,340,832, US 4,956,380, EP 0303434, JP 03056486, JP 01 143897, JP 1226880, EP 0280956, US 4,857,537, EP 0461677, EP 0345428, JP 02275858, US 5,506,256, JP 06192298, EP 0618193, JP 03255080, EP 0468469, US 5,1 18,81 1 , JP 05025125, WO 9313065, JP 05201970, WO 9412474, EP 0670309, EP 0451547, JP 06339390, US 5,073,549, US 4,999,349, EP 0268281 , US 4,743,616, EP 0232849, EP 0224272, JP 621 14978, JP 621 14957, US 4,757,083, US 4,810,721 , US 5,198,458, US 4,826,870, EP 0201742, EP 0201741 , US 4,873,342, EP 0172458, JP 61037764, EP 0201743, US 4,772,587, EP 0372484, US 5,028,604, WO 91 /18877, JP 04009367, JP 04235162, US 5,407,950, WO 95/01352, JP 01250370, JP 02207070, US 5,221 ,752, EP 0468339, JP 0421 1648, WO 99/46272, WO 2006/058720 and PCT/EP2006/061428.

Suitable prolyl endopeptidase inhibitors for the purpose of the present invention are, e.g. Fmoc-Ala-Pyrr-CN, Z-Phe-Pro-Benzothiazole (Probiodrug), Z-321 (Zeria Pharmaceutical Co Ltd.); ONO-1603 (Ono Pharmaceutical Co Ltd); JTP-4819 (Japan Tobacco Inc.) and S-17092 (Servier).

Other suitable compounds that can be used according to the present invention in combination with QC-inhibitors are NPY, an NPY mimetic or an NPY agonist or antagonist or a ligand of the NPY receptors.

Preferred according to the present invention are antagonists of the NPY receptors.

Suitable ligands or antagonists of the NPY receptors are 3a, 4,5,9b-tetrahydro-1 h- benz[e]indol-2-yl amine-derived compounds as disclosed in WO 00/68197.

NPY receptor antagonists which may be mentioned include those disclosed in European patent applications EP 0 614 91 1 , EP 0 747 357, EP 0 747 356 and EP 0 747 378; international patent applications WO 94/17035, WO 97/1991 1 , WO 97/19913, WO 96/12489, WO 97/19914, WO 96/22305, WO 96/40660, WO 96/12490, WO 97/09308, WO 97/20820, WO 97/20821 , WO 97/20822, WO 97/20823, WO 97/19682, WO 97/25041 , WO 97/34843, WO 97/46250, WO 98/03492, WO 98/03493, WO 98/03494 and WO 98/07420; WO 00/30674, US patents Nos. 5,552,41 1 , 5,663,192 and 5,567,714; 6,1 14,336, Japanese patent application JP 09157253; international patent applications WO 94/00486, WO 93/12139, WO 95/00161 and WO 99/15498; US Patent No. 5,328,899; German patent application DE 393 97 97; European patent applications EP 355 794 and EP 355 793; and Japanese patent applications JP 061 16284 and JP 07267988. Preferred NPY antagonists include those compounds that are specifically disclosed in these patent documents. More preferred compounds include amino acid and non-peptide-based NPY antagonists. Amino acid and non-peptide-based NPY antagonists which may be mentioned include those disclosed in European patent applications EP 0 614 91 1 , EP 0 747 357, EP 0 747 356 and EP 0 747 378; international patent applications WO 94/17035, WO 97/1991 1 , WO 97/19913, WO 96/12489, WO 97/19914, WO 96/22305, WO 96/40660, WO 96/12490, WO 97/09308, WO 97/20820, WO 97/20821 , WO 97/20822, WO 97/20823, WO 97/19682, WO 97/25041 , WO 97/34843, WO 97/46250, WO 98/03492, WO 98/03493, WO 98/03494, WO 98/07420 and WO 99/15498 ; US patents Nos. 5,552,41 1 , 5,663,192 and 5,567,714; and Japanese patent application JP 09157253. Preferred amino acid and non-peptide-based NPY antagonists include those compounds that are specifically disclosed in these patent documents.

Particularly preferred compounds include amino acid-based NPY antagonists. Amino acid- based compounds, which may be mentioned include those disclosed in international patent applications WO 94/17035, WO 97/1991 1 , WO 97/19913, WO 97/19914 or, preferably, WO 99/15498. Preferred amino acid-based NPY antagonists include those that are specifically disclosed in these patent documents, for example BIBP3226 and, especially, (R)-N2- (diphenylacetyl)-(R)-N-[1 -(4-hydroxy- phenyl) ethyl] arginine amide (Example 4 of international patent application WO 99/15498).

M1 receptor agonists and compositions containing such inhibitors are described, e.g. in WO2004/087158, WO91/10664. Suitable M1 receptor antagonists for the purpose of the present invention are for example CDD-0102 (Cognitive Pharmaceuticals); Cevimeline (Evoxac) (Snow Brand Milk Products Co. Ltd.); NGX-267 (TorreyPines Therapeutics); sabcomeline (GlaxoSmithKline); alvameline (H Lundbeck A/S); LY-593093 (Eli Lilly & Co.); VRTX-3 (Vertex Pharmaceuticals Inc.); WAY- 132983 (Wyeth), CI-101 7/ (PD-151832) (Pfizer Inc.) and MCD-386 (Mitridion Inc.), .

Acetylcholinesterase inhibitors and compositions containing such inhibitors are described, e.g. in WO2006/071274, WO2006/070394, WO2006/040688, WO2005/092009, WO2005/079789, WO2005/039580, WO2005/027975, WO2004/084884, WO2004/037234, WO2004/032929, WO03/101458, WO03/091220, WO03/082820, WO03/020289, WO02/32412, WO01/85145, WO01/78728, WO01/66096, WO00/02549, WO01/00215, WO00/15205, WO00/23057, WO00/33840, WO00/30446, WO00/23057, WO00/15205, WO00/09483, WO00/07600, WO00/02549, W099/47131 , WO99/07359, WO98/30243, W097/38993, W097/13754, W094/29255, WO94/20476, W094/19356, WO93/03034 and W092/19238. Suitable acetylcholinesterase inhibitors for the purpose of the present invention are for example Donepezil (Eisai Co. Ltd.); rivastigmine (Novartis AG); (-)-phenserine (TorreyPines Therapeutics); ladostigil (Hebrew University of Jerusalem); huperzine A (Mayo Foundation); galantamine (Johnson & Johnson); Memoquin (Universita di Bologna); SP-004 (Samaritan Pharmaceuticals Inc.); BGC-20-1259 (Sankyo Co. Ltd.); physostigmine (Forest Laboratories Inc.); NP-0361 (Neuropharma SA); ZT-1 (Debiopharm); tacrine (Warner-Lambert Co.); metrifonate (Bayer Corp.), INM-176 (Whanln), huperzine A (Neuro-Hitech / Xel Pharmaceutical), mimopezil (Debiopharm) and Dimebon (Medivation/Pfizer).

NMDA receptor antagonists and compositions containing such inhibitors are described, e.g. in WO2006/094674, WO2006/058236, WO2006/058059, WO2006/010965, WO2005/000216, WO2005/102390, WO2005/079779, WO2005/079756, WO2005/072705, WO2005/070429, WO2005/055996, WO2005/035522, WO2005/009421 , WO2005/000216, WO2004/092189, WO2004/039371 , WO2004/028522, WO2004/009062, WO03/010159, WO02/072542, WO02/34718, WO01/98262, WO01/94321 , WO01/92204, WO01/81295, WO01/32640, WO01/10833, WO01/10831 , WO00/5671 1 , WO00/29023, WO00/00197, W099/53922, W099/48891 , W099/45963, WO99/01416, WO99/07413, WO99/01416, WO98/50075, WO98/50044, WO98/10757, WO98/05337, W097/32873, W097/23216, W097/23215, W097/23214, W096/14318, WO96/08485, W095/31986, W095/26352, WO95/26350, W095/26349, W095/26342, W095/12594, WO95/02602, WO95/02601 , WO94/20109, W094/13641 , WO94/09016 and W093/25534.

Suitable NMDA receptor antagonists for the purpose of the present invention are for example Memantine (Merz & Co. GmbH); topiramate (Johnson & Johnson); AVP-923 (Neurodex) (Center for Neurologic Study); EN-3231 (Endo Pharmaceuticals Holdings Inc.); neramexane (MRZ-2/579) (Merz and Forest); CNS-5161 (CeNeS Pharmaceuticals Inc.); dexanabinol (HU- 21 1 ; Sinnabidol; PA-5021 1 ) (Pharmos); EpiCept NP-1 (Dalhousie University); indantadol (V- 3381 ; CNP-3381 ) (Vernalis); perzinfotel (EAA-090, WAY-126090, EAA-129) (Wyeth); RGH- 896 (Gedeon Richter Ltd.); traxoprodil (CP-101606), besonprodil (PD-196860, CI-1041 ) (Pfizer Inc.); CGX-1007 (Cognetix Inc.); delucemine (NPS-1506) (NPS Pharmaceuticals Inc.); EVT- 101 (Roche Holding AG); acamprosate (Synchroneuron LLC); CR-3991 , CR-2249, CR-3394 (Rottapharm SpA.); AV-101 (4-CI-kynurenine (4-CI-KYN)), 7-chloro-kynurenic acid (7-CI- KYNA) (VistaGen); NPS-1407 (NPS Pharmaceuticals Inc.); YT-1006 (Yaupon Therapeutics Inc.); ED-1812 (Sosei R&D Ltd.); himantane (hydrochloride N-2-(adamantly)-hexamethylen- imine) (RAMS); Lancicemine (AR-R-15896) (AstraZeneca); EVT-102, Ro-25-6981 and Ro-63- 1908 (Hoffmann-La Roche AG / Evotec), neramexane (Merz).

Furthermore, the present invention relates to combination therapies useful for the treatment of atherosclerosis, restenosis or arthritis, administering a QC inhibitor in combination with another therapeutic agent selected from the group consisting of inhibitors of the angiotensin converting enzyme (ACE); angiotensin II receptor blockers; diuretics; calcium channel blockers (CCB); beta-blockers; platelet aggregation inhibitors; cholesterol absorption modulators; HMG-Co-A reductase inhibitors; high density lipoprotein (HDL) increasing compounds; renin inhibitors; IL- 6 inhibitors; antiinflammatory corticosteroids; antiproliferative agents; nitric oxide donors; inhibitors of extracellular matrix synthesis; growth factor or cytokine signal transduction inhibitors; MCP-1 antagonists and tyrosine kinase inhibitors providing beneficial or synergistic therapeutic effects over each monotherapy component alone. Angiotensin II receptor blockers are understood to be those active agents that bind to the AT1 -receptor subtype of angiotensin II receptor but do not result in activation of the receptor. As a consequence of the blockade of the AT1 receptor, these antagonists can, e.g. be employed as antihypertensive agents. Suitable angiotensin II receptor blockers which may be employed in the combination of the present invention include ΑΤΊ receptor antagonists having differing structural features, preferred are those with non-peptidic structures. For example, mention may be made of the compounds that are selected from the group consisting of valsartan (EP 443983), losartan (EP 253310), candesartan (EP 459136), eprosartan (EP 403159), irbesartan (EP 45451 1 ), olmesartan (EP 503785), tasosartan (EP 539086), telmisartan (EP 522314), the compound with the designation E-41 77 of the formula

the compound with the designation SC-52458 of the following formula

and the compound with the designation the compound ZD-8731 of the formula

or, in each case, a pharmaceutically acceptable salt thereof.

Preferred AT1 -receptor antagonists are those agents that have been approved and reached the market, most preferred is valsartan, or a pharmaceutically acceptable salt thereof.

The interruption of the enzymatic degradation of angiotensin to angiotensin II with ACE inhibitors is a successful variant for the regulation of blood pressure and thus also makes available a therapeutic method for the treatment of hypertension.

A suitable ACE inhibitor to be employed in the combination of the present invention is, e.g. a compound selected from the group consisting alacepril, benazepril, benazeprilat; captopril, ceronapril, cilazapril, delapril, enalapril, enaprilat, fosinopril, imidapril, lisinopril, moveltopril, perindopril, quinapril, ramipril, spirapril, temocapril and trandolapril, or in each case, a pharmaceutically acceptable salt thereof. Preferred ACE inhibitors are those agents that have been marketed, most preferred are benazepril and enalapril.

A diuretic is, for example, a thiazide derivative selected from the group consisting of chlorothiazide, hydrochlorothiazide, methylclothiazide, and chlorothalidon. The most preferred diuretic is hydrochlorothiazide. A diuretic furthermore comprises a potassium sparing diuretic such as amiloride or triameterine, or a pharmaceutically acceptable salt thereof.

The class of CCBs essentially comprises dihydropyridines (DHPs) and non-DHPs, such as diltiazem-type and verapamil-type CCBs.

A CCB useful in said combination is preferably a DHP representative selected from the group consisting of amlodipine, felodipine, ryosidine, isradipine, lacidipine, nicardipine, nifedipine, niguldipine, niludipine, nimodipine, nisoldipine, nitrendipine and nivaldipine, and is preferably a non-DHP representative selected from the group consisting of flunarizine, prenylamine, diltiazem, fendiline, gallopamil, mibefradil, anipamil, tiapamil and verapamil, and in each case, a pharmaceutically acceptable salt thereof. All these CCBs are therapeutically used, e.g. as anti-hypertensive, anti-angina pectoris or anti-arrhythmic drugs. Preferred CCBs comprise amlodipine, diltiazem, isradipine, nicardipine, nifedipine, nimodipine, nisoldipine, nitrendipine and verapamil or, e.g. dependent on the specific CCB, a pharmaceutically acceptable salt thereof. Especially preferred as DHP is amlodipine or a pharmaceutically acceptable salt thereof, especially the besylate. An especially preferred representative of non-DHPs is verapamil or a pharmaceutically acceptable salt, especially the hydrochloride, thereof.

Beta-blockers suitable for use in the present invention include beta-adrenergic blocking agents (beta-blockers), which compete with epinephrine for beta-adrenergic receptors and interfere with the action of epinephrine. Preferably, the beta-blockers are selective for the beta- adrenergic receptor as compared to the alpha-adrenergic receptors, and so do not have a significant alpha-blocking effect. Suitable beta-blockers include compounds selected from acebutolol, atenolol, betaxolol, bisoprolol, carteolol, carvedilol, esmolol, labetalol, metoprolol, nadolol, oxprenolol, penbutolol, pindolol, propranolol, sotalol and timolol. Where the beta- blocker is an acid or base or otherwise capable of forming pharmaceutically acceptable salts or prodrugs, these forms are considered to be encompassed herein, and it is understood that the compounds may be administered in free form or in the form of a pharmaceutically acceptable salt or a prodrug, such as a physiologically hydrolyzable and acceptable ester. For example, metoprolol is suitably administered as its tartrate salt, propranolol is suitably administered as the hydrochloride salt, and so forth.

Platelet aggregation inhibitors include PLAVIX® (clopidogrel bisulfate), PLETAL® (cilostazol) and aspirin.

Cholesterol absorption modulators include ZETIA® (ezetimibe) and KT6-971 (Kotobuki Pharmaceutical Co. Japan).

HMG-Co-A reductase inhibitors (also called beta-hydroxy-beta-methylglutaryl-co-enzyme-A reductase inhibitors or statins) are understood to be those active agents which may be used to lower lipid levels including cholesterol in blood.

The class of HMG-Co-A reductase inhibitors comprises compounds having differing structural features. For example, mention may be made of the compounds, which are selected from the group consisting of atorvastatin, cerivastatin, fluvastatin, lovastatin, pitavastatin, pravastatin, rosuvastatin and simvastatin, or in each case, a pharmaceutically acceptable salt thereof.

Preferred HMG-Co-A reductase inhibitors are those agents, which have been marketed, most preferred is atorvastatin, pitavastatin or simvastatin, or a pharmaceutically acceptable salt thereof. HDL-increasing compounds include, but are not limited to, cholesterol ester transfer protein (CETP) inhibitors. Examples of CETP inhibitors include JTT705 disclosed in Example 26 of U.S. Patent No. 6,426,365 issued July 30, 2002, and pharmaceutically acceptable salts thereof. Inhibition of interleukin 6 mediated inflammation may be achieved indirectly through regulation of endogenous cholesterol synthesis and isoprenoid depletion or by direct inhibition of the signal transduction pathway utilizing interleukin-6 inhibitor/antibody, interleukin-6 receptor inhibitor/antibody, interleukin-6 antisense oligonucleotide (ASON), gp130 protein inhibitor/antibody, tyrosine kinase inhibitors/antibodies, serine/threonine kinase inhibitors/antibodies, mitogen-activated protein (MAP) kinase inhibitors/antibodies, phosphatidylinositol 3-kinase (PI3K) inhibitors/antibodies, Nuclear factor kappaB (NF-KB) inhibitors/antibodies, ΙκΒ kinase (IKK) inhibitors/antibodies, activator protein-1 (AP-1 ) inhibitors/antibodies, STAT transcription factors inhibitors/antibodies, altered IL-6, partial peptides of IL-6 or IL-6 receptor, or SOCS (suppressors of cytokine signaling) protein, PPAR gamma and/or PPAR beta/delta activators/ligands or a functional fragment thereof.

A suitable antiinflammatory corticosteroid is dexamethasone.

Suitable antiproliferative agents are cladribine, rapamycin, vincristine and taxol.

A suitable inhibitor of extracellular matrix synthesis is halofuginone.

A suitable growth factor or cytokine signal transduction inhibitor is, e.g. the ras inhibitor R1 15777.

A suitable tyrosine kinase inhibitor is tyrphostin.

Suitable renin inhibitors are described, e.g. in WO 2006/1 16435. A preferred renin inhibitor is aliskiren, preferably in the form of the hemi-fumarate salt thereof.

MCP-1 antagonists may, e.g. be selected from anti-MCP-1 antibodies, preferably monoclonal or humanized monoclonal antibodies, MCP-1 expression inhibitors, CCR2-antagonists, TNF- alpha inhibitors, VCAM-1 gene expression inhibitors and anti-C5a monoclonal antibodies. MCP-1 antagonists and compositions containing such inhibitors are described, e.g. in WO02/070509, WO02/081463, WO02/060900, US2006/670364, US2006/677365, WO2006/097624, US2006/316449, WO2004/056727, WO03/053368, WO00/198289, WO00/157226, WO00/046195, WO00/046196, WO00/046199, WO00/046198, WO00/046197, WO99/046991 , WO99/007351 , WO98/006703, WO97/012615, WO2005/105133, WO03/037376, WO2006/125202, WO2006/085961 , WO2004/024921 , WO2006/074265.

Suitable MCP-1 antagonists are, for instance, C-243 (Telik Inc.); NOX-E36 (Noxxon Pharma AG); AP-761 (Actimis Pharmaceuticals Inc.); ABN-912, NIBR-177 (Novartis AG); CC-1 1006 (Celgene Corp.); SSR-150106 (Sanofi-Aventis); MLN-1202 (Millenium Pharmaceuticals Inc.); AGI-1067, AGIX-4207, AGI-1096 (AtherioGenics Inc.); PRS-21 1095, PRS-21 1092 (Pharmos Corp.); anti-C5a monoclonal antibodies, e.g. neutrazumab (G2 Therapies Ltd.); AZD-6942 (AstraZeneca pic); 2-mercaptoimidazoles (Johnson & Johnson); TEI-E00526, TEI-6122 (Deltagen); RS-504393 (Roche Holding AG); SB-282241 , SB-380732, ADR-7 (GlaxoSmithKline); anti-MCP-1 monoclonal antibodies(Johnson & Johnson).

Combinations of QC-inhibitors with MCP-1 antagonists may be useful for the treatment of inflammatory diseases in general, including neurodegenerative diseases. Combinations of QC-inhibitors with MCP-1 antagonists are preferred for the treatment of Alzheimer's disease.

Most preferably the QC inhibitor is combined with one or more compounds selected from the following group:

PF-4360365, m266, bapineuzumab, R-1450, Posiphen, (+)-phenserine, MK-0752, LY-450139, E-2012, (R)-flurbiprofen, AZD-103, AAB-001 (Bapineuzumab), Tramiprosate, EGb-761 , TAK- 070, Doxofylline, theophylline, cilomilast, tofimilast, roflumilast, tetomilast, tipelukast, ibudilast, HT-0712, MEM-1414, oglemilast, Linezolid, budipine, isocarboxazid, phenelzine, tranylcypromine, indantadol, moclobemide, rasagiline, ladostigil, safinamide, ABT-239, ABT- 834, GSK-189254A, Ciproxifan, JNJ-17216498, Fmoc-Ala-Pyrr-CN, Z-Phe-Pro- Benzothiazole, Z-321 , ONO-1603, JTP-4819, S-17092, BIBP3226; (R)-N2-(diphenylacetyl)- (R)-N-[1 -(4-hydroxyphenyl) ethyl] arginine amide, Cevimeline, sabcomeline, (PD-151832), Donepezil, rivastigmine, (-)-phenserine, ladostigil, galantamine, tacrine, metrifonate, Memantine, topiramate, AVP-923, EN-3231 , neramexane, valsartan, benazepril, enalapril, hydrochlorothiazide, amlodipine, diltiazem, isradipine, nicardipine, nifedipine, nimodipine, nisoldipine, nitrendipine, verapamil, amlodipine, acebutolol, atenolol, betaxolol, bisoprolol, carteolol, carvedilol, esmolol, labetalol, metoprolol, nadolol, oxprenolol, penbutolol, pindolol, propranolol, sotalol, timolol, PLAVIX® (clopidogrel bisulfate), PLETAL® (cilostazol), aspirin, ZETIA® (ezetimibe) and KT6-971 , statins, atorvastatin, pitavastatin or simvastatin; dexamethasone, cladribine, rapamycin, vincristine, taxol, aliskiren, C-243, ABN-912, SSR- 150106, MLN-1202 and betaferon.

In particular, the following combinations are considered: a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1 , 2, 4, 5, 7, 9, 1 1 , 13, 15 and 17-29, in combination with Atorvastatin for the treatment and/or prevention of artherosclerosis, a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1 , 2, 4, 5, 7, 9, 1 1 , 13, 15 and 17-29, in combination with immunosuppressive agents, preferably rapamycin for the prevention and/or treatment of restenosis,

a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1 , 2, 4, 5, 7, 9, 1 1 , 13, 15 and 17-29, in combination with immunosuppressive agents, preferably paclitaxel for the prevention and/or treatment of restenosis,

a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1 , 2, 4, 5, 7, 9, 1 1 , 13, 15 and 17-29, in combination with AChE inhibitors, preferably Donepezil, for the prevention and/or treatment of Alzheimer's disease,

a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1 , 2, 4, 5, 7, 9, 1 1 , 13, 15 and 17-29, in combination with interferones, preferably Aronex, for the prevention and/or treatment of multiple sclerosis,

a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1 , 2, 4, 5, 7, 9, 1 1 , 13, 15 and 17-29, in combination with interferones, preferably betaferon, for the prevention and/or treatment of multiple sclerosis,

a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1 , 2, 4, 5, 7, 9, 1 1 , 13, 15 and 17-29, in combination with interferones, preferably Rebif , for the prevention and/or treatment of multiple sclerosis

a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1 , 2, 4, 5, 7, 9, 1 1 , 13, 15 and 17-29, in combination with Copaxone, for the prevention and/or treatment of multiple sclerosis,

a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1 , 2, 4, 5, 7, 9, 1 1 , 13, 15 and 17-29, in combination with dexamethasone, for the prevention and/or treatment of restenosis, a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1 , 2, 4, 5, 7, 9, 1 1 , 13, 15 and 17-29, in combination with dexamethasone, for the prevention and/or treatment of atherosclerosis,

- a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1 , 2, 4, 5, 7, 9, 1 1 , 13, 15 and 17-29, in combination with dexamethasone, for the prevention and/or treatment of rheumatid arthritis,

a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1 , 2, 4, 5, 7, 9, 1 1 , 13, 15 and 17-29, in combination with HMG-Co-A-reductase inhibitors, for the prevention and/or treatment of restenosis, wherein the HMG-Co-A-reductase inhibitor is selected from atorvastatin, cerivastatin, fluvastatin, lovastatin, pitavastatin, pravastatin, rosuvastatin and simvastatin,

- a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1 , 2, 4, 5, 7, 9, 1 1 , 13, 15 and 17-29, in combination with HMG-Co-A reductase inhibitors, for the prevention and/or treatment of atherosclerosis wherein the HMG-Co-A-reductase inhibitor is selected from atorvastatin, cerivastatin, fluvastatin, lovastatin, pitavastatin, pravastatin, rosuvastatin and simvastatin,

a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1 , 2, 4, 5, 7, 9, 1 1 , 13, 15 and 17-29, in combination with HMG-Co-A reductase inhibitors, for the prevention and/or treatment of rheumatoid arthritis wherein the HMG-Co-A-reductase inhibitor is selected from atorvastatin, cerivastatin, fluvastatin, lovastatin, pitavastatin, pravastatin, rosuvastatin and simvastatin,

a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1 , 2, 4, 5, 7, 9, 1 1 , 13, 15 and 17-29, in combination with amyloid-beta antibodies for the prevention and/or treatment of mild cognitive impairment, wherein the amyloid-beta antibody is Acl-24,

a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1 , 2, 4, 5, 7, 9, 1 1 , 13, 15 and 17-29, in combination with amyloid-beta antibodies for the prevention and/or treatment of Alzheimer's disease, wherein the amyloid-beta antibody is Acl-24, a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1 , 2, 4, 5, 7, 9, 1 1 , 13, 15 and 17-29, in combination with amyloid-beta antibodies for the prevention and/or treatment of neurodegeneration in Down Syndrome, wherein the amyloid-beta antibody is Acl- 24,

a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1 , 2, 4, 5, 7, 9, 1 1 , 13, 15 and 17-29, in combination with beta-secretase inhibitors for the prevention and/or treatment of mild cognitive impairment, wherein the beta-secretase inhibitor is selected from WY- 25105, GW-840736X and CTS-21 166,

a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1 , 2, 4, 5, 7, 9, 1 1 , 13, 15 and 17-29, in combination with beta-secretase inhibitors for the prevention and/or treatment of Alzheimer's disease, wherein the beta-secretase inhibitor is selected from WY- 25105, GW-840736X and CTS-21 166,

a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1 , 2, 4, 5, 7, 9, 1 1 , 13, 15 and 17-29, in combination with beta-secretase inhibitors for the prevention and/or treatment of neurodegeneration in Down Syndrome, wherein the beta-secretase inhibitor is selected from WY-25105, GW-840736X and CTS-21 166,

a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1 , 2, 4, 5, 7, 9, 1 1 , 13, 15 and 17-29, in combination with gamma-secretase inhibitors for the prevention and/or treatment of mild cognitive impairment, wherein the gamma-secretase inhibitor is selected from LY-450139, LY-41 1575 and AN-37124,

a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1 , 2, 4, 5, 7, 9, 1 1 , 13, 15 and 17-29, in combination with gamma-secretase inhibitors for the prevention and/or treatment of Alzheimer's disease, wherein the gamma-secretase inhibitor is selected from LY- 450139, LY-41 1575 and AN-37124,

a QC inhibitor, preferably a QC inhibitor of formula (I), more preferably a QC inhibitor selected from any one of examples 1 , 2, 4, 5, 7, 9, 1 1 , 13, 15 and 17-29, in combination with gamma-secretase inhibitors for the prevention and/or treatment of neurodegeneration in Down Syndrome, wherein the gamma-secretase inhibitor is selected from LY-450139, LY-41 1575 and AN-37124. Such a combination therapy is in particular useful for AD, FAD, FDD and neurodegeneration in Down syndrome as well as atherosclerosis, rheumatoid arthritis, restenosis and pancreatitis. Such combination therapies might result in a better therapeutic effect (less proliferation as well as less inflammation, a stimulus for proliferation) than would occur with either agent alone.

With regard to the specific combination of inhibitors of QC and further compounds it is referred in particular to WO 2004/098625 in this regard, which is incorporated herein by reference.

Pharmaceutical compositions

To prepare the pharmaceutical compositions of this invention, at least one compound of formula (I) optionally in combination with at least one of the other aforementioned agents can be used as the active ingredient(s). The active ingredient(s) is intimately admixed with a pharmaceutical carrier according to conventional pharmaceutical compounding techniques, which carrier may take a wide variety of forms depending of the form of preparation desired for administration, e.g., oral or parenteral such as intramuscular. In preparing the compositions in oral dosage form, any of the usual pharmaceutical media may be employed. Thus, for liquid oral preparations, such as for example, suspensions, elixirs and solutions, suitable carriers and additives include water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents and the like; for solid oral preparations such as, for example, powders, capsules, gelcaps and tablets, suitable carriers and additives include starches, sugars, diluents, granulating agents, lubricants, binders, disintegrating agents and the like. Because of their ease in administration, tablets and capsules represent the most advantageous oral dosage unit form, in which case solid pharmaceutical carriers are obviously employed. If desired, tablets may be sugar coated or enteric coated by standard techniques. For parenterals, the carrier will usually comprise sterile water, though other ingredients, for example, for purposes such as aiding solubility or for preservation, may be included. Injectable suspensions may also prepared, in which case appropriate liquid carriers, suspending agents and the like may be employed. The pharmaceutical compositions herein will contain, per dosage unit, e.g., tablet, capsule, powder, injection, teaspoonful and the like, an amount of the active ingredient(s) necessary to deliver an effective dose as described above. The pharmaceutical compositions herein will contain, per dosage unit, e.g., tablet, capsule, powder, injection, suppository, teaspoonful and the like, from about 0.03 mg to 100 mg/kg (preferred 0.1 - 30 mg/kg) and may be given at a dosage of from about 0.1 - 300 mg/kg per day (preferred 1 - 50 mg/kg per day) of each active ingredient or combination thereof. The dosages, however, may be varied depending upon the requirement of the patients, the severity of the condition being treated and the compound being employed. The use of either daily administration or post-periodic dosing may be employed.

Preferably these compositions are in unit dosage forms from such as tablets, pills, capsules, powders, granules, sterile parenteral solutions or suspensions, metered aerosol or liquid sprays, drops, ampoules, autoinjector devices or suppositories; for oral parenteral, intranasal, sublingual or rectal administration, or for administration by inhalation or insufflation. Alternatively, the composition may be presented in a form suitable for once-weekly or once- monthly administration; for example, an insoluble salt of the active compound, such as the decanoate salt, may be adapted to provide a depot preparation for intramuscular injection. For preparing solid compositions such as tablets, the principal active ingredient is mixed with a pharmaceutical carrier, e.g. conventional tableting ingredients such as corn starch, lactose, sucrose, sorbitol, talc, stearic acid, magnesium stearate, dicalcium phosphate or gums, and other pharmaceutical diluents, e.g. water, to form a solid preformulation composition containing a homogeneous mixture of a compound of the present invention, or a pharmaceutically acceptable salt thereof. When referring to these preformulation compositions as homogeneous, it is meant that the active ingredient is dispersed evenly throughout the composition so that the composition may be readily subdivided into equally effective dosage forms such as tablets, pills and capsules. This solid preformulation composition is then subdivided into unit dosage forms of the type described above containing from 0.1 to about 500 mg of each active ingredient or combinations thereof of the present invention.

The tablets or pills of the compositions of the present invention can be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action. For example, the tablet or pill can comprise an inner dosage and an outer dosage component, the latter being in the form of an envelope over the former. The two components can be separated by an enteric layer which serves to resist disintegration in the stomach and permits the inner component to pass intact into the duodenum or to be delayed in release. A variety of material can be used for such enteric layers or coatings, such materials including a number of polymeric acids with such materials as shellac, cetyl alcohol and cellulose acetate. This liquid forms in which the compositions of the present invention may be incorporated for administration orally or by injection include, aqueous solutions, suitably flavoured syrups, aqueous or oil suspensions, and flavoured emulsions with edible oils such as cottonseed oil, sesame oil, coconut oil or peanut oil, as well as elixirs and similar pharmaceutical vehicles. Suitable dispersing or suspending agents for aqueous suspensions, include synthetic and natural gums such as tragacanth, acacia, alginate, dextran, sodium carboxymethylcellulose, methylcellulose, polyvinylpyrrolidone or gelatin.

The pharmaceutical composition may contain between about 0.01 mg and 100 mg, preferably about 5 to 50 mg, of each compound, and may be constituted into any form suitable for the mode of administration selected. Carriers include necessary and inert pharmaceutical excipients, including, but not limited to, binders, suspending agents, lubricants, flavorants, sweeteners, preservatives, dyes, and coatings. Compositions suitable for oral administration include solid forms, such as pills, tablets, caplets, capsules (each including immediate release, timed release and sustained release formulations), granules, and powders, and liquid forms, such as solutions, syrups, elixirs, emulsions, and suspensions. Forms useful for parenteral administration include sterile solutions, emulsions and suspensions.

Advantageously, compounds of the present invention may be administered in a single daily dose, or the total daily dosage may be administered in divided doses of two, three or four times daily. Furthermore, compounds for the present invention can be administered in intranasal form via topical use of suitable intranasal vehicles, or via transdermal skin patches well known to those of ordinary skill in that art. To be administered in the form of transdermal delivery system, the dosage administration will, of course, be continuous rather than intermittent throughout the dosage regimen.

For instance, for oral administration in the form of a tablet or capsule, the active drug component can be combined with an oral, non-toxic pharmaceutically acceptable inert carrier such as ethanol, glycerol, water and the like. Moreover, when desired or necessary, suitable binders; lubricants, disintegrating agents and coloring agents can also be incorporated into the mixture. Suitable binders include, without limitation, starch, gelatin, natural sugars such as glucose or betalactose, corn sweeteners, natural and synthetic gums such as acacia, tragacanth or sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride and the like. Disintegrators include, without limitation, starch, methyl cellulose, agar, bentonite, xanthan gum and the like. The liquid forms in suitable flavored suspending or dispersing agents such as the synthetic and natural gums, for example, tragacanth, acacia, methyl-cellulose and the like. For parenteral administration, sterile suspensions and solutions are desired. Isotonic preparations which generally contain suitable preservatives are employed when intravenous administration is desired.

The compounds or combinations of the present invention can also be administered in the form of liposome delivery systems, such as small unilamellar vesicles, large unilamellar vesicles, and multilamellar vesicles. Liposomes can be formed from a variety of phospholipids, such as cholesterol, stearylamine or phosphatidylcholines.

Compounds or combinations of the present invention may also be delivered by the use of monoclonal antibodies as individual carriers to which the compound molecules are coupled. The compounds of the present invention may also be coupled with soluble polymers as targetable drug carriers. Such polymers can include polyvinylpyrrolidone, pyran copolymer, polyhydroxypropylmethacrylamidephenol, polyhydroxyethylaspartamid-ephenol, or polyethyl eneoxidepolyllysine substituted with palmitoyl residue. Furthermore, the compounds of the present invention may be coupled to a class of biodegradable polymers useful in achieving controlled release of a drug, for example, polyactic acid, polyepsilon caprolactone, polyhydroxy butyeric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanoacrylates and cross- linked or amphipathic block copolymers of hydrogels.

Compounds or combinations of this invention may be administered in any of the foregoing compositions and according to dosage regimens established in the art whenever treatment of the addressed disorders is required.

The daily dosage of the products may be varied over a wide range from 0.01 to 1 .000 mg per mammal per day. For oral administration, the compositions are preferably provided in the form of tablets containing, 0.01 , 0.05, 0.1 , 0.5, 1 .0, 2.5, 5.0, 10.0, 15.0, 25.0, 50.0, 100, 150, 200, 250 and 500 milligrams of each active ingredient or combinations thereof for the symptomatic adjustment of the dosage to the patient to be treated. An effective amount of the drug is ordinarily supplied at a dosage level of from about 0.1 mg/kg to about 300 mg/kg of body weight per day. Preferably, the range is from about 1 to about 50 mg/kg of body weight per day. The compounds or combinations may be administered on a regimen of 1 to 4 times per day. Optimal dosages to be administered may be readily determined by those skilled in the art, and will vary with the particular compound used, the mode of administration, the strength of the preparation, the mode of administration, and the advancement of disease condition. In addition, factors associated with the particular patient being treated, including patient age, weight, diet and time of administration, will result in the need to adjust dosages.

In a further aspect, the invention also provides a process for preparing a pharmaceutical composition comprising at least one compound of formula (I), optionally in combination with at least one of the other aforementioned agents and a pharmaceutically acceptable carrier.

The compositions are preferably in a unit dosage form in an amount appropriate for the relevant daily dosage. Suitable dosages, including especially unit dosages, of the the compounds of the present invention include the known dosages including unit doses for these compounds as described or referred to in reference text such as the British and US Pharmacopoeias, Remington's Pharmaceutical Sciences (Mack Publishing Co.), Martindale The Extra Pharmacopoeia (London, The Pharmaceutical Press) (for example see the 31 st Edition page 341 and pages cited therein) or the above mentioned publications.

Examples of the invention

Examples of compounds of formula (I)

Example Name Structure Formula Mol Weight

1-(1H- benzo [d] imidazol-5 -

1 yl)-4- C16H13N3O 263.294 phenylazetidin-2- HN

one

The invention further relates to the racemates, S-stereoisomers and R-stereoisomers of the compounds of the present invention as shown in the table above.

Synthesis of the Compounds of the linvention

General synthesis description: Method 1 Examples

Step A:

1 .2 equivalent of 5-amino-benzimidazole and 1 .0 equivalents of the respective aldehyde were dissolved in toluol and heated under reflux overnight. After completion of the reaction, the solvent was removed and the remaining oil was used for further steps without further purification

Step B:

2.0 equivalents of zinc-dust were suspended in benzol. 0.7 equivalents of TMS-CI were added under an argon atmosphere. The mixture was then stirred under reflux for 15 min. After cooling to rt, 1 equivalent of the Schiff-base and 1 .7 equivalents of brom-acetic acid ethylester were added. The mixture was kept under reflux overnight. Afterwards the solvent was removed and the remains were taken up in little methylene chloride, drawn on silica gel and subjected to column chromatography, applying a CH 2 CI 2 /MeOH gradient.

Method 2

Step A:

Chloro trimethylsilane (0.5eq) was added to a suspension of zinc dust (1 .5eq) in dry benzene (50ml_) and refluxed for " l Omin. The reaction mass was cooled to 00C, Ethyl bromo acetate (1 .5 eq) and a solution of I (1 eq) in dry benzene were added successively and refluxed for 2h. The reaction mass was quenched with saturated ammonium chloride solution and extracted with ethyl acetate. The combined organic layer was washed successively with water, brine; dried over anhydrous sodium sulfate and concentrated in vacuo to afford crude. Purification by column chromatography over silica gel (60-120 mesh) using 20-25% ethyl acetate in PET ether as eluent afforded II.

Step B

Pyridinium dichromate (4eq), Molecular sieves were added to a solution of II (1 eq) in dichloromethane (400ml_) at 00C and stirred for 4h at room temperature The reaction mass was filtered over celite and washed with dichloromethane. The combined filtrate and washings was concentrated in vacuo to afford crude. Purification by column chromatography over silica gel (60-120mesh) using 15% ethyl acetate in PET ether as eluent afforded III. Step C A solution of tert-butyl 5-amino-1 H-benzimidazole-1 -carboxylate (0.9 eq) in xylene was added to a solution of III (1 eq) and pyridine in Xylene and taken in a round bottomed flask fitted with a distillation condenser at 1400C. The reaction mass was further heated to 1600C and xylene was collected. The solvent was evaporated in vacuo to afford crude. Purification by column chromatography over silica gel (60-120mesh) using 5% methanol in DCM as eluent afforded IV.

Step D

Sodium borohydride (2eq) was added in one lot to a solution of IV (1 eq) in mixture of Tetrahydrofuran and Methanol at OOC and stirred for 15min. The reaction mass was quenched into saturated ammonium chloride solution and extracted with ethyl acetate. The combined organic layer was washed successively with water, brine; dried over anhydrous sodium sulfate and concentrated in vacuo, triturated with pet ether and dried under reduced pressure to afford V.

Step E

Diethylazodicarboxylate (1 .5eq) was added to a stirred solution of V (1 eq) and Triphenylphosphine (1 .5eq) in dry Tetrahydrofuran at OOC and stirred at room temperature for 30min. The reaction mass was quenched with water and extracted with ethyl acetate. The combined organic layer was washed with water, brine, dried over anhydrous sodium sulfate and concentrated in vacuo to crude. Purification by column chromatography over silica gel (60- 120mesh) using 35% ethyl acetate in pet ether as eluent afforded VI.

Step F

Trifluoroaceticacid acid was added to a solution of crude VI (1 eq) in Dichloromethane at 0°C and stirred for 2h at room temperature. The volatiles were evaporated in vacuo and the resulting residue was partitioned between saturated sodium bicarbonate solution and ethyl acetate. The organic layer was separated and the aqueous layer was extracted with ethyl acetate. The combined organic layer was washed successively with water, brine; dried over anhydrous sodium sulfate and concentrated in vacuo to afford crude. Purification by column chromatography over neutral alumina using 5-8% methanol in dichloromethane as eluent afforded VII.

Synthesis of Example Compounds 1 to 16: Example 1

The compound was synthesized according to method 1 : rad -(1H-benzoimidazol-5-yl)-4-phenylazetidin-2-one:

Step A: 5-aminobenzimidazole (1 .33g, 10 mmol), benzaldehyde (1 .27 g, 1 .22 mL, 12 mmol) in 80 mL of toluol, yielding 1 .62 g crude I;

Step B: I (0.433 g, 2 mmol), zinc dust (0.262 g, 4 mmol), TMS-CI (0.180 mL, 1 .4 mmol), bromo acetic acid ethyl ester (0.377 mL, 3.4 mmol).Yield: 0.089g (5.6%); MS m/z 285.1 (M+H) + ; 1 H NMR (DMSO, 400 MHz): δ 0.82-0.91 (m, H); 0.97-1 .16 (m, 4H); 1 .39-1 .42 (m, H); 1 .52-1 .69 (m, 5H); 3.24-3.27 (m, H); 3.42-3.46 (m, H); 4.48-4.52 (m, H); 6.92 (s, H); 7.56-7.59 (dd, H, 3 J=9.1 Hz, 4 J=2.1 Hz); 7.73-7.75 (d, H, 3 J=9.1 Hz); 7.94-7.95 (d, H, 4 J=2.1 Hz); 9.24 (s, H), HPLC 99%.

Examples 2 and 3

(R)- 1-( 1H-benzo[d]imidazol-6-yl)-4-phenylazetidin-2-one 2

Example 1 (0.089g) was subjected to chiral prep HPLC, affording compound 2. Yield: 0.025g, 1 H-NMR (DMSO-d6): δ 12.25(d, 1 H); 8.13(d, 1 H); 7.53(d, 1 H); 7.46-7.30(m, 6H); 7.12(d, 1 H); 5.25(t, 1 H); 3.63-3.57(m, 1 H); 2.92-2.86(m, 1 H); MS=264(M+1 ); HPLC~96.31 %: Chiral HPLC~92.48% (S)-1-(1H-benzo[d]imidazol-6-yl)-4-phenylazetidin-2-one 3

Example 1 (0.089g) was subjected to chiral prep HPLC, affording compound 3. Yield: 0.025g, 1 H-NMR (DMSO-d6): 5 12.25(d, 1 H); 8.13(s, 1 H); 7.46-7.30(m, 7H); 7.12(bs, 1 H); 5.25(q, 1 H); 3.63-3.57(m, 1 H); 2.90-2.86(m, 1 H); MS=264(M+1 ); HPLC~99.00%: Chiral HPLC~98.35%.

Example 4

Rac 1-(1 H-benzo[d]imidazol-5-yl)-4-(2,6-difluoro-4-methoxyphenyl)aze tidin-2-one 4

Step A: 5-aminobenzimidazole (0.690g, 5.2 mmol), 2,5-difluoro-4methoxy-benzaldehyde (1 .07 g, 6.2 mmol) in 80 mL of toluol, yielding 1 .66 g crude I;

Step B: I (1 .15 g, 4 mmol), zinc dust (0.524 g, 8.31 mmol), TMS-CI (0.360 mL, 2.8 mmol), bromo acetic acid ethyl ester (0.754 mL, 6.8 mmol). Yield: 0.022g (1 .2%); MS m/z 330.4 (M+H) + ; 1 H NMR (DMSO-d 6 , 400 MHz): δ 3.25-3.29 (m, 1 H), 3.63-3.67 (m, 1 H), 3.76 (s, 3H), 5.49-5.51 8m, 1 H), 6.80 (d, 2H, 3J=1 1 .2 Hz), 7.34 (dd, 1 H, 3J=8.8 Hz, 4J=2 Hz), 7.50 (d, 1 H, 4J=2 Hz), 7.74 (d, 1 H, 3J=8.8 Hz), 9.10 (s, 1 H) , HPLC [A]: 100%)

Examples 5 and 6

The compounds were prepared according to Method 2 rac-4-(4-(3,3-difluoropropoxy)-2-fluorophenyl)- 1-(1H-benzo[d]imidazol-5-yl)azetidi

Step A: Chloro trimethylsilane (1 .2ml_, 9.174mmol), zinc dust (1 .8g, 27.52mmol), benzene (50ml_), Ethyl bromo acetate (3.0ml_, 27.52mmol), 4-(3,3-difluoropropoxy)-2- fluorobenzaldehyde (4.0g, 18.34mmol) in dry benzene (20mL), Yield: 4g (73%)

Step B: Pyridinium dichromate (19.6g, 52.28mmol), Molecular sieves (19.6g) were added to a solution of ethyl 3-(4-(3,3-difluoropropoxy)-2-fluorophenyl)-3-hydroxypropanoa te (4.0g, 13.07mmol) in dichloromethane (400ml_), Yield 3.5g (88%)

Step C: tert-butyl 5-amino-1 H-benzo[d]imidazole-1 -carboxylate (1 .7g, 7.40mmol) in xylene (20ml_) was added to a solution of ethyl 3-(4-(3,3-difluoropropoxy)-2-fluorophenyl)-3- oxopropanoate (2.5g, 8.22mmol) and pyridine (0.5ml_) in Xylene (40ml_, Yield 2.2g (50.7%). Step D: Sodium borohydride (340mg, 8.944mmol), N-(1 -tert-butyl-oxycarbonyl-1 H- benzo[d]imidazole-5-yl-)3-(4-(3,3-difluoropropoxy)-2-fluorop henyl)-3-oxopropanamide (2.2g, 4.47mmol) in mixture of Tetrahydrofuran (35ml_) and Methanol (15ml_) at 0°C, Yield 1 .8g (77,6%)

Step E: Diethylazodicarboxylate (1 .2mL, 7.59mmol), N-(1 -tert-butyl-oxycarbonyl-1 H- benzo[d]imidazole-5-yl-)3-(4-(3,3-difluoropropoxy)-2-fluorop henyl)-3-hydroxypropanamide (2.5g, 5.060mmol) and Triphenylphosphine (1 .98g, 7.59mmol) Tetrahydrofuran (30mL) at 0°C- Yield: 1 .0g (42%)

Step F: Trifluoroaceticacid acid (2m L) was added to a solution of crude tert-butyl 6-(2-(4-(3,3- difluoropropoxy)-2-fluorophenyl)-4-oxoazetidin-1 -yl)-1 H-benzo[d]imidazole-1 -carboxylate (1 .0g, 2.08mmol) in Dichloromethane (20mL) at 0°C, Yield: 0.3g (40%).

The remains were purified by Chiral prep HPLC using the following conditions:

Column: Chiralcel-OX-H (250 * 30 * 5.0μ); Mobile phase: Hexane (0.1 %DEA): Ethanol (75:25); Flow rate: 30 mL/min; Diluent: Mobile phase, the prep fractions were evaporated in vacuo to afford 80mg each of the examples:

(R)-4-(4-(3,3-difluoropropoxy)-2-fluorophenyl)- 1-(1H-benzo[d]im (5) Melting range: 150-155°C; 1 H-NMR (DMSO-d6): δ 12.36(d, 1 H); 8.16(d, 1 H); 7.57-7.26 (m, 3H); 6.90(dd, 1 H); 6.79(d, 1 H); 6.21 (bt, 1 H); 5.36(d, 1 H); 4.1 1 (t, 2H); 3.58(q, 1 H); 2.97(dd, 1 H); 2.35-2.20(m, 2H); MS=376(M+1 ); HPLC [B]~98.54%: Chiral HPLC~99.87%.

(S)-4-(4-(3,3-difluoropropoxy)-2-fluorophenyl)-1-(1H-benz o[d]imida (6) Melting range: 137-142°C; 1 H-NMR (DMSO-d6): δ 12.39(s, 1 H); 8.15(s, 1 H); 7.53(d, 1 H); 7.37(t, 2H); 7.20(bs, 1 H); 6.93(dd, 1 H); 6.79(dd, 1 H); 6.21 (bt, 1 H); 5.36(d, 1 H); 4.1 1 (t, 2H); 3.58(q, 1 H); 2.98(d, 1 H); 2.35-2.20(m, 2H); MS=376(M+1 ); HPLC [B]~99.14%: Chiral HPLC~99.03%.

Examples 7 and 8

rac4-(4-(3,3-difluoropropoxy)-2,3-difluorophenyl)-1-(1H-b enzo[d]imidazol-5-yl)azetidi

Step A: Chloro trimethylsilane (0.8ml_, 6.35mmol), zinc dust (1 .16g, 7.79mmol), benzene (80mL), Ethyl bromo acetate (1 .69mL, 15.25mmol), 4-(3,3-difluoropropoxy)-2,3- difluorobenzaldehyde (3.0g, 12.71 mmol) in dry benzene (20ml_), Yield: 2g (51 %)

Step B: Pyridinium dichromate (9.28g, 24.69mmol), Molecular sieves (18.5g) were added to a solution of ethyl 3-(4-(3,3-difluoropropoxy)-2,3-fluorophenyl)-3-hydroxypropan oate (2g, 6.17mmol) in dichloromethane (300mL), Yield 1 .8g (91 %)

Step C: tert-butyl 5-amino-1 H-benzo[d]imidazole-1 -carboxylate (1 .17g, 5.03mmol) in xylene (30ml_) was added to a solution of ethyl 3-(4-(3,3-difluoropropoxy)-2,3-difluorophenyl)-3- oxopropanoate (1 .8g, 5.59mmol) and pyridine (0.5mL) in Xylene (40mL, Yield 2.6g (86.5%). Step D: Sodium borohydride (386mg, 10.22mmol)), N-(1 -tert-butyl-oxycarbonyl-1 H- benzo[d]imidazole-5-yl-)3-(4-(3,3-difluoropropoxy)-2,3-diflu orophenyl)-3-oxopropanamide

(2.6g, 5.1 1 mmol) in mixture of Tetrahydrofuran (35ml_) and Methanol (15mL) at 0°C, Yield 1 .4g (53.6%)

Step E: Diethylazodicarboxylate (0.64mL, 4.1 1 mmol), N-(1 -tert-butyl-oxycarbonyl-1 H- benzo[d]imidazole-5-yl-)3-(4-(3,3-difluoropropoxy)-2,3-fluor ophenyl)-3-hydroxypropanamide (1 .4g, 2.74mmol) and Triphenylphosphine (1 .08g, 4.1 1 mmol)) Tetrahydrofuran (30mL) at 0°C- Yield: 2.5g crude.

Step F: Trifluoroaceticacid acid (6m L) was added to a solution of crude tert-butyl 6-(2-(4-(3,3- difluoropropoxy)-2,3-difluorophenyl)-4-oxoazetidin-1 -yl)-1 H-benzo[d]imidazole-1 -carboxylate (2.5g, 5.08mmol) in Dichloromethane (30mL) at 0°C, Purification by column chromatography over neutral alumina using 1 -1 .5% methanol in dichloromethane as eluent afforded rac4-(4- (3,3-difluoropropoxy)-2,3-difluorophenyl)-1 -(1 H-benzo[d]imidazol-5-yl)azetidin-2-one. Yield: 0.45g. The remains were purified by Chiral prep HPLC using the following conditions: Column: Chiralcel-OX-H (250 * 30 * 5.0μ); Mobile phase: A: Hexane (0.1 % DEA); Ethanol (75:25); Flow rate: 30mL/ min; Diluent: Mobile phase.

Diluent: Mobile phase, the prep fractions were evaporated in vacuo to afford 80mg each of the examples:

(R)-4-(4-(3,3-difluoropropoxy)-2,3-difluorophenyl)- 1-(1H-benzo[d]im

(7) Melting range: 165-170°C; 1 H-NMR (DMS0-d6): δ 12.40(d, 1 H); 8.16(d, 1 H); 7.58-7.42 (m, 2H); 7.34(t, 1 H); 7.21 (bs, 1 H); 7.08(q, 2H); 6.21 (bt, 1 H); 5.41 (d, 1 H); 4.21 (t, 2H); 3.67-3.60(q, 1 H); 3.02(d, 1 H); 2.39-2.24(m, 2H); MS=394(M+1 ); HPLC [B]~98.21 %: Chiral HPLC~99.91 %. (S)-4-(4-(3,3-difluoropropoxy)-2,3-difluorophenyl)-1-(1H-ben zo[d]imidazol-5^

(8)

Melting range: 148-153°C; 1 H-NMR (DMS0-d6): δ 12.40(d, 1 H); 8.16(d, 1 H); 7.58-7.42 (m, 2H); 7.34(t, 1 H); 7.20(bs, 1 H); 7.08(q, 2H); 6.21 (bt, 1 H); 5.41 (d, 1 H); 4.21 (t, 2H); 3.67-3.60(q, 1 H); 3.04(q, 1 H); 2.36-2.26(m, 2H); MS=394(M+1 ); HPLC [B]~99.62%: Chiral HPLC~98.93%. Examples 9 and 10

rac-4-(4-(3,3-difluoropropoxy)-2fi-difluorophenyl)- 1-(1H-benzo[d]imidaz

Step A: Chloro trimethylsilane (1 .3mL, 10.58mmol), zinc dust (1 .93g, 29.66mmol), benzene (30mL), Ethyl bromo acetate (2.8mL, 25.14mmol), 4-(3,3-difluoropropoxy)-2,6- difluorobenzaldehyde (5g, 21 .18mmol) in dry benzene (20mL), Yield: 4.5g (66%)

Step B: Pyridinium dichromate (15.67g, 41 .65mmol) 3-(4-(3,3-difluoropropoxy)-2,6- fluorophenyl)-3-hydroxypropanoate (4.5g, 13.88mmol) in dichloromethane (400mL), Yield 1 .2g (28%)

Step C: tert-butyl 5-amino-1 H-benzo[d]imidazole-1 -carboxylate (0.92g, 3.975mmol) in xylene (30mL) was added to a solution of ethyl 3-(4-(3,3-difluoropropoxy)-2,6-difluorophenyl)-3- oxopropanoate (1 .6g, 4.968mmol) and pyridine (0.5mL) in Xylene (30mL), Yield 2.0g crude. Step D: Sodium borohydride (297mg, 7.858mmol), N-(1 -tert-butyl-oxycarbonyl-1 H- benzo[d]imidazole-5-yl-)3-(4-(3,3-difluoropropoxy)-2,6-diflu orophenyl)-3-oxopropanamide (2g, 3.929mmol) in mixture of Tetrahydrofuran (21 mL) and Methanol (9m L) at 0°C, Yield 1 .3g (53.62%)

Step E: Diethylazodicarboxylate (0.6mL, 4.07mmol), N-(1 -tert-butyl-oxycarbonyl-1 H- benzo[d]imidazole-5-yl-)3-(4-(3,3-difluoropropoxy)-2,6-diflu orophenyl)-3-hydroxypropanamide (1 .3g, 2.54mmol) and Triphenylphosphine (1 g, 4.07mmol) Tetrahydrofuran (30mL) at 0°C- Yield: 0.75g crude.

Step F: Trifluoroaceticacid acid (1 mL) was added to a solution of crude tert-butyl 6-(2-(4-(3,3- difluoropropoxy)-2,6-difluorophenyl)-4-oxoazetidin-1 -yl)-1 H-benzo[d]imidazole-1 -carboxylate (2.5g, 5.08mmol) in Dichloromethane (10mL) at 0°C, Purification by preparative TLC afforded 70mg of rac4-(4-(3,3-difluoropropoxy)-2,6-difluorophenyl)-1 -(1 H-benzo[d]imidazol-5- yl)azetidin-2-one as pale yellow solid. This was purified by Chiral prep HPLC using the following conditions: Column: Chiralcel-OX-H (250 * 30 * 5.0μ); Mobile phase: A: Hexane (0.1 % DEA); Ethanol (75:25); Flow rate: 30mlJ min; Diluent: Mobile phase.The prep fractions were evaporated in vacuo to afford 28mg and 18mg of the respective enantiomers

(R)-4-(4-(3,3-difluoropropoxy)-2fi-difluorophenyl)- 1-(1H-benzo[d]im

(9)

1 H-NMR (DMSO-d6): δ 12.42(s, 1 H); 8.18(s, 1 H); 7.53(d, 1 H); 7.33(s, 1 H); 7.15(bs, 1 H); 6.82(d, 2H); 6.19(bt, 1 H); 5.49(d, 1 H); 4.13(t, 2H); 3.62-3.58(m, 1 H); 3.22(q, 1 H); 2.33-2.22(t, 3H); MS=394(M+1 ); HPLC [B]~99.51 %: Chiral HPLC~99.63%.

(S)-4-(4-(3,3-difluoropropoxy)-2fi-difluorophenyl)-1-(1H- benzo[d]imidazo

(10)

1 H-NMR (DMSO-d6): δ 12.42(s, 1 H); 8.16(s, 1 H); 7.53(d, 1 H); 7.33(s, 1 H); 7.15(bs, 1 H); 6.82(d, 2H); 6.19(bt, 1 H); 5.49(s, 1 H); 4.13(t, 2H); 3.62-3.59(m, 1 H); 2.30(q, 2H); MS=393.9(M+1 ); HPLC [B]~99.66%: Chiral HPLC~99.62%.

Examples 11 and 12

rac4-(4-(2,2-difluoropropoxy)-2-fluorophenyl)- 1-( 1H-benzo[d]imidazol-5-yl)azetidin-2-one

Step A: Chloro trimethylsilane (0.81 mL, 6.88mmol), zinc dust (1 .26g, 19.26mmol), benzene (30mL), Ethyl bromo acetate (1 .9mL, 16.51 mmol), 4-(2,2-difluoropropoxy)-2- fluorobenzaldehyde (3g, 13.76mmol) in dry benzene (20mL), Yield: 3g (75%)

Step B: Pyridinium dichromate (13.80g, 36.72mmol), Molecular sieves (15g) were added to a solution of ethyl 3-(4-(2,2-difluoropropoxy)-2-fluorophenyl)-3-hydroxypropanoa te (2.8g, 9.18mmol) in dichloromethane (400mL), Yield 2.2g (79%)

Step C: tert-butyl 5-amino-1 H-benzo[d]imidazole-1 -carboxylate (830mg, 3.56mmol) in xylene (30mL) was added to a solution of ethyl 3-(4-(2,2-difluoropropoxy)-2-fluorophenyl)-3- oxopropanoate (1 .2g, 3.96mmol) and pyridine (0.5mL) in Xylene (30mL), Yield 1 .8g crude. Step D: Sodium borohydride (280mg, 7.33mmol), N-(1 -tert-butyl-oxycarbonyl-1 H- benzo[d]imidazole-5-yl-)3-(4-(2,2-difluoropropoxy)-2-fluorop henyl)-3-oxopropanamide (1 .8g, 3.66mmol) in mixture of Tetrahydrofuran (21 mL) and Methanol (9m L) at 0°C, Yield 0.75g (42.2%)

Step E: Diethylazodicarboxylate (0.36mL, 2.27mmol), N-(1 -tert-butyl-oxycarbonyl-1 H- benzo[d]imidazole-5-yl-)3-(4-(2,2-difluoropropoxy)-2-fluorop henyl)-3-hydroxypropanamide

(700mg, 1 .42mmol) and Triphenylphosphine (700mg, 1 .42mmol) Tetrahydrofuran (30mL) at 0°C- Yield: 1 .0g crude

Step F: Trifluoroaceticacid acid (4m L) was added to a solution of crude tert-butyl 6-(2-(4-(2,2- difluoropropoxy)-2-fluorophenyl)-4-oxoazetidin-1 -yl)-1 H-benzo[d]imidazole-1 -carboxylate (1 .0g, 2.08mmol) in Dichloromethane (20mL) at 0°C, Yield: 0.13g (40%). Same synthesis (on same quantities) was repeated to afford another 120mg of rac4-(4-(2,2-difluoropropoxy)-2- fluorophenyl)-1 -(1 H-benzo[d]imidazol-5-yl)azetidin-2-one. Both combined~250mg and purified by Chiral prep HPLC using the following conditions:

Column: Chiralcel-OX-H (250 * 30 * 5.0μ); Mobile phase: A: Hexane (0.1 % DEA); Ethanol (75:25); Flow rate: 30mlJ min; Diluent: Mobile phase.

The prep fractions were evaporated in vacuo and triturated with diethyl ether to afford 70mg of each isomer respectively.

(R)-4-(4-(2,2-difluoropropoxy)-2-fluorophenyl)- 1-(1H-benzo[d]imidazol-5-yl)azeM

(11 )

Melting range: 165-170°C; 1 H-NMR (DMSO-d6): δ 12.35(d, 1 H); 8.16(d, 1 H); 7.55(d, 1 H); 7.47-7.32(m, 2H); 7.07(q, 1 H); 6.87(q, 1 H); 5.37(d, 1 H); 4.29(t, 2H); 3.63-3.58(m, 1 H); 3.00(q, 1 H); 1 .71 (t, 3H); MS=376(M+1 ); HPLC [B]~99.17%: Chiral HPLC~99.79%. (S)-4-(4-(2,2-difluoropropoxy)-2-fluorophenyl)-1-(1H-benzo[d ]imidazol-5-yl)azetidin^

(12)

Melting range: 203-208°C; 1 H-NMR (DMSO-d6): δ 12.37(d, 1 H); 8.14(s, 1 H); 7.59-7.37(m, 3H); 7.32(bs, 1 H); 7.01 (dd, 1 H); 6.85(dd, 1 H); 5.37(q, 1 H); 4.29(t, 2H); 3.63-3.58(m, 1 H); 3.00(q, 1 H); 1 .70(t, 3H); MS=376(M+1 ); HPLC [B]~98.39%: Chiral HPLC~96.69%. Examples 13 and 14

Rac-4-(4-(2,2-difluoropropoxy)-2,3-difluorophenyl)-1-(1H-ben zo[d]im

one

Step A: Chloro trimethylsilane (0.79mL, 6.35mmol), zinc dust (1 .2g, 19.06mmol), benzene (30mL), Ethyl bromo acetate (1 .6mL, 15.25mmol), 4-(2,2-difluoropropoxy)-2,3- difluorobenzaldehyde (3g, 12.7mmol) in dry benzene (20mL), Yield: 3.1 g (79%)

Step B: Pyridinium dichromate (13.92g, 37.03mmol), was added to a solution of ethyl 3-(4- (2,2-difluoropropoxy)-2,3-fluorophenyl)-3-hydroxypropanoate (3g, 9.25mmol) in dichloromethane (400mL), Yield 1 .2 g (40%)

Step C: tert-butyl 5-amino-1 H-benzo[d]imidazole-1 -carboxylate (0.72g, 3.03mmol) in xylene (30mL) was added to a solution of ethyl 3-(4-(2,2-difluoropropoxy)-2,3-difluorophenyl)-3- oxopropanoate (1 .2g, 3.77mmol) and pyridine (0.5mL) in Xylene (30mL, Yield 1 .8g crude. Step D: Sodium borohydride (268mg, 7.07mmol), N-(1 -tert-butyl-oxycarbonyl-1 H- benzo[d]imidazole-5-yl-)3-(4-(2,2-difluoropropoxy)-2,3-diflu orophenyl)-3-oxopropanamide (1 .8g, 3.5mmol) in mixture of Tetrahydrofuran (21 mL) and Methanol (9mL) at 0°C, Yield 1 .2g (67.0 %) Step E: Diethylazodicarboxylate (0.6ml_, 3.67mmol), N-(1 -tert-butyl-oxycarbonyl-1 H- benzo[d]imidazole-5-yl-)3-(4-(2,2-difluoropropoxy)-2,3-fluor ophenyl)-3-hydroxypropanamide (1 .2g, 2.43mmol) and Triphenylphosphine (0.96g, 3.67mmol) Tetrahydrofuran (30mL) at 0°C- Yield: 0.4g crude.

Step F: Trifluoroaceticacid acid (1 ml_) was added to a solution of crude tert-butyl 6-(2-(4-(2,2- difluoropropoxy)-2,3-difluorophenyl)-4-oxoazetidin-1 -yl)-1 H-benzo[d]imidazole-1 -carboxylate (400mg, 0.81 1 mmol) in Dichloromethane (10mL) at 0°C, Yield: 0.07g (22%).

This was purified by Chiral prep HPLC using the following conditions: Column: Chiralcel-OX- H (250 * 30 * 5.0μ); Mobile phase: A: Hexane (0.1 % DEA); Ethanol (75:25); Flow rate: 30mlJ min; Diluent: Mobile phase. The prep fractions were evaporated in vacuo to afford 30mg and 34mg of the respective enantiomers.

(R)-4-(4-(2,2-difluoropropoxy)-2,3-difluorophenyl)- 1-(1H-benzo[d]imidazol-5-yl)azetidi

(13)

Melting range: 173-177°C; 1 H-NMR (DMSO-d6): δ 12.35(d, 1 H); 8.15(s, 1 H); 7.55-7.36(m, 2H); 7.22(t, 1 H); 7.10(t, 1 H); 5.44(d, 1 H); 4.39(t, 2H); 3.66-3.61 (m, 1 H); 3.00(q, 1 H); 1 .71 (t, 3H); MS=394(M+1 ); HPLC [B]~98.55%: Chiral HPLC~99.94%.

(S)-4-(4-(2,2-difluoropropoxy)-2,3-difluorophenyl)-1-(1H- benzo[d]imidazol-5-yl)azetidin

(14)

Melting range: 170-174°C; 1 H-NMR (DMSO-d6): δ 12.35(d, 1 H); 8.15(s, 1 H); 7.55-7.36(m, 3H); 7.22(t, 1 H); 7.10(t, 1 H); 5.42(d, 1 H); 4.40(t, 2H); 3.66-3.61 (m, 1 H); 3.00(q, 1 H); 1 .71 (t, 3H); MS=394(M+1 ); HPLC [B]~99.15%: Chiral HPLC~98.09%.

Examples 15 and 16

Rac-4-(4-(2,2-difluoropropoxy)-2^-difluorophenyl)- 1-(1H-benzo[d]imidazol-5-yl)azetidi one

Step A: Chloro trimethylsilane (0.7mL, 5.8mmol), zinc dust (1 g, 16.3mmol), benzene (30mL), Ethyl bromo acetate (2.33g, 13.9mmol), 4-(2,2-difluoropropoxy)-2,6-difluorobenzaldehyde (2.7g, 1 1 .6mmol) in dry benzene (20mL), Yield: 2.4g (67%).

Step B: Pyridinium dichromate (1 1 g, 29.6mmol), Molecular sieves (1 1 g) 3-(4-(2,2- difluoropropoxy)-2,6-fluorophenyl)-3-hydroxypropanoate (2.4g, 7.40mmol) in dichloromethane (400mL), Yield 1 .7g (71 ,2%)

Step C: tert-butyl 5-amino-1 H-benzo[d]imidazole-1 -carboxylate (984mg, 4.2mmol) in xylene (30mL) was added to a solution of ethyl 3-(4-(2,2-difluoropropoxy)-2,6-difluorophenyl)-3- oxopropanoate (1 .7g, 5.2mmol) and pyridine (1 .0 mL) in Xylene (30mL), Yield 1 .4g crude. Step D: Sodium borohydride (209mg, 5.5mmol), N-(1 -tert-butyl-oxycarbonyl-1 H- benzo[d]imidazole-5-yl-)3-(4-(2,2-difluoropropoxy)-2,6-diflu orophenyl)-3-oxopropanamide (1 .4g, 2.7mmol) in mixture of Tetrahydrofuran (14ml_) and Methanol (6ml_) at 0°C, Yield 1 .3g (94%).

Step E: Diethylazodicarboxylate (0.5mL, 3.2mmol), N-(1 -tert-butyl-oxycarbonyl-1 H- benzo[d]imidazole-5-yl-)3-(4-(2,2-difluoropropoxy)-2,6-diflu orophenyl)-3-hydroxypropanamide (1 .1 g, 2.15mmol) and Triphenylphosphine (845mg, 3.2mmol) Tetrahydrofuran (30mL) at 0°C- Yield: 1 .6g crude.

Step F: Trifluoroaceticacid acid (4m L) was added to a solution of crude tert-butyl 6-(2-(4-(2,2- difluoropropoxy)-2,6-difluorophenyl)-4-oxoazetidin-1 -yl)-1 H-benzo[d]imidazole-1 -carboxylate (1 .6g ) in Dichloromethane (20ml_) at 0°C. Yield: 0.3g of rac 4-(4-(2,2-difluoropropoxy)-2,6- difluorophenyl)-1 -(1 H-benzo[d]imidazol-5-yl)azetidin-2-one. This was purified by Chiral prep HPLC using the following conditions: Column: Chiralcel-OX-H (250 * 30 * 5.0μ); Mobile phase: A: Hexane (0.1 % DEA); Ethanol (75:25); Flow rate: 30mL/ min; Diluent: Mobile phase.The prep fractions were evaporated in vacuo and triturated with diethyl ether to afford 70mg of each enantiomer respectively.

(R)-4-(4-(2,2-difluoropropoxy)-2fi-difluorophenyl)- 1-(1H-benzo[d]imid^

(15)

Melting range: 158-162°C; 1 H-NMR (DMSO-d6): δ 12.40(d, 1 H); 8.15(d, 1 H); 7.57-7.46(m, 1 H); 7.38(s, 1 H); 7.28(t, 1 H); 6.91 (d, 2H); 5.45(d, 1 H); 4.33(t, 2H); 3.62-3.58(m, 1 H); 3.24(q, 1 H); 1 .69(t, 3H); MS=394(M+1 ); HPLC [B]~97.07%: Chiral HPLC~99.51 %.

(S)-4-(4-(2,2-difluoropropoxy)-2fi-difluorophenyl)-1-(1H- benzo[d]imidazo

(16)

Melting range: 163-166°C; 1 H-NMR (DMSO-d6): δ 12.40(d, 1 H); 8.14(bs, 1 H); 7.55-7.26(m, 2H); 7.04(s, 1 H); 6.90(d, 2H); 5.45(d, 1 H); 4.32(t, 2H); 3.63-3.58(m, 1 H); 3.24(d, 1 H); 1 .69(t, 3H); MS=394(M+1 ); HPLC [B]~98.92%: Chiral HPLC~99.69%. Activity screening

Fluorometric assays

All measurements were performed with a BioAssay Reader HTS-7000Plus for microplates (Perkin Elmer) at 30 °C. QC activity was evaluated fluorometrically using H-Gln- ?NA. The samples consisted of 0.2 mM fluorogenic substrate, 0.25 U pyroglutamyl aminopeptidase (Unizyme, Horsholm, Denmark) in 0.2 M Tris/HCI, pH 8.0 containing 20 mM EDTA and an appropriately diluted aliquot of QC in a final volume of 250 μΙ. Excitation/emission wavelengths were 320/41 0 nm. The assay reactions were initiated by addition of glutaminyl cyclase. QC activity was determined from a standard curve of ?-naphthylamine under assay conditions. One unit is defined as the amount of QC catalyzing the formation of 1 μηιοΙ pGlu- ?NA from H- Gln-y5 A per minute under the described conditions.

In a second fluorometric assay, QC was activity determined using H-Gln-AMC as substrate. Reactions were carried out at 30 °C utilizing the NOVOStar reader for microplates (BMG labtechnologies). The samples consisted of varying concentrations of the fluorogenic substrate, 0.1 U pyroglutamyl aminopeptidase (Qiagen) in 0.05 M Tris/HCI, pH 8.0 containing 5 mM EDTA and an appropriately diluted aliquot of QC in a final volume of 250 μΙ. Excitation/emission wavelengths were 380/460 nm. The assay reactions were initiated by addition of glutaminyl cyclase. QC activity was determined from a standard curve of 7-amino- 4-methylcoumarin under assay conditions. The kinetic data were evaluated using GraFit sofware.

Spectrophotometric assay of QC

This novel assay was used to determine the kinetic parameters for most of the QC substrates. QC activity was analyzed spectrophotometrically using a continuous method, that was derived by adapting a previous discontinuous assay (Bateman, R. C. J. 1989 J Neurosci Methods 30, 23-28) utilizing glutamate dehydrogenase as auxiliary enzyme. Samples consisted of the respective QC substrate, 0.3 mM NADH, 14 mM oc-Ketoglutaric acid and 30 U/ml glutamate dehydrogenase in a final volume of 250 μΙ. Reactions were started by addition of QC and persued by monitoring of the decrease in absorbance at 340 nm for 8-15 min.

The initial velocities were evaluated and the enzymatic activity was determined from a standard curve of ammonia under assay conditions. All samples were measured at 30 °C, using either the SPECTRAFIuor Plus or the Sunrise (both from TECAN) reader for microplates. Kinetic data was evaluated using GraFit software.

Inhibitor assay

For inhibitor testing, the sample composition was the same as described above, except of the putative inhibitory compound added. For a rapid test of QC-inhibition, samples contained 4 mM of the respective inhibitor and a substrate concentration at 1 KM. For detailed investigations of the inhibition and determination of K-values, influence of the inhibitor on the auxiliary enzymes was investigated first. In every case, there was no influence on either enzyme detected, thus enabling the reliable determination of the QC inhibition. The inhibitory constant was evaluated by fitting the set of progress curves to the general equation for competitive inhibition using GraFit software. The inhibitor assay was performed at two different pH levels, pH 6.0 and pH 8.0: The respective pH value in the assay solution was adjusted using conventional methods.

Pharmacokinetic parameters

Methods

Three mice (strain CD-1 ) were administered orally 30mg/kg of each test compound dissolved in 0.8% Methocel. Samples were taken at the time points 10 min, 0.5, 1 , 2, 4 and 8hr after test compound administration for plasma and brain collection.

Blood collection

The mice were anesthetized with Isoflurane. Approximate 200 μΙ_ of each blood sample were collected via cardiac puncture for terminal bleeding into K2EDTA tubes. Blood samples were put on ice and centrifuged at 2000 g for 5 min to obtain plasma sample within 15 minutes. CSF collection:The animals were euthanized with pure CO2 inhalation. A mid line incision was made on the neck. The muscle under the skin was cut to expose the cisterna magna. The cisterna magna was penetrated with the sharp end of a capillary and CSF was collected via capillarity.

Brain collection

After CSF collection, a perfusion with 7x total mouse blood volume (approximate 15 ml) of ice- cold PBS (pH 7.4) was conducted via cardiac puncture before brain collection. A mid-line incision was made in the animal scalp. The brain was removed and rinsed with cold saline. Brain was placed into a screw-top tube and weighed. Brain samples were homogenized for 2 min with 3 volumes (v/w) of PBS (pH 7.4) and then analyzed with LC-MS/MS. The brain concentration was corrected with a dilution factor of 4 as following:

Brain concentration = brain homogenate cone, x 4, assuming 1 g wet brain tissue equals to 1 ml.

Plasma, brain and CSF samples were stored at approximately -80°C until analysis. Sample preparation

For plasma samples: An aliquot of 20 μΙ sample was added with 200 μ IS (Diclofenac, 200 ng/mL) in ACN, the mixture was vortexed for 2 min and centrifuged at 12.000 rpm for 5 min. 1 μΙ supernatant was injected for LC-MS/MS analysis.

For diluted plasma samples: An aliquot of 4 μΙ sample was added with 16μΙ blank plasma, mixed well, added with 200 μΙ IS (Diclofenac, 200 ng/ml) in ACN.

For Brain samples: Brain tissue was homogenized for 2 min with 3 volumes (v/w) of PBS. An aliquot of 20 μΙ sample was added with 200 μΙ IS (Diclofenac, 200 ng/ml) in ACN, the mixture was vortexed for 2 min and centrifuged at 12000 rpm for 5 min. 1 μΙ supernatant was injected for LC-MS/MS analysis.

For CSF samples: The CSF sample was added with corresponding 20 fold volume of IS (Diclofenac, 200 ng/ml) in ACN, the mixture was vortexed for 2 min. 3μΙ supernatant was injected for LC-MS/MS analysis.

Tmax, T1/2, AUC and logBB values were calculated using conventional methods.

Results

Ki

Ki

hQPCTL

Example hQC pH6,0

pH6

[nM]

[nM]

1 170 33.4

2 20.8 6.9

4 8.6 2.5

5 3.3 0.86

7 2.4 1 .1

9 2.4 0.2

1 1 3.8 1 .3

13 2.3 2.0

15 3.6 0.3 Example compounds 4, 5, 7, 9, 1 1 , 13 and 15 showed a good blood-brain-barrier crossing behaviour and AUC-values in the brain, which support their use in the treatment of neurodegenerative diseases.

Analytical methods

Analytical HPLC

Method [A]: The analytical HPLC-system consisted of a Agilent MSD 1 100 utilizing a Waters SunFire RP 18 (2,5 μηι), analytical column (length: 50 mm, diameter: 2.1 mm), and a diode array detector (DAD) with λ = 254 nm as the reporting wavelength. The compounds were analyzed using a gradient at a flow rate of 0.6 mL/min; whereby eluent (A) was acetonitrile, eluent (B) was water and eluent (C) 2% formic acid in acetonitrile applying the following gradient:

The purities of all reported compounds were determined by the percentage of the peak area at 214 nm.

Method [B]: The analytical HPLC-system consisted of a Agilent MSD 1 100 utilizing a Waters SYMETRY RP 18 (3,5 μηι), analytical column (length: 75 mm, diameter: 4.6 mm), and a diode array detector (DAD) with λ = 254 nm as the reporting wavelength. The compounds were analyzed using a gradient at a flow rate of 1 .0 mL/min; whereby eluent (A) was acetonitrile, eluent (B) was water 0.01 M % ammonium acetate:

Time min % Solvent A % Solvent B

0 10 90

2 10 90

6 90 10 18 90 10

19 10 90

20 10 90

Chiral HPLC

The analytical chiral HPLC-system consisted of a Agilent MSD 1 100 utilizing a Waters Chiracel OX-H (5 μηι), analytical column (length: 250 mm, diameter: 4.6 mm), and a diode array detector (DAD) with λ = 254 nm as the reporting wavelength. The compounds were analyzed using an isocratic mixture of 0.1 %DEA in Hexane and Ehanole (70/30) at a flow rate of 1 .0 mL/min.

Mass-spectrometry, NMR-spectroscopy

ESI-Mass spectra were obtained with a SCIEX API 365 spectrometer (Perkin Elmer) utilizing the positive ionization mode.

The 1 H NMR-Spectra (500 MHz) were recorded at a BRUKER AC 500. The solvent was DMSO-De, unless otherwise specified. Chemial shifts are expressed as parts per million (ppm) downfiled from tetramethylsilan. Splitting patterns have been designated as follows: s (singulet), d (doublet), dd (doublet of doublet), t (triplet), m (multiplet) and br (broad signal). Mass-spectrometry, NMR-spectroscopy:

ESI-Mass spectra were obtained with a SCIEX API 365 spectrometer (Perkin Elmer) utilizing the positive ionization mode.

The 1 H NMR-Spectra (500 MHz) were recorded at a BRUKER AC 500. The solvent was DMSO-De, unless otherwise specified. Chemial shifts are expressed as parts per million (ppm) downfiled from tetramethylsilan. Splitting patterns have been designated as follows: s (singulet), d (doublet), dd (doublet of doublet), t (triplet), m (multiplet) and br (broad signal).

MALDI-TOF mass spectrometry

Matrix-assisted laser desorption/ionization mass spectrometry was carried out using the Hewlett-Packard G2025 LD-TOF System with a linear time of flight analyzer. The instrument was equipped with a 337 nm nitrogen laser, a potential acceleration source (5 kV) and a 1 .0 m flight tube. Detector operation was in the positive-ion mode and signals are recorded and filtered using LeCroy 9350M digital storage oscilloscope linked to a personal computer. Samples (5 μΙ) were mixed with equal volumes of the matrix solution. For matrix solution DHAP/DAHC was used, prepared by solving 30 mg 2 ' ,6 ' -dihydroxyacetophenone (Aldrich) and 44 mg diammonium hydrogen citrate (Fluka) in 1 ml acetonitrile/0.1 % TFA in water (1/1 , v/v). A small volume (~ 1 μΙ) of the matrix-analyte-mixture was transferred to a probe tip and immediately evaporated in a vacuum chamber (Hewlett-Packard G2024A sample prep accessory) to ensure rapid and homogeneous sample crystallization.

For long-term testing of Glu 1 -cyclization, Αβ-derived peptides were incubated in 10ΟμΙ 0.1 M sodium acetate buffer, pH 5.2 or 0.1 M Bis-Tris buffer, pH 6.5 at 30 °C. Peptides were applied in 0.5 mM [Ap(3-1 1 )a] or 0.15 mM [Ap(3-21 )a] concentrations, and 0.2 U QC is added all 24 hours. In case of A (3-21 )a, the assays contained 1 % DMSO. At different times, samples are removed from the assay tube, peptides extracted using ZipTips (Millipore) according to the manufacturer ' s recommendations, mixed with matrix solution (1 :1 v/v) and subsequently the mass spectra recorded. Negative controls either contain no QC or heat deactivated enzyme. For the inhibitor studies the sample composition was the same as described above, with exception of the inhibitory compound added (5 mM or 2 mM of a test compound of the invention). Compounds and combinations of the invention may have the advantage that they are, for example, more potent, more selective, have fewer side-effects, have better formulation and stability properties, have better pharmacokinetic properties, be more bioavailable, be able to cross blood brain barrier and are more effective in the brain of mammals, are more compatible or effective in combination with other drugs or be more readily synthesized than other compounds of the prior art.

Throughout the specification and the claims which follow, unless the context requires otherwise, the word 'comprise', and variations such as 'comprises' and 'comprising', will be understood to imply the inclusion of a stated integer, step, group of integers or group of steps but not to the exclusion of any other integer, step, group of integers or group of steps.

All patents and patent applications mentioned throughout the specification of the present invention are herein incorporated in their entirety by reference. The invention embraces all combinations of preferred and more preferred groups and embodiments of groups recited above.